Abstracts from the 57th European Society of Human Genetics (ESHG) Conference: e-Posters (2025)

Table of Contents
EP01 Cancer Genetics Olga Rodenko1, Anke Esber1, Irina Cepraga1, Stefanie Schütze1, Lars Jansen1, Mieczyslaw Romuald Gajda2, Nikolaus Gaßler2, Ingo B. Runnebaum1, Matthias Dürst1, Norman Häfner1, Claudia Backsch 1 Selin Fulya Toprak Yalçın 1, Esra Nur Demirtaş1, Tugba Sarac-Sivrikoz2, Selcuk Sözer Tokdemir1 Albertas Valintelis 1, Ieva Drejerienė1, Matas Norvydas1, Jūratė Gruodė1;2 Fotini Ieremiadou1, Kostantinos Tyrosvoutis2, Aspasia Divane 3 Mónica Arranz Ledo 1, Enrique Lastra Aras2, Amaya Olaverri Hernández3, Marta Orozco Belinchón3, Enrique Pérez Riesgo1, Lara Hernández Sanz1, Noemí Martínez Martín1, Mar Infante Sanz1, Mercedes Durán Domínguez1 Marija Staninova Stojovska 1, Elizabeta Krstevska Bozhinovkj1, Nadica Matevska-Geshkovska1, Nenad Mitrevski2, Biljana Grozdanovska2, Milco Panovski2, Aleksandar Dimovski1;3 Ieva Drejeriene 1, Matas Norvydas1, Albertas Valintelis1, Jūratė Gruodė1;2 Ian Babler-Madrid 1, Nara Sobreira1, Renan Martin1, Elizabeth Wohler1 Gulshara Abildinova1, Zhanna Zhabakova 1, Anna Borovikova1 Vincenzo Rallo 1, Matteo Massidda2, Xiaofen Wen3, Jiaxin Shen3, Manila Deiana1, Andrea Maschio1, Antonio Mario Scanu2, Maria Rosaria De Miglio2, Andrea Angius1 Agnieszka Podgorska 1, Ewa Kwiatkowska1, Renata Lotocka1, Pawel Wiechno2, Katarzyna Seliga1, Aleksandra Gos1, Katarzyna Zielinska-Musko1, Lukasz Fulawka1, Aleksandra Kuzniar1, Kinga Koszucka1, Andrzej Tysarowski1 marion rolain1, patricia faure1, Nathalie Parodi1, Maud Brauchaud1, Margaux Clement Le Choismier1, Stéphanie Baert-Desurmont2, Edwige Kasper1, Gaelle Bougeard2, Mariette Renaux-Petel3, Thomas Vermeulin4, Théry Jean-Christophe5, Claude Houdayer 1 Paula García-Vallés 1;2;3, Jésica Pérez-García1;2;3, Paloma Martín-Bejarano Soto1;2;3, Rosario Vidal-Tocino1;4, Óscar Javier Blanco Múñez5, Jose Perea1, Ana-Belén Herrero1;2;3, Rogelio González-Sarmiento1;2;3 Maria Michelli 1;1, Eirini Roupou1, Ioanna Giannopoulou1, Niki Prekete1, NIKOLAOS KAVANTZAS1, Angelica a Saetta1 Jeanette YUEN 1, Malikka Venkatramani1, Si Qin Zhou1, Jianbang Chiang1, Sock Hoai Chan1, Joanne Ngeow1;2 Pia Pužar Dominkuš 1, Marija Rogar1;2, Vita Dolzan1, Petra Hudler2 Tanja Blagus 1, Ana Mervič2, Katja Goricar1, Danijela Strbac3, Alenka Franko2;4, Viljem Kovac2;3, Vita Dolzan1 Maria Isabel Alvarez 1;2, Paula Trilla1, Marta Gracia1, Laura Muñoz1, Maria Pellise3, Belen Pastor Piñera3;4, Lorena Moreno3;4, Teresa Ocaña3;4, Francesc Balaguer3;4, Celia Badenas1;2 Alenka Bombač 1, Mojca Unk2, Vida Stegel1, Srdjan Novaković1 Anca Pavel 1, Petruta Gurban1, Andreea Tutulan-Cunita1, Danai Stambouli1 Hubert Chen 1 Nuria Arroyo-Garrapucho 1;2;3;4, Vera Sommers4, Hanne Willems4, Kaat Peperstraete4, Vanessa Dubois5, Ana-Belén Herrero1;2;3, Rogelio González-Sarmiento1;2;3, Frank Claessens4 Ava Kwong1;2;3, Chun Hang Au 4, Dona N Ho4, Elaine YL Wong4, Henry CM Leung4, Amy WS Leung4, Janet HY Law4, Fian BF Law4, Sze Keong Tey5, Cecilia YS Ho4, Edmond SK Ma3;4 Trayan Delchev 1, Tsvetina Veleva1, Maria Sredkova-Ruskova1, Nevyana Ivanova2, Kalina Mihova2, Radka Kaneva2, Daniela Avdjieva-Tzavella1 Svenja Holst1, Friedegund Meier2, Jörg Otte3, Patrick Petzsch4, Julia Reifenberger5, Thorsten Wachtmeister4, Dana Westphal2, Mirjana Ziemer6, Wasko Wruck3, James Adjaye3, Regina C. Betz7, Harald Surowy1, Arno Ruetten8, Silke Redler 1 Faezeh Ghazvini Zadegan 1, Franziska Fiedler1, Tino Schenk1 Merita Xhetani 1, Albina Hasa2, Blerta Laze3 Beatriz Hidalgo Calero 1, José Manuel Sánchez Zapardiel1, Sonia Rodriguez Novoa2, Montserrat de Miguel1, Ricardo Blázquez Martín2, Marina Ibáñez Vizcaíno1, Francisca Luengo Sainz de Baranda1, Victoria Carrero Blázquez1, Ainhoa Almeida Santiago2, Anna Esteve-Codina3, Luis Robles Díaz4 Jaouhara MAAMAR 1;2, Rajaa Chahboune1, Ouafae Kaissi2, Hasna Hamdaoui2, Badreddine Nouadi2, Fatima Zahra Laarabi1;2, Houda Jelti1;2, Sabrine Bouressas1;2, Abdelhaq Lamaibdel1;2, Oussama Kettani2, Rabie Rahhali3, Nabila Sellal4;5, Mounia Amzerin3;4, Mohamed El Hfid4;5, Fatima Zahra El m’rabet3;4, Afaf Lamzouri1;2 Clara Venegas 1;2;3, Elisa Izquierdo1;2;3, Maria Pilar Casares1, Beatriz Ruz1;2;3, Alicia de Pablo1, Elixabet Lopez-Lopez4, Angela Gutierrez-Camino5, Antonio Pérez2;3;6, Alvaro Martinez1, ADELA ESCUDERO1;2;3 Ieva Vaicekauskaitė1;2, Raimonda Kubiliūtė1;2, Kristina Žukauskaitė1;2, Algirdas Žalimas1;2, Albertas Ulys1, Rasa Sabaliauskaite1;2, Sonata Jarmalaite 1;2 varol güler 1, Funda Özkök2, Başak Günçer3, Sacide Pehlivan4 Hafdis T Helgadottir 1;2, Pär Lundin3, Annika Lindblom1;2 Magda Mielczarek 1, Bartosz Czech1, Rafał Stępień1, Joanna Szyda1 Anzhelika Chegodar 1, Natalia Bodunova1, Natalia Oparina1;2, Chinchi Babadzhanova1;3, Larisa Tsapkova1, Nikolay Karnaukhov1, Ainash Altayeva4 Paulina Slawnikowska1, Tomasz Konecki2, Piotr Kutwin2;3, Zbigniew Jablonowski2, Agnieszka Zmysłowska1, Edyta Borkowska 1 Elisa Izquierdo1;2, Clara Venegas1;2, Beatriz Ruz1;2, Antonio Pérez1;3, Ana Patiño4;5, Teresa Imizcoz6, Gorka Alkorta6, Elena Panizo7, Sergio Manresa8, Lucas Moreno Martin Retortillo9;10, Adela Cañete11, Maria Teresa Tormo12, Josep Escriva12, Piedad Alba Pavón13, Angela Gutierrez-Camino13, Olatz Villate13, Manuel Ramirez Orellana14, Ana Chamorro14, Cristina Saiz15, Andrea Vilaplana8, Asbleidy Carolina Torres8, Aroa Soriano8, Hector Salvador16, Laura Martí17, Diana Salinas-Chaparro17, Estela Carrasco López18;19, ADELA ESCUDERO 1;2 Maribel Soler1, Clara Venegas1;2;3, Beatriz Ruz1;2;3, Borja González1, Virginia Contreras1, Alicia de Pablo1, Núria Rodríguez-Salas1, Dolores Corral1, Sonsoles San Roman1, Jesus Peralta1;2, Susana Noval1;2, Antonio Pérez1;2;3, ADELA ESCUDERO1;2;3, Elisa Izquierdo 1;2;3 Laura Roht 1;2, Piret Laidre2, Mikk Tooming1;3, Hanno Roomere3, Kadri Rekker3, Kadri Toome3, Olga Fjodorova3, Ülle Murumets3, Jaan Soplepmann4;5, Katrin Ounap1;2, Tiina Kahre1;3 Irene Vanni1, Lorenza Pastorino1;2, Virginia Andreotti1, Danila Comandini3, Giuseppe Fornarini3, Massimiliano Grassi3, Alberto Puccini3, Enrica Teresa Tanda2;4, Alessandro Pastorino3, Valentino Martelli3, Luca Mastracci5;6, Federica Grillo5;6, Francesco Cabiddu6, Antonio Guadagno6, simona coco7, Eleonora Allavena2, Francesca Barbero1, William Bruno1;2, Bruna Dalmasso1, Sara Erika Bellomo8, Caterina Marchiò8;9, Francesco Spagnolo10;11, Stefania Sciallero3, Enrico Berrino8;9, Paola Ghiorzo 1;2 Maria Hellberg 1 Larissa Dias de Souza1, Alexandre Defelicibus2, Diogo Soares3, José Rocha3, Daniele Paixão Pereira3, Dirce Carraro1, Giovana Torrezan 1 Maria Makarova 1, Marina Nemtsova1, Maria Byakhova2, Anastasia Danishevich3, Denis Chernevskiy1, Olesya Sagaidak1, Maria Vorontsova4, Maxim Belenikin1, Anastasia Krinitsina1, Aleksey Antonenko1, Anna Semenova2, Natalia Bodunova3, Igor Khatkov3, Vsevolod Galkin2 Gergana Stancheva 1, Ivan Boronsuzov2, Veronika Petkova1, Kalina Mihova1, Margarita Kamenova3, Prof. Dobrin Konstantinov2;4, Maya Yordanova2;4, Radka Kaneva1 Rasene Gereisha 1, Gabteni SANA1;2, Ahlem Achour1;2, Firas Akrout3, Rym Meddeb1;2, Carli Tops4, Richard Gallon5, Mariem Ben Rekaya6;7, Soumaya Rammeh6;7, Ridha Chkili3, Nada Mansouri8, Neila Belguith1;9, Ridha Mrad1;2 Miriam Carriero 1, Ludovico Graziani1, Emanuele Agolini2, Mario Bengala3, Antonio Novelli2, Michela Biancolella4, Giuseppe Novelli1;3 Joshua Kraindler 1, Rupendra Shrestha1, Owen Tan1, Jayamala Parmar1, Reanu Gopal1, Natalie Hart1, Vanessa Tyrrell2;3, Tracey O’Brien3;4, Deborah Schofield1 Deimante Usaite 1, Julija Simiene1, Kestutis Suziedelis1;2 Ieva Vaicekauskaitė1;2, Paulina Kazlauskaitė1;2, Rugilė Gineikaitė1;2, Ruta Ciurliene3, Juozas Rimantas Lazutka2, Rasa Sabaliauskaite 1;2 Marta Heise 1, Piotr Jarzemski2, Aneta Bąk1, Anna Junkiert-Czarnecka1, Olga Haus1 Elizabeth Córdoba-Lanús1;2;3, Omar García-Pérez 1;2, Leticia Melgar-Vilaplana2;4, Noelia Hernández Hernández5, Ricardo Fernández-de-Misa2;5;6 olfa tahri1, Sabriye Kocatürk Sel 2, oya ışık3, leyla uslu3, choubaila reddad1, Bertan YILMAZ2 Niki Prekete 1, Eirini Roupou1, Maria Michelli1, Vasiliki Chaidou1, Aphrodite Nonni1, NIKOLAOS KAVANTZAS1, Angelica a Saetta1 Marjaana Pussila 1, Aleksi Laiho2, Petri Törönen2, Pauliina Björkbacka3, Sonja Nykänen1, Kirsi Pylvänäinen4, Liisa Holm2, Jukka-Pekka Mecklin5, Laura Renkonen-Sinisalo6, Taru Lehtonen6, Anna Lepistö6, Jere Linden3, Satu Mäki-Nevala7, Päivi Peltomäki7, Minna Nyström8 Sanja Kiprijanovska 1, Hristina Dichevska1, Aleksandar Trifunovski2, Ognen Ivanovski2, Skender Saidi2, Zivko Popov1;2;3, Aleksandar Dimovski1;4 Nuket Y Kutlay 1, Halil Gürhan Karabulut1, TIMUR TUNCALI1, Hatice Ilgın Ruhi1, Sadiye Ekinci1, Şule Altıner1, Arzu Vicdan1, Ezgi Gökpınar İli1 Piedad Alba Pavón 1, Lide Alaña1, Teresa Imizcoz2, Miriam Gutierrez-Jimeno3, Aizpea Echebarria-Barona1;4, Ricardo López Almaraz1;4, Itziar Astigarraga1;4;5, Ana Patiño6, Olatz Villate1 Anna Byrjalsen 1, Sara Garcia2, Line Borgwardt1, Karin Wadt1, Anne-Marie Gerdes1, Thomas van Overeem Hansen1 Roya Gasimli 1, Asli Subasioglu2 Sara Ebrahimi 1, Haniye Rahimi Kolour1, Ehsan Nazemalhosseini Mojarad2, maryam Ghanbari-Safari3 Che Kang Lim 1;2, Yi Tian Png3, Lee Yew Mun3, Zhi Yong Lim3, Wei Ying Sim1, Chwee Ming Lim1;2;3 Manon Reda1, Anes Hadjadj1, Juliette Sauge1, Laurent Arnould1, Juliette Albuisson 1 Nadja Nikolic 1, Jelena Carkic1, Violeta Sango2, Nicole Riberti3, Boban Anicic4, Jelena Milasin5 Dragana Jugović 1, Marija Vukelic-Nikolic2, Višnja Madić3, Ljiljana Branković1, Radovan Milićević1, Perica Vasiljević4 Mustafa Kaya1, Ibrahim Kaya 2, Dilek Colak3 Emanuela Bostjancic 1, Kristian Urh1, Nina Zidar1 yiannos kyprianou 1 Maria Glushkova 1, Tihomir Todorov1, Svetlana Gacheva2, Albena Todorova1;3 Yalda Hekmatshoar 1, Arzu Zeynep Karabay2, Asli Koc2, Tulin Ozkan3, Asuman Sunguroglu3 Renata Posmyk 1, Joanna Karwowska1, Klaudia Berk1 Anna Maria Cueto-González 1;2, Alejandro Moles-Fernandez1;2, Maria Camprodón-Gómez3, Anna Casteras-Román4, Teresa Vendrell-BAyona1, Elena García-Arumí1;2, Eduardo Tizzano1;2 Iulia Simina 1;2, Iulia Teodora Perva2, Catalin-Vasile Munteanu1;3, Oana Cristina Voinea4;5, Adrian Trifa1;6;7 Milos Lazarevic1, Dijana Trisic1, Milica Jaksic-Karisik1, Drago Jelovac1, Nadja Nikolic1, Jelena Carkic1, Jelena Milasin 1 Jorge Docampo-Cordeiro 1, Fransico Jose Garcia-Iñigo1, Elena Jaime-Lara1, Marta Casas-Rodriguez1, Rosa DeFrutos-Curiel1, Ana Santos-Corral1, Maria Luisa Casas-Losada1 Asli Subasioglu 1, Roya Gasimli2 AYBERK TURKYILMAZ 1, KUBRA ADANUR SAGLAM1, Oguzhan Demir1, Mustafa Yilmaz1, Tuna Apuhan1, Alperhan Cebi1 Busra Yasa Cevik 1;2, gülşah tuna1;2, fadime didem can trabulus3, Selcuk Sözer Tokdemir1;2 marie bequin 1;2, Luisa Vergori1;2, Vincent Milon1;2, Omar Soukarieh3;4, Caroline Meguerditchian4, Fida Khater1;2, Jonathan Dauvé1;2, Yves Delneste1, Louise-Marie Chevalier1;2, David-Alexandre Tregouët4, Gaelle Bougeard5;6, Alain Morel1;2, Isabelle Tournier1;2 Natali Schachter- Safrai1;2, Inbal Kedar1, lihi atzmoni3;4, adel Shalata5, Dvora Kidron4;6, lina basel-salmon1;4;7, Zohar Levi4;8, Yael Goldberg 1;4 Maryam Safa Isini 1, Hossein Najmabadi2, ali Salehzadeh1, Farzam Ajamian3 Arwa Babai 1, Vicky Goh2;3, Sabrina Talukdar1;4, vishakha tripathi1;4, Anjana Kulkarni1;4, Louise Izatt1;4 Hakan Tomac 1, Aysel Kalayci2, Deniz Agirbasli2 LUISA AYELEN RAMOS NAVAS 1;2, Beatriz Esquivel Vázquez3, Eduardo Salido Ruiz1;2;3 Beatriz Esquivel Vázquez 1, LUISA AYELEN RAMOS NAVAS2, Eduardo Salido Ruiz1;2 Javier Manuel Galego Carro 1;2, Marta Santamariña1;2;3, Miguel E. Aguado-Barrera1;2, Jorge Amigo-Lechuga1;2, Olivia Fuentes1;2, Ana Crujeiras1;2, Carlos López1;2, Carla Coedo-Costa1;2, Antonio Gomez-Caamaño1;4, Ana Vega1;2;3 Başak Günçer 1, varol güler2, Sacide Pehlivan3, Ahmet Mesut Şentürk4, Bilge Özerman Edis1 Shamila Ladak 1;1, Fizza Akbar2, alizeh fatimi1, bassim zahid1, Salman Kirmani2 EP02 Reproductive Genetics Svetlana Yovinska 1, Mariela Hristova-Savova2, Yuri Buchvarov2, Petya Andreeva2, Tanya Timeva2, Atanas Shterev2, Rumen Nikolov1, Ivanka Dimova3 Emanuele Micaglio1, Arianna Riva2, Alice Tamma 2, Marco Villa1, Sara Benedetti1, Carlo Pappone1, Ugo Sorrentino2, Daniela Zuccarello2 Emanuele Micaglio 1, Arianna Riva2, Alice Tamma2, Marco Villa1, Ludovico Sabatelli1, Rosanna Cardani1, Sara Benedetti1, Carlo Pappone1, Ugo Sorrentino2, Daniela Zuccarello2 Slavena Davidova 1;2, Mariela Hristova-Savova2, Kalina Belemezova2;3, Petya Andreeva2;4, Atanas Shterev2, Jasmina Jeleva5, Ivanka Dimova2;3 Clayton Axiak 1, Francesca Borg-Carbott1, Ritienne Attard1, Clara Lazzaretti2, Carmela Perri2, Lara Baschieri2, Karen Cassar3, Mark Gruppetta3;4, Josanne Vassallo3;4;5, Livio Casarini2, Stephanie Bezzina Wettinger1;5, Rosienne Farrugia1;5 Giulia Di Cola 1, Giuseppe Castello1, Chiara Faggionato1, Simone Bolli2, Cinzia Di Monte2, Monica Mazzola2, Federica Cariati3, Claudia Cartolano4, Jessica Parrotta4, Alessandro Conforti3, Alessandra Andrisani1, Roberta Venturella4, Giulia Scaravelli2, Carlo Alviggi3, Daniela Zuccarello1 Michala Hrabíková 1, Štěpán Chvojka1, Leona Cerna1, Jan Král1, Filip Zembol1, Barbora Honysová1, Martina Bittoova1, Monika Koudová1 Nouha ABDELMOULA 1, Balkiss Abdelmoula1 Balkiss Abdelmoula 1, Khaled Trigui1, Wided Ltaif1, Kays Chaker1, Yassine Ouanes1, Nouha ABDELMOULA1 Marta Olszewska 1, Marzena Kamieniczna1, Nijole Pollock2, Zuzanna Graczyk1, Monika Fraczek1, Katarzyna Kurek1, Alexander N. Yatsenko2, Maciej Kurpisz1 Yusuf Bahap 1, Meral Yirmibes Karaoguz1, Esra Tuğ1, Thomas Liehr2, Meral Yirmibes Karaoguz1 Sweta Das 1;2, Rekha Aaron2, Paramasivam g2, GAUTHAM ARUNACHAL UDUPI3, Sumita Danda2 Jelena Pozojevic 1, Radhika Sivaprasad2, Joshua Laß3, Franziska Haarich4, Joanne Trinh3, Neseebullah Kakar1, Kristin Schulz1, Kristian Händler1, Annemarie Verrijn Stuart5, Jacques C. Giltay6, Koen L. I. van Gassen7, Almuth Caliebe2, Paul-Martin Holterhus8, Malte Spielmann1, Nadine Hornig2 Jasenka Wagner 1, Deni Plecko1, Nena Arvaj1, Kristina Kralik2, Mirta Kadivnik3 Andrei Kullyev 1, Svetlana Avdeichik1, Yaron Goikhman1 Katarina Davalieva1, Gjorgji Bozhinovski 1, Sanja Kiprijanovska1, Katerina Kubelka-Sabit2;3, Dijana Plaseska-Karanfilska1 Mariela Hristova-Savova 1, Petya Andreeva2, Ivelina Oprova2, Atanas Shterev2, Ivanka Dimova3 Liliia Chorna 1, Danuta Zastavna1, Yaryna Zahanyach1, Oksana Kolodiy2, Halyna Makukh3 Michaela Blankenburg 1, Oriane Vedrines1, Markus Stumm1 Maria-Anna Kyrgiafini 1, Alexia Chatziparasidou2, Nikolaos Christoforidis2, Zissis Mamuris1 Evelina Dagyte 1, Algirdas Utkus1 Radostina Raynova 1;2, Tanya Milachich3, Petya Andreeva4, Tanya Timeva4, Maria Yunakova4, Atanas Shterev4, Alexey Savov1, Ivanka Dimova2;5 Petya Andreeva 1;1;2, Ivelina Oprova1;2, Mariela Hristova-Savova3, Kalina Belemezova3, Ivanka Dimova4 Matthias Linke 1, Charlotte Hewel1, Bartosz Linek2, Christine Skala2, Susanne Gerber1, Susann Schweiger1 abir talbi 1 Daniil Ferdman 1, Ekaterina Vasileva1, Tutakov Maksim2, Daniil Chebotarev2, Svetlana Pavliuchenkova2, Natalia Isaeva1, Roman Bikanov1 Ekaterina Vasileva 1, Nataliia Andreeva1, Daniil Ferdman1, Natalia Isaeva1, Roman Bikanov1 O. Sena AYDOS 1, nazila farhangzad1;2, Tulin Ozkan1, Asuman Sunguroglu1, Kaan Aydos3 Yevheniya Sharhorodska 1;2;3, Anna Ulrich2, Liliia Chorna1, Ivanna Shymanska4, Danuta Zastavna1, Oleg-Roman Gnateyko1, Chiara Scapoli3, Halyna Makukh4, Inga Prokopenko2;5 Natàlia Pujol Gualdo 1;2, Minna Karjalainen3;4, Urmo Võsa1, Riikka K. Arffman2, Reedik Mägi1, Justiina Ronkainen3, Triin Laisk1, Terhi Piltonen2 Anastasia Maltseva 1, Roman Illarionov1, Elena Vashukova1, Olga Pachuliia1, Andrey Glotov1 Roxana Borghese 1, Charlotte Sonigo2, Nelly Achour3, sophie monnot1, Nadine Girarel1, michael grynberg3, Alexandra Benachi2, Sophie Frank4, Chrystelle Colas4, Dominique Stoppa-Lyonnet4, Julie Steffann1 Christina Katsidi1;2, Haralambia Tsarouha2, elisavet kouvidi2, leandros lazaros2, Periklis Makrythanasis 3;4;5, amelia pantou2, emmanouil kanavakis2, ariadni mavrou2 Svetlana Deryabina 1, Olga Lagutina1, Elena Kudryavceva1 Fatemeh Ahangari 1, shima Dehdahsi1, Mahsa Fadaie1, Mehri Ashki1, Maryam Beheshtian1, Maryam Azad1, Masoumeh Akbari Kelishomi1, Negar Molaei2, Parnian Alagha2, Ariana Kariminejad1, Hossein Najmabadi1 sophie monnot 1, Nadine Gigarel1, anne mayeur2, Joana Bengoa1, Roxana Borghese1, Benoit Funalot1, Charlotte Sonigo2, nelly achour2, Julie Steffann1 Eloisa Caviglia 1, Micaela Galain1, Yannina Diaz Cano1, Mónica Fabbro1, Sergio Papier1, Florencia Nodar1, Cecilia Fernandez1 Micaela Galain 1, Eloisa Caviglia1, Yannina Diaz Cano1, Mónica Fabbro1, Sebastián Menazzi1, Valeria Paz2, Idelma Serpa2, Laura Sicaro3, Edgardo Young Obejero3, Andrea Dematteis4, Gustavo Estofan4, Sergio Papier1, Florencia Nodar1, Cecilia Fernandez1 Yannina Diaz Cano 1, Micaela Galain1, Eloisa Caviglia1, Mónica Fabbro1, Florencia Nodar1, Sergio Papier1, Cecilia Fernandez1 EP03 Prenatal Genetics Momen Kanjee 1, Nuran Ertargin1, Cigdem Yuce Kahraman1, Abdulgani Tatar1 Agnese Kalnina 1;2, Inguna Lubaua1;2, Ieva Grīnfelde1;2, Klinta Lisnere1, Maija Lubgane-Griķīte1, Elena Zeltina1, Gunta Kalnberza1 Manon Chretien1, Julien Osouf2, Carine Abel3, Alexandra Afenjar4, Tania Attie-Bitach5, elise brischoux-boucher6, lydie BURGLEN7, Nadège Calmels2, Nicolas Chassaing8, Thomas Courtin9, Julian Delanne10, Martine Doco-Fenzy11;12, Christele Dubourg13, Benjamin Durand1, Salima EL CHEHADEH1, Laurence Faivre10;14, Aurore Garde10, Emmanuelle Ginglinger15, Virginie Haushalter2, Damien Haye3, Solveig Heide9, Laurence Heidet16;17, Delphine Heron9, Clemence Jacquin12, Laetitia Lambert18;19, Vincent Laugel20, Daphné Lehalle9, Laurence Michel-Calemard21, Yohny Montoya22, Jean Muller2;23;24, Sylvie Odent25, Olivier Patat8, Juliette Piard14;26, Céline Poirsier12, Audrey Putoux3, Chloe Quelin25, Nicolas Sananes27;28, Audrey Schalk2, sophie scheidecker2;24, Christel Thauvin-Robinet10;14, Stéphanie Valence29, Anne-Sophie Weingertner27;28, Justine Wourms18, Hélène Dollfus1;24, Benedicte Gerard2, Caroline Schluth-Bolard2;24, Elise Schaefer 1 Rainer Wimmer 1, Miriam Kinzel2, Iris Dressler-Steinbach3, Holger Janke4, Andre Weber1, Markus Stumm1 Marta Palucci 1, Antonella Pizza2, Allessandra Terracciano3, Valeria Orlando3, antonella spagnuolo4, maria assunta spina4, Lucia Manganaro5, maria cristina muzi6, manuela di natale6, barbara raso6, carlotta vaccari6, sara nuovo6 Ilona Dietze-Armana 1, Andreas Gerhardinger1, Manuel Lüdeke1, Karl Mehnert1 Aliza Amiel 1, Mahdi Tarabeih2 Mohammad Reza Mahdavi1, Atefeh Khoshaein 2, Maryam Rahimi2, Hossein Jalali1, Mahan Mahdavi2 Zoi Koukou1, Eleftherios Panteris2, Emmanouel Manolakos3, Aristeidis Papadopoulos4, Ioannis Papoulidis3, Eleftheria Papadopoulou5, Konstantinos Relakis6, Stavros Sifakis 7 Ivana Joksic 1, Mina Toljic1, Andjela Stankovic1, Zagorka Milovanovic2;3 Monika Gorządek 1;2, Hanna Moczulska1;2, Karolina Gadzalska1;2, Paulina Jakiel1;2, Ewa Juścińska1;2, Tomasz Płoszaj1;2, Sebastian Skoczylas1;2, Maciej Borowiec1;2, Agnieszka Zmysłowska1;2 Lucie Faldynová1, Sylwia Walczysková 1, Dita Černá1, Monika Kudrejová1, Šárka Hilscherová1, Romana Kaniová1, Simona Širůčková1 Sara El-Dessouky1, Mona Aboulghar2, Reza Maroofian3, Ahmed Abdelfattah4, Maha Eid5, Wessam Sharaf-Eldin6, Maha Zaki7, Alaa Ebrashy8, Hassan Gaafar8, Ahmed Saad6, Mahmoud Issa7, Shahryar Alavi9, Zahra Firoozfar9, Mohamed Ateya8, Mona Fouad8, Rana Abdella8, Ahmed Ezz Elarab8, Ebtesam Abdalla10, Nahla Abdel-Aziz11, Haitham Khaled4, Mohamed Elhodiby12, Sameh Senousy4, Homa Tajsharghi 13 Kalina Belemezova 1, petya chaveeva2, Radostina Raynova3, Petya Andreeva2;4, Atanas Shterev2, Katerina Kavaldzhieva1, Ivanka Dimova5 Maria MALARSKA 1, Paulina Pachniak1, Hanna Moczulska1, Paulina Jakiel1, Karolina Gadzalska1, Agnieszka Pukajło-Marczyk2, Katarzyna Kiliś-Pstrusińska2, Katarzyna Rajfur3, Alicja Majos4, Agnieszka Zmysłowska1 Rebecca Gembicki 1, Michael Gembicki2, Jan Weichert2, Moneef Shoukier3, Yorck Hellenbroich1, Malte Spielmann1;4 Olga Bürger 1, Angelika Humbel1, Ivan Ivanovski1, Alessandra Baumer1, Anita Rauch1 Nader Khaleghi Hashemian 1, Gioia Mastromoro1, Daniele Guadagnolo1, Enrica Marchionni1, Allessandra Terracciano2, Antonio Novelli2, Antonio Pizzuti1 zohra hasan 1, MAHRUKH NASIR1;2, Fizza Akbar1, Salman Kirmani1 Kajsa Nilsson 1 cristina Ferreira 1;2, Ana Rita Tarelho1, Bárbara Marques1, Silvia Serafim1, Sónia Pedro1, Cristina Alves1, Mónica Viegas1, Ângela Ferreira3, Hildeberto Correia1 Carla Martín-Grau1, Carmen Orellana 2, Mónica Roselló Piera2, Laia Pedrola2, Juan Silvestre Oltra Soler2, Sandra Monfort Membrado2, Marta Domínguez Martínez1, Alfonso Caro Llopis1, Francisco Martinez2 Eusebiu Vlad Gorduza 1;2, Violeta Martiniuc2;3, Lavinia Caba1, Andreea Florea1, Ana Grigore1, Mihaela Gramescu1, Elena Mihalceanu1;2 Monika Jurkowska1, Ewa Matczyńska1, Anna Wąsowska1, Przemysław Łyszkiewicz1, Magdalena Kania1, Elżbieta Gregorczyk1, Paweł Ogrodnik1, Maria Jędrzejowska1, Marek Zagulski1, Anna Boguszewska-Chachulska 1 Luana Giovannangeli1, Virginie Magry 2, Florence Jobic1, Kahia Messaoudi2, Alexis Billes1, marianne perriere2, Ségolène Delmas-Lanta3, Alice Masurel4, Guillaume Jedraszak2, Gilles Morin1 Laura Feyereisen1, Tony Yammine 1, Leila Sahmoune Rachedi2, Jean-Paul Bory2, Victor Dupont-Gaudin3, Jean-Marc Dossot3, Véronique Dalstein1, Poirsier Céline4, Emilie Landais1 Mohamed AlHamed 1 Gema Escribano-Serrano 1, Diana Salinas-Chaparro1, mar borregán prats1, Judith Armstrong1;2;3, Delia Yubero1;2;3, Laura Martí1;2, Clara Xiol1;2, Guerau Fernandez1;2;3, Maria Edda Marimon Garcia4, Miriam Illa Armengol4;5, Miriam Perez Cruz4;5, Elisenda Eixarch Roca4;6, Mar Bennasar Sans4, Joan Sabria Bach4;5, Francesc Palau1;2;3;7 Panagiota Kekelou1, Cristos Chassanidis1, Thomai Tsantzali1, Evangelia-Eleni Delaki1, Effrossini Boutou1, Eleni Yfanti1, Maria Baou1, Panayiotis Kyriazopoulos1, Stamatia Theodoridou2, Veroniki Komninaka3, Spiridon Kanellakis4, Georgia Ikonomou5, Dimitra Pessini6, Maria Dimopoulou7, Angeliki Balassopoulou 1 Neeta Lakhani 1, Vijayalakshmi Salem Ramakumaran1, emily craft1 Laia Pedrola 1, Mónica Roselló Piera1, Carla Martín-Grau2, Juan Salvador Rubio Moll3, Rosa Portero Gómez3, Beatriz Marcos Puig3, Jose Vicente Cervera1, Ramiro Quiroga3, Carmen Orellana1 Kristina Aleknavičienė 1;2, Rasa Traberg1;2, Zivile Zemeckiene1;1, Rasa Ugenskiene1;2 Marina Martinez-Matilla 1, Alba Velo1, Claudia Abellán1, Iryna Boyko1, Gemma Cartagena1, Carmen Fernández-Vizcaino1, Lova Matsa2, Sandra Garcia-Herrero1 EP04 Sensory Disorders (Eye, Ear, Pain) Kristiyana Vitanova 1, Kunka Kamenarova1, Nevyana Veleva2, Kalina Mihova1, Yoanna Kaneva2, Reni Tzveova3, Martin Georgiev1, Ivanka Dimova1, Alexander Oscar2, Radka Kaneva1 Poh San Lai 1, Grace Tan1, Chunping Liu1, thuanchong Quah1 Basamat AlMoallem 1 Siying Lin 1;2, Ellie Hay3, Andrew Webster1;2, Omar Mahroo1;2, Robert Henderson1;2;4, Gavin Arno1;2 Tadeusz Kałużewski 1;2, Łukasz Kępczyński1;2, Karolina Matuszczyk2, Magdalena Markowska2, Jordan Sałamunia2, Bogdan Kałużewski2, Agnieszka Gach1 Raquel Muguerza 1;1, Nerea Bastida-Lertxundi1, julien crettaz1, Raquel Sáez-Villaverde1, yolanda ramírez1, jose luis eguiburu1, laura martinez1, irene perez1, carla lallave1, Zuriñe Martinez2, Marta Abrego2, Elena Beristain3, Blanca Gener Querol4 Ebrahim Shokouhian 1, Zohreh Fattahi1, Sanaz Arzhangi1, Kimia Kahrizi1, Hossein Najmabadi1, mojgan babanejad1 Niloofar Bazazzadegan 1, Kimia Kahrizi1, Hossein Najmabadi1, Kevin TA. Booth2;3, Sussan Banihashemi1, Sanaz Arzhangi1 Laura Mauring 1;2;3, Tiia Reimand1;3, Ülle Murumets1, Sander Pajusalu1;3, Mari Petraudze2, Kuldar Kaljurand2;3, Katrin Ounap1;3 Fulya Yaylacioglu Tuncay 1, burak acar2, murat yüksel2, Gülsüm Kayhan3, mehmet ali ergün3, Sengül Ozdek2 Raziye Rezvani Rezvandeh 1, negar kazemi1, Farzane Zare Ashrafi1, Masoud Edizadeh2, fatemeh ghodratpour1, Fatemeh Keshavarzi1, Khadijeh Jalalvand1, Sanaz Arzhangi1, Kimia Kahrizi1, Hossein Najmabadi1, Marzieh Mohseni1 negar kazemi 1, Raziye Rezvani Rezvandeh1, Farzane Zare Ashrafi1, fatemeh ghodratpour1, fatemeh keshavarzi1, Khadijeh Jalalvand1, Sanaz Arzhangi1, Masoud Edizadeh2, Kimia Kahrizi1, Hossein Najmabadi1, Marzieh Mohseni1 Ornella Rondinone 1, Giada Moresco1, Carola Conca Dioguardi2, Martina Miceli2, Rita Gorgoglione2, Chiara Pesenti2, Giovanni Marfia3;4, Stefania Navone3, Gianluca Tolva2, Leonardo Colombo5, Luca Rossetti5;6, Monica Miozzo1;2, Laura Fontana1;2 Mert Coşkun 1, Mehmet Erkan Dogan2, Alp Peker1, Aslı Toylu1, Özden Altıok Clark1 Vanessa Hochheimer 1, Zafer Yüksel1, Hanno Bolz1 Sinje Geuer 1, Cord-Christian Becker1 Luigi Monti 1;2, Elena Luppi1;2, Nicole Balducci3, Giovanni Innella1;2, Francesca Montanari1, Cesare Rossi1, Daniela Turchetti1;2, Marco Seri1;2 Tammy Benteau 1, Maxime Maheu2, David McComiskey3, Anne Griffin1, Kim Furlong4, Susan Stanton5, Terry-Lynn Young1 Neta Feinstein-Goren 1;2, Dalia Eli3, Daphna Prat2;4;5, Ortal Barel Barel6, Odelia Chorin2;7, Shelly Lev-Hochberg2;7, Abraham Spierer2;4, Guy Ben-Simon2;4;5, annick REIN ROTHSCHILD2;7, Ben Pode-Shakked2;5;7 Tyrni Pykäläinen1;2, Minna Kraatari-Tiri1;2, Pia Pohjola3, Sanna Häkli4, Elisa Rahikkala 1;2 Erlend Christoffer Sommer Landsend1, Charlotte von der Lippe 2, Line Merete Mediå3, Jeanette Ullmann Miller3, Knut Erik Berge4, Solrun Sigurdardottir3 Ewa Matczyńska 1;2, Marta Beć-Gajowniczek1, Larysa Sivitskaya1, Elżbieta Gregorczyk1, Ewa Suchecka1, Przemysław Łyszkiewicz1, Robert Szymańczak1, Maria Jędrzejowska1, Sławomir Teper2, Maciej Krawczyński3, Anna Boguszewska-Chachulska1 ofek freund 1, Baker Alsana2, Nadav Agam1, Matan M. Jean1, Amit Safran1, Tomer Poleg1, libe gradstein2, Erez Tsumi2, Ohad Shmuel Birk1;3 Michaela AH Hofrichter 1, Christian W Remmele1;2;3, Asuman Koparir1, Daniel Bengl1, Tabea Böttcher1, Tobias Müller4, Marcus Dittrich1;4, Wafaa Shehata-Dieler5, Sophie Flandin5, Thomas Haaf1 Catia Mio1, Jessica Zucco2, Carla Reale3, Concetta Ambrosino3, Federica Baldan1, Lorenzo Allegri1, Alessandra Franzoni2, Angela Valentina D’Elia2, Flavio Faletra4, Giuseppe Damante 1 Smaragda Kamakari 1, Stavrenia Koukoula2 Margherita Scarpato 1, Francesco Testa2, Roberta Zeuli1, Sandro Banfi1;3, Francesca Simonelli2, Marianthi Karali1;2 Federico Rondot1, Amedeo Primerano2, Patrizia Dentelli1, Enrico Grosso2, Alessandra Pelle 2, Barbara Pasini1;2 Sara Álvaro Sánchez 1, mar borregán prats1, Judith Armstrong1;2;3, Daniel Castillo1, Sara Cardelus Vidal4, Águeda Díaz Anadon4, Maurizio Levorato4, Francesc Palau1;5;6, Loreto Martorell Sampol1 Ayse Gurel 1, Omer Yildiz2 Stella Diogo-Cavassana1, Danillo Alencar-Coutinho1, Allan Anjos-Monteiro1, Thais Pailo-Carvalho2, Emi Z. Murano3, Jeanne Oiticica1;3, Ricardo F. Bento1;3, Regina Mingroni-Netto2, Ana Carla Batissoco1, Karina LEZIROVITZ 1 Silvia Borecka 1, Klaudia Cipkova1, Lukas Varga1;2, Dimitrios Paouris3, Martina Skopkova1, Miroslava Huckova1, Dominika Hromnikova1, Milan Profant2, Daniela Gasperikova1 Andri Miltiadous 1, Paul Costeas1, maria kanezou1, Petroula Gerasimou1, rafaella metaxa2, Emily Athanasiou3, George A. Tanteles4, Violetta Anastasiadou2 Elena Colastra Ugena 1, Ana Peña Cabia1, Irene Pereira Gonzalez1, Raquel Gomez Molina1, Maria Perez Gomez1, Maria Concepcion Calderon Alva1 Jennifer Brzezynski 1, Caroline Johnson1, Sandra Smieszek1, Changfu Xiao1, Christos Polymeropoulos1, Mihael Polymeropoulos1 Solomon Merepa 1, Karthikah Jeganathan1, Bertrand Chesneau1;2;3, Richard Holt1, Lidiya Talbot1, Fabiola Ceroni1, Dorine Bax1, Fiona Watkins1, Mira Kharbanda4, Katherine Lachlan4, Patrick Calvas3;5, Julie Plaisancié3;5, Nicolas Chassaing3;5, Elfride De Baere6;7, Nicola Ragge1;8 EP05 Internal Organs & Endocrinology (Lung, Kidney, Liver, Gastrointestinal) Nina Bögershausen 1, Alexander Wolff1, Yun Li1, Tassilo Wollenweber2, Julia Schmidt1, Silke Kaulfuß1, Karl Koehrer2, Arne Zibat1, Gökhan Yigit1;3, Bernd Wollnik1;3;4 Madeline Pearson 1, Ruth McGowan2, Philip Greene3, Zosia Miedzybrodzka4, jonathan berg1 Deimante Brazdziunaite 1, Gabija Mazur2, Algirdas Utkus1 lev dorfman1;2;3, Mansa Krishnamurthy4;5, Yael Haberman1;3;6;7, emily stenke8, Anat Guz Mark2;3, Hengameh Abdollahpour9, seba saleh nadeef10, Khaled Warasnhe11, Figen Ozcay12, vivien a. nguyen13, Martin G Martin14, stanley f. nelson15;16, Julian Martinez-Agosto14;15, james m. wells4;5;17, Fowzan Alkuraya10;18, Billy Bourke8;19, Yair Anikster3;20;21, Ben Pode-Shakked 3;20 aviva eliyahu 1;2;3, Danit Atias-Varon4, Ortal Barel Barel5, Yulia Khavin5, Elon Pras3, haike reznik wolf6, Lior Greenbaum3;6, Pazit Beckerman3;7, Nabil Abu amer3;7, Tamara Vignanski-Jaffe3;8, Asaf Vivante3;9;10 Yerai Vado1, Africa Manero Ruiz de Azua1, Arrate Pereda1, Guiomar Perez de Nanclares Leal 1 Ana C. Ribeiro 1, Najeeb Syed2, Luís R. Saraiva3, Manuel C. Lemos1 Anastasiia Buianova 1, Anastasiia Ryzhova2, Mariia Proskura2, Edita Petrosyan2, Valeriia Gavrilova2, Anna Shmitko1, Alina Samitova1, Oleg Suchalko1, Zhanna Repinskaia1, Galit Ilyina1, Anna Kuznetsova1, Iuliia Vasiliadis1, Natalia Ponikarovskaya1, Vera Belova1, Dmitriy Korostin1 Olga Martinez-Arroyo 1, Ana Flores-Chova1, Marta Mendez-Debaets1, Laia Garcia-Ferran1, Lorena Adan1, Lesley Escriva1, Sara Vela1, Carlos Bea1;2, Josep Redon1;2;3;4, Maria J Forner1;2;3, Raquel Cortés1, Ana Ortega1;5 Anney Yelena Rosario Vargas 1, María Jesus Izquierdo Ortiz1, Irene Oñate Alonso1, Badawi Hijazi Prieto1, María Isabel Sáez Calero1, Marta Boya Fernández1, Raquel Toro Casado1, Vanesa Camarero Temiño1, Basilia González Diez1, María Luisa Carrasco Prado1, Alejandra Martin Rosique1, Sheyla Cristal Álvarez Parra1 Olga Parshina 1, Anastasiia Buianova1, Ekaterina Filimonova2, Mariia Venediktova2, Alexandra Shagiazdanova2, Elena Dobronravova2, Stella Temurian2, Anna Shmitko1, Alina Samitova1, Oleg Suchalko1, Zhanna Repinskaia1, Galit Ilyina1, Iuliia Vasiliadis1, Anna Kuznetsova1, Mariia Sidorchuk1, Natalia Ponikarovskaya1, Vera Belova1, Dmitriy Korostin1 Ourayna Batta 1;2, lyahyai jaber3, siham chafai elalaoui4;5, abdelaziz sefiani2;4, imane jaouad cherkaoui4;5 Mariana Ryznychuk 1 Bojana Stevanovic 1, Ivan Milovanovic2, Marija Kosic3, Branka Popovic1 Sorina Schipor 1, Lidia Radomir2, Andrei Muresan1, Catalina Poalelungi1, Oana Popa1, Andreea Kremer1, Alina Dumitrica1, Luminita Udrea1, Adriana Padure1, Ioana Nedelcu1, Iuliana Gherlan2;3, ELENA EMANUELA BRAHA1 Lavinia Caba 1, MARIA CHRISTINA UNGUREANU2, Laura Florea3, Andreea Florea1, Eusebiu Vlad Gorduza1 Pruthvi Goparaju 1, Claudia Gragnoli2;3;4 Ewa Juścińska 1, Agata Pastorczak2, Karolina Gadzalska1, Paulina Jakiel1, Monika Gorządek1, Tomasz Płoszaj1, Sebastian Skoczylas1, Maciej Borowiec1, Agnieszka Zmysłowska1 Michaela Mihulová1, Karolina Kočandrlová 1;2, Veronika Moslerova1, Marek Turnovec1, Júlia Martinková1, Milan Macek1, Markéta Havlovicová1, Dana Thomasova1 Boodor Al-Kawlani 1, Uros Hladnik1, Nayla Leon Carlos1, Moneeb Othman1, Omid Paknia1, Peter Bauer1 Olga Beltcheva 1, Kunka Kamenarova1, Kalina Mihova1, Martin Georgiev1, Galia Zlatanova2, Simona Kerezieva3, Boriana Deliyska3, Maria Gaydarova2, Radka Kaneva1 Laura Skrabule 1, Dmitrijs Rots1, Ludmila Voložonoka1, Madara Mašinska1, Iveta Dzīvīte-Krišāne1, Ieva Mičule1 Kalina Mihova 1;2, Kunka Kamenarova1;2, Iva Stoeva3;4, Martin Georgiev1;2, Darina Kachakova-Yordanova1;2, Daniela Pencheva1;2, Radka Kaneva1;2 Domagoj Caban 1;2, Ana Merkler Sorgic1, Hana Ljubic1, Ana Acman Barisic1, Petra Anic1, Duška Tješić-Drinković3, Ana Mojsović Ćuić2, Ivna Kocijan2, Martina Ikić4, Ivana Rako1 Areej Alatawi 1, Mohammed Almannai2 Maryam Mirzazadeh 1, Naser Gilani2, Ariana Kariminejad3 Layal Shaheen 1;2, Anna Kim1, Elena Kovalenko1, Dmitrii Kharitonov1, Valeriya Rubinova1, Alexander Rakitko1 EP06 Skeletal, Connetive Tissue, Ectodermal and Skin Disorders Alexandra Yakovleva 1, Leonid Zhozhikov1, Roza Ivanova1;2, Aitalina Sukhomyasova1;2, Nadezhda Maksimova1 Zeynep Münteha Başer 1, Ceren Alavanda2, Vedat Yüce1, Esra Hilal Ceylan1, Ayşe Deniz Yücelten3, Bilgen Bilge Geçkinli1 Esra Dirimtekin 1, cekdar kapazan1, ahmet mert yanik2, barıs yılmaz3, Bilgen Bilge Geçkinli1 Alexandra Fountoulaki 1, Georgia Christopoulou2, angeliki erakleous1, maria kassotaki1, ILIANNA MANIADAKI1, Pantelis Constantoulakis2, Eleftheria Papadopoulou1 Łukasz Przesór 1, Małgorzata Piotrowicz1, Urszula Wysocka1, Iwona Pinkier1, Agata Kucińska1, Dominika Komorowska1, Wanda Hawuła1, Tadeusz Kałużewski1, Łukasz Kępczyński1, Agnieszka Gach1 Eyal Elron 1, Naama Orenstein1;2, lina basel-salmon1;2;3, Nurit Assia Batzir1 Pelin Simsek-Kiper 1, Merve Soğukpınar1, Beren Karaosmanoglu2, Gozel Jumayeva3, gizem ürel-demir1, rahsan gocmen4, Gulen Eda Utine1 Jenny Blechingberg 1, Thorkild Terkelsen1;2, Uffe B Jensen1, Mette Sommerlund3, Hanne Vinter4, Lise Graversen1 Nehal Hassib 1, rasha elhossini2, Inas Sayed3, Mohamad Abdelhamid3 Junya Adachi 1, Yoshihito Tokita2, Junichiro Machida3, Yoshihiko Aoki4, Michiyo Ando5, Yasuto Sano6, Mitsuo Goto5, Atsuo Kaetsu1, Takamasa Shirozu1, Eriko Osumi1, Michiko Matsuoka1, Nasir Maeda1 Yoshihiko Aoki 1, Junichiro Machida2, Junya Adachi3, Michiyo Ando4, Yasuto Sano4, Terumi Saito1, Mitsuo Goto4, Yoshihito Tokita5 Clarissa Modafferi 1, Elisabetta Tabolacci1, Arcangelo Fargnoli1, Ilaria Cassano1, Roberto Bertozzi1, Cristina Grippaudo2, Alessandro Arcovito3, Filomena Lo Vecchio1, Ettore Lo Cascio2, Pietro Chiurazzi1 irem kalay 1;2, Barbara Vona2;3 Lina Al Qadi 1, eric allain2;3;4;5, Haley McConkey6;7, Bekim Sadikovic6;7, Mouna Ben Amor1;8;9 Juliane Köhler 1, Malte Spielmann1;2, Nadine Hornig1 Valerie Katharina Berge 1;2, Nadine Hornig3, Yorck Hellenbroich1, Malte Spielmann1, Irina Hüning1;4 Robert Carlier1, Alice Porto Vasconcelos 2, Dominique P. GERMAIN2;3 Claudia Regina Bonini-Domingos 1, Lucas Ramos1, Flavia Cristina Rodrigues-Lisoni1 Tatiana Vasilyeva 1, Andrey Marakhonov2, Artem Borovikov3, Vitaly Kadyshev2, Olga Lenina4, Zhanna Markova5, Alena Chukhrova6, Inna Tebieva7, Rena Zinchenko2 KUBRA ADANUR SAGLAM 1, Semiha Bekfilavioglu2, Aysel Yıldız Boyraz2, Emine Ayça Cimbek2, Ayse Ozden3, AYBERK TURKYILMAZ1, Alperhan Cebi1, Hakan Doneray3, Gulay Karaguzel2 Merve Tanrısever Türk 1, gizem ürel-demir1;2, Gulen Eda Utine1, Pelin Simsek-Kiper1 Iveta Žukauskaitė 1, Rasa Traberg1, Rimvydas Jonikas1, Rasa Ugenskiene1 Nory Omayra Davalos Rodriguez 1;2, Rosa Esther Castañeda Arteaga3, Ricardo E’ Vega Hernandez4, José Guadalupe Ramiro Ortega Águila5, Ana Rosa Rincon Sanchez6, Cristian Víctor Ledezma Rodríguez7, Sergio Alberto Ramirez-Garcia8, Diana Garcia-Cruz8 Nela Maksimovic1, Biljana Jekic1, Milka Grk1, Tatjana Damnjanovic1, Dijana Perovic1, Biljana Ljujic2, Marija Dusanovic Pjevic1, Valentin Djonov3, Milica Rasic 1, Vladislav Volarevic2;4 Öznur YILMAZ BAYER 1, banu nur1, sezin yakut uzuner2, ercan mihci1 Ana Rosa Rincon Sanchez 1, Nory Omayra Davalos Rodriguez2;3, Diana Garcia-Cruz4, Rodrigo Hernández Ramírez5;6, Vianney Yolanda Jiménez Rodríguez7, Sergio Alberto Ramirez-Garcia4 Sofie Fredberg 1, Stine Bjoern Gram1;2;3, Helena Karstensen4, Ulrikke Lei5, Lilian Ousager1;2 Rana Alzahrani 1, Balsam Aleissa2, Sally Alyousef2, Norah Alsaleh1 Yucel Erbilgin 1, Kıvanc Cefle2, Sukru Ozturk2, Lala Soltanova3, Pinar Kadioglu4, sumeyye kocaağa1;5, Melisa Kılıç1;5, Muge Sayitoglu1 MAHRUKH NASIR 1;2, zohra hasan1, Fizza Akbar1, Salman Kirmani1 Elena Merkuryeva 1, Tatiana Markova2, Oksana Ryzhkova2, Yuri Buklemishev3, Vladimir Kenis4 Esra Hilal Ceylan 1, cekdar kapazan1, Zeynep Baser1, Ayşe Deniz Yücelten2, Esra Dirimtekin1, Bilgen Bilge Geçkinli1 MARIA TZETIS 1, Konstantina Kosma2, Periklis Makrythanasis1;3, Anastasios Mitrakos1;4, Thomas Mprantzos1;4, Nikolaos Marinakis1;4, Faidon-Nikolaos Tilemis1, ASPASIA TSEZOU5, Christalena Sofocleous1 Renata Szalai 1, Judit Bene1, Anna Zsigmond1, Krisztina Galimurka1, Kinga Hadzsiev1 khaled osman 1, adel Shalata2 Melek Trigui 1;2, Nathalie PALLARES-RUIZ1, David Geneviève2, Cyril Amouroux3, Thomas Edouard4, Sabine Sigaudy5, Marjolaine Willems2, Mouna Barat-Houari1 Seyedmohammad Seyedhassani 1, Massoud Neshan1;2, Neda Tavakkoli Banizi1 Bahriye Öykü Candan 1, Güven Toksoy2, Ayça Dilruba aslanger2, firdevs baş3, aslı al3, Birsen Karaman4 Renáta Michalovská 1, Helena Paszekova1, Kristýna Hanuláková1, Tomáš Píš1, Veronika Krulisova1, Ivo Mařík2, Elsa Zemankova3, Dominika Vallusova1, Terézia Haňová1, Zděnka Vlčková1 Ari Horton 1, Bryony Thompson2, Hnin Pwint Oo3, David Francis4, Ingrid Winship1 Ye Li 1, Maria Wehner1, Nicole Cesarato1, Yasmina Gossmann1, F. Buket Basmanav1, Regina C. Betz1 Stefanie Van de Voorde 1, Kathelijn Keymolen1, Frederik Hes1, Andrea Buysse1, Willem Verpoest2, Bert Callewaert3, Sofie Symoens3, Aude Beyens3, Ifigenia Spanoudi-Kitrimi4, Boyan Dimitrov1 Deepti Saxena 1, AMIT KUMAR TIWARI1, Amita Moirangthem1, Kausik Mandal1, Shubha Phadke1 Mobina Ghofrani Shadman 1, Nathalie Kruse1, Kristian Händler1, Saranya Balachandran1, Varun Sreenivasan1, M. Ijlal Haider2, Neseebullah Kakar1;3, Malte Spielmann1;4;5 Ruslan Minnebaev1, Anastasiia Kungurtseva2, Nina Demina1, Olga Shchagina1, Anna Orlova1, Alisa Vitebskaya2, Yulia Tikhonovich2, Petr Vasiliev 1 Sanda Huljev Frkovic 1, Marija Vidakovic1, Ivan Jovanovic2, David Ozretic2, Hrvoje Jednacak3, Marijan Frkovic1, Branka Runtić Jurić1 Rim Sghiri 1, Hana BenHassine1, Khadija Baccouche2, Nejla Elamri2, Adel Almogren3, Zahid Shakoor3, Foued Ben Haj Slama1, Elyes Bouajina2, Ramzi Zemni1 Betül Gerik-Celebi1, F. Sırrı Cam 2 Aleksandr Tyapkin 1, Artemii Ivanov1, Olga Leonova2, Aleksandr Krutko2, Alexey Peleganchuk3, Veniamin Fishman1, Elizaveta Elgaeva1;4, Yakov Tsepilov1 Azra Zejnelagic 1;1, Chanelle Cilia2, Jean Paul Ebejer1, Melissa Formosa2 Marlene Perez 1, Almudena Amor1, Laura Cazón1, Maria Sanchez1, Iria Gómez Díaz1, Rosalía Peteiro1, Ivonne Cárdenas Reyes1, María Valverde1, Soledad García Hernández1;2, Diego Cabrera Argaña1, Xusto Fernandez1, Martin Ortiz Genga1, Juan Pablo Ochoa1 Karolina Biel 1, Aleksander Jamsheer1 Diana Prepelita 1, Andreea Tutulan-Cunita1, Anca Pavel1, FLORINA MIHAELA NEDELEA2, VASILICA PLAIASU3, Ina Ofelia Focsa1, Danae Stampoulia1 Svetla Nikolova1, Martin Pasev 1, Milka Dikova2, Alexandre Loukanov3 Maria Sredkova-Ruskova 1, Todor Ruskov1, Tsvetina Veleva1, Trayan Delchev1, Darina Kachakova-Yordanova2;3, Kalina Mihova2;3, Kunka Kamenarova2;3, Radka Kaneva2;3, Daniela Avdjieva-Tzavella1 Francesco Marco Parodo 1;2, Giulia Gori2, Annarita Giliberti2, Giovanna Traficante2, Elia Dirupo2, Elena Andreucci2, Giorgia Mancano2, Sara Bargiacchi2, Rosangela Artuso2, Stefano Stagi3;4, Laura Papi1;5, Angela Peron1;6 Elif Yilmaz Gulec 1;2, Gulden Yorgancioglu Budak3 Ghada A. Otaify 1;2, Katta M. Girisha3, Khalid S. Al Thihli1, Ahmed Alsariri4 Enise AVCI DURMUŞALİOGLU 1, Yusuf Dogan1, Turkan Turkut Tan1, Dilsah Cogulu2, Esra Isik1, Ozgur Cogulu1, Tahir Atik1 EP07 Cardiovascular Disorders Ameema Asad1, Fizza Akbar1, Asra Wahid2, Yawer Saeed3, Fateh Ali Tipoo3, Salman Kirmani 1 Dalma Ruzsics 1, Katalin Csonka2, Viktória Szabó3, Vera Goda4, Rita Bertalan1, Éva Pállinger5, Luca Kamilla Li1, Zoltán Liptai1, Csaba Vilmányi6, Csaba Bödör2, Árpád Ferenc Kovács1 Aslıhan Gizem Bilgin 1;2, Berkay Ekici3, Aycan Fahri Erkan3, Neslihan Coban1;2 Gulshara Abildinova 1;1, Maxsim Solomadin2, Anel Rakhimova3, Aida Yessentayeva3, Anna Borovikova3;4, Zhanna Zhabakova3 Nadia Akawi 1, Fatma Aljasmi1 Nola Šiljić 1;2, Imke Christiaans1;2, Jan Jongbloed1;2 Meitsz Su 1, Hoi-Lam Tam2 Špela Stangler Herodež1, Tomaž Podlesnikar2, Lara Forstner1, Nadja Kokalj Vokač1, Matjaž Vogrin3, Danijela Krgović 1 Eero Aaltonen 1, Nina Mars1;2, Sanni Ruotsalainen1, Roope Ripatti1, Johanna Aro1, Kristina Hotakainen3;4, Marianne Leinikka3, Iiro Heikkilä3, Elisabeth Widén1, Samuli Ripatti1;2;5 Tuba Maras1, Sinem YALÇINTEPE 1, Hazal Sezginer Guler1, Murat Deveci2, Necdet Sut3 Eva Kutílkova 1, Alice Krebsová1, Pavel Votýpka2, Petra Peldová2, Terezia Tavacova3, Veronika Zoubková2, Miloš Kubánek1, Petr Peichl1, Marketa Segetova1, Jana Haskova1, Markéta Adamová1, Jan Janousek4, Marek Sramko1, josef Kautzner1, Milan Macek2 Noa Ruhrman Shahar 1;2, Lina Basel-Salmon1;2;3, Shay Ben-Shachar2;4, Dina Yagel5, sara hoss6, lily bazak1, Lena Sagi-Dain7, lilach Benyamini8, Sagi Josefsberg Ben-Yehoshua9;10, Rotem Greenberg4, Ofer Isakov2;4;5, elena friedman1, Michal Naftali5, daniel monakier6, moti haim11, amitai segev12, adel shalata13, nechama shalva14, alvit veber14 Jovana Kuveljic1, Ana Djordjevic 1, Milica Dekleva-Manojlovic2, Ana Kolakovic1, Ivan Zivotic1, Maja Zivkovic1, Tamara Djuric1 Pashmina Wiqar Shah 1, Tobias Reinberger1, Zouhair Aherrahrou1, Malte Spielmann2 NAZIMAH NIK-MAHMOOD1;2, Thiloka Ratnaike 2;3, Timothy Hearn1 Claire Hopton 1;2, Glenda Beaman1;2, Rahat Perveen2, Kate Chandler1;2, William Newman1;2 Laura Cazón 1;1, Noël Brögger1, Luis De la Higuera Romero1, Marlene Perez1, Iria Gómez Díaz1, Rosalía Peteiro1, Maria Sanchez1, Emilia Maneiro1, Xusto Fernandez1, Diego Cabrera Argaña1, Almudena Amor1, Ivonne Cárdenas Reyes1, María Valverde1, Soledad García Hernández1, Martin Ortiz Genga1, Juan Pablo Ochoa1 Megumi Shiomi 1, Yuki Nagata1;2, Takeaki Sudo3, Takamasa Ichikawa2, Kensuke Ihara4, Ken Asada5, Yasuaki Tanaka6, Yasuteru Yamauchi7, Takeshi Sasaki8, Hitoshi Hachiya9, Yasushi Imai10, Hideo Fujita11, Tetsuo Sasano12, Tetsushi Furukawa4, Toshihiro Tanaka1;2 Daniela González Fassrainer 1;1, Eva Wohlleber2, Martin Brauner2, Niels Vandendries2, Claudia Nevinny-Stickel-Hinzpeter1 Roman Praschberger 1, Johannes Zschocke1, Matthias Frick2, Sabine Rudnik1 Marius Šukys 1, Eglė Ereminienė2;3, Karolina Mėlinytė-Ankudavičė3, Inga Nasvytienė4, Rimvydas Jonikas4, Kristina Aleknavičienė1, Darius Čereškevičius2, Sandra Žėkienė4, Zivile Zemeckiene1, Rasa Ugenskiene1 Mahsa Tahmasebivand 1, fatemeh ghodratpour1, Farzane Zare Ashrafi1, Marzieh Mohseni1, Sanaz Arzhangi1, Yasser Riazalhosseini2, Mark Lathrop2, Kaveh Hosseini3, Hossein Najmabadi1, Kimia Kahrizi1;2 Jesica Ramirez 1, Magalí Born2, Antonella Villagra1, Eliel Ramirez1, Nicolas Renna1 Luisa Marsili 1;2, Melanie Rama3, Thomas Smol4, Guy Vaksmann5, Peter van Tintelen2, Jamal Ghoumid1 Elena Zaklyazminskaya 1, Mariam Sadekova1, Mikhaleva Ljudmila2, Sergey Dzemeshkevich3, Balashova Maria4;5 Mohammed Alghamdi 1, Patrice Bourgeois1, Emilie Consolino2, Pierre Jacques Ambrosi3, Karine NGUYEN PHONG2, Gilbert Habib3, Annachiara De Sandre-Giovannoli1 Johanne Tremblay 1, Redha Attaoua2, Mounsif Haloui2, Marie-Renée Guertin2, Edoh Kodji2, Pierre Dumas2, Janusz Kaczorowski1, Pavel Hamet1 Tigist Demssew Adane 1;1, Joyce Van Meurs1, André Uitterlinden1, Jeroen van Rooij1 Sladjana Teofilov 1, Olivera Miljanovic1, Tatjana Ostojic1, Milena Bulatovic1, sasa perovic1, natasa djordjevic2 Mariam Sadekova 1, Balashova Maria2;3, Anna Motreva4, Sergey Dzemeshkevich5, Elena Zaklyazminskaya2;5 Claire Fouquet 1, Nicole Revencu2, Elisa Docampo1, Vincent Bours1, Nele Boeckx3, Bart Loeys3, Julie Harvengt1 Ece Çepni 1, Serpil Eraslan2, Rukiye Eker Ömeroğlu3, Hülya Kayserili2;4 Madina Zhalbinova 1, Saule Rakhimova1, Ulan Kozhamkulov1, Kenes Akilzhanov2, Ayaulym Chamoieva1, Nazerke Satvaldina1, Zhanel Mirmanova1, Gulbanu Akilzhanova2, Makhabbat Bekbossynova3, Ainur Akilzhanova1 Botezatu Alina 1, Vlad Morhan2, Cecilia Lazea3, Florina-Victoria Nazarie4, Adina Chis4, Romana Vulturar4 Clara Herrero-Forte1, Raluca Oancea-Ionescu 1, Carmen Cotarelo-Pérez1, Victoria Cañadas-Godoy2, M. Alejandra Restrepo-Córdoba3, María Fenollar-Cortés1 Nazerke Satvaldina 1, Saule Rakhimova1, Diana Samatkyzy1, Asset Daniyarov2, Ulykbek Kairov2, Ainur Akilzhanova1 Sivan Koka 1;2, Moshe Rav Acha2;3;4, Tal Hasin2;3;4, Yoav Michowitz2;3;4, Bayya Feras2;3;4, Shemy Carasso2;3;4, Michael Glikson2;3;4, Ephrat Levy-Lahad1;2;4 Pawares Chitayanan 1 Miryam Rosa Stella Foti1, Naomi Fanciullo2, Camilla Lucca3, Carlotta Pia Cristalli4, Chiara Diquigiovanni4, Agnese Scatigno5, Maria Schiavo2, Maddalena Graziosi2, Silvia Palmieri2, laura pezzoli3, Francesco Lai6, Vera Uliana7, Federico Barocelli7, Elia De Maria8, Alessandro Fucili9, Biagio Sassone10, Giulia Parmeggiani11, Enrica Perugini12, Rita Selvatici1, Alessandra Ferlini1, Maria Iascone3, Elena Biagini2, Cesare Rossi4, Francesca Gualandi 1 Daniele Perrino 1, Monia Magliozzi1, Daniela D’Angelantonio1, Giovanni Parlapiano2, Anwar Baban2, Antonio Novelli1 Miriam Potrony 1;2;3, Irene Madrigal1;2, Maria Isabel Alvarez1;2;3, José Villanueva-Cañas4, Aina Montalban-Casafont4, Oscar Campuzano5;6, Aleksandra Mas-Stachurska7, Eduard Solé-González3;7, Aurora Sánchez Díaz1;2;3, Elena Arbelo3;6;7, Ana Garcia3;6;7, Celia Badenas1;2 Ludmila Voložonoka 1, Līvija Bārdiņa1, Egija Berga Svitina1, Irina Žiravecka1, Marija Jurcenko1, Ieva Mičule1, Ieva Grīnfelde1, Gita Taurina1, Dmitrijs Rots1 Maria Soares Nobrega1, maria abreu2, rui baptista3, rita cerqueira 1, joaquim sá1;4, marisa teixeira1 Jesús Wagih Gómez 1, Lidia María Carrillo2, Cristina Gil3, Elisa Nicolas Rocamora1, David López2, María del Carmen Olmo2, Carmen Muñoz2, Marina Navarro2, Serena Evelina Margaretha Munteanu1, Juan Ramón Gimeno Blanes2, Maria Sabater Molina1;3 Lenka Piherová 1, Petra Melenovská1, Viktor Stránecký1, Lenka Steiner-Mrazova1, Alena Vrbacká1, Helena Trešlová1, Alice Krebsová2, Miloš Kubánek2, Vojtech Melenovský2 Martin Bird 1, Antoine Rimbert2, Alan Pittman1, Steve Humphries3, Marta Futema1 Vincent van der Pas1, Christian van der Werf2, Alexa Vermeer3, Eline Nannenberg3, Jurren van Opstal1, Arthur Wilde2, Saskia Van der Crabben 3 Marie Arens1, Valerio Rhodio2, Elisabeth Graf2, Eimo Martens1, Juliane Winkelmann2, Dominik S. Westphal 1;2 Silvia Kalantari 1, Daniele Veraldi2, Davide Politano3, Gaia Visani1, Antonia Apicella2, Elisa Giorgio1;4, Enza Maria Valente1;4, angela berardinelli5, Claudia Codazzi2, Fabio Sirchia1;6 Dovile Zebrauskiene 1;2, Egle Sadauskiene2;3, Ruta Masiuliene4, Jurate Barysiene2;3, Egle Preiksaitiene1 Tatjana Damnjanovic 1, Dijana Perovic1, Nela Maksimovic1, Biljana Jekic1, Marija Dusanovic Pjevic1, Milka Grk1, Ana Djuranovic1 Maxime van Keulen1, Leonoor Wijdeveld 2, Dennis Dooijes1, Karin van Spaendonck-Zwarts3, Bianca Brundel2, Peter van Tintelen1 Feyza Altunbas Yalabik 1, Aslı Toylu1, Ibrahim Basarici2, ercan mihci3, banu nur3, Fırat Kardelen4, Özden Altıok Clark1 Irina Shilenok1;2, Ksenia Kobzeva 1;3, Olga Bushueva1 Aurelija Kemezyte 1, Raimonda Meskiene2, Birutė Burnytė2 Natalia Gallego 1;2;3, Lucía Miranda-Alcaraz1;2;3, Mónica Mora1;2;3, Alejandro Cruz-Utrilla4;5, Dra. María Jesús del Cerro6, Manuel López Meseguer7, Amparo Moya Bonora8, Nuria Ochoa4;5, alejandro parra1;2;3, Patricia Pascual Vinagre1;2;3, Mario Cazalla1;2;3, cristina silvan1;2;3, Pedro Arias1;2;3, julian nevado1;2;3, Pablo LAPUNZINA1;2;3, Pilar Escribano-Subias4;5, Jair Tenorio1;2;3 Lucía Miranda-Alcaraz1, Natalia Gallego1;2;3, Alejandro Cruz-Utrilla4;5, Dra. María Jesús del Cerro6, Manuel López Meseguer7, Amparo Moya Bonora8, Nuria Ochoa4, alejandro parra1;2, Patricia Pascual Vinagre1;2, mario cazalla1;2, cristina silvan1;2, Pedro Arias1;2, julian nevado1;2;3, Pablo LAPUNZINA1;2;3, Pilar Escribano-Subias4;5, Jair Tenorio 1;2;3 Marcela Galvez 1, jose geles1, Sandra Bello1 EP08 Metabolic and Mitochondrial Disorders Neslihan Cinkara 1, hacer ukba kına1, aslı durmuş2, Mustafa Yilmaz3 Petra Liskova 1;2, Marketa Tesarova1, Hana Štufková1, Jana Křížová1, Tomas Honzik1, Martin Meliska1, Marie Vajter1;2, Hana Hansikova1 Secu Doina 1, Daniela Blanita1, Natalia Usurelu1, Victoria Sacară1 Luca Kamilla Li 1, Nóra Fekete2, Dalma Ruzsics1, Péter Reismann3, Éva Pállinger2, Gyorgy Fekete1, Árpád Ferenc Kovács1 Olena Bahniuk 1, Nadiia Yusechko1, Inna Bahniuk1, Anna Yevstratyeva1, Maria Malaschynska2, Nataliia Olhovich3, Halyna Makukh1 Thamer Alghamdi 1, Bader AlQahtani2, Majid Al Eissa2;3;4, Abdul Rafiq Khan5, Majid Alfadhel1;3;6, Norah Alsaleh1;6 Zafer Yüksel 1, Steffen Kühner1, Nahid Nahavandi1, Roschan Salimi Dafsari2, Mojgan Drasdo1 Hazal Banu Olgun Celebioglu 1;2, Ayse Pinar Ozturk3, Şükran Poyrazoğlu3, Feyza Tuncer1 Sebastian Skoczylas 1, Tomasz Płoszaj1, Magdalena Traczyk-Borszyńska1, Karolina Gadzalska1, Paulina Jakiel1, Agata Pastorczak2, Ewa Juścińska1, Monika Gorządek1, Maciej Borowiec1, Agnieszka Zmysłowska1 Jennifer Goldstein 1, Heidi Wallis2, Emily Reinhardt2, Juliann Savatt3, Erin Rooney Riggs3, Amanda Thomas-Wilson4, Emily Groopman5, Vimla Aggarwal6, Simona Bianconi7, Raquel Fernandez8, Kim Hart9, Emily Kyle8, Nicole Liang10, Nicola Longo11, Christine Preston12, Daniel Reich13, Meredith Weaver8, Sarah Young14, Saadet Mercimek-Andrews15 Yasmina Elaribi 1, Sana Karoui2, Imen REJEB1, Alaa Ziadi1, Zohra Fitouri3, Manel Lajimi1, Syrine Hizem1, Houweyda Jilani1, Lamia BenJemaa1 guanjie chen 1, Ayo Doumatey1, jie zhou1, Lin Lei1, Charles Rotimi1, Adebowale Adeyemo1 Parnian Alagha 1;2, Fatemeh Ahangari1, Maryam Beheshtian1, Negar Molaei1;2, shima Dehdahsi1, Mahsa Fadaie1, Mehri Ashki1, Zohreh Elahi1, Maryam Mozaffarpour1, Parishad Saei1, Fatemeh Fatehi1, Khadijeh Noudehi1, Shima Zamanian Najafabadi1, Kimia Kahrizi2, Ariana Kariminejad1, Hossein Najmabadi1;2 amanda herranz cecilia1;2, Carmen Rodríguez Jiménez1;2;3, Ana Carazo1;2, natividad gallego onís3, María Esther Rubio Martín3, Maria Victoria Del Pozo-Gomez3, Juan Manuel Montejo4, laura García Fernández4, Carlos Rodríguez-Antolín5, Ana Bergua Martínez6, Jose David Andrade Guerrero6, Ana Morais López6, Sonia Rodriguez Novoa 1;1;2;2 Andžela Lazdāne 1, Svetlana Vorslova1, Zanda Daneberga1;2, Ieva Grīnfelde1;2, Madara Auzenbaha1;2, Ieva Pukite1;2 Karl Hackmann 1, Joseph Porrmann1, Sophie Henke2, Friederike Quitter2, Franziska Lange2, Evelin Schröck1;3, Doreen William1, Andreas Dahl4, Min Ae Lee-Kirsch5;6, Angela Hübner2, Katrin Koehler2 Frank Sleutels 1, S. Demirdas1, Esmee Oussoren2, Lies Hoefsloot1 Mustafa Yilmaz1, AYBERK TURKYILMAZ1, KUBRA ADANUR SAGLAM1, nazime cebi2, Alperhan Cebi 1 Florencia Epifani 1, Lluc Cabus2, Gregorio Nolasco1, Mercè Bolasell1, Adrián Alcalá San Martín1, Patricia Fernández1, Gisela Márquez1, Cristina Hernando-Davalillo1, Jennifer Pérez2, Esther Lizano2, Silvia Carbonell-Sala3, Joao Curado2, sonia belmonte2, Mercedes Serrano1;4 Laura Gort 1;2;3, José Manuel Gonzalez de Aledo1, Abraham J. Paredes-Fuentes1, Ana Argudo-Ramirez1, Laura Pacheco1, Rosa Maria López-Galera1;2;3, José Antonio Arranz4, Clara Carnicer4, Ana Felipe4, Mireia Del Toro4, Antonia Ribes2;3, Judit Garcia-Villoria1;2;3 Houyem Ouragini 1, Faten Fatnassi1, Ilhem BenFraj2, Dorra Chaouachi1, Monia Ouederni2, Samia Menif1 Katherine Schon 1, Peter Stilwell2, Jeanette Aston2, Katrina Lavelle2, Margreet Luchtenborg2, Robert Pitceathly3, Michael Hanna3, Carl Fratter4, Rita Horvath5, Mary Bythell2, Steven Hardy2, Patrick Chinnery5 Polina Tsygankova 1, Denis Chistol1, Tatiana Krylova1, Tatiana Markova2, Elena Dadali2, Ekaterina Zakharova1 Elif EROĞLU ŞİMŞEK 1, Ibrahim Akalin1, Murat Coskun2, Ayse Busra Akalin3, İlknur Sunar1, Nursel Elcioglu4 Mazhor Aldosary 1, Zuhair Alhassnan2, Dilek Colak3, Mohammad Al Muhaizea4, Namik Kaya1 Agnese Zarina 1;2;3, Agrita Puzuka1;2, Madara Auzenbaha1;2, Ieva Tolmane4;5, Zita Krumina6, Linda Gailite1, Dmitrijs Rots1 Ligia Almeida 1, Catarina Pereira1, Mariana Ferreira1, Lia Abbasi Moheb1, Jorge Pinto Basto1, Omid Paknia1, Peter Bauer1 Inga Poceviciene 1, Inga Nasvytienė1, Rasa Ugenskiene1;2 Daniela Arturo-Terranova1;2, Lina Moreno-Giraldo3, Jose Maria Satizabal Soto 1 Nina Maric 1, Bozica Kecman2, Slavica Ostojic2 EP09 Immunology and Hematopoietic System Rungroj Thangpong 1;2, Phawin Kor-anantakul1;2, Wuttichart Kamolvisit1;2, Chupong Ittiwut1;2, Rungnapa Ittiwut1;2, Narissara Suratannon3, Vorasuk Shotelersuk1;2, Kanya Suphapeetiporn1;2 Biljana Todorić-Živanović 1, Milica Strnad1, Lavinika Atanaskovic2, Marija Elez2 Rosa Santacroce 1, Giorgia Cordisco1, Alessandra Ranaldi1, Daniel Vazquez2, Anna Laura Colia1, Angelica Leccese1, Maria Francesca D’Ambrosio1, Giovanna D’Andrea1, Dario Josviack3, Maurizio Margaglione1, Maria d’Apolito1 İlknur Sunar1, Ayse Busra Akalin2, Ibrahim Akalin1, Fatih Demircioğlu 3, Mohammed Alchalabi4, Zeynep Derin5, Betül Tavil6 Gabriela Krasnanska 1;2, Lenka Wachsmannova1, Marian Baldovič1;3, Gabriela Bľandová1;4, Vladimír Eliáš1, Michal Konecny1;2 Manca Svetina 1, Mitja Košnik1;2, Renato Eržen1;3, Peter Kopač1;2, Peter Korošec1;4;5, Matija Rijavec1;6 Willemijn Kuper 1, Eva Rettenbacher2, Annet Van Hagen1 Victoria Paul 1, Nadia Korthals1, Judit Horvath1, Frank Tüttelmann1, Bodo Grimbacher2;3;4, Eva Eßeling5 Rasa Insberga 1;2, Ieva Mičule1, Ludmila Voložonoka1, Dmitrijs Rots1, Dace Enkure1, Anna Valaine3 Ana Flores-Chova 1, Olga Martinez-Arroyo1, Marta Mendez-Debaets1, Laia Garcia-Ferran1, Lorena Adan1, Liria Terrádez2, Josep Redon1;3, Maria J Forner1;3;4, Ana Ortega1, Raquel Cortés1 Lane Scheiber II1, Neha Gupta 2, Sarah Ford1, Evan Dombrosky1 Jasmin Blatterer 1, Sabrina Hammer1, Sophie Bierbaumer1, Rusmir Husic2, Johannes Schmid3, Elisabeth Schreiner1, Heidelis Tichy1, Klaus Wagner1, Sarah Verheyen1 Malena Gajate-Arenas 1, Ingrid Fricke-Galindo2, Omar García-Pérez1, Angélica Domínguez-de-Barros1, Gloria Pérez-Rubio2, Roberto Dorta Guerra1;3, Ivette Buendia Roldán4, Leslie Chavez-Galán5, Jacob Lorenzo-Morales1;6;7, Ramces Falfán Valencia2, Elizabeth Córdoba-Lanús1;6;7 Cristina-Loredana Pantea 1;2, Adela Chirita-Emandi1;2, Mihaela Bataneant3, Maria Puiu1;2 Katharina Schau-Römer 1, Simon Vieth2, Almuth Caliebe2, Malte Spielmann1;3, Inga Nagel1 Melania Eva Zanchetta 1, Serena Barozzi2, Federica Isidori3, Caterina Marconi3, Loredana Farinasso4;5, Roberta Bottega1, Anna Savoia6, Alessandro Pecci2;7, Michela Faleschini1 Sude sultan Özdemir 1, Ibrahim Akalin2;3, Fatih Demircioğlu4, İlknur Sunar5, AHMED AL-OMAR6, Ayşe Akalın7, Betül Tavil8 Quentin Sabbagh1, Jean-David Cohen2, Jeremie Mortreux3, Laure Raymond3, Guilaine Boursier4, Vanna geromel3, eric jeziorski5, Vincent Le Moing6, Didier Bessis7, David Geneviève 1 Ahmed Adel Gereisha 1, Rajaa Lahmar1, Rasene Gereisha1, Elyes Chabchoub1, Ramzi Zemni1, Anis Mzabi2, Neirouz Ghannouchi3, Foued Ben Hadj Slama1 Vivekananda Bhat 1, Namanpreet Kaur1, Amit Jairaman1, Jeevan Kumar1, Radhakrishnan Periyasamy1, Anju Shukla1 Betül Tavil 1, Motusheva Raushan Kimashbekovna2, İlknur Sunar3, Ayse Busra Akalin4, Ibrahim Akalin5 Volodymyr Kravets1;2, Maria Dushar1, Ivanna Shymanska1, Olena Boldyreva3, Liliya Nazarenko4, Halyna Makukh 1;2 Emine Göktaş 1, sümeyye şanal1, hüseyin tokgöz2, Ayse Gul Zamani1, selman yıldırım1 Antonia Vardeva 1, Hristo Ivanov1, Hassan Burnusuzov1, Vili Stoyanova1 Marieke De Bruyne 1;2, Lore Pottie1;2, Stijn Van de Sompele1;2, Nancy De Cabooter1;2, Delfien Bogaert3;4, Levi Hoste4, Toon Rosseel1;2, Kim De Leeneer1;2;5, Victoria Bordon3, Jana Neirinck6;7, Mattias Hofmans7, Carolien Bonroy6;7, Simon J. Tavernier1;2;4, Kathleen Claes1;2;5, Filomeen Haerynck4;8, Elfride De Baere1;2 Elisabeth Schreiner 1, Sophie Bierbaumer1, Jasmin Blatterer1, Sabrina Hammer1, Stefan Kuehberger1, Heidelis Tichy1, Klaus Wagner1, Sarah Verheyen1 Livia Varga 1;2, Nora Meggyesi1, Andras Bors1, Katalin Balassa2, Viktor Lakatos1, Melinda Paksi1, Laszlo Gopcsa1, Attila Tordai2, Masszi Tamás2, Peter Remenyi1, Hajnalka Andrikovics1 Omar García-Pérez1, Malena Gajate-Arenas1, Angélica Domínguez-de-Barros1, Candela Sirvent-Blanco1, Alma García-Ramos1, José E Piñero1;2;3, Jacob Lorenzo-Morales1;2;3, Elizabeth Córdoba-Lanús 1;2 Fatma Turki 1, hassen kamoun1, Rim Frikha1 Marija Rozevska 1;2, Viktorija Arnite2, Gita Taurina2, Ineta Grantina1;2, Inga Nartisa1;2, Dmitrijs Rots1;2, Natalja Kurjane1;2 Milica Strnad 1, Biljana Todorić-Živanović1, Lavinika Atanaskovic2, Marija Elez2 EP10 Intellectual Disability Olga Levchenko 1 Elsa Leitão 1, Sabine Kaya,1, Anne Faudet2, Frédéric Tran Mau-Them3;4, Isabelle An5, Perrine Charles2, Delphine Heron2, Christel Depienne1 Júlia Krisztina Erhardt 1, Krisztina Nemeth1, Éva Pállinger2, Gyorgy Fekete1, Árpád Ferenc Kovács1 Katarzyna Połatyńska 1, Tadeusz Kałużewski2, Agnieszka Gach2, Łukasz Kępczyński2 María Juliana Ballesta Martínez 1, María José Sánchez Soler1, Ana Teresa Serrano Antón1, Marta Domínguez Jiménez1, Vanessa Lopez1, Encarna Guillén-Navarro1 Bilgen Bilge Geçkinli 1, aysenur ersoy1, onur hanoglu1, Vedat Yuce1, mehmet ali soylemez1, Ahmet Arman1 Klaudia Berk 1, Renata Posmyk1 Özlem Baysal 1, Jikke-Mien Niermeijer2, Rolph Pfundt1;3, Bert de Vries1;3 Dmytro Mykytenko 1;2 Salwa Ben Yahia 1, Ahlem Achour1, Syrine Adhoum1, Amelie PITON2, faouzi maazoul1, jamel chelly2, ridha mrad1, Mediha Trabelsi1 Delia Sabau 1;2, Antoanela Curici2;3, Mihai Mitroi2 Paula Muñiz Sevilla1, alba navarro romero 1, Marián Lázaro1, Laura Rausell1, Pablo Gargallo1, Susana García-Linares2 Virginia Garcia-Solaesa 1, aranzazu perez-juana1, Mercè Artigas López1, Maria Ordóñez-Marina1, amaya bengoa alonso1, Fermin Garcia-Amigot1, Sara Pasalodos-Sanchez1, Lourdes Morales-Garófalo2, Eva Barbon-Alonso3, Maria Antonia Ramos-Arroyo1 Mohammad Reza Mahdavi 1, Atefeh Khoshaein2, Hossein Jalali1, Mohammad Amirzadegan2, Mahan Mahdavi2 Diogo Fernandes da Rocha 1, Vera Santos1, Sofia Dória2, Ana Grangeia1;2 Olga Shatokhina 1, Fatima Bostanova1, Maria Bulakh1, Oksana Ryzhkova1 Behzad Davarnia 1, farzad ahmadabadi2, mohammad jahanpanh1, diana mokhtari1, Sara Arish1 Charley Robert-Viard 1, Anne-Laure Soilly2, Celine Besse3, anne boland3, Delphine Bacq3, Alexia Benoit4, Hamza Hadj-Abdallah5;6, Benedicte Gerard4, Charlotte Poe7, Ange-Line Bruel7, Patrick Nitschké8, Alban Simon9, Stanislas Lyonnet5;6, Laurence Faivre7;10, Christel Thauvin-Robinet7;10, Sylvie Odent11, Delphine Heron12, Damien Sanlaville13, Thierry Frebourg14;15, Jean Muller4;9;16, Yannis Duffourd7, Marion Bouctot1, Niki Sabour1, Marie-Laure Humbert1, Christelle Delmas17, Jean-François Deleuze3, Francis Guillemin18, Valerie Seror19, Hamza Achit18, Helene Esperou17, Christine Binquet1, Hélène Dollfus9, Catherine Lejeune1 Michal Konecny 1;2, Gabriela Krasnanska1;2, Silvia Lakatosova3, Lenka Wachsmannova1, Marian Baldovič1;4, Vladimír Eliáš1, Gabriela Bľandová1;5, Gabriela Repiska6 Bashair Magadmi 1, Thomas Courtin1, Sophie Rondeau1, marion lesieur-sebellin1, Giulia Barcia1, Marlene Rio1 Asem Alkhateeb 1 Nuray Ozturk 1, Mert Coşkun2, Duygu Gamze Aracı2, sezin yakut uzuner3, Aslı Toylu2, Gokcen Karamik1, Öznur YILMAZ BAYER1, guldeniz oner1, banu nur1, ercan mihci1 Norah Alsaleh 1;2 RAYANA MAIA 1, Matheus Lira2, Erika Santos2 Teresa CARRION 1;2, Maria Antonia Grimalt3, Victor Asencio4, MARIA CAIMARI JAUME5, Jordi Roldan6 Zarah Kowalzyk 1, Marion Bermudez1, Joseph Porrmann1, Nataliya DiDonato1;2, Christoph Hübner2, Andreas Dahl3, PY Billie Au4, Lindsay Phillips4, Sanaa Choufani5, Rosanna Weksberg5;6, Doreen William1, Evelin Schröck1;7, Arne Jahn1 Ana Capela 1, Cláudia Falcão Reis1;2;3;4 Gergely Büki 1, Agnes Till1, Tímea Bujtás1, Anna Zsigmond1, Krisztina Galimurka1, Attila Gyenesei2, Kinga Hadzsiev1, Judit Bene1 Zivile Zemeckiene 1;2, Rasa Traberg1;2, Inga Nasvytienė1, Marius Šukys1;2, Rasa Ugenskiene1;2 Vladimira Vallova 1;2, Dominik Režný1, Jan Smetana1, Renata Gaillyova3, Petr Kuglik1;2 Fatma Majdoub 1, Imene Boujelbene2, Manel Guirat2, Alaa Ziadi2;3, Amal Souissi4, Ines Hsairi5, ridha mrad3, Hassen Kamoun2, Sabeur Masmoudi4, Mediha Trabelsi3, Ikhlas Ben Ayed2;4 Laura Trujillano 1, Irene Valenzuela Palafoll1, Mar Costa-Roger1, Ivon Cuscó2, Anna Maria Cueto-González1, Amaia Lasa-Aranzasti1, Barbara Masotto1, Anna Abulí1, Marta Codina1, Ana Maria Cordero1, M Luisa Carcas Espinosa1, Miguel del Campo3, Juan Antonio Moreno Ruiz4, Cristina Pardo Dominguez4, Carmen Palma Milla5;6, Rubén Pérez de la Fuente5;6, Juan Francisco Quesada Espinosa5;6, Blanca Gener-Querol7, María Juliana Ballesta Martínez8, Alejandro J Brea-Fernández9;10, Montse Fernández-Prieto9;10, Elisabet Gabau11, Anna Ruiz11, Juan Pablo Trujillo Quintero11, Fernando Santos-Simarro12, Mónica Roselló Piera13, Carmen Orellana13, Francisco Martinez13, Didac Casas-Alba14, Mercedes Serrano15, María Palomares-Bralo16, Emi Rikeros Orozco16, María de los Ángeles Gómez Cano16, Pilar Tirado-Requero17, Elena García-Arumí1, Eduardo Tizzano1 Teodora Barbarii 1, Diana Barca2;3 Karit Reinson 1;2, Kaisa Teele Oja1;2, Mihkel Ilisson2, Eleanor G. Seaby3, Monica H Wojcik3;4, Anne O’Donnell-Luria3;4, Bryan M Wittmann5, Adam Kennedy5, Karen DeBalsi5, Christiane Zweier6;7, Georgia Vasileiou6;7, Issam A. Alkhawaja8, Thomas Wirth9, Steffen Syrbe10, Suad Alyamani11, Fowzan Alkuraya11, David Ros-Pardo12, Iñigo Marcos-Alcalde12, Paulino Gomez-Puertas12, Ke Su13;14, Shaohua Fan13, Yu Ma15, Yi Wang15;16, Marie Vidailhet17, Alessandra Renieri18;19;20, Anna Maria Pinto20, Farah Almadhoun18, Andrea Petersen21, Kari Magnussen21, Olaf Bodamer21, Nicole Fleischer22, Amber Begtrup23, Sander Pajusalu1;2, Katrin Ounap1;2 Sarah Baer 1;2, Merol Jeanne1, Lamouche Jean-Baptiste1, Geoffroy Véronique3, Tarek Alouane1, Salima EL CHEHADEH1;4, Valérie Skory1, sophie scheidecker3;5, Benjamin Durand4, Elise Schaefer4, Jean Muller3;5, Amelie Piton1;5 Linda Sofan 1, Rebecca Buchert1, Wael Al-Ameri2, Olaf Riess1, Tobias Haack1, Tawfiq Froukh3 Louisa Sanchez 1, Evelyn Douglas1, Sin Lay Kang1, Kathy Cox1, Linda Burrows1, Abhi Kulkarni2, Karin Kassahn2, Lesley McGregor3, Jan Liebelt3, Suzanne Sallevelt3, Shannon LeBlanc3, Christopher P. Barnett3, Song Gao2, Evelyn Collen2, Kathryn Friend1, Sui Yu1 Judit Bene 1, Anna Zsigmond1, Gergely Büki1, Attila Gyenesei2, Zsuzsa Siegler3, Kinga Hadzsiev1 Eva-Maria Kraus 1, Robin-Tobias Jauss1, Konrad Platzer1, Franziska Schnabel1 Juliane Hoyer 1, Cornelia Kraus1, André Reis1;2 Maria Franaszczyk 1;1, Aleksandra Świeca1, Agnieszka Pollak1, Piotr Gasperowicz1, Grażyna Kostrzewa1, Piotr Stawiński1, Karolina Rutkowska1, Małgorzata Rydzanicz1, Krzysztof Szczałuba1, Rafał Płoski1 alejandro parra 1;2;3, Jair Tenorio1;2;3, julian nevado1;2;3, Mario Cazalla1;2;3, Patricia Pascual Vinagre1;2;3, Lucía Miranda-Alcaraz1;2;3, Natalia Gallego1;2;3, cristina silvan2, Pedro Arias3, Jesús Pozo-Román4;5, María Juliana Ballesta Martínez6;7, Encarna Guillén-Navarro1;7, ignacio arroyo8, Pablo LAPUNZINA1;2;3 Nada Amllal 1;2, maria zerkaoui3, Wafaa Jdioui3, abdelaziz sefiani1;2, siham chafai elalaoui3, lyahyai jaber1 IRATXE HUERTA 1, Mertxe Télez1, maría garcía barcina1 Barbara Golob 1, Mihael Rogac1, Aleš Maver1, Borut Peterlin1 Urszula Wysocka 1, Łukasz Kępczyński1, Agata Kucińska1, Katarzyna Połatyńska2, Tadeusz Kałużewski1, Agnieszka Gach1 Riina Žordania 1;2, Kati Kuuse3, Ülle Murumets3, Edgar Tarelkin4, Laura Roht2 Zianka Meyer 1, Stefan Weiß1, Douglas Friday1, Sophia Kummerow1, Irina Hüning2 Anna-Sophie Liegmann 1, Kirstin Hoff2, Britta Hanker1, Malte Spielmann1;2, Inga Nagel2 Juan Pablo Trujillo-Quintero 1, Carmen Manso2, Elisabeth Gabau Vila1, Neus Baena1, Núria Capdevila1, Anna Brunet-Vega1, Victor Martinez-Glez1, Nino Spataro1, Anna Ruiz1 Ramya Sukrutha 1, ananthapadmanabha kotambail1, GAUTHAM ARUNACHAL UDUPI1, CHETAN GHATI1 ABDULLAH SEZER1, Damla Puçak 2, Sebiha Cevik2, Oktay I. Kaplan2 Noel Macmanus1, Asunción Díaz1, Alícia Artigas Baleri2, Lucia Dougherty de Miguel1, Eulàlia Turón-Viñas1, Susana Boronat1, Ivon Cuscó 2;3 Marta Domínguez Jiménez 1, Ana Teresa Serrano Antón1, María José Sánchez Soler1, Julia Araújo De Castro2, Sonia Martínez Peñas2, Vanessa Lopez1;3, María Juliana Ballesta Martínez1;3, Encarna Guillén-Navarro1;3 Marine Tessarech 1;2, Gaelle Friocourt3, Florent Marguet4, Maryline Lecointre4, Le Mao Morgane2, Cyril Mignot5, Boris Keren5, Benedicte Heron6, Charlotte De Bie7, Koen Van Gassen7, Didier Loisel8, Benoit Delorme8, Steffen Syrbe9, Rodrigo Muñoz Díaz2, Klabunde-Cherwon Annick9, Rami Abou Jamra10, Meret Wegler10, Bert Callewaert11, Annelies Dheedene11, Merzouka Zidane-Marinnes12, Agnes Guichet1;2, Céline Bris1;2, Patrick Van Bogaert13, Florence Biquard14, guy lenaers2, Pascale Marcorelles15, Claude Férec3, Bruno Gonzales4, Vincent Procaccio1;2, Antonio Vitobello16, Dominique Bonneau1;2, Annie Laquerriere4, Salim Khiati2, Estelle Colin1;2 Solveig Heide1, Adrien Legrand2, Cristina Alina Birzu3, Daphné Lehalle1, Romain Duquet1, Anna Gerasimenko1, Delphine Heron1, Perrine Charles 1 Aïcha Boughalem 1, aziza belkhayate2, Laurence Lohmann1, Mylene Valduga1, Pascale Kleinfinger1, Aline Receveur1, stéphane serero3, Detlef Trost1 Ilona Jaszczuk 1, Monika Lejman2, Borys Styka2, Maciej Geremek3, Beata Nowakowska3, Izabela Winkler4, Agata Filip1 Marina Carapelle1, Silvia Frangella1, Anna Gloria Renzi1, Domizia Pasquetti1, Elena Sonnini1, Annalisa Gazzellone1, Clarissa Modafferi1, Pino D’Ambrosio1, VALENTINA TREVISAN1;2, Arianna Panfili3, Paolo Niccolò Doronzio1, Giuseppe Marangi1, Daniela Orteschi1;4, Emanuela Lucci Cordisco4, Pietro Chiurazzi4, Ilaria Contaldo5, Chiara Veredice6, Federica Francesca L’Erario 1, Marcella Zollino4;7 stéphanie moortgat 1, Anne Destree1, Deniz Karadurmus1, Olivier Monestier1 chitra ramalingam1, Jiin Ying Lim 2;3;4, sylvia kam2;3;4, Hai Yang Law2, nur afiqah binte mohd mislan2, weng khong lim3;5, ivy ng2, terrence thomas6, sandra sylvia mascarenhas1, saumya shekhar jamuar2;3;4;5 ANGEL ZUNIGA 1, Graciela Pi2, ANTONIO MARTINEZ CARRASCAL3 Arzu Guliyeva 1;2, Elif Yilmaz Gulec1;2, Filiz Ozen2 Ivan Tourtourikov1;2, Malina Stancheva3, Tanya Kadiyska 2;4 Susanne Ledig 1, Ann-Christin Tewes1, Frank Tüttelmann1, Oliver Schwartz2, Judit Horvath1 fatima ouboukss 1, ZHOUR EL AMRANI1, hicham bouchahta1, ilham ratbi2, Thomas Liehr3, abdelaziz sefiani1, Abdelhafid Natiq2 Camille Bergès 1, Clément SAUVESTRE2, Sophie Naudion2, Catherine Vincent-Delorme3, Thomas Smol3, melanie rama3, Muhammad Umair4, Reza Maroofian5, Tobias Haack6, Mona Grimmel6, Anne Slavotinek7, Stephanie Efthymiou5, Faisal Zafar8, Erum Afzal8, Tracy Dudding-Byth9, Vincent MIchaud1 EP11 Neurogenetic and Psychiatric Disorders Slavko Ormandzhiev 1;2, Tihomir Todorov2, Teodor Angelov3, Teodora Chamova3, Vanyo Mitev1, Albena Todorova1;2, Ivaylo Tarnev3;4 Erkan Koparir 1, Thomas König2, Erdmute Kunstmann1, Juliane Spiegler2, Thomas Haaf1, Eva Klopocki1 Christina Stoikova 1;1;1, Nevyana Ivanova1, Kalina Mihova1, Sashka Zhelyazkova2, MARIA PETROVA3, Genoveva Tacheva4, Ilyana Pacheva5, Iliyana Aleksandrova6, Daniela Deneva6, Ivan Litvinenko4, Ivan Ivanov5, Veneta Bojinova-Tchamova6, Ivaylo Tarnev2, Albena Jordanova7, Ivanka Dimova1, Radka Kaneva1 Nevyana Ivanova 1, Kalina Mihova1, Christina Stoikova2, Maria Sredkova-Ruskova3, Trayan Delchev3, Veneta Bojinova-Tchamova4, Daniela Avdjieva-Tzavella3, Vanyo Mitev2, Albena Jordanova1;5, Ivanka Dimova6, Radka Kaneva1 Lisa Pavinato 1;2;3, Antonietta Verrillo1;2, Chiara Leso4, Erika Ortolan3, Eleonora Campus5, Elena Rita Vecchi5, Andrea Angius6, Vincenzo Rallo6, Enza Ferrero3, Giovanni Battista Ferrero7, Luciano Conti5, Arianna Baggiolini1;2, Alfredo Brusco8;9 Laia Rodriguez-Revenga 1, Judit Garcia-Guallarte2, Germán Casabó-Vallés2, Trinitat Alberola-Pons2, Irene Madrigal1, Maria Isabel Alvarez1 Hasnaa Elbendary 1, maha zaki1, Karima Rafat1, Mohamad Abdelhamid2 Sabrina Stefanelli 1, Stephanie Jünemann2, Pascal Joset1, Isabel Filges1 Łukasz Kępczyński 1, Anna Łupińska2;3, Paula Smalczewska2, Katarzyna Połatyńska4, Jordan Sałamunia5, Tadeusz Kałużewski1, Renata Stawerska2;3, Arkadiusz Zygmunt2;3, Andrzej Lewiński2;3, Agnieszka Gach1 Antonio Federico Martínez-Monseny 1, Lourdes Vega Hanna1, didac casas-alba1, francesc palau martinez1, merce bolasell1, patricia rubio1, mar o’callaghan1, Ainhoa Pascual-Alonso1, sol balsells1, Judith Armstrong1 Nuno Ferreira 1 Ayfer Pazarbasi 1, Gulsevinc Aksoy1, İnayet Nur Uslu1 Lütfiye Özpak 1 Tarik Duzenli 1, gulsum kayhan1 Friedrich Stock 1, Jasmin Beygo1, Ilaria Parenti1, Harald Surowy1, Christopher Schröder1, Elsa Leitão1, Antje Kampmeier1, Christel Depienne1, Frank J. Kaiser1;2, Alma Küchler1 Fernanda Francis-Cartin 1;2, Esteban J. Rodriguez1;3;4, Gabriela Chavarria-Soley1;5, Alejandro Sanchez-Flores6;7, Henriette Raventós1;5 Esteban J. Rodriguez 1;2;3, Fernanda Francis-Cartin1;4, Gabriela Chavarria-Soley1;5, Henriette Raventós1;5 Timotej Glavač 1, Maruša Barbo2, Sara Redenšek Trampuž2, Metka Ravnik-Glavač2, Maja Zupančič1, Vita Dolzan2 Selinda Mascarenhas 1, Vaishnavi Badiger1, Sheeba Farooqui1, Anju Shukla1, Radhakrishnan Periyasamy1 Yasaman Pakdaman 1, Somayeh Bakhtiari2;3, Roseline Caumes4, paskal cullufi5, Yiran Xu6, Changlian Zhu6;7, Yangong Wang8, Thomas Smol9, Michael C. Kruer2;3, Aboulfazl Rad1, Shivendra Kishore1, Gabriela Oprea1 Odelia Chorin 1;2, Ben Pode Shakhed1, annick REIN ROTHSCHILD1, Lior Greenbaum2, Shelly Lev-Hochberg1 Carmen Manso1, Nino Spataro1, Lidia Torrent2, Laura Plans3, Mercè Casadesus3, Victor Martinez-Glez1, montserrat pamias2;4, Anna Ruiz 1 Pablo Iruzubieta 1;2, Cesar Augusto Pinheiro Ferreira Alves3, Aisha Mohamed Saeed Mohamed Al Shamsi4, Gehad ElGhazali5, Lorenzo Pinelli6, Diego Lopergolo7, Bernard P H Cho8, Amna Mohammed Al-Futaisi9, Fatema Al-Amrani9, Jessica Galli10, Katarina Vulin11, Elisa Fazzi10, Francisco Barajas-Olmos12, Holger Hengel13, Amy Jolly8, Maha Zaki14, Bayan Mohammed Aljamal15, Fowzan Alkuraya15, Michele Ragno16, Luigi Trojano17, Silvia Bianchi7, Francesca Pescini7, Amal Al Tenalji18, Majid Aziz18, Miriam Schmidts19;20, Giovanni Zifarelli21, Ludger Schöls22, Tobias Haack23, Adriana Rebelo24, Stephan Zuchner24, Lyn Griffiths25, Arman Cakar1, Lorena Sofía Orozco Orozco26, Karla García Helmes27, Meisam Babaei28, Peter Bauer21, Won Chan Jeong29, Ehsan Ghayoor Karimiani30, Wendy Chung31, Vahideh Nasr32, farhad assarzadegan32;33, Bita Shalbafan33, Hugh Markus8, Henry Houlden1, Reza Maroofian1 Zoia Rossokha 1, Maya Lafarenko2, Nazar Negrych3, Liliia Fishchuk1, Tetyana Nehrych4 Idris Janos Jimoh 1, Agnes Palasti1, Viktor Molnár1, Zsuzsanna Aranyi2, Zoltan Grosz1, Maria Judit Molnar1 Duygu Gamze Aracı 1, Özden Altıok Clark1, Öznur YILMAZ BAYER2, banu nur2, Senay Haspolat3, Aslı Toylu1, ercan mihci2 hacer ukba kına 1, Neslihan Cinkara1, Momen Kanjee2 András Szabó 1, Márta Czakó1, Gergely Büki1, Zsolt Bánfai1, Anita Maasz1, Agnes Till1, Barbara Patócs2, Judit Bene1, Kinga Hadzsiev1 Anita Maasz 1, Tímea Bodó2, Agnes Till1, Gábor Molnár3, György Masszi4, Gusztáv Labossa3, Zsuzsanna Herbert5, Judit Bene1, Kinga Hadzsiev1 Andrea Stoccoro 1, Eugenia Conti2, Elena Scaffei2, Laura Baroncelli3, Marianna Giangreco1, Sara Calderoni2, Fabio Coppedè1, Lucia Migliore1, Roberta Battini2 Lubaba Bintee Khalid1, Bilal Ahmad Mian1, Asmat Ali1, Mina Shahid2, Mathias Toft3;4, Zafar Iqbal4, Ambrin Fatima 1 Luciana Musante 1, Maria Teresa Bonati1, Giulia Maria Di Marzio1, Martina La Bianca1, Giorgia Girotto1;2, Caterina Zanus1 Yiwen Huang1, Irene Guerrini2;3, Iain Smith4, Mathis Heydtmann5, Marsha Morgan6, Allan Thomson1;3, Nicholas Bass1, Andrew McQuillin 1 Joseph Porrmann 1, Sylvia Hütter1, Jens Schallner2, Andreas Dahl3, Karl Hackmann1, Marcus Franke1, Evelin Schröck1;4 Mustafa Bakırtaş 1, Alara Akdeniz2, Hazel Delal Dara Kar3, Zerrin Çelik1, Yunus Kasım Terzi1, Halil İbrahim Aydın4 Houda Jelti 1;2, Sabrine Bouressas1;2, Abdelhaq Lamaibdel1;2, Oussama Kettani1, Jaouhara MAAMAR1;2, Ouafae Kaissi1, Badreddine Nouadi1, Hasna Hamdaoui1, Afaf Lamzouri1;2 David Vogrinc 1, Milica Gregorič Kramberger2;3;4, Andreja Emeršič2, Saša Čučnik2;5;6, Katja Goricar1, Vita Dolzan1 Enrico Ambrosini 1, Rita Cancilla2, Jefri Jeya Paul3, Peter Bauer3, Barbara Garavaglia4, Valeria Barili5, Antonio Percesepe1;5, Anna Negrotti6 Shokhijakhon Shokhimardonov 1, Nodira Tuychibaeva1 sara khadem ansari1, Ebru Erzurumluoglu Gokalp1, emre ozkara2, Ozlem Aykac3, Atilla Ozcan Ozdemir3, Sevilhan Artan 1 Buşra Kasap 1, Cengiz Yalçınkaya2, Dilek Uludağ Alkaya1, Filiz Geyik3, Nilay Güneş1, Sema Saltık4, Ahmet Okay Çağlayan5, Beyhan Tüysüz1 Engin Atli 1, Sinem YALÇINTEPE1, Veysel Oz2, Drenushe Zhuri1, Yasemin Ozen1, Emine Ikbal Atli1, Selma Demir1, Işık Görker3, HAKAN GURKAN1 Krisztina Galimurka 1, Renata Szalai1, Agnes Till1, Judit Bene1, Attila Gyenesei2, Kinga Hadzsiev1 Nino Spataro 1, Núria Capdevila1, Carmen Manso1, Viviana P Beltran-Salazar2, Victor Martinez-Glez1, Anna Ruiz1 Wissal Tergui 1, Ahlem Achour1;2, Nesrine Kerkeni1;2, Amira Zanati1, Wiem Essalah1, Zouhour Miladi3, Ichraf Kraoua3, Ilhem Turki3, ridha mrad1;2, Mediha Trabelsi1;2 Maya Atanasoska 1;2, Lubomir Balabanski1, Slavyana Yaneva Staykova1, Irena Bradinova1;3, Draga Toncheva1;4, Radoslava Vazharova1;5 Neus Baena 1, Montserrat Garcia-Puig2, Anna Brunet-Vega1, Laura Capel1, Carmen Manso1, Juan Pablo Trujillo Quintero1, Núria Capdevila1, Victor Martinez-Glez1 Lilla Buzai-Kiss 1;2, Barbara Trombitas1, Péter Balicza1;2, Judit C. Sági1, szabolcs udvari1, Maria Judit Molnar1;2 Eva Pinti 1, Anna Lengyel1, Agnes Sallai1, Andras Seres2, Gyorgy Fekete1, Iren Haltrich1, Kálmán Tory3 ASLI KARACAN 1;2, Ayça Dilruba aslanger1, Guven Toksoy1, Zehra Oya Uyguner1, Ayse Evrim Bayrak1 Dimitar Serbezov 1, Sena Karachanak-Yankova1;2, Dragomira Nikolova1, Marta Mihaylova1, Savina Hadjidekova1, Diana Belejanska3, Shima Mehrabian3, Mariya Petrova3, Latchezar Traykov3;4, Draga Toncheva1;4 Tamás Szlepák 1;2, Bernadette Kálmán3, Róbert Sepp4, Judit Mária Molnár1;2 Hülya Şenol1, Tahmina Chalabi Kurt1, Serdal Işıktaş1, Şükrü Tüzmen 2;3 Dragomira Nikolova 1, Sena Karachanak-Yankova1;2, Dimitar Serbezov1, Mikaela Stancheva2, Desislava Nesheva1, Marta Mihaylova2, Diana Belejanska3, Mariya Petrova3, Shima Mehrabian3, Latchezar Traykov3, Savina Hadjidekova1, Draga Toncheva4 Mohammad Al Muhaizea1, Ali Alshehri1, Hanan AlQudairy2, Sarah Alruways2, Nouf Almutairi2, Rawan Almas3, Albandary Al-Bakheet2, Aljouhra AlHargan2, Anoud Albader2, Ghadeer Alhamid2, Dilek Colak4, NAMIK KAYA 2 Andrea Pietra 1;2, Mina Grippa2, Elisabetta Spezia3, Patrizia Bergonzini3, Alberta Patanè4, Alberta Leon4, Lorenzo Iughetti3, Olga Calabrese2 Sena Karachanak-Yankova 1;2, Dimitar Serbezov1, Dragomira Nikolova1, Desislava Nesheva1, Marta Mihaylova1, Mikaela Stancheva2, Dimitrina Georgieva2, Slavica Josifovska3, Diana Belejanska4, Shima Mehrabian4, Mariya Petrova4, Latchezar Traykov4, Savina Hadjidekova1, Draga Toncheva5 Aurora Arghir 1, Sorina Mihaela Papuc1, erbescu alina1, Maria Dobre2, Budisteanu Magdalena1;3 Marinella Corbetta 1, Ettore Salsano2, Chiara Pisciotta2, Carla Zanferrari3, Alberto Albanese4, Giorgio Giaccone5, Stefania Magri1, Franco Taroni1 Geir Julius Braathen 1, Kristian Tveten1, Sarka Øygarden2, Elin Dahl2 Nataliia Andreeva 1, Roman Bikanov1 Amal Alhashem 1;2, Inesse Abdallah3, Hamoud alonazi3, Mishal Alsulami3, Hatem Elghezal3 Hajar AlAkeel 1, Mohammed Almannai1, Ahmad Abulaban2 Gokalp Celik1, Fatma Dereli1, Hasan Baş1, Gulay Ceylaner 1, Serdar Ceylaner1 Cecilia Evangelisti1, Sherin Ramadan2, Natalia Fava2, Marco Seri1, emanuele panza 1;2 Silvia Baratta 1, Davide Curti2, Salvatore D’Asero2, Marcello Tognozzi2, Luca Moschini3, Massimo Camerlingo2, Franco Taroni1 Milica Pešić 1, Milena Stevanović2, Nikola Andrejić3, Jovan Pesovic4, Sanja Cirkovic5, Sanja Dimitrijevic6, Danijela Bascarevic6, Natasa Dragasevic Miskovic7, Ivana Novakovic1;7, Dragana Protic6;8 Joana Vilela 1;2, Hugo Martiniano1;2, Ana Rita Marques1;2, João Xavier Santos1;2, Muhammad Asif1;2;3, Célia Rasga1;2, Guiomar Oliveira4;5, Astrid Vicente1;2 Beril Beray Soydemir 1, Ayse Busra Akalin1, Mehmet Ersahin2, Ibrahim Akalin3 Cecilie Pape Madsen 1, Allan Thomas Højland1;2, Thomas Blauenfeldt3 Vira Galagan 1, Nikita Pozhar1, Maryna Tsygankova1, Olga Zhurakhovska1, Valentyna Kurakova1, Viktoriia Serafymovych1 Lucie Sedlackova 1, Markéta Vlčková2, Petra Lassuthova1, Markéta Havlovicová2 Barbara Masotto 1;2;3, Eulàlia Rovira-Moreno1;2;3, Anna Esteve-García4, Ariadna Padró Miquel5, Cristina Sau-Puig4, Cinthia Aguilera5, Nuria Llecha4;5, Laura Trujillano1;2;3, Marta Codina1;2;3, Amaia Lasa-Aranzasti1;2;3, Berta Alemany6, Pedro Alía4, leticia Iranzo Nuez1;2;3, Elena García-Arumí1;2;3, Eduardo Tizzano1;2;3 Chi-Shin Wu1, Chia-Lin Hsu1, Mei-Chen Lin1, Mei-Hsin Su2, Yen-Fen Lin1, Chia-Yen Chen3, Po-Chang Hsiao4, Yi-Jiun Pan2, Pei-Chun Chen1, Yen-Tsung Huang5, Shi-Heng Wang 1 Iryna Lastivka 1, Vita Antsupova2, Ludmila Khlunovska1, Larysa Sheiko3, Ljudmila Brisevac3, Anastasiya Babintseva4, Iana Ushko2 Faezeh Sarayloo 1;2, Dan Spiegelman2, Daniel Rochefort2, Fulya Akcimen1, Rachel De Barros Oliveira2, Patrick Dion2, Guy Rouleau1;2 Karim Yanin 1, Olga Gumeniuk1, Yuriy Chernenkov1, Anna Fisun1, Olga Groznova2 Simge Tuana Ay 1;2, Sena Cetin1;2, Filiz Ozen1, Elif Yilmaz Gulec1;2 Mohnish Suri 1, Matthias De Wachter2, Sarah Weckhuysen3, Michael C. Kruer4, Somayeh Bakhtiari4, Henry Houlden5, Reza Maroofian5 Karin Skarping1, Larissa Larissa2, Åsa Petersén3, Huu Phuc Nguyen2, Samuel Gebre-Medhin 1 Giulia Barcia 1;2, Jean-Madeleine de Sainte Agathe2;3, Lionel Arnaud2;3, Marie Faoucher2;4, Anne-Sophie Lèbre5;6, Nicolas Chatron6;7, Christele Dubourg4, Cyril Mignot8, Laurence Perrin9, Patrick Van Bogaert10, Silvia Napuri4, Anne de Saint Martin11, Marie Therese Abi Warde11, Sarah Baer11, Mathieu MILH12, Nathalie Villeneuve12, Anne Lepine12, Dorothée Ville7, Anne-Lise Poulat7, Matthildi Athina Papathanasiou Terzi7, Eleni Panagiotakaki7, Claire Bar13, Frederic Villega13, Caroline Hachon-LeCamus14, Jennesson Mélanie15, Julien Thevenon16, Henri Margot17, Marine Legendre17, Elise Boucher18, Juliette Piard18, Olivier Patat14, Valentin Ruault19, Marine Lebrun20, Anaïs Lharidon2, Virginie Bernard6, Rima Nabbout21, Laurent Villard22, Gaetan Lesca23 EP12 Neuromuscular Disorders Kumiko Yanagi 1, Hiroshi Matsumoto2, Masahiko Yamamori1, Hajime Wakamatsu3, Tomomi Hidai1, Arisa Igarashi1, Yoichi Matsubara4, Tadashi Kaname1 Anastasia Maksimova 1, Polina Golikova1, Aitalina Sukhomyasova1;2, Nadezhda Maksimova1 Ruchen Zhang 1, Bisei Ohkawara1, Mikako Ito1, Akio Masuda1, Kinji Ohno1 Irene Madrigal 1;2, Andrés Nascimento3, Laia Rodriguez-Revenga1;2, Cristina Jou3, Daniel Natera-de Benito3, Giorgia Sebastiani4, Gemma Arca4 Ann-Kathrin Zaum 1, Natalie Pluta1, Anna Forster1, Astrid Pechmann2, Delia Lorenz3;4, Juliane Lippert1, Yvonne Jelting1, Simone Rost1;5 Dana Safka Brozkova 1, Anna Uhrova Meszarosova1 Parsa Soleimani 1, Aida Ghasemi2, Davood Zare-Abdollahi1, afagh alavi1;2 Maria Giertlova 1;2;3, Petra Drenčáková1, Lenka Nosková4, Jana Šaligová5, Ľudmila Potočňáková5, Simona Drobňáková5, Miriam Kolníková6, Katarína Kušíková6, Patrícia Balážová6, Angela Baranová7, Ladislav Gurčík8, Zuzana Mažeriková9, Martin Mistrík10, Paula Stretavská11, Marie Zikanova4, Viktor Stránecký4, Jakub Sikora4, Tomas Honzik4, Stanislav Kmoch4 Francesca Luisa Conforti 1;1, Martina Pecoraro2, Benedetta Perrone1, Viviana Mosca2, Paola Ruffo1, Vincenzo La Bella2 Maruša Barbo 1, Tanja Blagus1, Vita Dolzan1, Blaž Koritnik2;3, Metka Ravnik-Glavač1 Danyal Nassar 1;1, Khrystyna Kachurak1, Oleksandr Kornutii1, Anastasiia Dutchak1 Woori Jang 1, Jiyeon Kim2, Kyoung-Jin Park3, Joonhong Park4 Benjamin Durand 1, Aurélie Gouronc2, Aleksandra Nadaj Pakleza3;4, Nicolas Dondaine2, Claire Feger2, Maria Cristina Antal5;6, Béatrice Lannes7;8, sophie scheidecker2, Valerie Biancalana2 Hijab Zahra 1, Ghazala Zafar2, Haq Nawaz Khan2, lubaba khalid2, omar mahmud3, Mathias Toft4, Zafar Iqbal5, Ambrin Fatima2 Elena Zinina 1, Vyacheslav Tabakov1, Irina Panchuk1, Inna Sharkova1, Olga Shchagina1, Aleksander Polyakov1 Christina Tyner 1, Sandra Smieszek1, Bart Przychodzen1, Caroline Johnson1, Christos Polymeropoulos1, Gunther Birznieks1, Walker Hagan2, Leticia Lenkiu2, Caitlyn Niccum2, Heather Cannon2, Rocky Brighton2, Xiufang Guo3, Kenneth Hawkins3, Nesar Akanda3, James Hesperos2;3, Mihael Polymeropoulos1 Caroline Johnson 1, Sandra Smieszek1, Bart Przychodzen1, Christina Tyner1, Christos Polymeropoulos1, Gunther Birznieks1, Mihael Polymeropoulos1 Petra Lassuthova 1, Dagmar Grecmalova2, Annette Lischka3, Katja Eggermann3, Ingo Kurth3, Radim Mazanec4 Yasmina Rahmuni 1;2, Youssef El Kadiri1;2, lyahyai jaber1, birouk nazha3, nesnassi mounir3, abdelaziz sefiani1;2, ilham ratbi1;4 Fatemeh Fatehi 1, Zeinab Ghorbanoghli1, Mahdieh Kooshki1, Shima Zamanian Najafabadi1, Khadijeh Noudehi1, Mina Makvand1, Hossein Najmabadi1;2, Ariana Kariminejad1 María Fenollar-Cortés 1, Vicente Gajate-García2, Clara Herrero-Forte1, Raluca Oancea-Ionescu1, Carmen Cotarelo-Pérez1, Laura García-Reguero1, Juan Luis Rodríguez-Hermosa3, alejandro horga2 Selma Demir 1, Sinem YALÇINTEPE1, Veysel Öz2, Yasemin Karal2, Engin Atli1, Drenusha Zhuri1, Hazal Sezginer Guler1, Yelda Yalçın3, HAKAN GURKAN1 Emmanuelle Pion1, Mireille Cossee2;3, Valerie Biancalana4, Cecile Acquaviva5, Céline Bouchet6, Julien Fauré7, Roseline Froissart5, France Leturcq8, Rita Menassa9, Corinne Metay10, Laurence Michel-Calemard9, Emmanuelle Campana-Salort11, Juliette Nectoux8, François Petit12, John Rendu7, Pascale Richard10, Damien Sternberg13, Sandrine Vuillaumier-Barrot6, Shahram Attarian11, Svetlana Gorokhova14, Martin Krahn 14 Mar Costa-Roger 1;2, Ana Maria Cordero1;2, Ivon Cuscó1;2;3, Elena García-Arumí1;2, Eduardo Tizzano1;2 Maria Akhkiamova 1, Olga Shchagina1, Aleksander Polyakov1, Sergey Kutsev1, Sergey Voronin1, Andrey Marakhonov1 Alice Margutti1, Martina Mietto1, Vittoria Nagliati1, Maria Sofia Falzarano1, Gaetano De Feo1, Giusy Cavarretta1, Fernanda Fortunato1, Marcella Neri1, Francesca Gualandi1, Alessandra Ferlini1, Rita Selvatici 1 Lior Greenbaum 1;2, merav ben-david1;2, alex zvulunov1;3;4, amir dori1;2 KADRİYE FARYAP 1, Ayse Busra Akalin2;3, Elif EROĞLU ŞİMŞEK4, Ibrahim Akalin4 Graciela Pi 1, ANGEL ZUNIGA2, MLuisa Bueno-Sanchez1, Joana Ortega Sanz1 Bohdan Tretiak 1, Danuta Zastavna1, Veronika Hamkalo1, Khrystyna Bakum1, Hayane Akopyan1 Negar Molaei 1;2, Fatemeh Ahangari1, Maryam Beheshtian1, Parnian Alagha1;2, shima Dehdahsi1, Mahsa Fadaie1, Mehri Ashki1, Zohreh Elahi1, Maryam Mozaffarpour1, Parishad Saei1, Fatemeh Fatehi1, Khadijeh Noudehi1, Shima Zamanian Najafabadi1, Kimia Kahrizi2, Ariana Kariminejad1, Hossein Najmabadi1;2 Zeinab Ghorbanoghli 1, Fatemeh Ahangari1, Fariba Afroozan1, Mahtab Ramezani2, Hossein Najmabadi1;3, Ariana Kariminejad1 Maruša Barbo1, Tanja Blagus1, Vita Dolzan1, Blaž Koritnik2;3, Metka Ravnik-Glavač 4 Manar Kaptan 1, Beyza Yavuzcan1, Serpil Eraslan2, Şahin Avcı2, İlker Eren3;4, gulshan yunisova3;5, Mehmet Demirhan3;4, Piraye Oflazer3;5, Hülya Kayserili1;2;3;6 EP13 Multiple Malformation/Anomalies Syndromes Bruno Stephan1, Juliana Leao1, Debora Pegos1, Julia Abrahão1, Vitoria do Val1, Matheus Castro1, Rachel Honjo1, Debora Bertola1, Hiromi Aoi2, Saida Ken2, Rie Seyama2, Yuri Uchiyama2, Naomichi MATSUMOTO2, Chong Kim 1 aysenur ersoy 1, Bilgen Bilge Geçkinli1, Zeynep Baser1, onur hanoglu1, cekdar kapazan1 Georgia Christopoulou 1, Stavros Bournazos1, Alexandra Fountoulaki2, maria kassotaki2, Ioannis Goniotakis2, ILIANNA MANIADAKI2, Pantelis Constantoulakis1, Eleftheria Papadopoulou2 Ana-Maria Meašić 1;2;3, Katarina Vulin1;2;3, Mijana Kero4, Leona Morožin Pohovski1;2;3, Lara Samadan1, Adriana Bobinec1;2;3, Sanja Pejić Rosko5, Silvija Pušeljić6, Ljubica Odak1;2;3 Faidon-Nikolaos Tilemis 1, Nikolaos Marinakis1;2, Nikoletta Selenti1, Korina Karachristou3, Konstantina Katechi3, Konstantina Kosma1, Aphrodite Kampouraki1, Jan Traeger-Synodinos1 federica gaudioso 1, Camilla Meossi1, Cristina Gervasini2;3, Valentina Massa2;3, Romano Tenconi4, giulietta scuvera1, Paola Marchisio5;6, camilla lucca7, Maria Iascone7, laura pezzoli7, Donatella Milani5 Vita Antsupova 1, Iryna Lastivka2, Ludmila Khlunovska2, Anastasiya Babintseva3, Ljudmila Brisevac4, Larysa Sheiko4, Iana Ushko1 Alfredo Repáraz-Andrade 1, Cristina Torreira Banzas1, Milagros Blanco Pérez1, Begoña Durán Fernández-Feijoo2, Arturo Fernández Nogueira1 Carmen María Dolores Sánchez1, Daniel Doval Calvo2, María José Sánchez Soler1;3, Ana Teresa Serrano Antón 1;3, María Juliana Ballesta Martínez1;3, Vanesa López González1;3, Marta Domínguez Jiménez1;3, Marta Roldán Montero1, Elena Campillo Antón1, Encarna Guillén-Navarro1;3 Andreja Zagorac 1, Peter Gradišnik2, Faris Mujezinović3, Nadja Kokalj Vokač1 adina fuchs 1;2, Yonatan Rips2, shira Yanovsky-Dagan2, Tamar Harel1;2 Rosanna H.E. Krakowsky 1;1, Tobias Bartolomaeus1, Anne-Christin Teichmann1, Helene Faust1, Vincent Strehlow1, Franziska Schnabel1 Oksana Tyshchenko 1;2, Maria Dushar1, Nataliya Huleyuk1;3, Sofiia Levandivska1, Erika Patskun4, Halyna Makukh1;5 Jung Min Ko 1, A Young Park1, Naye Choi1 Marta Wójcik 1, Magdalena Badura-Stronka1;2, Aleksandra Wnuk-Kłosińska1;2, Dorota Simon3, Aleksandra Jakubczak3, Michał Piechota3, Aleksander Jamsheer1;2 Nataliya Kitsera1, Maya Bondarenko 2 Miruna Gug 1, Diana Miclea2;3, Nicoleta Andreescu4;5, Ioana Mozos6;7, Cristina Gug8 Ange-Line Bruel 1;2, Anil Kumar Ganga3, Lenka Nosková4, Irene Valenzuela Palafoll5, Jelena Martinovic6, Yannis Duffourd1;2, Marie Zikanova4, Filip Majer4, Stanislav Kmoch4, Marketa Mohler7, Jingbo Sun3, Lauren Sweeney3, Núria Martínez-Gil5;8, David Breslow3, Christel Thauvin-Robinet1;2;9 Eriko Nishi 1, Kumiko Yanagi2, Nobuhiko Okamoto1, Tadashi Kaname2 Sameeta Kumari 1, Fizza Akbar2, Seung Woo Ryu3, Arshalooz Rahman4, Salman Kirmani4 Somayyeh Heidargholizadeh 1, Birsen Karaman1, Tugba Kalayci1, Ayça Dilruba aslanger1, Gülnihal Bulut2, gözde tutku turgut1, Behiye Tuğçe yıldırım1, Bilge Ozsait Selcuk1 María de la Luz Arenas-Sordo 1;2, Isabel Barradas-Hernández3, J. Karina Peñuelas-Romero1, Kyrrck A Del Real-Martínez4, JImena Solorio-Fosado5 Sabina Nagieva 1, Oksana Ryzhkova1, Viktoriia Zabnenkova1, Anna Orlova1, Olga Shchagina1, Natalia Taran2, Irina Tin2, Tatyana Strokova2, Natalia Semenova1 Franziska Schnabel 1, Christina Kloetzer2, Helene Faust1, Robin-Tobias Jauss1, Denny Popp1, Anne Sophie Kubasch2, Madlen Jentzsch2, Georg-Nikolaus Franke1, Jens Uhlig3, Klaus Metzeler2, Vladan Vucinic2, Uwe Platzbecker2, Johannes Lemke1;4, Henry Oppermann1 Serap Ketenci İşlek 1, Merve Soğukpınar1, gizem ürel-demir1;2, Pelin Simsek-Kiper1, Gulen Eda Utine1 Dominique Braun 1, Anne Gregor1, Monika Haubitz2, Gabriela M Baerlocher2;3, Claudine Rieubland1, Cornelia Kraus4, Christiane Zweier1 Clemence Vanlerberghe 1, Frédéric Frénois1, Thomas Smol1, Anne-Sophie Jourdain1, Fabienne Escande1, Emilie Ait Yahya2, Abdulrahman Aldeeri3;4, Timothy Yu3, Valérie Cormier-Daire5, Jamal Ghoumid1, Maureen Jacob6, Ruth Newbury-Ecob7, Sylvie Manouvrier-Hanu1, Jessica Platon8, Sebastian Sailer9, Perrine Brunelle1, Lydie Da Costa8;10;11, Florence Petit1 Veronica Seidel 1, Julia Suárez González2, Alejandra Aguado del Hoyo3, Andrea Palacios Bermejo4, Raquel Rodrigo Fernández5, María Molina Bañón4, Ester Sanz López6 Nela Maksimovic 1, Tatjana Damnjanovic1, Biljana Jekic1, Milka Grk1, Marija Dusanovic Pjevic1, Ana Djuranovic1, Milica Rasic1, Parsa Barzegar2, Badr El Hayani1, Dijana Perovic1 Nazlı Büşra Açıkgöz 1, gizem ürel-demir1;2, Pelin Simsek-Kiper1, Gulen Eda Utine1 VASILICA PLAIASU 1, Diana Ozunu1, Gabriela Motei1, Rodica Cretu2, Florin Brezan2, Lucica Ghita2, Salwa Addilami2, Bogdan Pascu2, Monica Panzaru3, Roxana Popescu3, Cristina Rusu3 Theresia Herget 1, Christine Zühlke2, Maja Hempel1;3, Amelie van der Ven1 Elena Sukarova-Angelovska 1, Predrag Noveski2, Dijana Plaseska-Karanfilska2, Sara Veleska3, Slavica Trajkova4, Danilo Nonkulovski5, G. Ilieva3, Liljana Tasavska -Rmus3, Eugene Lee6, Jihye Kim6 Shelly Lev-Hochberg 1, annick REIN ROTHSCHILD1, Ben Pode Shakhed1, Odelia Chorin1 Olivia Kaas Laursen 1, Janni Majgaard Jensen1 Shima Zamanian Najafabadi 1, Zhila Ghaderi1, Zeinab Ghorbanoghli1, Fatemeh Fatehi1, Fatemeh Ahangari1, Hossein Najmabadi1;2, Ariana Kariminejad1 Kameliya Kercheva 1;2, Irena Bradinova1;2, Silvia Andonova1, Radostina Raynova1, Violeta Dimitrova2;3, Alexey Savov1;2 Gulnihal Bulut 1, Bilge Ozsait Selcuk2, Somayyeh Heidargholizadeh1, Tugba Kalayci2, Ayça Dilruba aslanger2, Selen Cerci1, Birsen Karaman3 Hilal Şentürk 1;2, burak eraslan3, sinan akbaş1, Turkan Uygur4, Ayça Dilruba Aslanger1, Volkan Karaman1, Gozde Yesil Sayin1, Guven Toksoy1, Zehra Oya Uyguner1 Inês Beleza 1, Ana Vilan2;3, Sofia Dória3, Ana Grangeia1;3 Zeyneb Berrin KOSEM 1, İlknur Sunar2, Ayse Sena Demircioglu3, Rukiye Akarsu4, İsmail Gencay5, Ibrahim Akalin6 Raquel Rodrigues 1, Catarina Macedo1, Mariana Soeiro e Sá1, José Paulo Monteiro2, Ana Sousa1, Ana Berta Sousa1 Laura van Zutven 1, Jeroen Knijnenburg2, Gerthe Kerkhof3, Eunice Stefanou1, Marielle Alders4, Sharmila Chitoe-Ramawadhdoebe2, Diana Rongen - van Nispen1, Marga Thomassen1, Yvette Van Ierland1, Hennie Brüggenwirth1 Darina Kachakova 1;2, Kunka Kamenarova1;2, Kalina Mihova1;2, Martin Georgiev1;2, Gergana Stancheva1;2, Valentina Peycheva1;2, Ivanka Dimova2;3, Radka Kaneva1;2 Karolis Slibinskas 1, Giedrė Kojelienė1;2, Roma Liutkeviciutė2, Ivan Gavriljev3, Rasa Ugenskienė1, Rasa Traberg1 Anastasios Mitrakos 1;2, Kyriaki Kekou1, Faidon-Nikolaos Tilemis1;2, Maria Svingou1, Georgios Papadimas3, Christalena Sofocleous1, Jan Traeger-Synodinos1, MARIA TZETIS1 chumei li 1, Sarab Mohamed2 Zeynep Esener 1, Nezir Özgün2, Ahmet Yaramis3 Estelle Colin 1;2, Clarisse Battault1;2, Magalie Barth1, Marine Tessarech1;2, Agnes Guichet1, Maud Blanluet1, Florence Biquard3, Françoise Boussion3, Clara Houdayer1;2, Pierre Blanc4, Alban Lermine4, Benoit Delorme5, Vincent Procaccio2;5, Céline Bris1;2 Michael da Silva1, Bruna Burssed1, Marco Antonio Ramos1, Eduardo Perrone1, Maria Isabel Melaragno 1 Deepthi De Silva 1, Nuwani Wijesooriya2, Romesh Gunasekera3, sriyani Basnayake4, Malka Jayathilake5 Mehmet Akif Yücesoy 1, sinan akbaş1, Gülhanım Memiş1, Esma Nur Konur1, Durmuş Durmaz1, Volkan Karaman1, Leyli Şentürk1;2, Umut Altunoğlu1;3, Esma Şengenç4, Hülya Maraş Genç5, Edibe Pempegül Yıldız5, Adnan Deniz6, Bülent Kara6, Ayça Dilruba aslanger1, Bilge Ozsait Selcuk1, Gozde Yesil Sayin1, Zehra Oya Uyguner1, Birsen Karaman1;7 Arezou Karamzade 1, Meisam Babaei2, Neda Golchin3, Saeed Talebi4, Aysun Khalil Nejad Sani Banaei3, Mohammad Keramatipour3;5 Júlia Martinková 1, Lukáš Ryba1, Emílie Vyhnálková1, Miroslava Balaščaková1, Martin Schwarz1;2, Romana Borská3, Lenka Fajkusova3, Anna Křepelová1 Ina Ofelia Focsa1;2, Andreea Tutulan-Cunita 2, Anca Pavel2, Diana Prepelita2, VASILICA PLAIASU3, Liana Ples1;4, Anca Neacsu2, Maria Stratan2, Daniela Blanita5, Natalia Usurelu5, Danai Stambouli2 Sónia Custódio 1, Raquel Gouveia Silva1, Oana Moldovan1, Eva Rolo1, Ana Sousa1, Ana Berta Sousa1 Dilek Uludağ Alkaya 1, Nilay Gunes1, Buşra Kasap1, Timur Yıldırım2, Aysegul Bursali2, Evren Akpınar2, Mehmet Müfit Orak3, Mehmet Bülent Balioğlu2, SEHIME TEMEL4, Beyhan Tüysüz1 Henrique Serigatto 1, Nancy Kokitsu-Nakata1, Siulan Vendramini-Pittoli1, Tania Kamiya2, Ana Krepischi3, Carla Rosenberg3, Roseli Zechi-Ceide1 Katarzyna Wojciechowska 1, Whitley Zie2, Anna Kutkowska-Kaźmierczak3, Małgorzata Rydzanicz4, Rafał Płoski4, Monika Lejman1 Hilal Onur 1, Nilay Güneş1, Dilek Uludağ Alkaya1, Ali Metin Kafadar2, Beyhan Tüysüz1 Amal Abd Mouleh 1;2, Syrine Hizem1;2, Yasmina Elaribi1, Houweyda Jilani1, Imen REJEB1;3, Ons Azzabi3;4, Sana Karoui1, Lamia BenJemaa1;3 Tim Bender 1, Kirsten Cremer1, Hartmut Engels1, Claudia Perne1, Isabelle Claus1, Jessica Trautmann1, Axel Schmidt1, Stefanie Heilmann-Heimbach2, Pietro Incardona3, Andreas Müller4, Hela Hundertmark1, Sophia Peters1 Jelena Jovanovic 1, Gordana Stojanovic1, Olivera Miljanovic1;2;3, Sladjana Teofilov1, Luca Lovrecic4, Alenka Hodžić4, Borut Peterlin4 Davide Calosci 1, Lisa Passaglia1, Annapaola Calò2, Chiara Rigon1;3, Dario Seif Alì4, Cinzia Bertolin2, Eva Trevisson1;2;3 Gokcen Karamik 1, sezin yakut uzuner2, Nuray Ozturk1, Öznur YILMAZ BAYER1, guldeniz oner1, banu nur1, ercan mihci1 Anna Hammarsjö 1;2, Karin Sollander1;2, Sofia Frisk1;2 Cherif Mortadha 1, Yasmina Elaribi1, Syrine Hizem1;2, Houweyda Jilani1, Sana Karoui1, yosra ben rejeb3, Sami Jebnoun4, Imen REJEB1;5, Lamia Ben Jamaa1;5 Ekin Cemre Bayram Tokaç 1, Filiz Mine cizmecioglu Jones2, Esra Koçyiğit2, Aslı Ece Solmaz1 Anna Pecková 1, Jana Lastuvkova1, Vlasta Cejnova1, Anna Uhrova Meszarosova2 Claudio Catalli 1, Isabel Llano Rivas1, Hiart Maortua Olabe1, Intza Garin Elkoro1, Blanca Gener Querol1 Natalia Braun-Walicka 1, Agnieszka Pluta2;3, Tomasz Wolak2, Edyta Maj2;4, Agnieszka Maryniak3, Monika Gos1, Anna Abramowicz1, Aleksandra Landowska1, Ewa Obersztyn1 Aleksandra Pietrzyk 1, Sabina Lichołai2, Patrycja Szymańska3, Damian Loska3, Agnieszka Sobczyńska-Tomaszewska3 Laimutis Kucinskas 1, Rasa Traberg1, Marius Sukys1, Inga Nasvytienė1, Rasa Ugenskienė1 Karolis Baronas 1, Aušra Matulevičienė1, Beata Aleksiūnienė1, Kamilė Šiaurytė1, Algirdas Utkus1 Anca Mitroi 1;2, Georgeta Cozaru1;2, Mariana Aschie1;2, Costel Brinzan1;2 Marta Souto1, Márcia Martins 2, Andreia Dias3, Ana Matos1, Osvaldo Moutinho4, Rosário Leite1 Anila Babameto-Laku 1;1, Aida Bushati2, Arlind Myftari3 Daniela Avdjieva-Tzavella 1, Tsvetina Veleva2, Trayan Delchev1, Maria Sredkova-Ruskova1 Olga Ivanova 1, Polina Tsygankova1, Ekaterina Zakharova1 Tsvetina Veleva 1, Daniela Avdjieva-Tzavella1, Trayan Delchev1, Maria Sredkova-Ruskova1 Salih Türk 1, Cengiz Yalçınkaya2, Buşra Kasap1, Filiz Geyik3, Dilek Uludağ Alkaya1, Nilay Güneş1, Esma Şengenç4, Sema Saltık5, Beyhan Tüysüz1 Gözde Tutku Turgut 1, Umut Altunoglu1;2, Tuğba Kalaycı1, Sahin Avci1;2, Ayça Dilruba Aslanger1, Volkan Karaman1, Zehra Oya Uyguner1, Hülya Kayserili1;2 Filiz Ozen 1, Arzu Guliyeva1;2 YoungBae Sohn 1, Yoong-a Suh2, Chang Ahn Seol3, Do-Wan Kim4 Rasa Traberg 1, Rimvydas Jonikas1, Jūratė Laurynaitienė2, Rasa Ugenskiene1 Wiem Essalah 1, Ahlem Achour1;2, Asma Tajouri2, Madiha Trabelsi1;2, Faouzi Maazoul1, lilia kraoua1;2, Imen Chelly2;3, Rym Meddeb1;2, Neila Belghith1, Sana Gabteni1;2, ridha m’rad1;2, Ines Ouertani1;2 Adriana Bobinec 1;2;3, Ivona Sansović1;2;3, Ana-Maria Meašić1;2;3, Katarina Vulin1;2;3, Leona Morožin Pohovski1;2;3, Ljubica Odak1;2;3 Beyza Yavuzcan1, Manar Kaptan1, Mert Kaya 1, Cigdem Arikan2, Umut Altunoglu3 EP14 Cytogenetics, Genome Variation and Architecture Darya Yurchenko 1, Zhanna Markova1, Marina Minzhenkova1, Nadezda Shilova1 Usha Dutta 1, Divya Bhanu1, Ashwin Dalal1 Clairo Lin Hui Ng 1, Mui Li Tan1, Lee Tian Toh1, Hui Yi Yon1, Min Hwee YONG1, Angeline Hwei Meeng Lai2 Ivana Tonkovic Djurisevic 1, Sanda Huljev Frkovic2, Morana Miklos1, Anita Pokupec Bilic1 Anita Pokupec Bilic 1, Morana Miklos1, Ivana Tonkovic Djurisevic1, Ivan Bilic2, Katja Dumic Kubat3 Diksha Jethnani1, Joanna Lubieniecka 1;2, Krzysztof P Lubieniecki2, Kilian Audoire2, Maria Yusenko2, Charlotte Hippert2, Kristina Döring2, Nikolina Jovancevic2, Sabine Hoffjan2, Patrick Weydt3, Jinko Graham1, Huu Phuc Nguyen2 marion lesieur-sebellin 1;2, Kristen Wigby3;4, Elise Schaefer5, Aurélie Gouronc6, Nicolas Chatron7;8, Anne-Lise Poulat9, Audrey Putoux7;10, Alice Goldenberg11;12, Mathilde Quibeuf11;13, Pascal Chambon11;12, Sophie Rondeau1, Giulia Barcia1;2, Jonathan Levy14, Juliette Piard15;16, Paul Kuentz17;18, Martine Doco19;20, Nathalie Bednarek19, Roseline Caumes21, Sonia Bouquillon22, Cedric Le Caignec23;24, Olivier Patat25, Philippe Khau Van Kien26, Jean Chiesa27, Geoffroy Delplancq28, Séverine Bacrot28, Sophie Brisset28, Vincent Cantagrel2, Jerica Lenberg4, Jennifer Friedman4;29, Marlene Rio1;2, sophie scheidecker6, VALÉRIE MALAN1;2 Thomas Liehr 1 Zhuo Qu 1, narumi sakaguchi1, Chie Kikutake1, Mikita Suyama1 Emine Atlı 1, Engin Atli1, Yasemin Ozen1, Sinem YALÇINTEPE1, HAKAN GURKAN1 Nadezda Shilova 1, Marina Minzhenkova1, Zhanna Markova1, Natalya Kuzina1, Nina Demina2, Artem Borovikov3 Zhanna Markova 1, Darya Yurchenko1, Artem Borovikov2, Nadezda Shilova1 Sara Veleska 1;1, Elena Sukarova1, Slavica Trajkova2, Learta Alili-Ademi3, Liljana Tasavska -Rmus1, Gordana Ilieva1 Sílvia Pires 1;2, Paula Jorge1;2, Thomas Liehr3, Natália Oliva-Teles1;2;4;5 Georgia Christopoulou1, Alexandra Fountoulaki2, maria kassotaki2, ioanna lygerou2, maria michailou2, Ioannis Goniotakis2, rozanna poulaki2, ILIANNA MANIADAKI2, maria bitsori2, PELAGIA VORGIA3, chrissoula perdikogianni2, emmanouil paraskakis2, ioannis germanakis2, Pantelis Constantoulakis1, Eleftheria Papadopoulou 2 Laura Formentini 1, Giulia Olivucci1;2, Anna Basile3, Mariia Zadorozhna3;4, Paola Dimartino3, Elisa Giorgio3;4, Malte Spielmann5;6;7, Tommaso Pippucci1 Caterina Marconi 1, Lina Quteineh1, Sebastian Suchet1, Frédéric Masclaux1, Jean-Louis Blouin1;2, Joël Fluss3, Marc Abramowicz1;2, Michel Guipponi1;2 Hiba Abu Khalil 1, Mohammed Albalwi2;3, Norah Alsaleh1;4 Omamah AlShehri 1, Abdulelah Abunadi2, Ashwaq Alghamdi2, Aziza AlKhalidi2, Bader AlMuzaini2;3, Mohammed Al Balwi2;3;4 Martine Doco-Fenzy 1;2, Olivier Pichon1, Faezeh Vasheghani1, Emilie Landais3, Tony Yammine2, Sylvie Bernard4, Poirsier Céline2, nathalie Le Du5, vincent jauffret4, Noemie Celton5, Kamran Moradkhani6, nicolas gruchy7, Agnes Guichet8, Sylvie Jaillard9, Sylvie Odent9, Marie Vincent1, Mathilde Nizon1, Bertrand Isidor1, Sandra Mercier1, celine bonnet10, Marc-Antoine Belaud-Rotureau11, Clemence Jacquin3, Nathalie Bednarek12, Jonathan Levy13, Laetitia Lambert10, Mylene Dexheimer14, stéphane bezieau1 Jakub Vavra1, Johana Alaverdyan 1, Artur Sergunin1, Daniela Ekendahl1 Aysel Kalayci 1, Deniz Agirbasli2, Nihal Serdengeçti3, Mustafa Tarık Alay2, Mahmut Cem Tarakçıoğlu3, Mehmet Seven2 Ingrid Bader 1, Kathrin Buder2, Stefanie Beck-Wödl1, Marc Sturm1, Petra Stöbe1, Olaf Riess1, Tobias Haack1 Gülhanım Memiş 1, hatice arslan1, sinan akbaş1, Ayça Dilruba aslanger1, Gozde Yesil Sayin1, sevgi yavuz2, aslı al3, bağdagül aksu4, zeynep yıldırım4, alev yılmaz4, firdevs baş3, Bilge Ozsait Selcuk1, Birsen Karaman1;5 Mohammed Al Balwi 1;2;3, Roaa Alahmadi4, Nedaa Aldossary4, Khalid Alshalan4, Albandri Alzaid4, Mohammed Siraj4, Mshari alhwiti4, Nabilah Alghamdi4, Razan Almudhhi1, Aziza AlKhalidi1, Malak Alghamdi5 Janne Sofie Wehnert 1, Yorck Hellenbroich2, Malte Spielmann2, Inga Nagel1 Azizah AlKhaldi 1, Mohammed Adam1, Alaa Almeleihi1, Salman Alsaad1, Areej AlAtawi2, Mohammed Al Balwi1;3;4 Kristina Gornik Crkvenac 1, Ana Juras1, Nevena Krnic2, Sanda Huljev Frkovic3 Irena Bradinova 1;2, Silvia Andonova2, Radoslava Vazharova3, Ralitsa Robeva4;5, Alexey Savov1;2 Aline Aywaz 1, Seda Hakobyan1, Susanna Midyan1, Tatevik Hovhannisyan1, Arpine Mkrtchyan1, Natella Kostandyan2 Derya Kan Karaer1, Kadri Karaer 1 Belmina Šarić Medić 1, Naida Lojo-Kadrić1, Jasmin Ramić1, Nikolina Tomić1, Lejla Pojskić1 Nuša Trošt 1, Luca Lovrecic1, Borut Peterlin1 Detlef Trost 1, Aline Receveur1, Laurence Lohmann1, Mylene Valduga1, Pascale Kleinfinger1, Aïcha Boughalem1 Eunice Matoso 1;2;3, Alexandra Estevinho2, Susana Isabel Ferreira4, Sara Ribeiro5, Daniela Couto6, Joana Barbosa de Melo3;4;7, Isabel Marques Carreira3;4;7, Jorge Saraiva5;8;9 Yury Barbitoff 1, Nadezhda Pavlova1, Polina Bogaichuk1 Tatyana Lazareva 1, Yuriy Barbitoff1, Yulia Nasykhova1, Andrey Glotov1 Valentyna Kurakova 1, Vira Galagan2, Olena Baronova1, Olga Miakushko1, Diana Mykhailova1, Maryna Tsygankova2, Olga Zhurakhovska2 panayiotis myrianthopoulos1, andria ketoni1, charithea ioannidou1, Liza Kokkinou1, efthymia constantinou1, elena panayiotou1, nicole salameh1, ioannis papaevripidou1, angelos alexandrou1, paola evangelidou1, carolina sismani 1 Julia Flunkert 1, Rainer Wimmer1, Markus Stumm1 Roberta Petillo1, Massimiliano Chetta2, Marina Tarsitano2, Serena Torre2, Elvira Sannino2, Marcella D’Antonio2, Maria Oro2, Maria Rivieccio2, Maria Cioce2, MariaRita Parisi2, Davide De Brasi3, Daniele De Brasi4, Manuela Priolo 2 Idit Maya 1, Michal Levy1, Lina Basel-Salmon1, Lena Sagi-Dain2 EP15 Diagnostic Improvements and Quality Control Antoine Stuitje 1, Zewdu Terefework2, Chris Hettinga1, Robert C. Akkers3, Rob Castel3 Charles Deng 1, Ryan Thompson1, Laura Sloofman1, Alexander Charney1, Noam Beckmann1 Mira Savvina 1, Nadezhda Maksimova1, Aitalina Sukhomyasova1 Céline Angin1, Monica Mazzucato2, Stefanie Weber3, Kurt Kirch3, Houda Ali4, Sylvie Maiella 4, ANA RATH4 Árpád Ferenc Kovács 1, Nóra Fekete2, Dalma Ruzsics1, Luca Kamilla Li1, Sándor Dávid Kovács1, Júlia Krisztina Erhardt1, Gergely Tibor Kozma3, Éva Pállinger2, Gyorgy Fekete1 gong meihua1, yi heng 1, rao junhua1, Xu chongjun1, Deng Ziqing1, Li Jiguang1, Wang Jingjing1, Ma Kexin1, Wang yicong1, Chen Fang1 Theresa Sujatta 1, Denny Popp1, Henry Oppermann1, Julia Hentschel1, Helene Faust1 Eva Graf 1, Whitney Pike2 Anna Shestak 1, Victoria Rumyantseva1, Elena Zaklyazminskaya1 Alvaro Arthur Soares1, Franklyn Emanuell Gomes dos Santos1, Josiel Santos do Nascimento1, Antonio Thomas da Silva1, Pollyanna Almeida dos Santos Abu Hana 2, Carlos Arthur Cardoso Almeida1 Nadav Agam 1;2, ruth schreiber2;3, Ohad Wormser1;2, ofek freund1;2, Matan M. Jean1;2, Amit Safran1;2, Tomer Poleg1;2, Ohad Shmuel Birk1;2;4 Jesper Eisfeldt 1;2, Kristine Bilgrav Saether1, Esmee ten Berk de Boer1;2, Marlene Ek1;2, Anna Lindstrand1;2 benedicta forson1, Melissa Berenger1, Krystyna Nahlik 1, Dana Ruminski Lowe1, Yves Konigshofer1, Dianren Xia1, Edward Davis1, Catherine Huang1 Frances Pitt1, Suhayl Mulla1, Denise Grovewood 2, Russell Buckley-Taylor2, Suki Lee3, Giorgio Fedele4, Graham Hill5, Paul Harper6, Donald Fraser7, Tamsin Redgwell2 Martin Ritthaler 1, Christine Froehlich1, Florian Battke2, Günter Jäger2, Heinz Gabriel1, Björn Schulte1, Martin Schulze1, Saskia Biskup1 Susan Louw 1, Ruth Cleaver2, Helen Tomkins3 Valérie FAUGERE 1, Christel Vache1, David BAUX1, Luke Mansard1, Charles VAN GOETHEM1, Claire-Marie Dhaenens2, Olivier Grunewald2, Isabelle Audo3, Christina Zeitz3, Isabelanne Meunier4, Beatrice Bocquet4, Mireille Cossee1, Anne BERGOUGNOUX1, Vasiliki Kalatzis5, Anne-Françoise Roux1 Graziella Camilleri 1;2, Rachelle Muscat2, Karen Muscat2, Jean Calleja Agius1, Edith Said1;2 Aleksey Antonenko 1, Ruslan Belov1, Maria Revkova1, Ekaterina Domoratskaya1, Alena Bartsis1, Anna Panferova1, Milana Gubona1, Polina Ulanova1, Maria Makarova1, Anastasia Krinitsina1, Maxim Belenikin1 Maria Revkova 1, Ekaterina Domoratskaya1, Dmitrii Markov1, Polina Ulanova1, Olga Chesnokova1, Natalia Sokolova1, Aleksey Antonenko1, Anna Panferova1, Natalia Doroshchuk1, Anastasia Krinitsina1, Maxim Belenikin1 Gregorio Nolasco 1, Monica Roldan2, Florencia Epifani1, Roser Urreizti3, LLuís Armengol4, Daniel Natera-de Benito5, Andrés Nascimento5, anna codina6, Maria Lluïsa Ramírez7, Cristina Hernando-Davalillo6, Carlos Ortez5, Loreto Martorell Sampol6, Mercedes Serrano8 Marian Baldovic 1;2, Gabriela Bľandová1;3, Gabriela Krasnanska1, Lenka Wachsmannova1, Vladimír Eliáš1, Vladimír Ferák1, Michal Konecny1;4 Jacob Day 1, Allison Newman1, Claire Salter1, Fathiya Al-Murshedi2, Almundher Al-Maawali2, Amna Mohammed Al-Futaisi2, Fowzan Alkuraya3, Lama Al-Abdi4, Nishanka Ubeyratna1, Joseph Leslie1, Richard Caswell5, Emma Baple1;6, Andrew Crosby1 Jerneja Debeljak 1, Nina Rupar1, Julij Šelb1;2, Peter Korošec1;3, Matija Rijavec1;4 Weihua Zhao1, Yanyan Song 2, Chuanfeng Huang3, Shan Xu2, Qi Luo1, Runsi Yao1, Nan Sun3, Bo Liang4;5, Jia Fei6, Fangfang Gao7, Jie Huang3, Shoufang Qu3 EP16 Bioinformatics, Machine Learning and Statistical Methods Noriko Tanaka 1;2, Yuki Onishi3, Maria Yamasaki4, Masako Kifushi3 Jaroslav Budiš 1;2;3, Tomáš Sládeček1;4, Michaela Gažiová1;5, Marcel Kucharik1;4, Zuzana Pös1;2;6, Ondrej Pös1;4, Zuzana Hanzlikova1;7, Jakub Styk1;2, Monika Kubanova4, Diana Rusnakova1;4, Jozef Martis1;4, Juraj Gazdarica1;3;4, Jozef Sitarcik1;4, Werner Krampl1;2;4, Ján Radvánszky1;2;6, Tomas Szemes1;2;4 María Aguirre1, Lina Moreno-Giraldo 1, armicson felipe solano1 Mert Polat 1, Zerrin Çelik1, Yunus Kasım Terzi1 Xavier Le Guillou Horn 1;2, Carole Guillevin2;3, Pierre Fayolle3, Clement Giraud2;3, Mathieu Naudin2;3, François Lecellier4, Christine Fernandez4, Remy Guillevin2;3 Vicente Yépez 1, Anna Esteve-Codina2, Julien Gagneur1 Maroua Chahdil 1;1, Carolina Fabrizzi1, Caterina Lucano1, Leslie Matalonga2, Charlotte Rodwell1, Marc Hanauer1, Laurent Tichit3, David Lagorce1, ANA RATH1 LiChao Huang 1, meng yang1, JiHong Wu2 Beatriz Sanz-Milián 1, Albert Ferran Martin1, Luisa Aparicio2, Aroa Barba2, Rebeca Blanco2, Laura Llaó2, Lídia Parra2, Rosella Soler2, Alba Flores Bonet3, Irina Royo3 Beatriz Ruz 1;2;3, Lucía de Dios1, Carmen Rodríguez Jiménez1;4, Victoria Eugenia Fernandez Montaño1, Maria Victoria Del Pozo-Gomez1, María Esther Rubio Martín1, Cristina Ortega-Patrón1, Elisa Izquierdo1;2;3, Clara Venegas1;2;3, Sonia Rodriguez Novoa1;5, adela escudero1;2;3, Antonio Pérez3;6;7, Carlos Rodríguez-Antolín1;8 Yeliz Ekici 1, Selcuk Candan2;3, Ikbal Billur Canbakan3, Feyza Tuncer4 Xiaofeng Liao 1, Yuliia Orlova2, Cenna Doornbos1, Anna Niehues3, Casper de Visser1, Junda Huang1, Thomas Ederveen1, Purva Kulkarni1, Joeri Van Der Velde4, Morris Swertz4, Martin Brandt2, Alain van Gool1, Peter-Bram t Hoen1 Albert Ferran Martin1, Anna Subirats2, Joana Fortuño2, Beatriz Rey-Fernández 2, Rebeca Blanco2, Jaume Torrents3, Albert Torrents3, Cristina Camprubí2, Manel Flores2, Irina Royo3 Tingyu Yang 1;2, Wen-Jing Wang2, Yulong Qin1;2, shuo yan1;2, Jinghua Sun2, Qing Zhou2 Rimvydas Jonikas 1, Marius Šukys1;2, Zivile Zemeckiene1;2, Rasa Ugenskiene1;2 T. madhusankha Alawathurage1, Kerstin Ludwig1, Axel Schmidt 1;2 Anton Changalidis 1, Yulia Nasykhova1, Andrey Glotov1, Yury Barbitoff1 Ashwini Shanmugam 1;2;3, Sophia Heneghan1;2;3, Kane Collins1;2;3, Elhussein Elhassan4;5, Omri Teltsh1;3, Sahin Sarihan1;6, Russell L. McLaughlin2;3;7, Ross Byrne7, Gianpiero Cavalleri1;2;3, Peter Conlon4;6, Katherine Benson1;3, Edmund Gilbert1;3 Aikaterini Lymperidou1;2, Judith Martinez-Gonzalez2;3, Guillem Bracons Cucó2, Xavier Escribà-Montagut4, Marta Huertas2;5, Ramon Mateo-Navarro2;4, David Sarrat-González4, Santiago Frid6, Juan Ramón González4, Alberto Labarga3, Susana Iraola-Guzmán 1, Rafael de Cid1 Eva Caceres1;2, Annick Renevey 1, Anders Jemt1;2, Yingbo Lin3, Ida Lindegaard1, Linnea La Fleur1, Susanne Månér1, Felix Haglund3;4, Henrik Stranneheim1;2;5, Anna Lyander1;6, Valtteri Wirta1;2;6 Adam Frankish1, Jane Loveland1, Shashikant Pujar2, Alex Astashyn2, Ruth Bennett1, Andrew Berry1, Eric Cox2, Claire Davidson1, Olga Ermolaeva2, Catherine Farrell2, Reham Fatima1, Tamara Goldfarb2, Jose M Gonzalez1, Diana Haddad2, Matt Hardy1, Toby Hunt 1, John Jackson2, Vinita Joardar2, Michael Kay1, Vamsi Kodali2, Kelly McGarvey2, Jonathan Mudge1, Michael Murphy2, Sanjida Rangwala2, Francoise Thibaud-Nissen2, Anjana Vatsan2, Craig Wallin2, David Webb2, Terence Murphy2 Mikhail Arbatskiy1;2, Veronika Sysoeva2, Polina Klimovich2, Kamilla Yamileva3, Ekaterina Semina3, Kseniya Rubina 3 Catalin-Vasile Munteanu 1;2, Adela Chirita-Emandi1;2, Maria Puiu1;2, Catalin Valer Marian2;3, Adrian Trifa2;3 Mariateresa Zanobio 1, Antonella Farina1, Claudia Santoro2, Marina Melone3, Silverio Perrotta2, Marco Benedetto4, Anna Biondi4, Stefano Tagliaferri4, Annalaura Torella1;5, Vincenzo Vincenzo Nigro1;5, Giulio Piluso1 Mikhail Arbatskiy 1, Veronika Sysoeva1, Ekaterina Semina1, Kseniya Rubina1 Alexander Schulz1, Dominik Seelow2, Denise Dawid1, Tanja Mederer1, Ralph Röth1;3, Patrick Günther3;4, Nagarajan Paramasivam3;5, Daniel Hübschmann3;5;6, Beate Niesler1;3, Gudrun Rappold1, Philipp Romero3;4, Stefanie Schmitteckert 1;3 Marta Ferreira 1;2;3, Celina São José1, francisco almeida1;3, joaquin jurado maqueda3, ana rita monteiro4, Pedro Ferreira2;5, carla oliveira1;3;6 Iurii Sadovnychenko 1, Nataliia Pastukhova2, Sofiia Rozumenko1 Mohammad Deen Hayatu 1, Rotraut Mößner2, Nina Magnolo3, Sandra Philipp4, Jörg C. Prinz5, Knut Schaekel6, Arif B Ekici1, Steffen Uebe1, Wiebke Sondermann7, Michael Sticherling8, Sascha Gerdes9, Dagmar Wilsmann-Theis10, Ulrike Hüffmeier1 Martin Georgiev 1;2, Kunka Kamenarova1;2, Darina Kachakova-Yordanova1;2, Neviana Ivanova1;2, Olga Beltcheva1;2, Kalina Mihova1;2, Valentina Peycheva1;2, Radka Kaneva1;2 Rahel Feleke 1, Bethany Quinton2, winston lau2, Toby Andrew1, nikolas maniatis2 Bianka Böllering 1 Arzu Zeynep Karabay 1, Asli Koc1, Yalda Hekmatshoar2 Radostina Valeva 1;2, Mariya Levkova1;2, Mari Hachmeriyan1;2, Milena Stoyanova1;2, Dinnar Yahya1;2, Lyudmila Angelova2 Francesco Ranucci 1, Daria Marzanati1, Elisa Giacomini2, Luca Pagliardini2, Valeria Stella Vanni3, Nicoletta Barberis3, Enrico Papaleo3, Massimo Candiani3, Maria Giulia Scotti4, Dejan Lazarevic4, Giovanni Tonon4, Davide Dragani5, Laura Privitera3, Patrizia Rovere Querini6, Davide Gentilini1, Paola Viganò7 Alexander Seitz 1, Anica Hoppe1, Mara Burkert1, Christoph Schmidt1 Rene Mulder 1, Jan Jongbloed1, Kristin Abbott1, B. Sikkema-Raddatz1, Joeri Van Der Velde1, Helga Westers1 Eylül Aydın 1;2, Berk Ergun3, Özlem Akgün Doğan2;4;5, Yasemin Alanay2;4;5, Ozden Hatırnaz Ng2;5;6, Özkan Özdemir2;6 Anna Soplenkova 1, Denis Maslov1, Anna Timoshchuk1, Tim Spector2, Michel Georges3, Sodbo Sharapov1, Gordan Lauc4, Yury Aulchenko1;5 Alexey Kamelin 1;2, Vladislav Perelygin1, Layal Shaheen2, Anna Kim2, Nicolay Plotnikov2, Alexander Rakitko2, Maria Poptsova1 Sergi Marí Alemany 1;2, Alba Hernangomez-Laderas1;2, Irati Miguel1, Ariadna Cilleros-Portet1;2, Bárbara P. González-García1;2, Iraia García-Santisteban1;2, Nora Fernandez-Jimenez1;2, Jose Ramon Bilbao1;2;3 EP17 Large Scale Association Studies (GWAS) Askar Akhmetshin 1, Alexandra Karunas1;2;3, Olga Savelieva1;2, Elza Khusnutdinova1;2 Niklas Worm Andersson 1;2, xiaoping wu1;2, Frank Geller1;2, Jan Wohlfahrt3, Mads Melbye3;4;5, Anders Peter Hviid1;6, Jonas Ghouse7;8, Ole Pedersen9;10, Erik Sørensen2, Sisse Rye Ostrowski2;9, Henning Bundgaard9;11, Marie Lund1;9;12, Bjarke Feenstra1;2;13 Mariia Proskura 1, Anastasiia Buianova2, Valery Cheranev2, Vera Belova2, Anna Shmitko2, Anna Pavlova2, Iuliia Vasiliadis2, Oleg Suchalko2, Denis Rebrikov2, Edita Petrosyan1, Dmitriy Korostin2 Chloé Pittman 1, Sofie Bliddal2;3, Ulla Feldt-Rasmussen2;4, Tugce Karaderi1 Taras Oleksyk 1;2, Khrystyna Shchubelka1;2, Walter Wolfsberger2, Olga Oleksyk1;3 Mikaela Stancheva 1, Lubomir Balabanski2, Dimitar Serbezov2, Dragomira Nikolova2, Radoslava Vazharova2, Draga Toncheva2, Sena Karachanak-Yankova1;2 Mark Seielstad 1 Sacha Bors 1, Sven Bergmann2 T. madhusankha Alawathurage 1, Ayda Abolhassani2, Axel Schmidt1, Laura Kilarski2, Thomas Illig3, Verena Kopfnagel3, Kerstin Ludwig1, Eva C. Schulte1;2 Nadezhda Belonogova 1, Elizaveta Elgaeva1;2, Irina Zorkoltseva1, Anatoly Kirichenko1, Gulnara Svishcheva1;3, Maxim Freidin4, Frances Williams5, Pradeep Suri6;7;8;9, Tatiana Axenovich1, Yakov Tsepilov1 Sodbo Sharapov1, Anna Timoshchuk 1, Olga Zaytseva2, Denis Maslov1, Anna Soplenkova1, Elizaveta Elgaeva3;4, Frano Vuckovic2, Irena Trbojević-Akmačić2, Frances Williams5, Dragan Primorac6, Jan van Zundert7;8, Michel Georges9, Karsten Suhre10, Massimo Allegri11, Nish Chaturvedi12, Malcolm Dunlop13, Matthias Bernd Schulze14;15;16, Tim Spector5, Yakov Tsepilov3;17, Gordan Lauc2, Yury Aulchenko1;3 EP18 Genetic Epidemiology and Pharmacogenomics Ivan B. Filippenkov 1, Natalia Yu. Glazova1;2, Elena A. Sebentsova1;2, Ivan V. Mozgovoy1, Vasily V. Stavchansky1, Nikolay F. Myasoedov1, Natalia G. Levitskaya1;2, Svetlana A. Limborska1, Lyudmila V. Dergunova1 Elizabeta Krstevska Bozhinovkj 1, Predrag Noveski2, Marija Staninova Stojovska1, Emilija Gjorgievska1, Angela Joskovska1, Nenad Mitreski3, Biljana Grozdanovska3, Aleksandar Dimovski1;2, Nadica Matevska-Geshkovska1 Svetlana A. Limborska 1, Ivan B. Filippenkov1, Yana Y. Shpetko1, Alina E. Denisova2, Vasily V. Stavchansky1, Leonid V. Gubsky2;3, Nikolay F. Myasoedov1, Lyudmila V. Dergunova1 Joan Fibla 1, Marina Laplana1 Ming-Wei Lin 1;2, Yu-Ting Lin1, Yung-Feng Lin3, Hui-Ying Weng2, Chien-Hsiun Chen4, Shih-Feng Tsai2;3;5 Hyojin Pyun 1, Soo Ji Lee1;2, Joohon Sung1;2 Žiga Doljak 1, Tanja Blagus1, Katja Goricar1, Miha Arnol2, Gregor Mlinšek2, Vita Dolzan1 Maxim Freidin 1, Nikolai Czajkowski2, Siri Solbakken2, Christopher Sivert Nielsen2 Luisa Foco 1, Eva König1, Reinhard Würzner2, Florian Kronenberg3, Peter Pramstaller1, Christian Fuchsberger1, Cristian Pattaro1, Fabiola Del Greco M.1 Chenxu Zhao 1, Elnaz Naderi1, Tesfa Dejenie Habtewold1, Richard Bruggeman2, Behrooz Z. Alizadeh1 Leon Ruiter-Lopez 1 Tetiana Harashchenko 1, Dmytro Krasnienkov2, Oleksandra Gorodna3, Anastasiia Mohylevets4, Tetiana Kaminska5, Vasiliy Podolskiy1, Tetiana Umanets1, Yuriy Antipkin1, Ludmila Livshits3 Silvia Carlés González1, Ana Fernandez-Montes2, Jose Antonio Trillo-Franco3, Paulina Piario4;5, Lorena Dieguez4;5, Roberto Piñeiro3, Ceres Fernandez-Rozadilla 1 Leonard Frach 1, Jean-Baptiste Pingault1 Hristo Ivanov 1;2, Aleksandar Linev1;2, Nelly Miteva-Marcheva1, Dimitar Dimitrov1, Elena Bangieva1, Antonia Vardeva1, Vili Stoyanova1;2 Natalia Gorovenko 1, Liliia Fishchuk2, Valeriy Pokylko3, Yuliia Cherniavska3, Liudmyla Zhuk3, Viktoriia Vershyhora4, Olena Popova4, Svitlana Podolska1, Olena Ievseienkova1, Oleksandr Buriak2, Volodymyr Olkhovych2, Zoia Rossokha4 Larisa Tsapkova 1, Natalia Bodunova1, Vera Polyakova1, Tatiana Yanova1, Anzhelika Chegodar1, Konstantin Rumyantsev1, Dmitriy Bordin1;2;3, Natalya Dekhnich4, Natali Ivanchik4 Maxim Solomadin 1, Abay Baigenzhin2;2, Lina Zaripova2, Larissa Kozina2, Alyona Boltanova2, Talgat Iglikov2 Isabel Erenas1, Nuria García2, Sandra Estepa1, Veronica Velasco3;4, Celia Chicharro1;5, María Sáinz4, Ana Fernández Araque3;4, Fernando Bandres5;6, ZORAIDA VERDE 1;4;5 Darius Čereškevičius 1, Kristina Zubielienė1, Ali Aldujeli1, Vacis Tatarūnas1 Maria Apellaniz-Ruiz 1, Edurne Urrutia Lafuente1;2, Alberto Maillo3, Oscar Teijido Hermida1, Maria Miranda Perez1, David Cobos Hermosa1, David Gomez-Cabrero2;3, Juan Jose Beloqui Lizaso4, Ander Ernaga Lorea5 Dauren Yerezhepov 1, Aidana Gabdulkayum1, Saya Amangeldikyzy2, Ainur Akilzhanova1 Haimeng Bai 1, Sandra Smiezek1, Rosarelis Torres1, Changfu Xiao1, Christos Polymeropoulos1, Gunther Birznieks1, Mihael Polymeropoulos1 Dmitrii Kharitonov 1, Elena Kovalenko1, Alexey Kamelin1, Anna Kim1, Nicolay Plotnikov1, Alexander Rakitko1 VIBHU JOSHI 1, kanika arora1, rakesh kumar pilania1, amit rawat1, Saniya Sharma1, Manpreet Dhaliwal1, surjit singh1, Deepti Suri1 Miska Kandolin 1;2, Minna Pöyhönen1;2, Eveliina Salminen1;2 Hasan Baş 1;2, Aysel Ünal1;3, Zeynep Esener1;4 Alessandra Franzoni 1, Jessica Zucco1, Dora Fabbro1, Elena Betto1, Giuseppe Damante1;2 Gregor Mlinšek1;2, Tanja Blagus3, Marija Malgaj Vrečko1, Andreja Aleš Rigler1, Vita Dolzan 3 Cita Zupanc1;2, Alenka Franko2;3, Danijela Strbac2;4, Viljem Kovac2;4, Vita Dolzan5, Katja Goricar 5 Jong Hun Kim 1 Daniela Zanetti 1, Mine Koprulu2, Fabio Busonero1, Andrea Maschio1, Francesca Crobu1, Claudia Langenberg2;3;4, Serena Sanna1;5 Carlos López 1;2, Miguel E. Aguado-Barrera1;2, Olivia Fuentes-Ríos1;2, Javier Manuel Galego Carro1;2, Carla Coedo-Costa1;2, Ana Crujeiras1;2, Patricia Calvo1;3, Ana Carballo1;3, Paula Peleteiro1;3, Begoña Taboada1;3, Antonio Gomez-Caamaño1;3, David Azria4, Manuel Altabas5, Jenny Chang-Claude6, Ananya Choudhury7, Dirk R. de Ruysscher8, Alison M Dunning9, Alexandra Giraldo10, Sara Gutiérrez-Enríquez5, Sarah Kerns11, Maarten Lambrecht12, Monica Ramos10, Tiziana Rancati13, Tim Rattay14, Petra Seibold15, Barry Rosenstein16, Victoria Reyes10, Elena Sperk17, Christopher Talbot14, Liv Veldeman18, Adam Webb14, Catharine West7, Ana Vega1;2;19 EP19 Population Genetics and Evolutionary Genetics Dubravka Havaš Auguštin 1, Brina Zagorc2;3, Jelena Šarac1, Mario Novak1, Mario Carić1, Andrea Rimpf4, Daniel Fernandes2;3;5, Inigo Olalde6;7;8, David Reich8;9, Ron Pinhasi2;3 Gabija Anikeviciute 1, Valentina Ginevičienė1;2 Marina Olkova 1, Igor Gorin1, Andrey Alimov1 Katažyna Samaitė 1, Ingrida Domarkienė1, Gabrielė Žukauskaitė1, Vaidutis Kučinskas1, Laima Ambrozaitytė1 Gabrielė Žukauskaitė 1, Ingrida Domarkienė1, Aušra Matulevičienė1, Vaidutis Kučinskas1, Laima Ambrozaitytė1 Jelena Šarac 1, Dubravka Havaš Auguštin1, Brina Zagorc2;3, Mario Novak1, Mario Carić1, Dinko Tresić Pavičić4, Daniel Fernandes2;3;5, Ron Pinhasi2;3 Alina Urnikyte 1, Laura Pranckeniene1, Vaidutis Kučinskas1 Hannes Gislason 1 Ela Zaimi 1, Merita Xhetani2, Alban Haxhia1, Zlatko Jakovski3 Ivanna Shymanska 1, Volodymyr Kravets1;2, Maria Dushar1, Marta Tyrkus1, Nataliya Matiytsiv1;2, Dmytro Mykytenko3, Halyna Makukh1 Emna BOUATROUS 1;2, Dorra Chaouachi1, imen boudrigua1, Samia Menif1, Houyem Ouragini1 Elvira Tahmazli 1, Gunay Akbarova - Ben Tzvi2 Zsolt Bánfai 1, Valerián Ádám1, Katalin Sümegi1;2, András Szabó1, Gergely Büki1, Krisztián Erős2, Kinga Hadzsiev1, Ferenc Gallyas2, Attila Miseta3, Miklós Kásler4, Bela Melegh1 Erinija Pranckevičienė 1;2, Judita Kasperiuniene2, Adomas Hendrixon3, Laimonas Januska1 Masoumeh Ghasemi 1, Marzieh Mohseni1, Masoud Edizadeh2, Yasser Riazalhosseini3;4, Kimia Kahrizi1, Hossein Najmabadi1 Hyun Jeong Kim1, Han Byul Jang1, Young-Youl Kim1, Hye-Ja Lee 1 Atanas Syarov 1;2, Adelina Yosifova3, Theodor Todorov2;3, Angelina Trifonova3, Velislava Terzieva3, Radoslava Vazharova2;3 Ketevan Kartvelishvili1;2, Nino Pirtskhelani 1;2;3, Nino Kochiashvili1, Tea Mukhuradze4, Tinatin Lezhava1 Marta Domínguez Martínez 1, Alfonso Caro Llopis1, Carla Martín-Grau1, Mónica Roselló Piera1, Juan Silvestre Oltra Soler1, Laia Pedrola1, Sandra Monfort Membrado1, Alba Gabaldon Albero1, Francisco Martinez1, Carmen Orellana1 SEHIME TEMEL 1, Mustafa Pir2, Feride Sahin3, Sebnem Ozemri Sag1, Yunus Kasım Terzi3 Saina Novgorodova 1, Aitalina Sukhomyasova1;2, Nadezhda Maksimova1 Eka Kvaratskhelia1;2, Mariam Ghughunishvili3, Sandro Surmava 1, Elene Abzianidze1;4, Arndt Rolfs5;6;7, Volha Skrahina6, Tinatin Tkemaladze1;3 EP20 Functional Genomics Hao Wu1;2, Jin-Huan Lin1;2, Xin-Ying Tang2;3, Gaëlle Marenne4, Wen-Bin Zou1;2, sacha schutz4;5, Emmanuelle Masson4;5, Emmanuelle Génin4, Yann Fichou4, Gerald Le Gac4;5, Claude Férec4, Zhuan Liao1;2, Jian-Min Chen 4 Antonio Gómez-Martín1;2, Marta Cosín-Tomàs3;4;5, Sofía Aguilar Lacasaña3;4;5, Olga Sánchez6;7, Zoraida García-Ruiz6;7, Hana Vespalcová3;4;5, Payam Dadvand3;4;5, Elisa Llurba6;7;8, Lola Gómez-Roig7;9, Jordi Sunyer3;4;5, Mariona Bustamante 3;4;5 PINAR KOSEOGLU-BUYUKKAYA 1, Neslihan Coban1 Ann-Cathrine Berking 1, Brigitte Pabst1, Uwe R. Kordes2, Lucas Müntnich3, Christian Kratz3, Jens Bohne4, Bernd Auber1, Tim Ripperger1 Ivan B. Filippenkov1, Ivan V. Mozgovoy 1, Yana Y. Shpetko1, Alina E. Denisova2, Vasily V. Stavchansky1, Leonid V. Gubsky2;3, Svetlana A. Limborska1, Lyudmila V. Dergunova1 Christian Stockhaus 1, Steffen Syrbe2, Alexandra Afenjar3, Francisco Martinez4, Denisa Weis5, Nicolas Chatron6, Arjan Bouman7, Konrad Platzer1 Liselot van der Laan 1, Rob Zwart1, Martin A. Haagmans1, Adri N. Mul1, David Dyment2, Pilar Caro3, Sebastian Sailer3, Christian Schaaf3, Marcel Mannens1, Bekim Sadikovic4, Mieke van Haelst1, Peter Henneman1 Christina Dillane 1, Juan Carmona2, Mike Tayeb1 Valentine Hoyau 1, clara blotas1, Claude Férec1, Stéphanie Moisan2 Valeria Morbidoni 1, Anais Franco Romero2, luca pannone3, Paolo Grumati4, Simone Martinelli3, Marco Sandri2, Eva Trevisson1 Hiba Alblooshi 1, Anjali Bharathan2, Anne Jhone1 Aleša Kristan 1, Aleš Maver2, Tadej Pajič2;3, Saša Anžej Doma4;5, Martina Fink4, Špela Žula4, Rok Količ6, Andrej Vuga6, Petra Hudler1, Tadeja Rezen1, Jurij Stojan1, Tanja Kunej7, Irena Preložnik Zupan4;5, Nataša Debeljak1 Sara Franco Freire 1, Maria Isabel Cabrera Gonzalez1, Marta García de Burgos1, Eva Pitters1, María del Carmen Benito López1, Francisco Javier Mérida de la Torre1, Elena Colastra Ugena1, Yolanda de Diego Otero1, Francisco Miguel rodriguez peña1, RAQUEL YAHYAOUI MACIAS1 Barbara Cugalj Kern 1;2, Jernej Kovač1;2, Robert Sket1;2, Barbara Jenko Bizjan1;2, Tine Tesovnik1;2, Klemen Dovč1;2, Tadej Battelino1;2, Nataša Bratina1;2 Friederike Ehrhart 1, Aishwarya Iyer1, Chris Evelo1, Cenna Doornbos2, Peter-Bram t Hoen2 Dilek Colak 1, Ibrahim Kaya2, Mustafa Kaya3, Hala Khalil4, Fatimah AlHamlan5, Ayodele Alaiya6, Olfat Alharazi1 Jolita Ciapaite1;2, Fried Zwartkruis3, Niklas Darin4, Ralf Husain5, Shimriet Zeidler6, Clara van Karnebeek2;7, Philippa Mills8, Judith Jans1;2, Nanda Verhoeven-Duif 1;2 Luisa Vergori 1;2, Frederic Brioude3;4, Omar Soukarieh5;6, Caroline Meguerditchian7, Yves Delneste1, Alain Morel1;2, David-Alexandre Tregouët5, Isabelle Tournier1;2 Karin Weiss 1;2, Karim Karimi3, Yael Lichtenstein1, Jack Reilly3, Haley McConkey3, Raissa Relator3, Jennifer Kerkhof3, Michael Levy3, Arjan Bouman4, Peter Turnpenny5, Joseph Symonds6, Philippe M. Campeau7, Matthew Tedder8, Tugce Balci9, Bekim Sadikovic3;10 Carlo Maj1, Gabriel Schweizer1, Johannes Schumacher1, Pouria Dasmeh 1;2 EP21 New Technologies Makbule SOMUNCU 1, Mahmut Selman Yıldırım1, Cihan Aydın2, Ayşe Gül Zamani1, Esra Albayrak3, Tuğçe Duran4, İbrahim Halil Kavaklı5 Hannah Titheradge 1;2, Lorraine Hartles-Spencer3, Jessica Woodley3, Emma Douglas1, Madhura Chandrashekara1, Derek Lim1, Esther Kinning1, Ataf Sabir1, Helen Brittain1, Deborah Osio1, Kai Ren Ong1, Mary O’Driscoll1, Helen Cox1, Joanna Jarvis1, Denise Williams1, Rebecca Igbokwe1, Joanne Stockton2, Luke Ames2, Andrew Beggs2 Yang Heng 1;2;3, Sha Liao1;2;3, Weiqing Liu1, Yong Ma1, Uma Sangumathi Kamaraj4, Dongmei Jia1, Xiuwen Feng1, Diyan Liang1, Jinqiong Xiang1, Caili Huang1, Jiajun Zhang1;2;3, Min Jian1, Kui Su1, Mei Li1, Yuin-Han Loh4, Ao Chen2;3, Xun Xu1 Anna Franzoni 1, Cristiano Sabelli1 Deniz Aksu1, Selin Akad Dincer1, Zerrin Çelik1, Feride Sahin1, Sema Karakus2, Yunus Kasım Terzi 1 Chris Kyriakidis 1, Ahmed Saad Elmorshedy2, Walaa Elamir Mohamed3, Carmen Garrido Navas1;4;5, Sara Chulián Prado4, Anne Kristine Schack1;6, Karmele Alapont1, Zoran Velkoski1, Marija Chaushevska1;7, Gjorgji Madjarov1;7 Carmen Hernandez 1, Virginia Perez1, alicia hernandez1, mario garcía mayo1, andreea lupsor1, candela montaner1, Inés Valledor1 Nicholas Kueng1, Daniel Sidler2, Banz Vanessa3, Carlo Largiadèr1, Charlotte Ng4, Ursula Amstutz 1 Nikolaos Marinakis 1;2, Anastasios Mitrakos1;2, Danai Veltra1, Maria Svingou1, Faidon-Nikolaos Tilemis1, Kyriaki Kekou1, Konstantina Kosma1, Aphrodite Kampouraki1, Hane Lee3, Yongjun Song3, Katherine Anagnostopoulou4, Yannis Loukas4, Periklis Makrythanasis1;5;6, Christalena Sofocleous1, MARIA TZETIS1, Jan Traeger-Synodinos1 Adrián Alcalá San Martín 1, Neus Fornés Garcia1, Didac Casas-Alba1, Patricia Fernández1, Patricia Peral García1, Sara Manresa García1, Gisela Márquez1, Enric Balius Fort1, Cristina Hernando-Davalillo1 Iris Pereira Caetano 1, Joana Mendonça1, Alice Mirante2, Rita Cardoso2, Marcia Rodrigues3, João Paulo Oliveira4, Ana Grangeia4, David Barbosa5, Luís Vieira1;6, João Gonçalves1;6 Krystal Sheridan1, Eleanor Matrullo2, Melissa Netwal3, Paul Lomax 3 Chaithanya Ponnaluri1, Vaishnavi Panchapakesa1, Daniel Evanich1, Matthew Campbell1, Bradley Langhorst1, Louise Williams 1 Wantanai Satheanwongsa1, Chayada Tangshewinsirikul2, Donniphat Dejsuphong3, Preyaporn Onsod1, Budsaba Rerkamnuaychoke1, Teerapong Siriboonpiputtana1, Takol Chareonsirisuthigul 1 Iván Monge Lobo 1, Barbara Fernández Garoz1, Diana Monclús Arroyo1, Elena M Ruiz Ballesteros2, Claudia Toledo Pacheco2, Ana Isabel Quinteiro García1, Nelmar Valentina Ortiz Cabrera1 Elian Rakhmanaliev 1, Hwei Ling Khor1, Swee Yin Lim1, Shi Hui Ang1, Mohammed Irfan1, Siti Rafei1;2, Karen Wang1;2, Mi Kyoung Park1 Angharad Roberts 1;2;3, Marina DiStefano4, Erin Rooney Riggs5, Katherine Josephs1, Fowzan Alkuraya6, Joanna Amberger7, Mutaz Amin8, Jonathan Berg9, Alison Coffey10, Fiona Cunningham11, Karen Eilbeck12, Helen Firth13, Julia Foreman14, Ada Hamosh7, Eleanor Hay2, Sarah Leigh15, Christa Martin5, Ellie McDonagh14, Daniel Perrett14, Erin Ramos16, Peter N. Robinson17, ANA RATH8, Zornitza Stark18, David Sant12, Nicola Whiffin19, Heidi Rehm4;20, James Ware3;4;21 Irina Panchuk 1, Svetlana Smirnikhina1 Cristina Rodilla 1;2, Gonzalo Nuñez Moreno1;2;3, Raquel Romero1;2;3, Pablo Mínguez1;2;3, Marta Corton1;2, Carmen Ayuso1;2 EP22 Treatments for Genetic Disorders Ekaterina Kondrateva 1;1, Anna Demchenko1, Olga Volodina1, Alexander Lavrov1, Svetlana Smirnikhina1 Anthony Goldstone 1, JESUS DOMINGUEZ RISCART2, Giusi Umano3, Orit Hamiel4, Melek Yildiz5, Melania Manco6, Patrick Sleiman7, Charles Savoie7, Dinesh Giri8, Jesús Argente9 Massimiliano Chetta 1, Marina Tarsitano2, Maria Oro3, Maria Rivieccio3, Liberato Marzullo4, Alessandra Rosati5, Nenad Bukvic6 Emanuela Pesce 1, Elisa Giorgio2;3, Tiziano Bandiera4, Stefano Ratti5, Ilaria Cani5, Pietro Cortelli5;6, Nicoletta Pedemonte1 Lorenzo Allegri1, Federica Baldan 1, Catia Mio1, Maimouna Shaar1, Giuseppe Damante1 Siobhan Kerr 1, Kinga Groß2, Johannes Zschocke2, jonathan berg1 Nilay Gunes 1, Dilek Uludag Alkaya1, Beyhan Tüysüz1 Carel Le Roux1, JESUS DOMINGUEZ RISCART2, Maria Rosaria Licenziati3, Leandro Soriano-Guillén4, Belma Haliloglu5, Anjali Zalin6, Simona Filomena Madeo7, Patrick Sleiman8, Charles Savoie8, Liya Kerem9, Jesús Argente 10 nasim mayeli fereydani 1, mansoureh ajami2, Elham Hoveizi1, Arash Alghasi3, Hamid Galehdari1 Orhan Kerim Inci 1, Volkan Seyrantepe1 EP23 Genetic Counselling / Services / Education Belinda McClaren 1;2;3, Melissa Martyn1;2;3, Jessica Ince1;2, Emma Weisz1;2;3;4, Katie Arkell1, Natasha J Brown3;5, Michael C. Fahey4, Clara Gaff1;2;3 Akane Kondo 1;2, Mikio Morine1, Takehiko Matsuyama3, Kazuhisa Maeda1 Mari Hachmeriyan 1;2, Mariya Levkova1;2, Trifon Chervenkov2;3, Eleonora Dimitrova4;5, Milena Stoyanova1;2, Dinnar Yahya2;4, Lyudmila Angelova2 Trang Do 1;2, Melissa Martyn1;2;3, Belinda McClaren1;2;3, Clara Gaff1;2;3, Alison McEwen4 Gulshara Abildinova1, Meruert Zhumazhanova2, Laura Ainabekova2, Dana Makhsutova2, Maral Muzar2, Manshuk Zhulkarova2, Tattikul Tazhenova3, Zhulduz Zhumadilova3, Nurbibi Suindikova4, Gulnara Abdresheva 5 Suzanne Nevin1;2, lauren kelada 1, Claire Wakefield1, gail hilton3, meg maack3, jason djafar1, kristina elvidge3, michelle farrar1 Monika Pokorna 1, Štefan Lednický1, Monika Koudová1, David Stejskal1 Angela Bedard1;2;3, Samantha Pollard4;5, Melanie O’Loughlin3;6, Anna Tsoy2, James Bedard2, Christina Rajsic7, Fabio Feldman6, Farah Jamal8, Alice Virani3;9, Sophie Sun6, Kasmintan Schrader3;6;10, Jennifer Nuk 3;11 Mariana Mazza Santos1, Pricila Bernardi2, Anelize Baranzeli1, Thais Assis1, Bruna Kellet Coelho1, Bruno Hech Dominski1, Sara Silva Lima1, Maristela Ocampos 1 Diana Salinas-Chaparro 1, Laura Ortega2, mar borregán prats1, Gema Escribano-Serrano1, alba pascual rodríguez3;4, Natalia Castejón-Fernández2, Guerau Fernandez1;3;4, Judith Armstrong1;3;4, Delia Yubero1;3;4, Cristina Hernando-Davalillo1, Adrián Alcalá San Martín1, Maria Victoria Dominguez5, Sandra González Castejón5, Laura Mallen5, Federico Mayor Zaragoza6, Magdalena Ugarte2, Belén Pérez2;4, Francesc Palau1;3;4;7 Emiko Kise 1;2, Tomomi Kojima1, Akiko Sakyu1, Tomoki Kosho1;3;4 Milena Stoyanova 1;2, Mari Hachmeriyan1;2, Mariya Levkova1;2, Dinnar Yahya1;2, Hristina Hristozova3;4, Lyudmila Angelova1 Konstantinos Agiannitopoulos 1, Natasa Katseli1, Kevisa Potska1, Christina Dogka1, Nikolaos Tsoulos1, Athanasios Papathanasiou1, Dimitrios Grigoriadis1, Georgios Tsaousis1, Konstantinos Papazisis2, Ioannis Natsiopoulos3, Ioannis Xanthakis4, Nikolas Bredakis5, Sofia Karageorgopoulou5, Dimitrios Mavroudis6, Christos Markopoulos7, Ioannis Boukovinas8, Christos Papadimitriou9, Vasileios Venizelos10, Athina Christopoulou11, Anna Koumarianou12, Eirini Papadopoulou1, George Nasioulas1 Milena Paneque 1;2, Maria Barbosa1;3, Angus Clarke4, Sofia Dias5, Filipa Júlio6, Alison Metcalfe7, Jorge Sequeiros1, Liliana Sousa5, Álvaro Mendes1 Eulàlia Rovira-Moreno 1, Anna Abulí1, Patricia Muñoz-Cabello1, Marta Codina1, Clara Serra-Juhé2, Eva Bailles3, Eduardo Tizzano1 Elena Baranova 1, Vera Polyakova2, Tatyana Yanova2, Marina Tripolskaya2, Ekaterina Efremova1, Vera Izhevskaya3, Natalya Bodunova2 rafaella metaxa 1, Andri Miltiadous2, Petroula Gerasimou2, natasa petridou1, Emily Athanasiou3, Paul Costeas4, Violetta Anastasiadou3 Alisa Förster 1, Clara Köppen1, Gunnar Schmidt1, Bernd Auber1 Rodica Dragotoiu 1 Marianne Le Dref 1, Alain Verloes1, Anne Hugon1, Nicholas Szeto1, Klea Vyshka1 Vijayalakshmi Salem Ramakumaran 1, Neeta Lakhani1, emily craft1, Pradeep Vasudevan1 LAI-PING LISA SIU 1, Min, Linda Ou1, Wing-Yiu, Yoyo Chu1, Stephanie Ho1, Wing-Yan, Sharon To1, Sze-Wing, Shirley Cheng1, Ho-Ming Luk1, Ivan Fai-Man Lo1 Nicholas Szeto 1, Marion Gerard2, Yves Gillerot3, jonathan berg4, Maurizio Genuardi5, Martin Krahn6, Ute Moog7, Edward S. Tobias8, Peter Turnpenny9, Johannes Zschocke10, Dorica Dan11, Sofia Douzgou HOUGE12, Marianne Le Dref1, Laurence Faivre13, Raoul Hennekam14, Anne Hugon1, Jean-Marie Jouannic15, Tjitske Kleefstra16, David Koolen16, Giovanni Mosiello17, Alessandra Renieri18, Marco Tartaglia17, Zeynep TÜMER19, Birute Tumiene20, Agnies van Eeghen14, Lisenka Vissers16, Klea Vyshka1, Dagmar Wieczorek21, Giuseppe Zampino5, Christiane Zweier22, Alain Verloes1 Anne Hugon 1;2, Dorica Dan3, Isabella Brambilla4, Vedrana Bibic5 Eka Kvaratskhelia 1;2, Sandro Surmava1, Tinatin Tkemaladze2, Nona Janikashvili3, Eka Ekaladze4, Elene Abzianidze1;5 Ludovico Graziani 1, Francesci Piceci Sparascio2, Mario Bengala3, Allessandra Terracciano2, Cinzia Galasso4, Antonio Novelli2, Giuseppe Novelli1;3 Evelina Vaitėnienė 1, Algirdas Utkus1 Iva Strnadova1;2, Karen-Maia Jackaman1, Jennifer Hansen1, Julie Loblinzk1, Manjekah Dunn1, Skie Sarfaraz1;2, Sam Hurd1, Joanne Danker1, Michelle Tso1, Sierra Angelina Willow1, Chloe Molnar1, Jackie Boyle3, Jackie Leach Scully1, Elizabeth Palmer1, Bronwyn Terrill 4;5 Zeynep Derin 1, ayşe büşra akalın2, Elif EROĞLU ŞİMŞEK3, İlknur Sunar3, Ibrahim Akalin3 EP24 Ethical, Legal and Psychosocial Aspects in Genetics Aina Isabel Gayá Barroso 1;1, Juan Gonzalez Moreno2, Adrián Rodríguez1, Milena Paneque3, Inés Losada López1, Eugenia Cisneros-Barroso1 Kristina Karrman 1 Lydia Seed 1;2, Anna Scott2;3, Michelle Peter4, Shereen Tadros5, Amanda Pichini6, Cristine Sortica da Costa2;7, Melissa Hill4 Sophie Stoesslein 1, Dagmar Wahl1, Konstanze Hörtnagel1 Larissa Mattern 1, Cordula Knopp1, Julia Suh1, Katja Eggermann1, Annette Lischka1, Jeremias Krause1, Robert Meyer1, Nergis Güzel1, Daniela Dey1, Eva Lausberg1, Thomas Eggermann1, Florian Kraft1, Matthias Begemann1, Ingo Kurth1, Miriam Elbracht1 Anne Cambon-Thomsen 1, Marc Billaud2, Catherine Bourgain3, Sarah Carvallo4, Nicolas Chatauret5, Isabelle Durand-Zaleski6, Christine Lemaitre7, Lucie Morillon7, Laurent Pasquier8, Clément Pimouguet9, Marie Préau10, Emmanuelle Rial-Sebbag1, Dominique Stoppa-Lyonnet11, Frédérique Nowak7 Lisa Feriol 1, Gauthier Chassang2, Emmanuelle Rial-Sebbag2 Ines Martin Villalba 1 Iva Ivanova 1;2, Galina Chaneva1, Dragomira Nikolova1;3 annick REIN ROTHSCHILD 1;2, michael tsur3, Ben Pode-Shakked1;2 Shiri Shkedi Rafid 1, roy gilbar2, aviad raz3 Kirsten Riggan1, Yvonne Bombard2, Natasha Bonhomme3, Roel Feys4, Heidi Carmen Howard5, McKenna Horstmann6, Karen Meagher1, Marsha Michie6, Kiran Musunuru7, Kelly Ormond 8, Sabina Rubeck6, Jane Yap9, Rosario Isasi4, Megan Allyse9 Salma Shickh 1, Viji Venkataramanan1, Katharine Fooks1, Karen MacDonald2, Trevor Seeger2, Taila Hartley3, Francois Bernier2, Kym Boycott3, Deborah Marshall2, Robin Hayeems1;4 Pauline Rateau 1, Emmanuelle Rial-Sebbag1 References

Volume XX | Supplement X

Berlin, Germany

June 1-4, 2024

© The Author(s), under exclusive licence to European Society of Human Genetics 2024

Sponsorship: Publication of this supplement was sponsored by the European Society of Human Genetics. All content was reviewed and approved by the ESHG Scientific Programme Committee, which held full responsibility for the abstract selections.

Disclosure Information: In order to help readers, form their own judgments of potential bias in published abstracts, authors are asked to declare any competing financial interests.

Contributions of up to EUR 10 000. (Ten thousand Euros, or equivalent value in kind) per year per company are considered “Modest”. Contributions above EUR 10 000.- per year are considered “Significant”.

Presenting author names are underlined in the contributor lists.

e-Posters

EP01 Cancer Genetics

EP01.001 Relevance of the gene ITIH5 in vulvar diseases

Olga Rodenko1, Anke Esber1, Irina Cepraga1, Stefanie Schütze1, Lars Jansen1, Mieczyslaw Romuald Gajda2, Nikolaus Gaßler2, Ingo B. Runnebaum1, Matthias Dürst1, Norman Häfner1, Claudia Backsch 1

1Department of Gynecology and Reproductive Medicine, University Hospital Jena, Jena, Germany; 2Section of Pathology, Institute of Legal Medicine, University Hospital Jena, Jena, Germany

Background/Objectives: The number of vulvar cancers (VC) and their precancers has increased worldwide in recent decades. To date, molecular genetic markers have not played a role in the differential diagnosis of vulvar intraepithelial neoplasias (VIN) and VC. The gene ITIH5 could be play a role as such a marker in vulvar carcinogenesis as like in other cancers. The aim of the present study is to determine the ITIH5 status on different vulvar patient material.

Methods: The copy number status of ITIH5 was analysed using fluorescence in situ hybridization in vulvar scrapes and vulvar tissues of patients with histologically confirmed VIN and VC and as well as Lichen sclerosus (LS) (scrapes n = 10, tissues n = 28). In parallel, promoter methylation was investigated by methylation specific PCR on vulvar scrapes and expression was determined using immunohistochemistry on paraffin embedded vulvar tissues.

Results: In vulvar scrapes, 60% of VIN and 60% of LS showed ITIH5 copy number loss and in 40% of VIN and in 20% of LS ITIH5 methylation were found. In paraffin sections, ITIH5 copy number losses were detected in 36.4% of VIN, in 40% of VC and in 14.3% of LS. Evaluation of ITIH5 expression in paraffin sections are still ongoing.

Conclusion: ITIH5 aberrations seems to be play a role in vulvar carcinogenesis. The use of ITIH5 as a molecular genetic marker could possibly enable a better treatment-relevant or prognostic differentiation of VIN, VC, LS and recurrences in the future.

Grants: Project is supported in part by the Brigitte and Dr. Konstanze Wegener Stiftung.

Conflict of Interest: None declared

EP01.002 Exploring the Impact of Secretome on Inflammation in Myeloproliferative Neoplasms (MPNs)

Selin Fulya Toprak Yalçın 1, Esra Nur Demirtaş1, Tugba Sarac-Sivrikoz2, Selcuk Sözer Tokdemir1

1Istanbul University Aziz Sancar Experimental Medicine Research Institute, genetic, Istanbul, Türkyie; 2Istanbul University Faculty of Medicine, Department of gynecology and obstetrics, Türkyie

Background/Objectives: Myeloproliferative Neoplasms (MPNs) are characterized by a critical inflammatory component that exacerbates disease progression. This is believed to stem from alterations in the bone marrow milieu. Our research focuses on assessing the influence of the secretome, released by mutant cells, on the inflammatory processes in MPNs.

Methods: We utilized HEL cells-derived spent media, introducing it to cord blood mononuclear cells (MNCs) in co-culture systems. Following a 48-hour incubation, the supernatants from these cultures were subjected to ELISA using the LEGENDplex™ cytokine array. Additionally, RT-PCR was employed to quantify changes in IRF3 and cGAS gene expression in the affected cells.

Findings: Significant enhancements in the secretion of cytokines IFN-γ, IL-1β, IL-12p70, and IL17A (P < 0.0001) were detected from infected cells compared to control groups. Concurrently, there was a marked upregulation in IRF3 (~3-fold) and cGAS (~1.5-fold) gene expressions, relative to control groups.

Conclusions: Our findings underscore the pivotal role of the secretome in promoting the cytokine release from healthy cells which may contribute to inflammation and immune response activation in MPNs. Moreover, this research enhances our understanding of genometastasis and potentially guiding the development of more precise and effective treatment modalities for this disease.

Grant: This project is supported by Istanbul University Project Support Office with TSA-2022-39111 grant

Conflict of Interest: None declared

EP01.003 Exploring prevalence of BARD1 pathogenic variant in breast cancer patients

Albertas Valintelis 1, Ieva Drejerienė1, Matas Norvydas1, Jūratė Gruodė1;2

1Klaipėda University Hospital, Klaipėda, Lithuania; 2University of Klaipėda, Klaipėda, Lithuania

Introduction: Founder mutations or population-specific variants are significant in disease diagnostics, especially in cancer genetics. BRCA1, BRCA2 gene mutations in Ashkenazi Jewish and Polish-Baltic populations allow for a more precise examination plan or even population-specific screening. We present a variant of the BARD1 gene identified in a cohort of Lithuanian women with breast cancer (BC), consistent with a hereditary predisposition to cancer. BARD1 is a known cancer susceptibility gene that moderately increases breast cancer risk. Among the BC patients BARD1 pathogenic variants were found to be around 0.25%. The aim of this study was to analyze pathogenic BARD1 variants among our cohort.

Methods We performed Ion Torrent on-demand panel testing for 883 patients diagnosed with BC, referred to Klaipėda University Hospital between 2018 and 2023. The library and template preparations were executed through the automated Ion Chef System or IonTorrent PGM, followed by sequencing on NGS Ion torrent platform, in accordance with the manufacturer’s guidelines. Results were analyzed using Ion Reporter Software.

Results In our patient cohort, we identified 5 unrelated patients with heterozygous pathogenic BARD1 frameshift variant (0.56%) NM_000465.4:c.2300_2301del (p.Val767AspfsTer4). No other BARD1 pathogenic variant was found. This variant was observed in the gnomAD controls 0.00002288% among European (non-Finnish) population. 3 of the 5 patients had triple negative BC, 2 patients had bilateral BC. 1 Patient also had BRCA1 pathogenic variant.

Conclusion: This study identified 1 pathogenic variant among the cohort that had high frequency rate among Lithuanian population. Further studies are required to determine this variant clinical management.

Conflict of Interest: None declared

EP01.004 Diagnostic value of NGS and Microarrays vs Conventional Karyotype and FISH in a case of hematological malignancy.

Fotini Ieremiadou1, Kostantinos Tyrosvoutis2, Aspasia Divane 3

1Life Code, Mol. Biol., Athina, Greece; 2Life Code, Cytogenetics, Athina, Greece; 3Life Code, Director, Athina, Greece

Background/Objectives: Present case is part of a larger effort on comparing diagnostic value of molecular techniques (NGS and Microarray) versus conventional analysis (karyotyping and FISH) in hematological malignancies.

Methods: A 71 years old CLL male patient came to our Lab for genetic testing.

Cytogenetic analysis performed on G-banded chromosome preparations from unstimulated BM culture. FISH performed using ZytoLight SPEC TP53/CEN 17 Dual Color Probe.

DNA extracted from peripheral blood using NucleoSpin blood extraction kit (Macherey-Nagel). NGS performed using custom panel (Agilent Technologies) and data analyzed by seqpilot software (JSI medical systems). CytoScan® 750K Array (Afffymetrix) and ChAS software used for detection of CNVs and LOH.

Results: Cytogenetic analysis revealed a 46,XY[9] karyotype.

I-FISH revealed one hybridization signal both for 17p13.1 and centromeric region in 90%.

NGS revealed: 1. the presence of TP53 p.R273L (c.818G>T) pathogenic variant (confirmed by sanger). 2. deletion of VHL, XPC, MLH1, MYD88, RET, BMPR1A, PTEN, DICER1 and TP53 and duplication of FANCD2, SMARCA4 and CALR genes.

Microarray analysis showed that these CNVs are part of larger multiple aberrations that include 3p and 14q terminal deletions, monosomy 10, 17p deletion and 19p terminal duplication.

Conclusion: NGS and microarray can play a vital role in the analysis of hematological malignancies. NGS can provide information both for chromosomal aberrations and mutational status eliminating the need for multiple testing. Detection of fusions by NGS is also possible but its efficiency remains to be tested.

Conflict of Interest: None declared

EP01.005 Retrospective study of non-informative BRCA1/2 families using NGS. The path towards preventive and personalized medicine.

Mónica Arranz Ledo 1, Enrique Lastra Aras2, Amaya Olaverri Hernández3, Marta Orozco Belinchón3, Enrique Pérez Riesgo1, Lara Hernández Sanz1, Noemí Martínez Martín1, Mar Infante Sanz1, Mercedes Durán Domínguez1

1Institute of Biomedicine and Molecular Genetics, University of Valladolid-Spanish National Research Council (IBGM, UVa- CSIC), Cancer Genetics Group, Unit of Excellence, Valladolid, Spain; 2Burgos University Hospital, Unit of Genetic Counseling in Cancer, Oncology, Burgos, Spain; 3Rio Hortega University Hospital, Unit of Genetic Counseling in Cancer, Oncology, Valladolid, Spain

Background/Objectives: It is widely accepted that analysing a greater number of predisposition genes benefits more families with suspected hereditary breast and ovarian cancer (HBOC). Since 2018, our Hereditary Cancer Laboratory has routinely used multigenic panels instead of BRCA1/2 screening alone, resulting in an increased diagnostic yield from 11% to 20%. However, many families included in the HBOC prevention program prior to its implementation did not have access to a more accurate diagnosis. Therefore, we conducted a retrospective study to evaluate the usefulness of reanalysing families with non-informative results from BRCA1/2 screening.

Methods: We selected a cohort of BRCA1/2 non-informative HBOC samples for them analyse using a multigene panel of 40 predisposition genes. Genomic DNA was extracted from peripheral blood and NGS was performed using ThermoFisher Scientific technology. Library and template preparation was performed on the Ion Chef System, followed by sequencing on the Ion S5. The results were analysed using Ion Reporter Software.

Results: In our study, 19% of the non-informative BRCA1/2 samples analysed were positive for a predisposition gene. The main mutated genes were ATM, CHEK2, BRIP1, PALB2 and RAD51D.

Conclusion: Our findings highlight the need to reanalyse suspected families for HBOC by using multigenic panels. The use of hereditary cancer testing can benefit a larger number of families by providing genetic counselling, preventive medicine for healthy carriers, and personalised medicine based on the mutated gene for those who have developed cancer.

Grants: Predoctoral fellowship co-financed by European Social Fund and Junta de Castilla y León (EDU/842/2022). Research Project PID2021-125909OB-100, IP Carlos Villalobos.

Conflict of Interest: None declared

EP01.006 Fumarate hydratase gene mutation in a family with familial colorectal cancer type X

Marija Staninova Stojovska 1, Elizabeta Krstevska Bozhinovkj1, Nadica Matevska-Geshkovska1, Nenad Mitrevski2, Biljana Grozdanovska2, Milco Panovski2, Aleksandar Dimovski1;3

1Faculty of Pharmacy, Ss. Cyril and Methodius University, Skopje; 2Faculty of Medicine, Ss. Cyril and Methodius University, Skopje; 3Macedonian Academy for Sciences and Arts, Skopje

Background/Objectives: Familial colorectal cancer type X (FCCX) is the most heterogeneous type of hereditary cancer of the large bowl whose genetic background has still not been determined.

Methods: We performed targeted NGS sequencing of 36 FCCX families from N. Macedonia. The inclusion criteria were absence of multiple polyps, MSS tumors and Amsterdam I/II positive family history. All patients were analyzed using multigene panels covering coding and exon/intron sequences of >100 genes implicated in hereditary cancers.

Results: We identified an in-frame insertion pathogenic variant c.1431_1433dupAAA p.Lys477dup in the fumarate hydratase (FH) gene which segregated with the disease in one of the examined families with multiple affected members over three generations. The propositus was a 38y old male who was diagnosed with advanced rectal cancer. The same mutation was detected in his sister (45y) who had several distal bowel polyps and his daughter who was diagnosed with Wilms tumor (WT-1 mutation negative) at the age of 10y. The patient’s father was affected with colorectal (56y) and lung cancer (71y). His uncle (56y) and aunt had prostate cancer and breast cancer (63y) + AML (70y), respectively. These two family members were not available for study. The known clinical features of the carriers of pathogenic FH mutations (HLRCC Sy - hereditary leiomyomatosis and renal cell cancer) were not present in the affected members of this family.

Conclusion: We describe a family with pathogenic FH mutation which co-segregated with the predisposition to various cancers, including colorectal FCCX cancer.

Grants: No

Conflict of Interest: None declared

EP01.007 Molecular characterization of tumor DNA and RNA from patients with lung cancer

Ieva Drejeriene 1, Matas Norvydas1, Albertas Valintelis1, Jūratė Gruodė1;2

1Klaipeda University Hospital, Klaipėda, Lithuania; 2Klaipeda University, Klaipėda, Lithuania

Introduction: Lung cancer stands out as the most prevalent cancer diagnosis as well as the leading cause of cancer-related deaths in the world. Targeted therapies guided by molecular diagnostics have become a standard treatment of lung cancer. On that note, molecular testing should be quick, comprehensive, and in accordance with clinically approved guidelines. The best option in lung cancer diagnostics is to use new generation sequencing (NGS) platform. It allows rapid and becomes economically viable testing of multiple targets (hotspot mutations, translocations, copy number variations) from one sample with a single assay.

Materials and Methods: DNA and RNA were extracted from 126 lung cancer formalin-fixed, paraffin-embedded (FFPE) tumour samples. Hotspot mutations, translocations and copy number variations were detected using NGS (Ion Torrent™ 5S System) Oncomine Focus Assay panel (ThermoFisher). Sequencing data were analyzed with Ion Reporter™ Software on ThermoFisher™.

Results: Hotspot mutations, translocations or copy number variations were detected in 80 (63,5%) of 126 lung cancer samples. Hotspot mutations were detected in KRAS, MAPK2K1, EGFR, BRAF, ERBB2, HRAS, MET, CTNNBI, PIK3CA genes and amounted to 50% of all variations. Translocations were identified in 16,25% and copy number variations in FGFR1, MYC, PIK3CA, BRAF, FGFR1, KRAS, AR, MET, MYCN, CCND1 genes constituted 33,75% of all genetic aberrations. Of all genetic findings, 44,5% of variants were clinically actionable driver mutations.

Conclusion: The findings underscore the practical value of our assay as a diagnostic tool capable of providing insights into cancer diagnosis, prognosis, and directing therapeutic approaches.

Conflict of Interest: None declared

EP01.008 Investigation of cancer related genes in patients with Enchondromatosis

Ian Babler-Madrid 1, Nara Sobreira1, Renan Martin1, Elizabeth Wohler1

1Johns Hopkins University

Background/Objectives: Ollier disease (OD) and Maffucci syndrome (MS) are cancer susceptibility syndromes, and their molecular bases are not fully understood. Approximately 50% of patients with these syndromes develop a malignancy, including chondrosarcomas and gliomas. The objective is to identify the molecular bases of unsolved OD and MS cases.

Methods: A burden test was conducted with a multisample VCF file containing WGS data from 638 samples (74 cases, 564 controls) and WES data from 2141 samples (46 cases, 2095 controls). The VCF was filtered based on the gene symbols of 1202 genes described in GeneDx, COSMIC, and glioma literature. Only single nucleotide variants (SNVs) were analyzed and filtered based on RefSeq and gnomAD annotations to include functional, rare (MAF ≤ 0.01) SNVs. The variants in genes that were more mutated among patients than controls with a p-value < 0.05 were further evaluated based on their gnomAD frequency, familial segregation, variant type, in silico predictions, ClinVar, HGMD, and ACMG classifications.

Results: Out of 534 genes with at least one variant in a case, 44 were more mutated among patients than controls with a p-value < 0.05. Further analysis identified strong candidate genes that are being further evaluated, including FGFR1 and TCF3.

Conclusion: The most robust candidate genes will be further investigated functionally and in unrelated cohorts of patients with sarcoma-related diseases to determine if they are involved with these malignancies.

Grants: National Human Genome Research Institute/National Heart, Lung, and Blood Institute (NHGRI/NHLBI) grant UM1 HG006542; Gabriella Miller Kids First Pediatric Research Program grant X01HL140517; NIH/NCI grant R03CA256535.

Conflict of Interest: None declared

EP01.009 Complex diagnostics of somatic mutations in tumor cells

Gulshara Abildinova1, Zhanna Zhabakova 1, Anna Borovikova1

1Medical Center Hospital of the President’s Affairs Administration of the Republic of Kazakhstan, Laboratory genetics, Astana, Kazakhstan

Consortium: Complex diagnostics of somatic mutations in tumor cells

The molecular profile of a tumor is necessary for personalized selection of drugs for cancer patients in order to establish the risk of tumor development, clarify the prognosis of the disease and predict the response to treatment.

Aim: search for mutations in paraffin-fixed tumor tissues in order to optimize targeted treatment.

Material and research methods: The research material was DNA and RNA isolated from tumor cells.

Method: DNA and RNA library preparation followed by sequencing of amplified fragments was carried out according to the manufacturer’s protocol.

Results: The study group consisted of 240 patients with tumors of various locations (70% solid, 30% gliomas). 140 patients with solid tumors used a set of 52 and 161 genes. 80% of patients had 1 mutation and 20% had 2 or more mutations

Missense mutations were identified in 75% of cases [BRAF (Val600Glu); KRAS (Gly12Val); IDH1(R132H); JAK3 (S493C); PIK3CA (E545Q; E542K; p.(H1047L), PTEN H93Y], CDK12 c.1047-1G > A, TERT c.-124C > T for the prognosis of glioblastoma.

In 10 patients, a combination of 2 or more mutations was identified [PIK3CA (E547K), FGFR3 (G697S); PIK3CA (E542K), KRAS (Gly12Val); ATR c.7762-2A > G, ATR c.5739-14_5739-6delinsT EGFR(A289T), JAK3(S493C); BRAF (G469V), KRAS (Gly12Val); TP53, FGFR4, CREBBP, SMARCA4].

In 10% of cases, an increase in the number of copies of the EGFR and KRAS genes was detected, and in 10% of cases, a fusion of the gene (RNA) MET-MET.M13M15, TMPRSS2-ERG, RBB2 amplification was detected.

Conclusion: For personalized prescription of targeted drugs, it is more effective to use multigene diagnostics for a comprehensive study of the mutational status of the tumor.

Conflict of Interest: None declared

EP01.010 Clinical relevance of immune microenvironment and gene-expression-based biomarkers in colorectal cancer

Vincenzo Rallo 1, Matteo Massidda2, Xiaofen Wen3, Jiaxin Shen3, Manila Deiana1, Andrea Maschio1, Antonio Mario Scanu2, Maria Rosaria De Miglio2, Andrea Angius1

1Institute for Genetic and Biomedical Research, Italian national research council, Monserrato, Italy; 2University of Sassari, Sassari, Italy; 3Cancer Hospital of Shantou University Medical College, Shantou, China

Background/Objectives: We explored genomic and immune landscape of colorectal cancer (CRC) compared to normal adjacent tissue (NCT). Starting from differentially expressed genes (DEGs) data, protein-protein interaction (PPI) network and deconvolution analysis were employed to unravel the intricate interplay between molecular alterations and immune cell CRC composition.

Methods: The PPI network was constructed using the STRING database and Cytohubba MCC algorithm was applied to identify hub genes. Deconvolution analysis utilizing CIBERSORTx estimated the immune cells proportions between CRC and NCT. Statistical analyses, including the Wilcoxon signed rank test, effect size measurement and Cox regression, were applied to determine the magnitude of variations in cell populations and their correlation with overall survival.

Results: The PPI network exploration revealed ten hub genes upregulated in CRC, showing significant enrichment (P < 1.0e-16). KEGG pathway analysis highlighted their involvement in critical cellular processes such as cell cycle, cell senescence, and p53 signaling pathways. Deconvolution analysis disclosed a distinct immune microenvironment, with fifteen cell types exhibiting significant differences in abundance and impact between CRC and NCT. Two correlated with a favorable and three with worse prognosis.

Conclusion: We provided insight into the molecular and immune dynamics in CRC, identifying potential prognostic biomarkers and therapeutic targets. The intricate interplay between specific immune cell populations and clinical outcomes underscores the importance of genetic and immune factors in the management of CRC. These findings contribute to advancing our knowledge of cellular heterogeneity and CRC pathogenesis toward personalized patient treatment strategies.

Grants: 2022WBLA3Z and B/2020/0094 from MIUR and Intesa Sanpaolo

Conflict of Interest: None declared

EP01.011 Prostate cancer genetic screening for BRCA1 and BRCA2 mutations

Agnieszka Podgorska 1, Ewa Kwiatkowska1, Renata Lotocka1, Pawel Wiechno2, Katarzyna Seliga1, Aleksandra Gos1, Katarzyna Zielinska-Musko1, Lukasz Fulawka1, Aleksandra Kuzniar1, Kinga Koszucka1, Andrzej Tysarowski1

1Maria Sklodowska-Curie National Research Institute of Oncology, Cancer Molecular and Genetic Diagnostics Department, Warsaw, Poland; 2Maria Sklodowska-Curie National Research Institute of Oncology, Department of Uro-Oncology, Warsaw, Poland

Background/Objectives: Prostate cancer is the most commonly diagnosed malignancy in men. Patients with diagnosed BRCA-positive, castration-resistant, metastatic prostate cancer can qualify for PARP-inhibitor targeted therapy. Therefore, genetic testing for at least BRCA1 and BRCA2 genes should become a routine practice, however available prostate tissue material is often difficult and test results are inconclusive.

Methods: Genetic testing was performed on DNA extracted from FFPE tissue from 211 prostate cancer patients using next-generation sequencing (NGS). Sequencing was run on Ion S5 (ThermoFisher) or Miniseq (Illumina) systems.

Results: Within the analyzed patient group 10 (6%) cancer samples were harboring a pathogenic variant in BRCA1/2 gene (one of which was carrier of two mutations): BRCA1 (1%) - c.5289dup, c.181T>G, c.3629_3630del; BRCA2 (5%) - c.5471dup, c.4176del, c.7251_7252del, c.9118-2A > G, c.9154C>T, c.2095C>T, c.3847_3848del, c.5621_5624del. These mutations indicate homologous recombination repair pathway (HRR) deficiency and sensitivity to PARP-inhibitor treatment.

Eligible analysis results could be obtained for 66% of prostate cases. However, tissue material occurred to be challenging and 34% of test results were unsuitable for molecular analysis. The main cause was poor fixation of cancer tissue and insufficient tumor content.

Conclusion: Frequency of BRCA-positive prostate cancers is low and amounts to approximately 6%. Due to high percentage of inconclusive results, some prostate cancer patients might miss the opportunity to qualify for targeted therapy. In this case BRCA testing on liquid biopsy (ctDNA) might be considered. There are no characteristic BRCA1/2 hotspot mutations in prostate cancers, therefore testing of the whole coding sequence is recommended using NGS technology.

Conflict of Interest: None declared

EP01.012 Li-Fraumeni syndrome : pathways of care for cancer prevention versus cancer treatment

marion rolain1, patricia faure1, Nathalie Parodi1, Maud Brauchaud1, Margaux Clement Le Choismier1, Stéphanie Baert-Desurmont2, Edwige Kasper1, Gaelle Bougeard2, Mariette Renaux-Petel3, Thomas Vermeulin4, Théry Jean-Christophe5, Claude Houdayer 1

1Univ Rouen Normandie, Normandie Univ, Inserm U1245, CHU Rouen, Department of Genetics, Rouen, France; 2Univ Rouen Normandie, Normandie Univ, Inserm U1245, Rouen, France; 3CHU Rouen, Department of Child and Adolescent surgery, Rouen, France; 4Centre Henri Becquerel, Department of Medical Information, Rouen, France; 5Univ Rouen Normandie, Normandie Univ, Inserm U1245, Centre Henri Becquerel, Department of Medical Oncology, Rouen, France

Consortium: the PREVENTABLE Consortium

Background/Objectives: Li-Fraumeni syndrome (LFS) represents one of the most severe hereditary predispositions to cancer, given the broad spectrum of tumours involved and their early age of onset. The Rouen genetics department is an expert centre for LFS, part of the European Reference Network for rare tumour risk syndromes (RTRS) GENTURIS. Prevention is a major aspect of health still underestimated by stakeholders and policymakers, which is why we joined the European PREVENTABLE H2022-2025 consortium. The aim of PREVENTABLE is to assess the clinical, social and financial benefits of applying multidisciplinary and specialized care to prevent advanced disease in families suffering from RTRS.

Methods: Based on experience and expertise of our clinical teams, we have identified all possible care pathways for the LFS patients, in order to create standard matrices for children and adults. Then matrices will be filled in with clinical data, lastly prices will be allocated to clinical procedures in order to compare prevention versus treatment in medico-economic terms.

Results: Matrices contained 124 procedures divided into 6 care modules for 58 different pathways of care. All the necessary ethical approvals were obtained, as patients must be informed about the opportunity to object. Thanks to the French LFS network, clinical data could be obtained from other caregivers, and from European centres involved in the project.

Conclusion: Despite the high complexity of LFS care, we demonstrated that consensus matrices are achievable. This project aims to demonstrate the virtuous practice of oncogenetics by highlighting the benefits of prevention in a complex, costly and deadly disease.

Grants: Funded by EU

Conflict of Interest: None declared

EP01.013 Whole exome sequencing shows recurrent abnormalities in members of the WNT pathway in familial polyposis

Paula García-Vallés 1;2;3, Jésica Pérez-García1;2;3, Paloma Martín-Bejarano Soto1;2;3, Rosario Vidal-Tocino1;4, Óscar Javier Blanco Múñez5, Jose Perea1, Ana-Belén Herrero1;2;3, Rogelio González-Sarmiento1;2;3

1Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; 2Molecular Medicine Unit, Department of Medicine, University of Salamanca, Salamanca, Spain; 3Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, Salamanca, Spain; 4Medical Oncology Service, University Hospital of Salamanca, Salamanca, Spain; 5Pathological Anatomy Service, University Hospital of Salamanca, Salamanca, Spain

Background/Objectives: Familial adenomatous polyposis (FAP) makes up about 1% of all colorectal cancers, which in turn are the second cause of cancer death. FAP is characterized by the early development of hundreds or thousands of adenomas throughout the colon. Germline mutations in APC or MUTYH account for the majority of FAP cases. However, there are patients with familial polyposis whose genetic cause is unknown. This project aims to analyze the whole exome of patients with polyposis without germline mutations in APC or MUTYH.

Methods: Two polyps and one healthy tissue sample were analyzed from three patients. Genomic DNA was extracted from formalin-fixed paraffin-embedded (FFPE) polyps and normal adjacent tissues using the QIAamp DNA FFPE tissue kit. The SureSelect Human All Exon V8 kit was used for library preparation and sequencing was performed on NovaSeq 6000 platform. Mutations of interest identified in this study were confirmed through Sanger sequencing.

Results: All the polyps from these patients display pathogenic somatic mutations or variants of uncertain significance in some member of the WNT pathway or the mTOR pathway. Notably, healthy tissues samples already presented alterations in two of the patients, a pathogenic LRP5 mutation in the first patient and a variant of uncertain significance in LGR4 in the second patient. These genes code a WNT pathway co-receptor and receptor, respectively.

Conclusions: Mutations in members of the WNT pathway appear to play a key role in those patients with familial polyposis without germline mutations in APC or MUTYH. However, further studies are required.

Grants: Study funded by Fundación Mutua Madrileña.

Conflict of Interest: None declared

EP01.014 Assessment of EGFR mutation status in cf-DNA from patients with NSCLC in a clinical setting

Maria Michelli 1;1, Eirini Roupou1, Ioanna Giannopoulou1, Niki Prekete1, NIKOLAOS KAVANTZAS1, Angelica a Saetta1

1Medical School of Athens, NKUA, 1st Department of Pathology, Athens, Greece

Background/Objectives: EGFR mutation analysis in circulating cell-free (cf-DNA) tumor DNA from plasma has shown to be a very promising approach for biomarker analysis of NSCLC patients with insufficient tumor tissue and disease monitoring due to several significant advantages (minimally invasive, potential for repeated sampling).

Methods: 595 cf-DNA samples from patients with NSCLC and 158 matched FFPE tumor tissues were analyzed using an IVD test, Cobas ® EGFR mutation v2. 8 samples were re-analyzed using Next Generation Sequencing.

Results: FFPE samples showed insufficient tumor cells in 16/158 cases (10%) and an invalid cobas test in 18/158 (11%). 3/34 total FFPE cases without a test result displayed EGFR mutation in cf-DNA. The presence of mutations in plasma from patients with primary NSCLC reached 9% (25/286). In total 153/595 cf-DNA samples (26%) displayed EGFR mutations. Exon 19 deletions were the most frequent mutations (66%), followed by point mutations in exon 21 (26%). The overall concordance of EGFR mutation status in plasma and tumor samples was 78% and the sensitivity 57%, specificity 100%, PPV 100%. The concordance of EGFR mutation status in plasma between Cobas and NGS was 100%.

Conclusion: EGFR mutation testing in cf-DNA using Cobas IVD test showed significant concordance with analysis of tissue samples and an optimal specificity and PPV. However, the sensitivity of the test has to be further improved indicating that a biopsy should be optimally analyzed for patients with an EGFR mutation-negative cf-DNA test.

Grants: no

Conflict of Interest: Maria Michelli full, Eirini Roupou full, Ioanna Giannopoulou full, Niki Prekete full, NIKOLAOS KAVANTZAS yes, Angelica a Saetta full

EP01.015 Multi-locus Inherited Neoplasia Alleles Syndromes in Cancer: An updated review and implications for clinical practice

Jeanette YUEN 1, Malikka Venkatramani1, Si Qin Zhou1, Jianbang Chiang1, Sock Hoai Chan1, Joanne Ngeow1;2

1National Cancer Centre Singapore, Division of Medical Oncology, Singapore, Singapore; 2Nanyang Technological University, Lee Kong Chian School of Medicine, Singapore, Singapore

Background/Objectives: The popularity of multi-gene panel testing has identified individuals with two or more pathogenic variants (PV). These carriers are at risk of suboptimal treatment, management and inaccurate cancer risk estimations from the lack of data. We reviewed published double heterozygote cases to identify possible predictors and associations with more severe phenotypes to understand the impact on clinical management.

Methods: We conducted a systematic review of published literature tracing double heterozygotes of cancer predisposition genes. Statistical tests were conducted to assess for predictors of double heterozygotes vs. monoallelic and non-carriers. Variables examined were: first age of diagnosis, early age of diagnosis defined as <5% age-stratified risk, number and type of primary cancers, and presence of unassociated phenotype.

Results: We identified 394 cases, with 33 patients from our local service. Results suggest that double heterozygotes presented with significantly more malignancies (32.0% vs 21.5% vs 10.3%), a younger median age of diagnosis (41 vs. 45 vs. 49 years) and an early age of diagnosis (24.9% vs 7.7% vs 4.7%) vs. monoallelic and non-carriers. Carriers with two dominant PVs had more malignancies (34.4% vs. 23.9% vs. 20.0%) and un-associated phenotypes (87.5% vs. 82.1% vs. 40.0%) vs. dominant-recessive and recessive-recessive PV carriers.

Conclusion: Our findings suggest that double heterozygotes have more severe phenotypes, with more than one primary cancer and younger ages of diagnosis. The nature of genes in double heterozygotes (i.e. dominant-dominant / dominant-recessive) may also have implications on cancer risk and clinical management. We propose a framework to evaluate double heterozygotes to guide clinical management.

Grants:

Conflict of Interest: None declared

EP01.016 Genetic variability in inflammation and oxidative stress genes affects gastric cancer risk and clinical features

Pia Pužar Dominkuš 1, Marija Rogar1;2, Vita Dolzan1, Petra Hudler2

1University of Ljubljana, Faculty of Medicine, Institute of Biochemistry and Molecular Genetics, Pharmacogenetics laboratory, Ljubljana, Slovenia; 2University of Ljubljana, Faculty of Medicine, Institute of Biochemistry and Molecular Genetics, Medical Centre for Molecular Biology, Ljubljana, Slovenia

Background/Objectives: Inflammation and oxidative stress are the hallmark of Helicobacter pylori infection, which is a leading risk factor for gastric cancer. The aim of our study was to evaluate the effect of single nucleotide polymorphisms in genes involved in inflammation and oxidative stress on gastric cancer risk, clinical-histopathological features and patient’s survival.

Methods: We enrolled 248 patients with gastric adenocarcinoma and 247 healthy control subjects in the study. Genotyping for two polymorphisms in GSTP1 and KEAP1, three in NFE2L2, and one in CAT, GPX1, IL-1β, TNFα, IL6, SOD2, and IL6R was performed using KASP assays. Logistic regression and Kaplan-Meier method were used for association and survival analysis.

Results: TNFα rs1800629 was significantly associated with higher gastric cancer risk (GG/GA + AA, OR = 1.62, P = 0.011). IL-1β rs1143623 (GG/CG + CC, OR = 0.37, P = 0.017), rs16944 (GG + AG/AA, OR = 0.28, P = 0.027) and IL6 rs1800795 (GG/CG, OR = 0.38, P = 0.010 and GG/CC, OR = 0.16, P = 0.010) were associated with tumour location. NFE2L2 rs6721961 was associated with tumour grade (GG + TG/TT, OR = 0.08, P = 0.007). KEAP1 rs9676881 (GG/GA + AA, OR = 0.36, P = 0.034) and IL-6 rs1800795 (GG + CG/CC, OR = 6.40, P = 0.027) were associated with vascular and perineural invasion, respectively. The CC genotype of IL-6 rs1800795 was associated with longer overall patient survival (P = 0.021).

Conclusion: Our results may be valuable in disease modelling and for the development of personalised approach in the risk assessment and disease management of gastric cancer.

Grants: ARIS grants P1-0170 and P1-0390.

Conflict of Interest: None declared

EP01.017 Telomeres and TERT polymorphisms as biomarkers of the risk of malignant mesothelioma

Tanja Blagus 1, Ana Mervič2, Katja Goricar1, Danijela Strbac3, Alenka Franko2;4, Viljem Kovac2;3, Vita Dolzan1

1University of Ljubljana, Faculty of Medicine, Institute of Biochemistry and Molecular Genetics, Parmacogenetics Laboratory, Ljubljana, Slovenia; 2University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia; 3Institute of Oncology Ljubljana, Ljubljana, Slovenia; 4University Medical Centre Ljubljana, Institute of Occupational, Traffic and Sports Medicine, Ljubljana, Slovenia

Background/Objectives: Malignant mesothelioma (MM), strongly associated with asbestos exposure, belongs to the group of telomerase-dependent cancers. The enzyme complex telomerase is activated due to increased expression of its main component, telomerase reverse transcriptase (TERT). We aimed to determine the associations between telomere length and TERT polymorphisms with the risk of developing MM.

Methods: Our retrospective case-control study, included 302 patients with MM, 386 subjects with pleural plaques (PP), and 86 subjects that were occupationally exposed to asbestos, but had no asbestos related diseases (controls). Relative telomere length was determined by MMQ-PCR, and the presence of TERT rs10069690, rs2736100, and rs2736098 was determined by KASP-PCR. Statistical analysis was performed using nonparametric tests and logistic regression.

Results: MM patients had shorter telomere length than subjects with PP (p < 0.001). Both TERT rs2736098 and rs10069690 were statistically significantly associated with a higher risk of MM in additive and dominant models (ORadd = 1.63; CI = 1.20–2.21; p = 0.002; ORdom = 1.46; CI = 1.09–1.96; p = 0.011; ORadd = 1.52; CI = 1.08–2.12; p = 0.017, ORdom = 2.2; CI = 1.10–4.48; p = 0.026, respectively). On the contrary, the presence of at least one polymorphic TERT rs2736100 allele reduced the risk of MM in additive and dominant models (ORadd = 0.56; CI = 0.35–0.90; p = 0.017; ORdom = 0.66; CI = 0.46–0.95; p = 0.026).

Conclusion: Telomere length and TERT polymorphisms may serve as biological markers of MM development.

Grants: ARIS grants P1-0170 and L3-2622.

Conflict of Interest: None declared

EP01.018 Challenges of PMS2 screening: technical procedures to validate the presence of PMS2 variants.

Maria Isabel Alvarez 1;2, Paula Trilla1, Marta Gracia1, Laura Muñoz1, Maria Pellise3, Belen Pastor Piñera3;4, Lorena Moreno3;4, Teresa Ocaña3;4, Francesc Balaguer3;4, Celia Badenas1;2

1Hospital Clínic de Barcelona, Biochemistry and Molecular Genetics, Barcelona, Spain; 2CIBER of Rare Diseases (CIBERER), Madrid, Spain; 3Hospital Clínic de Barcelona, Department of Gastroenterology, Barcelona, Spain; 4Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain

Background/Objectives: In the era of next generation sequencing technology (NGS), the molecular screening for PMS2 is challenging due to the presence of a large family of pseudogenes that are highly homologous. After routine genetic study we detected 6 patients in which neither long-range PCR nor MLPA could ensure the location of the variant detected. In this work, we present additional strategies to confirm PMS2 alteration.

Methods: Patients 1 to 3 presented an unique deletion of PMS2 exon 14, patient 4 showed a duplication of exons 7 to 11, patient 5 carried a deletion encompassing exons 14 and 15, and patient 6 presented a pathogenic variant in exon 12 in which long-range PCR was not conclusive. For copy number variants specific primers were used in order to confirm their presence by PCR and/or Sanger sequencing. For patient 6, RNA was extracted and cDNA was sequenced.

Results: All patients with single deletion of exon 14 were confirmed using the primers described previously identifying a recurrent deletion of approximately 2kb (c.2276-113_2245+159del). The duplication of exons 7 to 11 was confirmed using the combination of specific primers revealing the presence of an inverted tandem duplication of these exons. In patient 6, RNA assay suggested the presence of a PMS2- PMS2CL hybrid allele. Patient 5 remains under assessment.

Conclusion: All PMS2 variants require addition confirmation with specific approaches. Interestingly, the fact that all 6 patients showed isolated PMS2 loss which indicates that it could represent a highly indicator of a germline pathogenic variant.

Grants:

Conflict of Interest: None declared

EP01.019 Extended genetic testing of KIT/PDGFRA wild-type GISTs revealed NF1-associated GIST in one patient

Alenka Bombač 1, Mojca Unk2, Vida Stegel1, Srdjan Novaković1

1Institute of Oncology Ljubljana, Department of Molecular Diagnostics, Ljubljana, Slovenia; 2Institute of Oncology Ljubljana, Division of Medical Oncology, Ljubljana, Slovenia

Background/Objectives: Gastrointestinal stromal tumors (GISTs) represent a heterogeneous group of mesenchymal neoplasms with a spectrum of genetic alterations. Among these alterations, the association between GISTs and neurofibromatosis type 1 (NF1) mutation has garnered significant attention. NF1 mutation-related GISTs present distinct clinical and molecular characteristics compared to their counterparts. This abstract review a case of NF1-associated GIST from a larger study of 116 metastatic GIST patients, treated at Institute of Oncology Ljubljana.

Methods: Patient’s DNA was extracted from Formalin-fixed, paraffin-embedded (FFPE) primary tissue sample that was obtained prior to the imatinib treatment. Tumor genotyping was performed using Sanger and NGS sequencing with Illumina’s TruSight Oncology 500 DNA Kit. The expression of succinate dehydrogenase (SDH) complex was assessed by immunohistochemistry.

Results: From the initial study cohort of 116 patients twelve out of sixteen KIT/PDGFRA wild-type GISTs retained SDH expression. Additional NGS genotyping of KIT/PDGFRA wild-type GISTs revealed mutations in KIT or PDGRFA in seven patients and NF1 mutation in one patient with no prior association with Neurofibromatosis Type 1 syndrome.

Conclusion: For patients with KIT/PDGFRA wild-type GIST the reliability of direct Sanger sequencing results is insufficient therefore, it is recommended that NGS becomes a necessity for their treatment decision by its ability to detect low allele frequency mutations and potential other targetable alterations as well as identify germline mutations in genes associated with hereditary syndromes.

References: PMID: 35904169

Conflict of Interest: None declared

EP01.020 Germline genetic profiling using next-generation sequencing technology in a man with advanced prostate cancer: a case report

Anca Pavel 1, Petruta Gurban1, Andreea Tutulan-Cunita1, Danai Stambouli1

1Cytogenomic Medical Laboratory, Molecular Genetics, Bucharest, Romania

Background/Objectives: Recent developments in next-generation sequencing (NGS) technologies have contributed to our growing understanding of the prostate cancer (PC) biology, revealing the genetic foundation of the disease’s clinical variability. The aim of the current study was to identify potential genetic risk variants for PC using whole exome sequencing (WES) and to evaluate the effect of these variations on PC aggressiveness.

Methods: We present a case of advanced PC for which WES analysis was performed for the identification of potential genetic variants involved in PC pathology. To isolate the variants significant to our case, a PC susceptibility gene panel comprised of 164 genes was used. The obtained variants were then subjected to further analysis. A brief frequency data analysis was conducted. To assess the possible contribution of genetic variants to the PC disease, genotype-phenotype analysis was performed.

Results: WES revealed 291 nonsense and missense variants in 113 different genes, previously associated with PC. The results of frequency data analysis showed that most susceptibility genes identified were located on chromosomes 2 and 11 (24%). The most frequent variants were single nucleotide variants (SNV) (89.3%). Genotype-phenotype correlation analysis highlighted four variants that had a strong contribution to the PC phenotype, namely c.8965C>G in ATM gene, c.1385G>A in RNASEL gene, c.589G>A in TMPRSS2 gene and c.1501G>A in ELAC2 gene.

Conclusion: Our results suggest that genetic variants play an important role in the progression of PC and their confirmation could have future implications in PC diagnosis and prognosis, as well as therapy.

Grants: -

Conflict of Interest: None declared

EP01.021 Downregulation of SLC22A1 expression in hepatocellular carcinoma and its prognostic significance

Hubert Chen 1

1Vanderbilt University, College of Arts and Science, Nashville, United States

Background/Objectives: Hepatocellular carcinoma (HCC) is the most common primary liver malignancy that occurs predominantly in patients with underlying chronic liver disease and cirrhosis. Organic cation transporter 1(OCT1), encoded by SLC22A1 gene, is mainly expressed in the liver and is located at the basolateral membrane of hepatocytes. It plays a key role in the disposition and hepatic clearance of mostly cationic drugs and endogenous compounds. We aimed to systematically analyze SLC22A1expression and its prognostic role in HCC using various open databases.

Methods: Comparison of HCC and matched normal tissue gene expression was performed with the UALCAN and GEPIA online graphical user interfaces. Correlation between SLC22A1 gene expression and patient survival was evaluated with OncoLnc. SLC22A1 genetic alterations in HCC were also explored using cBioPortal.

Results: Expression of SLC22A1 is significantly down-regulated in the clinic-pathological characteristics (cancer stages, tumor grade, nodal metastasis status, and histological subtypes) examined in HCC patients compared to normal counterparts. Clinically, low expression of SLC22A1 was correlated with shorter overall survival in HCC patients (Log rank p-value = 0.00007). Analysis of the TCGA Liver Hepatocellular Carcinoma dataset (TCGA’s Pan-Cancer Atlas) revealed a low amplification (0.57%) and deep deletion (1.71%) frequency in HCC.

Conclusion: These observations indicate that SLC22A1 may function as a potential tumor suppressor gene. In HCC patients, SLC22A1 expression levels may serve as a prognostic predictive marker.

Grants: NA

Conflict of Interest: None declared

EP01.024 Hormone-induced intracellular interactions of androgen and glucocorticoid receptors

Nuria Arroyo-Garrapucho 1;2;3;4, Vera Sommers4, Hanne Willems4, Kaat Peperstraete4, Vanessa Dubois5, Ana-Belén Herrero1;2;3, Rogelio González-Sarmiento1;2;3, Frank Claessens4

1Biomedical Research Institute of Salamanca (IBSAL), University Hospital of Salamanca-USAL-CSIC, Salamanca, Spain; 2Molecular Medicine Unit, Department of Medicine, University of Salamanca, Salamanca, Spain; 3Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, Salamanca, Spain; 4Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; 5Basic and Translational Endocrinology (BaTE), Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium

Background/Objectives: Androgens are crucial for male sexual development, while glucocorticoids regulate energy homeostasis and immune responses. These hormones activate the androgen receptor (AR) and glucocorticoid receptor (GR). Despite tissue-specific crosstalk between glucocorticoids and androgens, the molecular mechanisms remain unclear. We explored AR and GR cooperation in mouse Fkbp5 transcription, a well-known gene responsive to both hormones, testing the hypothesis that this collaboration occurs via the FKBP5 androgen response element (ARE), which would reveal protein-protein interactions between the receptors. Enzalutamide effect, an AR antagonist, was tested on these interactions.

Methods: Glucocorticoids and androgens were tested in chemically castrated mice, supplemented with one or both steroids. Tissue qPCR was conducted. AR:AR, GR:GR, and AR:GR dimerization were studied using the NanoBiT system in HEK293T. Transcriptional activation via FKBP5 ARE was assessed through a luciferase reporter assay. Statistical significance was determined using Friedman test with Dunn multiple comparisons.

Results: In male mice, Fkbp5 responded to androgens and glucocorticoids in the gastrocnemius and adipose tissue. Treatment with both hormones increased expression levels in these tissues. NanoBiT confirmed rapid AR dimerization with dihydrotestosterone, while dexamethasone failed to induce detectable GR dimers. Combining DHT and dexamethasone resulted in AR:GR heterodimerization. AR and GR activation also additively induced the FKBP5-ARE-luciferase reporter gene. Enzalutamide effectively suppressed both AR:AR and AR:GR dimerization, while inhibiting AR- and GR-dependent FKBP5-ARE activation.

Conclusion: Our study suggests that AR:GR interaction could be involved in the interplay between androgens and glucocorticoids, which could be highly relevant during treatment of prostate cancer with AR inhibitors and glucocorticoids.

Grants: FIS-FEDER PI20/01569, FWO-1131720N, GOA/19/100.

Conflict of Interest: None declared

EP01.025 Germline long insertions in BRCA1/PALB2 exon revealed by PCR-free short-read and long-read whole-genome sequencing

Ava Kwong1;2;3, Chun Hang Au 4, Dona N Ho4, Elaine YL Wong4, Henry CM Leung4, Amy WS Leung4, Janet HY Law4, Fian BF Law4, Sze Keong Tey5, Cecilia YS Ho4, Edmond SK Ma3;4

1The University of Hong Kong and University of Hong Kong-Shenzhen Hospital, Department of Surgery, Hong Kong; 2Hong Kong Sanatorium & Hospital, Department of Surgery, Hong Kong; 3Hong Kong Hereditary Breast Cancer Family Registry, Hong Kong; 4Hong Kong Sanatorium & Hospital, Molecular Pathology Division, Department of Pathology, Hong Kong; 5The University of Hong Kong, Department of Surgery, Hong Kong

Background/Objectives: While to detect structural variants disrupting hereditary breast cancer genes is important, the detection of long insertion (>100 bp) is challenging.

Methods: Short-read PCR-free whole-genome sequencing (WGS) libraries were constructed using KAPA EvoPlus Kit (Roche), sequenced at 2x151 bp on NovaSeq 6000 S1 flow cells (Illumina), followed by alignment using Clara Parabricks v4.0.0 (Nvidia) and insertion detection using INSurVeyor v1.1.2. Long-read PCR-free WGS libraries were constructed using Ligation Sequencing Kit V14, sequenced on PromethION R10.4.1 flow cells (Oxford Nanopore), followed by Dorado v4.2.0 super-accurate basecalling, alignment using minimap2 v2.24 and consensus sequence generation using Flye v2.9.

Results: Under the hereditary breast cancer genetic testing program of Hong Kong Hereditary Breast Cancer Family Registry, we identified germline long insertions from two probands. Proband 1 was a Chinese female bilateral breast cancer patient with positive family history. A heterozygous 318 bp insertion with AluY and target site duplication at PALB2 exon 11 was identified by both short-read and long-read WGS. Proband 2 was a Chinese female early-onset triple-negative breast cancer patient with positive family history. An insertion at BRCA1 exon 24 was suspected by short-read WGS and fully elucidated by long-read WGS as a heterozygous complex deletion of 12 bp and insertion of 12,423 bp. It is among the longest insertions reported in hereditary breast cancer genes as deciphered by WGS.

Conclusion: Detection of the germline insertions enabled better-informed patient management and family member testing.

Grants: Dr. Ellen Li Charitable Foundation; Kerry Kuok Foundation; Hong Kong Hereditary Breast Cancer Family Registry

Conflict of Interest: None declared

EP01.026 Two cases of PTEN Hamartoma Tumor Syndrome caused by novel variants in the PTEN gene

Trayan Delchev 1, Tsvetina Veleva1, Maria Sredkova-Ruskova1, Nevyana Ivanova2, Kalina Mihova2, Radka Kaneva2, Daniela Avdjieva-Tzavella1

1University Children Hospital - SBALDB, Clinical Geneitcs, Sofia, Bulgaria; 2Molecular Medicine Center, Genome Diagsnostics Laboratory, Sofia, Bulgaria

Background: PTEN hamartoma tumor syndrome (PHTS) includes Cowden syndrome (CS), Bannayan-Riley-Ruvalcaba syndrome (BRRS), PTEN-related Proteus syndrome (PS), and PTEN-related Proteus-like syndrome.

PTEN (Phosphatase And Tensin Homolog) is a multi-functional tumor suppressor that is very commonly lost in human cancer. The protein encoded by this gene is a phosphatidylinositol-3,4,5-trisphosphate 3-phosphatase. It contains a tensin like domain as well as a catalytic domain. Unlike most of the protein tyrosine phosphatases, this protein preferentially dephosphorylates phosphoinositide substrates. It negatively regulates intracellular levels of phosphatidylinositol-3,4,5-trisphosphate and functions as a tumor suppressor by negatively regulating AKT/PKB signaling pathway.

We present two cases of PHTS. Case A – a 17-year-old female patient with macrocephaly, psychomotor regress, epileptic seizures, polymicrogyria and atypical autism. Case B, a 1-year-old male, who presented with psychomotor delay, muscle hypotonia, failure to thrive and macrocephaly.

Methods: Conventional cytogenetic assay, MLPA for microdeletions, subtelomeric deletions and duplications, as well as metabolic screening showed normal results in both cases. Subsequently we employed Whole Exome Sequencing in case A and B.

Results: In case A we found a novel, heterozygous, likely pathogenic variant c.158T>C (p.Val53Ala) in PTEN.

In case B we established a novel, heterozygous, likely pathogenic variant c.755A>G p.(Asp252Gly) in PTEN.

Conclusion: Two novel PTEN variants were found in two unrelated patients, who both manifested features of Cowden syndrome. These findings reaffirm the genetic and phenotypic variability of PHTS and expand our understanding of this rare condition.

Grants: N/A

Conflict of Interest: None declared

EP01.027 Gene expression profiling in porocarcinoma indicates heterogeneous tumour development and substantiates poromas as precursor lesions

Svenja Holst1, Friedegund Meier2, Jörg Otte3, Patrick Petzsch4, Julia Reifenberger5, Thorsten Wachtmeister4, Dana Westphal2, Mirjana Ziemer6, Wasko Wruck3, James Adjaye3, Regina C. Betz7, Harald Surowy1, Arno Ruetten8, Silke Redler 1

1University Hospital Düsseldorf, Institute of Human Genetics, Düsseldorf, Germany; 2University Hospital Dresden GmbH, Department of Dermatology, Dresden, Germany; 3University Hospital Düsseldorf, Institute for Stem Cell Research and Regenerative Medicine, Düsseldorf, Germany; 4Heinrich Heine University Düsseldorf, Biological and Medical Research Centre (BMFZ), Düsseldorf, Germany; 5University Hospital Düsseldorf, Department of Dermatology, Düsseldorf; 6University Medical Center Leipzig, Department of Dermatology, Leipzig; 7University of Bonn, Institute of Human, Bonn, Germany; 8Dermatopathology, Bodensee, Dermatopathology, Bodensee, Friedrichshafen

Background: Malignant sweat gland tumours are rare, with the most common form being eccrine porocarcinoma (EP). Its benign counterpart is the eccrine poroma (EPO). Approximately 18% of EPO cases transit to EP, suggesting that EPOs represent a transitional state in EP development.

Objectives: Generate in depth insights into EP biology via comprehensive analysis of tumour-specific gene expression properties. Generate insights into potential drivers of malignant transformation from EPO to EP.

Methods: Transcriptome profiling of 23 FFPE samples of primary EP and available SHT. Genome-wide transcriptome analysis in an exploratory cohort of 10 EPs. Validation via qPCR in an independent cohort of the remaining 13 primary EPs. All findings from the EP samples were then tested in a cohort of 17 EPOs.

Results: Transcriptome profiling revealed an overall diversity in gene expression, and indicated the existence of biologically heterogeneous sub-entities. Hierarchical clustering identified two key matrisomal clusters, which probably represent two major EP tumour subtypes. In both clusters, widespread gene downregulation was observed in EP tumours compared to SHT. Expression levels of CD74, NDGR1, SRRM2, ANXA2, and NOP53 showed a stepwise-reduction in expression from SHT via EPO to EP, thus supporting the hypothesis that EPO represents a transitional state in EP development. Besides providing further evidence implicating the p53 axis and the EGFR pathway, our results suggest new mechanisms of tumorigenesis, including the VEGF/VEGFR axis and the Endothelin pathway.

Conclusions: Larger samples are warranted to generate deeper insights into intra- and inter tumour heterogeneity.

Conflict of Interest: Svenja Holst: None declared, Friedegund Meier: None declared, Jörg Otte: None declared, Patrick Petzsch: None declared, Julia Reifenberger: None declared, Thorsten Wachtmeister: None declared, Dana Westphal: None declared, Mirjana Ziemer: None declared, Wasko Wruck: None declared, James Adjaye: None declared, Regina C. Betz: None declared, Harald Surowy: None declared, Arno Ruetten: None declared, Silke Redler Wilhelm Sander-Stiftung (Nr. 2015.042.1)

EP01.028 Blocking LSD1 and GCN5 enhances retinoid therapy in high RARA AML

Faezeh Ghazvini Zadegan 1, Franziska Fiedler1, Tino Schenk1

1Jena University Hospital, Jena, Germany

Objectives: Acute Myeloid Leukemia (AML) is characterized by a blockage in myeloid cell differentiation, primarily regulated by retinoic acid receptor transcription factors, including RARA, RARB, and RARG. While RARA is necessary for the induction of granulocytic differentiation, RARG plays a central role in maintaining the self-renewal of hematopoietic stem cells. To prevent the activation of RARG by ATRA, specific RARA-agonists, such as Am80, have been developed. Our results indicate that substituting ATRA with Am80, which specifically targets RARA, produces remarkably similar outcomes.

Methods: A promising approach involves using Am80, a specific RARA agonist, in combination with LSD1 and GCN5 inhibitors. The study conducted experiments on both AML cell lines and primary AML samples to investigate the effects of ATRA, Am80, and the inhibitors on myeloid differentiation and treatment response.

Results: The findings revealed that AML cell lines with higher RARA expression showed improved myeloid differentiation, including increased CD11b expression and reduced CD117 levels, with ATRA or Am80, and combining these retinoids with LSD1 and GCN5 inhibitors enhanced the response. In primary AML samples, a significant proportion exhibited positive responses, correlating with higher RARA levels, indicating better outcomes. The research underscored the importance of low LSD1 and GCN5 levels for RARA activity and myeloid differentiation.

Conclusion: The combination of Am80, LSD1 inhibitors, and GCN5 inhibitors holds promise in overcoming ATRA resistance in non-APL AML, particularly in cases with high RARA expression and low levels of LSD1 and GCN5. This approach presents potential for improved treatment responses, necessitating further clinical investigation. Grant: SCHE1909/2–3.

Conflict of Interest: Faezeh Ghazvini Zadegan SCHE1909/2–3, Franziska Fiedler: None declared, Tino Schenk SCHE1909/2–3, Universitätsklinikum Jena

EP01.030 BRCA1 mutations in family members with breast cancer history

Merita Xhetani 1, Albina Hasa2, Blerta Laze3

1University of Tirana, Faculty of Natural Sciences, Department of Biology and Center for Molecular Diagnostics and Genetic Research, Tirana, Albania; 2University of Tirana, Faculty of Natural Sciences, Department of Biology, Tirana, Albania; 3University of Vlora “Ismail Qemali”, Department of Biology, Vlora, Albania

Consortium: NanoAlb

Background/Objectives: Mutations in BRCA1 and BRCA2 genes are considered with a potential increased risk for developing early-onset breast cancer and familial ovarian cancer. The incidence of early-onset breast cancer depends largely on the type of mutation that causes it. This type of breast cancer follows an autosomal dominant inheritance pattern and tends to present as an early, high-intensity, bilateral form of the disease. We aim to investigate the BRCA 1 gene in a family with a breast cancer history.

Methods: Here we present a family study of 6 members with breast cancer history for detection of deletions or duplications in the human BRCA1 gene in genomic DNA isolated from human peripheral whole blood specimens. Copy Number Variations (CNVs) were detected by MLPA using P002 BRCA1 probemix and confirmation test with P087. Coffalyser.Net software was used for fragment analysis.

Results: We detected in all family members the heterozygous 40 nt deletion in BRCA1 exon 11. Two women age 32 and 35 yo (siblings) both without breast cancer clinical outcome, one women is 68 yo (mother) with early-onset breast cancer, her sister 58 years old diagnosed with breast cancer, mastectomy performed. Two other women (age 28 and 33 yo) first degree relatives, were diagnosed with breast cancer, has also heterozygous 40 nt deletion in BRCA1 exon 11.

Conclusions: Analysis of BRCA1 cancer families revealed a correlation between the mutation site and the relative risk of breast cancer development. We highlight the importance of screening for BRCA genes mutations in hereditary breast cancer.

Conflict of Interest: None declared

EP01.033 Advanced molecular diagnosis for type X adenomatous polyposis.

Beatriz Hidalgo Calero 1, José Manuel Sánchez Zapardiel1, Sonia Rodriguez Novoa2, Montserrat de Miguel1, Ricardo Blázquez Martín2, Marina Ibáñez Vizcaíno1, Francisca Luengo Sainz de Baranda1, Victoria Carrero Blázquez1, Ainhoa Almeida Santiago2, Anna Esteve-Codina3, Luis Robles Díaz4

1Hospital Universitario 12 de Octubre, Hereditary cancer laboratory, Madrid, Spain; 2Instituto de investigación sanitaria hospital 12 de Octubre (imas12), Hereditary cancer laboratory, Madrid, Spain; 3Centre Nacional d’Analisi Genòmica (CNAG), Barcelona, Spain; 4Hospital Universitario 12 de Octubre, Medical Oncology, Madrid, Spain

Background/Objectives: Adenomatous polyposis (AP) are diseases that predispose to develop tens to hundreds colorectal adenomatous polyps and thereby increase the risk of colorectal cancer. Many cases within hereditary cancer syndromes, are explained by the presence of deleterous variants in genes such as APC and MUTYH, among others. AP without identified mutation are called X adenomatous polyposis (XAP).

We performed multigenic NGS panel, targeting several genes involved in AP in 40 XAP cases. The unresolved XAP cases will be studied through a RNAseq approach.

The present Project will evaluate the relevance of implementing advanced molecular diagnosis through multigenic panels and RNAseq transcriptome sequencing in XAP cases.

Methods: NGS was performed using the Hereditary Cancer Solution kit (Sophia Genetics) and sequenced on the NextSeq platform (Illumina). Results were analyzed using Sophia-DDM software.

Detection of aberrant gene expression, aberrant splicing and monoallelic expression events in RNA sequencing data was carried out following DROP protocol. Also, fusion detection was carried out with starfusion

Results:

ID

Gene

Transcript

Zigosity

Variant

1 *

MUTYH

NM_001128425.1

Homozygous

c.1187G>A, p.(Gly396Asp)

2

MLH3

NM_001040108.1

Homozygous

c.3827+1G > A

3

MLH3

NM_001040108.1

Homozygous

c.3571-1G > T

4

AXIN2

NM_004655.3

Heterozygous

c.1092dupC, p.(Val365Argfs*9)

  1. * Also carrier of pathogenic variant in heterozygosity in the MSH6 gene: c.2932C>T, p.(Gln978*)

Conclusion: The use of multigenic panels in the study of XAP can improve diagnostic performance. RNAseq analysis as a second-line studies can help in the interpretation and classification of variants.

Grants: Project “PI19/00340” to L.R., funded by Instituto de Salud Carlos III (ISCIII) and co-funded by the European Union.

Conflict of Interest: None declared

EP01.034 Identification of Pathogenic BRCA1/BRCA2 Variants in Hereditary Breast Cancer Patients: Initial In-House NGS Results from the Northern Region of Morocco

Jaouhara MAAMAR 1;2, Rajaa Chahboune1, Ouafae Kaissi2, Hasna Hamdaoui2, Badreddine Nouadi2, Fatima Zahra Laarabi1;2, Houda Jelti1;2, Sabrine Bouressas1;2, Abdelhaq Lamaibdel1;2, Oussama Kettani2, Rabie Rahhali3, Nabila Sellal4;5, Mounia Amzerin3;4, Mohamed El Hfid4;5, Fatima Zahra El m’rabet3;4, Afaf Lamzouri1;2

1Life and Health Sciences Laboratory, Faculty of Medicine and Pharmacy of Tangier, Abdelmalek Essaâdi University, Morocco; 2Department of Medical Genetics and Oncogenetics, Mohammed VI University Hospital, Tangier, Morocco; 3Department of Medical Oncology, Mohammed VI University Hospital, Tangier, Morocco; 4Faculty of Medicine and Pharmacy of Tangier, Abdelmalek Essaâdi University, Morocco; 5Department of Radiotherapy, Mohammed VI University Hospital, Tangier, Morocco

Background/Objectives: Hereditary breast cancer (HBC) is characterized by the notable likelihood of being inherited across generations. Next-generation sequencing (NGS) has become an essential tool in the identification of pathogenic mutations within the responsible genes. The majority of HBC cases are associated with genetic mutations in the BRCA1/BRCA2 genes. With the establishment of the NGS platform in the northern region of Morocco, our study focuses on evaluating the mutational status of these two genes in women from this area, who present with suspected hereditary predisposition to breast cancer.

Methods: To achieve this objective, a targeted NGS panel focusing on BRCA1/2 genes was applied. This study was conducted with a cohort of eight patients from the northern region of Morocco, all of whom had undergone oncogenetic consultation. These women had been diagnosed with breast cancer and were deemed to be at significant hereditary risk.

Results: Among the 8 patients with HBC, 3 had pathogenic variants detected in BRCA1/2 genes (more than one-third). The identified mutations included c.798_799del (p.Ser267fs) and c.5309G>T (p.Gly1770Val) in the BRCA1 gene, as well as the c.1310_1313del (p.Lys437fs) mutation in the BRCA2 gene.

Conclusion: Our study emphasizes the high prevalence of BRCA mutations among patients with HBC, underscoring the necessity to establish at least one NGS platform in the Northern region of Morocco. This approach facilitates personalized management for these patients and enables presymptomatic diagnosis for at-risk family members.

Conflict of Interest: None declared

EP01.035 Pediatric cancer predisposition syndromes (pCPS) in an acute lymphoblastic leukemia cohort with no familial cancer history

Clara Venegas 1;2;3, Elisa Izquierdo1;2;3, Maria Pilar Casares1, Beatriz Ruz1;2;3, Alicia de Pablo1, Elixabet Lopez-Lopez4, Angela Gutierrez-Camino5, Antonio Pérez2;3;6, Alvaro Martinez1, ADELA ESCUDERO1;2;3

1Institute of Medical and Molecular Genetics (INGEMM), La Paz University Hospital, Madrid, Spain; 2Hospital La Paz Institute for Health Research - IdiPAZ, Madrid, Spain; 3Spanish National Cancer Research Centre (CNIO), Madrid, Spain; 4IIS Biobizkaia, Biochemistry & Molecular Biology, Barakaldo, Spain; 5IIS Biobizkaia, Pediatric Oncology, Barakaldo, Spain; 6Paediatric Haemato-Oncology Department, La Paz University Hospital, Madrid, Spain

Background/Objectives: Acute leukemia is the most frequent neoplasm in childhood. Although its etiology is largely unknown, genetic factors predisposing to childhood acute leukemia development have been described in 4-5% of patients. The identification of these pediatric cancer predisposition syndromes (pCPS) is crucial for the correct patient management and follow-up; however, most of them do not have recognizable clinical signs leading to underdiagnosis. This study aims to improve the diagnosis of pCPS associated with acute lymphoblastic leukemia (ALL) using a custom NGS panel.

Methods: Three hundred and fifteen patients diagnosed with ALL between 2014 and 2023, lacking familial cancer history, were sequenced using a hybridization customized NGS panel (Mut4Child) targeting 352 genes associated with pCPS. Bioinformatics analysis, classification, and interpretation of SNVs, indels and CNVs was performed in 264 patients using a tailor-made pipeline, VarSeq tool (Golden Helix), and the ACMG guidelines.

Results: The findings revealed 13 germline SNVs/CNVs classified as pathogenic or likely pathogenic (4.9%) in PAX5, TP53, MLH1, BRCA1, BRCA2, CHEK2, TSC2, BARD1 genes. These results allowed the diagnosis of 9 pCPS (3.4%), one of them predisposing exclusively to leukemia and the others to both leukemia and solid tumors.

Conclusion: These findings facilitated genetic counseling and individualized follow-up for patients based on their genetic diagnosis. Importantly, this analysis demonstrated the feasibility of implementing an NGS custom panel and in-house bioinformatics pipelines for the diagnosis of pCPS associated with ALL in routine clinical practice and highlights the relevance of the clinical management and follow-up of patients and their families.

Grants: Cris-Cancer and PMP21/00073.

Conflict of Interest: None declared

EP01.036 Genomic exploration of small renal masses in Lithuanian cohort

Ieva Vaicekauskaitė1;2, Raimonda Kubiliūtė1;2, Kristina Žukauskaitė1;2, Algirdas Žalimas1;2, Albertas Ulys1, Rasa Sabaliauskaite1;2, Sonata Jarmalaite 1;2

1National Cancer Institute, Vilnius, Lithuania; 2Vilnius University Life Sciences Centre, Institute of Biosciences, Vilnius, Lithuania

Background/Objectives: Small Renal Masses (SRMs) are a heterogeneous group of kidney lesions made up of about 20% of benign lesions, and 80% renal cell carcinoma (RCC) cases. 25% of RCCs are aggressive tumors. Currently, there are no reliable biomarkers for SRMs. The aim of the study was to explore the genomic landscape of SRMs to identify possible diagnostic and prognostic genetic biomarkers.

Methods: 52 SRMs biopsies (38 RCC and 14 benign) collected at the National Cancer Institute of Lithuania were first analyzed for common CHEK2 mutations by qPCR and then by targeted-next generation sequencing encompassing an expanded hotspot mutation gene panel made up of 50 commonly altered genes in cancer. The NGS was conducted on the Ion Torrent platform.

Results: Non-synonymous alterations were detected in 78% of SRMs in 16/51 genes. The presence of mutations listed as pathogenic in ClinVar database correlated with smaller tumor volume (p = 0.02). Mutations in KRAS, VHL, HNF1A, TP53, and ATM genes were predominantly detected in RCC cases rather than benign tumors. Together with BMI and artery hypertension status these mutations were predictive for RCC and rapidly growing tumors (AUC = 0.78 and AUC = 0.63 respectively).

Conclusion: This pilot study of Lithuanian patients with SRMs gave unique insights into the mutational landscape of these tumors. Mutational analysis of SRMs may provide an avenue for detection and prognosis of RCC cases.

Conflict of Interest: None declared

EP01.037 Exploring apoptotic responses and oxidative stress regulation by curcumin and a novel curcumin analogue (B3) in glioblastoma: Insights for therapeutic strategies

varol güler 1, Funda Özkök2, Başak Günçer3, Sacide Pehlivan4

1Institute of Graduate Studies in Health Sciences, Istanbul University, Medical Biology, istanbul, Türkyie; 2Istanbul University-Cerrahpasa, Department of Chemistry, İstanbul, Türkyie; 3Istanbul Faculty of Medicine, Istanbul University, Department of Biophysics, İstanbul, Türkyie; 4Istanbul Faculty of Medicine, Istanbul University, Medical Biology, İstanbul, Türkyie

Background / Objectives: Glioblastoma multiforme (GBM) presents a significant oncological challenge due to its aggressiveness and limited treatment options. This study investigates B3, a curcumin derivative, as a potential antitumor agent against U87 glioblastoma cells. Objectives encompass understanding B3’s molecular mechanisms in influencing cell proliferation, migration, apoptosis, and its synergistic potential with Temozolomide (TMZ).

Materials and Methods: We evaluated the impact of B3 on U87 cells through various assays. Proliferation was assessed using the MTT assay, and the DCFH-DA assay measured reactive oxygen species (ROS) elevation. Apoptosis induction was studied through Acridine Orange/Propidium Iodide dual staining, while migration was analyzed using the wound healing assay. Quantitative real-time PCR was employed to examine changes in RNA expression.

Results: B3 treatment demonstrated a significant inhibition of U87 cell proliferation, accompanied by elevated ROS levels. The compound exhibited anti-migratory effects, triggered apoptosis, and showed synergy with TMZ, rendering U87 cells less resistant. Molecular analysis revealed downregulation of cancer markers (Ki67, c-MYC, CYDD1) and upregulation of the tumor suppressor p53 at the RNA level. Additionally, migration-related genes (NFκB, MMP-2, MMP-9) showed decreased expression in B3-treated cells. Apoptosis pathway activation was confirmed with increased caspase-3 expression and an elevated Bax/Bcl-2 ratio at the RNA level.

Conclusion: B3 shows strong anti-tumor effects, impacting proliferation, migration, and apoptosis. Combined with TMZ, it enhances therapeutic potential. These results highlight B3’s promise for cancer treatment, urging further investigation into its mechanisms for translation to clinical use.

Grants: This study was supported by the Scientific Research Project Coordination Unit of Istanbul University under Grant [number 39220].

Conflict of Interest: None declared

EP01.038 Whole exome sequencing in a family with familial colorectal cancer

Hafdis T Helgadottir 1;2, Pär Lundin3, Annika Lindblom1;2

1Karolinska University Hospital, Clinical genetics and genomics, Stockholm, Sweden; 2Karolinska Institutet, Department of Molecular Medicine and Surgery, Stockholm, Sweden; 3Stockholm University, Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm, Sweden

Background/Objectives: Colorectal cancer (CRC) is a common cancer type with environmental and genetic risk factors. Family history is a strong risk factor where close relatives of CRC patients have an increased risk of CRC. Here, a family with several CRC patients was exome sequenced to identify genetic risk factors contributing to the disease.

Methods: Exome sequencing was performed on a large Swedish family with several individuals with CRC and polyps. In total, 16 family members from three generations were sequenced where seven had CRC. The sequencing data was analysed and downstream filtering was performed to identify candidate variants and genes, where only rare coding variants with severe in silico prediction were included.

Results: Several variants with allele frequency in gnomAD<0.1% and CADD > 20 were found to be shared by at least six of the CRC patients. All variants could be identified in at least one of the other family members, most of whom had been diagnosed with polyps. When less stringent filtering with gnomAD<1% and CADD > 20 was used, additional variants were found to be shared by at least six of the CRC patients. The most notable candidate variant was a frameshift variant in the AKR1B10 gene, but several additional candidate genes were identified.

Conclusion: In the search for a genetic cause for CRC in the family, several high-risk variants were found that could contribute to the disease.

Grants: The Nilsson-Ehle Endowments, Cancerfonden

Conflict of Interest: None declared

EP01.039 The effect of transcriptomic annotations in breast cancer DGE study

Magda Mielczarek 1, Bartosz Czech1, Rafał Stępień1, Joanna Szyda1

1Wrocław University of Environmental and Life Sciences, Wrocław, Poland

Background/Objectives: Gene expression profiling is crucial for understanding breast cancer biology and treatment individualization. The aim of this study was to elucidate transcriptome annotation effect on differential gene expression (DGE) and breast cancer survival prognosis.

Methods: DGE analyses were performed for MCF7 breast cancer (case) and normal tissues (control). The pipeline consisted of quality control, data editing, expression quantification, and DGE analysis. Two quantified transcripts expression outputs were used to apply four approaches defining DGE between (A1) case and control samples quantified based on GRCh37 assembly, (A2) case and control on GRCh38, (A3) case on GRCh37 and case on GRCh38 and (A4) control on GRCh37 and control on GRCh38.

Results: Identical Hallmark pathways resulted in Gene Set Enrichment Analysis for both A1 and A2, except Pancreas beta cells presented in A1 only. The Kyoto Encyclopedia of Genes and Genomes pathways presented only in one approach involved: Melanoma and Prostate cancer (A1) and ABC transporters, Acute myeloid leukaemia, Glycerophospholipid and retinol metabolism, Hedgehog and p53 signalling (A2). PCA determined that the greatest variability (97%) was found between cancer and normal samples (A1, A2) and GRCh37 and GRCh38 annotations (A3). For A4 the variability determined by the annotations was lower (40%). The difference between the average expression of prognostic genes associated with survival in breast cancer (NADERI) between GRCh37 and GRCh38 was not statistically significant (P-value = 0.91).

Conclusion: The overall DGE outcomes were not identical between GRCh37 and GRCh38 annotations, however, the transcriptome annotation had no effect on genes realted to survival prognosis in breast cancer.

Grants: No grant.

Conflict of Interest: None declared

EP01.041 Comparison of morphological patterns and genetic markers in non-small-cell lung cancer (NSCLC)

Anzhelika Chegodar 1, Natalia Bodunova1, Natalia Oparina1;2, Chinchi Babadzhanova1;3, Larisa Tsapkova1, Nikolay Karnaukhov1, Ainash Altayeva4

1Тhe Loginov Moscow Clinical Scientific Center, Moscow, Russian Federation; 2Petrovsky National Research Centre of Surgery Rams, Medical Genetic, Moscow, Russian Federation; 3City Clinical Oncological Hospital No. 1, Moscow, Russian Federation; 4Peoples’ Friendship University of Russia named after Patrice Lumumba, Moscow, Russian Federation

Background/Objectives: Histological types of NSCLC differ in genesis, genetic characteristics and clinical course, which affects the choice of treatment. We investigated histological types and several genetic markers of NSCLC.

Methods: Were examined FFPE tumor tissue samples of NSCLC from 182 patients. The patients’ ages were between 35 and 88 (median 66). There were 113 males and 69 females in our sample. Antibodies used for IHC studies: ALK(D5F3), PD-L1(SP263, SP142, 22C3), TTF-1(8G7G3/1), CK7 (SP52), p40 (BC28). RT-PCR was used to detect mutations in the EGFR (exons 18,19,20,21) and BRAF (V600E) genes. FISH identified ROS1 rearrangements.

Results: Histological types of NSCLC: adenocarcinoma - 161 (88.5%) cases, squamous cell carcinomas - 18 (10%) cases, NSCLC, NOS - 3 (1.5%) cases.

TTF-1 and CK7 positive adenocarcinomas were detected in 96/125 (77%) cases. All cases of squamous cell carcinomas were p40 positive. 44/121 (36%) adenocarcinomas and 10/121 (8%) squamous cell carcinomas were PD-L1 positive. 4/139 (2.8%) adenocarcinomas were ALK positive.

The BRAF mutation was detected in 5/125 (4%) adenocarcinomas.

42/161 (26%) adenocarcinomas had the EGFR mutation: 26 of Ex19del, 10 of L858R, 2 of Ex20ins, 1 of L861Q. 3 adenocarcinomas had double EGFR mutations: Ex19del and Ex20ins - 1; L861Q and G719X - 1; L858R and T790M - 1. ROS1 rearrangement - 2/132 (1.5%).

Conclusion: The EGFR L858R mutation was detected in lepidic and acinar types of adenocarcinomas only. A rare case of BRAF V600E and EGFR L858R co-mutation was discovered in G2 TTF-1 positive adenocarcinoma. The histological types and molecular genetic markers of NSCLC requires further in-depth study.

Conflict of Interest: None declared

EP01.042 PIK3CA expression as a prognostic factor for bladder cancer

Paulina Slawnikowska1, Tomasz Konecki2, Piotr Kutwin2;3, Zbigniew Jablonowski2, Agnieszka Zmysłowska1, Edyta Borkowska 1

1Medical University of Lodz, Department of Clinical Genetics, Łódź, Poland; 2Medical University of Lodz, Ist Clinic of Urology, Łódź, Poland; 3

Background & Objectives: Bladder cancer is the sixth most common cancer in Poland among men.The PI3K/AKT/mTOR signaling pathway, responsible for cell survival, growth and proliferation is one of the main pathways of intracellular transmission. Its regulation involves the proteins encoded by PIK3CA gene, the altered expression of which may contribute to the development of cancer, including bladder cancer. The aim of the study was to evaluate the expression of the PIK3CA in bladder cancer and to determine its correlations with clinicopathological parameters.

Materials & Methods: Tumor sections from 189 patients (81% of tissues came from patients with non-invasive cases) were examined. The control group consisted of 21 cases of normal urothelium from patients without cancer diagnosis (histopathologically confirmed). Real-time PCR reactions were performed using two TaqMan probes for PIK3CA and GAPDH. Expressions were assessed using the double delta method.

Results: The analysis showed that reduced expression PIK3CA dominates in the non-invasive stage, while in the invasive stage more often overexpression is observed (p = 0,003). There was also a correlation between the malignancy potential and the level of PIK3CA expression in the non-invasive stage (p = 0.01). Moreover, patients with reduced expression had a chance to live longer (p = 0,0003) and also had a greater chance of not having progression (p = 0,026).

Conclusions: Reduced expression of PIK3CA contributes to a milder course of diseases, while increased expression promotes tumor infiltration and corelates with an unfavorable prognosis.

Conflict of Interest: None declared

EP01.043 SEHOP-PENCIL project: Current status of the diagnosis and management of pediatric cancer predisposition syndromes in Spain

Elisa Izquierdo1;2, Clara Venegas1;2, Beatriz Ruz1;2, Antonio Pérez1;3, Ana Patiño4;5, Teresa Imizcoz6, Gorka Alkorta6, Elena Panizo7, Sergio Manresa8, Lucas Moreno Martin Retortillo9;10, Adela Cañete11, Maria Teresa Tormo12, Josep Escriva12, Piedad Alba Pavón13, Angela Gutierrez-Camino13, Olatz Villate13, Manuel Ramirez Orellana14, Ana Chamorro14, Cristina Saiz15, Andrea Vilaplana8, Asbleidy Carolina Torres8, Aroa Soriano8, Hector Salvador16, Laura Martí17, Diana Salinas-Chaparro17, Estela Carrasco López18;19, ADELA ESCUDERO 1;2

1Hospital La Paz Institute for Health Research - IdiPAZ, Translational Research in Pediatric Oncology Hematopoietic Transplantation & Cell Therapy, Madrid, Spain; 2La Paz University Hospital, Institute of Medical and Molecular Genetics (INGEMM), Madrid, Spain; 3La Paz University Hospital, Department of Pediatric Hematology and Oncology, Madrid, Spain; 4University Clinic of Navarra, Department of Pediatrics and Medical Genomics Unit, Pamplona, Spain; 5Center for Applied Medical Research and IdiSNA (CIMA), Solid Tumor Program, Pamplona, Spain; 6University of Navarra, CIMA-LAB Diagnostics, Pamplona, Spain; 7University Clinic of Navarra, Department of Pediatrics, Madrid, Spain; 8Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona., Personalised Medicine Programme, Childhood Cancer and Blood Disorders Research Group, Barcelona, Spain; 9Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona., Childhood Cancer and Blood Disorders Research Group, Barcelona, Spain; 10Vall d’Hebron University Hospital, Pediatric Oncology and Hematology Department, Barcelona, Spain; 11Hospital Universitari i Politècnic La Fe, Valencia, Spain; 12Instituto de Investigación Sanitaria La Fe, Valencia, Spain; 13Biobizkaia Health Research Institute, Pediatric Oncology Group, Barakaldo, Spain; 14Hospital Infantil Universitario Niño Jesús, Madrid, Spain; 15Fundación para la Investigación Biomédica del Hospital Infantil Universitario Niño Jesús., Madrid, Spain; 16Hospital Sant Joan de Déu, Pediatric Oncology Unit, Barcelona, Spain; 17Hospital Sant Joan de Déu, Genetic Unit, Barcelona, Spain; 18Vall d’Hebron Institute of Oncology, Herediatry Cancer Genetics, Barcelona, Spain; 19Vall d’Hebron University Hospital, Herediatry Cancer Genetics, Barcelona, Spain

Consortium: On behalf of the Group of Pediatric Cancer Predisposition Syndromes of SEHOP-PENCIL study- Personalised mEdicine for Cancer in children in Spain.

Background/Objectives: SEHOP-PENCIL is a nationwide project aiming to improve the genetic characterization of pediatric cancer in Spain. This includes optimizing the diagnosis of pediatric cancer predisposition syndromes (pCPS) caused by germline mutations detected in 10-12% of patients. pCPS identification is crucial to ensure effective patient management and personal and familial genetic counseling. However, the diagnosis of pCPS can be challenging as these patients are very heterogeneous and might not have a recognizable phenotype. This study aims to analyze the current status of pCPS diagnosis and management in Spain in order to improve the current diagnostic rate and to facilitate access to genetic testing and counselling by creating seven reference genomic centers focused on pCPS in Spain.

Methods: A national survey was conducted using the REDCap platform, targeting the oncohematology services of all SEHOP hospitals in Spain between 2022 and 2023.

Results: A total of 78% (35/45) Spanish centers completed the survey. Of these, 66% (23/35) perform germline genetic analyses related to pCPS. Jongmans/Ripperger/MIPOGG genetic testing criteria were followed by only 30% (7/23) of the centers in their clinical practice. The use of NGS panels was the most chosen technique by most centers, 52% (12/23). Surprisingly, less than 50% (11/23) referred patients and families to a genetic counseling unit.

Conclusion: The diagnosis and management of pCPS is already being implemented in some Spanish hospitals; however, it is necessary to standardise patient selection criteria, methodology and genetic counseling in order to offer a robust and uniform pCPS diagnosis across the country.

Grants: PMP21/00073.

Conflict of Interest: None declared

EP01.044 Improving the diagnosis of hereditary retinoblastoma using Mut4Child, a high depth NGS hybridization custom panel

Maribel Soler1, Clara Venegas1;2;3, Beatriz Ruz1;2;3, Borja González1, Virginia Contreras1, Alicia de Pablo1, Núria Rodríguez-Salas1, Dolores Corral1, Sonsoles San Roman1, Jesus Peralta1;2, Susana Noval1;2, Antonio Pérez1;2;3, ADELA ESCUDERO1;2;3, Elisa Izquierdo 1;2;3

1La Paz University Hospital, Madrid, Spain; 2Hospital La Paz Institute for Health Research - IdiPAZ, Madrid, Spain; 3Spanish National Cancer Research Centre (CNIO), Madrid, Spain

Background/Objectives: Retinoblastoma is the most frequent childhood eye malignancy. This tumor type usually appears in the first three years of life, with an overall incidence of 1 in 14000 live births in Spain. Retinoblastoma can occur either as a hereditary or nonhereditary form, depending on whether patients are carriers of a RB1 germline variant. This study includes a cohort of 317 pediatric patients diagnosed of retinoblastoma in a national reference hospital in Spain between 1998 and 2023. Of those, 176/317 (59%) and 130/317 (41%) patients were diagnosed with unilateral and bilateral retinoblastoma, respectively. The average age of unilateral and bilateral patients was 15 and 9 months, respectively (IQR = 17).

Methods: Conventional methods, such as Sanger and MLPA were performed in 238 patients, whereas 79 patients were anlyzed using a NGS hybridization custom panel at high depth (Median: 834X).

Results: Hereditary retinoblastoma was confirmed in 115/238 patients (48.6%), in which a RB1 germline variant was detected (87% SNVs, 13% deletions). Of note, 11/79 (14%) genetic alterations were detected by the custom NGS approach panel in patients who could not had been diagnosed by conventional methods, including two translocations and nine germline mosaic variants.

Conclusion: These results demonstrate the use of a high depth NGS custom panel improved the identification of heritable retinoblastoma. In addition, this retrospective study highlights the need for an accurate RB1 germline variant detection in the routine care of a public hospital to improve diagnosis, management, and outcome of children with retinoblastoma and their families.

Grants: Spanish Association of Retinoblastoma (AER), Cris-Cancer and PMP21/00073

Conflict of Interest: None declared

EP01.045 Lynch syndrome: one disease with different phenotypes or several different diseases?

Laura Roht 1;2, Piret Laidre2, Mikk Tooming1;3, Hanno Roomere3, Kadri Rekker3, Kadri Toome3, Olga Fjodorova3, Ülle Murumets3, Jaan Soplepmann4;5, Katrin Ounap1;2, Tiina Kahre1;3

1University of Tartu, Institute of Clinical Medicine, department of Clinical Genetics, Tartu, Estonia; 2Tartu University Hospital, Genetics and Personalized Medicine Clinic, Clinical Genetics, Tartu, Estonia; 3Tartu University Hospital, Genetics and Personalized Medicine Clinic, Laboratory Genetics, Tartu, Estonia; 4University of Tartu, Institute of Clinical Medicine, department of Hematology and Oncology, Tartu, Estonia; 5Tartu University Hospital, Surgery Clinic, Institute of Clinical Medicine, department of Hematology and Oncology, Tartu, Estonia

Background/Objectives: Lynch syndrome (LS) is the most frequent inherited colorectal cancer syndrome. The main cause of LS is the loss of function of DNA mismatch repair (MMR) genes. There are four key MMR genes: MLH1, MSH2, MSH6, PMS2, and one non-MMR gene (EPCAM). Recently, it has been hypothesized, that LS is not one disease, but encompasses different diseases.

Methods: We collected clinical data of 134 LS variant carriers during twelve years (2012-2023). There were 66 cancer patients and 68 healthy carriers of MMR genes disease-causing variants alone or in seven cases together with EPCAM gene deletion. Individuals were identified by Next-Generation Sequencing, using either Illumina’s TruSight Cancer (94 genes) or Trusight Hereditary Cancer (113 genes) panel. In case of family members, Sanger sequencing was used to detect the variant already known in the family.

Results: We identified 41 MLH1, 27 MSH2, 7 MSH2 + EPCAM deletion, 23 MSH6 and 36 PMS2 disease-causing variant carriers. In case of MLH1, MSH2 alone or together with EPCAM deletion and MSH6 pathogenic variants, colorectal cancer (CRC) or concurrent CRC only or together with other cancer types was the most common phenotype. The mean age of the first cancer diagnosis was 40.2, 49.2, 35.8 and 40.6 years, respectively. PMS2 mutated individuals had either CRC, breast or gynaecological cancers, the mean age of the first cancer was 54.5 years.

Conclusion: The phenotypic diversity of Lynch syndrome stems from the specific gene affected, causing variations in cancer risks, types, and mean ages of diagnosis.

Grants: PRG471

Conflict of Interest: None declared

EP01.046 Combining germline, tissue and liquid biopsy analysis by comprehensive genomic profiling to improve the yield of actionable variants in a real-world cancer cohort.

Irene Vanni1, Lorenza Pastorino1;2, Virginia Andreotti1, Danila Comandini3, Giuseppe Fornarini3, Massimiliano Grassi3, Alberto Puccini3, Enrica Teresa Tanda2;4, Alessandro Pastorino3, Valentino Martelli3, Luca Mastracci5;6, Federica Grillo5;6, Francesco Cabiddu6, Antonio Guadagno6, simona coco7, Eleonora Allavena2, Francesca Barbero1, William Bruno1;2, Bruna Dalmasso1, Sara Erika Bellomo8, Caterina Marchiò8;9, Francesco Spagnolo10;11, Stefania Sciallero3, Enrico Berrino8;9, Paola Ghiorzo 1;2

1IRCCS Ospedale Policlinico San Martino, Genetica dei Tumori Rari, Genoa, Italy; 2University of Genoa, Department of Internal Medicine and Medical Specialties, Genoa, Italy; 3IRCCS Ospedale Policlinico San Martino, Oncologia Medica 1, Genoa, Italy; 4IRCCS Ospedale Policlinico San Martino, Oncologia Medica 2, Genoa, Italy; 5University of Genoa, Dipartimento di Scienze Chirurgiche e Diagnostica Integrata, Genoa, Italy; 6IRCCS Ospedale Policlinico San Martino, Anatomia Patologica, Genoa, Italy; 7IRCCS Ospedale Policlinico San Martino, Tumori Polmonari, Genoa, Italy; 8Oncological Institute Candiolo, FPO-IRCCS, Pathology Unit, Candiolo, Italy; 9University of Turin, Department of Medical Sciences, Torino, Italy; 10IRCCS Ospedale Policlinico San Martino, Oncologia Medica 2; 11University of Genoa, Dipartimento di Scienze Chirurgiche e Diagnostica Integrata

Consortium: none

Background/Objectives: Comprehensive next-generation sequencing is widely used for precision oncology and precision prevention approaches. We aimed to determine the yield of actionable variants, the capacity to uncover hereditary predisposition and liquid biopsy appropriateness instead, or in addition to, tumor tissue analysis, in a real-world cohort of cancer patients, who may benefit the most from comprehensive genomic profiling.

Methods: Twenty-three patients were consecutively enrolled at our center, according to at least one of the following criteria: no available therapeutic options; long responding patients potentially fit for other therapies; rare tumor; suspected hereditary cancer; primary cancer with high metastatic potential; tumor of unknown primary origin. Matched germline/tumour/circulating-free DNA (cfDNA) profiling using the Hereditary Cancer Panel (germline) and the TruSight Oncology 500 panel (tissue/cfDNA) (Illumina) was performed. Variants were mapped by OncoKB and AMP/ASCO/CAP classification.

Results: The overall yield of actionable somatic and germline variants was 57% (13/23 patients), or 43.5%, excluding variants previously identified by routine testing. The accuracy of tumor/cfDNA germline-focused analysis was demonstrated by overlapping results of germline testing. Five germline variants in BRCA1, VHL, CHEK1, ATM genes would have been missed without extended genomic profiling. A previously undetected BRAF V600E mutation was emblematic of the clinical utility of this approach in a patient with a liver undifferentiated embryonal sarcoma responsive to BRAF/MEK inhibition.

Conclusion: Our study confirms the clinical relevance to perform extended parallel tumor DNA and cfDNA testing to broaden therapeutic options and to uncover hereditary predisposition following tumor sequencing in patient care.

Grants: Italian Ministry of Health-IRCCS Ospedale Policlinico San Martino, MUR-PRIN 2022.

Conflict of Interest: None declared

EP01.048 Familial leiomyosarcoma due to a pathogenic TP53 variant

Maria Hellberg 1

1, Department of Clinical Genetics, Pathology, and Molecular Diagnostics, Lund

Background/Objectives: Pathogenic variants in TP53 can cause Li-Fraumeni syndrome which is a condition that confers a high risk of many types of cancer. Sarcoma, breast cancer, adrenal cancer and brain cancer are considered the core cancers. Certain pathogenic variants in TP53 are known to confer a high risk of breast cancer but significantly less risk for other TP53-related cancers. Here we present a family with a cluster of four first degree relatives with leiomyosarcoma and one case of a malignant peripheral nerve sheath tumour (MPNST) but no other known malignancies.

Methods: Information regarding cancer diagnoses in the family was collected from family members. Germline whole genome sequencing was performed with DNA extracted from peripheral blood from one of the individuals with leiomyosarcoma. The data was filtered using a gene list consisting of four genes: FH, NF1, PTEN and TP53.

Results: Four family members with leiomyosarcoma were confirmed from the Swedish National Cancer Register. One of the individuals with leiomyosarcoma had previously been diagnosed with a MPNST. A pathogenic frameshift variant was detected in TP53 c.330del;p.(Leu111Trpfs*12) NM_000546.6 in the bloodsample from the individual with leiomyosarcoma and MPNST.

Conclusion: In this unusual family with four closely related individuals who had leiomyosarcoma but no other TP53-associated cancers a pathogenic TP53-variant was detected.

Conflict of Interest: Maria Hellberg Employed full time at the Department of Clinical Genetics, Pathology, and Molecular Diagnostics in Lund

EP01.049 Development of a targeted RNA sequencing panel to reclassify VUS in cancer predisposing genes

Larissa Dias de Souza1, Alexandre Defelicibus2, Diogo Soares3, José Rocha3, Daniele Paixão Pereira3, Dirce Carraro1, Giovana Torrezan 1

1A.C.Camargo Cancer Center, Clinical and Functional Genomics Group; 2A.C.Camargo Cancer Center, Bioinformatics facility; 3A.C.Camargo Cancer Center, Oncogenetics Department

Genetic tests often yield uncertain results, particularly with the identification of variants of unknown clinical significance (VUS), which poses a dilemma in clinical practice. This study aims to validate a set of computational and experimental tools to prioritize and reclassify predicted spliceogenic or non-coding VUS. Rare exonic and non-coding variants from Brazilian cancer patients who underwent DNA multigene panel testing were selected for this purpose. In silico tools (MaxEntScan, dbscSNV, SpliceAI and UTRannotator) were used for variant prioritization and targeted RNA-sequencing (RNA-cap) of 27 cancer predisposition genes was performed on predicted spliceogenic variants. Raw genetic test data from 2,082 patients revealed 1,954 rare non-benign or unclassified variants; among these 148 are predicted spliceogenic and 32 of impacting the 5’ UTR. We performed RNA-cap sequencing on 37 samples: 14 with Pathogenic/Likely Pathogenic (P/LP) variants, 11 with VUS and 12 negative controls. Splicing profiles were analyzed and compared using the rMATS software. Our findings validated the splicing effect in 12 out of 14 patients with P/LP variants. For the VUS group, only one significant splicing event was detected out of the total cases (10/11), suggesting that most may be downgraded to Benign/Likely Benign variants. This study has established criteria and an automated pipeline for selecting rare variants predicted to alter splicing, which contributes to standardized tools for differential splicing analysis. The anticipated data outcomes are aimed at improving clinical interpretation of hereditary cancer-related variants, thereby enhancing diagnostic accuracy and reducing risks for cancer patients.

Conflict of Interest: None declared

EP01.050 The most frequent germline pathogenic variants in Russian patients with breast cancer: whole genome sequencing results

Maria Makarova 1, Marina Nemtsova1, Maria Byakhova2, Anastasia Danishevich3, Denis Chernevskiy1, Olesya Sagaidak1, Maria Vorontsova4, Maxim Belenikin1, Anastasia Krinitsina1, Aleksey Antonenko1, Anna Semenova2, Natalia Bodunova3, Igor Khatkov3, Vsevolod Galkin2

1EVOGEN LLC, Moscow, Russian Federation; 2City Clinical Oncological Hospital №1, Moscow, Russian Federation; 3Тhe Loginov Moscow Clinical Scientific Center, Moscow, Russian Federation; 4Lomonosov Moscow State University, Moscow, Russian Federation

Objective: To analyze pathogenic variants (PV) in breast cancer patients (BC) and propose a novel target screening panel for individuals with suspected hereditary cancer syndromes (HCS).

Methods: 1514 patients with BC and suspected HCS (patient group, PG) and 5163 individuals without cancer (control group, CG) whole genome sequencing (WGS) were performed.

WGS: 30х, DNBseq-T7, EVOGEN LLC; bioinformatics analysis accelerators: EVA Pro, EVOGEN, Russia; MegaBOLT, MGI, China.

Results: 324 pathogenic variants (PV) in cancer-associated genes in 1514 BC patients were identified (21.4%).

Using PG ang CG WGS results 21 genes were determined for the most effective NGS-panel for suspected hereditary BC in Russia including ATM, BARD1, BRCA1, BRCA2, BRIP1, CDH1, CHEK2, FANCC, FANCM, FANCA, FANCI, FANCL, MLH1, MSH2, MSH6, PMS2, PALB2, PTEN, RAD51C, RAD51D, TP53. A significant association with BC was established for PV in ATM (p < 0.001), BARD1 (p = 0.003), BRCA1 (p < 0.001), BRCA2 (p < 0.001), BRIP1 (p = 0.013), MLH1 (p = 0.010), PALB2 (p < 0.001), TP53 (p < 0.001).

The most frequent PV in the PG were identified (hg38): BRCA1 (NM_007300.4) – c.181T>G, c.1961del, c.5329dup, c.4327C>T, c.5215+1G > T; BRCA2 (NM_000059.4) – c.7007G>A, c.658_659del; ATM (NM_000051.4)c.8147T>C; PALB2 (NM_024675.4) – c.172_175del, c.509_510del. Variants CHEK2 (NM_007194.4) c.444+1G > A, c.1100del, NBN (NM_002485.5) c.657_661del, did not demonstrate significant frequency differences between the PG and CG.

Conclusion: WGS results make it possible to assess the structure and frequency of the PV in BC patients and propose novel screening PCR- and NGS-based target panels.

Grants: Moscow City Health Department financial support.

Conflict of Interest: None declared

EP01.051 Expanding the possibilities for diagnosis, staging, prognosis and follow-up of patients with neuroblastoma through molecular genetic research

Gergana Stancheva 1, Ivan Boronsuzov2, Veronika Petkova1, Kalina Mihova1, Margarita Kamenova3, Prof. Dobrin Konstantinov2;4, Maya Yordanova2;4, Radka Kaneva1

1Medical University of Sofia, Medical Chemistry and Biochemistry, Molecular Medicine Center, Sofia, Bulgaria; 2University Hospital “Queen Johanna-ISUL”, Pediatric Oncohematology Department, Sofia, Bulgaria; 3University Multiprofile Hospital for Active Treatment and Emergency Medicine “N.I.Pirogov”, Department of Pathology, Sofia, Bulgaria; 4Medical University of Sofia, Department of Pediatrics

Background/Objectives: The relevance of the research topic is determined by the high incidence of paediatric solid tumours, their great malignancy potential, the high mortality rates and the possibility to offer the patients reliable therapy when timely and accurate diagnosis is made. Neuroblastoma(NB) is the most common solid extracranial tumor in children with incidence in Europe of 10/1 000 000. Approximately 40% of NB patients have high-risk disease with dissemination in bone marrow(BM), bone, distant lymph nodes, liver, and other organs. PCR-based detection of minimal residual disease(MRD) in NB patients can be used for initial staging and monitoring therapy response in BM and peripheral blood(PB).

Methods: The expression of TH and PHOX2B mRNA was analysed in materials from 11 patients(19BM, 11PB) and 7 healthy people by real-time quantification PCR(RQ-PCR).B2M was used as endogenous control gene.

Results: The expression of TH was confirmed in 14 of 19 BM and in 7 of 11 PB samples. The PHOX2B expression was detected in 15 of 19 BM and in 6 of 11 PB samples. Both BM and PB materials have been collected from nine patients. Combination of TH and PHOX2B expression in BM confirmed dissemination in 89% of the patients. It is interesting to note that in patients with metastatic disease, the studied markers are also found in the PB, which shows the potential of the method as non-invasive and gentle.

Conclusion:The combination of TH and PHOX2B expression has been identified as more sensitive and specific marker for monitoring MRD in NB than the routine investigations.

Grants: MES/Contracts D01-302/17.12.2021,D01-165/28.07.2022;MES,NSF/Contract-KP-06-N63/4-13.12.2022

Conflict of Interest: None declared

EP01.052 Eventual founder effect of a mutation in MLH1 gene within a large Tunisian family with CMMRD

Rasene Gereisha 1, Gabteni SANA1;2, Ahlem Achour1;2, Firas Akrout3, Rym Meddeb1;2, Carli Tops4, Richard Gallon5, Mariem Ben Rekaya6;7, Soumaya Rammeh6;7, Ridha Chkili3, Nada Mansouri8, Neila Belguith1;9, Ridha Mrad1;2

1Charles Nicolle Hospital, Department of Congenital and Hereditary Diseases, Tunis, Tunisia; 2Faculty of Medicine of Tunis, Laboratory of Human Genetics, Tunis, Tunisia; 3Military Hospital of Tunis, Department of Neurosurgery, Tunis, Tunisia; 4Leiden University Medical Center, Department of Clinical Genetics, Leiden, Netherlands; 5Faculty of Medical Sciences, Newcastle University, Translational and Clinical Research Institute, Newcastle upon Tyne, United Kingdom; 6Charles Nicolle Hospital, Department of Pathology, Tunis, Tunisia; 7Faculty of Medicine of Tunis, Research Unit of Onco-theranostic Biomarkers UR17ES15, Tunis, Tunisia; 8Military Hospital of Tunis, Department of Pathology, Tunis, Tunisia; 9Faculty of Medicine of Sfax, Laboratory of Human Molecular Genetics, Tunis, Tunisia

Background/Objectives: Constitutional mismatch repair deficiency (CMMRD) syndrome is a rare autosomal recessive genetic disorder caused by biallelic germline mutations in one of the mismatch repair genes.

Our study explores distinctive clinical, pathological, and genetic aspects of CMMRD, emphasizing the importance of comprehensive management for families meeting CMMRD criteria.

Methods: A 38 years old tunisian patient was referred to our genetic consultation for a suspicion of cancer predisposition syndrome. He presented at the age of 18 a colonic oligopolyposis and adenosquamous carcinoma of the caecum. He later developed an undifferentiated carcinoma of the parotid, an astrocytoma, and an ampulla of Vater adenocarcinoma.

Family history was significant for early-onset colorectal cancer and Lynch syndrome.

A CRC predisposition gene panel including MMR genes was performed.

Results: Molecular analysis identified a homozygous germline missense variant in the MLH1 gene: c.1918C>A; p.(Pro640Thr).

Functional impact assessment, immunohistochemistry and the detection of increased cMSI confirmed its pathogenicity.

The variant was also identified in a heterozygous state in the eldest brother and his non-consanguineous wife who also presented a family history of colorectal cancer. This finding suggests a potential hereditary risk for the couple’s offspring to develop the disease.

The incidence of this variant in Tunisian families with LS and CMMRD could be consistent with a founder effect.

Conclusion: Even though CMMRD syndrome is a rare cause of early-onset malignancy, the diagnosis of CMMRD is crucial for early cancer detection, effective cancer therapy, accurate genetic counseling, and the implementation of targeted preventive surveillance programs.

Grants: None

Conflict of Interest: None declared

EP01.053 Somatic mosaicism for NF1 pathogenic variant detected by peripheral-blood exome sequencing in a patient with colon and lung cancers and no apparent clinical signs of Neurofibromatosis type 1.

Miriam Carriero 1, Ludovico Graziani1, Emanuele Agolini2, Mario Bengala3, Antonio Novelli2, Michela Biancolella4, Giuseppe Novelli1;3

1Tor Vergata University of Rome, Department of Biomedicine and Prevention, Rome, Italy; 2Bambino Gesù Children’s Hospital, Translational Cytogenomics Research Unit, Rome, Italy; 3Tor Vergata University Hospital, Medical Genetics Unit, Rome, Italy; 4Tor Vergata University of Rome, Department of Biology, Rome, Italy

Background/Objectives: Mosaic neurofibromatosis type 1 (MNF1) is an uncommon form of neurocutaneous disorder caused by postzygotic mutation in the neurofibromin gene (NF1). MNF1 is characterized by clinical manifestations typical of neurofibromatosis type 1 (NF1), although localized to one or more body segments. Patients with NF1 pathogenic variants have an increased risk for various types of tumors; however, adenocarcinomas involving the colon or the lungs have rarely been reported.

Methods: A 64-year-old man was referred to genetic counselling for previous synchronous diagnosis of lung and rectal adenocarcinoma. The patient also had multiple colorectal hyperplastic/adenomatous polyps. Physical examination revealed asymptomatic brownish soft papules across the chest. Several other lesions, surgically excised, were reported to first appear after pulmonary lobectomy grouped in the surgical wound region. There was no evidence of typical features of NF1 and no family history of pigment disorders.

Results: Custom phenotype-focused exome sequencing detected an NF1 pathogenic variant (NM_001042492.3: c.61-2A > G) at a frequency of 33% in the blood. Functional mRNA studies in an individual suspected to have NF1 showed that the c.61-2A > G intronic variant causes aberrant splicing and partial deletion of exon 2.

Conclusion: MNF1 phenotypic expression is sometimes subtle and internal malignant tumors may be diagnostic clues to previously undiagnosed MNF1. Molecular studies in paraffin-embedded tumor and skin samples are needed to understand the association between MNF1 and lung/colorectal cancer or polyposis. Regardless, this finding highlights that NF1 screening should be considered in patients suspected of having cancer genetic susceptibility syndrome even in the absence of clinical symptoms of neurocutaneous disorder.

Conflict of Interest: None declared

EP01.055 Quality of life in children with cancer and their carers

Joshua Kraindler 1, Rupendra Shrestha1, Owen Tan1, Jayamala Parmar1, Reanu Gopal1, Natalie Hart1, Vanessa Tyrrell2;3, Tracey O’Brien3;4, Deborah Schofield1

1GenIMPACT: Centre for Economic Impacts of Genomic Medicine, Macquarie University, Australia; 2Children’s Cancer Institute, Kensington, Australia; 3School of Clinical Medicine, UNSW Medicine & Health, Sydney, Australia; 4Kids Cancer Centre, Sydney Children’s Hospital, Randwick, Australia

Background/Objectives: Cancer is the leading cause of disease related death in children. While there have been significant improvements in survival rates for many childhood cancers, similar progress has not occurred in the most aggressive and high-risk cancers, with survival rates of only 30%. Precision medicine brings hope for patients and families. However, to ensure funding is accessible, evidence of cost-effectiveness is needed, which in addition to survival rates, will require data on quality of life measured in health utilities.

Methods: Patients are those with aggressive and high-risk child cancer enrolled in the Zero Childhood Cancer National Clinical Trial in Australia. 105 carers completed the tailored questionnaire, which collected information on QoL and health utilities through the Assessment of Quality of Life (AQoL)-8D and the EQ-5D-Y for children, both validated measures of QoL.

Results: Both EQ-5D-Y values for children and AQoL-8D values for carers were lower than population norms. Bivariate and multivariate regression analysis will also report some factors associated with QoL in children with aggressive and high-risk cancer and for their carers.

Conclusion: Childhood cancer has a substantial impact on both patients and families. Caring for a child with cancer has substantial impacts on the carer’s quality of life. This paper will present QoL data measured in health utilities for both children and carers. These are key inputs for cost-effectiveness of precision medicine for childhood cancer.

Grants: National Health and Medical Research Council (NHMRC) (Partnership Grant ID: 1159004).

Conflict of Interest: None declared

EP01.056 miRNA expression testing could improve presurgical diagnosis of follicular thyroid neoplasm

Deimante Usaite 1, Julija Simiene1, Kestutis Suziedelis1;2

1National Cancer Institute, Laboratory of Molecular Oncology, Vilnius, Lithuania; 2Vilnius University, Department of Biochemistry and Molecular biology, Vilnius, Lithuania

Background/Objectives: Discrimination between follicular thyroid cancer and benign adenoma is the most difficult aspect of thyroid pathology. Only 30% of patients who are reported as Follicular neoplasm by cytopathology and have thyroidectomy, ends up as Follicular carcinoma by postsurgical histopathology. The aim of this study was to identify miRNAs differentially expressed in human thyroid carcinoma line cells established from metastases of a follicular thyroid carcinoma (RO82-w-1), lymph node metastasis (FTC-133) or normal thyroid follicular epithelial cells (Nthy-ori-3-1), cultivated as 3D in vitro cell cultures.

Methods: Triplicates of human thyroid FTC-133, RO82-w-1 and Nthy-ori-3-1 (Sigma-Aldrich, USA) cell line 3D in vitro cultures were used in this analysis. RNA has been extracted and sequenced by NextSeq 500/550. Analysis of differentially expressed miRNA between different cell lines have been performed using R package DESeq2 v1.40.2. Only highly abundant (base mean ≥ 100) with threshold of Bonferroni adjusted p value < 0.01 and absolute log2 fold change (FC) |log2FC | > 3 were selected as differentially expressed miRNAs.

Results: We have identified 44 differiantially expressed miRNAs. 12 of them were downregulated and 2 of them were upregulated in both FTC-133 and RO82-w-1 cell lines if compared to Nthy-ori-3-1. By raising the threshold to |log2FC | > 5, 4 miRNAs were chosen: hsa-miR-10b-3p, hsa-miR-10b-5p, hsa-miR-196b-5p, hsa-miR-126-5p.

Conclusion: We have found that 4 miRNAs were significantly differentially expressed in follicular thyroid cells compared to benign cells. These data suggest that follicular thyroid adenoma can be distinguished from follicular thyroid carcinoma by miRNA expression analysis.

Grants: EU grant no: J05-LVPA-K-01-0122

Conflict of Interest: Deimante Usaite J05-LVPA-K-01-0122, Julija Simiene: None declared, Kestutis Suziedelis: None declared

EP01.058 (Epi)genetic Biomarkers for High-Grade Serous Ovarian Carcinoma

Ieva Vaicekauskaitė1;2, Paulina Kazlauskaitė1;2, Rugilė Gineikaitė1;2, Ruta Ciurliene3, Juozas Rimantas Lazutka2, Rasa Sabaliauskaite 1;2

1National Cancer Institute, Genetic diagnostic laboratory, Vilnius, Lithuania; 2Vilnius University, Life Sciences Center, Vilnius, Lithuania; 3National Cancer Institute, Oncogynecology, Vilnius, Lithuania

Background/Objectives: Ovarian Cancer (OC) is the second deadliest oncogynecologic disease in the world. High-grade serous ovarian cancer (HGSOC) is the most common type of OC with the highest rates of mortality. Currently, there is no specific means of OC detection recommended for use in the clinical setting, thus accurate OC diagnostic biomarkers are in high demand.

Methods: Tissues from 51 patients with gynecologic tumors (32 HGSOC, 10 other gynecologic tumors, 9 benign) were analyzed for the NOTCH pathway (NOTCH1-4, DLL1, JAG2, and HES1), WNT pathway (CTNNB1, FBXW7) and ARID1A gene expression by RT-qPCR, and HOX family related gene (HOPX, ALX4, CDX2), and ARID1A gene promoter methylation status by MSP.

Results: A significant decrease in ARID1A, WNT pathway, NOTCH1-4, DLL1, and HES1 gene expression in HGSOC patients in comparison with the benign cases was detected (p < 0.02). The HOPX gene promoter methylation was significantly increased in HGSOC cases when compared to benign (p = 0.02), ALX4 and CDX2 showed tendency for promoter hypermethylation (p = 0.06), while ARID1A had no significant differences in promoter methylation.

Conclusion: Gene expression and promoter methylation status could be useful for HGSOC diagnosis. More extensive research on early stage patients and non-invasive biosample specimens are needed for biomarker validation for clinical use.

Conflict of Interest: None declared

EP01.060 Clinical significance of germline variants in MLH1, MSH2, MSH6 and PMS2 genes and their association with prostate cancer risk in Polish men - preliminary results.

Marta Heise 1, Piotr Jarzemski2, Aneta Bąk1, Anna Junkiert-Czarnecka1, Olga Haus1

1Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Department of Clinical Genetics, Faculty of Medicine, Bydgoszcz, Poland; 2Jan Biziel University Hospital, Department of Urology, Faculty of Health Science, Bydgoszcz, Poland

Background/Objectives: Prostate cancer (PC) is a complex disease that affects millions of men globally. The relationship of PC with Lynch syndrome is unresolved though molecular investigations and epidemiologic studies have suggested a potential link to the syndrome. Thus, we searched for germinal variants in MLH1, MSH2, MSH6 and PMS2 mismatch-repair genes in Polish prostate cancer patients and controls and analyzed their impact on clinical course of the disease, including overall survival time.

Material: DNA from 97 men with PC from all over Poland and DNA from 100 men - volunteers, healthy at the time of the study.

Methods: NGS, Sanger sequencing.

Results: In 11/97 (11,3%) PC patients 11 variants were detected. The MSH2 c.744G>A was predicted as likely pathogenic and MSH6 c.1969C>T as pathogenic. The MSH2 c.337A>G, MSH6 c.2017C>G, c.3257C>A, c.3557-7_3557-4del, c.1714C>G and PMS2 c.1268C>G, c.1162T>A were predicted as VUS. The MSH6 c.3257-65_3257-61del and c.2267-97T > C were predicted as benign. Two PC patients were carriers of PMS2 c.1268C>G. One PC patient was the carrier of two variants in MSH6. No variant was detected in MLH1 gene. All detected variants were described previously. Bioinformatic analysis of all changes was performed using Franklin database.

Conclusions: The results of the preliminary investigation point at the need to study germinal variants of tested genes to help fully understand the pathogenesis of prostate cancer, identify men at PC high risk and predict the disease recurrence risk after radical prostatectomy.

Grant: This study was supported by the fund of the CM UMK, Bydgoszcz, Poland.

Conflict of Interest: None declared

EP01.061 PDCDL1 Single Nucleotide Variants (SNVs) in relation with the evolution of Cutaneous Malignant Melanoma (CMM)

Elizabeth Córdoba-Lanús1;2;3, Omar García-Pérez 1;2, Leticia Melgar-Vilaplana2;4, Noelia Hernández Hernández5, Ricardo Fernández-de-Misa2;5;6

1Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), San Cristóbal de La Laguna, Spain; 2Universidad de La Laguna, San Cristóbal de La Laguna, Spain; 3Consorcio Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas (CIBERINFEC). Instituto de Salud Carlos III, Madrid, Spain; 4Departamento de Anatomía Patológica. Hospital Univ. Ntra. Sra. de Candelaria, Santa Cruz de Tenerife, Spain; 5Departamento de Dermatología, Hospital Univ. Ntra. Sra. de Candelaria, Santa Cruz de Tenerife, Spain; 6Unidad de Investigación. Hospital Univ. Ntra. Sra. de Candelaria, Santa Cruz de Tenerife, Spain

Background/Objetives: The search for more precise biomarkers to improve knowledge about the prognosis of cutaneous malignant melanoma (CMM) patients in routine clinical practice is still an important challenge. PD-L1 protein exerts a regulation of the antitumor response acting as a good prognostic marker for the response to immunotherapy treatment. Different single nucleotide variants (SNV) in its coding gene, PDCDL1, have been associated with a greater susceptibility to developing other tumors and can operate as prognostic biomarkers in the early neoplasm phases.

Methods: Allelic and genotype discrimination of five SNVs rs4143815, rs822336, rs822337, rs2297136, rs822338 in the PDCDL gene was performed in 338 patients with CMM by qPCR. Hardy-Weinberg equilibrium was tested. Clinical variables were recorded.

Results: rs2297136 harboring the minor allele homozygous genotype AA in the 3’-UTR of the gene is associated with a higher Breslow index (OR = 1.15;95%CI = 1.042-1.267;p = 0.005), presence of ulceration (OR = 1.77;95%CI = 1.00-3.13;p = 0.05) and stage II-IV (OR = 2.10;95%CI = 1.03-4.29;p = 0.04). However, Cox regression analysis revealed that none of the SNVs were associated with melanoma-specific survival or disease-free survival (p > 0.05). As expected, the clinical variables, age at diagnosis (>50 years) [HR = 1.04(1-02-1.06)], gender (male) [2.59(1.48-4.51)], higher tumor thickness (mm) [HR = 1.17 (1.12-1.22)], presence of ulceration [HR = 6.31(3.55-11.23) and stage (III-IV) [HR = 3.16(1.62-6.15)] significantly (p < 0.001) associated with an increased risk of death.

Conclusions: In CMM patients, SNV rs2297136 in PDCL1 gene is associated with a higher Breslow and ulceration, both risk factors of poor prognosis. Further studies are needed to confirm this novel finding.

Grants: PIFIISC21/15, FIISC; CB21/13/00100, CIBERINFEC, Instituto de Salud Carlos III, Spain; Cabildo de Tenerife 2023-2028 (PI-CC202302222, Cabildo.23), Spain.

Conflict of Interest: Elizabeth Córdoba-Lanús Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Omar García-Pérez Financiado por Cabildo Insular de Tenerife 2023-2028, PROYECTO CC20230222, Leticia Melgar-Vilaplana: None declared, Noelia Hernández Hernández: None declared, Ricardo Fernández-de-Misa: None declared

EP01.063 Evaluation of the anticancer effect of Spirulina platensis on the MCF7 human breast cancer cell line

olfa tahri1, Sabriye Kocatürk Sel 2, oya ışık3, leyla uslu3, choubaila reddad1, Bertan YILMAZ2

1Çukurova University, Institute of Natural and Applied Sciences, Biotechnology, Adana, Türkyie; 2Çukurova University, Faculty of Medicine, Medical Biology, Adana, Türkyie; 3Çukurova University, Faculty of Fisheries, Marine Biology, Adana, Türkyie

Background: Spirulina platensis (Arthrospira platensis), a blue-green alga, is extensively studied for its anti-inflammatory and antioxidant properties. It is rich in vitamins, minerals, protein, and pigments (phycocyanin). C-phycocyanin (C-PC) is a natural blue colorant extracted from Spirulina, which is used in food and cosmetics. It has been found to have a strong anti-cancer effect on various cancer types such as breast cancer, liver cancer, lung cancer, and colon cancer, both in vitro and in vivo. In this study, the effect of C-PC derived from Spirulina produced at Çukurova University, Adana, Turkey on human breast cancer cell line MCF7 was tested.

Methods: In this study, we evaluated the effects of both Spirulina aqueous extract and its active ingredient C-PC on the viability of NIH3T3 and MCF7 cells using the MTT assay. Different doses (5, 25 ve 45 μg/ml) of C-PC were exposed to MCF7 for 24 hours. Expression levels of apoptotic genes (caspase-3, -8, -9) were evaluated by Real-Time PCR.

Results: According to the results of the MTT test, it was observed that C-PC significantly reduced the viability of both NIH3T3 and MCF7 cells. Additionally, it was found that C-PC caused significant increases in the expression levels of apoptotic genes.

Conclusion: The number of natural and synthetic substances that can exhibit chemotherapeutic effects for the treatment of breast cancer is increasing every day. According to our results, it is considered that C-PC may be a natural and alternative therapeutic agent in cancer treatment.

Conflict of Interest: None declared

EP01.064 Detection of PIK3CA mutations on breast cancer: Comparison of NGS and Cobas mutation test to Sanger sequencing in a clinical setting

Niki Prekete 1, Eirini Roupou1, Maria Michelli1, Vasiliki Chaidou1, Aphrodite Nonni1, NIKOLAOS KAVANTZAS1, Angelica a Saetta1

11st Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, Greece

Background/Objectives: The PI3K pathway is involved in breast tumourigenesis and PIK3CA mutations mainly found in exons 9 and 20, are currently used as therapeutic biomarkers. We compared Next Generation Sequencing (NGS) and Cobas mutation test to Sanger sequencing for detecting PIK3CA mutations (exons 9 and 20) to determine the best approach for the analysis of HR + /HER2- advanced breast cancer patients.

Methods: 160 samples from women previously diagnosed with breast cancer were analysed through NGS and Sanger sequencing whereas a second cohort of 69 FFPE samples were analysed through Cobas and Sanger. The same extracted DNA was used for the methods in comparison to avoid biased results.

Results: 53 cases (33%) were excluded from NGS analysis after not meeting the pre-analytical quality criteria or having a non-informative result and thus were only analysed through Sanger sequencing. 28% and 26% of the cases were found mutated with NGS and Sanger respectively. The PIK3CA mutated cases in exons 9 and 20 detected with Cobas (42%) were also detectable with Sanger analysis. The most frequently mutated codons were 545 in exon 9 (NGS/Sanger 33.3%, Cobas/Sanger 29.4%) and 1047 in exon 20 (NGS/Sanger 50%, Cobas/Sanger 41%) for both cohorts.

Conclusion: NGS and Sanger sequencing showed an overall 94% concordance, whereas Cobas and Sanger sequencing showed 100% concordance for exons 9 and 20. In conclusion, all methods are useful in detecting SNVs in PIK3CA and can be complementary in everyday practice depending on clinical requirements.

Grant References: No

Conflict of Interest: None declared

EP01.065 Chromosomal instability precede microsatellite instability in Lynch syndrome-associated colorectal tumorigenesis

Marjaana Pussila 1, Aleksi Laiho2, Petri Törönen2, Pauliina Björkbacka3, Sonja Nykänen1, Kirsi Pylvänäinen4, Liisa Holm2, Jukka-Pekka Mecklin5, Laura Renkonen-Sinisalo6, Taru Lehtonen6, Anna Lepistö6, Jere Linden3, Satu Mäki-Nevala7, Päivi Peltomäki7, Minna Nyström8

1University of Helsinki, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, Helsinki, Finland; 2University of Helsinki, Organismal and Evolutionary Biology Research Program, Faculty of Biosciences, and Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), Helsinki, Finland; 3University of Helsinki, Department of Veterinary Biosciences, and Finnish Centre for Laboratory Animal Pathology (FCLAP), Helsinki Institute of Life Science (HiLIFE), Helsinki, Finland; 4University of Jyväskylä, Faculty of Sports and Health Sciences, Jyväskylä; 5University of Jyväskylä, Faculty of Sports and Health Sciences, Jyväskylä, Finland; 6University of Helsinki, Applied Tumor Genomics, Research Programs Unit, Helsinki, Finland; 7University of Helsinki, Department of Medical and Clinical Genetics, Helsinki, Finland; 8University of Helsinki, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, Helsinki, Finland

Background/Objectives: In Lynch syndrome (LS) inherited mutations in DNA mismatch repair (MMR) genes cause MMR deficiency and microsatellite instability (MSI) in tumors. Regular surveillance is crucial for cancer prevention due to high risk; however, frequent interval cancers suggest limitations of colonoscopy-based methods alone. This study aimed to identify precancerous functional changes in colonic mucosa that could facilitate the monitoring and prevention of cancer development in LS.

Methods: We examined 71 colon biopsy specimens from individuals with LS, either tumor-free, diagnosed with adenoma/carcinoma, along with a control group of sporadic cases without LS or neoplasia. The majority (80%) of LS carriers had an MLH1 mutation. RNA-sequencing was used to analyse the transcriptomes, followed by GOrilla ontology analysis, and Reactome Knowledgebase and Ingenuity Pathway Analysis to identify changes potentially linked to neoplastic initiation in LS.

Results: Using pathway and gene ontology analyses along with measuring mitotic perimeters from colonic mucosa and tumors, we discovered a tendency to chromosomal instability (CIN), evident already in colonic mucosa. Our findings imply that CIN precedes MSI and could be the primary driver of cancer initiation, with MSI speeding up tumor formation. Moreover, our results indicate that MLH1 deficiency contributes to CIN development.

Conclusion: The findings confirm our earlier discoveries in mice and underscore the significance of early CIN as a crucial factor and precancerous indicator in the development of colorectal tumors in Lynch syndrome.

Grants: This work was supported by grants from the Jane and Aatos Erkko Foundation, the Academy of Finland; Cancer Foundation Finland sr, and the Sigrid Juselius Foundation.

Conflict of Interest: Marjaana Pussila: None declared, Aleksi Laiho: None declared, Petri Törönen: None declared, Pauliina Björkbacka: None declared, Sonja Nykänen: None declared, Kirsi Pylvänäinen: None declared, Liisa Holm: None declared, Jukka-Pekka Mecklin: None declared, Laura Renkonen-Sinisalo: None declared, Taru Lehtonen: None declared, Anna Lepistö: None declared, Jere Linden: None declared, Satu Mäki-Nevala: None declared, Päivi Peltomäki: None declared, Minna Nyström Minna Nyström is a member of the board of directors and a shareholder in LS CancerDiag Ltd

EP01.066 Non-invasive detection of bladder cancer by digital PCR analysis of urine samples for the presence of the most common mutations: pilot study for the detection of S249C FGFR3 mutation

Sanja Kiprijanovska 1, Hristina Dichevska1, Aleksandar Trifunovski2, Ognen Ivanovski2, Skender Saidi2, Zivko Popov1;2;3, Aleksandar Dimovski1;4

1Research Center for Genetic Engineering and Biotechnology, Macedonian Academy of Sciences and Arts, Skopje, N.Macedonia; 2University Clinic for Urology, Faculty of Medicine, St. Cyril and Methodius University, Skopje, N.Macedonia; 3Clinical Hospital “Acibadem Sistina”, Skopje, N.Macedonia; 4Faculty of Pharmacy, University “St. Cyril and Methodius”, Skopje, N.Macedonia

Background/Objectives: Bladder cancer (BlCa) is one of the most common types of carcinomas which is characterized by a high level (>75%) of mutations in the CDKN2A/2B and FGFR3 genes. The presence of these mutations in early stages of the disease makes them an ideal target for the development of a non-invasive assay for their detection in urine.

Methods: Tissue samples from 254 early stage (BlCa) were included in the study. Urine samples were available for most patients. The CDKN2A/2B and FGFR3 mutation status of each tissue sample was determined previously. A specific digital PCR assay was developed for the detection of the most common FGFR3 S249C mutation, while the development of methods for the detection of other common FGFR3 mutations and CDKN2A/2B deletions is in progress.

Results: A total of 31 urine samples were analysed by digital PCR, of which 23 samples were from patients with S249C mutant tumours. The average age of patients was 64.5 ± 11.1 years (20 male and 3 female) and 2/3 had early-stage disease (Ta-T1). The S249C mutation was detected in urine of all patients with mutation positive and was absent in the urine of all mutation negative tumours (specificity/sensitivity of 100%). Serial dilution analysis revealed that the mutation could be detected at the frequency of 0.1%.

Conclusion: Digital PCR analysis for the presence of the common mutations in urine is a promising strategy for non-invasive early detection and monitoring of BlCa.

Grants:

Conflict of Interest: None declared

EP01.067 Could VUS be important in AML?

Nuket Y Kutlay 1, Halil Gürhan Karabulut1, TIMUR TUNCALI1, Hatice Ilgın Ruhi1, Sadiye Ekinci1, Şule Altıner1, Arzu Vicdan1, Ezgi Gökpınar İli1

1Ankara University Medical School, Medical Genetics, Ankara, Türkyie

Background/Objectives: Acute myeloid leukemia (AML) includes a heterogeneous group of immature myeloid cells. In addition to recurrent cytogenetic abnormalities, European LeukemiaNet and National Comprehensive Cancer Network guidelines also recommend the use of sequencing variants in key genes in classification of AML. At this point, considering different variants in the same gene or a combination of different mutated genes, VUS (Variant of Uncertain Significance) variants may also be important in classification.

Methods: NGS results covering IDH1, IDH2, TP53, RUNX1, ASXL1, WT1, KIT, FLT3, NPM1 and CEBPA genes were evaluated in 108 AML patients with at least one variant. Variant frequencies, different variants in the same gene and variants occurring together in different genes were determined. Only pathogenic (P), likely pathogenic (LP) and VUS variants with VAF scores greater than 1% were analyzed.

Results: Approximately half of the total 253 variants were VUS (128), whereas 80 P and 45 LP variants were detected. The two most frequent variants were in FLT3 > WT1 genes for P, CEBPA > RUNX1 genes for LP and RUNX1 > WT1 genes for VUS variants. In 31 cases, more than one variant was detected in one gene, and in three of them, the same condition was detected for two different genes. Repeated variants were found in all genes.

Conclusion: VAF values in some of the repeated VUS variants suggest the possibility of germline susceptibility. Considering that leukemia is a somatic, clonal and heterogeneous disease, it would be useful to evaluate variant combinations on patient basis to interprete data in diagnosis and prognosis.

References:

Grants: No

Conflict of Interest: None declared

EP01.068 Application of Next-Generation Sequencing-based panels to improve the diagnosis, prognosis, and treatment of pediatric sarcomas.

Piedad Alba Pavón 1, Lide Alaña1, Teresa Imizcoz2, Miriam Gutierrez-Jimeno3, Aizpea Echebarria-Barona1;4, Ricardo López Almaraz1;4, Itziar Astigarraga1;4;5, Ana Patiño6, Olatz Villate1

1Biobizkaia Health Research Institute, Pediatric Oncology Group, Barakaldo, Spain; 2University of Navarra, CIMA LAB Diagnostics, Pamplona, Spain; 3Hospital Clínico Universitario de Valladolid, Servicio de Pediatría, Valladolid, Spain; 4Hospital Universitario Cruces, Pediatrics Department, Barakaldo, Spain; 5University of the Basque Country, UPV/EHU, Pediatrics Department, Faculty of Medicine and Nursing, Leioa, Spain; 6University Clinic of Navarra, CUN and Cima/Solid Tumor Area, Dpt. Of Pediatrics and Medical Genomics Unit, Pamplona, Spain

Background/Objectives: Sarcomas are conventionally diagnosed according to morphological features and immunohistochemical techniques. However, some of them exhibit features that may overlap with other entities. Therefore, identifying tumor-specific genetic alterations is necessary for the classification of this group of tumors. The objective was to assess the implications of two NGS-based genomic panels in the diagnosis, prognosis, and treatment of pediatric patients diagnosed with sarcomas.

Methods: Forty-two tumor samples of pediatric and young adult patients diagnosed with sarcomas were sequenced using Oncomine Childhood Cancer Research Assay panel (OCCRA panel) (Life Technologies) and/or Fusion Plex Sarcomas Panel (FPS panel) (Invitae).

Results: Thirty tumor samples were sequenced using OCCRA Panel and 74 clinically relevant genetic alterations were identified. The most frequent genomic alterations were EWSR1::FLI1 fusion gene (20%), alterations in FGFR4 and TP53 genes, and PAX3::FOXO1 fusion gene (13% each).

Fourteen samples were sequenced with the FPS panel. Eleven patients were fusion-positive, of which two patients had been previously sequenced with the OCCRA panel. In these patients, TGF::ADGRG7 and TPR::NTRK1 fusion genes were identified.

The NGS sequencing tools used in this study identified a genetic alteration with potential clinical implications in 74% of patients. A variant with implication in the diagnosis or potential therapeutic value was identified in 27 and 12 of sequenced tumors, respectively.

Conclusion: NGS-based panels are tools that complement morphology, immunohistochemistry and FISH techniques to obtain accurate data for diagnosis, prognosis and the identification of potential therapeutic targets.

Grants: EITB Media AND BIOEF, SAU (BIO20/CI/011/BCB), Basque Government (2021111030).

Conflict of Interest: None declared

EP01.069 Rare germline chromosome 1 duplication identified in young male with colon cancer: a case report investigating causality

Anna Byrjalsen 1, Sara Garcia2, Line Borgwardt1, Karin Wadt1, Anne-Marie Gerdes1, Thomas van Overeem Hansen1

1Copenhagen University Hospital Rigshospitalet, of Clinical Genetics, Copenhagen east, Denmark; 2Danish Technical University

Background/Objectives: Colorectal cancer (CRC) occurrence among young adults is increasing. In the following we present geno- and phenotype of a patient with colon cancer (CC).

Methods and Results: A 24-year-old male presented with abdominal pain, and a CT-scan revealed a pre-stenotic iliac dilatation. A subsequent laparoscopy identified a tumor in the cecal wall. The tumor was an adenocarcinoma, and immunohistochemistry and somatic gene testing came back ia.

The patient underwent genetic testing with an in-house panel: APC, AXIN2, BMPR1A, EPCAM, GREM1, MLH1, MSH2, MSH3, MSH6, MUTYH, NTHL1, PMS2, POLD1, POLE, PTEN, SMAD4 and STK11. No pathogenic variants were detected. Subsequent, whole genome sequencing revealing a duplication on chromosome 1 spanning 200 kb and covering CD101, TTF2, MIR942, TRIM45, and parts of PTGFRN and VTCN1. The duplication had not previously been described in gnomAD/the literature. However, a similar duplication covering an overlapping area (TTF2, MIR942, and TRIM45) had been reported in another family with CRC.

Segregation analysis revealed the duplication was maternally inherited. On the maternal side of the family, there had been one case of a tubular adenoma with high-grade neoplasia and one case of CRC. This led to genetic testing of the patient’s maternal grandparents. The duplication proved to be inherited from an unaffected relative.

We calculated a polygenic risk score using 95 SNPs correlated to the risk of CRC. The PRS was 7.87, indicating an average risk of CRC compared to others with CRC.

Conclusion: Our findings do not support that this variant is the sole reason for early onset CC in our patient.

Conflict of Interest: None declared

EP01.070 The CDKN2A Gene Variants related with Cancer Predisposition Syndrome

Roya Gasimli 1, Asli Subasioglu2

1Ege University, Medical Biology, Izmir, Türkyie; 2Izmir Katip Çelebi University, Medical Genetics, Izmir, Türkyie

Background/Objectives: The CDKN2A gene, situated on chromosome 9p21, encodes vital proteins, namely p16INK4a and p14ARF, pivotal in regulating cell proliferation. Mutations in CDKN2A result in uncontrolled cell growth and cancer progression. Understanding its implications not only aids in diagnostics and therapies but also enhances our understanding of cancer. Our objective was to identify gene variants, particularly within CDKN2A, among 3036 genomic DNA samples obtained from individuals with a predisposition to hereditary cancer or with affected first-degree relatives.

Material and Methods: The study collected peripheral blood samples from 3036 individuals with hereditary cancer backgrounds or predisposition. DNA extraction utilized Qiaseq Targeted Panels kits, followed by sequencing on the Illumina Miseq platform. FastQ data were analyzed, resulting in the generation of a variant call format (VCF) file using QIAGEN CLC Genomics Workbench. The Qiagen Clinical Insight (QCI Interpret) system identified variants associated with familial cancer predisposition.

Results: The analysis, conducted from the VCF file using the QCI Interpret system, identified germline likely pathogenic (LP) mutations in 5 patients and variants of uncertain significance (VUS) in 20 patients, associated with melanoma, colorectal, and pancreatic cancers. Mutations primarily localize to exon 1, 2 and intron 1.

Conclusion: In light of the potential consequences at the post-transcriptional level, variants classified as LP, VUS variants, and even those reported to have no effect at the protein level hold significant importance with various genetic and epigenetic mechanisms. Additionally, with supporting evidence from in vitro and in vivo functional studies, these variants could potentially emerge as therapeutic targets for various cancers.

Grants: None.

Conflict of Interest: None declared

EP01.072 Autophagy and P62: A Collaborative Effort in CRC Chemoresistance to 5-FU Uncovered by Combined Bioinformatics and Laboratory Tests

Sara Ebrahimi 1, Haniye Rahimi Kolour1, Ehsan Nazemalhosseini Mojarad2, maryam Ghanbari-Safari3

1Research Institute for Gastroenterology and Liver Diseases, 1. Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, tehran, Iran; 2Research Institute for Gastroenterology and Liver Diseases, Gastroenterology and Liver Diseases Research Center, tehran, Iran; 3Tehran North Branch, Islamic Azad University, 3. Department of Microbial Biotechnology, Faculty of Biological Science, tehran, Iran

Background: In contemporary research, autophagy is recognized as a significant pathway implicated in chemoresistance. The P62/SQSTM1 gene, which encodes the P62 protein, is a key player in this pathway. Inhibition of this gene presents a potential strategy to enhance the anticancer efficacy of chemotherapy drugs. This investigation aimed to explore the association between autophagy and resistance to 5-FU chemotherapy in colorectal cancer by identifying a lncRNA and a miRNA linked to the P62 gene and autophagy.

Material and Methods: For the in-silico analysis, a specific lncRNA and miRNA associated with the P62 gene and autophagy pathway were chosen from databases. Subsequently, two cell lines, SW480 and Caco2, were chosen to assess resistance to the drug 5-fluorouracil (5-FU). Multiple laboratory assays were utilized to validate resistance to chemotherapy and autophagy. Finally, the expression levels of the target gene, lncRNA, and miRNA were quantified by realtime-PCR.

Results: The in-silico analysis identified LINC01962 and miR-17-5p. Western blot and immunocytochemistry demonstrated an increase in autophagy in chemoresistance cells compared to the untreated cell line. The study revealed an upregulation of P62 expression in both cell lines, a decrease in LINC01962 expression in Caco2/5-FU, and an increase in its expression in SW480/5-FU, as well as an increase in miR-17-5p expression in both cell lines.

Conclusion: In conclusion, the findings indicate that LINC01962 may modulate 5-FU drug resistance in metastatic CRC cells by influencing the P62 gene. Additionally, upregulation of miR-17-5p expression may enhance autophagy in these cells, suggesting a potential therapeutic target to enhance the effectiveness of 5-FU-based chemotherapy in CRC.

Conflict of Interest: None declared

EP01.073 Comprehensive Exploration of Nasopharyngeal Carcinoma through Whole Transcriptomic Sequencing and Single-Cell RNA Sequencing

Che Kang Lim 1;2, Yi Tian Png3, Lee Yew Mun3, Zhi Yong Lim3, Wei Ying Sim1, Chwee Ming Lim1;2;3

1Singapore General Hospital, Clinical Translational Research, Singapore; 2Duke-NUS Medical School, Singapore; 3Singapore General Hospital, Otorhinolaryngology-Head&Neck Surgery, Singapore

Background: Nasopharyngeal carcinoma (NPC), an epithelial malignancy prevalent in Southeast Asia and Southern China, is closely associated with Epstein-Barr virus (EBV). Despite therapeutic advancements, late diagnosis and drug resistance often lead to unsatisfactory clinical outcomes. Recent advances highlight single-cell RNA sequencing (scRNA-seq) as pivotal in oncology research, offering insights into cellular heterogeneity. Integrating scRNA-seq with bulk RNA-seq data holds promise to transform NPC research by providing a comprehensive understanding of gene expression patterns.

Methods: Whole transcriptomic sequencing was performed in eight NPC patients and three controls. Additionally, single cell RNA sequencing was conducted in the selected samples to assess the tumor microenvironment.

Results: From the whole transcriptomic sequencing analysis, PLLP, S100A9, and PLCG2 genes emerged as the top three significantly expressed genes, exhibiting notably elevated expression levels in the NPC patient cohort. Additionally, in conjunction with the epithelial cluster (EPCAM + , KRT5 + , KRT8 + , KRT13 + , KRT19 + ), single-cell RNA sequencing analysis revealed diverse immune cell clusters, comprising four subtypes of B cells (including plasma B cells), four subtypes of T cells (including NKT cells), NK cells, and mast cells. These findings indicate a complex and heterogeneous tumor microenvironment.

Conclusion: Our study offers a comprehensive perspective on Nasopharyngeal Carcinoma (NPC), shedding light on its cellular heterogeneity and molecular pathways. Further research is warranted to validate our findings and explore the interplay between NPC tumors and immune cells.

Grant: SRG-OPN-007

Conflict of Interest: None declared

EP01.074 Performance of the polygenic risk score PRS313 for breast cancer prediction in patients with atypical breast lesion

Manon Reda1, Anes Hadjadj1, Juliette Sauge1, Laurent Arnould1, Juliette Albuisson 1

1Centre Georges François Leclerc, Département de Biologie et Pathologie des Tumeurs, DIJON, France

Breast cancer (BC) genetic includes polygenic factors, combined in recently characterized polygenic risk scores (PRS). SPORA is a study based on PRS313, the most used PRS in breast cancer (Mavaddat et al, 2019).

SPORA aimed at determining PRS313 performance to predict the occurrence of breast cancer in patients with atypical breast lesions (ABL), presenting with intermediate BC risk (13-20%).

We conducted a monocentric retrospective study of PRS313 performances in cases and controls (patients with ABL and BC within 5 years, n = 73, and without BC within 5 years, n = 154, respectively). Genotyping was performed on FFPE normal tissues, using a microfluidic platform.

PRS313 could be calculated, with a median of 271 SNPs per patient. Mean PRS scores for cases and controls are respectively 0.046 and -0.1. The PRS threshold with the best sensitivity and specificity was -0.05 (AUC 0.73 [0,66 – 0,8], sensitivity 0.7, specificity 0.63, odds-ratio 3.94 [2.171 - 7.170] (p < .0001)). At 20% threshold (high risk of BC), PRS is -0.125 (AUC 0.6, sensitivity 0.83, specificity 0.38, NPV 0.82, odds-ratio 3 [1,526 - 6,182]).

PRS313 could represent valuable tool to detect high risk patients in this group with intermediate risk.

These results will be consolidated through the analysis of NOMAT study patients, including the evaluation of PRS performance in combination with NOMAT model (predictive model of BC risk based on radiological criteria and age, Uzan et al, 2021). This new combination of biomarkers, in the scope of personalized medicine, could help selecting patients who require breast surgery.

Grants: AOR Bourgogne-Franche-Comté 2020 ; AOI Biocollection 2020.

Conflict of Interest: Manon Reda AOR Bourgogne-Franche-Comté 2020 ; AOI Biocollection 2020.

, Anes Hadjadj: None declared, Juliette Sauge: None declared, Laurent Arnould: None declared, Juliette Albuisson: None declared

EP01.075 MicroRNAs as dual action players: Tumor suppressors and differential diagnosis markers in head and neck tumors

Nadja Nikolic 1, Jelena Carkic1, Violeta Sango2, Nicole Riberti3, Boban Anicic4, Jelena Milasin5

1University of Belgrade, School of Dental Medicine, Implant Research Center, Belgrade, Serbia; 2University of Belgrade, Faculty of Biology, Belgrade, Serbia; 3University G. d’Annunzio of Chieti-Pescara, Department of Neuroscience, Imaging and Clinical Sciences, Chieti, Italy; 4University of Belgrade, School of Dental Medicine, Clinic for Maxillofacial Surgery, Belgrade, Serbia; 5University of Belgrade, School of Dental Medicine, Human Genetics, Belgrade, Serbia

Background/Objectives. Belonging to the broader category of head and neck tumors along with oral squamous cell carcinomas, salivary gland tumors (SGTs) are a heterogenous group of pathologies which still represents a challenge regarding differential diagnosis and therapy. Detection of molecular alterations is emerging as an effective diagnostic tool. We aimed to analyze the relative expression levels of micro RNAs miR-26a, miR-26b and miR-191, and pro-oncogenic molecular markers PLAG1, MTDH and HIF2 in SGTs and normal salivary gland (NSG) tissues, and evaluate them as potential differential diagnosis markers.

Methods. This cross-sectional study included 58 patients with SGTs (23 pleomorphic adenomas, 27 Warthin tumors and 8 malignant salivary gland tumors) and 10 controls (normal salivary gland tissues). Relative gene expression levels of all investigated molecules were determined by reverse transcriptase-real-time polymerase chain reaction.

Results. All three micro RNAs exhibited highest expression levels in benign SGTs, while miR-26a and miR-191 were significantly more expressed in PAs compared to WTs (P = 0.045 and P = 0.029, respectively). PLAG1 and HIF2 were both overexpressed in WTs compared to PAs (P = 0.048 and P = 0.053, respectively). Bioinformatic analysis suggested that all investigated micro RNAs function as negative regulators of MTDH.

Conclusion. All three micro RNAs have a negative impact on MTDH oncogene expression in malignant tumors, while the differences between levels of mir-26a, miR-191, PLAG1 and HIF2, in PA and WT represent possible differential diagnosis markers.

Grants: Science Fund of the Republic of Serbia, GRANT 7750038.

Conflict of Interest: None declared

EP01.076 Correlation between relative telomere length and specific mutations of the KRAS gene in patients with metastatic colorectal carcinoma

Dragana Jugović 1, Marija Vukelic-Nikolic2, Višnja Madić3, Ljiljana Branković1, Radovan Milićević1, Perica Vasiljević4

1University Clinical Center Niš, Center for Medical and Clinical biochemistry, Niš, Serbia; 2University of Niš, Faculty of Medicine, Department of Biology and Human Genetics, Niš, Serbia; 3University of Niš, Faculty of Sciences and Mathematics, Department of Biology and Ecology, Niš, Serbia; 4University of Niš, Faculty of Sciences and Mathematics, Department of Biology and Ecology, Niš, Serbia

Background/Objectives: Telomeres, specialized repeat DNA sequences, functioning as a “protective cap” at the ends of linear chromosomes maintain chromosomal stability and genomic integrity. Telomere length anomaly is one of the earliest genetic changes in malignant transformation that may be accelerated by specific alterations in the genes involved in colorectal cancer (CRC). This study aimed to evaluate the possible relationship between the relative telomere length and specific KRAS mutations in CRC patients with developed metastases as well as with their clinical-pathological characteristics.

Methods: The genomic DNA of 89 CRC patients with mutated KRAS were extracted from the formalin-fixed paraffin-embedded tumor tissue sections while the relative telomere length was measured by real-time PCR and calculated using the formula T/S = 2-ΔΔCt.

Results: The most frequent types of mutations were G12D (31,4%), G12V (22,4%), and G12А (11,2%) in codon 12. There was no significant correlation between relative telomere length and clinical-pathological characteristics of CRC patients. However, patients with mutation in G12V had significantly longer telomeres than patients with other mutations (p < 0.05).

Conclusion: The longer telomeres length was associated with the presence of a G12V mutation in KRAS, suggesting that this mutation coupled with the length of telomeres might play an important role in the biological behavior of CRC.

Grants: This work was supported by the Ministry of Education, Science and Technological Development of the Republic of Serbia (Grant No. 451-03-47/2023-01/ 200124).

Conflict of Interest: None declared

EP01.078 Integrated transcriptomic analysis reveals blood-based gene signature with diagnostic and prognostic potential for patients with breast cancer

Mustafa Kaya1, Ibrahim Kaya 2, Dilek Colak3

1Hacettepe University Medicine Faculty, Türkyie; 2Al Faisal University, Riyadh, Saudi Arabia; 3King Faisal Specialist Hospital and Research Centre (KFSHRC), Department of Molecular Oncology, Riyadh, Saudi Arabia

Introduction: Breast cancer (BC) continues to be one of the most malignant tumors despite the advances in cancer therapies and raising awareness. It has utmost importance to detect the disease reliably and as early as possible in order to reduce the risk of mortality.

Materials and Methods: Integrated genomic analysis of BC was performed using genome-wide gene expression profiling datasets from both tissue and blood samples from patients with BC that were probed using Affymetrix HGU133 Plus 2.0 array. The diagnostic and prognostic potential of the identified gene signature are validated using transcriptomic datasets from patients with BC with detailed clinical information.

Results: The integrated analysis revealed a 60-gene signature that are significantly expressed in both tissue and blood of BC patients compared to healthy controls. Hierarchical clustering using 60 genes clearly separated patients from normal controls validating the gene signature’s diagnostic potential. The investigation of the 60-geneset score with the BC clinical and pathological features, including age, grade and HER2 indicated that high geneset score was significantly associated with aggressive disease characteristics. Moreover, high gene-set score is associated with a poor disease outcome.

Conclusions: The results suggest that integrated genomic analysis may provide a robust methodology to diagnose patients with BC non-invasively and lead to improved diagnosis and early detection for BC.

Acknowledgment: We would like to thank King Faisal Specialist Hospital and Research Center for supporting this study (RAC#2110006 to DC).

Conflict of Interest: None declared

EP01.079 Metastasizing colorectal cancer: intra-tumor heterogeneity of cancer stem cells related genes identified by bioinformatics analysis of publicly available RNAseq data

Emanuela Bostjancic 1, Kristian Urh1, Nina Zidar1

1University of Ljubljana, Faculty of Medicine, Institute of Pathology, Ljubljana, Slovenia

Background: Molecular pathways in colorectal cancer (CRC) are well understood; however, development of metastases and intra-tumor heterogeneity (ITH) are still largely unexplained. Our previous study using bioinformatics analysis of publicly available RNAseq data (GEO) suggest involvement of cancer stem cells (CSCs) related genes in metastasizing CRC. We therefore analyzed identified genes and their potentially regulatory miRNAs in metastasizing CRC in context of ITH, which is related to cancer progression, therapy resistance and recurrences.

Methods: Nineteen patients and 63 formalin-fixed paraffin-embedded tissue samples were included (central part and invasive front of primary tumor, lymph node and liver metastases). After RNA isolation, mRNAs and miRNAs expression was analyzed using qPCR. Expression gradient of six CSC-related genes (ANLN, CDK1, ECT2, PDGFD, TNC, TNXB) and their potential regulatory miRNAs (miR-199a-3p, miR-200b-3p, miR-217-5p, miR-223-3p, miR-335-5p, miR-425-5p) was investigated.

Results: We observed differential expression of PDGFD and TNXB (and miR-199a) at the invasive front in comparison to central part of tumor. Significant differences in expression of ANLN, CDK1 and ECT2 (and miR-199a, miR-200b-3p, miR-223-3p, miR-425-5p) was identified in lymph node metastasis when compared to central part of tumor, whereas ANLN, CDK1 and ECT2 as well as TNXB were differentially expressed in liver metastases (and miR-199a-3p, miR-425-5p). Meta-analysis of publicly available sequencing data (TCGA) confirmed correlation between miR-425-5p and TNC.

Conclusion: Our results suggest that expression of CSC‑related genes and their potential regulatory miRNAs contribute to ITH in CRC, lymph node and liver metastases.

Grant references: Slovenian Research Agency (ARRS) J3-1754, J3-3070, P3-0054.

Conflict of Interest: Emanuela Bostjancic Slovenian Research Agency (ARIS)

Project number: J3-1754, Kristian Urh Slovenian Research Agency (ARIS)

Project number: J3-1754, Young Researcher, Nina Zidar Slovenian Research Agency (ARIS)

Project number: J3-1754

Program number: P3-0054

EP01.082 Detection of a novel RUNX1::FOXJ2 fusion in acute myeloid leukemia

yiannos kyprianou 1

1Karaiskakio Foundation, Molecular Pathology-Oncology Department, Nicosia, Cyprus

Background/Objectives: Adult acute myeloid leukemia (AML) is a type of blood and bone marrow cancer characterized by the uncontrolled proliferation of myeloid precursor cells in the bone marrow, leading to the inhibition of normal hematopoiesis.

Methods: We herein describe a case of a 60-year-old female patient with AML relapse post-transplant. Upon diagnosis, molecular investigation was carried out on peripheral blood and an FLT3 ITD and NPM1, TET2 and DNMT3A mutations were detected.

The patient relapsed 8 months following diagnosis. Molecular investigation was repeated on bone marrow confirming the presence of the mutations also detected in the diagnostic sample. In addition, RNA NGS was also repeated.

Molecular RNA investigation employed the Archer® FusionPlex® Pan-Heme Kit for Illumina platform according to manufacturer’s recommendations.

Results: RNA NGS investigation revealed the RUNX1::FOXJ2 fusion. The identified fusion partners were RUNX1 exon 6 chr21 and FOXJ2 exon 2 chr12.

Conclusion: RNA sequencing revealed the RUNX1::FOXJ2 fusion gene transcript in the relapse sample. This fusion was not detected in the diagnostic sample.

FOXJ2 (Forkhead Box J2) is a transcriptional activator localized constitutively at the nucleus of the cell.

RUNX1, is a transcription factor that plays a crucial role in hematopoietic differentiation. Chromosomal rearrangements with RUNX1 have been identified with uncommon partners in various hematologic malignancies.

Well-powered studies indicate that patients with a RUNX1 alteration, experience a less favourable prognosis in alignment with the patient’s disease progression.

Accurate detection of RUNX1 alterations is essential for unravelling the etiology of blood disorders and determining disease prognosis.

Grants:

Conflict of Interest: None declared

EP01.083 Novel mutation in CHEK1 gene in Bulgarian patient with Poorly cohesive gastric carcinoma

Maria Glushkova 1, Tihomir Todorov1, Svetlana Gacheva2, Albena Todorova1;3

1GMDL Genica, Sofia, Bulgaria; 2Аджибадем Сити Клиник УМБАЛ Младост, Sofia, Bulgaria; 3Medical University of Sofia, Sofia, Bulgaria

Background/Objectives: Here we present a 48-years old Bulgarian woman referred for testing of oncogene panel by NGS in relation to future therapy.

The diagnosis is Poorly cohesive gastric carcinoma with metastases in retroperitoneum and peritoneum. Immunohistochemistry shows high-grade adenocarcinoma, HER2- and PD-L1(CPS > 10). She underwent two courses of treatment with Oxaliplatina, Fluorouracil and Nivolumab. Cytopenia and inflammatory infectious changes are observed.

Methods: Genetic testing includes generally 688 genes: 175 genes related to target therapy, 69 genes for hereditary cancer and 7 genes are drug-metabolizing enzymes.

Results: The genetic test show pathogenic germline mutation in CHEK1 gene (p.Y334Ter;c.1002C>G). The mutation has not been reported in the literature.

Four somatic clinically significant variations were found in the FFPE: TP53 (p.K164E;c.490A>G), KRAS (CopyNumberGain) and two variants in RHOA. Based on the somatic genetic variants there are some FDA approved therapies but for the other types of tumors (Level C and D).

Conclusion: CHEK1 gene is protein kinase and plays central role in DNA damage, replication, repair, checkpoint activation and it is essential for cell proliferation and survival. It belongs to DNA damage response and repair (DDR) related genes and the detected variant is from inactivation type with possible benefit of immunotherapy. Tumors with mutations in DDR demonstrated a higher survival rate with ICB (ImmuneCheckpointBlockade) therapy. In this case immunotherapy would be possible opportunity.

We recommended segregation analysis for patient’s relatives in order to understand the risk of the corresponding tumors. The genetic testing of oncogene panel is important for the genetic counseling and correct therapeutic approach.

Grants: No

Conflict of Interest: None declared

EP01.084 Investigation of the effects of dietary supplement Methylsulfonylmethane on apoptosis signals in acute myeloid leukemia cell lines

Yalda Hekmatshoar 1, Arzu Zeynep Karabay2, Asli Koc2, Tulin Ozkan3, Asuman Sunguroglu3

1Altinbas University, Department of Medical Biology, Faculty of Medicine, Istanbul, Türkyie; 2Ankara University, Department of Biochemistry, Faculty of Pharmacy, Ankara, Türkyie; 3Ankara University, Department of Medical Biology, Faculty of Medicine, Ankara, Türkyie

Background/Objectives: Acute myeloid leukemia (AML) is a heterogeneous disease in both biological and clinical concepts. Methylsulfonylmethane (MSM), an organic sulfur-containing dietary supplement, utilized for improving various clinical conditions particularly osteoarthritis. MSM can exert antitumor activity in various types of cancers. However, the molecular mechanisms of action underlying antileukemic activity of MSM remain unclear.

Methods: In this study, we explored the anticancer effect of MSM on human AML cell lines (U937 and HL60) with focus on underlying death mechanism. Anticancer activity of the MSM was examined employing MTT assay, Annexin V-PE/7AAD staining and real-time q-PCR. Both cell lines were treated with different concentrations (50-400 mM) of MSM for 24h.

Results: Results of MTT assay revealed that in both cell lines, the MSM markedly reduced cell viability in comparison to the control cells. Additionally, findings of Annexin V-7AAD staining represented that MSM triggered apoptosis in both cell lines significantly. Apoptotic genes expression levels were assessed by real-time PCR. Based on the real time q-PCR data, MSM increased the mRNA expression levels of BAX and BIM genes in treated cells.

Conclusion: Over all, our results indicated that MSM could induce apoptosis in AML cell lines in a dose-dependent manner, therefore could be utilized as antileukemic agent.

Grants: -

Conflict of Interest: None declared

EP01.085 Gorlin-Goltz syndrome in six Polish patients

Renata Posmyk 1, Joanna Karwowska1, Klaudia Berk1

1Medical University in Bialystok, Department of Clinical Genetics, Bialystok, Poland

Background/Objectives: Gorlin-Goltz syndrome (GGS) (OMIM#109400), (Basal Cell Nevus Syndrome or Nevoid Basal Cell Carcinomas Syndrome) is a rare genetic disorder characterized by basal cell carcinomas and nevi, pits of palms and soles, calcification of falx cerebri, odontogenic keratocysts of jaws, bone abnormalities and dysmorphic features. Evans diagnostic criteria are very useful for clinical diagnosis. GGS is inherited in autosomal dominant manner and caused by mutations in PTCH1, PTCH2 or SUFU genes.

Methods: We present the phenotypes of Polish (N = 6) participants of the project „Genetic Background of Overgrowth Syndromes in Polish and Lithuanian Populations: Basis for Rapid Genetic Testing to Prevent Neoplasms” with cardinal features of GGS.

Results: In 4 patients the diagnosis was molecularly confirmed by the pathogenic mutations in PTCH1 gene, and in 1 the small deletion in the same gene was found. Despite the wide molecular testing in one patients, the genetic confirmation has not been made.

Conclusions: All patients fulfill the diagnostic criteria of GGS, but their symptoms are of wide spectrum and molecular results are variable. Lack of detection of pathogenic variants in genes correlated with clinical symptoms of GGS may indicate the unknown molecular cause of the disorder which may be a field for the future research.

Grants: NCN/1/DA/21/001/1106

Conflict of Interest: Renata Posmyk NCN/1/DA/21/001/1106, Full-time Medical University in Bialystok, Joanna Karwowska: None declared, Klaudia Berk: None declared

EP01.086 First association of Heyn-Sproul-Jackson syndrome and paraganglioma in one patient

Anna Maria Cueto-González 1;2, Alejandro Moles-Fernandez1;2, Maria Camprodón-Gómez3, Anna Casteras-Román4, Teresa Vendrell-BAyona1, Elena García-Arumí1;2, Eduardo Tizzano1;2

1Vall d’Hebron Barcelona Hospital Campus (Barcelona), Clinical and Molecular Genetics Area., Barcelona, Spain; 2Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, Medicine Genetics Group, Barcelona, Spain; 3Vall d’Hebron Barcelona Hospital Campus (Barcelona), Rare diseases Unit., Barcelona, Spain; 4Vall d’Hebron Barcelona Hospital Campus (Barcelona), Endocrinology Unit., Barcelona, Spain

Introduction: autosomal dominant gain-of-function mutations (GoFm) in DNMT3A gene (Heyn-Sproul-Jackson syndrome (HESJAS); OMIM618724) have recently been described with reciprocal phenotype to Tatton-Brown-Rahman syndrome (OMIM615879) and it is characterized by microcephalic dwarfism.

At present only 3 patients have been reported with HESJAS (Heyn et al.,2019), all without oncological pathology. On the other hand, 3 cases with somatic GoFm in the DNMT3A gene associated with paragangliomas have also been reported, none with HESJAS diagnosis, and only one with intellectual disability (Remacha et al., 2018; Mellid et al., 2020).

Methods: we report a 34-year-old patient with global developmental delay, mild intellectual disability, postnatal short stature, microcephaly, Chiari malformation type I, and particular phenotypic characteristics corresponding to HESJAS. Multiple asymptomatic carotid gangliogliomas were detected in a control MRI scan.

Results: Having a previously had an inconclusive array-CGH and exome, but after exome reanalysis revealed a GoFm in the DNMT3A gene not described in population databases, which was also found in the tumour tissue of one of the paragangliomas.

Conclusions: So far, GoFm in the DNMT3A gene have been described at the germline level in 3 patients with HESJAS and in 3 patients with paraganglioma without diagnosis of HESJAS, only one with isolated intellectual disability. To our knowledge, this is the first patient with HESJAS and GoFm in the DNMT3A gene confirmed at both, germline and tumour. Documentation of paragangliomas in patients with HESJAS should be incorporated into the follow-up protocol. On the other hand, a complete anamnesis and physical examination of patients with paraganglioma to discard HESJAS is also necessary.

Conflict of Interest: None declared

EP01.088 Is the obvious enough when considering Lynch Syndrome diagnosis? Rare cancers in index cases and diagnostic challenges

Iulia Simina 1;2, Iulia Teodora Perva2, Catalin-Vasile Munteanu1;3, Oana Cristina Voinea4;5, Adrian Trifa1;6;7

1Victor Babeş University of Medicine and Pharmacy, Department of Genetics, Center of Genomic Medicine, Timișoara, Romania; 2Oncohelp Medical Center, Genetics, Timișoara, Romania; 3Emergency Hospital for Children Louis Ţurcanu, Medical Genetics, Timișoara, Romania; 4Cantacuzino National Institute of Research, București, Romania; 5Carol Davila University of Medicine and Pharmacy, Pathology, București, Romania; 6Victor Babeș Hospital, Medical Genetics, Timișoara, Romania; 7The Oncology Institute “Prof. Dr. Ion Chiricuţă” Cluj-Napoca, Breast Cancer Center, Cluj-Napoca, Romania

Background: When an index case presents with a rare cancer absent from the tumour spectrum of the most prevalent cancer predisposition, Lynch syndrome (LS), several additional information can improve diagnosis timing and prevention strategy. We raise an issue that emphasizes these diagnostic challenges by presenting a case series.

Methods: The clinical workup and family history was performed for the affected individuals. MMR studies have completed the analysis. Sequence analysis and deletion/duplication testing of an extended panel of genes for cancer predisposition syndromes was performed, using Illumina’s sequencing-by-synthesis method.

Results: We identified 3 case-index patients with rare cancers consecutive to high susceptibility of LS, and only because we chose to perform extended panel sequencing due to their personal or family history: P1 presented with an extrahepatic biliary duct adenocarcinoma at age 42yo, heterogeneous family history, and a CNV in MLH1 gene; P2 had a breast luminal cancer at age 47yo, a paternal cousin with endometrial cancer and a PMS2 pathogenic variant; P3 had breast bilateral adenoma at age 34yo with positive familial history for both breast and endometrial cancers and a likely pathogenic variant in PMS2. Other 2 cases who both presented early with thyroid cancer were not diagnosed until they have developed a suggestive colon cancer, empowering the need for an improved diagnosis strategy in uncommon LS cancers.

Conclusion: Oncogenetics has an essential role in identifying individuals at-risk for Lynch syndrome and planning a preventive strategy using family history. Sometimes, thinking outside the patterns can offer a diagnosis.

Conflict of Interest: None declared

EP01.089 Oncogenic miR-21 and miR-31 clinical relevance in oral cancer: tissue specimens vs. exosomes

Milos Lazarevic1, Dijana Trisic1, Milica Jaksic-Karisik1, Drago Jelovac1, Nadja Nikolic1, Jelena Carkic1, Jelena Milasin 1

1University of Belgrade, School of Dental Medicine, Belgrade, Serbia

Background/Objectives: Oral cancers may be very aggressive, and new predictors of their behavior are needed. MicroRNAs (miRNAs) are small, non-coding RNAs involved in gene expression regulation; their altered levels have been linked to cancerogenesis. Exosomes, l extracellular vesicles that carry different cargos including nucleic acids, proteins, etc. are important mediators of intercellular communication. We aimed to investigate two oncogenic miRNAs (miR-21 and miR-31) levels in oral squamous cell carcinomas (OSCCs) specimens and exosomes, and to establish whether they may predict OSCC behavior.

Methods: RNA was extracted from 20 OSCC tissue specimens and from exosomes, using trizol. Exosomes were previously isolated from cell cultures obtained from OSCC patients, by magnetic separation, and characterized by flow-cytometry, TEM and nanoparticle tracking analysis. Reverse transcription and qPCR were applied to determine micro RNAs levels, followed by statistical analysis.

Results: MiR-21 levels were significantly higher in tissue specimens of OSCC patients compared to control tissues (p < 0.0001). The levels were also higher in T3 compared to T1 (p < 0.05) and T2 (p < 0.01) stages. Surprisingly, miR-21 levels were significantly lower in cancer exosomes compared to control exosomes. MiR-31 levels were also higher in OSCC than in controls (0.05) and exosomes followed the same pattern. MiR-31 also showed increase with increasing stages. In cancer tissue there was no significant difference of miRNAs expression, while exosomes contained much higher levels of miR-21 than miR-31 (<0.01).

Conclusion: Tumor tissues’, rather than exosomes’ miR-21 and miR-31 levels might indicate OSCC course.

Grants: Science Fund of the Republic of Serbia, GRANT 7750038

Conflict of Interest: Milos Lazarevic School of Dental Medicine, Dijana Trisic School of Dental Medicine, Milica Jaksic-Karisik: None declared, Drago Jelovac School of Dental Medicine, Nadja Nikolic School of Dental Medicine, Jelena Carkic School of Dental Medicine, Jelena Milasin Science Fund of the Republic of Serbia, GRANT 7750038, School of Dental Medicine

EP01.090 Unveiling novel genetic variants in Peutz-Jeghers syndrome: A case report of a de novo mutation in the STK11 gene

Jorge Docampo-Cordeiro 1, Fransico Jose Garcia-Iñigo1, Elena Jaime-Lara1, Marta Casas-Rodriguez1, Rosa DeFrutos-Curiel1, Ana Santos-Corral1, Maria Luisa Casas-Losada1

1Hospital Universitario Fundación Alcorcón, Analisis Clínicos, Alcorcon, Spain

Background/Objectives: Peutz-Jeghers syndrome (PJS) is a rare autosomal dominant disorder characterized by the development of hamartomatous polyps in the gastrointestinal tract and distinctive mucocutaneous pigmentation. Genetic mutations in the serine/threonine kinase 11 (STK11) gene have been implicated in the pathogenesis of PJS. Here, we present a case report of a novel de novo mutation in the STK11 gene associated with PJS, highlighting the importance of genetic testing in diagnosis and management.

Methods: Sophia Genetics Custom Hereditary Cancer Solution targeted genomic sequencing (TS) was performed on Illumina MiSeq and the resulting data was processed and analyze using Sophia DDM software.

Results: A novel loss-of-function mutation was identified, NM_000455.4:c.701del p.(Phe234Serfs*53), in heterozygosity, in exon 5 of STK11 gene. Further analysis confirmed de novo nature of the mutation.

Conclusion: We report the case of an 16-year-old female patient presenting with hamartomatous polyps and mucocutaneous pigmentation consistent with a clinical diagnosis of PJS.

The identification of a novel de novo mutation in the STK11 gene expands the spectrum of genetic variants associated with PJS. This case underscores the importance of considering de novo mutations in the genetic evaluation of patients with suspected PJS, especially in the absence of familial history. Additionally, our findings highlight the utility of TS in uncovering rare genetic variants underlying rare hereditary syndromes like PJS, facilitating accurate diagnosis and personalized management strategies.

Grants:

Conflict of Interest: None declared

EP01.091 The Association between MUTYH Gene Variants and Colorectal Cancer

Asli Subasioglu 1, Roya Gasimli2

1Izmir Katip Çelebi University, Faculty of Medicine, Department of Medical Genetics, İzmir, Türkyie; 2Ege University, Faculty of Medicine, Department of Medical Biology, Izmir, Türkyie

Background/Objectives: Familial cancers arise from intricate genetic and epigenetic factors, with notable focus on MUTYH gene variants. This gene encodes the DNA glycosylase enzyme involved in DNA base excision repair and is specifically associated with colorectal cancer and adenomatous polyposis. Our primary objective is to systematically document varied pathogenicity profiles of MUTYH gene variants, aiming to identify specific variants for advancing diagnostic, therapeutic, and prognostic strategies in familial cancers, with a specific focus on colorectal cancers. Disseminating these findings in scholarly literature aims to enrich the scientific discourse on molecular markers indicative of familial cancer predisposition.

Methods: The study utilized peripheral blood samples from 3036 individuals with a hereditary cancer and colorectal cancer history. Library preparations involved DNA samples from peripheral blood, utilizing Qiaseq Targeted Panels kits under optimal conditions. After quality assessments meeting concentration criteria, sequencing occurred on the Illumina Miseq platform. Raw data underwent analysis, producing a variant call format (vcf) file via the QIAGEN CLC Genomics Workbench. The Qiagen Clinical Insight (QCI Interpret) system discerned variants potentially implicated in familial cancers.

Results: In accordance to our findings, a comprehensive analysis revealed a total of 155 mutations in the MUTYH gene, comprising 79 pathogenic (P), 5 likely pathogenic (LP), and 71 variants of uncertain significance (VUS).

Conclusion: These results offer valuable insights into the genetic landscape, deepening our understanding of the potential implications of MUTYH gene variations in the context of familial cancers and colorectal cancer susceptibility.

Grants: None.

Conflict of Interest: None declared

EP01.092 Actionable secondary findings in genes related to cancer phenotypes in 2020 whole exome sequenced Turkish participants

AYBERK TURKYILMAZ 1, KUBRA ADANUR SAGLAM1, Oguzhan Demir1, Mustafa Yilmaz1, Tuna Apuhan1, Alperhan Cebi1

1Karadeniz Technical University, Medical Genetics, Trabzon, Türkyie

Abstract

Background: Big data generated from whole exome sequencing (WES) and whole genome sequencing (WGS) analyses can be used to detect actionable and high-penetrance variants that are not directly associated with the primary diagnosis of patients but can guide their clinical follow-up and treatment. Variants that are classified as pathogenic/likely pathogenic and are clinically significant but not directly associated with the primary diagnosis of patients are defined as secondary findings (SF). The aim of this study was to examine the frequency and variant spectrum of cancer related SF in the Turkish population and to discuss the importance of the presence of cancer related SF in at-risk family members in terms of genetic counseling and follow-up.

Methods: A total of 2020 patients from 2020 different families were evaluated by WES.

Results: SF were detected in 20 unrelated cases (0.99%), and variants in BRCA2 (11 patients) and BRCA2 (2 patients) genes were observed most frequently. A total of 17 different variants were identified, with 4 of them (c.9919_9932del and c.3653delG in the BRCA2 gene, c.2002A>G in the MSH2 gene, c.26_29delTGCC in the TMEM127 gene) being novel variations. In 3 different families, c.1189C>T (p.Q397*) variation in BRCA2 gene was detected, suggesting that this may be a common variant in the Turkish population.

Conclusion: This study represents the largest cohort conducted in the Turkish population, examining the frequency and variant spectrum of cancer-related SF. Genetic testing conducted in family members is presented as real-life data, showcasing the implications in terms of counseling, monitoring, and treatment through case examples.

Grants: None.

Conflict of Interest: None declared

EP01.093 Exploring the Influence of HER2 Negative Breast Cancer Cell Secretome on Healthy Epithelial Cells

Busra Yasa Cevik 1;2, gülşah tuna1;2, fadime didem can trabulus3, Selcuk Sözer Tokdemir1;2

1Institute of Health Sciences, Istanbul University, Istanbul, Türkyie; 2Aziz Sancar Institute of Experimental Medicine, Istanbul University, Department of Genetics, Istanbul, Türkyie; 3Bahçeşehir University Faculty of Medicine, Department of General Surgery, Istanbul, Türkyie

Background/ Objectives: Breast cancer ranks as a leading cancer diagnosis among women. A key aspect of cancer progression is the ability of cancer cells to manipulate their surrounding micro and macroenvironments, facilitating metastasis. This study explores the functional impacts exerted by the secretome of the HER2 negative breast cancer cell line, MCF7, on healthy breast epithelial cells (MCF10A).

Methods: In our experimental setup, one-fifth of the spent media from MCF7 cells, known for their aggressive proliferation and secretion of various compounds, was introduced to MCF10A cultures. Post 48 hours of co-culturing, MCF10A cells were assessed for alterations in functional characteristics. We employed xCELLigence for real-time proliferation tracking (every 15 minutes over 144 hours), scratch assays for migration analysis (every 12 hours for 48 hours), LEGENDplex™ ELISA for cytokine profiling, and 7AAD-AnnexinV staining to evaluate apoptosis. All observations were benchmarked against control MCF10A cells.

Results: Notably, the MCF10A cells exhibited a marked increase in proliferation and IL-6 secretion (P < 0.0001). Initial 24-hour migration rates were significantly heightened. Interestingly, an increase in apoptosis rates (from 1.79% to 3.89%) was also observed.

Conclusions: The enhanced proliferation, migration, and IL-6 release in MCF10A cells suggest a potential transition towards a malignant state under the influence of the MCF7 cell secretome. Future research focusing on the composition of this spent media could illuminate the underlying mechanisms of cancerogenesis.

Grant: The study financially supported by Health Institutes of Turkey (TUSEB). (Project No: 27803)

Conflict of Interest: Busra Yasa Cevik 40, TUSEB, gülşah tuna: None declared, fadime didem can trabulus 20, TUSEB, Selcuk Sözer Tokdemir 40, TUSEB

EP01.095 uORF creation by 5’UTR variants: a new mechanism of TP53 loss-of-function

marie bequin 1;2, Luisa Vergori1;2, Vincent Milon1;2, Omar Soukarieh3;4, Caroline Meguerditchian4, Fida Khater1;2, Jonathan Dauvé1;2, Yves Delneste1, Louise-Marie Chevalier1;2, David-Alexandre Tregouët4, Gaelle Bougeard5;6, Alain Morel1;2, Isabelle Tournier1;2

1University of Angers, UMR Inserm 1307 - CNRS 6075, CRCI2NA, Innate Immunity and Cancer, Angers, France; 2Integrative Center for Oncology, Functional Genomics Unit, Angers, France; 3University of Bordeaux, INSERM, U1034, Biology of Cardiovascular Diseases, Pessac, France; 4University of Bordeaux, INSERM, UMR 1219, Bordeaux Population Health Research Center, Bordeaux, France; 5University of Rouen, UMR Inserm 1245, Cancer and Brain Genomics, Rouen, France; 6Rouen University Hospital, Genetics Department, Rouen, France

Background/Objectives: The TP53 gene is a major player in oncology, both in sporadic cancers and in hereditary cancer syndromes such as Li-Fraumeni (LFS). It is essential for cancer patients and their relatives to be able to detect and interpret all the variants detected in this gene, including alterations in non-coding regulatory regions such as the 5’ or 3’ untranslated regions (5’UTR, 3’UTR). Such variants can be pathogenic but can also correspond to genetic modifying factors. In particular, variants in the 5’UTR can affect mRNA stability, subcellular localization or its translation efficiency. They can also create upstream open reading frames (uORFs) that can compete with the main ORF and lead to haploinsufficiency by reducing the protein levels. Here, we set up a functional assay for TP53 UTR variants, and characterized the functional impact of 32 variants.

Methods: Using our luciferase reporter assay dedicated to p53, we analyzed 32 5’UTR variants detected in cancer patients or in LFS-evocating patients for their impact on protein levels.

Results: Six variants impacted the protein levels, two of them by creating uORFs. This impact could be predicted by bioinformatics tools. To evaluate these in silico predictions, we also tested a selection of artificial variants predicted to create different types of uORFs.

Conclusion: Our study highlights the need to explore non-coding regulatory regions of cancer genes both for genetic counselling and personalized medicine. It also underscores the necessity to integrate uORF prediction tools in diagnostic annotation pipelines.

Grants : La ligue contre le cancer

MB-LV-VM Equal contribution

Conflict of Interest: None declared

EP01.096 Lynch syndrome caused by di-genic events – inherited MSH6 pathogenic variant and mosaic MLH1 methylation

Natali Schachter- Safrai1;2, Inbal Kedar1, lihi atzmoni3;4, adel Shalata5, Dvora Kidron4;6, lina basel-salmon1;4;7, Zohar Levi4;8, Yael Goldberg 1;4

1Raphael Recanati Genetic Institute, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel; 2Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; 3Department of Dermatology, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel; 4Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; 5The Simon Winter Institute for Human Genetics, The Bnai Zion Medical Center Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel; 6Department of Pathology, Meir Hospital, Kfar Saba, Israel; 7Felsenstein Medical Research Center, Petah Tikva, Israel; 8The Gastroenterology Department, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel

Background/Objectives: Lynch syndrome (LS) is a cancer predisposition syndrome, associated with increased risk mainly for mismatch repair deficient (MMRd)-colorectal and endometrial cancer. LS results from a monoallelic germline mutations in one of four MMR-genes: MLH1, MSH2, MSH6 and PMS2. Inactivation of the MMR genes can also be mediated by epigenetic mechanisms, mainly EPCAM deletions or MLH1 methylation. We here report on a patient with co-occurrence of inherited MSH6 pathogenic variant and MLH1 germline methylation

Methods: A 28 y/o female presented with MMRd right CRC. She reported on a neurofibroma previously removed from her skull and had five café au lait spots. Family history was significant for cancer on both sides. Molecular analysis of the tumor included MMR immunohistochemistry, MSI analysis and NGS. Germline analysis included multigene panel testing and MLH1 promoter methylation analysis by MS-MLPA, done on two independent DNA extractions.

Results: Tumor molecular analysis showed loss of MLH1 and PMS2 expression with intact MSH2 and MSH6. Tumor molecular profile showed MSI-H, mutation burden of 22 (m/mb), wild-type BRAF, and MSH6 c.755C>G; p.Ser252* variant in 46% of the reads. There were no pathogenic variants in MLH1, PMS2 and NF1. Germline testing revealed the MSH6 c.755C>G; p.Ser252* variant, inherited from the proband’s father, along with increased MLH1 methylation, indicating mosaic germline methylation.

Conclusion: The discrepancy between tumor IHC for MMR and germline status was explained by an epigenetic event of MLH1 methylation. The combined comprehensive tumor-germline analysis contributed to understanding of the underlying genetic diagnosis.

Conflict of Interest: None declared

EP01.097 Ten years investigation of EGFR mutation screening in Non-Small Cell Lung Cancer Comparison of Real-time PCR and Reverse Strip Assay methods

Maryam Safa Isini 1, Hossein Najmabadi2, ali Salehzadeh1, Farzam Ajamian3

1Islamic Azad University, Rasht Branch, Department of Biology, Rasht, Iran; 2Faculty of Social Welfare and Rehabilitation Sciences - National University, Tajrish, Iran; 3University of Guilan - Faculty of Sciences, Department of Biology, Rasht, Iran

Background: Non-Small Cell Lung Cancer (NSCLC) is one of the cancers whose prevalence is increasing day by day. Based on studies, it has been shown that EGFR mutation is one of the main factors in the pathogenesis of NSCLC. Strip Assay method is usually used to detect EGFR mutation. Based on recent studies, it has been determined that the use of cell free DNA (cfDNA) in peripheral blood samples can detect mutations using Reverse Strip Assay (RSA), just like biopsy samples. Considering that cfDNA samples are easier to access than biopsy samples, therefore, in this study, the sensitivity of EGFR mutation detection was compared using cfDNA and biopsy samples though RSA.

Methods: Biopsy samples of 306 patients with NSCLC and 120 cfDNA sample of patients who had mutations in the EGFR gene were collected and using cfDNA by RSA and RT-PCR methods.

Results: The EGFR RSA analysis from biopsy samples, revealed 82 (26.8%) FFPE samples carry an EGFR mutation, of which the most prevalent was p.L858R (c.2573T>G) in exon 21 (5.9%), p.E746-A750del(c.2235-2249del)(5.2%) and p.E746-A750del(c.2235-2250del)(3.9%) in exon 19. One hundred and twenty cfDNA samples of affected patients were used to evaluate EGFR mutations. Also, mutations were re-checked by RT-PCR in parallel. The results showed that cfDNA StripAssay and RT-PCR methods had the same sensitivity by using cfDNA instead of biopsy.

Conclusion: In general, it can be said that cfDNA instead of biopsy samples are more suitable for EGFR mutation screening using StripAssay, which cfDNA is more convenient, non-invasive and economical for patients.

Conflict of Interest: None declared

EP01.098 Evaluation of brain and whole-body MRI surveillance service for adult individuals with pathogenic TP53 variants: Data from a regional clinical genetics service in London

Arwa Babai 1, Vicky Goh2;3, Sabrina Talukdar1;4, vishakha tripathi1;4, Anjana Kulkarni1;4, Louise Izatt1;4

1South East Thames Regional Genetics Service, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom; 2Guy’s and St Thomas’ NHS Foundation Trust, Department of Radiology, London, United Kingdom; 3King’s College London, School of Biomedical Engineering & Imaging Sciences, London, United Kingdom; 4Guy’s and St Thomas’ NHS Foundation Trust, Hereditary Breast and Ovarian Cancer Service, London, United Kingdom

Background/Objectives: Germline pathogenic variants in TP53 are associated with Li-Fraumeni syndrome and a higher lifetime risk of developing various types of cancers. The current UK Cancer Genetics Group (CGG) guidelines for carriers of clinically-actionable TP53 germline variants (‘TP53 carriers’) recommend annual screening, consisting of brain MRI (B-MRI) and whole-body MRI (WB-MRI). This retrospective service evaluation, aimed at understanding the factors that influence attendance for annual MRI screening and to assess the detection rate of cancer and incidental findings (IF) at our Centre.

Methods: The timeline to diagnosis and previous MRI reports for 26 adult TP53 carriers (7 males, 19 females) at Guy’s Hospital from 2020 to 2023 were reviewed. Data about the intervals between successive MRI scans, report findings and any extra investigations required were collected.

Results: Overall, 10 out of 26 TP53 carriers (38%) attended for screening at the recommended 12-month interval. Delayed screening was mostly due to patient-related factors, including appointment rescheduling requests, non-response to invitation letters or not tolerating the scan. IF were reported in 15 out of 26 TP53 carriers (57%). 87% of the detected IF were benign and 13% were malignant. The malignant IF were due to recurrence or metastatic disease in TP53 carriers with prior cancer diagnosis. 30% of IF required further investigations, to evaluate the nature of the screen-detected lesions.

Conclusion: A larger-scale study is recommended to further assess the detection of malignant versus benign lesions through annual MRI screening in TP53 carriers and the cost-effectiveness of annual brain and WB-MRI.

Conflict of Interest: None declared

EP01.099 Genetic Landscape of Hereditary Colorectal Cancer: Insights from Next-Generation Sequencing Analysis in a Retrospective Study

Hakan Tomac 1, Aysel Kalayci2, Deniz Agirbasli2

1Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Istanbul, Türkyie; 2Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Medical Genetics, Istanbul, Türkyie

Background/Objectives: Colorectal cancer is the third most common cancer worldwide. It is closely linked to hereditary cancer syndromes, with distinct molecular mechanisms. Our study retrospectively examined genetic variations in colorectal cancer patients, utilizing Next-generation sequencing (NGS) based-multigene panel data. We aim to uncover key genetic insights and demographic patterns, contributing to a better understanding of hereditary colorectal cancer in our population and facilitating personalized approaches for diagnosis and familial screening.

Methods: The NGS genetic panel results of 42 patients diagnosed with colorectal cancer or patients having polypoid lesions with a strong family history of hereditary colorectal cancer were retrospectively analyzed. Clinical and demographic data, along with family history, were obtained from patient reports. Variants are analyzed using the Cytoscape application to identify affected molecular networks.

Results: This study included 42 patients (n = 45.2% female and n = 54.8 % male with a mean age at diagnosis 46.3 ± 16.3 years). No identifiable variants were detected in 25 patients (59.5 %). Our findings reveal that 7 patients (16.7%) carried pathogenic variants, while 10 (23.8%) carried Variants of Unknown Significance (VUS). 5 patients (11.9%) carried multiple variants. Pathogenic/likely pathogenic variants were detected in GALNT12, MUTYH, APC, MSH6, CTNNB1 and VUS were detected in EGFR, BRCA1, APC, MLH1, NTHL1, STK11, PALB2, MUTYH genes.

Conclusion: As panel testing statistically significantly increases VUS rates, variant interpretation and appropriate genetic counseling are important for clinical decision-making. The identified variants were linked to pathways related to chromosomal and microsatellite instability, as well as the Wnt and base excision repair pathways.

Conflict of Interest: None declared

EP01.100 BRCAness Phenotype and Immune Microenvironment in Triple-Negative Breast Cancer

LUISA AYELEN RAMOS NAVAS 1;2, Beatriz Esquivel Vázquez3, Eduardo Salido Ruiz1;2;3

1Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), San Cristóbal de La Laguna, Spain; 2Universidad de La Laguna, Escuela de Doctorado y Estudios de Posgrado. Ciencias médicas y farmacéuticas de la vida, San Cristóbal de La Laguna, Spain; 3Complejo Hospitalario Universitario de Canarias, Anatomía Patológica, San Cristóbal de La Laguna, Spain

Background: Breast cancer, with over 2.26 million new cases in 2021, stands as the most prevalent cancer globally. Triple-negative breast cancers (TNBCs), constituting 10–20% of cases, present aggressive features, and a challenging prognosis, prompting intensive research into their characterization and identification of therapeutic targets. Recent studies have explored “BRCAness,” indicating defects in homologous recombination repair (HRR) in tumors. Notably, inactivation of BRCA1 may result not only from genetic mutations but also epigenetic alterations like promoter methylation. Simultaneously, other studies have delved into the tumor immune microenvironment’s crucial role, with immunotherapy emerging as a breakthrough in modern oncology. Understanding TNBC immune infiltrate aids in identifying immunotherapy beneficiaries and resistance factors as potential therapeutic targets.

Methods: We analyze BRCA1 gene promoter methylation by pyrosequencing, as a potential marker of BRCAness to assess its clinical relevance in therapeutic stratification. Additionally, we explore immune biomarkers (TILs, and CD8+ spatial distribution) and their potential correlation with BRCA1 methylation and HRD-related mutations.

Results: Our pyrosequencing analysis of 100 triple-negative breast cancer patients revealed that 65% of the samples exhibited methylation CpG island methylation above 15% (range 15-80%), which could indicate BRCAness phenotype. Also, preliminary findings suggest a lack of significant correlation between Tumor Infiltrating Lymphocytes (TILs) abundance and BRCA1 methylation in TNBC.

Conclusion: In our series, more than half TNBC samples show methylation of BRCA1 promoter and a similar percentage have medium-high TIL infiltration, but statistical analysis revealed no association. Despite the recognized importance of both factors, initial results hint at a complex relationship requiring further analysis.

Conflict of Interest: LUISA AYELEN RAMOS NAVAS Proyecto PMP22/00054, Beatriz Esquivel Vázquez: None declared, Eduardo Salido Ruiz Proyecto PMP22/00054

EP01.101 Detection of PDL1 by FISH and IHC in triple negative breast cancer

Beatriz Esquivel Vázquez 1, LUISA AYELEN RAMOS NAVAS2, Eduardo Salido Ruiz1;2

1Complejo Hospitalario Universitario de Canarias (CHUC), Anatomía Patológica, La Laguna; 2Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), U740, San Cristóbal de La LAguna, Spain

Background/Objectives: the aim of this study was to compare results obtained analyzing gene and protein status of PD-L1, by fluorescence in situ hybridization (FISH) and immunohistochemistry (IHC) techniques in 50 breast cancer samples previously categorized as triple negative. This is an aggressive and heterogeneous subtype, characterized by the absence of expression of HER2 and hormone receptors, estrogen and progesterone. PD-L1, as programmed death ligand 1, has been identified as a potential marker of response to immune checkpoint inhibitors in several cancer types, including triple-negative breast cancer. Then, it would be a prognostic and predictive biomarker.

Methods: for the FISH technique, the ZytoLight SPEC CD274, PDCD1LG2/CEN9 Dual Color Probe (Zytovision) was used, while IHC was performed using specific antibodies for PD-L1 (Roche, both SP142 and SP263). PD-L1 expression levels were evaluated and the concordance between both techniques will be considered.

Results: preliminary results showed high overall agreement between FISH and IHC in the detection of PD-L1. Only cases were observed where it was possible to differentiate by FISH whether it was a true amplification or polysomy of chromosome 9. We would like to check if the response to immunotherapy varies in PD-L1 positive due to amplification or positive due to polysomy.

Conclusion: it is concluded that IHC is an economical and sensible enough technique to be used in routine diagnostics. The study of this molecule using FISH could become clinically useful if prognostic or treatment variations are demonstrated depending on the biological event responsible for PD-L1 overexpression.

Conflict of Interest: None declared

EP01.102 HOXB13 new founder mutation in galician (North West Spain) prostate cancer patients?

Javier Manuel Galego Carro 1;2, Marta Santamariña1;2;3, Miguel E. Aguado-Barrera1;2, Jorge Amigo-Lechuga1;2, Olivia Fuentes1;2, Ana Crujeiras1;2, Carlos López1;2, Carla Coedo-Costa1;2, Antonio Gomez-Caamaño1;4, Ana Vega1;2;3

1Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS); 2Fundación Pública Galega de Medicina Xenómica (FPGMX); 3Centro de Investigación en Red de Enfermedades Raras (CIBERER); 4Servicio de Oncología Radioterápica, Complejo Hospitalario Universitario de Santiago (CHUS)

Background/Objectives: Recent studies have associated the HOXB13 gene with a significantly increased risk of hereditary prostate cancer and different variants have been identified in various populations. However, there is only evidence regarding the pathogenicity of the c.251G>A variant, and the clinical significance of the rest remains uncertain. In this study, we propose to investigate the HOXB13 gene in a cohort of 835 high-risk prostate cancer patients.

Methods: We studied the HOXB13 gene (NM_006361.6) in a cohort of 835 galician (North West Spain) patients diagnosed with high-risk prostate cancer. We employed next-generation technologies for DNA sequencing (Illumina Novaseq6000). Variant detection, annotation and classification was performed through the use of GATK, ANNOVAR and ACMG/AMP guidelines respectively.

Results: The pathogenic variant HOXB13 (NM_006361.6): c.251G>A, p.(Gly84Glu) was found in three patients.

We have also detected the c.383C>A, p.(Ala128Asp) variant in one patient, the c.649C>T, p.(Arg217Cys) variant in another, and the c.853T>C, p.Ter285Glnext* variant in nine patients.

Patients carrying c.853T>C variant had a significantly earlier age and advanced disease at diagnosis compared to patients without pathogenic germline cancer-predisposing variants. Moreover, we found this variant to be significantly associated with the presence of family history of prostate cancer.

Haplotype and functional analysis are ongoing and will be presented at the meeting.

Conclusion: We have identified a possible first-to-date galician founder mutation in prostate cancer. The identification of these variants opens the door to personalized risk assessment and expands the possibilities of targeted therapies.

Grants: PI19/01424,INT20/00071,IN607B,PRYES211091VEGA,FPU2022-04048.

Conflict of Interest: None declared

EP01.103 Unveiling novel antitumor properties of an anthraquinone derivative: In vitro and in silico investigations

Başak Günçer 1, varol güler2, Sacide Pehlivan3, Ahmet Mesut Şentürk4, Bilge Özerman Edis1

1Istanbul Faculty of Medicine, Istanbul University, Department of Biophysics, İstanbul, Türkyie; 2Institute of Graduate Studies in Health Sciences, Medical Biology, İstanbul, Türkyie; 3Istanbul Faculty of Medicine, Istanbul University, Medical Biology, İstanbul, Türkyie; 4Faculty of Pharmacy, Istanbul Biruni University, Pharmaceutical Chemistry, İstanbul, Türkyie

Background/Objectives: Anthraquinone derivatives have gained traction in cancer research due to their potent anticancer properties. Our study aims to explore the antitumor potential of compound 3, a newly synthesized anthraquinone derivative, specifically targeting MDA-MB-231 breast cancer cells.

Methods: We initiated our investigation by assessing compound 3’s drug-likeness through in silico ADME analysis. Furthermore, we investigated its interaction with therapeutic target proteins via molecular docking. To evaluate its antitumor effects, we conducted proliferation assays using MTT, ROS elevation assays using DCFH-DA, migration assays employing wound healing techniques, and cytoskeleton analysis via immunofluorescence microscopy.

Results: Compound 3 demonstrated lower energy scores compared to reference drugs for targets PCNA, CK2, LC3, and MMP2, indicating its potential efficacy. In vitro analysis revealed significant inhibition of cellular proliferation, accompanied by a notable increase in reactive oxygen species (ROS) generation. Furthermore, the derivative displayed a marked reduction in migration capability and induced morphological alterations in the actin filament of the cell cytoskeleton. These findings collectively highlight the robust antitumor effects of compound 3, suggesting its promising therapeutic potential in combating cancer progression.

Conclusion: The comprehensive results underscore compound 3’s multifaceted mode of action, revealing promising therapeutic mechanisms. These findings position it as a compelling candidate for further development as a targeted anticancer agent, with broad potential applications across various cancer types. Compound 3 shows promise in hindering proliferation, inducing ROS, and impeding migration, warranting deeper investigation in preclinical and clinical settings.

Grants: This study was supported by the Scientific Research Project Coordination Unit of Istanbul University under Grant [number 39220].

Conflict of Interest: None declared

EP01.104 PALB2 variants among suspected hereditary breast cancer patients in Pakistan

Shamila Ladak 1;1, Fizza Akbar2, alizeh fatimi1, bassim zahid1, Salman Kirmani2

1Medical College, Aga Khan University Hospital, Karachi, Pakistan; 2Division of Women and Child Health, Aga Khan University Hospital, Karachi, Pakistan

Background/Objectives: Partner and localizer of BRCA2 (PALB2) is a moderate-risk gene in hereditary breast and ovarian cancer (HBOC) syndromes, with an estimated penetrance at 41-60%. We retrospectively reviewed PALB2 variants and their clinicopathological features among suspected hereditary breast cancer patients at the Aga Khan University Hospital, Pakistan.

Methods: Patients were identified from a cohort of 751 individuals attending Genetics clinics between 2017 and 2023, who underwent multi-gene panel testing for breast and gynecologic cancers. Testing was outsourced to certified commercial genetics laboratories – Prevention Genetics and Invitae Genetics.

Results: 18 female patients (2.4%) had a PALB2 variant, of which eight (44%) harbored pathogenic/likely pathogenic (P/LP) variants while 10 (66%) carried Variants of Uncertain Significance (VUSs). Mean age at diagnosis was 51 ± 17 years. Among P/LP variants, 56% of tumors were HR+ Her2-, 22% were triple negative, and the rest exhibited varying immunohistochemistry patterns. Histologically, most prominent was invasive ductal carcinoma (62.5%), followed by invasive lobular carcinoma (25%). Most tumors were Grade 2 (56%),followed by Grades 3 (33%) and 1 (11%). Patients harboring P/LP variants were more likely to have Grade 2 or above cancers than those with VUSs (p < 0.05). Family history was positive among 75% for breast and/or gynecological cancers.

Conclusion: PALB2 variants constituted 5% of breast cancer patients with positive genetic results, the highest statistic ever reported from Pakistan. Including PALB2 in gene testing panels for breast cancer patients, for tailored surveillance and targeted interventions, will lead to a more comprehensive understanding of breast cancer susceptibility across various populations.

Grants: Non-funded.

Conflict of Interest: None declared

EP02 Reproductive Genetics

EP02.001 Screening for pharmacogenetic defects in men with azoospermia

Svetlana Yovinska 1, Mariela Hristova-Savova2, Yuri Buchvarov2, Petya Andreeva2, Tanya Timeva2, Atanas Shterev2, Rumen Nikolov1, Ivanka Dimova3

1Medical University Sofia, Department of Pharmacology and Toxicology, Sofia, Bulgaria; 2SAGBAL “Dr. Shterev” Hospital, Sofia, Bulgaria; 3Medical University Sofia, Department of Medical Genetics, Sofia, Bulgaria

Background/Objectives: Non-obstructive azoospermia is determined in 5-10% of the infertile men, being the most severe form of male factor infertility. Evidence is currently accumulating for the impact of pharmacogenetic defects on spermatogenesis, contributing for idiopathic azoospermia. The aim of our study was to provide a screening for pharmacogenetic defects in infertile men and to determine the relationship between pharmacogenetic polymorphisms and non-obstructive azoospermia among Bulgarian population.

Methods: We performed a real-time PCR for the detection of the following pharmacogenetic polymorphisms: CYP2C19 c.681G>A (n = 20), ABCB1 c.3435 C > T (n = 16), and MTHFR c.677C>T and c.1298A>C (n = 71) in men with non-obstructive azoospermia. Allele and genotype frequencies were compared between our patients and European population database.

Results: The results revealed no significant difference in the frequency of CYP2C19 and MTHFR C677T polymorphisms (alleles and genotypes) between Bulgarian azoospermia patients and European population. We detected statistically higher frequency for the ABCB1 c.3435 T/T homozygotes – 56% in our patients vs 27% in control European population (p < 0.01). Regarding MTHFR c.1298A>C polymorphism, there was a trend for significantly higher frequency of C/C genotype in patients compared to controls (17% vs 10%, p < 0.08).

Conclusion: The ABCB1 gene encodes the intestinal efflux transporter P-glycoprotein, which prevents the accumulation of toxic substances in different organs, including gonads. Its connection to male infertility is worthy of investigation. Methylenetetrahydrofolate reductase has a crucial role in the folate and homocysteine metabolism. Future studies are recommended to understand better the influence of MTHFR activity and its relation to azoospermia.

Grants:

Conflict of Interest: None declared

EP02.002 Association of autosomal recessive Von Willebrand Disease (VWD) and Recurrent Miscarriage (RM)

Emanuele Micaglio1, Arianna Riva2, Alice Tamma 2, Marco Villa1, Sara Benedetti1, Carlo Pappone1, Ugo Sorrentino2, Daniela Zuccarello2

1IRCCS Policlinico San Donato, Arrhythmia and Electrophysiology Department, Milan, Italy; 2Padua University Hospital, Women’s and Children’s Health Department, Padua, Italy

Background/Objectives: During pregnancy, VWF plays crucial roles in hemostasis and placentation. Its reduced availability can disrupt these processes in women affected by VWD. However, little is known regarding the association between VWD and RM.

Methods: We followed up 30 women (aged 18 - 34 years old) clinically affected by VWD, genetically confirmed in 28. We found 25 cases of two or more consecutive miscarriages, 20 in the first and 5 in the second trimester of pregnancy. Patients were matched with a control population of 30 either non-carrier or heterozygous women in a comparable age interval. Couple consanguinity, twin pregnancies, gluten sensitivity, Leiden F V, G20210A variant and common causes of autoimmunity have been ruled out. Only the five women with second-trimester miscarriages performed karyotyping, showing chromosomal abnormalities in two unrelated cases.

Results: Six recurrent pathogenic variants in the VWF gene (c.50dup, c.257T>A, c.2561G>A, c.3507T>G, c.6890C>T, c.7940delC) in 28 patients, either in homozygous or in compound heterozygous state, appeared to be correlated with miscarriages at an earlier age and time of pregnancy than in controls. Moreover, 11 out of 30 women showed thrombocytopenia associated with specific truncating mutations of the VWF gene (c.50dup, c.2435delC, c.7940delC).

Conclusion: Our results indicate that VWD seems to be a risk factor for RM, suggesting that early diagnosis and treatment could improve reproductive outcomes in affected women. However, genetic and chromosomal abnormalities could not be ruled out in all women because karyotyping was not available, and thus we could not exclude other causes of abnormal placentation.

Conflict of Interest: None declared

EP02.003 Investigating the association between DMPK CTG repeats and female hypogonadism in patients affected by Type 1 Myotonic Dystrophy

Emanuele Micaglio 1, Arianna Riva2, Alice Tamma2, Marco Villa1, Ludovico Sabatelli1, Rosanna Cardani1, Sara Benedetti1, Carlo Pappone1, Ugo Sorrentino2, Daniela Zuccarello2

1IRCCS Policlinico San Donato, Arrhythmia and Electrophysiology Department, Milan, Italy; 2Padua University Hospital, Women’s and Children’s Health Department, Padua, Italy

Background/Objectives: Type 1 Myotonic Dystrophy (DMT1) is a degenerative genetic disorder caused by CTG repeats in the DMPK gene. While DMT1 is a well-known cause of male hypogonadism, the relationship between DMPK CTG repeats and female hypogonadism is almost unexplored.

Methods: We performed a cross–sectional risk study in 60 women, with a mean age of 31.5 years (18 to 45 years), with either suspected DMT1 or hypogonadism. We carried out a DMPK–CTG analysis to investigate the correlation between CTG and hypogonadism. Based on the analysis, 11 women were diagnosed with DMT1 with associated hypogonadism, 11 with DMT1 without hypogonadism, 22 did not show DMPK expansion but were affected by hypogonadism, while the remaining 16 showed neither DMPK expansion nor hypogonadism. The patients affected by DMT1 performed standard karyotyping, FRAXA and FRAXE genetic testing to exclude alternative genetic causes of premature ovarian failure.

Results: Our results indicate that hypogonadism severity in DMT1 patients may correlate with CTG repeat size. DMPK CTG repeats ranging from 150–1000 (range E2) were associated with milder forms of hypogonadism, while 1000 CTG repeats or more (range E3) were correlated with hypergonadotropic hypogonadism with both infertility and assisted reproduction failure. However, no statistical significance was reached.

Conclusion: To the best of our knowledge, this is the first study to show a possible correlation between female hypogonadism and DMPK CTG repeat size in female patients with DMT1, representing a first step in understanding a hitherto misrecognized clinical consequence of the disease.

Conflict of Interest: None declared

EP02.004 Extended DNA screening for HPV infection in Bulgarian females and males

Slavena Davidova 1;2, Mariela Hristova-Savova2, Kalina Belemezova2;3, Petya Andreeva2;4, Atanas Shterev2, Jasmina Jeleva5, Ivanka Dimova2;3

1New Bulgarian University, Sofia, Bulgaria; 2Medical complex “Dr. Shterev”, Sofia, Bulgaria; 3Medical University of Sofia, Sofia, Bulgaria; 4South-West University Neofit Rilski, Blagoevgrad, Bulgaria; 5SMDL Kandilarov, Sofia, Bulgaria

Background/Objectives: Human papillomavirus (HPV) infection is the cause for more than 95% of cervical cancers. Vaccination can significantly reduce the number of HPV-related cancers in the future, but the vaccine does not protect against all types of HPV. With the extended HPV genotyping we can cover many more infected individuals and provide better prevention and early treatment.

Methods: We genotyped 21 HPV types by real-time polymerase chain reaction, using HPV Quant-21 kit (DNA Technology LCC), in 1000 patients, attended reproductive clinic – 931 women and 69 men.

Results: Totally, we detected a positive result for HPV in 40.9% of women and 36.2% of men. There were patients positive for multiple types of HPV: 19.4% of women and 28% of men were positive for two types; for more than three types - 17.9% and 20%, respectively. The most prevalent types in women were HPV 31, 16 and 45, and in men – HPV 16, 52 and 51. The types, included in the vaccine Cadasil-9, comprise 88% of male cases and 85% of female cases.

Conclusion: HPV affects men and women at almost the same rate. About half of the positive men and 37% of positive women are affected by more than one HPV type. Around 12-15% of positive cases are affected by types of HPV, not included in the current vaccines. HPV genotyping is an invaluable tool for prophylaxis of cervical cancer.

Conflict of Interest: None declared

EP02.005 Identification and characterisation of a novel pathogenic KISS1R in-frame deletion in two siblings with Idiopathic Hypogonadotropic Hypogonadism

Clayton Axiak 1, Francesca Borg-Carbott1, Ritienne Attard1, Clara Lazzaretti2, Carmela Perri2, Lara Baschieri2, Karen Cassar3, Mark Gruppetta3;4, Josanne Vassallo3;4;5, Livio Casarini2, Stephanie Bezzina Wettinger1;5, Rosienne Farrugia1;5

1L-Università ta’ Malta, Department of Applied Biomedical Science, Msida, Malta; 2University of Modena and Reggio Emilia, Department of Biomedical, Metabolic and Neural Sciences, Modena, Italy; 3L-Università ta’ Malta, Department of Medicine, Msida, Malta; 4Mater Dei, Division of Endocrinology and Diabetes, L-Imsida, Malta; 5L-Università ta’ Malta, Centre for Molecular Medicine and Biobanking, Msida, Malta

Background/Objectives: Idiopathic Hypogonadotropic Hypogonadism (IHH) is a rare genetic disorder characterized by delayed or absent puberty due to impaired gonadotropin-releasing hormone signalling. Pathogenic variants in KISS1R, a key regulator of reproductive function that plays a crucial role in the activation of the hypothalamic-pituitary-gonadal axis, cause autosomal recessive IHH. Our objective was to identify and functionally characterise IHH variants in two Maltese male siblings.

Methods: Illumina HTS data for known IHH genes was generated for the probands. Bioinformatic analysis and in silico predictions were employed to prioritize variants. Bioluminescence Resonance Energy Transfer (BRET) and Homogenous Time-Resolved Fluorescence (HTRF) assays for IP1, Ca2+ mobilisation and cAMP were used for functional characterisation in transfected HEK293 cells after metastin stimulation.

Results: We identified a novel 10 amino acid in-frame deletion: KISS1R p.Y190_A199del (rs770541847). In silico modelling suggested a deleterious effect on protein function. One proband is homozygous for the variant and with hCG treatment successfully achieved transient fertility, whilst the other brother is heterozygous and has not had offspring despite treatment. Functional studies confirmed impaired receptor signalling. These findings establish a direct link between the novel KISS1R variant and the pathogenesis of IHH in these siblings.

Conclusion: The KISS1R p.Y190_A199del is a novel in-frame pathogenic variant which deletes half of the third extracellular domain, altering the ligand binding cavity and ablating a critical disulphide bridge. The heterozygous sibling potentially has still unidentified variant(s) in a different IHH gene, further highlighting the complexity of IHH.

Grants: The Malta NGS project R&I-2012-024; Genomics of Rare Diseases I21LU04; Trialect Traineeship.

Conflict of Interest: None declared

EP02.006 NextGenerationEU — NRRP italian project “Development of the Italian Preimplantation Genetic Test (PGT) network”

Giulia Di Cola 1, Giuseppe Castello1, Chiara Faggionato1, Simone Bolli2, Cinzia Di Monte2, Monica Mazzola2, Federica Cariati3, Claudia Cartolano4, Jessica Parrotta4, Alessandro Conforti3, Alessandra Andrisani1, Roberta Venturella4, Giulia Scaravelli2, Carlo Alviggi3, Daniela Zuccarello1

1Azienda Ospedale Università, PGT Unit-UOC Genetica Clinica, Padova; 2Istituto Superiore di Sanità, Registro Nazionale PMA, Roma; 3Azienda Ospedaliera Universitaria Federico II, UOS PMA- DAI Materno-Infantile, Napoli; 4Università Magna Graecia, UO di Ginecologia Universitaria-Dip. Medicina Sperimentale e Clinica, Catanzaro

Background/Objectives:Preimplantation Genetic Testing (PGT) allows the analysis of embryo before its transfer into the uterus. In Italy, only 9 out of 71 public Assisted Reproduction Technology (ART) centers currently offer PGT, limiting access to patients. The NRRP project aims to enhance the accessibility and effectiveness of PGT across Italy, promoting a more widespread and equitable distribution.

Methods:In order to establish a comprehensive public network of ART-PGT centers, an HUB and SPOKE model will be set; standardized PGT protocols and key performance indicators (KPIs) will be defined; data from single PGT cycles will be collected by the Italian ART Registry (IARTR).

Results:Since the start of the project in May 2023, significant progresses have been made, including the opening of the PGT unit at Padua Hospital, the training of embryologists and molecular biologists in all 4 operative units, and the organization of embryo biopsy courses in collaboration with the Italian Society of Embryology (SIERR). PGT data collection started in January 2024, and four webinars on PGT applications and the National PGT Congress are planned for 2024.

Conclusion:The project will improve accessibility and effectiveness of PGT in Italy allowing access to ART-PGT in several public Hospitals. The identification of KPIs and protocol standardization will assess procedure quality, establishing certified guidelines. The IARTR’s single cycle data collection will provide transparent information to enable couples to make informed family planning decisions.

Grants:This work was funded by the European Union—NextGenerationEU—NRRP M6C2— Investment: 2.1, “Enhancement and strengthening of biomedical research within the NHS”, grant number PNRR-MR1-2022-12376108.

Conflict of Interest: None declared

EP02.007 Clinical applicability of next-generation sequencing in clinical exome

Michala Hrabíková 1, Štěpán Chvojka1, Leona Cerna1, Jan Král1, Filip Zembol1, Barbora Honysová1, Martina Bittoova1, Monika Koudová1

1GNTlabs by GENNET, Molecular Genetics Laboratory, Prague 5, Czech Republic

Background/Objectives: Next generation sequencing (NGS) and Franklin Genoox (for secondary and tertiary analysis) are together very good tools for analyzing and evaluating of germline variants from multiple genes associated with HPO terms of patients.

Methods: We use the clinical exome GERDA (Gennet Exome for Rapid Diagnostic Assessment) for prenatal and postnatal diagnosis suitable especially in cases with a clear phenotype for which we assume a genetic etiology. It includes 7961 genes. We evaluate genes connected to HPO term including CNV analysis (sequencing coverage analysis and Franklin-Rainbow). Our standard is reporting variants for the reproductive risk of partners from the CarrierTest by Gennet [1], variants from the panel Czecanca (CZEchCAncerpaNelforClinicalApplication) [2] and variants from ACMG guidelines for secondary findings. Franklin integrates publicly available data from population, disease, and sequence databases and published literature, and employs machine-learning to derive a prediction aggregation score and adjust the weighting of ACMG classification criteria. It helps in order to reach a conclusion about the patogenicity of variants.

Results: The total number of so far processed samples was 670 (458 families). 61 families of them formed the prenatal clinical exome where we found genetic causality in 33%. 397 families of them formed the postnatal clinical exome where we found genetic causality in 19%.

Conclusion: The success of finding a causal variant is dependent on the precise definition of the phenotype in particular in prenatal analysis. We would like to present some interesting case history of our clinical exome.

[1] https://www.gennet.cz/en/carriertest

[2] Soukupova et al., 2018, https://doi.org/10.1371/journal.pone.0195761

Grants: none

Conflict of Interest: None declared

EP02.008 BMP15 genetic variants associated to Mayer-Rokitansky-Küster-Hauser syndrome

Nouha ABDELMOULA 1, Balkiss Abdelmoula1

1Medical University of Sfax, Genomics of Signalopathies at the Service of Precision Medicine LR23ES07, Sfax, Tunisia

Background/Objectives: We reported a Tunisian female patient harboring Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome and we discussed genotype-phenotype correlations related to two BMP15 genetic variants.

Methods: Clinical characteristics of the affected patient were identified. Cytogenetic and molecular analysis of the BMP15 gene were carried out. Genotype-phenotype correlation was investigated using literature and mutations databases.

Results: A 20 -year-old Tunisian female consulted for primary amenorrhea. She had a facial dysmorphism and features suggestive of Turner syndrome including short stature, low posterior hairline, neck webbing, broad chest, numerous pigmented naevi and hemangiomas, cubitus valgus and dental overcrowding. Her external genitalia and breast development appeared as completely normal feminine structures. Hormonal analysis confirmed a normogonadotropic normogonadism. US exploration revealed intra-abdominal normal ovarian anatomy with a complete aplasia of the uterus and the superior part of the vagina. Coelioscopy showed two ovaries with normal size and follicles with rudimental uterine hypoplasia and two rudimental Fallopian tubes. The two kidneys were normal. The diagnosis of a MRKH II was established. The karyotype revealed a 46,XX formula. The BMP15 sequencing revealed two heterozygous genetic variants c. 788insTCT and c. 852C > T.

Conclusion: Here, we demonstrated the presence of BMP15 genetic variants associated to Turner stigmata and Müllerian duct development disorders, while ovaries and kidneys development and functions were in the normal range. BMP15 variants were reported in many populations, but it is unproved whether these variants are unique to POF cases or present in controls as well. The variant c. 852C > T was observed in patients and controls in a recent Syrian study.

Grants: No

Conflict of Interest: None declared

EP02.009 Genotype-phenotype correlation in pure and mosaic trisomy X

Balkiss Abdelmoula 1, Khaled Trigui1, Wided Ltaif1, Kays Chaker1, Yassine Ouanes1, Nouha ABDELMOULA1

1Medical University of Sfax, Genomics of Signalopathies at the Service of Precision Medicine LR23ES07, Sfax, Tunisia

Background/Objectives: Although pure 47,XXX triple X syndrome is rare, numerous mosaic trisomy X formulas exist, encompassing diverse combinations. This study sought to identify the types and frequency of trisomy X observed in women undergoing cytogenetic investigation for reproductive issues.

Methods: We conducted a retrospective analysis of patients referred to our genetic counseling at the medical university for female reproductive issues and selected women harboring 47,XXX cell lines. Clinical data were extracted from cytogenetic reports.

Results: Seven women (average age: 32.85 years) were enrolled in the present study. Trisomy X was homogeneous in only one patient, predominant in a case of 45,X/47,XXX mosaicism, while the remaining five cases exhibited low-level trisomy X mosaicism ranging from 4 to 8% with two types: 46,XX/47,XXX and 45,X/46,XX/47,XXX. The individual with pure triple X syndrome exhibited unusual physical dysmorphia and a tall stature. The individual with a 45,X[5]/47,XXX[45] karyotype displayed a short stature and several Turner’s syndrome features, excepting dysmorphic facial appearance. For patients with low-level mosaicism of 47,XXX, with and without 45,X cell line, the phenotype appeared normal, but reproductive issues varied from amenorrhea to polycystic ovaries, unexplained infertility and ART failure, to the conception of malformed children.

Conclusion: The phenotypic variability observed in patients with trisomy X poses challenges in establishing genotype-phenotype correlations. Recognizing reproductive issues as indicative clinical signs of trisomy X emphasizes the need to enhance its effective cytogenetic diagnosis in female reproductive failures. The consideration of systematic FISH analysis recommendations may be proposed to improve diagnosis accuracy.

Grants: No

Conflict of Interest: None declared

EP02.010 Genomic variants and changes in methylation level in sperm DNA of infertile brothers with oligoasthenoteratozoospermia

Marta Olszewska 1, Marzena Kamieniczna1, Nijole Pollock2, Zuzanna Graczyk1, Monika Fraczek1, Katarzyna Kurek1, Alexander N. Yatsenko2, Maciej Kurpisz1

1Instytut Genetyki Człowieka PAN, Poznań, Poland; 2Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, United States

Background/Objectives: Male infertility represents varying degrees of severity, mostly with decreased semen parameters. Considering a fact that approx. half of the infertility cases has male background, and ~15% is related to genetic causes, it is essential to investigate both: genomic and epigenomic novel causative variants.

Our aim was to find genetic variants and altered methylation level regions causative for observed decreased sperm quality in two familial cases of infertile brothers with decreased sperm concentration and similar head and acrosome deformations.

Methods: Blood DNA samples from members of two families (F1: 3 brothers, F2: 4 brothers, father) were sequenced by Whole genome sequencing (WGS, Illumina NovaSeq6000), while sperm DNA were evaluated by whole genome methylome sequencing (WGMS, Illumina Infinium MethylationEPIC BeadChip 850k array). Immunofluorescence on spermatozoa for candidate genes was applied with proper antibodies (Leica DM5500, LASX software). Chromatin integrity tests on sperm were also performed (TUNEL, aniline blue, acridine orange).

Results: We have found genomic variants in a.o: ADCY10, DDX34, DDX3X, TCTN1, PABPC3, LENG9, USP8, while decresed methylation was documented for: SLC35F2, ATF3, GYS2, TBCD and VOPP1. Revealed mutations can clearly suggest the observed sperm phenotypes.

Conclusion: This study empasizes the strong need for genomic and epigenomic surveys in patients with severely decreased sperm parameters. Collating of WGS and WGMS data shows the complexity of genomic background for particular sperm parameters, microscopically identical.

Grants: National Science Centre in Poland: 2020/38/E/NZ2/00134 (MO), 2015/17/D/NZ5/03442 (MO), 2015/17/B/NZ2/01157 (MK)

Conflict of Interest: None declared

EP02.011 Revealing invisible chromosomal alterations involving pericentromeric regions of acrocentric chromosomes using specially designed FISH probes in patients with reproductive failure

Yusuf Bahap 1, Meral Yirmibes Karaoguz1, Esra Tuğ1, Thomas Liehr2, Meral Yirmibes Karaoguz1

1Gazi University Medical Faculty, Department of Medical Genetics, ANKARA; 2Friedrich Schiller University Jena University Hospital, Institute of Human Genetics, Jena, Germany

Background: Failure to achieve a healthy pregnancy or losing consecutive pregnancies is called a reproductive problem, and affects approximately 15% of couples. Conventional cytogenetic techniques are still crucial for uncovering microscopic chromosomal changes in these patients, but there is still unclear territory for non-microscopic ones. This project aims to identify submicroscopic changes in the pericentromeric regions of acrocentric chromosomes.

Methods: In this ongoing project, 20 couples with infertility or recurrent pregnancy loss (without evidence of non-genetic factors or on routine genetic testing), were re-examined. Fluorescent in situ hybridization (FISH) analysis was performed by means of newly designed probes that stain the pericentromeric regions of acrocentric chromosomes (13-15, 21, and 22) and their alpha-satellite regions. The brief abstract of the first results was presented at 15th National Congress of Medical Genetics in May 2022.

Results: In two couples we found aberrant patterns with the applied FISH-probe sets. An invisible reciprocal translocation emerged between Chr.15 and Chr.22. One couple never had a live birth, while the other had a dysmorphic child due to adjacent-2 segregation resulting from translocation. Additional FISH analysis of the affected child revealed three copies of the Histone Cell Cycle Regulator gene located in the 22q11 region.

Conclusion: With this study, the detection of hidden microscopic changes in acrocentric chromosomes investigates the causal cause of reproductive problems and also offers the opportunity for preimplantation and/or prenatal genetic diagnosis.

Grants: The Scientific and Technological Research Council of Turkey (TÜBİTAK) supported this project with no. 123S240 as 1002-Short-Term Support Module.

Conflict of Interest: None declared

EP02.013 Glass Syndrome Unveiled: A Unique Journey through Assisted Reproduction.

Sweta Das 1;2, Rekha Aaron2, Paramasivam g2, GAUTHAM ARUNACHAL UDUPI3, Sumita Danda2

1university of texas at austin, department of molecular biosciences, austin, United States; 2christian medical college and hospital, department of medical genetics, vellore, India; 3National Institute of Mental Health and Neuro-Sciences, department of human genetics, bengaluru, India

Glass Syndrome Unveiled: A Unique Journey through Assisted Reproduction.

Background/Objectives: Glass syndrome (OMIM #612313) is a rare genetic disorder characterized by intellectual disability and dysmorphic facial features, including down-slant of palpebral fissures, crowded teeth, cleft palate, and micrognathia. Initially described by Glass IA et al. in 1989, the syndrome was associated with an abnormal karyotype 46, XY, del(2)(q32.2q33.1). This study presents the case of a 7-year-old Indian girl diagnosed with Glass syndrome, revealing variations in clinical manifestations compared to previously reported cases. Notably, the patient was conceived through intrauterine insemination (IUI) using donor sperm.

Methods: The patient was diagnosed by clinical examination. She underwent Multiplex Ligation Probe Amplification (MLPA), identifying a 2q33.1 deletion. The finding was further confirmed through real-time polymerase chain reaction (PCR).

Results: The patient displayed classic Glass syndrome features, such as intellectual disability and dysmorphic facial characteristics. Deviations included the absence of short stature and microcephaly, alongside the presence of telecanthus and up-slanting palpebral fissures. Molecular analysis through MLPA and real-time PCR confirmed the 2q33.1 deletion, validating the Glass syndrome diagnosis.

Conclusion: This case emphasizes the clinical heterogeneity of Glass syndrome, as seen in the unique presentation of our patient. The use of IUI with donor sperm in conception added complexity in genetic counselling. The use of assisted reproductive technology, specifically IUI with donor sperm, in conception raises questions about potential genetic influences.

Grants: None

Conflict of Interest: None declared

EP02.014 LINE1-mediated epigenetic repression of androgen receptor transcription causes androgen insensitivity syndrome

Jelena Pozojevic 1, Radhika Sivaprasad2, Joshua Laß3, Franziska Haarich4, Joanne Trinh3, Neseebullah Kakar1, Kristin Schulz1, Kristian Händler1, Annemarie Verrijn Stuart5, Jacques C. Giltay6, Koen L. I. van Gassen7, Almuth Caliebe2, Paul-Martin Holterhus8, Malte Spielmann1, Nadine Hornig2

1Institute of Human Genetics, Lübeck, Germany; 2Institute of Human Genetics, Kiel, Germany; 3Institute of Neurogenetics, Lübeck, Germany; 4Institute of Cardiogenetics, Lübeck, Germany; 5, Department of Pediatric Endocrinology, Utrecht, Netherlands; 6Division Laboratories, Pharmacy and Biomedical Genetics, Utrecht, Netherlands; 7, Department of Genetics, Utrecht, Netherlands; 8, Department of Paediatrics, Division of Paediatric Endocrinology and Diabetes, Kiel, Germany

Background/Objectives: Androgen insensitivity syndrome (AIS) is a difference of sex development characterized by different degrees of undervirilized external genitalia in individuals with a 46,XY karyotype. Here, we present an undervirilized patient with a 46,XY karyotype, assigned with a female gender, with normal-for-male AMH levels at birth and normal testosterone/dihydrotestosterone ratio at age of one month following hCG stimulation.

Methods: Whole exome sequencing (WES), nanopore sequencing, quantitative PCR (qPCR), western blotting

Results: WES detected a maternally inherited L1 retrotransposon insertion in the 5´ untranslated region (5´UTR) of the androgen receptor (AR) on chromosome X. This finding coincided with reduced AR transcript and protein levels in patient-derived genital skin fibroblasts (GSF). Long-read nanopore sequencing determined the sequence and epigenetics of the L1 element and the AR promoter region where it is inserted, revealing a truncated L1 element of ≈2.7 kb. Interestingly, DNA methylation of the CpGs proximal to the L1 insertion was increased in the patient-derived GSF compared to healthy controls, suggesting that the L1 decreases AR levels by affecting its promoter methylation. To confirm this hypothesis, we compared the effects of wild-type and patient-derived AR 5´UTR, by inserting each of these sequences into a vector that contains the AR cDNA. mRNA and protein levels were decreased in the cells transfected with the L1-containing AR 5´UTR, in accordance with the results from patient-derived cells.

Conclusion: Our results show the relevance of retrotransposons in human disease. They comprise almost half of the human genome and can interfere with gene expression, especially when inserted at transcriptionally active regions.

Conflict of Interest: None declared

EP02.017 The role of gene variants for IL-6, IL-10 and TNF-α in the incidence of preterm birth

Jasenka Wagner 1, Deni Plecko1, Nena Arvaj1, Kristina Kralik2, Mirta Kadivnik3

1Faculty of Medicine Osijek, Department of medical biology and genetics, Osijek, Croatia; 2Faculty of Medicine Osijek, Department of medical statistics and medical informatics, Osijek, Croatia; 3Faculty of Medicine Osijek, Department of gynecology and obstetrics, Osijek, Croatia

Background/Objectives: The association of gene variants for IL-6 (rs1800796), IL-10 (rs1800896) and TNF-alpha (rs1800629) with the occurrence of spontaneous preterm birth was examined to determine whether these genetic variants are a risk factor or not.

Methods: 175 blood samples of pregnant women who gave birth prematurely and 169 corresponding control blood samples were analyzed with the aim of determining gene variants for IL-6 (rs1800796), IL-10 (rs1800896) and TNF-alpha (rs1800629). Control samples were samples of pregnant women with term delivery. DNA isolation was performed on mini spin columns according to the manufacturer’s protocol. The quality and purity of the isolated DNA was tested with a Qubit 3 Fluorometer. Genotyping was performed on ABI PRISM 7500 SDS using Taqman SNP Genotyping Assays. Obtained genotypes were analyzed using program package 7500 Software v2.3.

Results: Carriers of the A/A genotype for rs1800629 compared to carriers of the G/G genotype, have a 5.5 times greater chance of late preterm birth. The presence of the A/A or A/G genotype for rs1800896 in the recessive model of inheritance compared to the G/G genotype represents a potentially protective factor in which the test subjects have a 6.5 times lower chance of early preterm birth. In this study, no link was established between premature birth and variants in rs1800796.

Conclusion: Rs1800629 in mothers was associated with preterm birth. Rs1800896 in this study shows a potentially protective effect for the incidence of preterm birth. No association was established between premature birth and rs1800796.

Grants: IP3-2017 Medical Faculty Osijek

Conflict of Interest: None declared

EP02.019 Establishing a quality control in PGT-A: parameters and reference values.

Andrei Kullyev 1, Svetlana Avdeichik1, Yaron Goikhman1

1Israeli Medical Center-Reproductive Medicine and Family Health, Genetics, Tashkent, Uzbekistan

Background/Objectives: While PGT-A is arguably the most complex and challenging procedure in IVF, the only relevant KPI written in the Vienna consensus is the successful biopsy rate/tubing rate. As a result, the reported efficacy of PGT-A programs varies widely. That, in turn, led to a heated discussion about whether PGT-A is a useful tool or an unnecessary add-on in IVF treatment.

Methods: We have chosen PGT-A efficiency, defined by R.J. Paulson as post–PGT-A implantation rate/(pre–PGT-A implantation rate/euploidy rate), as our main KPI, defining competency value at ≥ 75%. Other KPIs are the percentage of euploid embryos in a group of patients under 35 years of age (competency value ≥ 60%), and the clinical pregnancy rate after transferring euploid embryos (> than the clinical pregnancy rate after transferring untested embryos for all age groups).

Results: Designing our workflow from the get-go to meet defined KPIs (non-traumatic biopsy by Flicking technique, biopsied samples storage in the freezer for no longer than 6 weeks before processing, re-testing of aneuploid embryos to detect and eliminate possible false positive results) allowed us to reach PGT-A efficiency of 86.13%. The euploidy rate for patients under 35 years was kept at over 60% from the year 2022, when our laboratory was founded.

Conclusion: For the PGT-A program to be beneficial to patients, a set of quality control points and reference values should be established in the PGT-A laboratory, and efforts should be made to reach the competency value for each KPI.

Grants: none

Conflict of Interest: None declared

EP02.020 Comparative proteomics analysis of chorionic villi reveals altered complement and coagulation cascades, TCA cycle and ferroptosis in recurrent pregnancy loss

Katarina Davalieva1, Gjorgji Bozhinovski 1, Sanja Kiprijanovska1, Katerina Kubelka-Sabit2;3, Dijana Plaseska-Karanfilska1

1Research Centre for Genetic Engineering and Biotechnology “Georgi D Efremov”, Macedonian Academy of Sciences and Arts (MASA), 1000 Skopje, North Macedonia; 2Clinical Hospital “Acibadem Sistina”, Skopje, North Macedonia; 3Faculty of Medical Sciences, University “Goce Delcev”, Stip, North Macedonia

Background/Objectives: Recurrent pregnancy loss (RPL) represents the most common disorder during pregnancy and affect ~5% of the couples aiming at childbirth with long-term consequences on family and society. As more than half of the RPL cases do not have identified cause, uncovering the mechanisms behind the idiopathic RPL is urgently needed.

Methods: Using label-free data-independent LC-MS/MS acquisition coupled with ion mobility, we compared the proteome of 13 first trimester placental chorionic villi from RPL cases with 10 age and gestational week matched chorionic villi from normal, elective pregnancies. Transcriptional levels of selected biomarkers were determined by qPCR in extended cohort of 35 RPL and 25 controls.

Results: From 1554 proteins identified based on ≥2 peptides, statistically significant difference in abundance adjusted for multiple testing (B-H p ≤ 0.05) and fold change ≥1.5 showed 128 proteins. Bioinformatics analysis identified complement and coagulation cascades, platelet activation, TCA cycle and ferroptosis as enriched pathways with the highest significance. NEFL and NOS3 were identified with highest differential abundance between RPL and controls. The results showed that the transcription levels of NEFL, DLST, NOS3 and CP were significantly increased in RLP group, consistent with the proteomics findings.

Conclusions: While blood coagulation, complement and coagulation cascades have well established associations with the pathogenesis of RPL, our study pointed to pathways such as TCA cycle and Ferroptosis, for which at present, very limited association exists. The current study brings novel insights behind the altered pathways in RPL and opens a way for investigations regarding the clinical management.

Grants: 08-707/2023 from MASA

Conflict of Interest: None declared

EP02.021 HLA-DRB1 genotyping in Bulgarian couples with recurrent implantation failure

Mariela Hristova-Savova 1, Petya Andreeva2, Ivelina Oprova2, Atanas Shterev2, Ivanka Dimova3

1SAGBAL “Dr Shterev”

, Genetic Laboratory, Sofia, Bulgaria; 2SAGBAL “Dr Shterev”

; 3Medical University Sofia

Background/objectives: The difference in spouses regarding HLA class II gene variants is one of the important conditions for successful implantation and pregnancy realization. The similarity of spouses in terms of HLA gene variants increases the probability of the appearance of an embryo with a double set of identical gene variants, i.e. HLA homozygosity, which is an unfavorable factor carrying the risk of reproductive losses. We aimed in analyzing the HLA-DRB1 genotypes in selected couples with recurrent implantation failure (RIF), i.e. more than 3 miscarriages or unsuccessful ART procedures.

Method: We used Real time PCR analysis for simultaneous detection of 13 DRB1 alleles (DNA Technology LCC), after isolation of DNA from peripheral venous blood.

Results: We analyzed eight couples for the allele variants in DRB1 locus – totally 16 genotypes and 32 alleles were detected. We established the highest frequency for DRB1*11 allele (34.4%), followed by DRB1*16 (15.6%) and DRB1*03 (12.5%). In 2 couples (25%) there was a risk for HLA-DRB1 homozygosity – the risk was 50% in one couple (genotypes of partners *03/*11 and *11/*11) and 25% in another one (genotypes *04/*11 and *04/*16, respectively).

Conclusion: Spousal HLA typing is used in diagnosing causes of reproductive failure to identify similarities in HLA gene variants in the couple. We suggest that in one fourth of couples with unexplained RIF, the contributing factor could be the HLA-II similarity.

Conflict of Interest: None declared

EP02.022 Distribution of PAI-1 5G/4G and ACE I/D polymorphism in women with recurrent pregnancy loss

Liliia Chorna 1, Danuta Zastavna1, Yaryna Zahanyach1, Oksana Kolodiy2, Halyna Makukh3

1State Institution Institute of Hereditary Pathology of the National Academy of Medical Sciences of Ukraine, Diagnostics of hereditary pathology, Lviv, Ukraine; 2KNP “3rd city clinical hospital of Lviv”, Women’s consultation, Lviv, Ukraine; 3KNP “Lviv Regional Clinical Perinatal Centre”, Regional centre of newborn screening, Lviv, Ukraine

Background/Objectives: Recurrent Pregnancy Loss (RPL) defined as two or more consecutive pregnancy losses, affecting 1–5 % of reproductive-age women. Current diagnostic procedures can identify etiologic factors in approximately 50% of RPL cases. The study aimed to assess the distribution of PAI-1 (675 5G/4G) and ACE (intron16 I/D) polymorphism in women with RPL.

Methods: A study group of 55 women with a history of at least two consecutive miscarriages was compared with a control group of 57 healthy women with normal pregnancies and without miscarriages. Polymerase chain reaction (PCR-RFLP) was used to identify the polymorphism.

Results: The frequency of 4G allele of PAI gene was higher in the RPL group (67%) compared to the control group (54%). It was found that the presence of 4G allele increases the risk of RPL by 2 times (Р = 0.01). DD genotype and D allele of ACE gene were more prevalent in RPL women (31%) than in controls (19%) although the difference was not significant. The combination of DD genotype with 4G4G genotype was significantly more frequent in RPL group compared with controls (16.6% versus 2.3%). The results showed that the presence of homozygotes for two 4G alleles of the PAI-1 gene and D alleles of the ACE gene leads to an 8-fold increased risk of RPL (OR = 8.6, CI: 0.98 -75.15, P = 0.04).

Conclusion: Our study suggest that the combination of homozygosity for the PAI-1 4G and ACE D alleles could be a risk factor to RPL for Ukrainian women.

Conflict of Interest: None declared

EP02.023 Significance of the sperm DNA fragmentation index (DFI) for male fertility diagnostics - results from more than 800 cases

Michaela Blankenburg 1, Oriane Vedrines1, Markus Stumm1

1Medicover Humangenetik Berlin-Lichtenberg MVZ, Berlin, Germany

Background/Objectives: Classical sperm analysis is based on the three semen parameters concentration, morphology and motility. The current edition of the WHO Laboratory Manual for the Examination and Preparation of Human Semen also contains a chapter on advanced tests. One group of these additional analyses are DNA fragmentation tests, which are a good extension of the basic tests.

Methods: The DFI of more than 800 semen samples were analysed with the Haloperm® test. The test results were divided into three fertilisation categories (normal, impaired, severely impaired) and correlated with the patient’s age, sperm concentration, sperm morphology and sperm motility. To analyse the significance of the results, a two-tailed t-test was performed and the correlation coefficient r was calculated for each of the parameters.

Results: The data showed a significant positive correlation between the DFI and the age of the patients, but a significant negative correlation between the DFI and the basic semen parameters concentration, morphology and motility. An exception were 15 infertile patients with high sperm DNA fragmentation and normal semen parameters. All these results indicate that an elevated DFI is a good additional parameter for poor sperm quality.

Conclusion: The Haloperm® test provides additional information on sperm quality and can help to refer patients to the most appropriate assisted reproductive technology. This is especially important for infertile patients with normal sperm parameters but with high sperm DNA fragmentation.

Grants:

Conflict of Interest: None declared

EP02.024 Deciphering the role of microRNAs (miRNAs) in azoospermia: A small RNA transcriptome analysis in testicular tissue

Maria-Anna Kyrgiafini 1, Alexia Chatziparasidou2, Nikolaos Christoforidis2, Zissis Mamuris1

1Laboratory of Genetics, Comparative and Evolutionary Biology, Department of Biochemistry & Biotechnology, University of Thessaly, Larissa, Greece; 2Embryolab IVF Unit, Kalamaria, Thessaloniki, Greece

Background/Objectives: Azoospermia, the most severe form of male infertility, affects approximately 1% of all men and 15% of those seeking infertility treatment. While genetic factors are identifiable in 25% of these cases, the vast majority remain idiopathic, posing a challenge for diagnosis and treatment. Our study aimed to identify differentially expressed microRNAs (miRNAs) in the testicular tissue of azoospermic males, aiming to reveal tissue-specific expression signatures associated with idiopathic azoospermia.

Methods: Small RNA sequencing was conducted on testicular samples from 18 IVF (in vitro fertilization) patients, organized into six groups according to the presence of spermatozoa and pregnancy outcomes. This included groups for samples with no spermatozoa, high presence of spermatozoa, and rare presence of spermatozoa with varying pregnancy results, along with specific pools for cystic fibrosis carriers and cryptozoospermic patients. Bioinformatic analysis was undertaken to identify the differentially expressed miRNAs between the different samples as well as the main pathways that are affected.

Results: Overall, approximately 20,000 miRNAs were detected and significant differences in small RNA expression (log2fold change≥2, p-value < 0.05) were observed across the analyzed pools. Bioinformatic analysis indicated that these altered miRNAs play crucial roles in metabolic processes, immune regulation, and spermatogenesis-related signaling pathways.

Conclusion: Our study investigates the complete miRNA landscape in testicular samples, revealing that variations in miRNA expression are tightly linked to sperm presence and IVF success rates, underscoring key molecular pathways that may affect male fertility. These findings suggest miRNAs as promising biomarkers and therapeutic targets for addressing male infertility.

Grants: Grant number: T1E∆K-02787

Conflict of Interest: None declared

EP02.026 Copy number variants (CNV) characterization in recurrent pregnancy losses

Evelina Dagyte 1, Algirdas Utkus1

1Vilnius University, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius, Lithuania

Background/Objectives: Recurrent pregnancy losses (RPL) is an important global health issue and carries psychological and financial burden for affected couples. Embryonic chromosomal abnormalities (both in the number of chromosomes and the structure) account for 50% of early pregnancy losses. Etiology of RPL in 40-60 % of couples remains idiopathic.

The aim of this study was to evaluate the role of embryonic chromosomal abnormalities and CNVs in the etiology of RPL.

Methods: SNP-genotyping was performed for patients with unexplained RPL (anatomy factors, antiphospholipid syndrome, chromosomal and endocrinological abnormalities were excluded). 32 products of conception (POCs) from miscarriage specimens were investigated using single nucleotide polymorphism array (SNP-array). Genomic DNA was extracted from all POCs using the phenol-chloroform extraction method. Genotyping was performed with Illumina HumanCytoSNP-12 v2.1 and Infinium Global Diversity Array with Cytogenetics-8 arrays according to standard protocols provided by the manufacturer.

Results: Normal results were identified in 15 (46,88%) cases and abnormal results were identified in 17 (53,13%) cases. In cases with abnormal results, aneuploidy was the most common finding with 11 (64,71%) cases of autosomal trisomy and 3 (17,65%) cases of monosomy X. Triploidy was found in 3 (17,65%) cases. 15 cases with normal results were subjected to further CNV analysis. 11 CNVs were identified, including 5 deletions and 6 duplications.

Conclusion: Wider comprehension of the genetic mechanisms involved in RPL may lead to the establishment of a diagnostic panel of genetic markers for screening for individuals at risk of RPL.

Grants:

Conflict of Interest: None declared

EP02.027 Preimplantation genetic screening in partners with chromosomal polymorphisms

Radostina Raynova 1;2, Tanya Milachich3, Petya Andreeva4, Tanya Timeva4, Maria Yunakova4, Atanas Shterev4, Alexey Savov1, Ivanka Dimova2;5

1University Hospital of Obstetrics and Gynecology, National Genetic Laboratory, Sofia; 2Shterev Hospital, Genetic Laboratory, Sofia, Bulgaria; 3Shterev Hospital, Embryology, Sofia, Bulgaria; 4Shterev Hospital, Obstetrics and Gynecology, Sofia, Bulgaria; 5Medical University, Sofia, Clinical Genetics, Sofia

Background/Objectives: Chromosomal polymorphisms (CPM) are variants in chromosomes generally considered “normal” karyotypes. Studies indicate that CPM may be associated with abnormal spermatogenesis, infertility, and recurrent miscarriages. Preimplantation genetic testing (PGT) is a useful approach for reducing miscarriage and increase successful pregnancy rate in such couples. The aim of our study was to determine the frequency and spectrum of chromosomal aberrations in embryos after PGT in carriers of CPM.

Methods: Nineteen couples underwent IVF cycles, followed by PGT (Perkin Elmer protocol) due to CPM - 13 with CPM of acrocentric chromosomes (Group I – average age of 33.6 years) and 6 with chromosome 9th CPM (Group II – 35.3 years on average).

Results:Overall, 92 embryos were subjected to trophectoderm biopsy – 4.8 embryos/cycle on average. We found 31 euploid embryos in total (44.1% in Group I and 33.3% in Group II) and lack of euploid in 7.7% and 33.3%, respectively; mosaic embryo was detected only in Group I (11%). Pregnancy was achieved in 58% and 50% of transfers in both groups. Single and complex aneuploidies were detected as follows: 17% and 12% in Group I, and 26% and 18% in Group II. The most frequently involved chromosomes were 2, 14 and 19 in Group I, and chromosomes 10, 19, 20 and 21 in Group II.

Conclusion:The polymorphism of chromosome 9 is associated with less euploid embryos than acrocentric chromosomal polymorphism. The last was associated with the presence of mosaic embryos. There is no correlation between polymorphic chromosome and aneuploidy

Grants: No grants.

Conflict of Interest: None declared

EP02.028 KIR AA1 genotype and recurrent implantation failure

Petya Andreeva 1;1;2, Ivelina Oprova1;2, Mariela Hristova-Savova3, Kalina Belemezova3, Ivanka Dimova4

1Shterev Hospital, IVF Unit, Sofia, Bulgaria; 2South-West University Neofit Rilski, Blagoevgrad, Bulgaria; 3Shterev Hospital, Genetic Department, Sofia, Bulgaria; 4Medical University of Sofia, Genetic Department, Sofia, Bulgaria

Background/Objectives: The killer cell immunoglobulin-like receptors (KIRs) of a subpopulation of uterine natural killer cells (uNK cells) is made up of 16 highly polymorphic genes classified into two haplotypes: KIRAA and KIRBx. The KIRAA haplotype has predominantly inhibitory KIRs and only one activating receptor, KIR2DS4, with uncertain function. The key distinction is the balance of KIRs, which can activate or inhibit uNKcells’ ability to create components essential for embryo implantation. The study intended to explore the distribution of KIR haplotypes and genotypes in patients with recurrent implantation failure (RIF).

Methods: In total, 82 patients were genotyped for activating or inhibiting KIR genes, with 28 women with RIF serving as the study group and 32 as controls. The individual KIRAA and KIRBx haplotypes have been identified via PCR with Sequence-Specific Primer (SSP).

Results: RIF was discovered in a higher proportion in haplotype KIR AA (60% RIF vs 40% controls; P = 0.2) than in KIR Bx (37.5% RIF vs 62.5% controls; p = 0.026). Furthermore, two genotypes were identified: KIR AA1 (31.82%) and KIR AA195 (68.18%). In haplotypes KIRAA, the KIR2DS4 locus in the full-length receptor (KIR2DS4-norm) was only expressed in KIR AA1. The RIF numbers were significantly higher in KIR AA1 (75% vs 15%, p = 0.01), while there was no difference between the two observed groups in KIR AA195.

Conclusion: The KIR AA1 genotype substantially increases the risk of recurrent implantation failures. The presence of the full-length receptor KIR2DS4norm in KIRAA haplotype patients implies RIF development.

Grants: Not applicable

Conflict of Interest: None declared

EP02.029 Preimplantation Genetic Testing of Aneuploidy (PGT-A) and Structural Chromosomal Imbalances (PGT-SR) by Nanopore Sequencing – initial validation experience on archival whole genome amplification samples

Matthias Linke 1, Charlotte Hewel1, Bartosz Linek2, Christine Skala2, Susanne Gerber1, Susann Schweiger1

1Institute for Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz; 2Clinic and Polyclinic for Obstetrics and Women’s Health, University Medical Center of the Johannes Gutenberg University Mainz, Mainz

Background/Objectives: Current strategies for preimplantation genetic testing for aneuploidy or structural rearrangements (PGT-A/SR) are mostly based on next-generation sequencing (NGS) and microarray platforms. Some studies have already demonstrated the feasibility of Nanopore sequencing as a PGT-A/SR screening platform for both aneuploidy and segmental imbalances.

Methods: Archival SurePlex-amplified (Whole Genome Amplification, WGA) trophectoderm biopsy samples (n = 28) previously analyzed using Vitrolife/Illumina VeriSeq PGS™ were reanalyzed using Nanopore sequencing on a PromethION P24. Libraries were prepared by using the Native Barcoding Kit 24 V14. The number of barcoded WGA products per R10.4.1 flow cell was 10 and 18 samples. Bioinformatic processing included wf-cnv based on the R package QDNAseq to call copy number aberrations.

Results: PGT-A/SR analysis based on Nanopore sequencing identified known specific aberrations in 100% of numeric chromosomal imbalances (27/27) as well as 100% (1/1) of samples with a segmental duplication (Chr.3p26.3, 2.73 Mb size). Due to the known decreasing sensitivity of NGS and nanopore-based copy number calling for imbalances smaller than 5 Mb, this copy number change was additional validated in a classical PGT-M approach. Chromosomal mosaicism previously detected by VeriSeq™, could also be validated in all of the reanalyzed samples (5/5 embryos of the PGT-A group).

Conclusion: Nanopore sequencing represents a viable alternative to current NGS-based PGT-A/SR solutions for aneuploidy and segmental imbalance screening of trophectoderm biopsy samples. Additionally, we currently investigate long-read sequencing-based SNP haplotyping for monogenic disease to replace linkage analysis and the direct readout of a classical PGT-M approach.

Grants: -

Conflict of Interest: None declared

EP02.030 Genital developement variations in a large french cohort : clinical features and genetic findings

abir talbi 1

1Robert Debré Hospital, paris, France

Background/Objectives: Genital development Variations (VDG) are a rare group of conditions affecting gonadal development, sexual differentiation, or chromosomal sex. The etiology of DSDs is very heterogenous and a precise diagnosis is essential for management of genetic, endocrine, surgical, reproductive, and psychosocial issues. The main goal of this present study was to characterize genetic defects in a large cohort of French VDG patients using a targeted NGS panel.

Methods: a cohort of 133 unrelated patients (109 Males/ 24females), recruited between 2022 and 2023, was studied using a panel of 59 known diagnostic genes of VDG. Gonadal dysgenesis, severe hypospadias and cryptorchidism are the most common clinical features of male patients while the primary amhenorrhea is the most frequent for the female VDG patients

Results: Pathogenic or probably pathogenic variants were identified in 28(21%)VDG patients but a molecular diagnosis is made in only 23(17%) of them. Variants in the NR5A1, WT1, SRY, CYP21A2, HSD17B3, AR, and SRD5A2 genes were the most common causes of VDG. 8(30%) of them are novel. Other variants were identified in genes associated with congenital hypogonadotropic hypogonadism (CHH), including the PROKR2 and GNRHR.

Forty five patients (33.8%) had rare variants including variants of uncertain significance (VUS) in more than one candidate gene

Conclusion: Genetic sequencing is increasingly applied to rare disease such as VDG but the achieved diagnostic rate of a targeted NGS panel is still insufficient to explain all the cases and must be completed with exome and whole genome analysis in order to unravel the complexity of this condition.

Grants:

Conflict of Interest: None declared

EP02.031 Non-invasive PGT: Risks and hopes

Daniil Ferdman 1, Ekaterina Vasileva1, Tutakov Maksim2, Daniil Chebotarev2, Svetlana Pavliuchenkova2, Natalia Isaeva1, Roman Bikanov1

1FIRST GENETICS JSC; 2medical genetics research center MedicaMente

Background/Objectives: Despite the high accuracy of PGT-A, it still has been remaining a significant problem associated with embryo damage risk. Non-invasive PGT (niPGT) is a technique focused on the study of extracellular DNA in a spent cultural medium (SCM). However, chromosomal DNA profile in the SCM and the real karyotype matching is questioned.

Methods: 89 SCM samples collected on the 5th or 6th day of fertilization. Whole genomic amplification (WGA) was performed using the ReproSeq™ PGS kit. WGA success revealed by agarose gel electrophoresis. Chromosome profiles were analyzed for all WGA positive SCM samples paired with invasive PGT-A using F-Genetics high-throughput sequencing system.

Results: WGA was successful for 60% SCM samples. Samples were stratified by day of SCM collection, storage duration, and chromosomal status by PGT-A (euploid vs aneuploid). The subgroup analysis demonstrated that day 6 of fertilization, less than 3 week storage and aneuploidy presence is the subgroup with minimal lack of amplification rate. Only 47% of niPGT was completely matched the PGT-A results. Contamination with cumulus or spermatozoa DNA was detected for at least 7% of niPGT samples.

Conclusion: We suggest that apoptosis in embryonic cells until the 6th day of development predominantly occurs in aneuploid cells.

The possible reason of the lack of amplification in niPGT is an euploid karyotype.

Storage SCM samples at -20°C for more than 3 weeks negatively influence on technical opportunities niPGT testing, giving higher lack of amplification rate.

Collection SCM on day 6 it better for amplification success in comparison with day 5.

Conflict of Interest: None declared

EP02.032 A successful case of preimplantation genetic testing for Hunter syndrome

Ekaterina Vasileva 1, Nataliia Andreeva1, Daniil Ferdman1, Natalia Isaeva1, Roman Bikanov1

1FIRST GENETICS JSC, Moscow, Russian Federation

Background/Objectives: Mucopolysaccharidosis type II (MPS II), also known as Hunter syndrome, is an X-linked recessive multisystem disorder. A couple with a female carrier of MPS II requested preimplantation genetic testing for monogenic disorder (PGT-M) in order to prevent pathogenic variant transmission to their offspring. Hunter syndrome with the hemizygous frameshift variant c.546dup (p.L183Afs*16) in the IDS gene was earlier diagnosed in proband’s brother. Preimplantation genetic testing for aneuploidies (PGT-A) was carried out before PGT-M because of advanced maternal age.

Methods: DNA of the proband, her partner and her parents was analyzed. The mother of the proband was also confirmed as heterozygous carrier of the same variant. Trophectoderm samples of each embryo undergone a whole-genome amplification (WGA). Fluorescent multiplex PCR followed by fragment analysis as indirect analysis and Sanger sequencing as direct analysis was used. Custom panel with fourteen polymorphic short tandem repeat (STR) markers was developed in order to determine the haplotype that associated with the disease. 4 out of 14 STR markers were defined as uninformative due to the homozygous status in the proband.

Results: One out of three embryos were identified by PGT-A as euploid with 46,XY karyotype. As a result of the subsequent PGT-M we revealed by both direct and indirect analyses that the pathogenic variant L183Afs*16 was not transmitted. No allele dropout was detected. This embryo was recommended for transfer.

Conclusion: We have developed and applied PGT-M panel for mucopolysaccharidosis type II with a high efficiency and accuracy, resulted in identification of healthy embryo in a high-risk couple.

Conflict of Interest: None declared

EP02.033 The role of mitochondrial dynamics genes in NOA

O. Sena AYDOS 1, nazila farhangzad1;2, Tulin Ozkan1, Asuman Sunguroglu1, Kaan Aydos3

1, Ankara University, Faculty of Medicine, Department of Medical Biology, Ankara, Türkyie; 2Ankara University, Institute of Health Sciences, Ankara, Türkyie; 3, Ankara University, Faculty of Medicine, Department of Urology, Ankara, Türkyie

Background: Mitochondria is a dynamic organelle that constantly undergoes fusion and fission. These dynamic processes work in concert to maintain functions of cell and cellular homeostasis. In process of spermatogenesis, the morphology of mitochondria undergoes significant modifications, and spermatogonia develop into highly specialized sperm cells that able to fertilize with perfectly coordinated, healthy mitochondria. Some of major molecules that mediate mitochondrial fusion and fission have been discovered in mammals, but there are not yet sufficient studies during spermatogenesis in humans.

Methods: Expression of mitochondrial fusion (OPA1) (MFN1) and mitochondrial fission (MFF) genes as biomarkers in mitochondrial dynamics was evaluated by RT-PCR in testicular cells taken from 8 infertile men with non-obstructive azoospermia (NOA) and 4 men with obstructive azoospermia (OA) as the control group.

Results: The expressions of MFN1, OPA1 genes were found to be increased in the NOA group compared to the OA group. Although the increase in OPA1 gene expression was not statistically significant, the expression of the MFN1 gene was found to be statistically significantly increased in the NOA group compared to the OA group.

Conclusion: MFN1 gene expression, which has been investigated for the first time in testicular cells of NOA patients, may play an important role in mitochondrial fusion in infertile men and may contribute to the identification of new biomarkers and the development of treatment protocols for infertile patients with mitochondrial dynamic defects. More clear results will be obtained when the study is concluded by increasing the number of patients.

Grants: This study was supported by Ankara University(BAP) (Project No:TDK-2022-2640)

Conflict of Interest: None declared

EP02.034 Ten infections before pregnancy and risk of pregnancy loss

Yevheniya Sharhorodska 1;2;3, Anna Ulrich2, Liliia Chorna1, Ivanna Shymanska4, Danuta Zastavna1, Oleg-Roman Gnateyko1, Chiara Scapoli3, Halyna Makukh4, Inga Prokopenko2;5

1Institute of Hereditary Pathology, National Academy of Medical Science, Clinical Genetics, Lviv, Ukraine; 2University of Surrey, Department of Clinical and Experimental Medicine, Guildford, United Kingdom; 3University of Ferara, Department of Life Sciences and Biotechnology, Ferrara, Italy; 4Scientific Medical Genetic Center “LeoGENE”, Lviv, Ukraine; 5University of Surrey, People-Centred AI Institute, Guildford, United Kingdom

The loss of two or more pregnancies before 24 weeks/at any time of gestation is defined as recurrent pregnancy loss (RPL)/recurrent miscarriage (RM). Susceptibility to infections can modify individual risk of RM/RPL. We investigated the causality between ten infectious diseases – chickenpox, shingles, mononucleosis, mumps, measles, scarlet fever, bacterial meningitis, hepatitis B, hepatitis A, COVID-19 – and subsequent risk of pregnancy loss.

For outcome, we used two RPL/RM datasets: (1)LUCAR (Lviv Ukrainian Cohort for Advancing Reproductive Health) study from the Western Ukraine, including 350 women with clinically confirmed idiopathic RPL and 454 control women with at least one healthy child; (2)UK Biobank (UKBB) dataset, consisting of 556 women with RM and 2,928 control women with at least one (self-reported) healthy-born child. The LUCAR/UKBB genome-wide array datasets were QCed, imputed to TopMED/HRC reference panels density. PLINK1.9 was used for clumping (P-value = 1×10-5, r2 = 0.5, wc = 1000kb) of infection’s GWAS summary statistics. We performed two‐sample Mendelian randomization (MR, TwoSampleMR 0.5.2 package) analysis. As instruments for infections, we used summary statistics from genome‐wide association studies (GWAS; PMID:28928442, and 7th release of COVID19 Host Genetics Initiative).

The MR using 210/20 independent shingles/mononucleosis SNPs as instruments showed protective causal effect of shingles/mononucleosis on risk of RPL (OR[95%CI] = 0.40[0.32-0.50], P-value = 6.81×10-17/ OR[95%CI] = 0.34[0.14-0.80], P-value = 0.014). Then, we performed MR analyses for UKBB, meta-analysed results and detected significant (OR[95%CI] = 0.98[0.96-0.995], P-value = 0.013) protective from RPL/RM causal effect of shingles. Other infections did not show causality (P-value > 0.05) on RPL/RM risk. Our findings suggest that exposure to herpes zoster infections before pregnancy are protective from idiopathic pregnancy loss.

Grant: EMBO SLG 5423-2023

Conflict of Interest: None declared

EP02.035 Circulating anti-Müllerian hormone levels in pre-menopausal women: novel genetic insights from a GWAS meta-analysis

Natàlia Pujol Gualdo 1;2, Minna Karjalainen3;4, Urmo Võsa1, Riikka K. Arffman2, Reedik Mägi1, Justiina Ronkainen3, Triin Laisk1, Terhi Piltonen2

1Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia; 2Research Unit of Clinical Medicine, Department of Obstetrics and Gynecology, Oulu, Finland; 3Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland; 4. Northern Finland Birth Cohorts, Arctic Biobank, Infrastructure for Population Studies, Faculty of Medicine, University of Oulu, Oulu, Finland

Background/Objectives: AMH is expressed by preantral and small antral stage ovarian follicles in women, and variation in circulating AMH levels has been associated with several conditions. However, the biological mechanisms underlying this variation are not fully understood. Previous GWAS have identified three loci associated with AMH levels in women.

Methods: We performed a GWAS meta-analysis (n = 9,668) in which we combined 2,619 AMH measurements from a founder population cohort (NFBC1966) with a previous GWAS meta-analysis that included 7,049 AMH measurements. We annotated the genetic variants, combined different data layers to prioritise potential candidate genes, described significant pathways and tissues enriched by the GWAS signals, identified plausible regulatory roles using colocalization analysis and leveraged publicly available datasets to assess genetic and phenotypic correlations.

Results: Three novel genome-wide significant loci were identified. One of these is in complete linkage disequilibrium with c.1100delC in CHEK2, which is found to be 4-fold enriched in the Finnish population compared to other European populations. We propose a plausible regulatory effect of some of the GWAS variants linked to AMH, as they colocalise with GWAS signals associated with gene expression levels of BMP4, TEX41 and EIFBP41. Gene set analysis highlighted significant enrichment in renal system vasculature morphogenesis and tissue enrichment analysis ranked the pituitary gland as the top association.

Conclusion: Our results highlight the increased power of founder populations and larger sample sizes to boost the discovery of novel trait-associated variants underlying variation in AMH levels.

Grants: EU Horizon 2020 under the MATER Marie Sklodowska-Curie grant agreement No. 813707 and Oulu university scholarship foundation.

Conflict of Interest: None declared

EP02.036 First and second trimester urine microRNA profiles in preeclampsia

Anastasia Maltseva 1, Roman Illarionov1, Elena Vashukova1, Olga Pachuliia1, Andrey Glotov1

1D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproduction, Department of Genomic Medicine, Russian Federation

Background/Objectives: Preeclampsia (PE) is a severe complication of pregnancy, affecting 2-8 % of all pregnancies. PE is characterized by a high incidence of maternal and child morbidity and mortality. We have previously studied the microRNA profile in plasma samples from pregnant women at high risk of preterm birth. In this study, we have performed a prospective study of the miRNA profile in urine, the most non-invasive biomaterial, during the first and second trimesters in pregnant women with PE.

Methods: Total of 48 urine samples were taken from 24 women (PE = 12, CTRL = 12) in the first and second trimesters. Small RNA isolation and library preparation for sequencing was performed using miRNeasy Serum/Plasma Kit (Qiagen) and QIAseq miRNA Library Kit (Qiagen). Libraries were sequenced on a HiSeq 2500 (Illumina). Bioinformatic data analysis was performed using the GeneGlobe Data Analysis Center and DESeq2 R Package.

Results: We detected differences in the levels of 5 miRNAs (4 upregulated – hsa-miR-205-5p, hsa-miR-203a-3p, hsa-miR-223-3p, hsa-miR-184; 1 downregulated – hsa-miR-1-3p) (log2(FC) ≥ 1.5; FDR ≤ 0.05) during the first trimester compared with the control (healthy pregnant women). During the second trimester 5 miRNAs was dysregulated (3 upregulated – hsa-miR-1-3p, hsa-miR-206, hsa-miR-9-5p; 2 downregulated – hsa-miR-205-5p, hsa-miR-223-3p). Hsa-miR-205-5p, hsa-miR-223-3p, and hsa-miR-1-3p are differentially expressed in both trimesters, but in reverse regulation.

Conclusion: Our results suggest that the miRNA profile in urine may predict a PE. Identified miRNAs in urine can be used as PE biomarkers.

Grants: This study was financially supported by a Russian Science Foundation grant 19-75-20033.

Conflict of Interest: None declared

EP02.037 Pre-implantation Genetic Testing for Hereditary Breast and Ovarian Cancer Syndrome: The Parisian Experience

Roxana Borghese 1, Charlotte Sonigo2, Nelly Achour3, sophie monnot1, Nadine Girarel1, michael grynberg3, Alexandra Benachi2, Sophie Frank4, Chrystelle Colas4, Dominique Stoppa-Lyonnet4, Julie Steffann1

1Necker Enfants Malades, Medecine Genomique des Maladies Rares, Paris, France; 2Antoine Béclère, Gynecologie Obstétrique, Paris, France; 3Antoine Béclère, Biologie de la reproduction, Paris, France; 4Institut Curie, Service de génétique, Paris, France

Background/Objectives: In France, Preimplantation Genetic Testing for Monogenic disease(PGT-M) is reserved for couples at high risk of having a child with a severe genetic disease deemed incurable, attested by a multidisciplinary prenatal diagnosis center (CPDPN). Hereditary breast and ovarian cancer syndrome is categorized as a late-onset disease for which access to PGT is disputed, in part because of the low success rate of this procedure.

Methods: Retrospective study of the 46 requests we received since 2015.

Results: A total of 27 requests (55%) were considered eligible, 10 were denied because of CPDPN refusal (4) or ovarian reserve alteration (6), and 9 couples dropped out. A total of 33 ovarian stimulation cycles were started, leading to 196 mature oocytes, 83 biopsied and 38 healthy embryos. Fresh or frozen embryos (n = 21) were transferred (n = 18), resulting in 8 pregnancies and 5 live births (23% of couples who started a PGT cycle). So far, all couples have requested to transfer embryos devoid of the pathogenic variant, regardless of gender, but none of the 42 couples who could not have a birth through PGT, requested a prenatal diagnosis.

Conclusion: PGT provides an alternative for couples who find it more “acceptable” to avoid transferring an embryo rather than opting for a pregnancy termination in the event of a predisposition to early-onset cancers. To increase the PGT pregnancy rate, the transfer of male embryos carrying the variant, assumed to have low tumor risks, should be considered.

Grants: No grants

Conflict of Interest: None declared

EP02.038 Complex chromosomal rearrangements in first trimester products of conception

Christina Katsidi1;2, Haralambia Tsarouha2, elisavet kouvidi2, leandros lazaros2, Periklis Makrythanasis 3;4;5, amelia pantou2, emmanouil kanavakis2, ariadni mavrou2

1National and Kapodistrian University of Athens, Medical School, Athens, Greece; 2Genesis Genoma Lab, Genetic Diagnosis, Clinical Genetics & Research, Athens, Greece; 3Biomedical Research Foundation of the Academy of Athens, Athens, Greece; 4National and Kapodistrian University of Athens, Laboratory of Medical Genetics, Athens, Greece; 5University of Geneva, Medical School, Genetic Medicine and Development, Geneva, Switzerland

Background/Objectives: Multiple or complex chromosomal abnormalities in products of conception are rare and are correlated with advanced maternal age and an early week of gestation. This study investigates their frequency in the first trimester of pregnancy.

Methods: 554 samples from first trimester aborted tissues were analyzed with conventional cytogenetic or molecular techniques. The average maternal age was 39 years (range 23-54). From each sample at least ten metaphases were analyzed using standard GTG banding. In cases where tissue samples failed to grow, molecular techniques, such as Quantitative Fluorescent Polymerase Chain Reaction (QF-PCR), were applied. Statistical analysis was performed using Pearson Chi-square exact tests to assess the association of the week of gestation and the age of the mother with single or complex chromosomal abnormalities.

Results: The success rate was 70,76%. Abnormal karyotypes were detected in 255 embryos (65%) and a normal karyotype was identified in 137 (35%). Single chromosomal abnormalities were identified in 235 cases (69,4%) with single trisomy 16 being the most frequent. Complex abnormalities were identified in 25 cases (9,8%), the most common finding was double trisomy. Polyploidies and monosomy 45,X occurred in 7,8%. Structural rearrangements and mosaicism were detected in 2,8% and 2,4%. Complex chromosomal abnormalities were more frequent among women older than 40 years old.

Conclusion: Embryos with complex chromosomal abnormalities were more frequent among women with advanced maternal age and were aborted one week earlier than embryos with single chromosomal abnormalities. Preimplantation genetic testing may be recommended in order to deliver a healthy offspring.

Grants: There were no grants

Conflict of Interest: None declared

EP02.039 Study of heterozygous carriage of genetic pathology among infertile couples in the Sverdlovsk region

Svetlana Deryabina 1, Olga Lagutina1, Elena Kudryavceva1

1Institute of medical cell technologies, Laboratory of molecular genetic researches, Yekaterinburg, Russian Federation

Background/Aims: The number of infertile patients planning to use assisted reproductive technologies (ART) is increasing significantly worldwide. However, the use of ART cannot automatically guarantee the birth of healthy children. The aim of this study was to determine the number of heterozygous carriers of severe hereditary pathology among infertile patients before in vitro fertilisation.

METHODS: In our study, we tested the first 30 married couples for carriage of fifty monogenic diseases by next generation sequencing (NGS). Informed voluntary consent was obtained from all patients.

Results: Our panel identified 28 carriers, that is, in almost every family one of the parents (and sometimes both) had an altered DNA sequence. The highest frequency of altered genetic variants was observed for congenital hearing loss (6 heterozygous carriers for GJB2 and SLC26A4 genes), cystic fibrosis and phenylketonuria (4 heterozygous carriers each). Also 3 future parents have mutations in the gene associated with biotinidase deficiency, 2 people are carriers of Wilson’s and Willebrand’s diseases. In addition, pathogenic variants in genes associated with such rare diseases for the Sverdlovsk region as isovalerian acidemia, recessive polycystic disease, metachromatic leukodystrophy, mitochondrial DNA depletion syndrome, and alpha-1-antitrypsin deficiency were identified. So far, no concordant heterozygotes have been identified in any of the pairs.

Conclusions: The findings indicate a high frequency of carrier genetic pathology among infertile couples, which makes further carrier screening appropriate.

Conflict of Interest: None declared

EP02.040 Implementation of Expanded Carrier Screening for Genetic Diseases: Report from Iran

Fatemeh Ahangari 1, shima Dehdahsi1, Mahsa Fadaie1, Mehri Ashki1, Maryam Beheshtian1, Maryam Azad1, Masoumeh Akbari Kelishomi1, Negar Molaei2, Parnian Alagha2, Ariana Kariminejad1, Hossein Najmabadi1

1Kariminejad-Najmabadi Medical Center, Tehran, Iran; 2Faculty of Social Welfare and Rehabilitation Sciences - National University, Tajrish, Iran

Background: Reproductive carrier screening (RCS) is designed to inform couples about their chances of having children with particular monogenic recessive conditions. Screening options have been revolutionized by NGS-based technologies and expanded carrier screening (ECS) is increasingly used in different populations. Here, we describe the results of the ECS test developed in Kariminejad-Najmabadi Pathology &Genetics Center, Tehran, Iran.

Methods: DNA samples of 225 Iranian cases (including 52 couples) were sequenced by NGS for 433 genes which designed for in house carrier panel considering ACMG recommended genes and genes with high carrier frequency in Iranian population. Also, deletions/duplications in DMD, SMN1 and repeats for FMR1 genes have been investigated by MLPA and TP-PCR techniques, respectively. Various routine filters applied to variants, the remaining, classified according to ACMG guideline criteria. Only variants that met the ACMG criteria for pathogenic/ likely pathogenic were reported.

Results: Of the 225 individuals, 83.11% were found to be carrier of at least one pathogenic/likely pathogenic variant. Twenty five out of the 52 couples (~48%) were carrier in same genes. The most frequent occurrence of carriers was recorded in the commonly investigated genes (CFTR, PAH, SMN1), consistent with the high carrier rates reported in the Iranian population.

Conclusion: NGS-based carrier screening offers a powerful approach for assessing carrier status to decrease the burden of genetic diseases, especially in populations with a high rate of consanguinity. The strategies for variant interpretation in a healthy population are challenging, therefore, the importance of access to studies from different populations is useful for a better interpretation of the results.

Conflict of Interest: None declared

EP02.041 Preimplantation diagnosis for mitochondrial DNA m.8344A > G (MERRF) mutation : challenge and success

sophie monnot 1, Nadine Gigarel1, anne mayeur2, Joana Bengoa1, Roxana Borghese1, Benoit Funalot1, Charlotte Sonigo2, nelly achour2, Julie Steffann1

1Necker Hospital, paris, France; 2Antoine Beclere Hospital, Clamart, France

Background/Objectives: Mitochondrial DNA (mtDNA) mutations cause a wide range of serious genetic diseases with high transmission risk, due to their maternal inheritance. Genetic heterogeneity of these diseases is due to the co existence of wild-type and mutated mtDNA (heteroplasmy). There is a threshold below which clinical symptoms are not likely to occur. At risk couples often ask for preimplantation genetic diagnosis (PGD) which is based on mutant load assessment in blastomeres.

Methods: Three women, asymptomatic carriers of m.8344A > G in the MT-TK gene, responsible for MERRF syndrome (Myoclonic Epilepsy with Ragged-Red Fibers), underwent PGD in our centre. Heteroplasmy level was measured on 75 cells from 40 embryos at day 3 of developpement (D3).

Results: The mutant load was stable among blastomeres from a given embryo at D3, with the exception of one embryo. We observed one mutation-free embryo. The remaining ones were all heteroplasmic (10 to 97%), suggesting random segregation of mutated DNA during maternal oogenesis. Five of them were transferred resulting in 2 pregnancies and the birth of two healthy children. Mutant load was measured at birth on cord blood showing a stable level in one case, and an increase of 18% in the other.

Conclusion: These results 1/ validate technical feasibility of PGD for the MERRF mutation on D3 embryos 2/ demonstrate clinical interest of PGD for women carrying high level of mtDNA mutations 3/ illustrate the possible variation of the mutant load during human embryo-foetal development. These data have an important impact in terms of genetic counselling for patients carrying mtDNA mutation.

Grants:

Conflict of Interest: None declared

EP02.042 Analysis of aneuploidies in products of conception: effect of maternal and gestational age

Eloisa Caviglia 1, Micaela Galain1, Yannina Diaz Cano1, Mónica Fabbro1, Sergio Papier1, Florencia Nodar1, Cecilia Fernandez1

1Novagen Genetics Laboratory. Cegyr-Eugin Group, Buenos Aires, Argentina

Background/Objectives: Between 15-20% of pregnancies result in miscarriages, of which 50% are due to embryonic chromosomal abnormalities. The aim was to describe the results of products of conception (POC) analysis and to assess the impact of maternal age and gestational age on the presence of aneuploidies.

Methods: 93 POC were studied by NGS. STRs in fetal and maternal DNA were examined to rule out contamination with maternal cells and XXX triploidies. Results were stratified according to maternal age into ≤37 (57) and >37 years (36).

Results: Results were obtained in 87% of the samples. Chromosomal abnormalities were identified in 63.4%. The most frequent aneuploidies were trisomies in chromosomes 16, 15, 21, 22, and 13. In ≤37 chromosomal abnormalities were detected in 56.1% of cases, while in >37 in 75%. Abnormalities occurred in a greater variety of chromosomes in ≤37, with trisomies predominating in chromosomes 14 and 22. Additionally, X monosomies and triploidies were identified. In >37, trisomies were more frequent in chromosomes 16, 15, and 21. The gestational age varied from 5 to 22 weeks in ≤37 and from 5 to 13 in >37. However, regardless of maternal age, 80% of abortions occurred between weeks 6-9.

Conclusion: Advanced maternal age increases the risk of aneuploidies. These abnormalities would explain the earlier pregnancy losses in women >37 years. NGS proved to be a useful tool for the study of POC. Knowing the cause of abortion allows estimating the risk of recurrence, determining the need for additional studies, and planning new reproductive strategies.

Grants:

Conflict of Interest: Eloisa Caviglia CEGYR, Micaela Galain CEGYR, Yannina Diaz Cano CEGYR, Mónica Fabbro CEGYR, Sergio Papier CEGYR, CEGYR, Florencia Nodar CEGYR, CEGYR, Cecilia Fernandez CEGYR

EP02.043 Multicenter study assessing the reproductive outcomes of mosaic embryo transfer in Argentina

Micaela Galain 1, Eloisa Caviglia1, Yannina Diaz Cano1, Mónica Fabbro1, Sebastián Menazzi1, Valeria Paz2, Idelma Serpa2, Laura Sicaro3, Edgardo Young Obejero3, Andrea Dematteis4, Gustavo Estofan4, Sergio Papier1, Florencia Nodar1, Cecilia Fernandez1

1Novagen Genetics Laboratory. Cegyr-Eugin Group, Buenos Aires, Argentina; 2Grupo Gamma, Servicio de Medicina Reproductiva, Rosario, Argentina; 3IFER, Instituto de Ginecología y Fertilidad, Buenos Aires, Argentina; 4CIGOR, Clínica de Fertilidad, Cordoba, Argentina

Background/Objectives: Chromosomal mosaicism is defined as the presence of two or more different cell lines within an embryo. Several publications report that mosaic embryo (ME) transfer can lead to successful pregnancies and healthy deliveries. However, the impact of mosaicism on IVF outcomes remains a matter of controversy. The aim was to assess if low-level ME transfer affects reproductive outcomes.

Methods: The study included 6241 embryos from 4 fertility clinics who underwent PGT. Patients in group A (n = 894) were transferred an euploid embryo, while in group B (n = 55), a low-grade ME (<40%, 2016-2021 or <50%, 2021-2023) aneuploid cells, was transferred.

Results: 46.2% embryos were euploid, 31.3% aneuploid and 19.3% mosaic (69.5% classified as low-grade and 30.5% as high-grade) and in 3.2% no results were obtained. Fifty five low-grade ME were transferred. The positive beta-hCG rates were 50.3% and 49.1%, while the clinical pregnancy rates were 36.9% and 36.4% for Groups A and B, respectively. Regarding live births, in Group A 328 (36.7%) children were born, and in Group B,16 (29.1%) with 2 ongoing pregnancies. Conversely, pregnancy losses in Group B (9.1%) were higher compared to the euploid group (2.9%). In all cases the newborns were apparently healthy after ME transfer, with an average gestational week at birth of 39 (37-41), and an average weight of 3197 (2900-4390) grams.

Conclusion: Low-grade ME transfer would not affect reproductive outcomes, as live birth rate appears to be like those observed with euploid embryos. Therefore, mosaic embryos are a valid alternative for couples lacking euploid embryos.

Grants:

Conflict of Interest: Micaela Galain Cegyr-Eugin Group, Eloisa Caviglia Cegyr-Eugin Group, Yannina Diaz Cano Cegyr-Eugin Group, Mónica Fabbro Cegyr-Eugin Group, Sebastián Menazzi Cegyr-Eugin Group, Valeria Paz GAMMA, Idelma Serpa GAMMA, Laura Sicaro IFER, Edgardo Young Obejero IFER, IFER, Andrea Dematteis CIGOR, Gustavo Estofan CIGOR, CIGOR, Sergio Papier Cegyr, Cegyr, Florencia Nodar Cegyr, Cegyr, Cecilia Fernandez Cegyr

EP02.044 PGT-SR: effect of chromosomal rearrangement type and carrier gender on the acquisition of transferable embryos.

Yannina Diaz Cano 1, Micaela Galain1, Eloisa Caviglia1, Mónica Fabbro1, Florencia Nodar1, Sergio Papier1, Cecilia Fernandez1

1Cegyr-Eugin Group, Novagen Genetics Laboratory, Buenos Aires, Argentina

Background/Objectives: Translocation carriers have an increased risk of having unbalanced embryos. PGT-SR is a crucial tool for selecting embryos to transfer. This study aimed to explore how chromosomal rearrangement type and carrier gender affect the ability to obtain transferable embryos.

Methods: 265 embryos from 71 cycles (47-couples) were divided according to the parental rearrangement into Robertsonian translocation carrier (ROB-T, 105) and Reciprocal translocation carrier (REC-T, 160) groups. Data were stratified by maternal age (≤37 and >37) and carrier gender: male carrier (MC) and female carrier (FC). Embryos were classified as transferable if euploid/balanced or showed low-grade mosaicism (<50%). Non-transferable embryos included unbalanced, aneuploid, or displayed high-grade mosaicism.

Results: Among ROB-T embryos, 43% (45/105) were transferable, compared to only 22% (35/160) in the REC-T group (p = 0.0004612). Subgroup analysis by maternal age revealed that in the ≤37, 43% (39/91) of ROB-T embryos were transferable versus 25% (30/122) in the REC-T group (p = 0.007583), and in the >37, 43% (6/14) of ROB-T embryos were transferable compared to 13% (5/38) in the REC-T group (p = 0.04951). Within the MC, 47% (36/76) of ROB-T embryos were transferable versus 24% (22/90) in the REC-T (p = 0.003467). Conversely, in the FC, 31% (9/29) of ROB-T embryos and 19% (13/70) of REC-T embryos were transferable (p = 0.2749). Unbalanced embryos, comprising 71% of non-transferable embryos, were more prevalent in REC-T carriers across all analyzed groups.

Conclusion: The type of rearrangement is relevant to obtaining transferable embryos. Carriers of REC-T, especially female carriers, could have a higher risk of having non-transferable embryos compared to carriers of ROB-T.

Conflict of Interest: Yannina Diaz Cano Full-time employee at Novagen, Micaela Galain Full-time employee at Novagen, Eloisa Caviglia Full-time employee at Novagen, Mónica Fabbro Full-time employee at Novagen, Florencia Nodar Shareholder of Cegyr, Sergio Papier Shareholder of Cegyr, Cecilia Fernandez Full-time employee at Novagen

EP03 Prenatal Genetics

EP03.001 A decade of insights: prenatal cytogenetic diagnosis results of high-risk pregnancies at Atatürk University Medical Genetics laboratory (ATAGEN)

Momen Kanjee 1, Nuran Ertargin1, Cigdem Yuce Kahraman1, Abdulgani Tatar1

1Atatürk University, Faculty of Medicine, Department of Medical Genetics, Erzurum, Türkyie

Background/Objectives: Prenatal diagnosis is the process of performing genetic testing on fetal samples to determine whether the fetus is affected by a genetic disorder. Conventional karyotype analysis is helpful in detecting large chromosomal aberrations (CA). Here, we summarize the indications and karyotype results of patients who underwent amniocentesis or cordocentesis in our clinic.

Methods: The karyotype findings of patients who underwent amniocentesis/cordocentesis in our clinic between 2014 and 2023 were evaluated retrospectively. The results were reported according to the International System for Human Cytogenomic Nomenclature (ISCN).

Results: In this study, the results from 1,330 samples were analyzed: 137(10.3%) cordocentesis and 1,193(89.7%) amniocentesis samples. Our culture success rate was 89%(1,184/1,330). The median maternal and gestational ages were 33 years(18-58) and 18 weeks(11-39), respectively. The most common indication was a positive screening result (43.1%;573/1,330), followed by fetal structural abnormalities (30.6%;407/1,330) and advanced maternal age (23.5%;313/1,330).

The prevalence of CA was 7.7%(103/1330), with aneuploidies being the most common aberrations (83.5%;86/103) followed by structural abnormalities (9.7%;10/103) and mosaicisms (6.8%;7/103). Overall, trisomy 21 was the most frequently detected alteration (n = 67; 61 standard trisomies, 5 mosaic, 1 standard trisomy with structural aberration) followed by trisomy 18 (n = 21; 20 standard trisomies, 1 standard trisomy with structural aberration), trisomy 13 (n = 3; all standard trisomies), sex chromosome aneuploidies (n = 4; 2 mosaic monosomy Xs, 1 47,XXY, 1 47,XYY), and other structural aberrations(n = 8).

Conclusion: Our study illustrated that invasive prenatal cytogenetic analysis remains a diagnostic method for detecting CA. Although additional diagnostic tests were offered to some patients, this study shows only the karyotype results.

Grants: None.

Conflict of Interest: None declared

EP03.002 A Rare Case Report: Trisomy 22 in the Second Trimester

Agnese Kalnina 1;2, Inguna Lubaua1;2, Ieva Grīnfelde1;2, Klinta Lisnere1, Maija Lubgane-Griķīte1, Elena Zeltina1, Gunta Kalnberza1

1Children’s Clinical University Hospital, Rīga, Latvia; 2Riga Stradins University, Rīga, Latvia

Background/Objectives: Trisomy 22, a chromosomal abnormality, often results in severe medical complications, typically leading to first-trimester spontaneous abortion. Live births and continued pregnancies beyond 12 weeks of gestation are exceedingly rare. We present a unique second-trimester case of trisomy 22 at our hospital.

Methods: The patient was examined at the Children’s Clinical University Hospital of Latvia’s Medical Genetics and Prenatal Diagnostics Department.

Results: At 13 weeks of gestation, the patient sought consultation at our medical genetics clinic following abnormal findings from first-trimester ultrasonography. Noninvasive prenatal testing (NIPT) indicated the possibility of trisomy 22. The fetus displayed increased nuchal translucency thickness, renal hyperechogenicity, and a complex congenital heart defect known as tetralogy of Fallot. Ultimately, the family made the decision to terminate the pregnancy at 15 weeks. Patho-anatomical examination revealed distinctive phenotypical features, such as excess nuchal skin, low-set ears, and micrognathia. The internal organs developed properly, except the prenatally diagnosed tetralogy of Fallot. Karyotype analysis from chorionic biopsy subsequently confirmed non-mosaic trisomy 22: 47,XY, + 22.

Conclusion: This case highlights the atypical presentation of trisomy 22 in later gestation, emphasizing the importance of early prenatal screening. While tetralogy of Fallot, potentially correctable postnatally, is commonly associated with genetic syndromes, its previous correlation with trisomy 22 has not been described. Early identification enables families to prepare for a child with a genetic syndrome or an elevated risk of fetal demise. Full genome NIPT played a crucial role in diagnosing this rare genetic aberration, highlighting its ability to detect conditions that might otherwise go unnoticed.

Conflict of Interest: None declared

EP03.003 Evaluation of the contribution of trio exome sequencing in targeted prenatal indications.

Manon Chretien1, Julien Osouf2, Carine Abel3, Alexandra Afenjar4, Tania Attie-Bitach5, elise brischoux-boucher6, lydie BURGLEN7, Nadège Calmels2, Nicolas Chassaing8, Thomas Courtin9, Julian Delanne10, Martine Doco-Fenzy11;12, Christele Dubourg13, Benjamin Durand1, Salima EL CHEHADEH1, Laurence Faivre10;14, Aurore Garde10, Emmanuelle Ginglinger15, Virginie Haushalter2, Damien Haye3, Solveig Heide9, Laurence Heidet16;17, Delphine Heron9, Clemence Jacquin12, Laetitia Lambert18;19, Vincent Laugel20, Daphné Lehalle9, Laurence Michel-Calemard21, Yohny Montoya22, Jean Muller2;23;24, Sylvie Odent25, Olivier Patat8, Juliette Piard14;26, Céline Poirsier12, Audrey Putoux3, Chloe Quelin25, Nicolas Sananes27;28, Audrey Schalk2, sophie scheidecker2;24, Christel Thauvin-Robinet10;14, Stéphanie Valence29, Anne-Sophie Weingertner27;28, Justine Wourms18, Hélène Dollfus1;24, Benedicte Gerard2, Caroline Schluth-Bolard2;24, Elise Schaefer 1

1Service de Génétique Médicale, Institut de Génétique Médicale - Hôpitaux Universitaires de Strasbourg, Strasbourg, France; 2Laboratoire de Diagnostic Génétique, Nouvel Hopital Civil - Hôpitaux Universitaires de Strasbourg, Strasbourg, France; 3Service de Génétique, Groupement Hospitalier Est, Hospices Civils de Lyon, Lyon, France; 4Centre de Référence Malformations et Maladies Congénitales du Cervelet, UF de génétique clinique, Hôpital Trousseau, APHP, Sorbonne Université, Paris; 5Service de Médecine Génomique des Maladies Rares, Hôpital Necker-Enfants Malades, Paris, France; 6Centre de Génétique Humaine, Centre Hospitalier Universitaire, Université de Bourgogne Franche-Comté, Besançon; 7Laboratoire de Neurogénétique Moléculaire, Hôpital d’Enfants Armand-Trousseau, APHP, Paris, France; 8Service de Génétique Médicale, Hôpital de Purpan, CHU de Toulouse, Toulouse, France; 9Service de Génétique Clinique et Médicale, Hôpital de la Pitié Salpêtrière, APHP, Paris, France; 10Service de Génétique Médicale, Hôpital du Bocage, CHU de Dijon, Dijon, France; 11Service de Génétique, Institut de Biologie, CHU Nantes, Nantes, France; 12Service de Génétique, Hôpital Maison Blanche, CHU de Reims, Reims, France; 13Laboratoire de Génétique Moléculaire et Génomique Médicale, Hôpital Pontchaillou, CHU de Rennes, Rennes, France; 14UMR 1231 GAD, Inserm, Université de Bourgogne Franche-Comté, Dijon, France; 15Service de Génétique Médicale, Hôpital Emile Muller, GHRMSA, Mulhouse, France; 16Service de Néphrologie Pédiatrique, Centre de Référence des Maladies Rénales Héréditaires de l’Enfant et de l’Adulte (MARHEA), Hôpital Necker-Enfants Malades, APHP-centre, Université Paris Cité, Paris, France; 17Laboratoire des Maladies Rénales Héréditaires, Inserm UMR 1163, Institut Imagine, Paris; 18Service de Génétique clinique et Médecine Infantile, Hôpital d’Enfants CHU de Nancy - Hôpitaux de Brabois, VANDOEUVRE-LÈS-NANCY, France; 19INSERM-U1256 NGERE, Université de Lorraine, Vandœuvre-lès-Nancy, France; 20Service de Pédiatrie 1, Hôpital de Hautepierre, CHU de Strasbourg, Strasbourg, France; 21Service de Biochimie et Biologie Moléculaire, Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Lyon, France; 22Service d’Obstétrique, Hôpital du Hasenrein, GHRMSA, Mulhouse, France; 23Unité de Bio-informatique Médicale Appliquée au Diagnostic, Hôpitaux Universitaires de Strasbourg, Strasbourg, France; 24Laboratoire de Génétique Médicale, INSERM UMRS_1112, Institut de Génétique Médicale, CRBS, Université de Strasbourg, Strasbourg, France; 25Service de Génétique Clinique, Hôpital Sud, CHU de Rennes, Rennes, France; 26Service de Génétique Médicale, Hôpital Jean Minjoz, CHU de Besançon, Besançon, France; 27Service de Gynécologie obstétrique, CMCO, CHU de Strasbourg, Schiltigheim, France; 28Centre Pluridisciplinaire de Diagnostic PréNatal, Département de Médecine Fœtale, CMCO, CHU de Strasbourg, Schiltigheim, France; 29Service de Neuropédiatrie, Hôpital d’Enfants Armand-Trousseau, APHP, Paris

Background/Objectives: Congenital malformations affect approximately 3% of pregnancies. The etiologies are diverse, and, in most cases, constitutional genetic abnormalities are sought.

Methods: In a multicenter, comparative, prospective study, we evaluated the yield of high-throughput sequencing analyses (in silico versus exome panel) in targeted prenatal indications strongly suggesting a monogenic pathology.

Results: Trio exome analysis of 86 fetuses (mean duration of 21 weeks of amenorrhea) led to a certain etiological diagnosis in 28% of cases (CGPA classes 4/5) and uncertain in 11.6% (VUS, class 3). The yield varied according to the type of malformation: 41.6% for vermian hypoplasia, 31% for polymalformative syndromes, 25% for hygroma colli / generalized hydrops, 23.5% for hyperechoic large kidneys, 20% for agenesis of the corpus callosum, 16.7% for holoprosencephaly, 1/2 for gyration anomalies and 1/4 for ophthalmological anomalies. The identified variants are mainly missense, in ciliopathy genes (19%) or DNA metabolism genes (16%). The mode of transmission is predominantly autosomal dominant, with 47% of variants arising de novo and 16% inherited from a pauci or asymptomatic parent. We detected a postzygotic mosaic (13%) and 4 double diagnoses. The primary in silico panel analysis provided 19% of the unambiguous molecular diagnoses; the secondary analysis of the whole exome allowed the diagnosis of a further 9% of cases, but with identification of numerous VUS.

Conclusion: This study demonstrates the added value of exome sequencing in comparison with the use of in silico panels but in targeted indications, demonstrating the value of a clear definition of the malformations justifying this approach.

Grants: No

Conflict of Interest: None declared

EP03.004 Tissue-Specific Aneuploidy in Mosaic Trisomy 18: Prenatal and Postnatal Insights

Rainer Wimmer 1, Miriam Kinzel2, Iris Dressler-Steinbach3, Holger Janke4, Andre Weber1, Markus Stumm1

1Medicover Humangenetik Berlin Lichtenberg MVZ, Cytogenetics, Berlin, Germany; 2Medicover Humangenetik Berlin Lichtenberg MVZ, Genetic Counseling, Berlin, Germany; 3Charité Universitätsmedizin Berlin, Department of Obstetrics, Berlin, Germany; 4Praxis für Pränatale Diagnostik, Berlin, Germany

Background/Objectives: Edwards’ syndrome, resulting from trisomy 18, is commonly considered as incompatible with life, leading to pregnancy terminations following prenatal detection. However, mosaic trisomy 18 exhibits significant clinical heterogeneity. We report on a male pregnancy at high risk for trisomy 18, double non-invasive prenatal testing was conducted. Initial results indicated a false normal female, followed by a male result positive for trisomy 18. Fetal ultrasound revealed ventricular septal defect, thymic hypoplasia, overthrown fingers and a megacisterna magna. Subsequent amniocentesis confirmed mosaic trisomy 18. Despite the diagnosis, the parents chose to continue the pregnancy, opting for comprehensive medical care. The boy was delivered by Cesarean section and underwent heart surgery at 4 months. At 8 months, his development was evaluated as age-appropriate. This case study presents unprecedented pre- and postnatal insights into the tissue-specific distribution of his aneuploid cells.

Methods: Chromosomal and/or FISH analyses were conducted on various tissues from all germ layers.

Results: The distribution of trisomy 18 cells varied significantly: amniotic fluid (28%), heart (96%), thymus (7%), skin (5%), and buccal smear (1%). Notably, the congenital heart malformation correlated with a high percentage of aneuploid cells in the right ventricle.

Conclusion: In cases of mosaic trisomy 18, the presence of a normal cell line with unknown tissue distribution complicates predicting pregnancy outcomes. Theoretical prognostic considerations should primarily focus on confirmed fetal malformations. Clinicians should be aware of the potential for varied outcomes and consider a multidisciplinary approach when counseling parents facing similar situations.

Grants:-

Conflict of Interest: None declared

EP03.005 Expanding the allelic spectrum of the PACS1 gene: a prenatal case report with brain malformation

Marta Palucci 1, Antonella Pizza2, Allessandra Terracciano3, Valeria Orlando3, antonella spagnuolo4, maria assunta spina4, Lucia Manganaro5, maria cristina muzi6, manuela di natale6, barbara raso6, carlotta vaccari6, sara nuovo6

1Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Department of Science of Life and Public Health, Rome, Italy; 2University of Campania “Luigi Vanvitelli”, Department of Precision Medicine, Naples, Italy; 3Bambino Gesù Children’s Hospital, IRCCS, Rare Diseases and Medical Genetics Unit, Rome, Italy; 4Centro Sant’Anna ASL Roma1, UOSD Diagnosi Prenatale, Rome, Italy; 5Sapienza University of Rome, Department of Radiological, Oncological and Pathological Sciences, Rome, Italy; 6Centro Sant’Anna ASL Roma1, UOSD Genetica Medica, Rome, Italy

Background/Objectives: Schuurs-Hoeijmakers syndrome (SHMS, OMIM #615009) is a rare autosomal dominant disorder characterized by mild-to-severe psychomotor delay/intellectual disability, abnormal craniofacial features and congenital malformations. SHMS is caused by heterozygous mutations in the PACS1 gene. Less than 90 cases have been reported worldwide (usually diagnosed postnatally), almost all sharing the p.Arg203Trp missense change.

Methods: We report a novel PACS1 variant, detected in a prenatal setting. A 32-year-old woman was referred to our prenatal diagnosis center at 20 weeks of gestation (WG) because of suspected corpus callosum (CC) hypoplasia and abnormal width of cavum septum pellucidi (CSP) appearing on ultrasound. Fetal brain MRI (at 21 WG) revealed a short, thickened and dysmorphic CC, associated with enlarged CSP and “teardrop” appearance of the frontal horns.

Results: Standard karyotype and CGH-array analysis performed on fetal DNA following amniocentesis did not reveal pathogenic alterations. Trio-based Clinical Exome Sequencing identified de novo heterozygous PACS1 variant c.2126G>A, causing the aminoacidic change p.(Arg709Gln). This variant is not described in scientific literature nor in international databases and is classified as “probably pathogenetic” (Class-IV) according to American College of Medical Genetics and Genomics (ACMG) guidelines.

Conclusion: We focused on the brain malformations observed in PACS1-mutated cases and recorded MRI findings of patients reported in the literature. Comprehensive evaluation was available only for 36 of them, mainly presenting with cerebellar hypoplasia (27%), ventricular abnormalities (19%), white matter defects (14%) and CC/CSP abnormal appearance (11%).

This work expands the allelic spectrum of PACS1, whose mutation should be considered also in cases with apparently isolated CC/CSP abnormalities.

Conflict of Interest: None declared

EP03.006 Comparisons of results from non-invasive prenatal testing (NIPT) and our cytogenetic analyses

Ilona Dietze-Armana 1, Andreas Gerhardinger1, Manuel Lüdeke1, Karl Mehnert1

1genetikum, Neu-Ulm

Background/Objectives: Non-invasive prenatal testing (NIPT) is a well-established option for prenatal screening, primarily for the detection of trisomy 21, 18, 13 using Cell-free DNA (cfDNA) in maternal plasma. cfDNA is derived from the outer trophoblast cell layer of the placenta. NIPT tests vary in their exact methodology and there are various assays available. In a pooled meta-analysis, the detection rate across different NIPT methods was 99% (T21), 96% (T18) and 91% (T13). The cumulative false positive rate (FPR) was less than 0.1% for T21 and 0.26% for T13/18 (Gil et al. 2015). In addition to false positive detection rates, the positive and negative predictive values (PPV and NPV) of a screening test are important clinical parameters. These values depend partly on the performance characteristics of the test, but also vary with the prevalence of the tested condition in the population.

Methods: Here we report a cohort of 535 cases from 2021 to 2023 with abnormal NIPT for trisomy 13/18/21 for which we performed verification using QF-PCR and fetal karyotype.

Results: NIPT results for 418 of 535 cases could be confirmed with QF-PCR and cytogenetic analysis, whereas in 117 cases (45xT13, 43xT18, 29xT21) no chromosomal abnormality was detected.

Conclusion: The discrepancy between NIPT and cytogenetic methods is due to the fact that the cell-free fetal DNA does not always correspond to the karyotype of the fetus, which can partly be attributed to placenta mosaics.

NIPT has limitations and complexities that requesting clinicians and their patients should consider.

Conflict of Interest: None declared

EP03.007 Prenatal diagnosis and pregnancy termination in Muslims living in mixed cities versus Muslims living in isolated habitats in Israel.

Aliza Amiel 1, Mahdi Tarabeih2

1Tel Aviv-Yafo, School of Nursing Research, Tel Aviv-Yafo, Israel; 2School of Nursing Research The Academic College Tel Aviv-Jaffa, Tel Aviv-Jaffa, Israel

Objective: Pregnancy termination is forbidden according to the laws of Islam, thus, the need for prenatal tests is inconclusive in Muslim society. In this research, we explored whether Jewish society norms influence Muslim women living in mixed cities to undergo prenatal testing and pregnancy terminations compared to Muslims women living in their own habitats.

Methods: The sample consisted of 1081 Israeli Muslim women between 18-49 years of age. 52% living in mixed cities (i.e., Muslim and Jews living in the same city in coexistence), and 48% Muslim responders living in their own habitats.

Results: Muslim women living in mixed cities performed more prenatal tests and pregnancy terminations than in isolated Muslim-only habitats. Most pregnancy terminations, with abnormal results, were performed in the center of the country. Participants living in the center, both mixed-cities and Muslim-only habitats performed more noninvasive prenatal testing (NIPT), compared to participants living in other parts of the country. Only participants living in mixed cities and Muslim-only habitats in the center of the country did receive genetic consultancy before undergoing the different prenatal tests.

Conclusions: Muslim women living in mixed cities performed more prenatal tests and pregnancy terminations than in isolated Muslim-only habitats, due to the influence of the Jewish society living in the same city and to more genetic consultancy. Additional medical health care and genetic counselling are needed in Muslim only habitant cities in other parts than the center of the country.

Conflict of Interest: Aliza Amiel full time, Mahdi Tarabeih full time

EP03.008 A novel α1-globin gene mutation reported from north of Iran: Hb Mazandaran (HbA1 c.154 GGC > TGC)

Mohammad Reza Mahdavi1, Atefeh Khoshaein 2, Maryam Rahimi2, Hossein Jalali1, Mahan Mahdavi2

1Thalassemia Research Center, Hemoglobinopathies Institute, Mazandaran University of Medical Sciences, Sari, Iran; 2Sinaye Mehr Research Center, Sari,Iran

Background/Objectives: Alpha thalassemia is one of the most common human genetic abnormalities and identification of all variants of Hemoglobin in different regions helps to get comprehensive knowledge about thalassemia disease and it can be used in preventive programs, and prenatal diagnosis (PND). In the present study, we describe a new α1 gene mutation (HbA1 c.154 GGC > TGC).

Methods: As a program for prevention of thalassemia a couple was referred to Fajr medical genetics laboratory (Sari, Iran) for routine hematological analysis. At first routine Cell Blood Count (CBC) and Hb capillary electrophoresis were applied. After taking written informed consent, molecular analysis was conducted on genomic DNA extracted from peripheral blood using QIAamp DNA Mini Kit (Qiagen, Germany). For identifying common Mediterranean α-Globin gene deletion multiplex Gap-PCR was performed and for detecting other mutations in the α and β-Globin genes DNA-Sequencing method was used.

Results: The results of CBC and capillary electrophoresis tests were compatible with β-thalassemia carrier in female subject. The case did not have common α-Globin gene deletions. Sequencing of α-Globin gene showed that subject carried c.315 + 1G > A and c.154G > T mutations of the β and α1-globin genes, respectively. Both of the mutations were also detected in the subject’s mother.

Conclusion: The c.154G > T variant (Hb Mazandaran) gives rise to a novel hemoglobin variant that was detected in two cases in combination with β-globin mutation, and it does not seem to be associated with severe hematological abnormalities in the carriers.

Grants: Not-Applicable

Conflict of Interest: None declared

EP03.010 Cell free fetal DNA at 11-13 weeks of gestation is not altered in complicated pregnancies

Zoi Koukou1, Eleftherios Panteris2, Emmanouel Manolakos3, Aristeidis Papadopoulos4, Ioannis Papoulidis3, Eleftheria Papadopoulou5, Konstantinos Relakis6, Stavros Sifakis 7

1School of Health Sciences, International Hellenic University (IHU), Sindos Thessaloniki, Greece; 2Aristotle University of Thessaloniki, Laboratory of Forensic Medicine and Toxicology, Thessaloniki, Greece; 3, Access to Genome P.C, Clinical Laboratory Genetics, Thessaloniki, Greece; 4Aristotle University of Thessaloniki, School of Medicine, Thessaloniki, Greece; 5University Hospital of Heraklion, Department of Pediatrics, Heraklion, Crete, Greece; 6Department of Obstetrics & Gynecology, University Hospital of Heraklion, Heraklion, Crete, Greece; 7, Mitera Maternity Hospital, Heraklion, Crete, Greece

Background/Objectives: Non-invasive maternal cell-free fetal DNA (cffDNA) is a promising biomarker for screening common genetic syndromes. Alterations in the expression levels of cffDNA in the maternal circulation have been demonstrated in abnormal pregnancies. However, the results are conflicting. The present study aimed to investigate whether cffDNA levels correlate with pregnancy complications.

Methods: The study group comprised pregnant women who presented with pregnancy complications, such as preterm birth, gestational hypertension, intrauterine growth retardation, gestational diabetes, polyhydramnios, oligohydramnios, vaginal bleeding, and placental abruption. The control group comprised women who had a normal pregnancy course. Blood samples were obtained from 500 pregnant women between 11-13 weeks of gestation. cffDNA was amplified, sequenced, and analyzed using the Next-generation Aneuploidy Test of Panorama-Natera kit. Nuchal translucency (NT) thickness as well as Pregnancy Associated Plasma Protein-a (PAPP-a) and β-human chorionic gonadotropin (β-hCG) levels were also measured. Statistical analysis was performed in 494 out of the 500 samples collected with SPSS v.26 (SPSS, Inc. Chicago IL, USA) using non-parametric methods. The parameters were normalized by the Multiples of Median (MoM) method.

Results: The expression levels of PAPP-A, β-hCG, and the NT mean MOM values were significantly different between the study and control groups (p = 0.005, p < 0.001, and p = 0.007, respectively). However, the expression levels of cffDNA and the mean MOM values were not significantly different between these two groups (p = 0.687).

Conclusion: The results of the present study support the conclusion that cffDNA expression is not altered in a series of pregnancy complications. The prognostic value of cffDNA in predicting adverse pregnancy outcomes requires further investigation.

Conflict of Interest: None declared

EP03.011 Prenatal diagnosis of Zellweger spectrum disorder caused by novel variant in PEX6 gene

Ivana Joksic 1, Mina Toljic1, Andjela Stankovic1, Zagorka Milovanovic2;3

1Gynaecology and Obstetric Clinic “Narodni front”, Genetic laboratory, Belgrade, Serbia; 2School of Medicine University of Belgrade, Belgrade, Serbia; 3Gynaecology and Obstetrics Clinic “Narodni front”, High risk pregnancy department, Belgrade, Serbia

Background/Objectives: Zellweger spectrum disorder (ZSD) is autosomal recesive disorder of peroxisome biogenesis caused by mutations in PEX genes. The clinical manifestations are very variable with hypotonia, facial dysmorphism, developmental delay, renal and liver dysfunction as most common features. We present a rare case of prenatally diagnosed ZSD caused by novel variant in PEX6 gene.

Methods: A 30-year old women was refered to genetic counseling in 27th gestation week of her third pregnancy due to presence of multiple fetal anomalies. Unilateral ventriculomegaly, cystic hygroma, enlarged microcystic kidneys and bilateral club foot were detected by ultrasound. Additionally, microcephaly, pachygyria and lysencephaly were noted on fetal brain MRI. Fetal blood sampling and clinical exome sequencing (CES, TruSight One, Illumina) were done. Variants were classified according to ACMG/AMP classification system.

Results: CES revealed two variants in PEX6 gene, c.1314_1321del and c.830del (NM_000274.4), classified as pathogenic and likely pathogenic, respectfully. Compound heterozygosity for PEX6 gene variants is consistent with diagnosis of ZSD in proband.

Conclusion: Before introduction of exome sequencing (ES) in prenatal setting, ZSD was rarely diagnosed antenatally due to unspecific and late presentation of symptoms. Frameshift variant located in exon 1, c.830del, found in our patient was previously unreported in literature and databases, and is predicted to lead to nonsense mediated decay. This case confirms utility od ES in prenatal diagnosis of fatal childhood conditions and adds to mutational spectrum of PEX6 gene.

Grants: None

Conflict of Interest: None declared

EP03.013 Arthrogryposis linked to the PIEZO2 gene causing fetal birth defects

Monika Gorządek 1;2, Hanna Moczulska1;2, Karolina Gadzalska1;2, Paulina Jakiel1;2, Ewa Juścińska1;2, Tomasz Płoszaj1;2, Sebastian Skoczylas1;2, Maciej Borowiec1;2, Agnieszka Zmysłowska1;2

1; 2Medical University of Lodz, Department of Clinical Genetics, Lodz

Background/Objectives: Arthrogryposis (OMIM:114300, 108145; ORPHA:376, 1154) is an extremely rare multiple congenital syndrome, inherited autosomal dominantly, characterized by contractures of the hands and feet of varying severity, camptodactyly, clubfoot and, less commonly, cleft palate. A fetus with arthrogryposis carries a heterozygous mutation in the PIEZO2 gene. The PIEZO2 protein plays a role in the rapid adaptation of mechanically activated (MA) currents in somatosensory neurons.

Methods: The material for the study was DNA isolated from Sherlock AX amniotic fluid (A&A BIOTECHNOLOGY). Whole exome sequencing (WES) was performed using the Twist Human Core Exome kit (Twist Bioscience). The result will be confirmed by Sanger sequencing, also with family segregation.

Results: We present a case report conducted during fetal ultrasound in the second trimester of pregnancy. Cleft palate, intrauterine growth retardation, fetal hydrocephalus, “club hand” sign, club foot, overlapping toes of the lower limbs make up the picture of abnormal fetal growth. Whole exome sequencing revealed that the fetus carries a heterozygous variant in the PIEZO2 gene (NM_001378182.1 p.Arg2799Cys), which is the genetic cause of arthrogryposis. Bioinformatics analysis identified the PIEZO2 variant as pathogenic, according to ACMG recommendations.

Conclusions: In conclusion, we found a pathogenic variant of the PIEZO2 gene in a fetus with multiple malformations. The data obtained confirms the whole exome sequencing is a highly useful tool to identify genetic background of complex defects in the fetus.

Grants:

Conflict of Interest: None declared

EP03.014 Non-invasive prenatal testing (NIPT): combination of CNV and gene analyses using an “in-house” target enrichment NGS—solution for non-centralized NIPT laboratory?

Lucie Faldynová1, Sylwia Walczysková 1, Dita Černá1, Monika Kudrejová1, Šárka Hilscherová1, Romana Kaniová1, Simona Širůčková1

1University Hospital Ostrava, Department of Molecular and Clinical Pathology and Medical Genetics, Ostrava, Czech Republic

Background/Objectives: Recent studies have integrated copy number variant (CNV) and gene analysis using target enrichment. Here we transferred this concept to our routine genetics laboratory, which is not linked to centralized non-invasive prenatal testing facilities.

Methods: From a cohort of 100 pregnant women, 22 were selected for analysis of maternal gDNA along with fetal cfDNA. Using targeted enrichment, 135 genes were analyzed, combined with aberrations of chromosomes 21, 18, 13, X, and Y. The data were subjected to specificity and sensitivity analyses, and correlated with the results from invasive testing methods.

Results: The sensitivity/specificity was determined for the CNV analysis of chromosomes: 21 (80%/75%), 18 (-/82%), 13 (100%/67%), and Y (100%/100%). Gene detection was valid for maternal gDNA. However, for cffDNA, it was not possible to determine the boundary between an artifact and a real sequence variant.

Conclusion: The target enrichment method combining CNV and gene detection seems feasible in a regular laboratory. However, this method can only be responsibly optimized with a sufficient number of controls and further validation on a strong bioinformatic background. The present results showed that NIPT should be performed in specialized centers, and that its introduction to isolated laboratories may not provide valid data.

Grants: This work was supported by Ministry of Health, Czech Republic – conceptual development of research organization (FNOs/2019).

Conflict of Interest: None declared

EP03.015 Integration of prenatal exome sequencing with extensive clinical phenotyping to assess fetal structural anomalies in a consanguineous cohort from Egypt

Sara El-Dessouky1, Mona Aboulghar2, Reza Maroofian3, Ahmed Abdelfattah4, Maha Eid5, Wessam Sharaf-Eldin6, Maha Zaki7, Alaa Ebrashy8, Hassan Gaafar8, Ahmed Saad6, Mahmoud Issa7, Shahryar Alavi9, Zahra Firoozfar9, Mohamed Ateya8, Mona Fouad8, Rana Abdella8, Ahmed Ezz Elarab8, Ebtesam Abdalla10, Nahla Abdel-Aziz11, Haitham Khaled4, Mohamed Elhodiby12, Sameh Senousy4, Homa Tajsharghi 13

1National Research Centre, Prenatal Diagnosis and Fetal Medicine Department, Human Genetics and GenomeResearch Division, Cairo, Egypt; 2Cairo University, Cairo,Egypt, Department of Obstetrics and Gynecology, Fetal Medicine Unit, Cairo, Egypt; 3, Department of Neuromuscular Disorders, london, United Kingdom; 4National Research Centre, Prenatal Diagnosis and Fetal Medicine Department, Human Genetics and GenomeResearch Division, Cairo, Egypt; 5National Research Centre, Departments of Human Cytogenetics, Human Genetics and GenomeResearch Division, Cairo, Egypt; 6National Research Centre, Medical Molecular Genetics Department, Human Genetics and GenomeResearch Division, Cairo, Egypt; 7National Research Centre, Clinical Genetics Department, HumanGenetics and Genome Research Division, Cairo, Egypt; 8Cairo University, Department of Obstetrics and Gynecology, Fetal Medicine Unit, Cairo, Egypt; 9UCL Queen Square Institute of Neurology, Department of Neuromuscular Disorders, London, United Kingdom; 10Alexandria University, Department of Human Genetics, Medical Research Institute, Cairo, Egypt; 11National Research Centre, Medical Molecular Genetics Department, Human Genetics and Genome Research Division, Cairo, Egypt; 12M.U.S.T. University, Department of Obstetrics and Gynecology, Faculty of Medicine, Cairo, Egypt; 13University of Skovde, Div of Biomedicine, Skovde, Sweden

Consangeniuos unions are associated with unfavourable genetic and perinatal outcomes in the affected offspring, raising significant public health concerns due to increased rates of infant mortality and disability resulting from congenital anomalies. The additional risk of recurrent fetal structural anomalies, a major contributor to perinatal mortality, further amplifies these concerns.

The objectives of this study were to assess diagnostic outcomes in Egyptian consangeuinous couples presenting with fetal structural anomalies and to obtain insights into the clinical subtypes of these anomalies.

The cohort consisted of 250 fetuses exhibiting sonographic abnormalities. The diagnostic approach involved prenatal comprehensive clincal phenotyping, conventional karyotyping, followed by whole exome sequencing (WES) and for cases with a normal karyotype. This combined approach yielded an overall diagnostic rate of 88% (220/250). Prenatal karyotyping detected chromosomal aberrations in 20% (50/250) of fetuses, with trisomy 18 and 13 being the most frequent anomalies. For cases with a normal karyotype, WES revealed underlying molecular genetic defects in 85% (170/200) of fetuses, primarily associated with central nervous system, skeletal and renal abnormalities. Among the molecularly diagnosed cases, 74% (125/170) exhibited pathogenic or likely pathogenic variants, or variants of uncertain significance (VUS), fully correlated with the observed defects. Inconclusive diagnoses included variants partially correlated with fetal defects and VUS.

In conclusion, the integrated application of karyotyping and WES successfully identified likely causative genetic defects in 88% of consangeuinous families with fetal anomalies, underscoring the diagnostic effectiveness of these tests in the prenatal diagnosis of fetal anomalies, especially within populations characterised by high consanguinity.

Conflict of Interest: None declared

EP03.016 A rare case of Y;14 chromosome translocation in a female fetus, detected in prenatal diagnostics

Kalina Belemezova 1, petya chaveeva2, Radostina Raynova3, Petya Andreeva2;4, Atanas Shterev2, Katerina Kavaldzhieva1, Ivanka Dimova5

1Medical University of Sofia, Biology, Sofia, Bulgaria; 2Medical Complex Dr. Shterev, Sofia, Bulgaria; 3National genetic laboratory, Sofia, Bulgaria; 4South-West University “Neofit Rilski”, Blagoevgrad, Bulgaria; 5Medical University of Sofia, Medical Genetics, Sofia, Bulgaria

Background/Objectives: We present the case of a 35-year-old pregnant woman with a second spontaneous pregnancy with no previous history of fetal abnormalities. The first trimester biochemical screening test detected increased nuchal translucency and jugular lymphatic sacs bilaterally, increasing the risk for Turner and Down syndromes.

Methods: A noninvasive prenatal test was performed, followed by amniocentesis. Amniotic fluid samples were tested using QF-PCR, and chromosome analysis was performed according to standard methods on cultured cells (GTG method).

Result: The NIPT result showed gender disagreement compared to the second fetal morphology exam which was consistent for a female fetus. Following the amniocentesis, the QF-PCR showed the presence of genetic material consistent with 47, XXY karyotype. Finally, the cytogenetics showed a very rare non-balanced translocation between chromosomes Y (missing the SRY region) and chromosome 14 (karyotype: 46,XX,t(Y;14)(q10;q10)).

Discussion: Cases of Y chromosome/autosome translocation are very rare. There are only a few cases in the literature reporting a similar unbalanced translocation, usually with the presence of the SRY region, resulting in a phenotypically normal male with infertility. Moreover, cases of non-SRY Y to autosome translocation are extremely rare and are related to gonad dysgenesis and high risk of gonad malignancy.

Conclusion: The case we report shows the importance of different testing approaches whenever there is inconsistency between the results from the NIPT and the fetal morphology. There is a need for further studies of patients with Y chromosome translocation to better understand the possible complications and outcomes in those cases.

Conflict of Interest: None declared

EP03.017 Alport Syndrome and Pregnancy: Clinical and Genetic Analysis

Maria MALARSKA 1, Paulina Pachniak1, Hanna Moczulska1, Paulina Jakiel1, Karolina Gadzalska1, Agnieszka Pukajło-Marczyk2, Katarzyna Kiliś-Pstrusińska2, Katarzyna Rajfur3, Alicja Majos4, Agnieszka Zmysłowska1

1Medical University of Lodz, Lodz, Poland, Department of Clinical Genetics, Łódź, Poland; 2Wroclaw Medical University, Wroclaw, Poland, Department of Pediatric Nephrology, Wrocław, Poland; 3Institute of Health Sciences, University of Opole, Poland, Opole, Poland; 4Medical University of Lodz, Lodz, Poland, Department of General and Transplant Surgery, Łódź, Poland

Background: Alport syndrome (AS) is a rare genetic disorder characterized by progressive damage to the basement membrane of the kidney, as well as the middle ear and retina. The purpose of this study was to analyze the impact of Alport syndrome on the course of pregnancy and to identify potential pregnancy complications associated with a genetic diagnosis of Alport syndrome.

Methods: The analysis included 12 pregnancies in 6 patients with confirmed Alport syndrome without clinical symptoms and 46 pregnancies in 23 patients with symptoms, taking into account various pregnancy parameters and molecular diagnosis of AS. Genetic testing was performed in all children using next-generation sequencing or Sanger sequencing method.

Results: Among the 12 children whose mothers were asymptomatic, 9 (75%) had genetic confirmation of Alport syndrome. Polyhydroamniosis was found in 16.67% of cases, especially in pregnancies with male fetuses and genetically confirmed Alport syndrome. In the analyzed group of 23 pregnant women with symptomatic AS, 85.29% of the children received genetic confirmation of the diagnosis. In the analyzed group, 14 pregnancies (37.84%) had polyhydroamniosis, the cause of which was not determined. Genetic testing of the children confirmed Alport syndrome in 85.29% of them.

Conclusion: The occurrence of polyhydroamniosis in pregnancies with confirmed Alport syndrome could suggest a possible influence of the syndrome on the presence of this complication. It is worth continuing research to understand the impact of Alport syndrome on pregnancy, with an emphasis on identifying potential risk factors and better managing the care of patients with this condition.

Conflict of Interest: None declared

EP03.018 Prenatal case with multiple sulfatase deficiency caused by homozygous SUMF1 mutation

Rebecca Gembicki 1, Michael Gembicki2, Jan Weichert2, Moneef Shoukier3, Yorck Hellenbroich1, Malte Spielmann1;4

1Institute of Human Genetics, University of Lübeck, Lübeck, Germany; 2Division of Prenatal Medicine, Department of Obstetrics and Gynecology, University Hospital of Schleswig-Holstein, Lübeck, Germany; 3Pränatal-Medizin München, München, Germany; 4Institute of Human Genetics, University of Kiel, Kiel, Germany

Multiple sulfatase deficiency (MSD) is a rare autosomal recessive metabolic disease caused by pathogenic homozygous or compound heterozygous variants in the sulfatase modifying factor 1 (SUMF1) gene. This gene encodes for formylglycine generating enzyme (FGE) which is involved in posttranslational modification and catalytic activation of the entire family of sulfatases.

The wide spectrum of clinical features combines symptoms of single sulfatase deficiencies whereby protein stability and residual activity influence disease severity. The most common ones are neurologic deterioration with mental retardation, skeletal anomalies, organomegaly, and ichthyosis.

Different clinical subtypes of MSD (neonatal, late infantile and juvenile) have been described based on the age of onset, the rate of progression and the severity of the disease.

The neonatal is the most severe with rapid progression and death within the first two years of life. Late infantile, the most common one, can subdivided into mild (onset beyond the second year of life) and severe (skeletal changes, progressive loss of mental/motor abilities by age 5 years, death within the first decade of life). The juvenile form is the rarest with later onset and slow progression.

Here, we report a prenatal case of MSD caused by the homozygous missense gene mutation c.739G>C,p.(Gly247Arg) in SUMF1 detected by exome analysis which was previously described with the late infantile subtype. The fetus shows multiple ultrasonographic abnormalities: decreased growth of all long bones, multiple cerebral anomalies (agenesis of the corpus callosum, colpocephaly, cerebellar vermis hypoplasia, frontal bossing), short ribs and a mild cardiomegaly.

Conflict of Interest: None declared

EP03.020 Further evidence for an attenuated phenotype of in-frame DMD deletions affecting the central rod domain of dystrophin around exon 48

Olga Bürger 1, Angelika Humbel1, Ivan Ivanovski1, Alessandra Baumer1, Anita Rauch1

1University of Zurich, Institute of Medical Genetics, Zürich, Switzerland

Background: Alterations in the X-linked recessive DMD gene cause dystrophinopathies with a broad clinical spectrum most commonly ranging from Duchenne muscular dystrophy (DMD) or Becker muscular dystrophy (BMD) to cardiomyopathy or intellectual disability (ID). Carrier females are commonly unaffected but may show signs of dystrophinopathies. In addition, few asymptomatic male carriers with elevated creatine kinase levels have been described possibly related to deletions around exon 48.

Results: We report an attenuated phenotype in a three-generation family with a deletion of exon 48 of the DMD-gene with clinically unaffected carrier males and females. We confirmed deep intronic breakpoints in this family by genome sequencing.

Conclusion: Predicting the phenotype in DMD-sequence alterations is challenging. The “reading frame rule” suggests that Duchenne muscular dystrophy results from DMD-mutations causing an out-of-frame transcript, whereas the milder Becker muscular dystrophy results from mutations causing an in-frame transcript. Exceptions to the “reading frame rule” are estimated at a rate below 10% [Aartsma-Rus et al 2006].

Deletions in similar regions (between exons 45 – 48 of the DMD-gene) have also been reported in patients with a severe Duchenne phenotype. This discrepancy may result from unprecise breakpoint definitions in the past that may not sufficiently discriminate between in-frame and out-of frame copy number variants, but additional modifiers may also be possible. Until more cases are characterized in more details, predicting the individual phenotype requires multiple carriers across different age groups and both genders. Cautious counselling is warranted especially regarding prenatal diagnostics.

Grants: University of Zurich clinical research priority program praeclare

Conflict of Interest: None declared

EP03.021 Paternally Inherited NFIA-Related Disorder: Prenatal and Postnatal Diagnosis at Once

Nader Khaleghi Hashemian 1, Gioia Mastromoro1, Daniele Guadagnolo1, Enrica Marchionni1, Allessandra Terracciano2, Antonio Novelli2, Antonio Pizzuti1

1Sapienza University of Rome, Department of Experimental Medicine, Rome, Italy; 2Bambino Gesù Children’s Hospital, Laboratory of Medical Genetics, Rome, Italy

Background/Objectives: We report on a male fetus exhibiting mild unilateral isolated cerebral ventriculomegaly detected through morphological ultrasonography and confirmed by magnetic resonance imaging (MRI). Upon genetic counseling, the 30-year-old father of the fetus referred a personal history of congenital hydrocephalus treated surgically at 9 months and posterior agenesis of the corpus callosum, without specific dysmorphic features or neurodevelopmental disorders.

Methods: Amniocentesis with standard karyotype and chromosomal microarray investigation yelded normal results. Exome sequencing targeting genes associated with brain anomalies was performed on the father of the fetus and his parents.

Results: A de novo heterozygous likely pathogenic variant c.82_83dup; p.(Trp30HisfsTer28) in the NFIA gene (NM_001134673) was identified in the father of the fetus. Monoallelic loss-of-function variants in NFIA are associated with “Brain malformations with or without urinary tract defects” (MIM#613735), aligning with his clinical presentation. Segregation analysis identified the variant also in the fetus. Subsequent fetal MRI at 27 weeks showed hydrocephaly-induced macrocephaly, thin cerebral cortex, agenesis of the septum pellucidum, thin corpus callosum, bilateral frontal polymicrogyria with abnormal sulcation, and reduced fetal movements. The couple opted for pregnancy termination. Fetal anatomopathological evaluation revealed hypertelorism, prominent nasal bridge, long philtrum, micrognathia, microstomia, low hairline, and low, posteriorly rotated poorly rimmed ears. Internal organ examination showed undescended testes and anomalies in pulmonary lobulation and bronchial segmentation, including an additional left lung lobe.

Conclusion: We report a hereditary NFIA-related disorder, typically arising de novo. Family history prompted exome sequencing investigation which identified a paternally trasmitted variant to the fetus.

Grants: None

Conflict of Interest: None declared

EP03.022 Whole Genome Sequencing Reveals Incidental Findings That Are Actionable In The Prenatal Setting – Case Report From a Consanguineous Population

zohra hasan 1, MAHRUKH NASIR1;2, Fizza Akbar1, Salman Kirmani1

1Aga Khan University, Division of Women and Child Health, Karachi, Pakistan; 2International Center for Chemical and Biological Sciences (ICCBS), Dr.Panjwani Centre for Molecular Medicine and Drug Research, Karachi, Pakistan

Background: Whole Exome Sequencing (WES) and Whole Genome Sequencing (WGS) are becoming primary tools for diagnosing genetic disorders. Incidental findings can be crucial, particularly in consanguineous families, providing actionable insights for both the proband and family members. This case highlights the significance of WGS in identifying incidental pathogenic variants with immediate clinical implications.

Methods: A consanguineous couple, having experienced unexplained neonatal deaths, presented their 5-month-old with severe hypotonia and myopathic changes. WGS was employed for comprehensive genomic analysis. A homozygous pathogenic variant in TK2 was identified, confirming Mitochondrial DNA Depletion Syndrome Type 2. Incidentally, a pathogenic variant in BRCA2 was also discovered. Parental testing revealed heterozygosity for both TK2 and BRCA2 variants. Subsequent prenatal testing for both variants was offered in the next pregnancy.

Results: Prenatal testing in the subsequent pregnancy unveiled a fetus homozygous for the BRCA2 pathogenic variant, associated with Fanconi Anemia, congenital anomalies, and early cancer predisposition. Faced with this diagnosis, the family opted to terminate the pregnancy at 21 weeks. This case underscores the power of WGS in not only identifying primary genetic disorders but also revealing secondary findings with immediate clinical relevance.

Conclusion: WGS emerges as a potent diagnostic tool, capturing both primary and incidental findings. In this case, relying solely on targeted gene panels would have overlooked the BRCA2 variant. The case emphasizes the importance of comprehensive genomic analysis in offering timely interventions and appropriate genetic counseling for families facing hereditary conditions.

Conflict of Interest: None declared

EP03.023 Assessment of maternal cell contamination in all prenatal samples by quantitative fluorescent PCR

Kajsa Nilsson 1

1Skåne University Hospital, Clinical genetics, Lund, Sweden

Background/Objectives: The contamination of fetal samples with maternal cells, Maternal Cell Contamination (MCC), can interfere in diagnostic prenatal testing. MCC is thus recommended to be assessed in clinical practice regardless of the genetic disorder or mode of inheritance. At the Department of Clinical genetics in Lund, MCC analysis was introduced for all prenatal samples in June 2021. Here we present the outcome of this clinical approach.

Methods: Quantitative Fluorescent PCR (QF-PCR) was performed on all fetal samples obtained by amniocentesis or chorionic villus sampling and on maternal blood sample between June 2021 and November 2023 as part of clinical routine.

Results: Out of overall 1624 pregnancies analyzed, significant maternal cell contamination (MCC) was detected in 25 cases (1.5%).

Conclusion: A significant proportion of the prenatal samples were contaminated by maternal cells. To determine the pure fetal origin of fetal samples undergoing genetic analysis and to provide good laboratory practice as well as correct result interpretation, it is of high importance that MCC is assessed in all fetal samples.

Grants:

Conflict of Interest: None declared

EP03.024 From a NIPS false positive to a chromosomal rearrangement: a case report

cristina Ferreira 1;2, Ana Rita Tarelho1, Bárbara Marques1, Silvia Serafim1, Sónia Pedro1, Cristina Alves1, Mónica Viegas1, Ângela Ferreira3, Hildeberto Correia1

1Instituto Nacional de Saúde Doutor Ricardo Jorge, I.P., Departamento de Genética Humana, Lisboa, Portugal; 2; 3Centro Hospitalar Universitário do Algarve – Hospital de Faro E.P.E., Serviço de Obstetrícia / Diagnóstico Pré-Natal, Faro, Portugal

We report a case of a pregnant woman at 21 weeks and six days of gestation who underwent cell-free fetal DNA based non-invasive prenatal screening (NIPS) without prior aneuploidy screening.

The initial NIPS result indicated a 3% fetal fraction with a high risk for chromosome X monosomy. To confirm this finding, amniocentesis was performed, and subsequent QF-PCR rapid aneuploidy test revealed a normal result for chromosome X in a female fetus, contradicting the NIPS outcome. Unusually, all polymorphic markers on chromosome X were non-informative, prompting additional investigation using multiplex-ligation dependent probe amplification (MLPA), which results revelled a terminal deletion in Xq22.3.

In order to characterize the deletion, microarray analysis was performed. The comprehensive analysis revealed a 74.51 Mb terminal deletion in Xq21.1q28 and an 85.52 Mb terminal duplication in 2p25.3p11.2, indicative of a derivative chromosome resulting from a translocation between the short arm of chromosome 2 and the long arm of chromosome X. This complex genomic rearrangement was confirmed by karyotyping. Parental karyotyping will be conducted promptly to ascertain the recurrence risk.

This case underlines the necessity of confirming NIPS results considered as high risk. While the initial suspicion of monosomy X was not substantiated, the pursuit of a deeper investigation revealed an entirely unexpected genomic anomaly with a more severe prognosis. This highlights the importance of thorough investigation following a NIPS high risk result, as QF-PCR rapid aneuploidy test, used many times as standalone method in prenatal diagnosis following NIPS high risk results, would not have solved this case.

Conflict of Interest: None declared

EP03.025 Necessity to define prenatal phenotypes to enhance diagnosis in multiple congenital anomalies.

Carla Martín-Grau1, Carmen Orellana 2, Mónica Roselló Piera2, Laia Pedrola2, Juan Silvestre Oltra Soler2, Sandra Monfort Membrado2, Marta Domínguez Martínez1, Alfonso Caro Llopis1, Francisco Martinez2

1Instituto de Investigación Sanitaria La Fe (IISLAFE), Genetics Unit, Translational Genetics Research Group, Valencia; 2Hospital Universitario y Politecnico La Fe, Genetics Unit, Translational Genetics Research Group, Valencia

Background/Objectives: The Human Phenotype Ontology (HPO) provides a standardized vocabulary of phenotypic abnormalities related to human diseases. It is widely-known that HPO can be used to support differential diagnoses and translational research. HPO currently contains over 13,000 terms that mostly correspond to medically relevant postnatal phenotypes. However, HPO database only contains 262 terms directly correlated with congenital anomalies (CA). In the absence of specific fetal HPOs, comprehensive multidisciplinary knowledge about prenatal phenotypes is crucial to improve diagnostics tests.

Methods: 32 fetal samples with ultrasound and/or anatomopathological diagnosis of CA were included with normal results in the previous prenatal karyotype and 750K SNP-array performed. WES was performed using SureSelectXT HS ExonV8 (Agilent Technologies) for enrichment and NextSeq (Illumina) for sequencing. Segregation studies were confirmed by Sanger sequencing.

Results: WES diagnostic yield was 43.8% in our cohort. In 5/14 patients (35.7%), the clinical phenotype standardized by HPO terms used did not facilitate the interpretation of genetic variants detected due to the lack of genotype-phenotype correlation in fetal patients. Novel phenotype terms were described in PIGP, EP300, ROBO1, FOXP3 and ARMC9 genes.

Conclusion: Detailed fetal phenotypes in HPO terms will support filtering and prioritization of variants in order to increase diagnostic yield in CA using WES. However, a significant percentage of fetal cases may be underdiagnosed. Our results illustrate the complexity of prenatal phenotype and underline the need for further researches to better reinforce prenatal usage of HPO terms in unreported prenatal diseases.

Grants: Supported by Instituto de Salud Carlos III through “PI22/01127” and “PI22/00272” projects.

Conflict of Interest: None declared

EP03.027 A retrospective study on 162 patients that benefited of a NIPT analyze

Eusebiu Vlad Gorduza 1;2, Violeta Martiniuc2;3, Lavinia Caba1, Andreea Florea1, Ana Grigore1, Mihaela Gramescu1, Elena Mihalceanu1;2

1“Grigore T. Popa” University of Medicine and Pharmacy, Medicine of Mother and Children, Iasi, Romania; 2“Cuza Voda” Obstetrics and Gynecology Hospital, Iasi, Romania; 3SC Cytogenis SRL, Iasi, Romania

Background/Objectives: To analyze the results of NIPT test.

Methods: We made a NIPT retrospective study between August 2022 and January 2024on patients from Romania.

Results: The cohort has 162 patients, with 3 cases inconclusive (BMI ≥ 28.58). 104 patients benefited from a full test (through which full and partial aneuploidies of all chromosomes were analyzed) while 55 cases opted for the identification of only chromosomes 21, 18, 13, X and Y. We analyzed 8 twin pregnancies with normal results. We found 4 pregnancies with high risk: monosomy X, trisomy 15, microdeletion 1q21 and trisomy 18, but only the last was confirmed by chromosomal analyze after amniocentesis. No chromosomal diseases were identified in all the children born. The age of women that applied for NIPT was between 26 and 49 years: 31 (<30 years; 19.49%) 65 (30 – 35 years; 40.88%) 47 (36 – 40 years; 29.55%) 17 (>40 years; 10.69%). Between different age interval we did not found significative differences that concern medium value of free fetal DNA fraction (FFDNA) (<30 years – 13.91%; 30 – 35 years – 14.06%; 36 – 40 years – 13.20%; >40 years – 11.26%). The tests were applied at: 11 WA (18 cases; 11.32%; medium FFDNA - 13.86%); 12 WA (33 cases; 20.75%; medium FFDNA - 13.66%); 13 WA (48 cases; 30.18%; medium FFDNA - 12.96%); 14 WA (22 cases; 13.83%; medium FFDNA - 11.94%); 15 WA (19 cases; 11.94%; medium FFDNA – 12.02%); ≥16 WA (19 cases; 11.94%; medium FFDNA – 14.21%).

Conclusion: Our study confirmed the quality of NIPT as prenatal screening for chromosomal disorders.

Conflict of Interest: None declared

EP03.028 Similarities and differences in performance between two WGS-based NIPT tests – 10 years of experience of the Genomed SA laboratory

Monika Jurkowska1, Ewa Matczyńska1, Anna Wąsowska1, Przemysław Łyszkiewicz1, Magdalena Kania1, Elżbieta Gregorczyk1, Paweł Ogrodnik1, Maria Jędrzejowska1, Marek Zagulski1, Anna Boguszewska-Chachulska 1

1Genomed SA, Warsaw, Poland

Non-invasive prenatal testing (NIPT) becomes a routine method of prenatal screening. Numerous testing approaches, methodologies, ranges, report options are being applied worldwide. The objective of this study was to compare data on the performance of two large series of different NIPTs provided by the Polish pan-country laboratory.

Altogether 41064 reports, issued between 2014 and 2023, were analysed. Data have been extracted from a test-dedicated database, order forms, final reports, follow-up sheets and personal communications. Two NGS-based NIPT technologies were applied: BGI (NIFTY – 15850 samples) and Illumina (VeriSeq v1 - 5191 samples and v2 – 20020 samples, under the SANCO brand). The tests differed in the applied chemistry, sequencers, bioinformatic algorithms and range of analysis.

Despite similar populations tested, significant differences in performance were noticed between tests (Table 1, in bold).

Feature (% if not specified otherwise)

BGI

Illumina

v1

v2

Common range

T21

1.58

1.86

1.54

T18

0.42

0.33

0.40

T13

0.23

0.19

0.21

SCA

0.98#

0.29

0.45

T9, T16, T22

0.12

NA

0.14

Other RAAs (excluding T7)

0.06

NA

0.25

CNV

0.18

NA

0.56

feedback

17.24

NA

19.64

true positive CNV

NA

54.55

false negative T21

0.044

0.028

turnaround time (days)

5.5

2.5

no-call rate

0.44

0.14

0.20

  1. # 50% of FP samples re-tested with Illumina turned out to be negative
  2. The Illumina technology, based on PCR-free, low-coverage WGS, with genome-wide reporting and dynamic fetal fraction incorporation in metrics, enables superior performance in turnaround time, no-call rate as well as false positive and false negative ratios.

Conflict of Interest: None declared

EP03.029 Prenatal diagnosis of Proteus Syndrome

Luana Giovannangeli1, Virginie Magry 2, Florence Jobic1, Kahia Messaoudi2, Alexis Billes1, marianne perriere2, Ségolène Delmas-Lanta3, Alice Masurel4, Guillaume Jedraszak2, Gilles Morin1

1CHU Amiens-Picardie, Genetics, Amiens, France; 2CHU Amiens-Picardie, Constitutional Genetics Laboratory, Amiens, France; 3CHU Amiens-Picardie, Gynecology, Amiens, France; 4CHU Amiens-Picardie, Neuropediatry, Amiens, France

Background/Objectives: Proteus syndrome is a rare disorder (<1/1,000,000) characterized by progressive overgrowth which affects commonly the skeleton, skin, adipose and central nervous system. It results from a somatic activating variation in the AKT1 gene. The c.49G>A (p.Glu17Lys) variation is the most reported. Proteus syndrome has a variable severity, with a mortality rate of approximately 25% before 22 years of age. We report the second case of Proteus syndrome prenatally diagnosed with whole exome sequencing.

Clinical Report: A 34-year-old woman was referred to the prenatal diagnosis department for non-visualized anterior brain structures and suspicion of micro-penis. Subsequent ultrasonography at 21 weeks of gestation (WG) confirmed absent septum lucidum, thick corpus callosum, genital abnormalities and macrosomia. Array-CGH did not found pathogenic imbalances (46,XX). Follow-up ultrasounds and MRI at 25 and 27 WG showed persistent abnormalities and megalencephaly, leading to exome sequencing on amniocytes. A mosaic variation in AKT1 (c.49G>A) with an allele frequency of 25% was identified. Medical termination of pregnancy was performed at 30 + 1 WG.

Conclusion: Only one case of prenatal diagnosis with exome sequencing has been reported to date with similar clinical presentation (Abell et al. 2020). The diagnosis challenges lie in the clinical variability with usual postnatal beginning and the mosaic nature of the syndrome, notably in case of low mosaicism. Whole exome sequencing, with a coverage depth of 130X, was efficient in this case, offering a precise diagnosis.

Conflict of Interest: None declared

EP03.030 Prenatal diagnosis of a mosaic rearrangement of chromosome 18: the result of dicentric chromosome breakage?

Laura Feyereisen1, Tony Yammine 1, Leila Sahmoune Rachedi2, Jean-Paul Bory2, Victor Dupont-Gaudin3, Jean-Marc Dossot3, Véronique Dalstein1, Poirsier Céline4, Emilie Landais1

1CHU de Reims, Structure de Génétique-Hématologie-Immunologie, Pôle de Biologie Territoriale, Reims, France; 2CHU de Reims, Diagnostic Prénatal - Service de Gynécologie Obstétrique, Reims, France; 3Laboratoire de Biologie Médicale Bioxa, Reims, France; 4CHU de Reims, Service de Génétique Clinique, Reims, France

Background: A primiparous 19-year-old patient was referred to our pluridisciplinary center for prenatal diagnosis at 26-weeks of gestation. Ultrasound examination showed severe asymmetrical intrauterine growth restriction (IUGR; with decreased abdominal and head circumferences, but spared femoral length), a single umbilical artery, and pericardial effusion.

Methods/Results: An amniocentesis was performed; rapid prenatal aneuploidy testing by fluorescence in situ hybridization (FISH) found no anomaly of chromosomes 13, 18 and 21. However, microarray-based comparative genomic hybridization (aCGH) of uncultured amniocyte DNA showed Profile A: a partial trisomy 18 with a terminal 18q monosomy (cf. table below). Conversely, the karyotype performed on short-term cell culture found 18q monosomy without partial trisomy, as confirmed by aCGH profile B. These seemingly contradictory findings made genetic counseling challenging. Due to the poor prognoses of both chromosome 18 rearrangements and the severity of IUGR, the patient elected for a therapeutic termination of pregnancy. Additional samples were collected for further testing. Array CGH of umbilical cord found Profile B. Placenta and amniocyte analyses found Profile C, a mosaic near-complete trisomy 18 with 18q monosomy.

Profile-A

Profile-B

Profile-C

- 18p11.32q21.31 trisomy (55Mb, 50% mosaic)

- 18q23qter monosomy (3Mb, 100%)

- 18q21.32qter monosomy (23Mb,100%)

- 18p11.32q23 trisomy (75Mb, 50-70% mosaic)

- 18q23qter monosomy (3Mb, 100%)

Conclusion: We hypothesize that profile A reflects two concurrent cellular populations: one presenting a dicentric chromosome 18 (profile C), and another with a derivative resulting from dicentric chromosome breakage (profile B). This case’s complexity would not be apparent had only cultured amniocytes been analysed.

Conflict of Interest: None declared

EP03.031 ANKS6 variants underlie polycystic kidneys in prenatal and neonatal cases

Mohamed AlHamed 1

1King Faisal Specialist Hospital & Research Centre, Clinical Genomics, Riyadh, Saudi Arabia

Background/Objectives: Nephronophthisis (NPHP) is an autosomal recessive disorder that can result in severe renal failure. ANKS6 gene has an important role for early renal development and encodes a protein that interacts with other proteins of the cilium. Fetal ultrasound imaging and diagnosis of fetuses with ANKS6 variants still underestimated. Here, we report the detection of ANKS6 variants in consanguineous families with polycystic kidney at early stages of life.

Methods: Three unrelated Saudi Arabian patients (2 prenatal and 1 neonate) were investigated. These cases were referred due to a history of echogenic kidneys. After clinical evaluation and fetal ultrasound imaging, whole-exome sequencing (WES) was performed to identify molecular genetics causes associated with echogenic kidney phenotype.

Results: Two sequence variants were detected in ANKS6 gene. A homozygous missense novel variant ANKS6: c.1159A>C was detected in families 1 and 2. The third family harboured a homozygous loss of function known variant ANKS6: c.907+2T > A.

Conclusion: We have identified 2 ANKS6 variants in all 3 polycystic kidney families. The findings provide an expanded clinical manifestation of NPHP and emphasize the utility of WES in the phenotype of echogenic kidney at prenatal settings.

Grants: None

Conflict of Interest: None declared

EP03.032 Prenatal genetic testing in the era of next-generation sequencing: experience of a Fetal Medicine Unit in a Spanish hospital

Gema Escribano-Serrano 1, Diana Salinas-Chaparro1, mar borregán prats1, Judith Armstrong1;2;3, Delia Yubero1;2;3, Laura Martí1;2, Clara Xiol1;2, Guerau Fernandez1;2;3, Maria Edda Marimon Garcia4, Miriam Illa Armengol4;5, Miriam Perez Cruz4;5, Elisenda Eixarch Roca4;6, Mar Bennasar Sans4, Joan Sabria Bach4;5, Francesc Palau1;2;3;7

1Dept. of Genetic Medicine - IPER, Hospital Sant Joan de Déu, Barcelona, Spain; 2Institut de Recerca Sant Joan de Déu, Barcelona, Spain; 3CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain; 4Fetal Medicine Unit. BCNatal. Centre de Medicina Maternofetal i Neonatal, Hospital Sant Joan de Déu - Hospital Clínic, Barcelona, Spain; 5Maternal and Child Health and Development Network II (SAMID II), Instituto de Salud Carlos III (ISCIII), Sub-Directorate General for Research Assessment and Promotion and the European Regional Development Fund (ERDF), Madrid, Spain; 6Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; 7Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain

Background/Objectives: Foetal abnormalities affect approximately 2–3% of pregnancies. In prenatal diagnosis, clinical exome sequencing (CES) is commonly offered in complex cases with normal karyotyping and chromosomal microarray (CMA). Prenatal CES (pCES) poses inherent challenges, associated with gene filtering strategies, unrecognized foetal phenotypes, and/or genetic variant interpretation. This study aims to understand the diagnostic yield of pCES in a Women’s and Children’s Hospital.

Methods: From 2017 to 2023, 125 foetal samples obtained through amniocentesis and chorionic villus sampling, underwent pCES analysis, either singleton (74) or trio (51). Samples originated from pregnancies with foetal anomalies, where both QF-PCR and CMA studies were negative. Searching for candidate genes was performed by virtual gene panels genes and/or HPOs. We compared indications of ultrasound anomalies and virtual gene panel or pCES according to HPOs criterion. Pregnancies with identified genetic conditions received genetic counselling.

Results: Regarding the most prevalent clinical indications, 29 foetus (23%) had more than one foetal anomaly, followed by 18 (14%) which were clinically suggestive of a possible skeletal dysplasia, and 16 (13%) of cardiac alteration. Both virtual panels and HPO selection yielded similar diagnostic rates: 38% (15/39) of variants through gene panel and 33% (13/39) by HPOs.

Conclusion: The data suggest that both disease-related gene panel and HPO-addressed genetic analysis have similar diagnostic yield. Precision or deep imaging phenotyping is the main criterion to improve the diagnostic rate of pCES and pCES-trio, which would facilitate prenatal genetic counselling and informed decision-making.

Conflict of Interest: None declared

EP03.033 Rare CNVs of HBA and HBB genes identified in a group of thalassemic patients and heterozygotes of Greek origin

Panagiota Kekelou1, Cristos Chassanidis1, Thomai Tsantzali1, Evangelia-Eleni Delaki1, Effrossini Boutou1, Eleni Yfanti1, Maria Baou1, Panayiotis Kyriazopoulos1, Stamatia Theodoridou2, Veroniki Komninaka3, Spiridon Kanellakis4, Georgia Ikonomou5, Dimitra Pessini6, Maria Dimopoulou7, Angeliki Balassopoulou 1

1Laiko Hospital, Unit of Genetics, Athens, Greece; 2Hippokration General Hospital, Thalassemia Prevention Unit, Thessaloniki, Greece; 3Laiko Hospital, Thalassemia Prevention Unit, Athina, Greece; 4Karamandaneio Prefecture Children Hospital of Patras, Thalassemia Prevention Unit, Patra, Greece; 5Larissa General Hospital Koutlimbaneio & Triantafylleio, Thalassemia Prevention Unit, Larisa, Greece; 6Agrinio General Hospital, Thalassemia Prevention Unit, Agrinio, Greece; 7Laiko Hospital, Unit for Thalassemias, Athens, Greece

Background/Objectives: Copy Number Variants (CNVs) are frequently identified in α thalassemia but they are rarely found in β thalassemia. Only some of them are routinely screened particularly in α thalassemia. This study aims to analyze the HBA and HBB CNV’s in thalassemic patients and heterozygotes whose genotypes were not defined/fully defined after routine genetic analysis

Methods: 59 DNA samples were studied. SALSA® MLPA® kits (Probemix P140, P102) MRC Holland, were used while fragment analysis was carried out in SeqStudio™ Genetic Analyzer (Thermo-Fischer Scientific). Analysis was performed using the Coffalyser.Net™ software.

Results: 9 CNV’s were identified in 16 samples. 4 deletions with sizes at least 257kb, 11,1kb, 131kb, 9,8kb, 1 duplication of the entire α globin cluster and 1 homozygous α gene triplication were identified in α globin cluster. The duplication and triplication were in compound heterozygosity with typical β thalassemia mutations giving rise to β thalassemia intermedia phenotypes. 2 deletions, 7.2kb (discontinuous) and 173kb and 1 duplication of the entire β globin cluster containing two β globin genes one carrying the wild type sequence an the other the HBB:c.93-21G > A variant, were identified in β globin cluster.

Conclusion: CNV’s analysis revealed nine rare variants three of which (deletion 9.8kb, deletion 173kb and β locus duplication) have not been previously reported. The duplication of entire globin gene clusters -particularly β- is a rare genetic phenomenon of great interest with few references so far. Characterization of these molecular lesions allows the accurate diagnosis and enables appropriate genetic counseling in case of prenatal diagnosis.

Conflict of Interest: None declared

EP03.034 Silent cis should not be silent

Neeta Lakhani 1, Vijayalakshmi Salem Ramakumaran1, emily craft1

1Leicester Royal Infirmary, Clinical Genetics, United Kingdom

Background/Objectives: Silent cis carriers of Spinal Muscular Atrophy (SMA) present a critical yet often overlooked aspect of the disease landscape. With the increased frequency of SMA carriers in our region, understanding the implications of silent cis carriers becomes imperative. Silent carriers, while phenotypically normal, harbor pathogenic heterozygous SMA alleles, which are not identified using current testing techniques. This poses significant implications for reproductive options and genetic counselling.

Methods: The estimated prevalence in our cohort is more than twice the reported prevalence of 4%. The chance of false negative parental carrier status results due to test limitations will result in erroneously concluding that the affected individual has a de novo variants and inaccurate recurrence estimation for the family. This raises ethical and emotional considerations regarding reproductive decision-making.

Results: Furthermore, the haplotyping of silent carriers are crucial adjunct to carrier testing, for accurate risk assessment and genetic counselling. Through this we can predict the likelihood of disease transmission and guide personalized reproductive strategies.

Conclusion: Here we present a case series of families with silent cis carrier status and how the family journey, though extended, significantly changes their reproductive outcomes.

Grants: None

Conflict of Interest: None declared

EP03.035 NIPT high-risk of rare autosomal aneuploidies and copy number variations are related to adverse pregnancy outcomes

Laia Pedrola 1, Mónica Roselló Piera1, Carla Martín-Grau2, Juan Salvador Rubio Moll3, Rosa Portero Gómez3, Beatriz Marcos Puig3, Jose Vicente Cervera1, Ramiro Quiroga3, Carmen Orellana1

1Hospital Universitario y Politecnico La Fe, Genetics Unit, Translational Genetics Research Group, Valencia; 2Instituto de Investigación Sanitaria La Fe (IISLAFE), Genetics Unit, Translational Genetics Research Group, Valencia; 3Hospital Universitario y Politecnico La Fe, Obstetrics and Gynaecology Unit, Valencia

Background/Objectives: Genome-wide prenatal cell-free DNA (cfDNA) screening can be used to screen for a wide range of fetal chromosomal anomalies in pregnant patients. In recent years, the scope of prenatal cfDNA screening has expanded to include optional screening for rare autosomal aneuploidies (RAAs) and copy number variations (CNVs). Recent publications have reported that these rarer chromosomal abnormalities have a clinical impact.

Methods: Blood samples were obtained from pregnant patients at 21 public maternity health centers from 1st March 2020 to 31st December 2022. For the cfDNA screening analysis, frozen plasma samples were processed using the VeriSeq™ NIPT Solution v2 assay (Illumina). Clinical follow-up was attempted for all high-risk cases. Confirmatory methodologies used included fluorescent in situ hybridization (FISH), karyotyping, 750 K SNP-array and quantitative fluorescent PCR (QF-PCR).

Results: A total of 6098 samples were analysed during the study period and 5880 (96.4%) had genome-wide cfDNA screening. A total of 31 RAAs and 31 CNVs cases were detected. The most common RAAs in our cohort were trisomy 7 and trisomy 16. RAA and CNV cases were related with pregnancy and perinatal adverse outcomes such as preeclampsia, gestational diabetes, spontaneous abortion, and congenital structural abnormalities.

Conclusion: Currently, professional medical societies do not recommend genome-wide cDNA screening in general pregnant population. However, our results show the importance of screening for less common chromosomal anomalies because it could identify patients at risk of perinatal complications who may require additional screening or clinical management during pregnancy.

Conflict of Interest: None declared

EP03.036 X chromosome rearrangement in familial Turner syndrome

Kristina Aleknavičienė 1;2, Rasa Traberg1;2, Zivile Zemeckiene1;1, Rasa Ugenskiene1;2

1Lithuanian University of Health Sciences, Department of Genetics and Molecular Medicine, Kaunas; 2Hospital of Lithuanian University of Health Sciences Kauno klinikos, Department of Genetics and Molecular Medicine, Kaunas

Background/Objectives: Turner syndrome, which affects 1 in 2500 females, involves the partial or complete absence of one X chromosome, resulting in diverse developmental abnormalities.

Methods: A 34-year-old woman, with a history of five miscarriages, autoimmune thyroiditis, and V-factor mutation, underwent Non-Invasive Prenatal Testing (NIPT) during her sixth pregnancy. Despite a low risk for T13, T18 and T21, the information on sex chromosomes was not provided. Fetal polyhydramnios was diagnosed, and at the 38th week, a neonate (2380 g, <3% hypotrophic) was born. At 3-4 months, an atrial septal defect was identified. Infant molecular karyotyping based on whole-genome sequencing (3x coverage, >50 kb resolution) revealed a derivative X chromosome with a 5283 kb terminal deletion (p22.33p22.32) and a 15026 kb duplication (q27.1q28), impacting 30 and 156 genes, respectively. Mother’s FISH analysis confirmed a subtelomeric X chromosome change, identical to her daughter.

Results: X chromosomal rearrangement (46,X,der(X)t(X;X)(p22.32;q27.1)) was detected. A terminal deletion involving SHOX, CSF2RA and ARSE genes established the Turner syndrome diagnosis. This type of change could lead to growth retardation, developmental delay, hypotonia, hypogonadism, ichthyosis, Kallmann syndrome, and skeletal abnormalities. In our case, the mother presented with short stature only, and the daughter had an atrial septal defect and hypotrophy. Variable expressivity was probably due to uneven X chromosome inactivation.

Conclusion: The identified terminal deletion prompted a Turner syndrome diagnosis. With a 50% likelihood of transmitting the derivative X chromosome, this case emphasizes the importance of comprehensive genetic evaluation for informed reproductive decision-making (prenatal invasive testing, preimplantation diagnostics) in high-risk pregnancies.

Grant References: -

Conflict of Interest: None declared

EP03.037 Prenatal molecular diagnosis WES vs CMA: What is the most cost-effective approach?

Marina Martinez-Matilla 1, Alba Velo1, Claudia Abellán1, Iryna Boyko1, Gemma Cartagena1, Carmen Fernández-Vizcaino1, Lova Matsa2, Sandra Garcia-Herrero1

1Igenomix. Part of Vitrolife Group, Paterna, Spain; 2Genetics Laboratory in Dubai, UAE - Igenomix, Dubai, United Arab Emirates

Background/Objectives: The chromosomal microarray analysis (CMA) testing is still more used than whole-exome sequencing (WES) as a second-line approach in pregnancies with ultrasound abnormalities. However, the prenatal diagnostic yield could be increased, and the approach could be more cost-effective by selecting WES instead of CMA, as WES allows the study of copy number variations (CNV) and single-nucleotide variations (SNV) simultaneously. Our objective was to assess which is the most cost-effective approach based on our experience.

Methods: Prenatal cases for WES from 2021 to 2023 were evaluated. WES was performed from fetal DNA using Nextera DNA Flex Pre-Enrichment Library Prep and Illumina Exome Panel and sequenced on the NovaSeq 6000 System (Illumina). Raw data were processed by the in-house bioinformatics pipeline. Detected variants were classified according to the recommendations of the American College of Medical Genetics (ACMG).

Results: A total of 53 prenatal cases were selected. Of them, 23 (43.4%) were positive, six (11.3%) non-conclusive, and 24 (45.3%) negative. Among the positive cases, in seven (30.4%) a pathogenic CNV was detected, while a pathogenic SNV was identified in 16 (69.6%) cases. All the CNV would be detected by CMA.

Conclusion: In our cohort, prenatal molecular diagnostic yield by WES technology was approximately of 43%, of which CMA would have missed approximately 70% (those related with SNV). Our results show that WES approach used as a second-line technology provides a more comprehensive diagnosis than CMA and is a more cost-effective approach, since provides CNV and SNV information in a single test.

Grants:

Conflict of Interest: None declared

EP04 Sensory Disorders (Eye, Ear, Pain)

EP04.001 Inherited cataracts and its genetic connection to glaucoma and other ocular phenotypes in Bulgarian patients

Kristiyana Vitanova 1, Kunka Kamenarova1, Nevyana Veleva2, Kalina Mihova1, Yoanna Kaneva2, Reni Tzveova3, Martin Georgiev1, Ivanka Dimova1, Alexander Oscar2, Radka Kaneva1

1Medical University of Sofia, Department of Medical Chemistry and Biochemistry, Sofia, Bulgaria; 2University Hospital “Aleksandrovska”, Department of Ophthalmology, Sofia, Bulgaria; 3UMHAT “Queen Joanna – ISUL”, Department of General and Clinical Pathology Department of Molecular Biology, Sofia, Bulgaria

Background/Objectives: Inherited cataracts are clinically and genetically heterogeneous cause of visual impairment at an early age. It can be associated with various ocular and systemic abnormalities. Early diagnosis and treatment are crucial for the visual prognosis. The purpose of this study was to identify the disease-causing mutations in Bulgarian patients with non-syndromic and syndromic forms of inherited cataract accompanied by glaucoma using next-generation sequencing of clinical exome (CES).

Methods: Six unrelated patients and their relatives were selected on the basis of diagnosis of cataracts. Genomic DNA of all probands was subjected to genetic analysis using clinical exome sequencing on MiSeq platform. Variants in selected genes associated with glaucoma, optic atrophy, anterior-segment dysgenesis, and inherited retinal degeneration, were extracted from the WES data and filtered using systematic bioinformatics analysis and the American College of Medical Genetics and Genomics criteria. Candidate variants were verified by Sanger sequencing and MLPA assay.

Results: Using CES, 7 potential pathogenic variants (PPVs) and 2 variants of uncertain significance were found in 9 genes, including TRPM3, MYH9, CRYAA, CLCN1, CACNA1S, COL11A1, TRPV4, CRYBB1, RP2. One patient expressing a mixed phenotype of cataracts and retinitis pigmentosa, carried a novel deletion in the RetNet-gene, RP2. Four novel variants, TRPM3- p.Val1002Met, MYH9- p.Asp25Asn, CRYAA - p.Arg116Leu, and CRYBB1- p.Arg132His, were found in non-syndromic cases presenting with congenital or nuclear cataracts, myopia, retinopathy, and glaucoma.

Conclusion: Determining the genetic cause may not only help to identify concomitant ocular and/or systemic disorders but also enable individualized genetic counselling.

Grants: KP-06-N33/12/18.12.2019, D01-285/17.12.2019, D01-395/18.12.2020

Conflict of Interest: None declared

EP04.002 Identification of low penetrance RB1 variants and carrier screening in two families

Poh San Lai 1, Grace Tan1, Chunping Liu1, thuanchong Quah1

1National University of Singapore, Paediatrics, Singapore, Singapore

Background: Retinoblastoma (RB) is a rare intraocular tumour caused by biallelic inactivation of RB1 gene. Although RB is a highly penetrant autosomal dominant disease, some families show low-penetrance phenotypes with reduced expressivity and incomplete penetrance of mutant RB1 alleles. In this study, we analysed two patients clinically diagnosed with unilateral and bilateral disease respectively.

Methods: Tumour and blood samples were sequenced using NGS and Sanger methods.

Results: The first proband carried a heterozygous splice mutation (NM_000321.3:c.607+1G > T) inherited from his asymptomatic father. He has a younger brother with unilateral RB harbouring the same variant. The disease-eye to carrier ratio (der) for this family is 0.66 (2/3) which is within the expected low penetrance values. The second proband inherited a germline missense variant (NM_000321.3:c.1981C>T:(p.Arg661Trp) from his unaffected father. His unaffected brother carried the same variant leading to a der = 0.66 in the family. Identification of low penetrance variants presents significance challenges in genetic counselling for both families. RB survivors are at risk of secondary malignancies while recently, there has been rare observation of asymptomatic RB1 mutation carriers developing primary osteosarcoma. In both families, highlighting the risk and implications of transmitting the low penetrance variants to future offsprings is important and life-long surveillance is indicated for asymptomatic carriers.

Conclusion: Patients with low penetrance mutations and asymptomatic members of their families remain at risk for malignancies. Genetic testing is important to identify carriers in families of patients with germline mutations as they could be asymptomatic but carry known or novel RB1 low-penetrance variants.

Grants: NMRC/0021/12

Conflict of Interest: None declared

EP04.003 the clinical and molecular Spectrum associated with isolated Wolfram Syndrome Type 1

Basamat AlMoallem 1

1College of Medicine, King Saud University, Department of Ophthalmology, Riyadh, Saudi Arabia

Consortium: none

Background/Objectives Wolfram Syndrome (WS) known as DIDMOAD syndrome (Diabetes Insipidus, Diabetes Mellitus, Optic Atrophy, and Deafness) (OMIM# 222300). It is the third most common hereditary optic neuropathy that comprises three major types. WS-Type 1, is the commonest form caused due to biallelic mutations in WFS1 with neuropathy constituting one of its major diagnostic criteria. Here we aim to characterize a 45-year-old gentleman complaining of gradual loss of vision at a near distance even with glasses and intolerance to bright light that started when he was 31 years old despite corrective LASIK surgery.

Methods Detailed ophthalmogical and systemic examinations followed by Whole exome sequencing (WES)

Results Examination showed a VA 20/400 OD and 20/200 OS. He scored 1/15 on the Ishihara color vision test OU. Slit lamp examination was unremarkable except for diffuse disc pallor and cupping of 0.8-0.9 in both eyes. Visual field testing and OCT showed a significant reduction in RNFL bilaterally. WES revealed a known pathogenic biallelic frameshift variant in the WFS1 gene: NM_001145853.1:c.2673A>C, (Arg859 Pro*). Systemic evaluation including hearing assessment and neurological examination were normal. Other laboratory tests including CBC, ESR, renal, and liver function panels were within normal limits, folate = 14.8 (normal), random glucose = 6.6 (normal).

Conclusion Our case represents the first report of a patient with isolated autosomal recessive Wolfram syndrome type 1 due to confirmed biallelic pathogenic mutations in the WFS gene with isolated optic nerve atrophy and glaucomatous cupping in the absence of other systemic features and expanded its mutational spectrum.

Grants None

Conflict of Interest: None declared

EP04.004 DYRK1A syndrome presenting with familial exudative vitreoretinopathy

Siying Lin 1;2, Ellie Hay3, Andrew Webster1;2, Omar Mahroo1;2, Robert Henderson1;2;4, Gavin Arno1;2

1National Institute of Health Research Biomedical Research Centre at Moorfields Eye Hospital and the UCL Institute of Ophthalmology, London, United Kingdom; 2UCL Institute of Ophthalmology, London, United Kingdom; 3Great Ormond Street Hospital, Department of Clinical Genetics, London, United Kingdom; 4Great Ormond Street Hospital, Department of Ophthalmology, London, United Kingdom

Background/Objectives: The DYRK1A gene encodes a proline-directed kinase crucial for central nervous system development, with haploinsufficiency resulting in DYRK1A-related intellectual disability syndrome. Ocular manifestations occur in two-thirds of affected individuals, primarily involving refractive error and strabismus. Retinal involvement is rare, with only isolated cases of retinal detachment (2 patients) and peripheral retinal avascularity (1 patient) described. This study reports two patients with familial exudative vitreoretinopathy (FEVR) associated with DYRK1A disease variants.

Methods: Whole genome sequencing (WGS) was performed for 2 FEVR patients, along with comprehensive ophthalmic and systemic evaluations.

Results: Patient 1 (3 years, female) presented at 3 weeks of age with microphthalmia, microcornea and total retinal detachment in the right eye, whilst the left eye was myopic with macular dragging. There was a history of intrauterine growth retardation, and she was later noted to have short stature, microcephaly with cerebellar hypoplasia, generalised seizures and global developmental delay (GDD).

Patient 2 (19 years, female) presented at age 6 with a fixed funnel retinal detachment in the left eye; the right eye was myopic with optic atrophy, peripheral retinal ischaemia and fibrosis. Her medical history included microcephaly with cerebellar anomalies, generalised seizures, GDD, and distinctive facial features.

WGS identified heterozygous de novo DYRK1A variants in both patients; a known pathogenic variant NM_001347721.2:c.1282C>T p.(Arg428Ter) in patient 1, and a rare, novel missense variant c.857T>C p.(Leu286Pro), predicted damaging by in silico algorithms, in patient 2.

Conclusion: These cases confirm vitreoretinal involvement in DYRK1A syndrome, and highlight FEVR as a useful diagnostic handle in individuals with neurodevelopmental disorders.

Grants: none

Conflict of Interest: None declared

EP04.005 Biallelic mutations of CEP290 as a molecular background of isolated retinal dystrophy

Tadeusz Kałużewski 1;2, Łukasz Kępczyński1;2, Karolina Matuszczyk2, Magdalena Markowska2, Jordan Sałamunia2, Bogdan Kałużewski2, Agnieszka Gach1

1Polish Mother’s Memorial Hospital Research Institute, Department of Genetics, Łódź; 2Genos Sp. z o.o., Laboratory of Medical Genetics, R&D Department, Łódź

Background: The CEP290 gene encodes a centrosomal protein involved in ciliary assembly and trafficking. Homozygous or compound heterozygous mutations are well-known molecular backgrounds of ciliopathies, e.g., Joubert syndrome type 5 (OMIM: #610188), Meckel syndrome type 4 (OMIM: #611134), Senior-Loken syndrome type 6 (OMIM: #610189) and Leber congenital amaurosis 10 (OMIM: #611755). The phenotype of CEP290-related disorders ranges from isolated retinal dystrophy to severe multisystemic involvement.

Methods: This report presents two affected brothers of Polish origin, children of non-consanguineous parents. Both patients have abnormal refraction (HP:0000539) and visual field defect (HP:0001123). They do not present any syndromic features of ciliopathies. The high-throughput molecular diagnosis was carried out to investigate the molecular background of their disease.

Results: Whole exome sequencing detected compound heterozygosity for CEP290 variants: c.1992del p.(Pro665Leufs*10) and c.104T>C p.(Val35Ala) in both affected brothers. Parental testing confirmed in trans arrangement. The c.1992del variant was reported in the literature in connection with severe manifestations of ciliopathies (PMID:17345604, PMID:16909394, and PMID:23559409), while the c.104T>C was not described until now. The c.1992del was classified as pathogenic, and c.104T>C as a variant of uncertain significance according to ACMG guidelines.

Conclusion: The phenotype of the patients is consistent with the clinical presentation of isolated inherited retinal disorders caused by the CEP290 mutations. Given the shortage of data regarding genotypic-phenotypic correlations of this gene defects, clinical and molecular findings described here expand the molecular and clinical spectrum of CEP290-related disorders.

Conflict of Interest: None declared

EP04.007 Genetic spectrum of 381 patients with hearing loss diagnosed with a comprehensive approach in a clinical setting

Raquel Muguerza 1;1, Nerea Bastida-Lertxundi1, julien crettaz1, Raquel Sáez-Villaverde1, yolanda ramírez1, jose luis eguiburu1, laura martinez1, irene perez1, carla lallave1, Zuriñe Martinez2, Marta Abrego2, Elena Beristain3, Blanca Gener Querol4

1Donostia University Hospital, Clinical Genetics, Donostia, Spain; 2Donostia University Hospital, Otorhinolaryngology Department, Donostia, Spain; 3Araba University Hospital, Molecular Genetics Laboratory, Gasteiz, Spain; 4Cruces University Hospital, Clinical Genetics, Barakaldo, Spain

Background/Objectives: Deafness, the most common sensorineural impairment, is genetic in 60% of cases. It can be syndromic (30%) or non-syndromic (70%), which is genetically highly heterogeneous. Genetic diagnosis has become an essential test in the diagnosis of deafness.

Methods: 381 patients (2014-2023) from the Basque Public Health System were studied in Donostia University Hospital with a comprehensive sequential approach: 1) screening of frequent genes: GJB2 (Sanger sequencing), GJB6 (MLPA kit P163, MRC Holland), CNV of STRC (digital droplet-PCR) and mitochondrial variants (SNaPshot); 2) if negative result in the screening: Clinical Exome Sequencing TruSight One (Illumina) (65 patients) or Clinical Exome Solution-v2 (159 patients) or -v3 (Sophia Genetics) (77 patients) and virtual panel filtering (89, 91 and 109 genes, respectively).

Results: The overall diagnostic rate was 43% (163/381): 21% (80/381) in the screening and 22% (83/381) in the expanded analysis. Thirty-one different genes were involved (72.4%, (118/163) autosomal recessive, 17.2% (28/163) dominant, 9.2% (15/163) mitochondrial and 1.2% (2/163) X-linked). The more frequent autosomal recessive genes were: GJB2 (44.1%, 52/118), STRC (12.7%, 15/118), OTOF (6.8%, 8/118) and BSND (5.9%, 7/118), and among the autosomal dominant: TECTA (14.3%, 4/28), ACTG1 and WFS1 (10.7%, 3/28 each).

Conclusion: These results confirm the great genetic variability of deafness and suggest there are still many genes to be discovered. The great importance of the STRC gene and of the mitochondrial inheritance in our environment is striking, greater than that described in the literature. Moreover, the OTOF cases could be candidates for gene therapy in the near future.

Conflict of Interest: None declared

EP04.008 Genetic etiology of perrault syndrome in Iranian families: first report from Iran

Ebrahim Shokouhian 1, Zohreh Fattahi1, Sanaz Arzhangi1, Kimia Kahrizi1, Hossein Najmabadi1, mojgan babanejad1

1University of Social Welfare and Rehabilitation Sciences, Genetics Research Center, Tehran, Iran

Background/Objectives: Perrault syndrome (PS) is an extremely rare autosomal recessive disorder characterized primarily by bilateral sensorineural hearing loss in both genders, and primary, or secondary ovarian failure in females. Moreover, neurological features, such as cerebral ataxia, peripheral neuropathy, epilepsy and intellectual disability are frequent manifestations of PS as well. So far, six genes have been reported to cause PS, and globally almost 100 families have been identified with this syndrome.

Methods: Here we report the first cases of Perrault syndrome from two Iranian families, with novel and reported variants in CLPP and TWNK genes, that was identified by Whole Exome Sequencing (WES).

Results: Family A, a non-consanguineous marriage, consisted of three affected individuals presented with bilateral severe to profound congenital hearing loss, cerebral ataxia, epilepsy, and intellectual disability. In Family B, with a consanguineous marriage and one affected female presented with bilateral moderate to severe hearing loss, and peripheral neuropathy. WES revealed compound heterozygous variants for the proband of Family A; a previously reported c.21delA, and a novel missense variant (c.512C>G) in the CLPP gene. In Family B, a previously reported homozygous mutation, c.874C>A, was identified in the TWNK gene.

Conclusion: This is the first report of genetic variations of CLPP and TWNK genes in affected patients with PS from Iran. We expect that our detailed clinical study will improve the genotype-phenotype interpretation of causal PS variants and the analysis of next-generation sequencing data, leading to clarification of the cause of complicated cases of rare diseases.

Grants: USWR/GRC/2020-T-2405

Conflict of Interest: None declared

EP04.009 A novel candidate gene -MACF1- associated with autosomal dominant non-syndromic hearing loss in an Iranian family

Niloofar Bazazzadegan 1, Kimia Kahrizi1, Hossein Najmabadi1, Kevin TA. Booth2;3, Sussan Banihashemi1, Sanaz Arzhangi1

1Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran; 2Department of Medical and Molecular Genetics, Indiana School of Medicine, Indianapolis, IN, United States; 3Department of Otolaryngology—Head and Neck Surgery, Indiana School of Medicine, Indianapolis, IN, United States

Background: Cytoskeletal dynamics, the interplay of actin and microtubules is a tightly regulated process. Defects in the proteins involved can result in a wide range of cellular consequences. Hearing loss exhibits genetic and phenotypic heterogeneity. Among 140 genes casually linked to non-syndromic hearing loss, 63 are associated with autosomal dominant inheritance. Here we add to this growing number by implicating MACF1 (reported with lissencephaly with MIM # 618325), as a novel candidate gene for autosomal dominant non-syndromic hearing loss (ADNSHL).

Methods: A large Iranian family segregating progressive ADNSHL (without any cognitive or brainstem malformation) was recruited for this study. Affected and unaffected individuals underwent clinical evaluation and after informed consent peripheral blood samples were collected. Following DNA isolation, exome sequencing (ES) was performed on proband. After read mapping and variant calling, variants were annotated with a custom bioinformatic pipeline. Variants were filtered based on quality, minor allele frequency and predicted variant impact. Subsequently, variants were further filtered and prioritized. Sanger sequencing was utilized for segregation analysis of all candidate variants in available samples.

Results: The proband had bilateral mild-moderate sensorineural hearing loss and was negative for GJB2 mutations. After applying ES on family proband, a missense mutation c.C1378T (p .H460Y) was found in a novel candidate gene, MACF1 and co-segregated with the hearing loss in the extended family.

Conclusion: We speculated that MACF1 mutations probably cause non-syndromic hearing loss inherited in an autosomal dominant manner. The potential functional impact of the identified variant will be investigated through further analysis.

Grant references: USWR: 2875

Conflict of Interest: None declared

EP04.011Missense variant in COL18A1 causing Knobloch syndrome

Laura Mauring 1;2;3, Tiia Reimand1;3, Ülle Murumets1, Sander Pajusalu1;3, Mari Petraudze2, Kuldar Kaljurand2;3, Katrin Ounap1;3

1Tartu University Hospital, Genetics and Personalized Medicine Clinic, Tartu, Estonia; 2Tartu University Hospital, Eye Clinic, Tartu, Estonia; 3University of Tartu, Institute of Clinical Medicine, Tartu, Estonia

Missense variant in COL18A1 causing Knobloch syndrome

Background/Objectives: Knobloch syndrome (OMIM # 267750) is a rare recessively inherited condition with significant phenotypic heterogeneity characterized by high myopia, vitreoretinal degeneration, and variable occipital skull abnormalities, with or without neurodevelopmental delay. Knobloch syndrome is caused by pathogenic variants in the COL18A1, which results in the aberrant synthesis of type XVIII collagen.

Case report: A 9-year-old boy underwent genetic consultation due to low vision, high axial myopia since early childhood, and bilateral retinal detachment. He is the second child of a healthy couple, born after uneventful pregnancy at term, with unremarkable early development and no relevant family history. The fundus demonstrates signs characteristic to high myopia, and undifferentiated fovea bilaterally, but no anterior segment defects. No occipital abnormalities are evident or palpable. He has neither cutis aplasia nor epilepsy.

Results: Next-generation sequencing of a targeted gene panel from the DNA extracted from the blood identified two heterozygous variants in trans in COL18A1: NM_001379500.1(COL18A1):c.2673dup, p.(Gly892Argfs*9) (pathogenic), and c.2443G>A, p.(Gly815Arg) (variant of uncertain clinical significance (VUS)). This variant has never been reported and is ultra-rare from the gnomAD database. Furthermore, all computational prediction tools support a deleterious effect on the gene of this variant. It is also an evolutionarily highly conserved nucleotide.

Conclusion: This case emphasizes the need for more functional data about the missense variants found in COL18A1. According to ClinVar, 1030 different missense variants have been identified, with all but one classed as VUS/benign. We are seeking collaborators to further investigate its functional effects.

Grants: PRG471, PRG2040, PSG774

Conflict of Interest: Laura Mauring: None declared, Tiia Reimand: None declared, Ülle Murumets: None declared, Sander Pajusalu PSG774, Mari Petraudze: None declared, Kuldar Kaljurand: None declared, Katrin Ounap PRG471, PRG2040

EP04.012 Genetic test results of patients clinically diagnosed with Stargardt disease: Novel ABCA4 variants from Turkey

Fulya Yaylacioglu Tuncay 1, burak acar2, murat yüksel2, Gülsüm Kayhan3, mehmet ali ergün3, Sengül Ozdek2

1University of Health Sciences, Gülhane Medical Faculty, Medical Biology, Ankara, Türkyie; 2Gazi University, School of Medicine, Ophthalmology, Ankara, Türkyie; 3Gazi University, School of Medicine, Medical Genetics, Ankara, Türkyie

Background/Objectives: This study aims to contribute to the disease-related variant data from Turkey by evaluating the genetic test results of patients clinically diagnosed with Stargardt disease.

Methods: The genetic test results of 42 patients diagnosed with Stargardt disease in the ophthalmology department of Gazi Medical Faculty were retrospectively evaluated. The genetic testing methods, reported gene variants, and variant classifications were recorded. The disease-related variants were investigated in the Human Gene Mutation Database (HGMD), ClinVar and literature to decide whether the variant was novel.

Results: Retina panel (n = 23) or clinical exome panel (n = 19) was applied as the genetic testing methods in patients. Twenty-nine ABCA4 variants were reported in 42 unrelated patients. Genetic test results supported the clinical diagnosis in 31 patients. The most frequently detected variant type was missense variants (16/29). Three variants were not available in the HGMD and ClinVar databases and were not reported in the literature: c.466_467dupAT, p.Leu157SerTer2; c.4540-1G > C; c.878delC, p.Met293SerfsTer7. Although seven variants were in the ClinVAR database, their association with a retinal disease was not mentioned in the literature and databases: c.4529C>T, p.Pro1510Leu; c.6121G>A, p.Gly2041Ser; c.6568C>T, p.Gln2190Ter; c.1916A>G, p.Tyr639Cys; c.6568C>T, p.Gln2190Ter; c.5018+81C>T; c.303-181G>A. c.5882G>A, p. Gly1961Glu was detected in 7 patients in this study and was the most frequently recurring variant similar to different patient populations.

Conclusion: This study is the most extensive series providing variant data on Stargardt disease from Turkey. Newly detected variants contribute to the variant spectrum of ABCA4 in Stargardt disease.

Conflict of Interest: None declared

EP04.013 Whole-exome sequencing revealed a novel candidate gene, ARHGAP22, as potential cause of non-syndromic hearing loss in an Iranian family

Raziye Rezvani Rezvandeh 1, negar kazemi1, Farzane Zare Ashrafi1, Masoud Edizadeh2, fatemeh ghodratpour1, Fatemeh Keshavarzi1, Khadijeh Jalalvand1, Sanaz Arzhangi1, Kimia Kahrizi1, Hossein Najmabadi1, Marzieh Mohseni1

1Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran; 2Genoks Genetic Diagnosis Center, Department of Bioinformatics, Ankara, Türkyie

Background: Non-syndromic hearing loss (NSHL) affects one out of every 500 newborns worldwide. In Iran, it is the second most frequent disability due to the high rate of consanguineous marriages.

Methods: Whole exome sequencing (WES) was performed to identify the disease-causing gene in a consanguineous family with hereditary bilateral severe sensorineural NSHL in three affected members.

Results: WES revealed a homozygous missense variant c.1873G>A in the ARHGAP22 gene (NM_021226.4) for the patients. Co-segregation analysis revealed that all affected individuals were homozygous for this variant with the heterozygous carrier parents.

Conclusion: The c.1873G>A variant located in the Coiled Coiled structure at position 625 that substitutes lysine for glutamic acid. The protein that is expressed by ARHGAP22 is found in the Rho GTPase protein cluster and interacts with proteins that lead to hearing loss; such as CDC42, JAG1, ROCK2, and DIAPH3. The bioinformatics databases indicate that ARHGAP22 is expressed in the ear. We propose that the ARHGAP22 gene could be a candidate as a new hereditary hearing loss gene. Further studies need to confirm the effect of ARHGAP22 on the auditory system.

Grant number: USWR/GRC/2024 /3072

Institutional ethical approval number: IR.USWR.REC.1402.187

Conflict of Interest: None declared

EP04.014 DBX2: A novel candidate gene for non-syndromic hearing loss identified in an iranian family

negar kazemi 1, Raziye Rezvani Rezvandeh1, Farzane Zare Ashrafi1, fatemeh ghodratpour1, fatemeh keshavarzi1, Khadijeh Jalalvand1, Sanaz Arzhangi1, Masoud Edizadeh2, Kimia Kahrizi1, Hossein Najmabadi1, Marzieh Mohseni1

1Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran; 2Genoks Genetic Diagnosis Center, Department of Bioinformatics, Ankara, Türkyie

Background: More than 70 percent of pathogenic hearing loss genes are involved in autosomal recessive non-syndromic hearing loss (ARNSHL). In Iran, due to the high rate of consanguineous marriage, hearing loss is the second most common neurosensory disorder, affecting, approximately 1 in 166 individuals.

Methods: In this study, after excluding GJB2 and OtoSCOPE mutations, we performed Whole Exome Sequencing (WES) of two Iranian siblings born to a consanguineous family, with late-onset moderate to severe sensorineural hearing loss.

Results: We identified a homozygous missense variant (c.28G>A (p.Ala10Thr)) in the DBX2 gene (NM_001004329.3) as a novel candidate gene.

Conclusion: DBX2, known as developing Brain Homeobox 2, possesses DNA-binding transcription factor activity, and RNA polymerase II-specific. This gene interacts with LS1, LMX1A, and LMX1B in auditory pathways. Additionally, it is a crucial component of the superior olivary complex, responsible for sound localization. The c.28G>A (p.Ala10Thr) variant leads to the formation of a larger and more hydrophobic amino acid potentially altering the protein structure and its hydrophobic interactions. Further investigation of the DBX2 gene may contribute to our understanding of the genetic basis of auditory function.

Grant number: USWR/GRC/2024 /3072

Institutional ethical approval number: IR.USWR.REC.1402.187

Conflict of Interest: None declared

EP04.015 Comprehensive Molecular Diagnosis of Inherited Retinal Diseases: Use of an Extended Virtual NGS Panel for Enhanced Detection of Genetic Variants

Ornella Rondinone 1, Giada Moresco1, Carola Conca Dioguardi2, Martina Miceli2, Rita Gorgoglione2, Chiara Pesenti2, Giovanni Marfia3;4, Stefania Navone3, Gianluca Tolva2, Leonardo Colombo5, Luca Rossetti5;6, Monica Miozzo1;2, Laura Fontana1;2

1University of Milan, Medical Genetics, Department of Health Sciences, Milan, Italy; 2ASST Santi Paolo e Carlo Hospital, Medical Genetics Unit, Milan, Italy; 3Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Milan, Italy; 4Aeronautica Militare, Clinical Pathology Unit, Istituto di Medicina Aerospaziale “A. Mosso”, Milan, Italy; 5ASST Santi Paolo e Carlo Hospital, Department of Ophthalmology, Milan, Italy; 6University of Milan, Department of Health Sciences, Milan, Italy

Background/Objectives: Inherited retinal diseases (IRDs) are a clinically and genetically heterogeneous group of disorders characterized by photoreceptor degeneration. Currently, over 300 genes have been identified to cause both non-syndromic and syndromic retinal degeneration, and this number is increasing. Molecular diagnosis is fundamental for clinical management, prognostic assessment, and treatment prospects, as gene therapy is available for some types of IRDs.

Methods: We developed a comprehensive in-house extended virtual NGS panel that includes both SNV/indels and CNV in coding and specific non-coding regions of 332 IRDs-genes. Using eVai software (enGenome), we analyzed a cohort of 52 patients affected by rod-cone and other retinal dystrophies, suspected of having a genetic etiology. Some of these patients has previously undergone genetic testing, yielding either negative or inconclusive results.

Results: Our extended approach enabled conclusive diagnoses in 31 patients, achieving a diagnostic rate of 60%. We identified 35 variants in total: 2 CNVs (deletions), 17 missense, 10 frameshift/nonsense, 7 splicing, and 2 intronic variants. Of these, 23 variants are classified as pathogenic/likely pathogenic (P/LP), 2 as VUS/LP, and 10 as VUS strongly correlating with phenotype. The most frequently mutated genes were USH2A (16% of cases) and EYS (10% of cases). In addition, this in-house panel facilitated the diagnosis of tricky cases, including the identification of a deep intronic variant in USH2A and the re-diagnosis of Pseudoxanthoma elasticum in a patient with biallelic ABCC6 variants.

Conclusion: Our study supports the use of a comprehensive diagnostic approach, extending also to non-coding regions, to achieve an accurate molecular diagnosis of IRDs.

Grants: None.

Conflict of Interest: None declared

EP04.016 A novel frameshift variant in compound heterozygous state in the USH2A gene associated with Usher syndrome

Mert Coşkun 1, Mehmet Erkan Dogan2, Alp Peker1, Aslı Toylu1, Özden Altıok Clark1

1Akdeniz Üniversitesi Hastanesi, Medical Genetics, Antalya, Türkyie; 2Akdeniz Üniversitesi Hastanesi, Ophthalmology Department, Antalya, Türkyie

Background/Objectives: Usher syndrome (USH) is an autosomal recessive disorder characterized by bilateral sensorineural hearing loss and progressive retinitis pigmentosa. USH2A, one of the causative genes of this syndrome, codes a protein named Usherin which is important for the functions of photoreceptor cells and cochlear hair cells. In this study, we present a patient with clinical findings of USH. She had congenital hearing loss and experienced progressive visual impairment beginning in the second decade. We aimed to evaluate the sequence variants of the USH associated genes in order to confirm the patient’s diagnosis.

Methods: Genomic DNA was extracted from the patient’s peripheral blood sample. Exons and exon-intron transition regions of the USH associated genes were investigated by next generation sequencing. Variants were analyzed using Sophia DDM program. Variant information servers dbSNP, ClinVar, Ensembl and ACMG 2015 criteria were used for evaluations.

Results: In USH2A gene, c.2845dup (p.Leu949Profs*12) frameshift and c.4628-2A > T splice site variants were detected in compound heterozygous state. The splice site variant was reported as a “pathogenic” change in ClinVar database. The frameshift variant which is predicted to cause premature termination of protein translation was not reported in population or disease-specific databases.

Conclusion: The detected novel frameshift mutation is evaluated as a candidate variant that may be associated with USH, due to its predicted functional effects and presence with a known pathogenic variant. To better understand genotype-phenotype correlation, segregation of the variant in the patient’s family and larger population data will be examined.

Grants: None

Conflict of Interest: None declared

EP04.017 Diagnosis of Smith-Magenis syndrome in an adult patient with rapidly progressive profound hearing loss

Vanessa Hochheimer 1, Zafer Yüksel1, Hanno Bolz1

1Bioscientia Institute for Medical Diagnostics GmbH, Bioscientia Human Genetics, Ingelheim, Germany

Background/Objectives: Smith-Magenis syndrome (SMS) is an autosomal dominant multisystem disorder initially found to be caused by microdeletions on chromosome 17p11.2. Since then, whole-exome sequencing (WES) has revealed an increasing number of point mutations in RAI1, the disease-causing gene contained in the SMS deletion locus. Symptoms include typical facial features, intellectual disability, sleep disturbances and behavioral problems; diagnosis is usually given in childhood. Hearing loss (HL) occurs in about 80%, as conductive HL in childhood and sensorineural HL in adulthood. It is progressive and usually mild.

We report a 27-year-old male for whom parents reported lip reading since early infancy, and hearing aids were provided in the 1st decade for his mild-to-moderate HL. Progression to profound HL required bilateral cochlear implantation (CI) at 20 years. Previous genetic testing for Usher syndrome, optic atrophy and hearing loss because of night blindness and visual field constriction revealed a heterozygous pathogenic USH2A variant.

Methods: Trio-WES on DNA from peripheral blood samples (patient, parents) was conducted on an Illumina NovaSeq 6000 system.

Results: Medical history and physical examination revealed moderate developmental delay, hypotonia, bilateral iris coloboma, brachycephaly, brachydactyly, and facial dysmorphism. The cause of the visual complaints remained elusive (ophthalmological assessment did not confirm retinal dystrophy). We identified a novel heterozygous truncating de novo c.106_116del p.(Ala36Leufs*13) variant in RAI1. Reverse phenotyping revealed onychotillomania and heavy sleep disturbance.

Conclusion: SMS may atypically present with predominant progressive HL requiring bilateral CI and should thus be considered in genetic analysis for profound sensorineural HL.

Conflict of Interest: None declared

EP04.019 Double trouble, incidental finding or clinical irrelevant – the interpretation challenge

Sinje Geuer 1, Cord-Christian Becker1

1MVZ genetikum GmbH, Neu-Ulm, Germany

Background: We report on a 4-year-old female child with hearing impairment without any signs of syndromic involvement. The family history is empty for early onset hearing impairment.

Methods: In-silico panel analysis for syndromic and non-syndromic hearing impairment based on whole exome sequencing (TWES, Twist Human Core Exome with additional customized probes and NGS with NovaSeq Illumina and deep sequencing of mitochondrial DNA) was performed.

Results: Panel-analysis revealed potentially relevant variants in three genes. A pathogenic variant and a variant of unclear significance (VUS) in MYO15A might be causative for autosomal recessive non-syndromic hearing loss if present in compound- heterozygosity. A heterozygous VUS in WFS1 could be causative for autosomal dominant non-classical Wolfram syndrome if arisen de-novo. A pathogenic variant in mt-TL1 with low heteroplasmy of 7% and known to cause MELAS-Syndrome might cause syndromic or (rarely) non-syndromic hearing impairment. Noteworthy, we cannot exclude that MELAS will never manifest in our patient. The relevance of the variants detected might be partly resolvable by segregation analysis. Nevertheless, the MELAS-variant presents an interpretation challenge due to its extremely low heteroplasmy and tissue specific variability.

Conclusion: This case report highlights the challenge to interpret multiple variants of which each might explain the patient phenotype while certain syndromic characteristics are not present (yet), and leaves us with the question: Is this double trouble or an incidental finding? Technological improvement leads to a third option: A known pathogenic variant might be clinical irrelevant due to its low allele frequency.

Conflict of Interest: None declared

EP04.020 De novo variants in PDE6B retinitis pigmentosa

Luigi Monti 1;2, Elena Luppi1;2, Nicole Balducci3, Giovanni Innella1;2, Francesca Montanari1, Cesare Rossi1, Daniela Turchetti1;2, Marco Seri1;2

1Medical Genetics Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy, Bologna, Italy; 2Alma Mater Studiorum University of Bologna, Bologna, Italy, Department of Medical and Surgical Sciences, Bologna, Italy, Bologna, Italy; 3Ophtalmology Unit, S.Orsola-Malpighi Hospital, University of Bologna, Department of Medical and Surgical Sciences, Bologna, Italy, Bologna, Italy

Background/Objectives: PDE6B pathogenic variants are associated with autosomal recessive Retinitis Pigmentosa (RP) and occur in a small proportion of affected patients (2-5%). In this study, we aim to expand the clinical and mutational spectrum of PDE6B-associated RP by presenting two new unrelated cases and investigating the occurrence of novel variants in the gene.

Methods: Patients’ histories were recorded by medical charts. Both patients underwent ophthalmic examinations and a hereditary eye disease NGS panel analysis. Variant validation and segregation analysis were performed through Sanger-sequencing.

Results: Both patients, a 42-year-old woman and a 13 years-old boy, reported the onset of symptoms in childhood/adolescence (narrow visual field and hemeralopia) and share preserved visual acuity, extrafoveal bone spicules at the ocular fundus, atrophy of the bilateral extrafoveal photoreceptors and intraretinal cysts at OCT. Both resulted carriers of a known pathogenic variant inherited from a parent and a novel variant (c.928-1G > A;p.? and c.340C>T;p.Gln114Ter) of de novo origin respectively on the maternal and the paternal allele (false paternity was excluded).

Conclusion: PDE6B-associated disease is a classic RP with relatively preserved central vision at older ages. Intraretinic cysts are common findings. Our identification of two novel mutations in PDE6B increases the spectrum of genetic variations. Their de novo origin may not be coincidental, suggesting a more common finding than previously thought, and could explain the high prevalence of novel variants in PDE6B (up to 43% in some studies). Further research will be needed to understand the mechanism underlying this phenomenon.

Grants:

Conflict of Interest: None declared

EP04.021 Natural history of DFNA9 (COCH) in a six-generation family from North America segregating the Belgian founder mutation (c.151C>T; p.P51P/S)

Tammy Benteau 1, Maxime Maheu2, David McComiskey3, Anne Griffin1, Kim Furlong4, Susan Stanton5, Terry-Lynn Young1

1Memorial University, Faculty of Medicine, St. John’s, Canada; 2Universite’ de Montreal, Ecole d’orthophonie d’audiologie, Montreal, Canada; 3University of Ottawa, Neuroradiology, Ottawa, Canada; 4NL Balance and Dizziness Centre, St. John’s, Canada; 5Western University, National Centre of Audiology, London, Canada

Background/Objectives: DFNA9 hearing loss (HL) due to COCH (c.151C>T; p.P51P/S) also causes severe vestibular dysfunction and an increased risk of dementia. Potential treatment targets anti-sense oligonucleotides (AONs) to silence the mutant allele. Success, in part, depends on the identification of SNPs that increase efficacy and a deeper understanding of the full natural history to identify the precise therapeutic window. This study describes the DFNA9 haplotype and natural history in a multigenerational North American family.

Methods: We identified a six-generation family from the genetic isolate of Newfoundland, Canada with late onset SNHL segregating as an AD trait due to COCH (c.151C>T). To determine if this is the Belgian founder allele, microsatellite markers spanning DFNA9 were compared with 3 Dutch mutation carriers. Phenotypic investigations on mutation carriers will be presented, including nature of HL and vestibular disturbance.

Results: Genotyping revealed this family segregates the Belgian founder mutation, based on a shared 1.2 MB haplotype. The proband experienced bilateral HL (sloping to moderate loss at high frequencies) that worsened over a 15-year period and was treated with a cochlear implant (CI). Although CT scan at age 40 detected no vestibular abnormalities, by age 56, the proband required supports for daily activities.

Conclusion: Extended families with founder mutations are particularly useful for capturing the natural history of adult-onset HL and are a valuable source of pre-symptomatic carriers for prospective studies required for targeted gene therapies.

Grants: ACOA (Atlantic Canada Opportunities Agency); CIHR (Canadian Institutes for Health Research); CFI (Canadian Foundation for Innovation).

Conflict of Interest: Tammy Benteau I am employed as a Research Assistant with the senior author, Maxime Maheu: None declared, David McComiskey: None declared, Anne Griffin: None declared, Kim Furlong: None declared, Susan Stanton: None declared, Terry-Lynn Young: None declared

EP04.022 GLRA2-Associated X-linked dominant high myopia in a multigenerational family

Neta Feinstein-Goren 1;2, Dalia Eli3, Daphna Prat2;4;5, Ortal Barel Barel6, Odelia Chorin2;7, Shelly Lev-Hochberg2;7, Abraham Spierer2;4, Guy Ben-Simon2;4;5, annick REIN ROTHSCHILD2;7, Ben Pode-Shakked2;5;7

1Sheba Medical center, Tel-Hashomer, The Danek Gertner Institute of Human Genetics, Ramat Gan, Israel; 2Tel Aviv University, Faculty of Medicine, Tel Aviv, Israel; 3Sheba Medical center, Tel-Hashomer, The Opthalmo-Genetics Clinic, Department of Ophthalmology, Goldschleger Eye Institute, Ramat Gan, Israel; 4Sheba Medical Center, Tel-Hashomer, Department of Ophthalmology, Goldschleger Eye Institute, Ramat Gan, Israel; 5Sheba Medical center, Tel-Hashomer, The Talpiot Medical Leadership Program, Ramat Gan, Israel; 6Sheba Medical center, Tel-Hashomer, The Genomic Unit, Sheba Cancer Research Center, Ramat Gan, Israel; 7Edmond and Lily Safra Children’s Hospital, Sheba Medical center, Tel-Hashomer, The Institute for Rare Diseases, Ramat Gan, Israel

Background: High myopia is a complex ocular condition influenced by both genetic and environmental factors. The global rise in myopia has positioned it as a significant public health concern. Understanding its genetic basis is crucial for unraveling its etiology. To date, over 100 genes have been associated with myopia (with only 17 considered causal). We sought to investigate the molecular basis of isolated high myopia affecting multiple individuals across four generations of a non-consanguineous Moroccan-Jewish family, with an X-linked dominant inheritance pattern.

Methods: Targeted gene panel sequencing was conducted on a multigenerational family with isolated high myopia. Comprehensive clinical ophthalmic examinations were performed to characterize the myopic phenotype.

Results: Our investigation revealed a pathogenic variant in the GLRA2 gene, implicating its involvement with isolated high myopia within the family. Notably, GLRA2 was previously associated with a neurodevelopmental phenotype. In addition to our findings, only recently (February 2023) was GLRA2 identified as a player in the genetic landscape of isolated high myopia in two large Chinese families and a simplex case, collectively emphasizing the significance of this gene in the manifestation of high myopia across diverse populations.

Conclusion: This study advances our understanding of GLRA2-related high myopia, underscoring its distinctiveness from the syndromic neurodevelopmental phenotype previously associated with this gene. Our findings advocate for the inclusion of GLRA2 in the differential diagnosis of isolated high myopia cases, marking a step toward unravelling the complex genetic landscape of high myopia, enhancing diagnostic precision, prognosis accuracy, and holding promise for targeted therapeutic interventions.

Conflict of Interest: None declared

EP04.023 Characterization of sensorineural hearing loss in Finland reveals enrichment of pathogenic founder variants

Tyrni Pykäläinen1;2, Minna Kraatari-Tiri1;2, Pia Pohjola3, Sanna Häkli4, Elisa Rahikkala 1;2

1University of Oulu, Research Unit of Clinical Medicine and Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland; 2Oulu University Hospital, Dept of Clinical Genetics, Oulu, Finland; 3Turku University Hospital, Dept of Genomics, Turku, Finland; 4Oulu University Hospital, Dept of Otorhinolaryngology and Phoniatrics, Oulu, Finland

Background/Objectives: To examine the clinical and genetic characteristics of childhood-onset bilateral sensorineural hearing loss (SNHL) in Finland.

Methods: A retrospective cohort study was conducted in the Oulu University Hospital from 2017 to 2022. Inclusion criteria were children younger than 18 years with diagnosed bilateral SNHL (ICD-10: H90.3).

Results: The cohort consisted of 249 children. Pathogenic or likely pathogenic sequence variants explaining the SNHL were identified in 34 % (N = 84/249) of children, including 60 non-syndromic SNHL patients (71 %, N = 60/84) and 24 syndromic (29 % N = 24/84) SNHL patients. Biallelic pathogenic variants in GJB2 were the most common etiology, explaining 16 % of SNHL in this cohort (N = 40/249). The most common hearing loss syndrome in this study was Usher type 3 (N = 5). Seventeen (N = 17/249, 7 %) children had chromosome abnormalities or copy number variants explaining their SNHL, including deletion of STRC-CATSPER2 genes (N = 4), other deletion (N = < 3), Down syndrome (N = 6), Turner syndrome (N = 4), or duplication (N = < 3). Pathogenic or likely pathogenic variants enriched in the Finnish population were identified in TMC1, CABP2, MYO7A, TWNK, SUCLA2, and CLRN1 genes.

Conclusion: SNHL is genetically and clinically heterogeneous. The most common causative gene was GJB2. Hearing loss syndromes explained 29 % of all SNHL caused by pathogenic or likely pathogenic sequence variants. The Finnish-enriched variants comprised of 23 % (N = 19/84) of all likely causative variants.

Grants: Research Council of Finland (grant number 338446), Uolevi Mäki Foundation, State Subsidy of Oulu University Hospital.

Conflict of Interest: None declared

EP04.024 Relationship between physical and psychological functioning and health-related quality of life in congenital aniridia

Erlend Christoffer Sommer Landsend1, Charlotte von der Lippe 2, Line Merete Mediå3, Jeanette Ullmann Miller3, Knut Erik Berge4, Solrun Sigurdardottir3

1Oslo University Hospital, Department of Ophthalmology, Oslo, Norway; 2Telemark Hospital Trust, Department of Medical Genetics, Skien, Norway; 3Oslo University Hospital, Centre for Rare Disorders, Oslo, Norway; 4Oslo University Hospital, Department of Medical Genetics, Oslo, Norway

Background: Congenital aniridia is a serious eye disease characterized by absence of iris to various degrees. Genetic causes may be pathogenic variants in PAX6 or in regulatory regions of PAX6. We will describe health-related quality of life (HRQoL) in adults with PAX6-related aniridia and assess the relationships between HRQoL, psychological status, ocular health and obesity.

Methods: Twenty-nine adults with PAX6-related aniridia (48% male, aged 18-79 years) participated. HRQoL was measured with SF-36 and the EQ visual analogue scale (VAS). Symptoms of anxiety and depression were measured using the Hospital Anxiety and Depression Scale (HADS). Obesity was assessed with the Patient-Reported Outcomes in Obesity (PROS). Sociodemographic characteristics, genetic variants and ocular and medical health variables were also analysed.

Results: The participants scored significantly lower in the general health domain of the SF-36 than the general population (65.2 vs. 75.3, p = 0.017). The EQ VAS score was also lower in the aniridia group (64.9 vs. 77.9, p = 0.021). Low PCS score was correlated with presence of ocular pain (p = 0.019), high HADS score (p = 0.017) and high PROS score (p = 0.009). High HADS and PROS scores were both related to low EQ VAS scores.

Conclusion: Adults with PAX6-related aniridia scored worse on certain measures of HRQoL than the general population. Poorer HRQoL was associated with increased symptoms of anxiety, depression and obesity and with presence of ocular pain.

Reference: Published in PMID: 38131258 https://doi.org/10.1111/aos.16615

Conflict of Interest: Erlend Christoffer Sommer Landsend Landsend is member of the scientific committee of the patient organization Aniridia Norway., The work was funded by the National Advisory Unit on Rare Disorders in Norway. The funding organization had no role in the design or conduct of this research., Charlotte von der Lippe: None declared, Line Merete Mediå: None declared, Jeanette Ullmann Miller: None declared, Knut Erik Berge: None declared, Solrun Sigurdardottir: None declared

EP04.025 Optimisation of the custom IRD NGS panel - a further step in the improvement of a diagnostic tool

Ewa Matczyńska 1;2, Marta Beć-Gajowniczek1, Larysa Sivitskaya1, Elżbieta Gregorczyk1, Ewa Suchecka1, Przemysław Łyszkiewicz1, Robert Szymańczak1, Maria Jędrzejowska1, Sławomir Teper2, Maciej Krawczyński3, Anna Boguszewska-Chachulska1

1Genomed S.A., Warsaw, Poland; 2Medical University of Silesia in Katowice, Chair and Clinical Division of Ophtalmology, Katowice, Poland; 3Center of Medical Genetics Genesis, Poznań, Poland

We present data on the genetic diagnosis of inherited retinal dystrophies (IRD), including the diagnostic yield, resulting from the panel design improvement, as well as bioinformatic pipeline development.

NGS analysis based on an annualy optimised and expanded Twist Bioscience custom panel (v3), targeting coding regions of 360 IRD genes, along with over 400 selected deep intronic variants and mtDNA sequence, was applied to identify genetic causes of IRD for 378 Polish patients. Bioinformatics involved the GATK(v4) best practice pipeline. CNV analysis was conducted using GATK GermlineCNVCaller. The variant classification was performed according to ACMG/AMP guidelines with the help of the in-house interpretation tool BroVar(v3). mtDNA analysis was routinely performed and included into the standard diagnostic pipeline.

Positive results were provided for 243 patients (64.29%). In 8 cases, this was possible due to the analysis of deep intronic regions. An increase in mean coverage (247x, SD 37) and uniformity improved CNV analysis resulting in 13 patients with the diagnosis confirmation based on CNVs. Coverage in the difficult RPGR ORF15 region was further improved with additional probe tiling. Pathogenic variants in the RPGR gene were identified in 25(6.6%) patients, 13 of them were uncovered in the ORF15 region.

The described changes in panel construction, in particular the inclusion of an updated list of deep intronic variants, together with improvements in the sensitivity of CNV analysis, should provide a higher diagnostic yield than current IRD diagnostics based on standard exome enrichments.

Partially funded by Ministry of Education and Science, applied PhD project PCTT/1874/2019.

Conflict of Interest: Ewa Matczyńska Full time employment in Genomed S.A., Marta Beć-Gajowniczek Full time employment in Genomed S.A., Larysa Sivitskaya Full time employment in Genomed S.A., Elżbieta Gregorczyk Full time employment in Genomed S.A., Ewa Suchecka Full time employment in Genomed S.A., Przemysław Łyszkiewicz Part time employment in Genomed S.A., Robert Szymańczak Full time employment in Genomed S.A., Maria Jędrzejowska Part time employment in Genomed S.A., Sławomir Teper: None declared, Maciej Krawczyński Employment in Diagnostyka GENESIS Sp. z o.o, Anna Boguszewska-Chachulska Ownership of Genomed S.A. stocks, Full time employment in Genomed S.A.

EP04.026 Variable expression of novel FGF10 splice-site mutation in large kindred with aplasia of lacrimal and salivary glands (ALSG).

ofek freund 1, Baker Alsana2, Nadav Agam1, Matan M. Jean1, Amit Safran1, Tomer Poleg1, libe gradstein2, Erez Tsumi2, Ohad Shmuel Birk1;3

1ben gurion university, Faculty of Health Sciences, beer sheva, Israel; 2soroka medical center, Department of Ophthalmology, beer sheva, Israel; 3soroka medical center, genetic institute, beer sheva, Israel

Purpose: Twelve individuals from 6 generations of single Bedouin kindred presented with epiphora from an early age. We aimed to delineate the disease’s clinical phenotype and identify the underlying genetic defect.

Methods: Clinical phenotyping was determined by senior ophthalmologists and geneticists following informed consent and IRB approval. Genetic studies combined linkage analysis and whole exome sequencing, followed by filtration of candidate variants and segregation analysis. Aberrant splicing of mutant FGF10 exon 2 was assayed in HEK-293 cells transfected with plasmids harboring the wild-type or mutant sequences, followed by RT-PCR using primers from exon 1 and 3.

Results: Ophthalmologic examination of the affected individuals revealed tear deficiency and congenital punctal atresia. Multiple caries with no concomitant abnormalities of the ears or digits were also noted, determining a diagnosis of aplasia of the lacrimal and salivary glands (ALSG). Variable expression and penetrance were evident between affected individuals and bilaterally within individuals. Genetic analysis identified a disease-causing novel heterozygous splice-site mutation in intron 2 of FGF10, segregating through the kindred as expected for dominant heredity with incomplete penetrance. Transfection of HEK-293 cells with plasmids expressing wildtype or mutant FGF10 sequences proved that the mutation eliminates exon 2 in the mutant RNA.

Conclusions: We report a novel dominant splice-site mutation in FGF10, abrogating transcription of exon 2 and causing ALSG with epiphora from a young age, with variable expression and incomplete penetrance.

Conflict of Interest: None declared

EP04.027 Recurrent MYO6 variants as a result of a small genome cohort study for hearing loss

Michaela AH Hofrichter 1, Christian W Remmele1;2;3, Asuman Koparir1, Daniel Bengl1, Tabea Böttcher1, Tobias Müller4, Marcus Dittrich1;4, Wafaa Shehata-Dieler5, Sophie Flandin5, Thomas Haaf1

1Institute of Human Genetics at the University of Würzburg, Würzburg, Germany; 2Center for Rare Diseases, University Hospital Würzburg, Würzburg, Germany; 3Bavarian Genomes Network for Rare Diseases, Germany; 4Department of Bioinformatics, Würzburg, Germany; 5Comprehensive Hearing Center, Department of Otorhinolaryngology, University Hospitals, Würzburg, Germany

Background/Objectives: Hearing loss (HL) is one of the most common sensory disorders, affecting around 5% of the world’s population. A genetic predisposition is suspected in 60% of cases. Because of the considerable heterogeneity in HL, pinpointing the genetic source poses a challenge. The implementation of advanced technologies such as whole genome sequencing (WGS) consequently enhances the efficacy of elucidating the root cause of HL.

The scope of this genome cohort study with 15 patients is to find the causative HL variants.

Methods: DNA samples from each index patients were prepared according to the Illumina WGS protocol and sequenced on the NovaSeq6000. Most patients were already pre-tested at least for GJB2. Analyses for SNVs, INDELs and structural variations were performed using in-house genome analysis pipeline. Disease causing variants were validated by Sanger sequencing and segregation analysis.

Results: Overall, a causing variant was discovered in 5/15 patients (33%). However, the analysis is still ongoing. Interestingly, we also detected a heterozygous variant c.884_893del in MYO6 (NM_004999.4) in two index patients with expected autosomal dominant trait for HL. Pre-analysis of the haplotype suggest a hotspot or founder effect.

Conclusion: Overall, a genetic diagnosis was contributed for 1/3 of our patients. In half of these patients, this was only possible using WGS analysis after preliminary exome/panel analysis failed. This allowed the detection of the MYO6 variant and determined the recurrent character, which expand the genetic landscape of the gene MYO6 as well as hereditary HL.

Grants:

Conflict of Interest: None declared

EP04.029 20 years of aniridia testing in Udine: more than meets the eye

Catia Mio1, Jessica Zucco2, Carla Reale3, Concetta Ambrosino3, Federica Baldan1, Lorenzo Allegri1, Alessandra Franzoni2, Angela Valentina D’Elia2, Flavio Faletra4, Giuseppe Damante 1

1Università degli Studi di Udine, Dipartimento di Medicina, Udine, Italy; 2Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Istituto di Genetica Medica, Udine; 3Biogem Scarl, Istituto di Ricerche Genetiche, Ariano Irpino, Italy; 4Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Istituto di Genetica Medica, Udine, Italy

Background/Objectives: Anterior segment dysgenesis (ASD) refers to a spectrum of congenital disorders characterized by a wide range of heterogeneity that affect the development of the cornea, the iris and the lens. The most well-known anterior segment abnormality affecting the iris is aniridia, which is associated with heterozygous loss of function variants in PAX6. Genomic research has indeed boosted our understanding in the molecular basis of ASD. Here we describe the molecular characterization of a cohort of 162 patients displaying isolated or syndromic congenital ASD (i.e., isolated or syndromic aniridia, Peters anomaly, Axenfeld-Rieger and Gillespie syndromes).

Methods: Sanger sequencing and multiplex ligation-dependent probe amplification were used to analyse the 11p13 locus. Whole exome sequencing (WES) was performed on PAX6-negative samples.

Results: Our data reiterate the notion that PAX6 alterations are primarily associated with ASD since the majority of the cohort (66.7%) has a pathogenic or likely pathogenic variant in the 11p13 locus. WES allowed us to assess variants in known genes (i.e., CYP1B1, ITPR1, MAB21L1, PXDN and PITX2) and to identify rarer phenotypes (i.e., MIDAS and OGIN syndromes). Besides, a heterozygous MAF deletion was identified in a patient with bilateral aniridia. Morpholino treatments in Danio Rerio suggested a putative involvement of MAF in lens dysgenesis.

Conclusion: Our data clearly suggest that WES allows expanding the analytical portfolio of ocular dysgenesis, both isolated and syndromic, and that is pivotal for the differential diagnosis of those conditions in which there may be phenotypic overlaps and in general in ASD.

Grants: n.a.

Conflict of Interest: None declared

EP04.030 Genetic Profile of Retinopathies Due to Disease-Causing Variants in Leber Congenital Amaurosis (LCA)-Associated Genes in a Greek Cohort

Smaragda Kamakari 1, Stavrenia Koukoula2

1OMMA Ophthalmological Institute of Athens, Ophthalmic Genetics, Athens, Greece; 2Ophthalmica Institute of Ophthalmology and Microsurgery, Electrophysiology, Thessaloniki, Greece

Background/Objectives: Leber congenital amaurosis (LCA) is the most frequent cause of congenital blindness in children and most severe form of inherited retinal dystrophies. To date, more than 25 genes have been implicated in the pathogenesis of LCA and Early-Onset Severe Retinal Dystrophy (EOSRD). The aim of the study was to report the molecular basis of LCA/EOSRD in 51 Greek families.

Methods: DNA microarrays and/or Next Generation Sequencing panel for LCA genes were performed to identify potentially pathogenic variants. Sanger sequencing was used for identification of the 2nd allele (2 cases) and segregation analysis to confirm biallelism within the families.

Results: The molecular background was established in 51 independent patients. 48 distinct variants were identified in 12 genes: CEP290, CRB1, GUCY2D, RDH12, RPGRIP1, AIPL1, RPE65, NMNAT1, TULP1, SPATA7, LCA5 and IQCB1. Almost 1/5 of the patients were affected with LCA10 due to CEP290 gene variants with its intronic mutation c.2991+1655A > G being the most frequent. CEP290 was the most prevalent gene in our cohort (17.6%), followed by CRB1 and GUCY2D (11.8% each), RDH12 and RPGRIP1 (9.8% each), AIPL1 and RPE65 (7.8% each), NMNAT1, TULP1 and SPATA7 (5.9% each), LCA5 (3.9% each) and IQCB1 (2%).

Conclusion: This study provides the first molecular genetic characteristics of patients with LCA from the previously unexplored Greek population. Our study expands the mutational spectrum as we report novel variants in LCA genes and allows for the formation of a cohort for target therapy of the disorder.

Grant: EU FP6, Integrated Project ‘EVI-GENORET’ (LSHG-CT-2005-512036; S.K)

Conflict of Interest: None declared

EP04.031 A novel disease-causing TUBB4B variant in an Italian family diagnosed with progressive cone dystrophy and early onset sensorineural hearing loss

Margherita Scarpato 1, Francesco Testa2, Roberta Zeuli1, Sandro Banfi1;3, Francesca Simonelli2, Marianthi Karali1;2

1Università degli Studi della Campania ‘Luigi Vanvitelli’, Medical Genetics, Department of Precision Medicine, Naples, Italy; 2Università degli Studi della Campania ‘Luigi Vanvitelli’, Multidisciplinary Department of Medical, Surgical and Dental Sciences, Eye Clinic, Naples, Italy; 3Telethon Institute of Genetics and Medicine, Naples, Italy

Background: Leber congenital amaurosis with early-onset deafness (LCAEOD) is a rare autosomal dominant syndrome manifesting retinal degeneration and sensorineural hearing loss (SHL). LCAEOD was first described in 2017 in four families segregating heterozygous missense mutations in TUBB4B, a gene encoding a β-tubulin isotype. To date, only three other families with TUBB4B-associated LCAEOD have been reported. All described cases harboured missense variants affecting the same amino acid (Arg391).

Methods: Whole Exome Sequencing (WES) was performed on a 45-year-old female and her 9-year-old son, both diagnosed with progressive cone dystrophy and early onset bilateral SHL.

Results: Ophthalmological assessment of the proband showed macular dystrophy with osteoblast-type pigmentation at the posterior pole, reduced visual acuity, nonrecordable photopic and subnormal scotopic electroretinography (ERG) responses. Her son presented diffuse dystrophy of the retinal pigment epithelium, thinning of the macular area and markedly reduced photopic ERG responses. WES of the proband and her son identified a shared c.1049A>C point mutation in TUBB4B (NM_006088) which causes the substitution of a highly conserved Lysine with Threonine at amino acid position 350. The variant is novel, absent from population databases, and classified as Likely Pathogenic according to the ACMG criteria.

Conclusion: Here, we report a novel, disease-causing, missense variant in TUBB4B in two affected members of an Italian family segregating with progressive cone dystrophy and early onset SHL. Our findings corroborate the recently reported association of TUBB4B variants with sensorineural syndromic forms, expand the list of pathogenic TUBB4B variants, and demonstrate that TUBB4B mutations can also cause cone-dominated retinal phenotypes.

Conflict of Interest: None declared

EP04.032 Unveiling Genetic Insights: NGS Analysis of 57 Subjects with Inherited Retinal Disorders

Federico Rondot1, Amedeo Primerano2, Patrizia Dentelli1, Enrico Grosso2, Alessandra Pelle 2, Barbara Pasini1;2

1University of Turin, Department of Medical Sciences, Turin, Italy; 2AOU Città della Salute e della Scienza di Torino, Turin, Italy

Background/Objectives: Inherited Retinal Disorders (IRD) constitute a heterogeneous group of genetic conditions marked by retinal dystrophy, resulting in progressive visual field impairment, whether presented in a syndromic or isolated clinical context. This study presents a cohort of 57 subjects clinically diagnosed with IRD, exhibiting a spectrum of clinical presentations, including retinitis pigmentosa, Stargardt disease, macular dystrophy, and less specific retinal dystrophy. Employing a Clinical Exome platform and a virtual panel approach in Singleton, Duo, and Trio settings, subjects underwent Next-Generation Sequencing (NGS) analysis, enhancing the comprehensive genetic evaluation of IRD phenotypes.

Methods: SureSelect-Agilent Custom-Constitutional-Panel-17Mb encompassing 5219 genes. Virtual panel “Inherited Retinal Disorder” (157 genes).

Results: A definitive molecular diagnosis was successfully established in 30 out of 57 patients. However, in 27 patients, conclusive molecular diagnosis remained elusive, attributed to uncertain significance variants (6/27), heterozygous pathogenic variants in autosomal recessive genes (7/27), and the absence of detectable candidate variants (14/27). Despite the cohort’s heterogeneity, a mutation detection rate of 53% was achieved, aligning with published data on genetic diagnoses in Inherited Retinal Disorders (IRD).

Conclusion: This study deepens our understanding of Inherited Retinal Disorders (IRD), underscoring the necessity for integrated clinical and genetic approaches in both diagnosis and management. The identification of unreported pathogenic variants broadens insights into the pathological molecular mechanisms of these disorders, laying the groundwork for further functional and diagnostic investigations in unresolved cases.

Grants:

Conflict of Interest: None declared

EP04.033 Our experience in analysing stereocilin (STRC) gene alterations detected by next-generation sequencing

Sara Álvaro Sánchez 1, mar borregán prats1, Judith Armstrong1;2;3, Daniel Castillo1, Sara Cardelus Vidal4, Águeda Díaz Anadon4, Maurizio Levorato4, Francesc Palau1;5;6, Loreto Martorell Sampol1

1Sant Joan de Déu Barcelona Hospital, Molecular and Genetic Medicine Department, Esplugues de Llobregat, Spain; 2Institut de Recerca Sant Joan de Déu (IRSJD), Genómica para el diagnóstico de enfermedades raras, Barcelona, Spain; 3CIBER - Center for Biomedical Research Network, CB06/07/0061, Madrid, Spain; 4Sant Joan de Déu Barcelona Hospital, Otorhinolaryngology Department, Esplugues de Llobregat, Spain; 5Institut de Recerca Sant Joan de Déu (IRSJD), Neurogenética y Medicina Molecular, Barcelona, Spain; 6CIBER - Center for Biomedical Research Network, CB06/07/0001, Madrid, Spain

Background/Objectives: Approximately 60% of hearing loss cases have a genetic basis, identifying it is crucial for prognosis and treatment.

Biallelic mutations in STRC gene result in DFNB16. STRC is located in a complex chromosomal region, which comprises its pseudogene (STRCP1).

Acknowledging NGS limitations in detecting Copy-Number-Variants (CNVs), we aim to elucidate STRC alterations identified through short-read exome sequencing by comparing them with multiplex ligation-dependent probe amplification (MLPA) technique.

The goal is to provide an accurate genetic diagnosis for patients with DFNB16.

Methods: Exomes were analysed using a custom Hearing~Loss panel. Cases without STRC alterations were excluded.~The remaining were grouped and CNVs were validated using MLPA.

Results: DFNB16 patients were categorized into six groups:

Biallelic STRC variants at 15:43890086-43939998 region

Homozygous

1

*Large CNVdeletion = 48.5kb

Compound heterozygous

2

CNVdeletion = 48.5kb + CNVdeletion≠48.5kb

3

CNVdeletion = 48.5kb + hemizygous SNV

4

SNV1 + SNV2

Homozygous

5

SNV

Other

6

**Potential DFNB16 patients: carriers or with atypical alterations and duplications

  1. *Males are at risk of suffering from Deafness-Infertility-Syndrome (DIS).
  2. **Patients under study to clarify their status.

Results were not always coincident; for solving challenging cases, MLPA and familial segregation studies were necessary. The main cause of DFNB16 was large homozygous CNVdeletion of 48.5kb, encompassing STRC and the adjacent CASPER2 gene.

Conclusion: While NGS has limitations in detecting CNVs at STRC, complementing with MLPA allows for precise genetic diagnoses. Careful interpretation of the results by experienced professionals is required to provide accurate information and, consequently, appropriate genetic counselling for these families.

Grants: Not aplicable.

Conflict of Interest: None declared

EP04.034 Blended phenotypes in rare Inherited Retinal Dystrophies; Coinheritance of CNGB3-associated Achromatopsia and RLBP1-related Retinitis Punctata Albescens

Ayse Gurel 1, Omer Yildiz2

1Abant Izzet Baysal University Izzet Baysal Training and Research Hospital, Department of Medical Genetics, Bolu, Türkyie; 2Abant Izzet Baysal University Izzet Baysal Training and Research Hospital, Department of Ophthalmology, Bolu, Türkyie

Background/Objectives: Achromatopsia (ACHM, MIM #262300) is a congenital cone dystrophy characterized by poor color discrimination, low visual acuity, nystagmus and photoaversion. It is caused by biallelic mutations in CNGB3, a gene encoding cone photoreceptor channel subunits. Concurrently, Retinitis Punctata Albescens (RPA, MIM #136880) is a fleck retina disease presenting with yellow-white deposits throughout the retina and night blindness, caused by biallelic mutations in the RLBP1 gene. This study reports the coinheritance of homozygous pathogenic mutations in CNGB3 and RLBP1 genes within the proband, clarifying the notable clinical variability observed among affected siblings.

Methods and results: The proband, 34-year-old female born to consanguineous parents, displayed poor visual acuity and absent color discrimination since birth, along with photosensitivity. Ophtalmological evaluation revealed myopia, nystagmus and flecks over the fundus. The proband’s brother presented with photosensitivity, absent color vision and reduced visual acuity while the affected sister, clinically diagnosed with retinitis pigmentosa, exhibited progressive vision impairment and night blindness. Clinical exome sequencing of the proband revealed a homozygous splice acceptor mutation in CNGB3 c.1579-1G > A and a homozygous missense mutation located in the last nucleotide of exon 5 of RLBP1 c.346G>C (p.Gly116Arg), presumably affecting the normal splicing mechanism.

Conclusion: To the best of our knowledge, this study demonstrates the coinheritance of ACHM and RPA for the first time in the literature, accentuating mutually deleterious outcome in the proband. The blended phenotypes observed in Inherited Retinal Dystrophies, which are clinically and genetically heterogeneous, highlight the importance of comprehensive gene panels and detailed evaluation of intrafamilial phenotypic variability.

Grants:

Conflict of Interest: None declared

EP04.035 Prevalence of STRC variants among mild-moderate hearing loss or presumptive autosomal recessive inheritance in the Brazilian population

Stella Diogo-Cavassana1, Danillo Alencar-Coutinho1, Allan Anjos-Monteiro1, Thais Pailo-Carvalho2, Emi Z. Murano3, Jeanne Oiticica1;3, Ricardo F. Bento1;3, Regina Mingroni-Netto2, Ana Carla Batissoco1, Karina LEZIROVITZ 1

1Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Laboratório de Otorrinolaringologia/LIM32, São Paulo, Brazil; 2Instituto de Biociências, Universidade de São Paulo, Departamento de Genética e Biologia Evolutiva, Centro de Pesquisas sobre o Genoma Humano e Células-Tronco, São Paulo, Brazil; 3Hospital das Clínicas HCFMUSP, Faculdade de Medicina da Universidade de São Paulo, ENT Department, São Paulo, Brazil

Background/Objectives: Hereditary non-syndromic hearing loss has 80% of its inheritance transmitted recessively. Regarding mild to moderate hearing loss, studies have suggested that pathogenic variants in STRC (DFNB16), especially large deletions, should be considered as the second major cause of this phenotype, with an estimated frequency between 1% to 5%. Due to the high prevalence of STRC causative variants, STRC screening is recommended in laboratory routines around the world. A genome sequencing study of 2097 normal-hearing Brazilian individuals revealed a heterozygous ~57.9kb STRC microdeletion in 38 individuals; which suggested it as the most recurrent CNV (AF = 0.91%) in Brazilians. The present study aims to determine the frequency of STRC deletions by investigating selected Brazilian subjects with mild-moderate hearing loss or presumptive autosomal recessive inheritance.

Methods: The study subjects were selected for STRC deletion screening based on mild-moderate hearing loss or presumptive autosomal recessive inheritance. A TaqMan-based qPCR assay was used to identify STRC CNVs. In cases with heterozygous CNVs, a long-range PCR and sequencing assay screened the hemizygous allele.

Results: Our cohort comprised 66 mild-moderate and 70 presumptive autosomal recessive cases. To date, three Brazilian families showed homozygosity for STRC deletions, all exhibiting familial recurrence of moderate deafness.

Conclusion: Given the high prevalence of STRC deletions among subjects with moderate hearing loss, we suggest screening as a routine investigation. Further studies will aim to find if it is the same deletion found in the general Brazilian population.

Grants: The São Paulo Research Foundation FAPESP funded this research (numbers 23/07188-7 and CEPID 2013/08028-1).

Conflict of Interest: None declared

EP04.036Frequency of STRC-related hearing loss in GJB2 negative individuals

Silvia Borecka 1, Klaudia Cipkova1, Lukas Varga1;2, Dimitrios Paouris3, Martina Skopkova1, Miroslava Huckova1, Dominika Hromnikova1, Milan Profant2, Daniela Gasperikova1

1Institute of Experimental Endocrinology, Biomedical Research Center SAS, Bratislava, Slovakia; 2Medical Faculty of Comenius University and University Hospital, Department of Otorhinolaryngology – Head and Neck Surgery, Bratislava, Slovakia; 3Medical Faculty of Comenius University and National Institute of Children’s Diseases, Clinic of Pediatric Otorhinolaryngology, Bratislava, Slovakia

Background/Objectives: Deletions and mutations in the STRC gene are the second most common cause of autosomal recessive, mild to moderate, nonsyndromic sensorineural hearing loss (SNHL). Larger deletions overlapping into the CATSPER2 gene may lead to a syndromic hearing disorder associated with male infertility (DIS syndrome). Exploring these changes is methodologically complicated due to a high homology between the STRC gene and its pseudogene. The aim of our work was to identify the copy number variations and pathogenic variants in the STRC gene in a selected population of Slovak patients.

Methods: 371 GJB2 negative probands with mild to moderate SNHL onset up to 15 years were selected out of the 1 200 probands registered in the SNHL biobank. For determination of the genetic etiology, MLPA, long-range PCR, nested PCR, Sanger sequencing and KASP genotyping assay were used.

Results: We identified a homozygous deletion of the STRC, CKMT1B and CATSPER2 region in 8 probands. Five of these probands are male, suggesting the DIS syndrome. Homozygous deletions of separate STRC exons of different lengths were observed in another 8 probands. Eight probands were compound heterozygotes for deletion of the STRC gene and pathogenic variants on the second allele. The variant R1073* was the most frequent one and was detected in five probands.

Conclusion: The genetic etiology of hearing impairment was determined in 24 out of 371 probands (6,5%). Our results confirmed that changes in the STRC gene are the second most common cause of hearing loss in Slovakia.

Grants: APVV-20-0236, VEGA 1/0572/21

Conflict of Interest: None declared

EP04.038 Genetics of eye disorders in Cypriot patients

Andri Miltiadous 1, Paul Costeas1, maria kanezou1, Petroula Gerasimou1, rafaella metaxa2, Emily Athanasiou3, George A. Tanteles4, Violetta Anastasiadou2

1Karaiskakio Foundation, Molecular Diagnostic Laboratory; 2Karaiskakio Foundation, Genetics Clinic; 3Makario Children’s Hospital, Genetics Department, Cyprus; 4The Cyprus Institute of Neurology and Genetics, Egkomi Lefkosias, Cyprus

Background/Objectives: Inherited eye diseases are the leading cause of blindness in adults and in children. Diagnosis of these disorders has witnessed significant progress the last decade in the advent of next generation sequencing.

Methods: Patients affected with eye diseases including but not limited to retinitis pigmentosa, glaucoma, cone rod dystrophy, leber congenital amaurosis, and ocular albinism were referred to our laboratory and NGS sequencing was performed with Agilent’s Exome V8 panel and data were analysed using Franklin software.

Results: Diagnostic rate in eye disease genetic investigation was >80% and causative or likely causative variants were reported in the following genes:ABCA4, CRB1, TYR, MAK, FOXC1, POC1B, NR2E3, GRK1, CFH, EYS, GPR143, RPE65. In addition syndromic cases with eye abnormalities were reported to harbour variants in BBS7 and CEP41.

Conclusion: Eye disease is a significant cause of vision loss and therefore it’s crucial that is is diagnosed early on the course of disease for the correct type of treatment and/or surveillance. Non syndromic eye diseases with known monogenic causes have a relatively high diagnostic rate due to the very specific phenotype that is investigated compared to broader or more complex phenotypes.

Grants: No grants were received for this study

Conflict of Interest: None declared

EP04.039 Oculocutaneous albinism

Elena Colastra Ugena 1, Ana Peña Cabia1, Irene Pereira Gonzalez1, Raquel Gomez Molina1, Maria Perez Gomez1, Maria Concepcion Calderon Alva1

1Hospital Virgen de la Luz, Análisis Clínicos, Cuenca, Spain

Background/Objectives: 19-year-old woman with a personal history of hypermetropia and astigmatism, with a suspected diagnosis of macular dystrophy.

Methods: A clinical exome was performed to test for genes related to hereditary retinal dystrophies, and no variant was detected that could explain its phenotype.

It was decided to extend the study to genes related to albinism, since it presented flat macules with absence of fovea.

Results: In this reanalysis, three genetic variants were identified in the TYR gene:

  • c.405del(p.Phe135Leufs*6) in heterozygosis, classified as probably pathogenic.

  • c.575C>A(p.Ser192Tyr) in heterozygosity and c.1205G>A(p.Arg402Gln) in homozygosis, both classified as of uncertain clinical significance.

The detection of these variants is compatible with the patient’s clinical features.

A family segregation study is requested from the parents to determine the phase of the variants identified, since pathogenic variants in the TYR gene are associated with oculocutaneous albinism type IA and IB (autosomal recessive pattern).

After analysis, the p.Arg402Gln variant is in cis phase with the p.Ser192Tyr variant, and both are in trans phase with the p.Phe135Leufs variant. The p.Arg402Gln variant is considered a hypomorphic allele and when combined with the p.Ser192Tyr variant in cis, it is estimated to have a loss of function effect equal to or greater than a pathogenic variant in the TYR gene.

Conclusion: The patient is a carrier in compound heterozygosis for variants probably pathogenic in the TYR gene, diagnosis of the disease.

Family segregation study plays a fundamental role in the diagnosis of this disease, thus it is essential to provide adequate genetic counseling.

Grants: None

Conflict of Interest: None declared

EP04.042 A patient with ATOH7 variants with Delayed Sleep-Wake Phase Disorder and Optic Nerve Hypoplasia

Jennifer Brzezynski 1, Caroline Johnson1, Sandra Smieszek1, Changfu Xiao1, Christos Polymeropoulos1, Mihael Polymeropoulos1

1Vanda Pharmaceuticals Inc., Washington, DC, United States

Background/Objectives: We are conducting a study to evaluate the effects of tasimelteon in Delayed Sleep-Wake Phase Disorder (DSWPD) patients. We report our first completed patient from the Open-Label Extension (OLE), a 24-year-old female diagnosed with DSWPD and Optic Nerve Hypoplasia (ONH), with delayed Dim Light Melatonin Onset.

Methods: The study consists of screening and treatment, followed by an OLE to explore the safety and efficacy of daily dosing with tasimelteon over 11 months. During the OLE, patients answer sleep diaries and take one dose of tasimelteon 1 hour before their desired bedtime.

Results: The patient’s average sleep onset was 01:27 during screening and 00:42 during the OLE, an improvement of 45 minutes. At screening, the patient reported their symptoms as moderate on the Patient Global Impression of Severity (PGI-S). On average during the OLE, the patient reported their symptoms as mild on the PGI-S.

The patient’s history of ONH could indicate lack of proper optic response to light. They carry two variants in the ATOH7 gene, rs61854782 and rs7916697, known to be associated with ONH. They have a VNTR PER35/5 genotype, associated with normal sleep patterns, and no identified predicted loss-of-function mutations within other circadian genes.

Conclusion: This case illustrates the positive effect of tasimelteon on a patient diagnosed with DSWPD, based on an earlier sleep onset shift and improvement in PGI-S responses. It also provides the opportunity for research into ONH and ATOH7 variants, and the relationship with DSWPD and sighted Non-24.

Grants: Not Applicable.

Conflict of Interest: Jennifer Brzezynski Employee and stock owner., Employee and stock owner., Caroline Johnson Employee and stock owner., Employee and stock owner., Sandra Smieszek Employee and stock owner., Employee and stock owner., Changfu Xiao Employee and stock owner., Employee and stock owner., Christos Polymeropoulos Employee and stock owner., Employee and stock owner., Mihael Polymeropoulos CEO and stock owner., CEO and stock owner.

EP04.043 Optical Genome Mapping facilitates rapid characterisation of structural variants in families with developmental eye anomalies

Solomon Merepa 1, Karthikah Jeganathan1, Bertrand Chesneau1;2;3, Richard Holt1, Lidiya Talbot1, Fabiola Ceroni1, Dorine Bax1, Fiona Watkins1, Mira Kharbanda4, Katherine Lachlan4, Patrick Calvas3;5, Julie Plaisancié3;5, Nicolas Chassaing3;5, Elfride De Baere6;7, Nicola Ragge1;8

1Oxford Brookes University, Faculty of Health and Life Sciences, Oxford, United Kingdom; 2Université de Toulouse, Molecular, Cellular and Developmental Biology Unit (MCD), Centre de Biologie Intégrative (CBI), Toulouse, France; 3CHU de Toulouse, Centre de Référence pour les Affections Rares en Génétique Ophtalmologique (CARGO), Toulouse, France; 4University Hospital Southampton NHS Foundation Trust, Wessex Clinical Genetics Service, Southampton; 5Laboratoire National de Référence (LBMR), Génétique des anomalies malformatives de l’œil, Toulouse, France; 6Ghent University, Department of Biomolecular Medicine, Ghent, Belgium; 7Ghent University Hospital, Center for Medical Genetics Ghent (CMGG), Ghent, Belgium; 8Birmingham Women’s and Children’s NHS Foundation Trust, West Midlands Regional Clinical Genetics Service and Birmingham Health Partners, Birmingham, United Kingdom

Introduction: Developmental eye anomalies, including anophthalmia, microphthalmia and coloboma (AMC), are a genetically heterogeneous group of disorders. Structural variants (SVs) affecting coding and non-coding regions of the genome can alter expression, function and regulation of genes implicated in eye development. Conventional diagnostic methods, including array CGH and WGS, can limit resolution and/or exact nature of rearrangements. Here, we use Optical Genome Mapping (OGM) to identify and characterise SVs in a cohort of individuals with AMC.

Methods: Using ultra-high molecular weight DNA, we screened 53 AMC families for SVs using OGM (Bionano Genomics, USA).

Results: We identified heterozygous deletions involving known eye development genes in the retinoic acid pathway (ALDH1A3 and STRA6) in two independent families with bilateral anophthalmia. These SVs were inherited in trans with corresponding rare SNVs, providing diagnosis. In three further families, OGM characterised complex rearrangements involving genes part of pathways relevant to eye development; i) a complex SV with two deletions flanking an inversion affecting CYP26A1 and CYP26C1, genes encoding enzymes involved in retinol metabolism, affecting two unrelated families; ii) an individual with 3 separate SVs, including a chromosome 11q13 duplication containing SIMO and other cis regulatory elements of PAX6; and iii) an insertional translocation involving chromosomes 3 and 5 which may impact important regulatory elements.

Conclusion: We demonstrate that OGM can easily identify clinically relevant SVs, including complex rearrangements, in individuals with AMC. This technology has the potential to complement clinical genetic testing, and support research into the mechanisms underlying such genetic disorders.

Funding: Baillie Gifford

Conflict of Interest: None declared

EP05 Internal Organs & Endocrinology (Lung, Kidney, Liver, Gastrointestinal)

EP05.001 Long read genome sequencing facilitates CNV mapping at highly homologous loci

Nina Bögershausen 1, Alexander Wolff1, Yun Li1, Tassilo Wollenweber2, Julia Schmidt1, Silke Kaulfuß1, Karl Koehrer2, Arne Zibat1, Gökhan Yigit1;3, Bernd Wollnik1;3;4

1Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany; 2Genomics and Transcriptomics Laboratory, Biologisch-Medizinisches-Forschungszentrum (BMFZ), Düsseldorf, Germany; 3German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany; 4Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany

Background/Objectives: High sequence similarity in segmental duplications can facilitate copy number variation (CNV). Within such regions, sequence identity may hinder exact short read mapping and consequently, the detection of CNV boundaries by short read sequencing (SRS) approaches. Here, we present a family with two siblings, affected by juvenile onset exocrine pancreatic insufficiency (EPI), in whom long read genome sequencing (LRGS) was successfully applied.

Methods: We initially employed short read exome sequencing and/or qPCR in both siblings and their parents. Additionally, we performed HiFi LRGS in one sibling, using the PacBio Sequel II system, and analyzed the data with our in-house pipeline.

Results: Exome sequencing identified a homozygous deletion at the CELA3A / CELA3B locus in both affected siblings. CELA3A and CELA3B are highly homologous genes, likely owing to gene duplication. The boundaries of the deletion could not be determined with SRS due to misalignment. LRGS, with an average read length of ~20.000 bp, allowed us to map the breakpoints more precisely. The deletion creates a CELA3A/CELA3B fusion gene, possibly disrupting the function of both genes. CELA3A and CELA3B code for digestive enzymes that are detected clinically as stool elastase. The fact that the deletion affects both genes, likely explains the observation of extremely low stool elastase levels in both siblings.

Conclusion: We present a biallelic deletion at the CELA3A/B locus as a potential novel cause for EPI and demonstrate the efficiency of LRGS for breakpoint mapping in highly homologous regions. We aim to identify further individuals with CELA3A/B associated EPI.

Grants: Deutsche Forschungsgemeinschaft (Project 417959134)

Conflict of Interest: None declared

EP05.002 Outcomes of patients with Juvenile Polyposis - Hereditary Haemorrhagic Telangiectasia caused by pathogenic SMAD4 variants in a pan-Scotland cohort.

Madeline Pearson 1, Ruth McGowan2, Philip Greene3, Zosia Miedzybrodzka4, jonathan berg1

1University of Dundee, School of Medicine, United Kingdom; 2West of Scotland Centre for Genomic Medicine, United Kingdom; 3South East of Scotland Clinical Genetics Services, United Kingdom; 4University of Aberdeen, School of Medicine, United Kingdom

Background/Objectives: SMAD4 is integral to mediating signal transduction for serine threonine kinase receptors. Germline loss of SMAD4 function (LOF) results in Juvenile Polyposis-Hereditary Haemorrhagic Telangiectasia Overlap Syndrome (JP-HHT). JP-HHT is associated with life-threatening complications including gastrointestinal cancers and pulmonary arteriovenous malformations (PAVMs). We investigated the JP-HHT phenotype in a Scottish cohort.

Methods: A retrospective multi-centre case-note review identified 28 patients with a pathogenic LOF SMAD4 variant from 13 families across all Scottish Clinical Genetics Centres, providing a complete clinical picture of the Scottish JP-HHT cohort.

Results: Colonic polyps were identified in 87%(20/23) of screened patients. 43%(10/23) of patients with polyps required a colectomy at a median age of 24years (IQR20.0–47.0). Colorectal cancer occurred in 25%(7/28) of patients at a median age of 33years (IQR29.5–45.0).

Gastric polyps were identified in 67%(12/18) of screened patients. 42%(5/12) of patients with gastric polyps required a gastrectomy at a median age of 47years (IQR37.0–59.0). 1 patient was diagnosed with gastric cancer, at age 66 years.

PAVMs were identified in 47%(9/19) of screened patients. When investigated for, epistaxis and telangiectasia were present in 75%(15/20) and 29%(4/14) of patients respectively.

88%(23/26) and 81%(17/21) of patients exhibited JP and HHT features respectively, with 70%(14/20) demonstrating features of both conditions.

Conclusion: SMAD4 variant carriers are at very high risk of life-threatening gastrointestinal disease, including early onset bowel cancer and gastric obstruction caused by non-malignant polyposis. PAVM incidence was at least equivalent to that seen in HHT type 1, if not higher. All individuals with pathogenic LOF SMAD4 variants require comprehensive screening.

Grants:

Conflict of Interest: None declared

EP05.003 Defining PKD1 variants in patients with cystic kidney

Deimante Brazdziunaite 1, Gabija Mazur2, Algirdas Utkus1

1Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania; 2Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania

Background: PKD1 gene pathogenic variants are the most common cause of autosomal dominant polycystic kidney disease (ADPKD; OMIM #173900; ORPHA:730), which is the most common in the group of cystic kidney diseases. The ADPKD database counts more than 1200 pathogenic/likely pathogenic variants in PKD1, while more than 200 remain as variants of uncertain significance (VUS).

Aim: To identify and classify genetic variations within PKD1 gene among patients exhibiting multiple renal cysts.

Methods: 62 patients with multiple renal cysts were included in the study. The study group consisted of 35 women and 27 men; 57 adults and 5 children. Patients were tested with a kidney-focused next-generation sequencing panel of 498 genes, including the main polycystic kidney disease genes PKD1, PKD2, PKHD1, ALG5, DZIP1L, DNAJB11, GANAB. Pathogenicity of detected variants was assessed using the American College of Medical Genetics and Genomics guidelines.

Results: Variants in PKD1 gene were identified in 34 patients, of which 18 were not described in the literature. Family history was negative for 11 PKD1-patients. After evaluating pathogenicity of the variants, 26 were classified as pathogenic/likely pathogenic, and 8 as VUS. In 12 patients clinically significant variants were not identified in the analyzed genes. However, some of those patients were not clinically diagnosed with polycystic kidney disease or had an atypical form of the disease.

Conclusion: A genetic diagnosis was established for the majority of patients in the conducted study. All PKD1 variants identified in the study are unique and did not repeat between families.

Conflict of Interest: None declared

EP05.004 PERCC1-associated congenital enteropathy: Delineating the natural history of a new disorder of enteroendocrine cell function

lev dorfman1;2;3, Mansa Krishnamurthy4;5, Yael Haberman1;3;6;7, emily stenke8, Anat Guz Mark2;3, Hengameh Abdollahpour9, seba saleh nadeef10, Khaled Warasnhe11, Figen Ozcay12, vivien a. nguyen13, Martin G Martin14, stanley f. nelson15;16, Julian Martinez-Agosto14;15, james m. wells4;5;17, Fowzan Alkuraya10;18, Billy Bourke8;19, Yair Anikster3;20;21, Ben Pode-Shakked 3;20

1Cincinnati Children’s Hospital Medical Center, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati, United States; 2Schneider’s Children’s Medical Center, Institute of Gastroenterology, Nutrition and Liver Diseases, Petach Tikva, Israel; 3Tel-Aviv University, Faculty of Medicine, Tel Aviv, Israel; 4Cincinnati Children’s Hospital Medical Center, Division of Endocrinology, Cincinnati, United States; 5Cincinnati Children’s Hospital Medical Center, Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati, United States; 6Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Pediatric Gasteroenterology Unit, Ramat Gan, Israel; 7University of Cincinnati College of Medicine, Department of Pediatrics, Cincinnati, United States; 8Children’s Health Ireland-Crumlin, National Centre for Paediatric Gastroenterology, Dublin, Ireland; 9University Medical Center Hamburg-Eppendorf, Institute of Human Genetics, Hamburg, Germany; 10King Faisal Specialist Hospital and Research Center, Department of Translational Genomics, Center for Genomic Medicine, Riyadh, Saudi Arabia; 11Başkent University Faculty of Medicine, Ankara, Turkey, Department of Pediatrics, Ankara, Türkyie; 12Başkent University Faculty of Medicine, Department of Pediatric Gastroenterology and Hepatology, Ankara, Türkyie; 13UCSF Benioff Children’s Hospital, Division of Pediatric Gastroenterology, Hepatology and Nutrition, Oakland, United States; 14University of California Los Angeles, Department of Pediatrics, Los Angeles, United States; 15University of California Los Angeles, Department of Human Genetics, David Geffen School of Medicine, Los Angeles, United States; 16University of California Los Angeles, Department of Neurology, David Geffen School of Medicine, Los Angeles, United States; 17Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, United States; 18College of Medicine, Alfaisal University, Department of Anatomy and Cell Biology, Riyadh, Saudi Arabia; 19University College Dublin, School of Medicine, Dublin, Ireland; 20Sheba Medical Center, Metabolic Disease Unit, Edmond and Lily Safra Children’s Hospital, Ramat Gan, Israel; 21Sheba Medical Center, The Wohl Institute for Translational Medicine, Ramat Gan, Israel

Consortium: Undiagnosed Diseases Network

Abstract

Background: Congenital diarrheas and enteropathies (CODEs) constitute a heterogeneous group of individually rare disorders, characterized by infantile-onset chronic diarrhea. Recently, biallelic deletions encompassing an unannotated open reading frame termed PERCC1, were implicated in an autosomal-recessive CODE among Iraqi-Jewish families. Disruption of PERCC1 was further shown to hamper enteroendocrine cell (EEC) function in organoids and in vivo models. We sought to elucidate the natural history of this newly-recognized disorder.

Methods: Clinical data was obtained directly from the patient and/or their parents, via a virtual semi-structured medical interview. When a direct interview was not possible, clinical data was collected by the physician caring for the patient, based on their medical records.

Results: An international cohort of n = 11 patients was recruited, at an average age of 18.5 years (range, 2-37). Molecular diagnosis was reached at an average age of 14.8 years (range, 1.5m-36y). TPN was required in all patients, and was successfully weaned off in 9/11, at an average age of 8 years (range, 10m-20y). The PERCC1-related genotype underlying their disease varied between biallelic chromosomal deletions (6/11 patients; 54%); a recently identified stopgain mutation (c.390C>G) shared by unrelated Irish patients (3/11; 27%); a novel c.555C>G point mutation (1/11; 9%); or maternal UPD of chromosome 16 encompassing PERCC1 harboring a novel c.348C>G truncating variant (1/11; 9%).

Conclusions: PERCC1-associated congenital enteropathy is an ultra-rare, underdiagnosed disorder, typically manifesting at early infancy with persistent watery diarrhea and hypernatremic dehydration. Our findings expand the current knowledge regarding both the molecular basis and phenotypic spectrum of this disorder.

Conflict of Interest: None declared

EP05.005 Broadening the phenotype of MAFB disease spectrum- kidney, eye, ear and nervous system involvement

aviva eliyahu 1;2;3, Danit Atias-Varon4, Ortal Barel Barel5, Yulia Khavin5, Elon Pras3, haike reznik wolf6, Lior Greenbaum3;6, Pazit Beckerman3;7, Nabil Abu amer3;7, Tamara Vignanski-Jaffe3;8, Asaf Vivante3;9;10

1Sheba, The Danek Gertner Institute of Human Genetics, Ramat Gan, Israel; 2Bar Ilan university, The Goodman faculty of life science, Ramat Gan, Israel; 3Tel-Aviv University, Faculty of Medicine, Tel aviv, Israel; 4Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Pediatric Nephrology Unit, Ramat Gan, Israel; 5Sheba Medical Center, Tel-Hashomer, The Genomic Unit, Sheba Cancer Research Center, Ramat Gan, Israel; 6Sheba Medical Center, Tel-Hashomer, The Danek Gertner Institute of Human Genetics, Ramat Gan, Israel; 7Sheba Medical Center, Tel-Hashomer, Institute of Nephrology and Hypertention, Ramat Gan, Israel; 8Sheba Medical Center, Tel-Hashomer, Goldschlager Eye Institute, Ramat Gan, Israel; 9Sheba Medical Center, Tel-Hashomer, Department of Pediatrics B, Edmond and Lily Safra Children’s Hospital, Ramat Gan, Israel; 10Sheba Medical Center, Tel-Hashomer, Pediatric Nephrology Unit, Edmond and Lily Safra Children’s Hospital, Ramat Gan, Israel

Background/Objectives: Focal segmental glomerulosclerosis (FSGS) is a leading cause of steroid resistant nephrotic syndrome (SRNS) and chronic kidney disease (CKD). Monogenic etiologies are detected with increasing frequency for FSGS. Many of the disease-causing genes are involved in the structure of the slit diaphragm and podocyte maintenance.

Missense variants in the MAF BZIP Transcription Factor B (MAFB) gene have been recently reported as a cause of a dominantly inherited syndrome with variable expressivity, ranging from syndromic FSGS to isolated renal or ocular disease.

Methods: We report herein five affected individuals, members of an extended family, presenting with proteinuria and variable renal and extra-renal phenotype.

Results: A novel heterozygous missense variant c.797T>C p(Leu266Pro) in MAFB was made in the proband and in a sibling by Exome Sequencing (ES) and subsequent variant segregation by Sanger sequencing in the mother.

In addition to CKD and FSGS, the proband presented with congenital auricular malformations, hearing loss and neurodevelopmental delay. Her male sibling had Duane retraction syndrome (DRS) and neurodevelopmental involvement, while an additional family member presented with an isolated renal phenotype. Some of these clinical features have not been previously determined as part of the MAFB phenotypic spectrum.

Conclusion: Taken together, MAFB-related disease spectrum is broader than previously described. Awareness of this may promote accurate diagnosis and improve medical treatment.

Grants: N/A

Conflict of Interest: None declared

EP05.006 Multi-locus imprinting disturbances in patients with PHP1B/iPPSD3

Yerai Vado1, Africa Manero Ruiz de Azua1, Arrate Pereda1, Guiomar Perez de Nanclares Leal 1

1Bioaraba Health Research Institute, Genetics, Epigenetics and Cellular Biology, Gasteiz, Spain

Background/Objectives: iPPSD3 (previously known as pseudohypoparathyroidism 1B) is a rare epigenetic disorder characterized by PTH resistance, hypocalcemia, hyperphosphatemia and, sometimes, TSH resistance. It is caused by epigenetic variants occurring in one or more GNAS-DMR, with hypomethylation at GNAS A/B:TSS-DMR always present. In some imprinting disorders, methylation disturbances at multiple loci (MLID) have been described and genetic defects at genes involved in methylation establishment/maintenance reported. Our interest was to determine if MLID is present in iPPSD3 and its genetic causes.

Methods: 90 patients with epigenetically confirmed iPPSD3 were checked for MLID by MS-MLPA (kit ME034-C1). 49 of these individuals were also studied with a custom targeted methylation sequencing panel (ImprintSeq). Samples of 30 trio or, at least duo (index and mother) were analysed by NGS with a custom panel of imprinting regulator genes, specifically looking for maternal effect genes.

Results: From the 90 patients who presented an epigenetic defect, the genetic cause was identified in 24 (18 with STX16 deletions and 6 with upd(20)pat). MLID was not observed by ME034-C1, but ImprintSeq detected it in 18 patients affecting different DMRs with (mainly) no known clinical effect. Preliminary analyses of maternal effect genes have allowed the identification of some VUS variants.

Conclusion: In MLID-iPPSD3 cases (36.7% of our series) the involved DMRs are not included in currently available MS-MLPA kits and their clinical significance is unknown. Further clinical studies are needed to evaluate their impact.

Grants: Institute of Health Carlos III (PI20/00950); Basque Department of Health (GV2021/111056).

Conflict of Interest: None declared

EP05.007 Growth Hormone deficiency: spectrum of causal genetic variants in Portuguese patients

Ana C. Ribeiro 1, Najeeb Syed2, Luís R. Saraiva3, Manuel C. Lemos1

1CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal; 2Laboratory of Genomic Medicine, Sidra Medicine, Doha P.O. Box 26999, Qatar; 3Translational Medicine Division, Sidra Medicine, Doha P.O. Box 26999, Qatar

Background/Objectives: Growth Hormone (GH) deficiency is a rare disorder which in the absence of treatment can result in severe short stature. Combined (CPHD) or isolated (IGHD) GH deficiency can have a genetic origin, due to variants in genes affecting the hypothalamic-pituitary development and/or function. We aimed to characterize the causal variants of GH deficiency in a cohort of 205 Portuguese patients.

Methods: This study included 81 IGHD and 124 CPHD patients. The GH1 and GHRHR or PROP1 genes were analysed by Sanger sequencing in these groups, respectively. In patients without causal variants, and in 324 controls, whole exome sequencing (WES) was performed. Putatively pathogenic variants were filtered in 46 known GH deficiency-related genes.

Results: Causal variants were identified in 20% of GH deficiency patients. These variants were distributed across the PROP1, GLI2, PROK2, PROKR2, CDON, CHD7, GHRHR, and HESX1 genes. Regarding controls, only 2% had variants that would potentially originate this disorder. Comparing the frequency of individuals with variants of uncertain significance (VUS), the difference between patients and controls was not statistically significant. However, VUS in the RBM28 and GLI2 genes were significantly more frequent in GH deficiency patients.

Conclusion: A genetic diagnosis was established in 20% of the studied patients. At least for the RBM28 and GLI2 genes, identified VUS may also be involved in the pathogenesis of GH deficiency. Further studies are needed to confirm this hypothesis.

Grants: Portuguese Foundation for Science and Technology (PTDC/SAU-GMG/098419/2008, UIDB/00709/2020, and UI/BD/151021/2021), and Sidra Medicine (SDR200059).

Conflict of Interest: None declared

EP05.008 Dent disease is effectively identified in clinical practice and is well confirmed by whole-exome sequencing: report of seven cases

Anastasiia Buianova 1, Anastasiia Ryzhova2, Mariia Proskura2, Edita Petrosyan2, Valeriia Gavrilova2, Anna Shmitko1, Alina Samitova1, Oleg Suchalko1, Zhanna Repinskaia1, Galit Ilyina1, Anna Kuznetsova1, Iuliia Vasiliadis1, Natalia Ponikarovskaya1, Vera Belova1, Dmitriy Korostin1

1Pirogov Russian National Research Medical University, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Moscow, Russian Federation; 2Pirogov Russian National Research Medical University, Russian Children’s Clinical Hospital, Moscow, Russian Federation

Background/Objectives: Dent disease is a rare renal tubular disorder characterized by X-linked recessive inheritance. It primarily affects the proximal tubules of the kidneys and is divided into two types (associated with CLCN5 and OCRL genes, respectively). The accessibility and high effectiveness of whole-exome sequencing (WES) in patients provide the opportunity for early diagnosis and more timely treatment of such children, delaying end-stage renal disease.

Methods: WES analysis paired with comprehensive clinical evaluation was assessed in seven pediatric male patients with suspected Dent disease.

Full size table

Results: The patient group is characterized by minor clinical heterogeneity (see table). Variants of patients 1-3 were previously undescribed.

Conclusion: In this study, the preliminary diagnosis was confirmed in all patients, so we ensured that biochemical analysis of blood, urine tests, kidney ultrasound, as well as a physical examination combined with history-taking are exhaustive differential diagnostic methods for Dent disease.

Grants: №075-15-2019-1789.

Conflict of Interest: None declared

EP05.009 Novel targets for unravelling renal tubular injury in diabetes and hypertension: Effect of increased exosomal levels of miR-200a-3p

Olga Martinez-Arroyo 1, Ana Flores-Chova1, Marta Mendez-Debaets1, Laia Garcia-Ferran1, Lorena Adan1, Lesley Escriva1, Sara Vela1, Carlos Bea1;2, Josep Redon1;2;3;4, Maria J Forner1;2;3, Raquel Cortés1, Ana Ortega1;5

1Incliva Biomedical Research Institute, Cardiometabolic and Renal Risk Research Group, Valencia; 2Clinic Hospital of Valencia, Internal Medicine Unit, Valencia; 3Medicine’s Faculty, Department of Medicine, Valencia; 4Carlos III Health Institute, CIBER OBN; 5Carlos III Health Institute, CIBER CV

Background/Objectives: Hypertension and diabetes mellitus (DM) are major risk factors of renal damage. Evidence places the renal tubule as a driving force in kidney disease. Exosomes are disease biomarkers mediating specific cell communication by transferring molecules such as microRNAs. Previously, we found that miR-200a-3p has promising roles in renal injury. We aimed to analyse the levels of miR-200a-3p in exosomes from patient’s urine and deepen into its role in tubular damage.

Methods: Urinary exosomal levels of miR-200a-3p were measured by RT-qPCR in hypertensive patients with and without DM (n = 69) and increased UAE (n = 42). We investigated the exosomal and pellet levels of miR-200a-3p and their targets at mRNA and protein levels (Western blot) in tubular cells (RPTECs) subjected to treatments and observed their influence on apoptosis (flow cytometry), injury and RPTEC markers. Furthermore, we performed mimic and inhibitor miRNAs transfection in treated RPTECs.

Results: We found increased levels of miR-200a-3p in urinary exosomes from patients with increased UAE. Further, this miRNA was able to discriminate UAE with an AUC of 0.75 (p = 0.003). In in vitro experiments, miR-200a-3p and renal injury markers are increased and RPTEC markers and apoptosis decreased, under glucose and angiotensin-II conditions. miR-200a-3p mimic and inhibitor experiments showed an important influence on its targets involved in tubular damage (SIRT1 and CLDN1) and in injury markers.

Conclusion: Increased exosomal levels of miR-200a-3p emerge as a potential disease marker and treatment target to decrease renal tubular injury associated to hypertension and diabetes.

Grants: PI19/01796; PI21/00249; PI23/01179; FI20/00096; FI22/00032; CD00069; IJC2020-045308-I. ERDF.

Conflict of Interest: None declared

EP05.010 Involvement of TBC1D8B novel variant as a cause of x-linked corticosteroid resistant nephrotic syndrome.

Anney Yelena Rosario Vargas 1, María Jesus Izquierdo Ortiz1, Irene Oñate Alonso1, Badawi Hijazi Prieto1, María Isabel Sáez Calero1, Marta Boya Fernández1, Raquel Toro Casado1, Vanesa Camarero Temiño1, Basilia González Diez1, María Luisa Carrasco Prado1, Alejandra Martin Rosique1, Sheyla Cristal Álvarez Parra1

1Burgos University Hospital, Burgos, Spain

Background/Objectives: Steroid-resistant nephrotic syndrome (SRNS) is a glomerular disease characterized by massive proteinuria, most often associated with a focal and segmental glomerulosclerosis.1 This histological pattern has variety of potential causes, including rare variants of podocyte-related genes. TBC1D8B plays a regulatory role by inhibiting endogenous Rab11, a key protein in vesicular reycling cells, that interacts with nephrin dynamics in the glomerular podocyte.3

Variants in TBC1D8B gene on the X chromosome can lead to SRNS type 20 and its progression to kidney failure in individuals under 25 years-old.1 Its most commonly in boys 2, women should not express severe manifestations unless the phenomenon of lyonization is present.

We identified novel TBC1D8B variant in a 32 years-old, spanish woman on hemodialysis for unrelated chronic kidney disease since 18 years-old. Additionally, she carries a pathogenic variant in KMT2D, associated with congenital malformations of the kidney-urinary tract and the Kabuki phenotypic spectrum not present in the patient.

Methods: We carried out a genetic study with a global panel of nephropathy (529 genes).

Results: We found that the patient is a heterozygous carrier of the possibly pathogenic variant c.1426C>T:p.Arg476 in the geneTBC1D8B (NM_017752.2).

Conclusion: Pathogenic variants in the TBC1D8B gene are associated with the development of nephrotic syndrome type 20, a kidney disorder with an X-linked inheritance pattern, of variable expression, especially in women in whom the severity of the disease is assumed by the pattern of chromosome X inactivation.

Grants: No grants to declare.

Conflict of Interest: None declared

EP05.012 Exploring genetic variants for the diagnosis of chronic liver disease in the pediatric population

Olga Parshina 1, Anastasiia Buianova1, Ekaterina Filimonova2, Mariia Venediktova2, Alexandra Shagiazdanova2, Elena Dobronravova2, Stella Temurian2, Anna Shmitko1, Alina Samitova1, Oleg Suchalko1, Zhanna Repinskaia1, Galit Ilyina1, Iuliia Vasiliadis1, Anna Kuznetsova1, Mariia Sidorchuk1, Natalia Ponikarovskaya1, Vera Belova1, Dmitriy Korostin1

1Pirogov Russian National Research Medical University, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Moscow, Russian Federation; 2Pirogov Russian National Research Medical University, Russian Children’s Clinical Hospital, Moscow, Russian Federation

Background/Objectives: Genetic factors contribute to nearly half of the chronic liver diseases diagnosed in childhood [1]. Genetic testing is vital in the diagnosis process, significantly influencing the disease’s progression and improving the overall quality of life for affected children. Approximately 30% of cases employing whole-exome sequencing (WES) have proven beneficial in reaching a diagnosis [2,3].

Methods: We conducted WES on pediatric patients afflicted with chronic liver diseases.

Results: The examined patient group comprised 45 children (median 5 years, age range 1 month−17 years), consisting of 25 boys and 20 girls. Family history of liver diseases was present in only 5 children, and 13 patients had comorbidities. Liver cirrhosis was diagnosed in 4 children, while idiopathic cholestasis was identified in 5 cases. Additionally, 2 children were found to have alpha-antitrypsin deficiency. Among the remaining 34 patients, unclassified hepatitis was predominant. A genetic cause of the disease was identified in 12 cases, with 6 out of 45 patients (13,3%) exhibiting at least one heterozygous genetic variation. The study revealed 19 distinct pathogenic variants, including 8 novel ones, distributed across 11 different genes (JAG1, ATP7B, ABCC2, ATP8B1, KI12, PKD1, SBDS, ABCB11, ABCB4, HADHA, IFIH1) were identified.

Conclusion: Even in cases with a limited degree of confirmed diagnoses, WES emerges as a valuable tool for a thorough clinical evaluation of patients grappling with liver diseases.

Grants: №075-15-2019-1789.

References:

  1. 1.

    Nicastro E, et al. Liver Transpl. 2018;24(2):282-293.

  2. 2.

    Chen CB, et al. J Pediatr. 2023;258:113408.

  3. 3.

    Chen HL, et al. J Pediatr. 2019;205:153-159.e6.

Conflict of Interest: None declared

EP05.013 TTC21B gene-related nephropathies: Recurrence of the c.626C>T gene variant in Mediterranean populations

Ourayna Batta 1;2, lyahyai jaber3, siham chafai elalaoui4;5, abdelaziz sefiani2;4, imane jaouad cherkaoui4;5

1Faculty Of Medicine And Pharmacy Of Rabat, research team in genomics and molecular epidemiology of genetic diseases, genomics center of human pathologies, Rabat, Morocco; 2National Institute of Hygiene, Medical Genetics department, Rabat; 3Faculty Of Medicine And Pharmacy Of Rabat, research team in genomics and molecular epidemiology of genetic diseases, genomics center of human pathologies; 4Faculty Of Medicine And Pharmacy Of Rabat, research team in genomics and molecular epidemiology of genetic diseases, genomics center of human pathologies, Morocco; 5Ibn Sina CHU. Faculty of Medicine and Pharmacy of Rabat, University Mohammed V, Rabat, Morocco, Medical Genetics Unit, Rabat, Morocco

Background: The TTC21B gene, responsible for encoding the retrograde intraflagellar transport protein IFT139 and initially linked to nephronophthisis, has recently been associated with a homozygous c.626C>T(p.P209L) in families with segmental and focal glomerulosclerosis and tubulointerstitial lesions.

Patients: We report here the case of four Moroccan families: three families with several subjects suffering from early-onset hypertension and end-stage renal failure, and segmental and focal glomerular on renal biopsy. In the fourth family, two siblings were observed with infantile nephronophthisis.

Results: Through clinical exome sequencing, we identified the pathogenic variant c.626C>T (p.P209L) at homozygous state in two families with end-stage renal disease and early-onset hypertension. Additionally, this mutation was found in a composite heterozygous state, paired with another splicing mutation, in the family with infantile nephronophthisis. Furthermore, we identified this homozygous mutation in another family through a targeted sequencing for this variant using conventional Sanger sequencing.

This c.626C>T mutation of the TTC21B gene has already been described, in the homozygous state, in 3 Moroccan families with end-stage renal failure. It has also been reported in North African and Mediterranean families, confirming the founder effect of this pathogenic variant in Mediterranean populations.

Conclusion: We propose to look for this founding mutation, firstly, in families with renal failure of undetermined origin, tubuloglomerular renal lesions, with or without extra-renal manifestations such as high myopia and arterial hypertension at an early age. In instances where this mutation is absent, employing next-generation sequencing becomes imperative to prevent diagnostic uncertainties, enable specific treatment initiation, and facilitate appropriate genetic counseling.

Conflict of Interest: None declared

EP05.014 Association of vitamin D receptor Apa l rs7975232 polymorphism with growth hormone deficiency in children

Mariana Ryznychuk 1

1Bukovinian State Medical University, Department of Pediatrics and Medical Genetic, Chernivtsi, Ukraine

Background/Objectives: to investigate the Apa I polymorphism of the VDR gene in children with GHD

Methods: We studied 36 children with growth hormone deficiency (GHD). To determine polymorphic variants of Apa I (rs 7975232), modified protocols of VDR gene PCR technique with subsequent determination of polymorphism were used.

Results: In the group of patients with GHD, the proportion of A/C genotype carriers is 72.22% (26 children), C/C genotype carriers - 19.45% (7 children), and A/A - 8.33% (3 children).

Allelic analysis in patients with GHD revealed the following data: Carriage of the A allele of the Apa I polymorphic locus (rs 7975232) of the Vit. D VDR gene is significantly associated with the risk of GHD – OR = 1.62 (95% CI 0.97-2.68; p = 0.06).

In patients with GHD and in the presence of A/C and A/A genotypes, the risk of the disease is high OR = 1.68 (95%CI 0.77-3.68; p = 0.19) and OR = 1.58 (95%CI 0.63-3.97; p = 0.33), respectively.

In our study, children with the C/C polymorphism were found to be deficient in vit. D deficiency (44.77 ± 4.05 nmol/L), and in children with A/C and A/A polymorphisms, insufficiency of this vitamin was detected (52.42 ± 5.52 and 57.94 ± 4.49 nmol/L, respectively).

Conclusion: Carriage of the A allele of the rs 7975232 Apa I polymorphic locus of the vitamin D receptor VDR gene is significantly associated with the risk of developing GHD, and in the presence of A/C and A/A genotypes, the risk of this pathology is significantly higher.

Grants: none

Conflict of Interest: None declared

EP05.015 RTL in peripheral blood of CD patients as a potential predictor of the disease outcome

Bojana Stevanovic 1, Ivan Milovanovic2, Marija Kosic3, Branka Popovic1

1Institute of Human Genetics, School of Dental Medicine, University of Belgrade; 23University Children’s Hospital “Tiršova”, Belgrade; 3Institute of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade

Background/Objectives: Crohn’s disease (CD) and ulcerative colitis (UC) are two principal forms of inflammatory bowel disease (IBD). It is well-documented that relative telomere length (RTL) is used as a prognostic tool and Its reduction in intestinal epithelium can initiate inflammation in IBD. Since bowel tissue sampling is a highly invasive diagnostic procedure, our aim was to estimate the prognostic potential of RTL measurement in peripheral blood of IBD pediatric patients.

Methods: Blood and bowel tissue samples were collected from young patients experiencing chronic abdominal pain and swelling symptoms in University Children’s Hospital. Intestinal tissue was submitted to pathohistological analysis for diagnostics and three examination groups were established: control, CD, UC. Peripheral blood leukocytes and bowel tissue were used as a source of DNA for RTL estimation. Relative telomere to single copy gene (T/S) ratio was determined by qPCR. Pearson’s Correlation Coefficient was employed to summarize the strength of linear relationship between blood and tissue samples.

Results: Our results imply a decreasing RTL trend in line with inflammation occurrence, in both blood and bowel tissue of IBD patients compared to controls. Further, a moderate positive correlation (p < 0.01, r = 0.67) between RTL measured in blood and tissue of CD (N = 16) pediatric patients. No parallel correlation could be made in the control (N = 14) and UC (N = 15) group of young patients. Supplementary, sex differences in RTL were not observed.

Conclusion: This study suggests that RTL measurement in blood of CD patients can be used for the prediction of the disease course and severity as less invasive approach.

Grants: No. 451-03-47/2023-01/ 200129

Conflict of Interest: None declared

EP05.016 The landscape of novel causal variants by clinical WES in two siblings with concomitant pseudohypoparathyroidism type 1A and growth hormone insensitivity syndrome with immune dysregulation-2

Sorina Schipor 1, Lidia Radomir2, Andrei Muresan1, Catalina Poalelungi1, Oana Popa1, Andreea Kremer1, Alina Dumitrica1, Luminita Udrea1, Adriana Padure1, Ioana Nedelcu1, Iuliana Gherlan2;3, ELENA EMANUELA BRAHA1

1National Institute of Endocrinology “C. I. Parhon”, Research Department, Bucharest, Romania; 2National Institute of Endocrinology “C. I. Parhon”, Pediatrics Department, Bucharest, Romania; 3University of Medicine and Pharmacy “C. Davila”, Bucharest, Romania

Background. Pseudohypoparathyroidism type 1A (PHP1A) is a rare autosomal dominant inherited disease caused by maternal transmission of GNAS mutations. It is characterized by multihormonal resistance and several phenotypic features. The concomitant appearance of PHP1A with growth hormone insensitivity syndrome with immune dysregulation-2 (GHISID2) due to a heterozygous STAT5B variant has never been reported. Case presentation. We present two affected sisters (6 y.o and 4 y.o) with PHP1A caused by a novel splice mutation c.212+3A > C in exon 2 of GNAS gene, classified as VUS according to ACMG guidelines. They also have a heterozygous variant also classified as VUS in STAT5B gene, c.599G>A (p. Arg200Gln). Patients genomic variants were identified using Illumina Trusight One sequencing kit. The siblings developed a slightly different presentation in clinical condition. Both patients presented with PTH resistance, which is the hallmark of PHP, as well as TSH resistance, severe hypocalcaemia not corrected on Ca supplements and hyperphosphatemia, eczema and low-normal IGF1. Both had language delayed and mild intellectual disability, but the younger sister had significant calcifications on basal nuclei (putamen and globus pallidum). The patients inherited the GNAS variant from their mother, but not the variant in STAT5B gene. Conclusions. Our cases highlight the significance of clinical WES testing in patients with admixed phenotypes for a more precise diagnosis and treatment scheme. This are first clinically and genetically identified PHP1A cases with a novel GNAS gene mutation and concomitant GHISID2 due to a rare STAT5B gene variant in Romania.

Conflict of Interest: None declared

EP05.017 Molecular testing established the diagnosis in a challenging case of Gitelman Syndrome

Lavinia Caba 1, MARIA CHRISTINA UNGUREANU2, Laura Florea3, Andreea Florea1, Eusebiu Vlad Gorduza1

1University of Medicine and Pharmacy “Grigore T. Popa” Iasi, Medical Genetics, Iasi, Romania; 2University of Medicine and Pharmacy “Grigore T. Popa” Iasi, Endocrinology, Iasi, Romania; 3University of Medicine and Pharmacy “Grigore T. Popa” Iasi, Nephrology, Iasi, Romania

Background/Objectives: Gitelman syndrome (GS) is a rare salt-losing tubulopathy, characterized by renal potassium wasting, hypokalemia, metabolic alkalosis, hypocalciuria, hypomagnesemia, and hyperreninemic hyperaldosteronism. The inheritance is autosomal recessive. Patients may experience muscle spasms, cramps, pain caused by chondrocalcinosis, fatigue, polydipsia, polyuria, paresthesias, palpitations. In some cases, they can associate hypostature, thyroid damage (hypothyroidism/hyperthyroidism). We present a case with molecularly confirmed diagnosis of Gitelman at the age of 21 years.

Methods: The patient was clinically diagnosed at 10 years of age with pituitary dwarfism for which he was treated with growth hormone. During her teenage years, she presented marked asthenia, vertigo, polyuria, night sweats, chronic constipation. The patient presented episodes of hyperreninemia, hypokalemia, hypocalciuria, hypomagnesemia. At the age of 20, the clinical diagnosis of salt-losing tubulopathy Bartter Syndrome/ Gitelman Syndrome was discussed and the molecular testing for the genes involved in tubulopathies was done.

Results: Molecular testing in a panel of 39 genes involved in tubulopathies detected a homozygous pathogenic variant in the SLC12A3 gene: c.1924C>G (p.Arg642Gly). Biallelic inactivating mutations in the SLC12A3 gene are associated with Gitelman Syndrome. The result of molecular testing confirms the diagnosis of Gitelman Syndrome in our case.

The patient does not come from a consanguineous family or geographically isolated region.

Conclusion: Genetic testing is important for the diagnosis of Gitelman Syndrome which has clinical and paraclinical signs that overlap with other salt-losing tubulopathy/other diseases. It is also important for the appropriate genetic counseling.

Conflict of Interest: None declared

EP05.018 Implication of vasopressin receptor genes (AVPR1A and AVPR1B) in the susceptibility to Polycystic ovarian syndrome

Pruthvi Goparaju 1, Claudia Gragnoli2;3;4

1Creighton University, Endocrinology, Omaha, United States; 2Creighton University, Endocrinology, Omaha, United States; 3Penn state college of Medicine, Department of Public Health Sciences, Hershey, United States; 4Bios Biotech Multitech Diagnostic Health Center, Molecular Biology Laboratory, Rome, Italy

Background/Objectives: Polycystic ovarian syndrome (PCOS) is a complex heterogenous disorder manifesting with various reproductive, endocrine, and metabolic derangements such as insulin resistance and hyperglycemia. The arginine vasopressin peptide (AVP), also called the antidiuretic hormone (ADH), modulates metabolic functions such as glucose homeostasis, insulin sensitivity, and lipid metabolism via binding to two central and peripheral receptors (AVPR1A and AVPR1B. In the present study, we aimed to detect whether the AVPR1A and AVPR1B genes confer risk for PCOS.

Methods: In peninsular Italian families, we tested 7 single nucleotide polymorphisms (SNPs) in the AVPR1B gene and 2 SNPs in the AVPR1A gene via Pseudomarker for parametric linkage and linkage joint to association (i.e., linkage disequilibrium) with PCOS

Results: We identified 2 risk variants in each gene that were significantly associated with the risk of PCOS.

Conclusion: To the best of our knowledge, this is the first study to report risk variants in AVPR1A and AVPR1B genes in association with PCOS. Replication in other ethnic groups as well as functional studies are needed to confirm these results.

Grants: N/A

Conflict of Interest: None declared

EP05.019 Case report: Triple A syndrome in a Polish siblings carrying a ultra-rare homozygous variant of the AAAS gene

Ewa Juścińska 1, Agata Pastorczak2, Karolina Gadzalska1, Paulina Jakiel1, Monika Gorządek1, Tomasz Płoszaj1, Sebastian Skoczylas1, Maciej Borowiec1, Agnieszka Zmysłowska1

1Medical University of Lodz, Department of Clinical Genetics, Lodz, Poland; 2Medical University of Lodz, Department of Genetic Predisposition to Cancer, Lodz, Poland

Background/Objectives: Triple A syndrome is a rare, multisystem, autosomal recessive disorder characterized by the triad: achalasia, alacrimia and ACTH- resistant adrenal insufficiency. Various neurological symptoms may be present or evolve over time. The incidence of this condition is unknown, but fewer than 100 cases have been published, according to Orphanet. Patients with Allgrove syndrome requires hydrocortisone replacement therapy to reduce the risk of potentially life-threatening adrenal insufficiency and may require esophageal surgery. We report the family with two brothers carrying a homozygous variant of the AAAS gene who presented mainly neurological symptoms.

Methods: Genomic DNA was extracted from a peripheral blood sample (EDTA) on a Maxwell RSC 48 using a semi-automated method (Blood DNA Kit AS1400). WES was performed on a NovaSeq 6000 using a library preparation kit: Twist Human Core Exome and RefSeq and Mitochondrial Panel (Twist Bioscience).

Results: A 40-year-old twins were referred to the Genetic Outpatient Clinic due to similar symptoms including: sensorimotor polyneuropathy, hemiparesis, dysphagia, defective tear formation, muscle weakness and limb muscle abnormalities. The first manifestations were observed in childhood as gait abnormalities. WES revealed in both brothers the presence of a homozygous c.787T>C (p.Ser263Pro) missense variant in the AAAS gene (NM_015665.5) encoding aladin. Bioinformatics analysis of the identified variant confirmed its pathogenicity as recommended by the ACMG.

Conclusion: This report describes the usefulness of WES as a molecular method that plays a key role in finishing the “diagnostic odyssey” process and providing appropriate diagnosis and treatment, especially in patients with multisystem disorders with a wide range of different symptoms.

Grants:

Conflict of Interest: None declared

EP05.020 3D facial gestalt analysis of ADPKD patients

Michaela Mihulová1, Karolina Kočandrlová 1;2, Veronika Moslerova1, Marek Turnovec1, Júlia Martinková1, Milan Macek1, Markéta Havlovicová1, Dana Thomasova1

12nd Faculty of Medicine, Charles University and University Hospital in Motol, Department of Biology and Medical Genetics, Prague, Czech Republic; 2Faculty of Science, Charles University, Department of Anthropology and Human Genetics, Prague, Czech Republic

Background/Objectives: Autosomal dominant polycystic kidney disease (ADPKD) is predominantly caused by variants in PKD1 gene, affecting not only the kidneys but also other vital organs. Molecular genetic analysis of ADPKD patients is often intricate, necessitating innovative diagnostic approaches. The association of PKD1 pathogenic variants with 3D facial gestalt remains unexplored. Here, we present the results of 3D facial morphometry in a cohort of Czech ADPKD patients.

Methods: We enrolled 37 ADPKD cases and conducted facial scanning using the 3dMD Facial System based on stereophotogrammetry. Subsequently, scans were edited in Geomagic Wrap 2021 and analyzed using CPD-DCA in the Morphome3cs software, comparing patients to average 3D facial models of women and men. The study included 25 females (mean age 33y) and 12 males (mean age 23y), all with PKD1 mutations.

Results: Our analysis revealed distinctive 3D facial gestalt features in ADPKD patients. In females, a more protrusive buccal area was observed. The facial profile exhibited increased convexity, accompanied by a more prominent chin. In males, more pronounced supraorbital arches, eyes, and infraorbital regions, along with a prominent lower jaw area was observed. Additionally, in both sexes notable prominence in the nose, particularly in the nasal bridge, was evident.

Conclusion: Our findings suggest the existence of a distinct 3D facial gestalt in ADPKD patients with PKD1 pathogenic variants, which may serve as a diagnostic aid or facilitate risk stratification, particularly in presymptomatic cases.

Grants: Supported by grant 44120 from The Charles University Grant Agency and grant NV19-06-00443 from the Czech Medical Research Council

Conflict of Interest: None declared

EP05.021 Detection of novel homozygous intronic variant in the CFTR gene in families with severe cystic fibrosis

Boodor Al-Kawlani 1, Uros Hladnik1, Nayla Leon Carlos1, Moneeb Othman1, Omid Paknia1, Peter Bauer1

1centogene, rosotock, Germany

Background/Objectives: We describe four patients from two unrelated consanguineous families of Saudi origin who were tested at CENTOGENE.Two siblings and a cousin are from family 1: Patient 1 is the older sister, a three-month-old girl with a history of severe metabolic alkalosis, hypokalemia, hyponatremia, hypochloremia, shortness of breath and chronic diarrhea and excessive sweeting. Patient 2 is the younger sister, a two-month-old girl, and she is similarly affected. The oldest sister died at five-month-old of age due to a chronic disease of vomiting. Patient 3 is the cousin, a two-year-old male, and he has recurrent hypernatremia, hypokalemia and recurrent chest infections. Patient 4 is from family 2, two-year-old girl, with asthma, recurrent lower respiratory tract infections, vomiting, cyanosis and failure to thrive.

Methods: Sanger sequencing was performed for three patients from family 1. NGS pulmonary panel was performed for patient 4 from family 2 using Illumina platform at CENTOGENE.

Results: The analysis identified a novel homozygous intronic variant in the CFTR gene [NM_000492.3:c.3140-10T > G T; p.(?)] in all four affected patients. The mother of the affected siblings of family 1 was a heterozygous carrier. This variant was never described in GnomAD, ClinVar, or HGMD, and is classified as likely pathogenic (PM2_Supporting, PP3, PM3, PP1_Moderate) according to ACMG/AMP ClinGen SVI general recommendations.

Conclusion: The novel intronic CFTR variant is segregated recessively in family 1 and associated with a severe cystic fibrosis. Splice AI predicted a probability of aberrant splicing. It was suggestive of pathogenicity of this variant. RNA studies are needed to assess the effect of the variant.

Conflict of Interest: None declared

EP05.022 Genetic testing for glomerular disease - implications for heterozygous carriers of pathogenic variants in collagen IV genes

Olga Beltcheva 1, Kunka Kamenarova1, Kalina Mihova1, Martin Georgiev1, Galia Zlatanova2, Simona Kerezieva3, Boriana Deliyska3, Maria Gaydarova2, Radka Kaneva1

1Medical University - Sofia, Molecular Medicine Center, Deptartment of Medical Chemistry and Biochemistry, Sofia, Bulgaria; 2Medical University of Sofia, SBAL Pediatric Diseases, Nephrology and Hemodialysis Clinic, Department of Pediatrics, Sofia, Bulgaria; 3Medical University of Sofia, UMHAT “Tsarica Yoanna-ISUL”, Department of Nephrology, Sofia, Bulgaria

Background/Objectives: Mutations in type IV collagen genes are associated with Alport syndrome. While providing patients with homozygous or compound heterozygous variants in COL4A3 and COL4A4, or hemizygous mutations in COL4A5, with genetic diagnosis is pretty straightforward, the status of carriers of single variants in the autosomal genes as well as female carriers in the X-linked gene is more complicated.

Methods: Next generation sequencing (Illumina) was used for screening glomerular disease patients. In addition to the COL4A genes, an extended panel comprising of more than 200 (for the clinical exome, TSO, MiSeq) or more than 700 genes (for WES, NovaSeq 6 000), previously associated with kidney pathologies, was analysed. The pathogenicity of relevant variants was evaluated based on ACMG criteria.

Results: Heterozygous pathogenic or likely pathogenic mutations in the three type IV collagen genes were found in 25 individuals (22 families). Those were both truncating and non-truncating variants, some of them novel. In 3 additional cases the disease-causing variant was accompanied with a variant of unknown significance (VUS) in the same or second COL4A gene and in 6 other – with a VUS in non-collagen gene. The renal phenotype of these carriers varied widely – early and late-onset micro- and macroscopic hematuria, nephrotic syndrome and/or kidney failure.

Conclusion: Despite the possibilities provided by NGS, no second collagen mutation is identified in a significant number of cases with clinically-significant renal disease. This has considerable implications for both counselling and treatment.

Grant references: D01-302/17.12.2021, D01-165/28.07.2022

Conflict of Interest: Olga Beltcheva D01-302/17.12.2021, D01-165/28.07.2022, Kunka Kamenarova D01-302/17.12.2021, D01-165/28.07.2022, Kalina Mihova D01-302/17.12.2021, D01-165/28.07.2022, Martin Georgiev D01-302/17.12.2021, D01-165/28.07.2022, Galia Zlatanova: None declared, Simona Kerezieva: None declared, Boriana Deliyska: None declared, Maria Gaydarova: None declared, Radka Kaneva D01-302/17.12.2021, D01-165/28.07.2022

EP05.023 Potential founder mutation in HESX1 causing congenital hypopituitarism in the Latvian population.

Laura Skrabule 1, Dmitrijs Rots1, Ludmila Voložonoka1, Madara Mašinska1, Iveta Dzīvīte-Krišāne1, Ieva Mičule1

1Children’s Clinical University Hospital, Latvia

Background/Objectives: Congenital hypopituitarism is a rare endocrine disorder characterized by deficient hormone production from the pituitary gland since birth. The protein product of HESX1 plays a pivotal role in the regulation of tissue-specific gene expression, contributing to the differentiation and function of the developing pituitary. Deficiency can result in clinical spectrum including isolated growth hormone deficiency, congenital hypopituitarism, and septooptic dysplasia. It has been implicated in approximately 1% of sporadic septooptic dysplasia cases.

Methods: Six of Latvian congenital hypopituitarism and septooptic dysplasia patients have been analysed by whole exome sequencing.

Results: In three of six patients homozygous HESX1 variant NM_003865.2:c.326G>A, p.(Arg109Gln) was detected. This variant has exhibited a noteworthy prevalence among affected individuals, suggesting a plausible founder mutation specific to the Latvian population. In ClinVar database this variant is described as variant of uncertain significance. We classified as likely pathogenic, as it is reported in individuals with similar phenotype. Based on Gnomad database highest frequency (0,017%) of the variant is in East Asian population. Upon examining data of our internal database of patients, we observed a carrier frequency of 1,2% and an allele frequency of 0,59%, suggesting a plausible founder mutation specific to Latvian population correlating with findings in our hypopituitarism patients.

Conclusion: These findings highlight the widespread presence of the identified variant in Latvia. Emphasizing its significance and potential implications for genetic screening and diagnostic approaches for hypopituitarism cases. Further studies are warranted to define frequency of this variant in congenital hypopituitarism and septooptic dysplasia patients and general population.

Conflict of Interest: None declared

EP05.024 Two novel mutations in patient with congenital hypothyroidism and cardiac abnormalities

Kalina Mihova 1;2, Kunka Kamenarova1;2, Iva Stoeva3;4, Martin Georgiev1;2, Darina Kachakova-Yordanova1;2, Daniela Pencheva1;2, Radka Kaneva1;2

1Medical University of Sofia, Molecular Medicine Center, Department of Medical Chemistry and Biochemistry, Sofia, Bulgaria; 2Medical University of Sofia, Genome Diagnostics Laboratory, Department of Medical Chemistry and Biochemistry, Sofia, Bulgaria; 3University Pediatric Hospital “Prof. Ivan Mitev, Screening and Functional Endocrine Diagnostics Laboratory, Sofia, Bulgaria; 4Medical University of Sofia, Department of Pediatrics, Sofia, Bulgaria

Background/Objectives: Congenital hypothyroidism refers to deficiency of thyroid hormones presenting at birth. Thyroid hormone has essential role in neurodevelopment, growth and energy metabolisms and is one of most common preventable causes of intellectual disability. Unravelling the genetic etiology of the disease is important for the timely diagnosis in children, their treatment, outcome and genetic counselling in affected families.

Methods: A 7-years-old boy, was referred after NTSH screening with hormonal constellation of elevated TSH hormone levels, and decreased levels of T4, hypoplastic orthotopic thyroid gland and coarctation of the aorta and heart murmur with family history of mother with Hashimoto’s thyroiditis and sister with congenital hypothyroidism and short stature. DNA from blood samples was isolated and whole exome sequencing (WES) was performed on Novaseq 6000, Illumina platform.

Results: Genetic analysis found two novel mutations - heterozygous deletion in gene GLI2 (OMIM #165230) c.423delG (p.Met141Ilefs*40), likely pathogenic according ACMG criteria, playing a key role in pituitary development. Second mutation is heterozygous deletion in gene MYRF (OMIM #608329) c.350delG (p.Gly117Alafs*36), likely pathogenic according ACMG criteria, mutations are responsible for autosomal dominant cardiac anomalies. Combination of two mutation reveals the causes of complex phenotype

Conclusions: In this case genetic diagnoses is revealed with two novel likely pathogenic mutations GLI2-c.423delG (p.Met141Ilefs*40) and MYRF-c.350delG (p.Gly117Alafs*36). WES is a powerful tool for detecting mutations in patients with congenital diseases whit complex phenotype, negative in former molecular studies. Therefore in such cases the WES would be in favour for an earlier molecular diagnosis, subsequent personalized treatment and genetic counselling in affected families.

Grant:D01-302/17.12.2021, D01-165/28.07.2022

Conflict of Interest: None declared

EP05.025 Next generation sequencing in molecular diagnosis of cystic fibrosis in Croatian patients

Domagoj Caban 1;2, Ana Merkler Sorgic1, Hana Ljubic1, Ana Acman Barisic1, Petra Anic1, Duška Tješić-Drinković3, Ana Mojsović Ćuić2, Ivna Kocijan2, Martina Ikić4, Ivana Rako1

1University hospital centre Zagreb, Department of Laboratory Diagnostics, Zagreb; 2University of Applied Health Sciences, Department of Biology and Physics, Zagreb, Croatia; 3University hospital centre Zagreb, Department of Pediatrics, Zagreb, Croatia; 4University of Applied Health Sciences, Department of Anatomy and Physiology, Zagreb, Croatia

Background/Objectives: Cystic fibrosis (CF) is a chronic, life-threatening genetic disease caused by loss-of-function mutations in the transmembrane conductance regulator gene (CFTR). CF is one of the most prevalent autosomal recessive inherited conditions in the Caucasian population with an incidence ranging from 1 in 2500 to 1 in 3600. The objective was to perform a complete screening of CFTR gene in Croatian patient population using next generation sequencing (NGS).

Methods: After clinical examination, 50 patients with clinical manifestations suggestive of CF, positive sweat test or suggestive clinical features following the most recent diagnosis of CF guidelines, were tested for CF. For detection of single nucleotide variations (SNVs), indels and quantitative detection of exon spanning copy number variations (CNVs), all appearing in the coding regions and adjacent exon-intron boundaries in the CFTR gene we used Devyser CFTR kit for sequencing on a MiSeq instrument (Illumina).

Results: Fifteen patients were initially screened for 36 known CF variants using multiplex allele specific polymerase chain reaction (MAS-PCR) method (Devyser CFTR Core). The NGS sequencing data correctly confirmed previously detected variants. In the remaining 35 patients we identified seven different CF-causing variants, which four of them we couldn’t identified with previously used method. The most common identified variants was F508del, D1152H and S466X.

Conclusion: Modern NGS technology and variant interpretation enable accurate sequencing-based CF screening which leads to the diagnosis and the possibility of a specific treatment for each patient through precision medicine.

Grants:

Conflict of Interest: None declared

EP05.027 Heterozygous variants in the COL4A4 gene can cause Alport syndrome.

Areej Alatawi 1, Mohammed Almannai2

1king Abdullah specialized children hospital, king Abdullah International medical research center., Genetics and Precision medicine department, Saudi Arabia; 2king Abdullah specialized children hospital, king Abdullah International medical research center., Genetics and Precision Medicine Department, Saudi Arabia

Background/Objectives: Alport syndrome (AS) shows a broad phenotypic spectrum ranging from isolated microscopic hematuria (MH) to end-stage kidney disease. Biallelic disease-causing variants in COL4A3/COL4A4 have been associated with autosomal recessive AS, while the association of monoallelic variants is not well characterized. In this case report, we presented a female with hematuria, massive proteinuria, and focal segmental glomerulosclerosis (FSGS) on renal pathology who had a variant in COL4A4 that was previously reported in individuals with autosomal recessive AS.

Methods: Case report

Results: Herein, we report a case of Alport syndrome in a 38-year-old female with clinical hematuria, massive proteinuria, and FSGS on renal pathology. There is a positive history of ESRD, including her father and two oldest sisters; all of them passed away due to ESRD. A clinical exome and genome were done for her, which reported a heterozygous pathogenic variant in COL4A4, c81_86del p. (Ile29_Leu30del); the same variant was reported before in biallelic form. Segregation analysis of the siblings is under process.

Conclusion: We propose that heterozygous variants in the COL4A4 gene can cause Alport syndrome, albeit less severe than biallelic, but can reach end-stage renal disease. While we don’t have enough data, the penetrance could be incomplete given the reported healthy heterozygous carriers. Possible interactions with other modifier genes or environmental factors could contribute to the phenotype.

Conflict of Interest: None declared

EP05.028 Bi-allelic variants in LIPN1 in two families with sudden death without myoglobinuria

Maryam Mirzazadeh 1, Naser Gilani2, Ariana Kariminejad3

1Kariminejad-Najmabadi Pathology & Genetics Center, Tehran, Iran; 2Farabi Medical Laboratory, Erbil, Iraq; 3Kariminejad-Najmabadi Pathology & Genetics Center, Tehran, Iran

Mutations in the LIPN1 gene play a crucial role in human metabolic traits, contributing to conditions like metabolic syndrome, type 2 diabetes, and early-onset rhabdomyolysis1. LPIN1-related acute recurrent myoglobinuria (OMIM #268200) arises from bi-allelic mutations in LPIN1, representing an autosomal recessive form of rhabdomyolysis characterized by early and recurring episodes, often emerging before the age of 5, posing a life-threatening risk.

Two cases within consanguineous families are presented, both marked by sudden deaths. In the first family, a 3-year-old female child experienced sudden death without a history of previous illness. Whole exome sequencing revealed no pathogenic variants, leading to whole genome sequencing, which identified a homozygous pathogenic 1.8 kb deletion of exon 18 of LPIN1. The first and second child from this family had died at the ages of 2 and 3 years without any previous illness or symptoms associated with myoglobinuria.

In the second case, a 10-year-old asymptomatic child exhibited a homozygous likely pathogenic variant (c.2768+1G > A) in LPIN1 gene. The child’s consanguineous parents carried a heterozygous pathogenic variant in LPIN1 and had previously lost two offspring to sudden death without reported rhabdomyolysis or LPIN1-associated symptoms.

This emphasizes that, while rhabdomyolysis is the primary presentation of LPIN1 gene mutations, there are instances where this symptom is absent. It underscores the importance of considering a broader spectrum of manifestations associated with rhabdomyolysis for accurate diagnosis and management. A comprehensive understanding of varied clinical presentations is crucial, emphasizing the need to explore a wider range of symptoms associated with LPIN1 gene mutations

Conflict of Interest: None declared

EP05.029 Prediction of diabetes type 1 and diabetes type 2 in Eastern European population using polygenic risk scores.

Layal Shaheen 1;2, Anna Kim1, Elena Kovalenko1, Dmitrii Kharitonov1, Valeriya Rubinova1, Alexander Rakitko1

1Genotek, Moskva, Russian Federation; 2Moscow Institute of Physics and Technology, Dolgoprudny, Russian Federation

Background/Objectives: Type 1 (T1D) and Type 2 (T2D) diabetes pose significant health risks if not promptly detected and managed. In our study we employ polygenic risk score (PRS) approach to facilitate early detection of T1D and T2D in the Eastern European population to improve long-term health outcomes.

Methods: We conducted genome-wide association studies (GWAS) to identify genetic mutations associated with T1D and T2D in a cohort of Eastern European descent. The study included 1030 T2D cases and 7758 controls aged 40-80 years, and 421 T1D cases and 4040 controls aged 8-40 years. PRS calculations were performed using LDpred and PRSice software.

Results: The T1D GWAS revealed significant associations with the following genes: BCL2L15, MUCL1, INS, TAP2, HLA-DQA1, and HLA-DQB1. For T1D, PRSice yielded AUC of 0.73, where for bottom 10% of PRS strata the OR compared to the median is 0.27 (CI: 0.12-0.6), and the top 10% has OR of 5.1 (CI: 3.3-7.9). LDpred2 best model AUC = 0.76.

For T2D, PRSice PRS has AUC = 0.6 with OR = 1.92 (CI: 1.5-2.5) of the top 10% compared to the median, LDpred2 with a grid model had AUC of 0.62, outperforming other models.

Conclusion: Our findings underscore the utility of PRS in the early detection of T1D and T2D within the Eastern European population, with LDpred2 demonstrating superior performance in risk assessment. These insights could guide personalized interventions and contribute to better management strategies for diabetes in underrepresented populations.

Conflict of Interest: None declared

EP06 Skeletal, Connetive Tissue, Ectodermal and Skin Disorders

EP06.001 Clinical case of multiple hereditary exostoses: identification of a major pathogenic variant in the EXT2 gene

Alexandra Yakovleva 1, Leonid Zhozhikov1, Roza Ivanova1;2, Aitalina Sukhomyasova1;2, Nadezhda Maksimova1

1Research Laboratory of “Molecular Medicine and Human Genetics”, Institute of Medicine, North-Eastern Federal University, Yakutsk, Russian Federation; 2Medical Genetic Center, Republic Hospital No1 – National Center of Medicine

Background/Objectives: Multiple hereditary exostoses type I and type II are autosomal dominant diseases associated with variants in the EXT1 (OMIM *608177) and EXT2 (OMIM *608210) genes, respectively. A clinical case of a Yakut family with multiple exostotic chondrodysplasia is presented.

Methods: The proband is seven years old. Phenotypic signs comprise multiple exostoses in the humerus, elbow, femur, and tibia; arthralgia of the elbow, shoulder, and knee joints. X-rays of the knee joints revealed multiple osteochondral exostoses of the femur and tibia of both lower extremities. From the family history, it is known that the father and grandfather also exhibit the exostoses phenotype. A child is from the 3rd pregnancy, which proceeded without complications—delivery at term, birth weight 4,1 kg, length 52 cm. The disease manifested during the patient’s second year. When he was six years old, he began to complain of pain in his arms and legs when walking.

Results: Previously we identified candidate variants in the EXT1 and EXT2 in the Yakut population. We searched for the causative variant using Sanger sequencing which revealed a frameshift variant c.310delA, p.Ile104fs*8, ENST00000533608 (c.409delA, p.Ile137fs, NM_000401.3) of EXT2 in the proband and father.

Conclusion: In our previous studies, this variant was identified in 12 families. Therefore, we can make a presumption that this pathogenic variant is possibly major for the Yakut ethnic group.

Grants: This work was supported by the Ministry of Science and Higher Education of the Russian Federation, “Genomics of the Arctic: epidemiology, heredity and pathology” (No. FSRG-2020–0014).

Conflict of Interest: None declared

EP06.002 A novel missense DSG1 mutation in a patient with SAM syndrome

Zeynep Münteha Başer 1, Ceren Alavanda2, Vedat Yüce1, Esra Hilal Ceylan1, Ayşe Deniz Yücelten3, Bilgen Bilge Geçkinli1

1Marmara University Hospital, Medical Genetics, Istanbul, Türkyie; 2Health Sciences University Van Education and Research Hospital, Medical Genetics, Van, Türkyie; 3Marmara University Hospital, Dermatology, Istanbul, Türkyie

Background/Objectives: Desmoglein 1 protein, encoded by DSG1 gene, is a major glycoprotein component of desmosome which forms cell adhesion proteins with desmocollins. While heterozygous DSG1 mutations cause striate palmoplantar keratoderma type 1 (MIM:#148700), biallelic mutations cause severe dermatitis, multiple allergies and metabolic wasting (SAM) syndrome (MIM:#615508).

Methods: After DNA isolation from peripheral blood Clinical Exome Sequencing (CES) was performed by next-generation sequencing (Illumina Nextseq 500).

Results: A 10 -year- old girl was directed to our clinic with diffuse erythroderma, anhidrosis, scaling and growth retardation. She was born to consanguineous parents at 36th weeks of gestation via C/S and hospitalized for 10 days due to skin lesions. She didn’t have recurrent infection history. She was allergic to packaged foods. Her neuromotor developmental milestones were normal. At the time of application her weight was 26 kg (3 P), height was 124 cm (<3 P) and head circumference was 50 cm (75-90 P). Generalized erythroderma, scaling, hypotrichosis, hyperkeratotic lesions under her feet were noted. Her blood tests showed increased IgE. Other system evaluations were normal. Molecular analysis of CES revealed a novel homozygous c.909G>C p.(Trp303Cys) variant in the DSG1(NM_001942.4) gene. The variant was considered as likely pathogenic according to ACMG criteria. Segregation analysis of the parents was planned.

Conclusion: Here, we report the clinical findings of a patient with a novel variant in the DSG1 gene. As the clinical findings can be reminiscent of Netherton Syndrome, genetic testing is important for accurate diagnosis.

Grants: None.

Conflict of Interest: None declared

EP06.004 A family of LBR biallelic variants resulting in rhizomelic skeletal dysplasia with pelger-huet anomaly

Esra Dirimtekin 1, cekdar kapazan1, ahmet mert yanik2, barıs yılmaz3, Bilgen Bilge Geçkinli1

1Marmara University Faculty of Medicine, Department of Medical Genetics, istanbul, Türkyie; 2Marmara University Faculty of Medicine, Department of Hematology, istanbul, Türkyie; 3Marmara University Faculty of Medicine, Department of Pediatric Hematology, istanbul, Türkyie

Background: Rhizomelic skeletal dysplasia with or without Pelger-Huet anomaly (OMIM #618019) (SKPHA) is characterized by rhizomelic skeletal dysplasia of variable severity with or without abnormal nuclear shape and chromatin organization in blood granulocytes. The lamin-B receptor gene (LBR; NM_002296) encodes a bifunctional LBR protein. Heterozygous pathogenic variants of LBR cause Pelger-Huet anomaly (PHA) (OMIM #169400), while biallelic mutations cause a spectrum of rare skeletal dysplasias.

Method: Genomic DNA was extracted from peripheral blood. We sequenced skeletal dysplasia panel of 88 genes with Next Generation Sequencing (NGS). Family segregation analysis were performed using Sanger sequencing.

Case: A 4-year-old boy was referred to our medical genetics clinic because of short stature and short limbs. The mother had milder symptoms. He was born to consanguineous parents at 36 weeks of gestation with a birth weight of 2400 gr (SDS-0,81) and length of 43 cm (SDS-2,05). He had severe short stature (height: 94cm, SDS-3,17), low weight (weight: 14 kg, SDS-1,85) and microcephaly (head circumference: 46 cm, SDS-3,39). Physical examination showed frontal bossing, small-narrow thorax, rhizomelic shortening of limbs. X-rays showed short and horizontal ribs, short, bowed radii, humeri and ulnae and widened and irregular metaphyses. Previously reported heterozygous c.1640A>G (p.Asn547Ser) variation and heterozygous c.43C>T (p.Arg15*) variation was detected in LBR. Mother was compound heterozygous and father heterozygous carrier for c.43C>T (p.Arg15*) variation. Peripheral blood smears revealed PHAin the index case and his parents.

Conclusion: Here, we report a family with biallelic and heterozygous variation of LBR to contribute to genotype -phenotype associations.

Conflict of Interest: None declared

EP06.005 Whole exome sequencing as a tool in the diagnosis of ectodermal dysplasias : a case report

Alexandra Fountoulaki 1, Georgia Christopoulou2, angeliki erakleous1, maria kassotaki1, ILIANNA MANIADAKI1, Pantelis Constantoulakis2, Eleftheria Papadopoulou1

1University Hospital of Heraklion, Pediatrics, Heraklion, Greece; 2, GENOTYPOS Science Labs, Athens, Greece

Background/Objectives: Ectodermal dysplasias (ED) is a group of approximately 180 disorders with structural and functional abnormalities in various tissues originating from the ectodermal layer. Despite some of the syndromes having different genetic causes, the symptoms are sometimes very similar. Hypohidrotic ED (HED) is characterized by hypohidrosis, hypotrichosis, and hypodontia, with estimated incidence 1-9:100,000. We present a case of X-linked HED (XLHED), which was diagnosed by Whole Exome Sequencing (WES).

Methods: A 2.5-year-old male was referred to a pediatrics hospital unit for the investigation of heat intolerance and episodes of hyperpyrexia. Physical examination revealed peculiar facies including loss of eyelashes and eyebrows, periorbital hyperpigmentation, oligodontia with few peg-shaped teeth, depressed nasal bridge, everted lower lip and sparse, thin, light-colored scalp hair. Nails and physical growth were normal, with delayed speech development. The sweat test showed anhidrosis. Proband’s mother and maternal grandmother are reported to have isolated hypodontia. WES (Twist Bioscience) was applied on DNA extracted from peripheral blood. Bioinformatics analyses were conducted by SOPHiA DDM® pipelines.

Results: WES revealed a hemizygous, EDA variant; c.895G>A. This is located in a mutational hot-spot and a different variant at the same codon is classified as pathogenic. It is absent from GnomAD and detected in patients with similar phenotype. Therefore, it is classified as pathogenic leading to genetic diagnosis.

Conclusion: WES consists a tool for the identification of the molecular etiology of ectodermal dysplasias. Early diagnosis of XLHED is essential for the patient’s early management, to prevent hyperpyrexia, to restore dental defects and to provide genetic counseling to the family.

Conflict of Interest: None declared

EP06.006 A novel uncommon copy number variations in the 11p15.5 imprinting region causing Beckwith–Wiedemann and Silver–Russell Syndrome.

Łukasz Przesór 1, Małgorzata Piotrowicz1, Urszula Wysocka1, Iwona Pinkier1, Agata Kucińska1, Dominika Komorowska1, Wanda Hawuła1, Tadeusz Kałużewski1, Łukasz Kępczyński1, Agnieszka Gach1

1Institute of Polish Mother’s Health Center, Department of Clinical Genetics, Łódź, Poland

Consortium: Departament of Genetics, Mother’s Memorial Hospital Research Institute, Łódź, Poland

Background/Objectives: Defects of 11p15.5 imprinting result in two growth disorders with opposite phenotypes: Beckwith–Wiedemann syndrome (BWS) and Silver–Russell syndrome (SRS). One is associated with overgrowth, the other with growth retardation. This region is organized in two imprinted domains (IGF2/H19 and KCNQ1OT1/CDKN1C).

Here we report uncommon copy number variation of 11p15.5 region, responsible for growth abnormalities consistent with BWS and SRS diagnosis.

Methods: SRS case patient was 1 year old female, presenting intrauterine growth retardation, low birth weight, feeding difficulties, and dysmorphic features and fifth finger clinodactyly. The aberration was identified by commercially available MS-MLPA test, and was further confirmed by aCGH testing. BWS case was a 1 year old male, presenting with cholestasis, abnormal muscle tonus, overgrowth, inguinal hernia and cryptorchidism. The aberration was identified by whole exome sequencing, the methylation status and parental origin was confirmed by MS-MLPA.

Results: MS-MLPA in SRS case revealed reduced methylation levels in the H19 gene region and CNV affecting IGF2 (duplication) and partially H19 (triplication). Three copies of IGF2 and four copies of H19 have been noticed. In BWS case the aberration included whole imprinted region.

Conclusion: Changes in copy number involving the 11p15.5 region may be associated with an abnormal phenotype, and their effect depends on the location, size, parental origin, and methylation status. BWS/SRS locus duplication is a rare cause of Beckwith-Wiedemann syndrome. To our best knowledge the IGF2 duplication with partial H19 triplication with clinical features of Silver-Russell syndrome is here described for the first time.

Grants: None

Conflict of Interest: None declared

EP06.007 AXIN1-related osteosclerosis in a young adult – a case report and phenotypic expansion of a novel disorder

Eyal Elron 1, Naama Orenstein1;2, lina basel-salmon1;2;3, Nurit Assia Batzir1

1Schneider- Children’s Medical Center, Pediatric Genetics Unit, Petah Tikva, Israel; 2Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel; 3Felsenstein Medical Research Center, Israel

Background/Objectives: AXIN1 encodes a cytoplasmic protein that plays a crucial role in several biological processes. It has a regulatory role in the Wnt-beta-catenin signaling pathway, influencing embryonic development and tissue regeneration and maintenance, including bone homeostasis. Recently, homozygous truncating variants in AXIN1 were recently implicated by Terhal et al. (2023) in craniometadiaphyseal osteosclerosis with hip dysplasia.

Methods: Trio exome sequencing was performed for a 17 year-old female presenting with sclerosing bone dysplasia, bilateral optic disc coloboma, short stature, precocious adrenarche with polycystic ovaries and hypertension. A head CT performed at the age of 14 years revealed a increased density of the cranial bones, vertebrae and long bones, with abnormal fusion of the C1 vertebra. On a skeletal survey, an Erlenmeyer flask deformity was noted in both femurs and distal forearms.

Results: A homozygous in-frame deletion variant in AXIN1 was identified in the patient: NM_003502.4:c.2468_2470del; p.Tyr823del. This variant is absent from gnomAD and affects the DIX domain of AXIN1 which has been shown to play an important part in homodimerization. The variant was classified as a variant of uncertain significance leaning towards likely pathogenic.

Conclusion: This is the second report of autosomal recessive osteosclerosis associated with AXIN1, broadening the genotypic and phenotypic spectrum.

Grants: No grants

Conflict of Interest: None declared

EP06.008 ERF-related craniosynostosis in a patient with hypochondroplasia: a rare coincidence

Pelin Simsek-Kiper 1, Merve Soğukpınar1, Beren Karaosmanoglu2, Gozel Jumayeva3, gizem ürel-demir1, rahsan gocmen4, Gulen Eda Utine1

1Hacettepe University Faculty of Medicine, Pediatric Genetics, Ankara, Türkyie; 2Hacettepe University Faculty of Medicine, Medical Genetics, Ankara, Türkyie; 3Hacettepe University Faculty of Medicine, Pediatrics, Ankara, Türkyie; 4Hacettepe University Faculty of Medicine, Radiology, Ankara, Türkyie

Background/Objectives: Hypochondroplasia is a FGFR3-related disorder characterized by short-limbed severe short stature. Craniosynostosis is not among the typical clinical findings of hypochondroplasia. Craniosynostosis, a genetically heterogenous condition, is a primary abnormality of skull growth involving premature fusion of the cranial sutures. ERF mutation is one of the most recently identified genetic mutations associated with syndromic craniosynostosis (OMIM# 600775).

Methods: We describe a 6-year-old patient who was referred to our centre with the findings of dysmorphic facial features and proportionate short stature. The molecular etiology was investigated with exome sequencing after informed consent was taken from the parents.

Results:The physical examination revealed short-limbed short stature and dysmorphic findings including frontal bossing, dolicocephaly, midface hypoplasia, bilateral proptosis, low-set ears, depressed nasal root, high palate, pectus carinatum, narrow thorax, pes planus, and brachydactyly. He had moderate intellectual disability. The radiographic findings were compatible with craniosynostosis and hypochondroplasia. EEG detected epileptiform anomaly and cranial MRI displayed developmental cortical anomaly, diffuse polymicrogyria, and hypo-dysplasia of corpus callosum. Exome sequencing revealed a previously reported pathogenic heterozygous FGFR3 (c.1626C>G) and ERF (c.256C>T) variants in the patient. While FGFR3 variant was de novo, segregation analysis revealed the same ERF variant in the mother and maternal uncle with similar facial features.

Conclusion: Hypochondroplasia and craniosynostosis are rare clinical entities that require appropriate diagnosis and management. Additional genetic etiologies should be considered in patients with atypical findings. Exome sequencing aids in the identification of rare coincidental genetic disorders.

Conflict of Interest: None declared

EP06.009 Novel deep-intronic variant in ATP2C1 as the cause of Hailey-Hailey disease

Jenny Blechingberg 1, Thorkild Terkelsen1;2, Uffe B Jensen1, Mette Sommerlund3, Hanne Vinter4, Lise Graversen1

1Aarhus University Hospital, Department of Clinical Genetics, Aarhus N; 2Aarhus University, Department of Biomedicine, Aarhus C, Denmark; 3Aarhus University Hospital, Department of Dermatology, Aarhus N; 4Aarhus University Hospital, Department of Pathology, Aarhus N

Background/Objectives: Hailey-Hailey disease (HHD) is an autosomal dominantly inherited genodermatosis characterized by skin blisters in the flexural areas of the body, that change into painful plaques. HHD is caused by mutations in ATP2C1, but for around 10% of patients, no causative variant is found in the coding region of ATP2C1. We present a family with three affected family members in two generations. No genetic cause was found in the coding region of ATP2C1, but a deep-intronic variant, likely to be causative, was identified.

Methods: Whole genome sequencing of DNA from whole blood was done. Sequencing data of all exon and intron sequences of ATP2C1 were analysed.

Results: In intron 7 of ATP2C1, the variant c.532-560T > G (NM_014382.5) was identified, and predicted by in silico prediction tools to create a new deep intronic donor splice site. The variant is not present in frequency databases containing variants in the general population, or in databases containing pathogenic variants. Segregation analysis has identified the variant in the three affected family members. Functional studies of the effect of the variant upon RNA splicing of the ATP2C1 messenger RNA are currently awaiting.

Conclusion: We have identified a deep-intronic variant in ATP2C1 likely causative of HHD. This is the first report of a deep-intronic variant as the cause of HHD and shows that whole genome sequencing with analysis of the intronic sequences could be adopted to increase the genetic diagnostic yield for HHD patients.

Grants: Aage Bangs Fond, Aase og Ejnar Danielsens Fond

Conflict of Interest: None declared

EP06.010 Unclassical OI like manifestations caused by COL1A variants: New insights on C1ROD classification

Nehal Hassib 1, rasha elhossini2, Inas Sayed3, Mohamad Abdelhamid3

1National research centre, Giza, Egypt; 2National research centre, clinical genetics, Giza, Egypt; 3National research centre, Orodental genetics, Giza, Egypt

Abstract

Background/ObjectivesCollagen type 1(Col1) is the principal connective tissue forming the body skeleton, the sclera, the skin, and the teeth. Collagen type 1 is a protein fundamentally formed of alpha 1 and alpha 2 chains. Pathogenic variants in COL1A1/2 are responsible for various bone disorders presented as osteogenesis imperfecta (OI) or Ehler danlos syndrome (EDS). This report presents eighteen patients from four unrelated families, with a homozygous COL1A1 variant in one family and heterozygous COL1A1/2 variants in three families by Whole exome sequencing (WES). Additionally, a new insight on COL1-related overlap disorder (C1ROD)” has been conveyed.

Methods: Clinical manifestations in all the studied patients resemble osteogenesis imperfecta (OI) in the presence of variable degrees of osteoporosis, bone deformities, gray sclera, wormian bones in the skull and oro-dental abnormalities. However, no bone fractures were documented in any of these cases, which is a cardinal feature in OI patients. They also lack the characteristics of EDS.

Results: COL1A1/2 variants were the only possible cause justifying their clinical manifestations.

Conclusion: Thus; we suggest expanding COL1-related overlap disorder (C1ROD)” to include OI like disease in addition to the previously reported EDS like disease. Moreover, we present a new patient harboring a homozygous COL1A1 gene variant with distinctive phenotype in comparison to the few previously reported patients.

Grant: This work was funded by a research grant from the Science and Technology Development Fund (STDF), Academy of Science Research and Technology, Egypt (Grant number: 33458).

Keywords: Dentinogenesis imperfecta, COL1A1, COL1A2, C1ROD, bone fragility, osteoporosis

Conflict of Interest: None declared

EP06.012 MSX1 variant in Japanese nonsyndromic oligodontia case’

Junya Adachi 1, Yoshihito Tokita2, Junichiro Machida3, Yoshihiko Aoki4, Michiyo Ando5, Yasuto Sano6, Mitsuo Goto5, Atsuo Kaetsu1, Takamasa Shirozu1, Eriko Osumi1, Michiko Matsuoka1, Nasir Maeda1

1Toyohashi Municipal Hospital, Toyohashi, Japan; 2Aichiken Iryoryoikusogo Center Hattatsushogai Research Institute, Kasugai, Japan; 3Toyota Memorial Hospital, Toyota, Japan; 4Okazaki Municipal Hospital, Okazaki, Japan; 5Aichi-Gakuin University Dental Hospital, Nagoya, Japan; 6Aichi Gakuin University Dental Hospital, Nagoya, Japan

Background/Objectives: Congenital tooth agenesis is a common human anomaly which is called oligodontia, when the number of missing teeth is 6 or more. Although a series of genetic studies have revealed the causative genes of human tooth agenesis, the etiology remains largely unknown. The MSX1 gene, a member of the homeobox gene family, encodes a DNA-binding protein, which is involved in many epithelial-mesenchymal interactions, leading to vertebrate organogenesis, and appears to be most critical during early tooth development.

The aim of the present study was to identify genetic clues for a 19-year-old female with 14 missing teeth, without systemic abnormality.

Methods: Mutational analysis by whole-exome sequencing was performed to identify the cause of oligodontia in a Japanese family.

Results: The heterozygous MSX1 gene variant c.434G>A was identified in patients. It is a nonsense variant and produces the C-terminal cleavage gene product p.Trp145*, which lacks a homeodomain.

Conclusion: It has previously been demonstrated that the homeodomain (amino acids 175–229) plays a pivotal role in molecular interactions with DNA and other transcription factors related to tooth development. Functional analysis suggested that the variant of MSX1 lost its function as a transcription factor due to the loss of the homeodomain, causing tooth agenesis.

Grants: This work was supported in part by AMED under grant numbers JP17nk0101334 and JP20ek0109397 (to YT).

Conflict of Interest: None declared

EP06.013 Japanese Cleidocranial Dysplasia family diagnosed by molecular genetic analysis

Yoshihiko Aoki 1, Junichiro Machida2, Junya Adachi3, Michiyo Ando4, Yasuto Sano4, Terumi Saito1, Mitsuo Goto4, Yoshihito Tokita5

1Okazaki City Hospital, Oral and Maxillofacial Surgery, Okazaki, Japan; 2Toyota Memorial Hospital, Oral and Maxillofacial Surgery, Toyota, Japan; 3Toyohashi Municipal Hospital, Oral and Maxillofacial Surgery, Toyohashi, Japan; 4Aichi Gakuin University of Dentistry, Maxillofacial Surgery, Nagoya, Japan; 5Aichi Developmental Disability Center Institute for Developmental Research, Disease Model, Kasugai, Japan

Background/Objectives: The classic of cleidocranial dysplasia (CCD) spectrum disorders includes delayed closure of the cranial sutures, hypoplastic or aplastic clavicles, and dental abnormalities, though they range from mild CCD to those with no skeletal abnormalities. The gene responsible for CCD is the runt-related transcription factor 2 (RUNX2) gene. A 11-year-old girl was clinically diagnosed with CCD because of short stature, open fontanelles, and delayed eruption of permanent teeth. The purpose of this study is to determine the cause of the disease by molecular genetic analysis in this family.

Methods: Saliva samples were collected from the family members, and genomic DNA was purified. The exon region of the RUNX2 gene was sequenced from the genomic DNA. From the mutation information obtained, wild-type and mutant vectors were constructed and functionally analyzed.

Results: The c.614C>G nucleotide substitution in the RUNX2 gene was found in the affected patient and her father. This mutation caused an amino acid substitution of p.T205R in the runt domain of the RUNX2 gene. Wild-type RUNX2 showed concentration-dependent activity, while mutant RUNX2 was found to be dysfunctional.

Conclusion: We identified a missense mutation in RUNX2 as the cause of CCD in this case. Most of the mutations disrupt the runt domain, which is critical for the biological function of the RUNX family of transcription factors. It is important to establish early diagnosis in these patients in order to offer a better quality of life.

Grants: This work was supported in part by AMED under grant numbers JP17nk0101334 and JP20ek0109397 (to YT).

Conflict of Interest: None declared

EP06.014 New Insight into the Genotype-Phenotype Correlation of PTH1R variants and Primary Failure of tooth eruption on an Italian Cohort and Literature Review

Clarissa Modafferi 1, Elisabetta Tabolacci1, Arcangelo Fargnoli1, Ilaria Cassano1, Roberto Bertozzi1, Cristina Grippaudo2, Alessandro Arcovito3, Filomena Lo Vecchio1, Ettore Lo Cascio2, Pietro Chiurazzi1

1Gemelli, Dipartimento di Scienze della Vita e Sanità Pubblica, Roma, Italy; 2Gemelli, Department of Head and Neck, Division of Oral Surgery and Implantology, Roma, Italy; 3Gemelli, Dipartimento di Scienze Biotecnologiche di Base, Roma, Italy

Background/Objective: Primary failure of tooth eruption (PFE) is an autosomal dominant disease with penetrance defect. We established clinical criteria to better identify PFE patients carrying PTH1R gene variants. In order to better understand the genetic causes of this disease we examined a new cohort of 32 patients, including 7 familial cases, referred to have PFE from our Hospital and from external collaborators. PTH1R gene was analysed through Sanger sequencing, then we evaluated the possible impact of single variants on the clinical phenotype using online databases and in silico prediction tools.

Methods: DNA was collected from patients using cytobrushes. Nucleotide sequencing of intronic/exonic PTH1R candidate regions (NM_000316.3) was performed using ABI3500 Genetic Analyzer.

Results: Sequencing analysis of the PTH1R coding sequence in a cohort of 32 patients revealed 9 different variants, 4 exonic and 5 intronic. Through in silico prediction tools and databases, 3 of them (1 exonic, 1 intronic and 1 in a splicing site) has been considered potentially involved in the PFE phenotype. Sequencing of cDNA was unsuccessfully attempted due to the low levels of PTH1R expression in the different tissues analyzed.

Conclusions: The yield of the genetic test increases when the clinical selection of the patients with PFE is well-characterized. Dental eruption failure with pure clinical findings of PFE associated with familial history revealed variants in PTH1R gene, offering a diagnostic test for the family.

Conflict of Interest: None declared

EP06.015 Homozygosity for a Dominant Novel NPR2 Splice-Altering Variant in a Turkish Family

irem kalay 1;2, Barbara Vona2;3

1Sağlık Bilimleri Üniversitesi Ümraniye Eğitim ve Araştırma Hastanesi, Department of Medical Genetics, Istanbul, Türkyie; 2Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany; 3Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany

Background: NPR2 encodes natriuretic peptide receptor, a regulator of skeletal growth. Homozygous and/or heterozygous loss-of-function variants in NPR2 are associated with acromesomelic dysplasia, Maroteaux type (AMDM), or short stature with nonspecific skeletal abnormalities. Gain-of-function variants result in epiphyseal chondrodysplasia, Miura type, which is an overgrowth disorder. This study explores the clinical characteristics of individuals in a family segregating a novel splice-altering variant in heterozygous and homozygous states.

Methods: The proband underwent evaluation using a virtual panel with 131 skeletal dysplasia genes applied to Clinical Exome Sequencing data. A candidate homozygous putative splice-altering variant was detected and confirmed by Sanger sequencing. The variant underwent segregation testing in family members with short stature.

Results: We identified a novel homozygous intronic NPR2 variant (NM_003995: c.1218+4A > G) in the proband that has an AMDM phenotype. Her sisters and parents were heterozygous and presented only short stature without other dysmorphic features. The c.1218+4A > G variant is absent in gnomAD and multiple splice prediction tools predict it is likely to abolish the splice donor site of exon 5.

Conclusion: This study highlights the importance of genetic testing in individuals with skeletal dysplasias. The splice-donor variant c.1218+4A > G causes a loss of C-type natriuretic peptide-dependent cGMP generation capacities. With this study, we introduce a novel likely pathogenic variant in intron 5 of NPR2 in a Turkish family. Finally, we present an unusual circumstance of a family segregating both dominant and recessive modes of inheritance with an opportunity for deep phenotyping.

Conflict of Interest: None declared

EP06.016 Novel variant c.148A>G, p.(Lys50Glu), in the NFIX gene linked to Malan syndrome

Lina Al Qadi 1, eric allain2;3;4;5, Haley McConkey6;7, Bekim Sadikovic6;7, Mouna Ben Amor1;8;9

1Centre de formation médicale du Nouveau-Brunswick, Université de Sherbrooke, Moncton, NB, Canada; 2Vitalité Health Network, Dr. Georges-L.-Dumont University Hospital Centre, Department of Molecular Genetics, Moncton, NB, Canada; 3Atlantic Cancer Research Institute, Moncton, NB, Canada; 4Université de Moncton, Department of Chemistry and Biochemistry; 5New Brunswick Center for Precision Medicine, Moncton, NB, Canada; 6Western University, Department of Pathology and Laboratory Medicine, London, Ontario, Canada; 7London Health Sciences Centre, Verspeeten Clinical Genome Centre, London, Ontario, Canada; 8Vitalité Health Network, Dr. Georges-L.-Dumont University Hospital Centre, Department of Medical Genetic, Moncton, NB, Canada; 9Université de Sherbrooke, Department of Pediatrics, Sherbrooke, QC, Canada

Background/Objectives: Heterozygous pathogenic variants in the NFIX gene are linked to Malan Syndrome characterized by postnatal overgrowth, gestalt, skeletal defects, developmental delay/intellectual disability. Approximately, 90 affected individuals have been documented. We report an 18-year-old female with a syndromic overgrowth and a de novo likely pathogenic variant in the NFIX gene with an unusual epigenetic result.

Methods: The investigation included microarray, metabolic workup, fragile X, and EpiSignTM DNA methylation analysis, followed by a targeted exome. The parents were offered targeted genetic testing.

Results: Micro-array, metabolic workup, and fragile X were normal. EpiSignTM testing unexpectedly revealed methylation changes suggestive of Beckwith Weideman syndrome (BWS), and borderline multilocus imprinting disturbances. Clinical testing for BWS was negative. Targeted exome revealed a heterozygous variant of uncertain significance in the NFIX gene: c.148A>G, p.(Lys50Glu). Parental testing confirmed de novo inheritance. Based on the clinical phenotype and the characteristics of the novel variant, it has been reclassified to likely pathogenic.

Conclusion: We report a novel likely pathogenic variant in a patient with Malan syndrome. Our patient also demonstrated abnormal methylation at multiple ICRs, suggestive of low level of mosaicism for BWS in the absence of BWS clinical features. To the best of our knowledge, these two molecular findings had not been previously reported and the relationship is unclear. It’s an interesting observation that can be incidental or a subject for future research that might shed light on NFIX gene and epigenetic mechanisms.

Grants: Genome Canada GAPP grant (OGI-188). Centre de formation médicale du Nouveau Brunswick. Research NB

Conflict of Interest: None declared

EP06.017 Male patient with a novel likely pathogenic variant in LRP4 causing Sclerosteosis 2

Juliane Köhler 1, Malte Spielmann1;2, Nadine Hornig1

1University of Kiel, Institute of human genetics, Kiel, Germany; 2University of Lübeck, Institute of human genetics, Lübeck, Germany

Background/Objectives: Sclerosteosis is an extremely rare skeletal dysplasia featuring a diffusely radiodense skeleton (SOST2; #614305). It is transmitted in an autosomal dominant or autosomal rezessive manner. So far one heterozygous and two homozygous loss-of-functions variants in LRP4 are known to cause SOST2. LRP4 encodes for a sclerosin interaction protein the low-density lipoprotein receptor-related protein 4. LRP4 mRNA is, especially during embryogenesis, expressed in many tissues. The sclerostin protein expression has been reported only postnatally in terminally differentiated cells enveloped with a mineralized matrix, such as osteocytes, mineralized hypertrophic chondrocytes and cementocytes.

Methods: We acquired the patient’s family history and clinical data. Chromosome banding analysis, array-CGH and exome sequencing were performed using blood samples. The disease’s severity of our patient was assessed and we did literature research for similar patients.

Results: The patient presented with increased stature and head circumference, frontal bossing and broad clavicles. Some fingernails and toenails seemed dysplastic. He also presented with motor delay. Chromosome banding analysis and array-CGH showed unremarkable findings. Whole-exome sequencing revealed a heterozygous likely pathogenic variant in LRP4 (NM_002334.4:c.4693C>T; p.Gln1565*) predicted to lead to a stop gain at position 1565 of in total 1905 amino acids. The variant was not present in the gnomAD database. A segregation analysis and X-rays are pending.

Conclusion: Here we report an additional patient with Sclerosteosis and a novel heterozygous stop gain variant in LRP4. The variant has so far not been described. Our case report emphasizes the heterogeneity and the variability of the phenotype of variants in LRP4.

Conflict of Interest: None declared

EP06.018 Expanding the phenotypic spectrum of Smith-McCort dysplasia 2

Valerie Katharina Berge 1;2, Nadine Hornig3, Yorck Hellenbroich1, Malte Spielmann1, Irina Hüning1;4

1University of Lübeck, Institute of Human Genetics, University Hospital Schleswig-Holstein, Lübeck, Germany; 2University Zürich, Institute of Medical Genetics, Schlieren, Switzerland; 3University of Kiel, Institute of Human Genetics, University Hospital Schleswig-Holstein, Kiel, Germany; 4MVZ für Humangenetik und Molekularpathologie, Rostock, Germany

Background: Smith-McCort dysplasia 2 (SMC2) is a rare autosomal-recessive osteochrondrodysplasia caused by homozygous or compound heterozygous pathogenic variants in RAB33B. Main clinical characteristics include a disproportionate short stature of varying severity with a short neck and trunk, protruding thorax, rhizomelic shortening of the limbs, progressive genua valga as well as elbow and finger joint stiffness with first symptoms usually presenting within the second to fourth year of life. Radiological findings resemble Dyggve-Melchior-Clausen syndrome which additionally presents with intellectual disability.

Methods: We report three siblings (one girl and two boys) aged 7, 12 and 16 years from a consanguineous Iraqi family with clinical features aligning the above described. Growth was normal until the age of three. Two of the affected siblings presented with microcephaly. Exome sequencing (ES) was performed in the most severely affected sibling as well as a penta-based whole genome sequencing (WGS) within a study including the other two affected siblings as well as the parents.

Results: By ES the pathogenic homozygous RAB33B variant NM_031296.3:c.253C>T; p.(Gln85*) was detected in the most severely affected sibling. Using penta-based WGS we were able to confirm the homozygous variant in RAB33B in the two affected siblings and identify the parents as heterozygous carriers.

Conclusion: To date no microcephaly had been described for patients with SMC2. Two of the three here presented siblings showed a microcephaly ranging between -2,3 to -2,6 SD while having an unaffected intellectual development. These cases widen the phenotypic spectrum of SMC2 and demonstrate the intrafamiliar phenotypic heterogeneity of SMC2.

Conflict of Interest: None declared

EP06.019 Carotid Rete Mirabile in Pseudoxanthoma Elasticum

Robert Carlier1, Alice Porto Vasconcelos 2, Dominique P. GERMAIN2;3

1Raymond Poincaré Hospital (APHP), Department of Radiology, Garches, France; 2APHP University Paris Saclay, Division of Medical Genetics, Garches, France; 3Paris-Saclay University, University of Versailles, UFR des Sciences de la Santé Simone Veil, Montigny-le-Bretonneux, France

Background/Objectives: Pseudoxanthoma elasticum (PXE, OMIM #264800) is a rare genetic metabolic disorder of connective tissue caused by bi-allelic pathogenic variants in the ABCC6 gene. The lack of functional ABCC6 protein leads to progressive ectopic mineralization of soft connective tissues, that is most apparent in the skin, eyes and cardiovascular system. The first clinical sign of PXE is almost always small yellow papules on the nape and sides of the neck and in flexural areas. Dystrophic calcification of Bruch’s membrane of the retina, may trigger choroidal neovascularization and, ultimately, blindness in late-stage disease. Lesions in medium-sized artery walls may result in peripheral artery disease. Transient ischemic attacks and ischemic strokes have been reported but cerebrovascular abnormalities have not been extensively characterized so far although in a recent study heterozygous ABCC6 mutations were amongst the most common in a large series of >4,000 young stroke patients. Carotid rete mirabile (CRM), an embryonic abnormal carotid malformation, has been independently reported in association with PXE in the literature.

Methods: In this report, brain MRI investigation was performed in a large cohort of 40 PXE patients.

Results: Carotid rete mirabile was identified in 4 patients (10%).

Conclusion: The results further highlight the occasional association between the two conditions. PXE should be considered in case of identification of carotid rete mirabile on brain MRI. The pathophysiological mechanisms that support the embryonic malformation in a few PXE patients warrants further investigation, as well as the potential role of ABCC6 variants as a risk factor for ischemic stroke.

Conflict of Interest: None declared

EP06.020 Markers in leg ulcer manifestation for sickle cell anemia patients

Claudia Regina Bonini-Domingos 1, Lucas Ramos1, Flavia Cristina Rodrigues-Lisoni1

1Ibilce-Unesp, Biological Sciences, Sao José do Rio Preto, SP

Consortium: Postgraduate program in Biosciences

Background/Objectives: This study investigates leg ulcers in sickle cell anemia (SCA) patients, a condition stemming from a beta globin gene mutation resulting in abnormal hemoglobin S (Hb S). Hb S induces structural changes in red blood cells, causing hemolysis and vasocclusion, yet the etiology of leg ulcers in SCA remains unclear. The research aims to identify markers linked to leg ulcer occurrence in SCA. Hemoglobin profiles were confirmed through HPLC and PCR-RFLP, exploring angiogenesis-related gene polymorphisms (TGF-β1, VEGFA1, HIF-1α) and assessing protein expression via Western blotting.

Methods: Hemolysis markers were evaluated from medical records. The study involved 24 Hb SS adults with active leg ulcers and 33 without ulcers. Results revealed a significant association between anemia severity and leg ulcers, with strong correlations for hemoglobin, aspartate aminotransferase, and indirect bilirubin. However, no significant relationship emerged between investigated polymorphisms and leg ulcers.

Results: Protein expression analysis indicated elevated VEGFA1 and TGFB1 levels in ulcer patients, suggesting a pro-angiogenic state. This implies that heightened expression of these proteins, coupled with severe anemia, may disrupt homeostasis, impeding wound healing and triggering leg ulcers.

Conclusion: In conclusion, the study establishes a connection between anemia severity, elevated VEGFA1, TGFB1 proteins, and leg ulcer occurrence in SCA patients, contributing to a deeper understanding of the mechanisms behind leg ulcer development in this population.

Grants:

Conflict of Interest: None declared

EP06.021 The different origin of the common genetic cause of X-linked ichthyosis in distinct ethnic groups of the Republic of the North Ossetia – Alania

Tatiana Vasilyeva 1, Andrey Marakhonov2, Artem Borovikov3, Vitaly Kadyshev2, Olga Lenina4, Zhanna Markova5, Alena Chukhrova6, Inna Tebieva7, Rena Zinchenko2

1Research Centre for Medical Genetics, Genetic epidemiology laboratory, Moscow, Russian Federation; 2Research Centre for Medical Genetics, Genetic epidemiology laboratory, Moscow; 3Research Centre for Medical Genetics, Moscow; 4Research Centre for Medical Genetics, Moscow, Russian Federation; 5Research Centre for Medical Genetics, Cytogenetics laboratory, Moscow; 6Research Centre for Medical Genetics, DNA diagnostics laboratory, Moscow; 7Republican Children’s Clinical Hospital, Vladikavkaz, Russian Federation

Background/Objectives: Recessive X-linked ichthyosis (XLI) (OMIM #308100) represents one of the most common genodermatoses. XLI is characterized by dark brown scales and skin dryness, other systems can be involved. The cause of XLI is LoF variants, predominantly deletions of the STS gene. Clinical and genetic heterogeneity of ichthyoses demands genetic testing. During expedition to the Republic of North Ossetia–Alania, one of the Northern Caucasus Republics, six unrelated families from different ethnic groups with hereditary ichthyosis were identified.

Methods: Genetic counseling; WES, CMA (implied in one of the index patients); multiplex PCR, PDAF; Sanger sequencing (in 1 negative for STS deletion family); Xp22.31 polymorphic STR loci analysis.

Results: Pedigrees showed that only men were affected. Manifestations included generalized skin dryness and grayish-brown scales, in one case, skin lesions were combined with mental retardation and cryptorchidism.

A hemizygous chromosome Xp22.31 deletion encompassing the STS gene of 1.9 Mb was defined in Kumyk family. The deletion was screened amongst rest families by PDAF and was detected in 4 more families (Turkish, and 3 Ossetian-Ironian). In Ossetian-Digorian family a hemizygous substitution c.1109G>C p.(Gly370Ala) in the STS gene was detected by Sanger sequencing.

Different length of STR markers suggested different origin of the deletion in different families.

Conclusion: Xp22.31 deletion is thought to have a recurrent character, it occurred in a hot spot at a locus with an increased recombination rate near pseudoautosomal region, and independently in different ethnic groups.

Grants: This study was supported by the state assignment of the Ministry of Science and Higher Education of Russia.

Conflict of Interest: None declared

EP06.022 Clinical and genetic characteristics of Turkish pediatric short stature cohort

KUBRA ADANUR SAGLAM 1, Semiha Bekfilavioglu2, Aysel Yıldız Boyraz2, Emine Ayça Cimbek2, Ayse Ozden3, AYBERK TURKYILMAZ1, Alperhan Cebi1, Hakan Doneray3, Gulay Karaguzel2

1Karadeniz Technical University Faculty of Medicine, Medical Genetics, Trabzon, Türkyie; 2Karadeniz Technical University Faculty of Medicine, Department of Pediatrics, Division of Pediatric Endocrinology, Trabzon, Türkyie; 3Atatürk University Faculty of Medicine, Department of Pediatrics, Division of Pediatric Endocrinology, Erzurum, Türkyie

Background/Objectives: Children with <-2 SDS height for age and ethnicity are defined as short stature. Here we aim to present genetic etiologies in a Turkish pediatric patient cohort with short stature.

Methods: Genetic testing and clinical follow-up of pediatric patients with short stature were retrospectively reviewed. Patients tested with Whole Exome Sequencing (WES) and SNP-array analyses were included. Patients with secondary causes of short stature and those with additional chromosomal diseases and methylation defects that may cause short stature were not included in the study. SNP-array and WES were performed in 40 and 76 patients, respectively.

Results: Of the total cases, 9 (37.5%) were female and 15 (62.5%) were male. The median age at initial presentation was 1,81 years. The median height of boys was -2.86 SDS and that of girls was -3.68 SDS. 24 unique variations of ACAN(n = 1), ACP5(n = 1), BLM(n = 2), BRAF(n = 1), COL10A1(n = 1), CUL7(n = 2), DYNC2H1(n = 2), EXT1(n = 1), FGD1(n = 2), GNPAT(n = 1), LRP5(n = 1), NF1(n = 1), OBSL1(n = 2), PGAP1(n = 1), SRCAP(n = 2), TRIM37(n = 1), TRPV4(n = 2) genes were found in 22 patients from 20 different families. Diagnostic success rate was 28.94% in patients who were tested with WES. 4p16 and SHOX deletions were found in 2 patients from 2 different families. Growth hormone therapy was administered to patients with variations in ACAN, ACP5, COL10A1 genes and SHOX deletion.

Conclusion: It is necessary to reveal the genetic causes of short stature for managing treatable causes in a timely manner, having information about the prognosis and providing accurate genetic counselling to families, including the risk of recurrence.

Grants: No

Conflict of Interest: None declared

EP06.023 Insights into cartilage-hair hypoplasia and anauxetic dysplasia spectrum through five unrelated patients

Merve Tanrısever Türk 1, gizem ürel-demir1;2, Gulen Eda Utine1, Pelin Simsek-Kiper1

1Hacettepe University Faculty of Medicine, Department of Pediatrics, Division of Pediatric Genetics, Ankara, Türkyie; 2Mersin City Training and Research Hospital, Pediatric Genetics, Mersin, Türkyie

Background/Objectives: Cartilage-hair hypoplasia (CHH,MIM#250250) and Anauxetic dysplasia (AD,MIM#607095) belong to a spectrum of autosomal recessive spondylometaepiphyseal dysplasias characterized by prenatal onset of severe short stature. AD is marked by severe skeletal manifestations, whilst CHH is associated with extra-skeletal features, including hypotrichosis, immunodeficiency, and hematological abnormalities. AD is caused by variants in RMRP, POP1, and NEPRO genes, whereas CHH results from variants in RMRP. Here, we present the clinical, radiologic and molecular findings of five unrelated patients diagnosed with AD or CHH.

Methods: Three patients were diagnosed with AD and one with CHH through RMRP sequencing, while the fifth patient received a diagnosis with exome sequencing, supported by a preliminary diagnosis of AD based on clinical and radiological findings.

Results: All patients presented with severe short stature, short neck, brachydactyly, scoliosis, and joint laxity. In three patients harboring a RMRP variant and diagnosed with AD; rhizomelic shortening, iliac bone hypoplasia, shortened long tubular bones, delayed carpal bone age, and metaphyseal flaring were observed. Another patient with a RMRP variant, exhibiting mild metaphyseal involvement with sparse eyebrows and hair, was diagnosed with CHH. The patient with compound heterozygous variants in the POP1 gene had metaphyseal dysplasia, hypoplastic femoral necks, delayed carpal bone age, and flexion contractures in the elbows.

Conclusion: AD and CHH represent a heterogeneous spectrum with varying degrees of metaphyseal dysplasia and extra-skeletal manifestations. Given the spectrum of clinical and radiological variations, a high suspicion index and accurate interpretation are essential for a precise diagnosis and appropriate genetic counseling.

Grants:None.

Conflict of Interest: None declared

EP06.024 Case report of rare Trichothiodystrophy syndrome

Iveta Žukauskaitė 1, Rasa Traberg1, Rimvydas Jonikas1, Rasa Ugenskiene1

1Lithuanian University of Health Sciences, Department of Genetics and Molecular medicine, Kaunas, Lithuania

Background/Objectives: Trichothiodystrophy (TTD) is a rare, genetic, syndromic hair shaft abnormality disorder characterized by short, dry, sulfur-deficient, brittle hair usually associated with highly variable neuroectodermal manifestations, such as ichthyosis, photosensitivity, and intellectual disability. Photosensitive trichothiodystrophy-1 (TTD1) is caused by homozygous or compound heterozygous mutations in the ERCC2 gene.

Methods: The proband, a 42-year-old female, born at 33-34 weeks of gestation with a birth weight of 1400 g, from the first pregnancy complicated by polyhydramnios and poor fetal growth. The patient exhibited failure to thrive, dry skin, curly hair and photophobia. Due to global developmental delay, a severe mental disability was diagnosed. The patient also had congenital hip dysplasia, which has now progressed to arthrosis of the hip joint. Cataracts were diagnosed and treated at 16-17 years of age. On physical examination, the patient presented with short stature, microcephaly, caries, dry skin with hyperkeratosis, and coarse, brittle, curly hair. Genealogy was negative.

Results: Whole exome sequencing (WES) revealed a homozygous missense variant in the ERCC2 (NM_000400.4) gene, c.1459C>T (p.Arg487Trp), which we have classified as VUS, based on ACMG 2015 criteria PM2, PP3, PM3 (supporting). This is the first homozygous c.1459C>T variant in a TTD1 patient reported so far. A previously published case involved a TTD patient with the c.1459C>T variant in a compound heterozygous state.

Conclusion: Lacking functional evidence, we classified the homozygous c.1459C>T variant as a VUS. However, considering high phenotypic compatibility, we believe this variant to be causative for the patient’s symptoms. Periodic variant reassessment is highly recommended.

Grants: No

Conflict of Interest: None declared

EP06.025 Hypophosphatemic rickets. Case family report with pathogenic variant in the PHEX gene

Nory Omayra Davalos Rodriguez 1;2, Rosa Esther Castañeda Arteaga3, Ricardo E’ Vega Hernandez4, José Guadalupe Ramiro Ortega Águila5, Ana Rosa Rincon Sanchez6, Cristian Víctor Ledezma Rodríguez7, Sergio Alberto Ramirez-Garcia8, Diana Garcia-Cruz8

1Institute of Human Genetics, CUCS, University of Guadalajara, Department of Molecular Biology and Genomics, Guadalajara, Jalisco; 2Valentín Gómez Farias Hospital, ISSSTE., a) Genetics, Zapopan, Jalisco; 3Valentín Gómez Farias Hospital, ISSSTE., b) Pediatric nephrology, Zapopan, Jalisco, Mexico; 4Valentín Gómez Farias Hospital, ISSSTE., c) Head of outpatient consultation, Zapopan, Jalisco; 5Medicine Degree, CUCS, University of Guadalajara, Guadalajara, Jalisco, Mexico; 6Institute of Molecular Biology in Medicine and Gene Therapy, Molecular Biology and Genomic, Guadalajara, Jalisco; 7Specialty Hospital, CMNO, IMSS, Plastic and Reconstructive Surgery, Guadalajara, Jalisco; 8Benito Juárez Autonomous University of Oaxaca, Faculty of Chemical Sciences, Oaxaca, Oaxaca, Mexico

Background/Objective: Hypophosphatemic rickets (HR) is a heterogeneous group of inherited disorders characterized by hypophosphatemia and bone hypomineralization. The most common is X-linked hypophosphatemia (XLH, MIM 307800). The diagnosis includes the identification of Phosphate-Regulating endopeptidase, X-linked (PHEX) gene variant. The present report refers a family with XLH carried a variant in PHEX gene.

Methods: Case 1: Female 7-years old, product of the first pregnancy of non-consanguineous parents. Normal development until at 4-month age, she began with progressive curvature of the lower extremities. Tooth eruption Delay.

Physical examination. Dental abnormalities. Disproportionate short stature, genu varum. X-ray. Distal medial femoral and tibial bowing with widening and fraying of the distal epiphyseal plate of the femur. Nephrology. Cystic kidney disease and nephrocalcinosis. Low levels of 25-hydroxy vitamin D and phosphorus in serum and urine.

Case 2. 35-year-old mother, with disproportionate short stature, due to shortening of lower extremities. The genu varum and gait improved using orthopedic devices in childhood. No other associated findings.

Results: Genetic study was carried out analyzing 16 genes related to renal tubular function. Detected mutation of the PHEX gene. Mother and daughter are heterozygous of the variant PHEX, Exon 4, c.367del (p. lLe123Serfs*21).

Conclusion: XLH is the most common form of familial hypophosphatemia, with complete penetration and variable expressivity. More than 300 pathogenic variants of the PHEX gene have been described. The variant in PHEX gene described in this family has not been reported in the literature in individuals affected; for this reason, it has been classified as pathogenic.

Conflict of Interest: None declared

EP06.026 Association of LGALS3 gene polymorphisms with methotrexate treatment efficacy in patients with rheumatoid arthritis

Nela Maksimovic1, Biljana Jekic1, Milka Grk1, Tatjana Damnjanovic1, Dijana Perovic1, Biljana Ljujic2, Marija Dusanovic Pjevic1, Valentin Djonov3, Milica Rasic 1, Vladislav Volarevic2;4

1Institute of Human Genetics, Faculty of Medicine, University of Belgrade, Serbia; 2Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Serbia; 3Institute of Anatomy, University of Bern, Switzerland; 4Department of Microbiology and Immunology, Faculty of Medical Sciences, University of Kragujevac, Serbia

Background/Objectives: In the treatment of rheumatoid arthritis (RA) methotrexate (MTX) is a commonly used drug. However, this treatment is ineffective in approximately 30% of patients. Galectin-3 is detected in the synovial tissue of RA patients and accumulates at sites of cartilage invasion. Serum levels of galectin-3 are also elevated in these patients. Based on this knowledge, we investigated the possible impact of LGALS3 gene polymorphisms (rs4644, rs4652 and rs11125) on disease activity at the beginning and after six months of MTX therapy, and MTX efficacy after six months of treatment, in a group of 205 RA patients.

Methods: Patients were genotyped for selected polymorphisms by Real-time PCR method, using standardized TaqMan assays.

Results: Based on the EULAR criteria, after 6 months of MTX therapy, 173 patients (84.4%) were classified as MTX responders, while 32 patients (15.6%) were MTX non-responders. Analysis of the association of LGALS3 gene polymorphisms with activity of the disease, measured by DAS28 score, did not show statistically significant results. However, the carriers of rs4644 C allele (CC or CA genotype) and rs4652 A allele (AA or AC genotype) had more often poor responses to MTX therapy (p = 0.01 and p = 0.008, respectively). Also, all three analyzed polymorphisms were in haplotype block and rs4644/rs4652/rs11125 CAA haplotype is significantly more frequent in patients with poor response to MTX therapy (p = 0.012).

Conclusion: In conclusion, in RA patients, LGALS3 gene polymorphisms may be associated with MTX therapy response.

Grants: This work was supported by European Crohn’s and Colitis Organization (ECCO) and Faculty of Medical Sciences University of Kragujevac (MP01/18).

Conflict of Interest: Nela Maksimovic: None declared, Biljana Jekic: None declared, Milka Grk: None declared, Tatjana Damnjanovic: None declared, Dijana Perovic: None declared, Biljana Ljujic: None declared, Marija Dusanovic Pjevic: None declared, Valentin Djonov: None declared, Milica Rasic: None declared, Vladislav Volarevic This work was supported by the European Crohn’s and Colitis Organization (ECCO) (grant “The role of galectin 3 in acute colitis”) and the Faculty of Medical Sciences University of Kragujevac (MP01/18)

EP06.027 A case of Brachiolmia type 4 resulting from a novel variant of PAPSS2 gene

Öznur YILMAZ BAYER 1, banu nur1, sezin yakut uzuner2, ercan mihci1

1akdeniz university, pediatric genetics, antalya, Türkyie; 2akdeniz univesity, medical biology, antalya, Türkyie

Background/Objectives: Brachiolmia is a genetically heterogeneous skeletal dysplasia characterized by disproportionate short stature, short trunk, platyspondyly, and often mild long bone abnormalities. Brachyolmia can be classified into various types, including Brachyolmia type-1 (Toledo/Hobaek type), Brachyolmia type-2 (Maroteaux type), Brachyolmia type-3, Brachyolmia type-4 and Brachyolmia type-5. Here we present our rare case diagnosed with Brachiolmia type 4, which is associated with the PAPSS2 gene and is inherited as an autosomal recessive disease.

Methods: We presented a 6,5 years old boy born at 38 weeks of gestation from 35-year-old mother by cesarean section. There was parental consanguinity. On physical examination, he had short stature (his height was 104,2 cm, -3.07 SDS), synophyrs, thick eyebrows, long philtrum, simian crease on right hand, scoliosis, lomber lordosis. The upper segment/lower segment ratio was determined as 0.85 (<-2 SDS). Psychomotor development was normal. He had normal intelligence.

Results: Clinical-exome sequencing analysis displayed a novel homozygous pathogenic variant of PAPSS2 gene (NM_001015880.2, c.1753C>T, p.(Arg585*)).

Conclusion: Brachiolmia type 4 includes skeletal and radiographic findings such as platyspondyly, irregular end plates, kyphoscoliosis, lumbar scoliosis, short and bowed lower limbs, enlarged knee joints, mild metaphyseal changes of knees and hips, precocious osteoarthropathy, mild brachydactyly. Short stature is present from prenatal. So far 79 patients with PAPSS2 deficiency were reported in the literature. We have report a novel nonsense variant of PAPSS2 which caused an autosomal recessive form of Brachyolmia in a consanguineous Turkish family.

Key words: Brachiolmia type 4, PAPSS2, short stature

References:

- https://doi.org/10.1016/j.heliyon.2023.e23688

- https://doi.org/10.1002/ajmg.a.61282

Grants: None

Conflict of Interest: None declared

EP06.028 Ehlers-Danlos syndrome type III with haploinsufficiency associated with compound heterozygous genotype of the TNXB gene. Mexican case report.

Ana Rosa Rincon Sanchez 1, Nory Omayra Davalos Rodriguez2;3, Diana Garcia-Cruz4, Rodrigo Hernández Ramírez5;6, Vianney Yolanda Jiménez Rodríguez7, Sergio Alberto Ramirez-Garcia4

1Institute of Molecular Biology in Medicine and Gene Therapy, Molecular Biology and Genomic, Guadalajara, Jalisco; 2Institute of Human Genetics, CUCS, University of Guadalajara, Department of Molecular Biology and Genomics, Guadalajara, Jalisco; 3Valentín Gómez Farias Hospital, ISSSTE., Genetics, Zapopan, Jalisco; 4Benito Juárez Autonomous University of Oaxaca, Faculty of Chemical Sciences, Oaxaca, Oaxaca, Mexico; 5Specialty Hospital, CMNO, IMSS, General Surgery, Guadalajara, Jalisco, Mexico; 6Institute of Human Genetics, CUCS, University of Guadalajara, Molecular Biology and Genomic, Guadalajara, Jalisco, Mexico; 7Institute of Human Genetics, CUCS, University of Guadalajara, Medicine degree, Guadalajara, Jaisco, Mexico

Background/Objective: Ehlers-Danlos syndrome (EDS) is a group of genetic connective tissue disorders, characterized by skin hyperextensibility, generalized joint hypermobility, chronic joint pain, and tissue fragility. It affects 1:10,000 to 1:25,000 individuals. One of these variants, EDS hypermobility type (EDS III, MIM 130020), comprises 30% of EDS cases. This report describes a male with TNXB gene mutation.

Methods: 18-year-old male, referred for chronic generalized musculoskeletal pain and joint hypermobility since childhood. Product of the IV pregnancy of 7 siblings, from non-consanguineous parents. Hyperextensible skin in mother, maternal grandmother, and brother. Rheumatoid arthritis was referred.

Physical examination. Soft and hyperextensible skin. Brachycephaly, flat frontal, high nasal bridge, hypoplastic nares, short philtrum, large mouth, thick upper lip. Bilateral neck hyperextensibility. Upper extremities with significant joint hypermobility shoulders. Hands with camptodactyly. Thoracic and lumbar hyperextension spine.

Laboratory. Negative rheumatoid factor. Low levels of Tenascin XB (TNXB) 6.5 ng/ml.

Electron microscopy. The ultrastructural level, the skin showed irregular and immature elastin fibers and fibers devoid of microfibrils were observed.

Results: PCR-GAP and Sanger Sequencing showed compound heterozygous genotype; deletion of 2 bp, AA56063 in exon 8 and deletion of 30Kb in 6p21.3, (RCV002051614 ID 23588), aberrant truncated proteins are formed, decreases function, and leads to haploinsufficiency, even though the mutations are recessive.

Conclusion: This is the first mutation described in TNXB gene in Mexican case with combination of mutations. The diagnosis was based on clinical major and minor criteria and family history. Genetic testing was used to confirm the diagnosis of the specific subtype.

Conflict of Interest: None declared

EP06.029 A novel homozygous DST variant in a patient with Epidermolysis Bullosa Simplex 3

Sofie Fredberg 1, Stine Bjoern Gram1;2;3, Helena Karstensen4, Ulrikke Lei5, Lilian Ousager1;2

1Odense University Hospital, Department of Clinical Genetics, Odense, Denmark; 2University of Southern Denmark, Department of Clinical Research, Odense, Denmark; 3, European Reference Network for Rare Skin Diseases (ERN-Skin); 4Copenhagen University Hospital Rigshospitalet, Department of Clinical Genetics, Copenhagen, Denmark; 5Herlev and Gentofte Hospital, Department of Dermatology and Allergy, Gentofte, Denmark

Background/Objectives: Epidermolysis bullosa is a heterogeneous group of skin diseases characterized by bullae and blistering of skin exposed to mechanical stress. The mild autosomal recessive subtype Epidermolysis Bullosa simplex 3 (EBS3) (OMIM #615425) mainly affects the feet. In the Human Gene Mutation Database (HGMD), a total of six disease-causing and four possibly disease-causing variants in DST have been associated with EBS3.

Methods: A woman born to non-consanguineous parents presented at 18 years of age with blistering and keratoderma isolated to the skin of her feet. She had no infections or scarring and no symptoms from the hair, teeth or nails. Diagnostic analysis was performed by next-generation sequencing, using an exome-based in-silico panel targeting 352 genes related to dermatological diseases.

Results: We identified a homozygous frameshift variant in DST (c.7544dupA, p.(Gln2516Alafs*6), NM_001723.6). The variant introduces a premature stop codon in the last coding exon, which theoretically leads to a truncated dystonin protein product or alternatively nonsense mediated decay. The variant is reported with a minor allele frequency (0.0008%,13/1.614.078 heterozygous alleles) in gnomAD v4.0.0 with no homozygous individuals. To the best of our knowledge, the variant has not been reported in the literature, though it has been reported once in ClinVar as a variant of uncertain significance. Most variants related to EBS3 reported in HGMD, were either frameshift or nonsense mutations, supporting our interpretation of the variant as likely pathogenic.

Conclusion: We identified a novel likely pathogenic homozygous variant in DST in a woman with EBS3.

Grants: None

Conflict of Interest: None declared

EP06.030 Homozygous SMOC2 Deletion: A Novel Variant Associated with Dentin dysplasia, type I

Rana Alzahrani 1, Balsam Aleissa2, Sally Alyousef2, Norah Alsaleh1

1Ministry of National Guard Health Affairs, Genetics and Precision Medicine Department, Riyadh, Saudi Arabia; 2Ministry of National Guard Health Affairs, College of Dentistry, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Riyadh, Saudi Arabia

Background/Objectives:

  • Dentin dysplasia is a rare disturbance of dentin formation characterized by normal enamel but atypical dentin formation, which leads to premature exfoliation of the teeth. Inherited dental malformations constitute a clinically and genetically heterogeneous group of disorders. Dentin dysplasia type 1 is an autosomal recessive disorder associated with homozygous variants in the SMOC2 gene.

  • Here, we are reporting a 20-year-old male was referred from the dental clinic with a complaint of dental malocclusion, hypodontia, and microdontia. He was born from a highly consanguineous family with similar findings in 2 other cousins. His neonatal history and developmental history were unremarkable. His examination was remarkable for short stature, strabismus and dysmorphic features with protruding lips and severe open bite. Difficulty swallowing and long epiglottis. He has Anodontia and microdontia with slight calculus and thick plaque and is missing all teeth in the lower anterior segment.

    Methods/ Results:

  • Exome sequencing was unremarkable, followed by Genome sequencing, which reported a homozygous copy loss encompassing the entire SMOC2 gene. This variant is absent from both local and international databases.

    Conclusion:

  • SMOC2 gene encodes a member of the SPARC family, which is highly expressed during embryogenesis and wound healing. Animal studies have shown the effect of SMOC2 depletion on odontogenesis. To date, only 4 pathogenic and likely pathogenic variants have been reported in cases with Dentin dysplasia. Here, we report a novel homozygous copy number loss involving the entire SMOC2 gene, which explains the index’s phenotype and severe presentation.

Grants: None

Conflict of Interest: None declared

EP06.031 A Novel FBN1 Variant in A Large Marfan Family with High Penetrance of Aortic Features

Yucel Erbilgin 1, Kıvanc Cefle2, Sukru Ozturk2, Lala Soltanova3, Pinar Kadioglu4, sumeyye kocaağa1;5, Melisa Kılıç1;5, Muge Sayitoglu1

1Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Genetics, Istanbul, Türkyie; 2Istanbul University, Istanbul Medical Faculty, Division Of Medical Sciences, Department Of Internal Medicine, Istanbul, Türkyie; 3Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Department Of Internal Medicine, Istanbul, Türkyie; 4Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Department Of Internal Medicine, Istanbul, Türkyie; 5Istanbul University, Institute of Graduate Studies in Health Science, Istanbul, Türkyie

Background/Objectives: Marfan syndrome (MFS) is an autosomal dominantly inherited connective tissue disorder that caused by pathogenic FBN1 variants. Although, over 3077 variants have been reported in FBN1, genotype-phenotype correlations have not been defined for the all variants. Here, we report a large Turkish Marfan family with a rare FBN1 variant.

Methods: The index patient who had an atypic Cushing disease and a strong family history with MFS were referred to our department. The patients’ DNA sample was analyzed by whole exome sequencing. Furthermore, 16 relatives were screened for FBN1 variant by Sanger sequencing and clinically evaluated according to Ghent II criteria.

Results: The proband and nine relatives were heterozygous for NM_000138.4:c.5330G>T, p.(Cys1777Phe) variant. The variant was classified as pathogenic which has been reported in one patient in Uniprot but no other databases such as gnomAD and ClinVar. The index case had dilated cardiomyopathy, pectus carinatum, pes planus, scoliosis, skin striae, myopia and severe apnea. None of the patients had hindfood deformity, pneumothorax, reduced elbow extension. Her family history revealed that two family members died from aortic rupture at age 19 and 32, her mothers’ aunt had surgery due to aortic dilatation and additional two family members had also had aortic dilatation.

Conclusion:This work reports for the first time a family with the FBN1 p.(Cys1777Phe) variant and a high penetrance of aortic rupture or aorta dilatation in the family.

Grants: This work was supported by Ministry of Industry and Technology, Istanbul Development Agency (ISTKA), Project No:TR10/22/TNH/0001.

Conflict of Interest: Yucel Erbilgin Ministry of Industry and Technology, Istanbul Development Agency (ISTKA), Project No:TR10/22/TNH/0001, Full Time, Kıvanc Cefle Full time, Sukru Ozturk Full Time, Lala Soltanova Full Time, Pinar Kadioglu Full Time, sumeyye kocaağa Part Time, Melisa Kılıç Part Time, Muge Sayitoglu Ministry of Industry and Technology, Istanbul Development Agency (ISTKA), Project No:TR10/22/TNH/0001, Full Time

EP06.032 Warburg-Cinotti Syndrome in a Pakistani male – Recognizing an ultra-rare syndrome in an under-represented population.

MAHRUKH NASIR 1;2, zohra hasan1, Fizza Akbar1, Salman Kirmani1

1Aga Khan University, Women and Child Health, Karachi, Pakistan; 2International Center for Chemical and Biological Sciences (ICCBS), Dr. Pajwani center for molecular medicine and drug research, Karachi, Pakistan

Background/Objectives: Warburg-Cinotti syndrome (WCS) is an ultra-rare connective tissue disorder characterized by corneal vascularization, keloid formation, chronic skin ulcers, subcutaneous tissue wasting, hand contractures and acro-osteolysis. It is caused by a heterozygous pathogenic (P)/ likely pathogenic (LP) variant in DDR2, which encodes discoidin domain collagen-responsive receptor tyrosine kinase 2 that regulates connective tissue formation. Less than 10 cases have thus far been reported. We report the clinical details of the first Pakistani case of WCS, caused by de-novo variant in DDR2.

Methods: Retrospective review of records– Case report.

Results: A 27-year-old male, born to non-consanguineous parents, presented with early-onset bilateral congenital blepharophimosis, progressive visual impairment, loss of subcutaneous fat in the extremities, progressive acro-osteolysis, conductive hearing loss along with impaired wound healing. Dysmorphic features included abnormal nasal bridge, high-arched palate, micrognathia and severe midface retrusion.

Whole Genome Sequencing identified a heterozygous missense variant in DDR2 (c.1829T>C, p.Leu610Pro) classified initially as a variant of uncertain significance (VUS) but later reclassified as likely pathogenic. Parental testing confirmed the de-novo occurrence of the variant. The phenotypic presentation matched with the genetic diagnosis of autosomal dominant WCS, providing clinical evidence for the variant to be disease causing.

Conclusion: This is first reported instance of WCS associated with a DDR2 variant in a Pakistani, expanding the clinical and genetic understanding of this rare syndrome in a diverse population. WGS and parental analysis confirmed a de-novo autosomal dominant inheritance pattern, confirming the molecular basis of WCS in our patient.

Grants: It is not a funded project.

Conflict of Interest: None declared

EP06.033 Identification of two novel pathogenic variants in the PLOD2 gene in a case of Bruck syndrome type 2

Elena Merkuryeva 1, Tatiana Markova2, Oksana Ryzhkova2, Yuri Buklemishev3, Vladimir Kenis4

1Research Centre for Medical Genetics, Moscow, Russian Federation; 2Research Centre for Medical Genetics, Moscow; 3N.N. Priorov National Medical Research Center of Traumatology and Orthopaedics, Moscow; 4H. Turner National Medical Research Center for Children’s Orthopedics and Trauma Surgery

Background/Objectives: Bruck Syndrome Type II (OMIM #609220) is a profoundly rare autosomal recessive condition characterized by osteogenesis imperfecta-like symptoms, severe congenital joint contractures, skin pterygia, short stature, significant limb deformities, and progressive scoliosis. Pathogenic variants in the nucleotide sequence of the PLOD2 gene lead to the development of the syndrome.

This study aims to delineate the molecular profile and clinical-radiographic manifestations of a patient with Bruck Syndrome Type 2, attributed to newly identified compound heterozygous mutations in the PLOD2 gene.

Methods: The patient, a 10-year-old, underwent an extensive evaluation involving pedigree analysis, clinical and neurological assessments, radiography, dual-energy X-ray absorptiometry, targeted panel sequencing, and direct Sanger sequencing to establish a precise diagnosis.

Results: We report two previously undocumented PLOD2 variants in the patient: c.8dupG (p.Cys4MetfsX35) in exon 1 and c.2222G>A (p.Gly741Glu) in exon 20. Clinical hallmarks included recurrent fractures, deformities of the long bones and spine, and stunted growth, with the child being non-ambulatory and unable to sit unassisted. Notably, joint contractures were absent at the age of ten. Radiographic indicators comprised multi-fractures, extreme multiplanar deformities exceeding 90 degrees, cortical layer thinning, thoracolumbar kyphosis, platyspondyly, vertebral compression fractures. Pelvic radiographs revealed protrusion of the acetabulum. Radiological examination of the skull showed Wormian bones, open sutures, platybasia, and basilar invagination.

Conclusion: The discovery of two novel PLOD2 variants expands the known mutational spectrum and provides valuable insights for future research on genotype-phenotype correlations in Bruck Syndrome Type 2.

Conflict of Interest: None declared

EP06.034 A novel mutation of LOR gene in a family with Vonwinkel syndrom, ichthyosiform variant

Esra Hilal Ceylan 1, cekdar kapazan1, Zeynep Baser1, Ayşe Deniz Yücelten2, Esra Dirimtekin1, Bilgen Bilge Geçkinli1

1Marmara University Hospital, Medical Genetics; 2Marmara University Hospital, Dermatology

Objectives:Vonwinkel Syndrome (Vsiv) is a rare autosomal dominant syndrome,characterized by diffuse generalized ichthyosiform dermatosis,palmoplantar keratoderma,digital constricting bands, and without hearing loss. Heterozygous mutations in the LOR gene encoding the loricrin protein cause VSIV. Loricrin promotes the maturation of corneocytes and extracellular adhesion structure and is critical for the terminal differentiation of the keratinocytes.Our aim is to present a novel pathogenic variant of LOR to discuss the clinical and molecular findings.

Material/Method: DNA was isolated from peripheral blood of the patient and his parents. Dermatosis panel of 90 genes were sequenced by Next Generation Sequencing (NGS). Family segregation analysis of the identified variant was performed using Sanger sequencing.

Results:The proband is a 7 -days-old boy who was the first child of consanguineous Turkish parents. He was referred to our clinic because of diffuse generalized ichthyosiform dermatosis. Physical examination revealed palmoplantar hyperkeratosis and generalized ichthyosiform dermatosis like his mother. Echocardiography, abdominal USG,bone survey and BERA test were normal. Molecular analysis revealed a novel heterozygous c.718_719del (p.Cys240Leufs*95) pathogenic variant in the LOR gene (NM_000427). According to ACMG criteria, this variant was evaluated as pathogenic. Segregation analysis revealed that her affected mother had the same mutation in heterozygous form.

Conclusion: The clinical appearance of VSIV occurs in infancy and early childhood, when differential diagnosis is difficult.We conclude that this study submits a novel pathogenic variant to the literature and contributes to the genotype-phenotype correlation.

Grants: None.

Conflict of Interest: None declared

EP06.035 The genetic landscape of RASopathies (neurocutaneous disorders) in Greek index cases

MARIA TZETIS 1, Konstantina Kosma2, Periklis Makrythanasis1;3, Anastasios Mitrakos1;4, Thomas Mprantzos1;4, Nikolaos Marinakis1;4, Faidon-Nikolaos Tilemis1, ASPASIA TSEZOU5, Christalena Sofocleous1

1National and Kapodistrian University of Athens, Medical Genetics, Athens, Greece; 2National and Kapodistrian University of Athens, Medical Genetics, Athens, Greece; 3Biomedical Research Foundation of the Academy of Athens, Genetics, Athens, Greece; 4EPIKNIPI research Institute, Genetiics, Athens, Greece; 5University of Thessaly, Laboratory of Cytogenetics and Molecular Genetics, Larissa, Greece

Background/Objectives: RASopathies or neurocutaneous diseases belong to a group characterized by dysregulation of RAS/MAPK pathway. Although each may have unique phenotypes, there are many overlapping characteristics, including heart defects, short stature, neurocognitive impairment, musculoskeletal abnormalities, craniofacial malformations, cutaneous lesions and cancer predisposition. RASopathies have been associated with mutations in about twenty genes including: Neurofibromatosis Type 1 (NF1): NF1, Neurofibromatosis type 2 & schwannomatosis: NF2, SMARCB1, LZTR1, Noonan syndrome (NS) & LEOPARD: PTPN11, SOS1/2, RAF1, KRAS, NRAS, SHOC2, CBL, LZTR1, Legius syndrome (LS): SPRED1, Costello syndrome (CS): HRAS, Noonan-like syndrome with loose anagen hair (NS-LAH): SHOC2, Cardio-facio-cutaneous syndrome (CFCS):KRAS, BRAF, MEK1/2, and Tuberous sclerosis: TSC1/2.

Methods: A total of 840 patients were referred to the Laboratory of Medical Genetics for RASopathy testing. Phenotypic and clinical characterization was performed during pre-test counseling. WES was performed using IDT xGen Exome Research v2 kit and running on Illumina NextSeq-500 system. For bioinformatic analysis VarSome Clinical and Franklin platforms were used. Unresolved cases were further analyzed using the ExomeDepth WES-based CNV-calling algorithm. All variants including CNVs were classified following ACMG recommendations and ClinGen criteria.

Results: Amongst the 840 patients, 426 carried causative pathogenic variants. Of these 367 carried an NF1 pathogenic variant (50% de novo and 180 different NF1 variants). For the remaining 58 patients pathogenic variants were as follows: twelve LZTR1, five NF2, eight SPRED1, nine PTPN11, two SMARCB1, fourteen TSC2 and eight TSC1.

Conclusion: WES with CNV calling proved helpful in addressing management for these conditions providing differential diagnosis and targeted therapeutic options.

Conflict of Interest: None declared

EP06.036 Identification of a novel FBN1 gene mutation in a patient with ectopia lentis

Renata Szalai 1, Judit Bene1, Anna Zsigmond1, Krisztina Galimurka1, Kinga Hadzsiev1

1University of Pécs, Department of Medical Genetics, Pécs, Hungary

Background/Objectives: Marfan syndrome (MFS) is an autosomal dominant connective tissue disease encompassing characteristic features of the cardiovascular, skeletal, and ocular systems. Heterozygous mutations in FBN1 gene lead to haploinsufficiency of fibrillin-1, result in distinct overlapping conditions, including; classical and neonatal MFS, autosomal dominant ascending aortic aneurysms, familial arachnodactyly, Shprintzen–Goldberg syndrome, the ‘MASS’ phenotype (myopia, mitral valve prolapse, borderline aortic root enlargement, skin and skeletal findings), mitral valve prolapse syndrome, and autosomal dominant ectopia lentis (EL). EL with the genetic and cardiovascular features form the major manifestations of the diagnostic Ghent criteria of MFS. Fibrillin is a relevant component of the ciliary zonules, therefore EL manifests in up to 60% of MFS cases.

Methods: Here we report a six years old Hungarian patient with ectopia lentis and marfanoid habitus. Sequencing analysis was performed using QIAGEN QIAseq Targeted DNA Pro Panel library kit and Illumina MiSeq sequencing technology. Sanger sequencing confirmed the presence of the pathogenic variant.

Results: NGS analysis of FBN1, TGFBR1 and TGFBR2 gene panel identified a novel missense c.3427G>T (p.Gly1143Cys) variant in FBN1 gene in a heterozygous state. The classification of the detected variant is likely pathogenic based on the American College of Medical Genetics and Genomics (ACMG) guideline.

Conclusion: EL is a condition that may be a part of numerous syndromes or be isolated. In young patients with EL symptom, FBN1 mutation analysis should be performed to help establishing a rapid and correct diagnosis and identify patients at risk of potentially life-threatening complications.

Grants: PTE ÁOK KA-2023-26.

Conflict of Interest: None declared

EP06.037 A family with OBSL1-related disorder and a unique presentation of 3-M syndrome associated with respiratory distress and early death.

khaled osman 1, adel Shalata2

1The Simon Winte Institute For Human Genetics, Bnai Zion Medical Center, Haifa, Israel, genetics, haifa, Israel; 2The Simon Winte Institute For Human Genetics, Bnai Zion Medical Center, Haifa, Israel, Haifa, Israel

Background: 3-M syndrome is an autosomal recessive disorder caused by bi-allelic pathogenic variants in CCDC8, CUL7, or OBSL1. Classically, it is characterized by growth deficiency with distinctive facial appearance, characteristic skeletal features and occasionally systemic involvement.

Early mortality rate is considered to be low.

Herein, We report a unique presentation of an individual with severe pre and postnatal course resulting in early death.

Materials: A Muslim family was referred to genetic counseling due to their newborn daughter with severe signs of skeletal dysplasia.

Fetal course was noted with shortening of long bones. Postnatally she presented with short stature and macrocephaly associated with suggestive skeletal and dysmorphic features.

Later, she developed a respiratory distress (secondary to hypoplastic lung) and pulmonary hypertension leading to early death. brain and abdominal US showed no anomaly.

Genome sequencing (national project) revealed a likely pathogenic homozygous OBSL1 variant c.844G>T;p.(Gly282Cys) inherited from both parents.

Discussion: The OBSL1 variant is not observed in databases, was predicted to be deleterious by Splice in-silico models (causing an alternative splice in exon 3 -SpliceAI (0.99), DANN (1)) and the Patient’s phenotype/ family history is highly specific for 3-M syndrome. RNA sequencing are planned to be done.

Respiratory complications were described previously in subjects with 3-M syndrome. death was rarely reported, typically in probands with CUL7 mutations rather than OSBL1.

In conclusion, we present a novel homozygous OBSL1 variant in a pedigree with unique presentation:- skeletal dysplasia with respiratory distress and pulmonary hypertension causing early death, thus expanding the phenotype of OBSL1- Related disorder.

Conflict of Interest: None declared

EP06.038 Clinical and molecular characterization of 24 patients with ACAN variations

Melek Trigui 1;2, Nathalie PALLARES-RUIZ1, David Geneviève2, Cyril Amouroux3, Thomas Edouard4, Sabine Sigaudy5, Marjolaine Willems2, Mouna Barat-Houari1

1CHU de Montpellier, Department of Molecular Genetics and Cytogenomics, Rare and Auto inflammatory Diseases Unit, CHU Montpellier, University of Montpellier, Montpellier, France; 2CHU de Montpellier, Montpellier University, Reference Center for Rare Disease skeletal dysplasia, Medical Genetic department, CHU Montpellier, Montpellier, France; 3CHU de Montpellier, Department of Pediatric Endocrinology, Arnaud de Villeneuve Hospital, Montpellier France; 4CHU de Toulouse, Endocrine, Bone Diseases and Genetics Unit, Toulouse University Hospital, Toulouse, France; 5Hôpital Timone Enfant, Marseille, Département de Génétique Médicale

Background/Objectives: Short stature (SS) is a prevalent clinical manifestation in children. While certain causes of SS can be readily identifiable through routine biological tests, often physicians struggle to ascertain any underlying pathogenic cause, resulting in the diagnosis of idiopathic short stature (ISS). Aggrecan, encoded by ACAN, serves as a major proteoglycan component in the extracellular matrix of the growth plate and plays a crucial role in cartilage function. The aim of our study is to establish a genotype-phenotype correlation in 24 patients carrying distinct ACAN variants.

Methods: We conducted a panel-based analysis, including 82 genes associated with constitutional bone diseases, in 292 French patients who consulted for difficulty to thrive. Genotype-phenotype correlation analysis was performed for all included subjects.

Results: Panel analysis unveiled a genetic aetiology in 72 probands, implicating various genes. Twenty-four carried a pathogenic or probably pathogenic ACAN variant (33 %), of which 20 were novel. Two patients harbored the same novel nonsens variant, yet exhibited different phenotypes. Familial studies performed in 17 families, demonstrated that ACAN variants were inherited in 16/17 cases, associated with short stature (16/16), brachydactyly (4/16), early-onset osteoarthritis (3/16), advanced bone age (3/16), obesity (2/16) and midfacial hypoplasia (2/16). Only one variant was found de novo in a patient displaying scoliosis and short stature.

Conclusion: A comprehensive genomic approach is crucial to identify the true proportion of ISS with a monogenic condition.

Aggrecanopathies are heterogeneous and particularly frequent in apparently ISS, sometimes lacking skeletal dysplasia.

Conflict of Interest: None declared

EP06.039 Case report: A rare case of anauxetic dysplasia 3

Seyedmohammad Seyedhassani 1, Massoud Neshan1;2, Neda Tavakkoli Banizi1

1Dr. Seyedhassani Genetic Center, Medical Genetic, Yazd, Iran; 2Yazd welfare Organization, Molecular genetic, Yazd, Iran

Objectives: Anauxetic dysplasia 3 (ANXD3) is a rare spondyloepimetaphysial dysplasia characterized by severe short-limb, short stature, joint hypermobility, dental abnormalities, dysmorphic facial features, other skeletal disorders, and mild intellectual disability. The case was a 27-year-old girl born out of a consanguineous marriage suffering from short stature (70 cm), microcephaly (35 cm), sparse scalp hair, broad eyebrows, low set ears, short metacarpals, trident hands, kyphoscoliosis, pectus excavatum, bowing of the femur, hip dislocation, short toes and brachydactyly and has insulin-dependent diabetes. Evidence of left ventricular hypertrophy and aortic insufficiency was also reported in her echocardiogram. Differential diagnoses of skeletal dysplasia or achondroplasia have been proposed.

Methods: Whole exome sequencing (WES) was done using the Illumina platform and Agilent SureSelect V7 library preparation kit. Confirmation of found mutation and evaluation of the unaffected parents and segregation study of ten relatives were done by Sanger sequencing.

Results: We identified a homozygous NEPRO mutation (c.280C>T, p.Arg94Cys) in the girl by WES. This mutation was categorized as variation unknown significant (VUS). Parents were heterozygous for this mutation. We found heterozygosity in the parents and an inheritance pattern of autosomal recessive.

Conclusions: In this study, we identified a novel mutation in a rare Iranian patient with ANXD3 by whole exome sequencing. The results for the patient and the members of his family will help understand the etiology of the disease, diagnose carriers, assist in genetic counseling, and emphasize the need for DNA-based prenatal screening for families with a history of the disease.

Conflict of Interest: None declared

EP06.040 Investigation of the genetic etiology of short stature

Bahriye Öykü Candan 1, Güven Toksoy2, Ayça Dilruba aslanger2, firdevs baş3, aslı al3, Birsen Karaman4

1Institute of Aziz Sancar Experimental Medicine, Genetics, İstanbul, Türkyie; 2Istanbul Faculty of Medicine, Medical Genetics, İstanbul, Türkyie; 3Division Of Medical Sciences, Child Health and Diseases, İstanbul, Türkyie; 4Child Health Institute, Basic Pediatric Sciences, İstanbul, Türkyie

Background/Objectives: Short stature is defined as height below the 3rd percentile and growth rate below the 25th percentile. Short stature is related to genetic, chromosomal, single gene, and multifactorial factors. This study aims to investigate and elucidate the genetic etiology of short stature.

Methods: In this study, in order to elucidate the genetic etiology of 10 clinically diagnosed short stature patients with unknown underlying causes were evaluated with cytogenetic and molecular tests. Patients who had normal karyotypes were included to the analysis of 25 genes (BMP4, FGF8, FGFR1, GH1, GHR, GHRH, GHSR, HESX1, HHIP, IGF1, IGF1R, IGFALS, IGFBP3, IGSF1, LHX3, LHX4, OTX2, POU1F1, PROKR2, PROP1, SHH, SHOX, SOX3, STAT5B, WDR11) which was carried out on the Ion Torrent platform. Mutations that were thought to have clinical importance were confirmed by Sanger sequencing.

Results: A likely pathogenic novel variant was found at c.412G>T in the SHOX gene of one case and confirmed by Sanger sequencing. Additionally, a pathogenic variant was detected at c.1439del in the WDR11 gene. As a result of the research comparing the reading quality of the variant with each case, it was concluded that it was a fake variant. Additionally, VUS variants were detected in four genes in three cases.

Conclusion: Since short stature has wide genetic background, a gene panel is not sufficient, and a proper approach would be to investigate index individuals, their parents, and healthy controls using whole exome or whole genome sequencing.

Grants: The present work was supported by the Research Fund of Istanbul University (project no: TYL-2022-39328)

Conflict of Interest: None declared

EP06.041 Correlation between genotype and phenotype - some variants in the COMP gene cause MED, others PSACH

Renáta Michalovská 1, Helena Paszekova1, Kristýna Hanuláková1, Tomáš Píš1, Veronika Krulisova1, Ivo Mařík2, Elsa Zemankova3, Dominika Vallusova1, Terézia Haňová1, Zděnka Vlčková1

1GHC GENETICS, Prague; 2Ambulant Centre for Defects of Locomotor Apparatus (ACDLA) in Prague, Prague, Czech Republic; 3E-med, s.r.o., Genetics Benesov, Benesov, Czech Republic

Background/Objectives: Causal variants in the COMP gene encoding cartilage oligomeric matrix protein can cause two skeletal dysplasias, the milder multiple epiphyseal dysplasia (MED) and pseudoachondroplasia (PSACH). We have identified 4 different patients with a causal de novo variant in the COMP gene with different manifestations.

Methods: We used the Clinical Exome Solutions (CES v3) panel by Sophia Genetics to perform massive parallel sequencing on NextSeq550. CES v3 covers the coding regions of 5500 genes, the entire mitochondrial genome and non-coding variants known to be associated with rare and inherited disorders. The SOPHiA DDM™ platform was used to analyze the data and identify variants, including SNVs, Indels, and CNVs.

Results: All 4 causal variants identified in COMP were in the TSP type-3 domain. 3 patients have MED and 1 patient has PSACH. The sequencing variant c.1450T>G(p.Cys484Arg) was previously described in a patient with PSACH, in our patient it corresponds to the diagnosis of MED. Thus, there is a phenotypic continuum between the diseases. The clinical picture of the patients manifests after 2 years of life, but one of our patients (after embryo transfer) has clinical manifestations from 18 months, together with facial dysmorphia and psychomotor retardation.

Conclusion: Mutations in the COMP gene are associated with PSACH and MED syndromes. The diagnosis of these patients is challenging, due to the wide phenotypic range of these syndromes. Therefore, the use of clinical exome sequencing has proven to be beneficial in determining the final diagnosis in patients with skeletal disorders with different clinical manifestations caused by rare inherited disorders.

Conflict of Interest: None declared

EP06.042 Osteoma and Calcinosis Cutis – New Molecular Mechanisms for GNAS-related disorders

Ari Horton 1, Bryony Thompson2, Hnin Pwint Oo3, David Francis4, Ingrid Winship1

1The Royal Melbourne Hospital, Department of Genomic Medicine, Parkville, Australia; 2The Royal Melbourne Hospital, Department of Pathology, Parkville, Australia; 3The Royal Melbourne Hospital, Dermatology Department, Parkville, Australia; 4Murdoch Children’s Research Institute, Victorian Clinical Genetics Services, Parkville, Australia

Background/Objectives: Genetic testing in inherited skin disorders is having greater clinical relevance, as therapeutics become available. Osteoma cutis (OC) and Calcinosis Cutis (CC) are subcutaneous nodules with deposition of bone or calcium below the skin, but can represent multisystemic conditions with enodcrine and neurodevelopmental implications. High-density chromosomal microarray (GDA-cyto, Illumina) can identify pathogenic copy number variation, mosaicism, and inheritence patterns, with major implications for individuals health and reproductive risk.

Methods: Patients attending clinic with mutlifocal OC or CC, with or without family history, are offered genetic counselling and testing. Initial testing involves exome sequencing with application of curated panels including calcium and phosphate disorders, Pseudohypoparathyroidism, Albright Hereditary Osteodystrophy and ACVR1. Subsequent testing in gene elusive patients, includes obtaining a GDA-cyto.

Results: A 41 year old female presented with childhood onset, progressive and multifocal OC confirmed on histopathology and short stature. Her 7 year old daughter was born with multiple congenital and skeletal anomalies. Exome sequencing was uninformative. Subsequent GDA-cyto, demonstrated a 20q13.32 deletion involving deletion of GNAS and STX16. GNAS inactivation can be associated with pseudopseudohypoparathyroidism (PPHP), progressive osseous heteroplasia (POH) and osteoma cutis (OC). Imprinting of the allele has phenotypic implications, with paternal inheritence associated with PPHP compared with the pseudohypoparathyroidism (PHP) of maternal inheritence. A second patient, a 51 year old with facial OC, is currently under investigation.

Conclusion: Osteoma Cutis and calcinosis cutis represent an important phenotype for the utility of genomic sequencing using exome and GDA-cyto to inform care, with imprinting involved in phenotype.

Grants: No grants were obtained for this report.

Conflict of Interest: None declared

EP06.043 The first case of uncombable hair syndrome of Asian origin with pathogenic variants in PADI3

Ye Li 1, Maria Wehner1, Nicole Cesarato1, Yasmina Gossmann1, F. Buket Basmanav1, Regina C. Betz1

1Institute of Human Genetics, University Hospital of Bonn, Bonn, Germany

Background/Objectives: Uncombable hair syndrome (UHS) is a rare hair shaft abnormality diagnosed in children who have dry, frizzy, and often light-colored hair that cannot be combed flat. We have the world’s largest UHS cohort with over 100 affected individuals/pedigrees and had previously discovered that UHS is caused by biallelic pathogenic variants in the genes PADI3, TGM3, or TCHH. So far, UHS has only been described in individuals of European, American and middle Eastern origin. In this study, we report the first case of UHS in an affected girl of Asian origin.

Methods: A 3-year-old girl of Asian origin was referred to us for genetic counseling based on the clinical presentation of frizzy, difficult to comb, dark hair since birth. Sanger sequencing of PADI3 was performed on the DNA of the patient and her parents.

Results: Sequencing analysis of the patient revealed compound heterozygous pathogenic variants, namely, c.363C>A;p.(Cys121Ter) and c.1374dup;p.(Val459Argfs*15) in PADI3. The parents were each heterozygous for one of the two variants. Both of the variants had not been reported for UHS and were either absent (c.363C>A;p.(Cys121Ter)) or rare in genetic databases (c.1374dup;p.(Val459Argfs*15); allele frequency < 0.00001 in gnomAD v.4.0.0).

Conclusion: Hereby, we report the first case of UHS in an individual of Asian origin who carries compound heterozygous pathogenic variants in PADI3; neither of which had previously been found in UHS affected persons of other ethnic origin. Therefore, it remains to be discovered whether these variants could be population specific and/or explain other cases of UHS in Asian populations.

Conflict of Interest: None declared

EP06.044 Focal facial dermal dysplasia type 4 - a case report

Stefanie Van de Voorde 1, Kathelijn Keymolen1, Frederik Hes1, Andrea Buysse1, Willem Verpoest2, Bert Callewaert3, Sofie Symoens3, Aude Beyens3, Ifigenia Spanoudi-Kitrimi4, Boyan Dimitrov1

1Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Clinical Sciences, Research group Genetics, Reproduction and Development, Centre for Medical Genetics, Laarbeeklaan 101, 1090 Brussels, Belgium; 2Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Clinical Sciences, Research Group Genetics of Reproduction and Development, Brussels IVF Centre for Reproductive Medicine, Brussels, Belgium.; 3Center for Medical Genetics, Ghent University Hospital, 9000 Ghent, Belgium; 4Department of Dermatology, Katholieke Universiteit Leuven, Leuven, Belgium

Background/Objectives: Focal facial dermal dysplasia type 4 (FFDD4) is a rare genetic disorder characterized by linear preauricular skin lesions that arise from embryonic fusion defects. FFDD4 is caused by homozygous or compound heterozygous variants in the CYP26C1 gene.

Methods: We performed exome-based virtual gene panel analysis for genodermatoses including 337 genes following detailed clinical phenotyping in an individual with FFDD4. Variants were classified according to the guidelines of the American College of Medical Genetics and Genomics (ACMG).

Results: A 3-month-old boy, born to nonconsanguineous parents, presented with well-defined, hypopigmented skin lesions along the preauricular region. Further clinical assessment showed also bilateral dysplastic ears and a hypopigmented macula with centrally positioned haemangioma on the left shoulder. Genetic analysis confirmed the presence of a homozygous pathogenic variant in exon 4 of CYP26C1 (NM_183374.3): c.845_851dup, p.(Gln284HisfsTer129).

Conclusion: FFDD4 is a distinct subtype of focal facial dermal dysplasia characterized by preauricular skin lesions. Interestingly, the reported herein individual and patients from the literature present with some additional clinical features such as haemangiomas, dysplastic ears and cleft lip/palate. Pending confirmation in additional case series, these findings may represent rare features of this condition associated with CYP26C1 pathogenic variation.

Grants:

/

Conflict of Interest: None declared

EP06.045 Genetics of fetal skeletal dysplasia - our experience

Deepti Saxena 1, AMIT KUMAR TIWARI1, Amita Moirangthem1, Kausik Mandal1, Shubha Phadke1

1Sanjay Gandhi Postgraduate Institute of Medical Sciences, Medical Genetics, Lucknow, India

Background/Objectives: Fetal skeletal disorders are a heterogeneous group of disorders affecting bone development. Without molecular diagnosis, definitive prenatal diagnosis in next pregnancy cannot be provided. Here, we aim to describe our experience regarding the genetic spectrum of fetal skeletal dysplasia.

Methods: The cases were selected based on antenatal ultrasound findings or postnatal examination and the clinical details were collected. Targeted Sanger sequencing or Whole exome sequencing (trio/ solo) based on clinical diagnosis was performed on cases with fetal skeletal dysplasia detected on antenatal ultrasound, selected over a period of two years from September, 2020 to September, 2022.

Results: A total of 23 cases were selected accordind to the inclusion criteris. Majority of the cases (20/23) had short long bones as the presenting feature, some of them had additional findings such as polydactyly, bent bones or evidence of fracture in long bones, increased nuchal fold thickness or hydrops. Three cases had radial ray defect with some extraskeletal manifestations. A pathogenic/ likely pathogenic variation related to the phenotype was detected in 16/23 (69.6%) cases, variant of uncertain significance was detected in 1/23 (4.3%) cases and no variant could be identified in 6/23 (26%) cases.

Conclusion: Our study further emphasizes the role of whole exome sequencing in fetal abnormalities. Confirmation of molecular diagnosis improves pregnancy management, prenatal counselling, and assessment of recurrence risk in further pregnancies. Identification of novel variants further expanded the mutational spectrum of fetal disorders.

Grants: Indian Council of Medical Research (Grant no. 33/2/2019-TF/ Rare/ BMS)

Conflict of Interest: None declared

EP06.046 Whole Exome Sequencing in highly consanguineous families with congenital limb malformation

Mobina Ghofrani Shadman 1, Nathalie Kruse1, Kristian Händler1, Saranya Balachandran1, Varun Sreenivasan1, M. Ijlal Haider2, Neseebullah Kakar1;3, Malte Spielmann1;4;5

1Universitätsklinikum Schleswig-Holstein (UKSH), University of Lübeck and University of Kiel, Institute of Human Genetics, Lübeck, Germany; 2UKSH, Institute for Cardiogenetics, Lübeck, Germany; 3BUITEMS, Department of Life Sciences & Informatics, Quetta, Pakistan; 4Max Planck Institute for Molecular Genetics, Human Molecular Genetics, Berlin, Germany; 5DZHK e.V. (German Center for Cardiovascular Research), Cardiovascular Research, Hamburg

Background/Objectives: Multiple studies have shown that consanguinity could lead to or contribute to an elevated incidence of autosomal recessive genetic disorders. Here, we selected 48 affected individuals from different highly consanguineous Pakistani families with recurring congenital limb malformations, in an effort to identify the genetic etiology.

Methods: Whole exome sequencing (WES) and subsequent variant calling was performed in 48 affected individuals, silent mutations and variants with high prevalence were filtered out and remaining variants were prioritized with respect to their causative potential based on various variant databases using VarFish. This was followed by subsequent variant validation and segregation analysis by Sanger sequencing.

Results: WES analysis yielded a multitude of possible limb and bone associated variants to prioritize, including variants that have previously not been associated with the respective malformations. Validation is ongoing and will be presented.

Conclusion: In conclusion, WES proved to be a highly effective way to decipher the genetic basis of highly consanguineous families with congenital limb malformation. Furthermore, our findings expand the allelic spectrum of skeletal dysplasias.

Grants: M.S. is a DZHK principal investigator and is supported by grants from the Deutsche Forschungsgemeinschaft (DFG) (SP1532/3-2, SP1532/4-1, SP1532/5-1, and SP1532/13-1) and the Deutsches Zentrum für Luft- und Raumfahrt (DLR 01GM1925)

Conflict of Interest: None declared

EP06.047 New PDGFRB variants associated with Kosaki Overgrowth Syndrome: case reports from two unrelated families.

Ruslan Minnebaev1, Anastasiia Kungurtseva2, Nina Demina1, Olga Shchagina1, Anna Orlova1, Alisa Vitebskaya2, Yulia Tikhonovich2, Petr Vasiliev 1

1Research Centre for Medical Genetics, Moscow; 2Sechenovskiy Universitet, Moscow, Russian Federation

Background/Objectives: Kosaki overgrowth syndrome (KOGS) is an extremely rare overgrowth syndrome arising from heterozygous activating variants of PDGFRB. The KOGS phenotype includes distinctive facial features, advanced growth and bone age, abnormalities on brain MRI. Fewer than 15 patients with Kosaki syndrome have been described worldwide at date.

Methods: CES (for patient 1), WGS (for patient 2) and Sanger sequencing were performed.

Results: Here we present two patients from unrelated families.

Patient 1 (8 y.o., male): height – 140 cm(+2.06 SD), head circumference – 49 cm.(-2.39 SD). The patient has pronounced sutures of the skull, rapid growth, skull tuberosity, prominent supraorbital ridge, large hands and feet, pectus excavatum. Brain MRI: partial hypoplasia of the cerebellum and multiple foci of white matter gliosis. New, previously undescribed missense variant in the PDGFRB (NM_002609.4):c.2558G>C p.(Arg853Pro) in heterozygous state was revealed.

Patient 2 (14 y.o., female): height – 186 cm(+3.89 SD), head circumference – 57 cm.(+2.54 SD). The patient has prominent supraorbital ridges, macrosomia, macrocephaly, marbled skin, large hands and feet, and advanced bone age. Brain MRI: multiple foci of white matter gliosis.A variant c.1997A>C(p.Asn666Thr) was identified in PDGFRB gene. This variant was previously described once and associated with Castleman disease. We identified this variant as Likely Pathogenic (PM2/PM5/PP3/PM1) and linked it to Kosaki syndrome.

In both cases, Sanger sequencing was performed, and de novo status was established.

Conclusion: Here, we report two patients with extremely rare Kosaki syndrome from unrelated families and report two new variants in PDGFRB gene, that wasn’t previously associated with a KOGS phenotype.

Grants: no

Conflict of Interest: None declared

EP06.048 The importance of early neuroimaging evaluation and neurosurgical treatment in children with achondroplasia – referral centre experience

Sanda Huljev Frkovic 1, Marija Vidakovic1, Ivan Jovanovic2, David Ozretic2, Hrvoje Jednacak3, Marijan Frkovic1, Branka Runtić Jurić1

1UHC Zagreb, Department of Pediatrics, Zagreb, Croatia; 2UHC Zagreb, Clinical Department for Diagnostic and Interventional Neuroradiology, Zagreb, Croatia; 3UHC Zagreb, Department of Neurosurgery, Zagreb, Croatia

Background/Objectives: Achondroplasia (ACH) is the most common skeletal dysplasia associated with disproportionate short stature. It is caused by a heterozygous pathogenic variant in the FGFR3 gene, which encodes fibroblast growth factor receptor 3. In 80% of cases, the disease is the result of a de novo mutation and is not inherited from the parents. Because the cranial base develops by endochondral ossification, a process that is impaired in ACH, subsequent stenosis of the foramen magnum with associated compression of the medulla oblongata and spinal cord exposes patients at great risk of early neurological complications, including sudden infant death.

Methods: In eight of our patients with ACH, brainstem MRI was performed during the early period of life. Except for stenosis of the foramen magnum, six patients had neuroradiological signs of compression of the brainstem and spine at the time of evaluation. In the four children, compressive myelopathy was found, and only one patient presented with clinical signs of compression with central apnoea, whereas the others had asymptomatic ventriculomegaly with slightly slowed motor development.

Results: In all children with neuroradiologically verified compression at the cervicomedullary joint level, neurosurgical decompression of the foramen magnum and laminectomy were performed. The youngest patient was 7 months old, and the oldest patient was 35 months old at the time of the procedure.

Conclusion: This case series emphasises the need for early neuroimaging evaluation and timely neurosurgical intervention in all children with ACH to prevent potential neurological complications typical of this condition.

Grants: None

Conflict of Interest: None declared

EP06.049 IFIH1 rs1990760 and pulmonary radiological findings among patients with rheumatoid arthritis

Rim Sghiri 1, Hana BenHassine1, Khadija Baccouche2, Nejla Elamri2, Adel Almogren3, Zahid Shakoor3, Foued Ben Haj Slama1, Elyes Bouajina2, Ramzi Zemni1

1Faculty of Medicine of Sousse, Immunogenetics Unit UR14-ES18, Faculty of Medicine, Sousse, Tunisia, Sousse, Tunisia; 2Farhat Hached Hospital, Rheumatology, Sousse, Tunisia; 3King Saud University, Pathology, Riyadh, Saudi Arabia

Background/Objectives: Melanoma Differentiation-Associated protein 5 (MDa5) is a cytoplasmic molecule encoded by the gene IFIH1. Patients with anti-MD5a are at high risk of developing rapidly progressive interstitial lung disease (ILD). Several variants of IFIH-1 have been associated with autoimmune diseases such as rheumatoid arthritis (RA)/ To assess the association between a functional variant of IFIH1, rs1990760 and radiological manifestations among patients with RA.

Methods: rs 1990760 was genotyped in 210 Tunisian RA patients and 224 matched controls by mutagenically separated PCR (MS-PCR). RA patients were assessed by chest radiograph and computed tomography.

Results: Fifty-seven (27.1%) RA patients had pulmonary radiologic changes including ILD in 48 (22.8%) cases, pulmonary nodules in 5 (2.4%) cases, and pleural effusion in 3 (1.4%) cases. Among patients with ILD, 11 (5.2%) had diffuse interstitial pulmonary fibrosis. C allele was present in 52% of RA patients and 49.1% of controls. rs 1990760 was associated neither with RA risk nor with radiological findings except pulmonary fibrosis. C allele was found to be protective against lung fibrosis (OR = 0.2; IC95% = 0.05-0.7; p = 0.007) in the recessive model.

Conclusion: s1990760 C allele was found to be protective against lung fibrosis in RA patients. Large scale studies are needed to confirm this association and to elucidate the exact role of this variant.

Grants: None.

Conflict of Interest: None declared

EP06.050 A very rare disease of Lethal congenital contracture syndrome 9: novel clinical features and ADGRG6 gene variant

Betül Gerik-Celebi1, F. Sırrı Cam 2

1Balıkesir Atatürk City Hospital, Department of Medical Genetics, balikesir; 2Manisa Celal Bayar University, Department of Medical Genetics

A very rare disease of Lethal congenital contracture syndrome 9: novel clinical features and variant

Hamide Betül Gerik-Çelebia, Fethi Sırrı Çamb

aDepartment of Medical Genetics, Balıkesir Atatürk Şehir Hastanesi, Balıkesir, Turkey

bDepartment of Medical Genetics, Manisa Celal Bayar University Faculty of Medicine, Manisa, Turkey

Background/Objectives: Lethal congenital contracture syndrome 9 (LCCS9; MIM no # 616503) is characterized by pulmonary hypoplasia, several skeletal anomalies, flexion contractures, and polyhydramnios. Adhesion G protein-coupled receptor G6 (ADGRG6 gene; MIM no* 612243) variants have been associated with this autosomal inherited disease.

Methods: We performed WES analysis on probands with first-degree consanguineous parents. The proband was a baby girl, the fifth child of this family. She had dysmorphic features, pulmonary hypoplasia, esophageal atresia, bilateral talipes equinovarus, and polyhydramnios. Familial segregation was analyzed by Sanger sequencing.

Results: We identified a novel homozygous ADGRG6 (NM_198569.3): c.3264G>A (p.Trp1088*) variant. This gene variant has not been previously reported in public population databases (Genome Aggregation Database (gnomAD, https://gnomad.broadinstitute.org/), Leiden Open Variation Database (LOVD, https://www.lovd.nl)) and associated current literature.

Conclusion: This study contributed to the phenotypic and mutation spectrum of LCCS9. The novel ADGRG6: c.3264G>A variation leads to a premature stop codon that result in a shorter protein product. It was thought that it may be associated with the poor prognosis of this family. In this study, the tenth family with a history of LCCS9 is presented. This family had esophageal/ gastric atresia, which has not been previously reported in the literature.

Conflict of Interest: None declared

EP06.052 Differential gene expression in degenerative lumbar discs from the Russian disc degeneration study (RuDDS) biobank

Aleksandr Tyapkin 1, Artemii Ivanov1, Olga Leonova2, Aleksandr Krutko2, Alexey Peleganchuk3, Veniamin Fishman1, Elizaveta Elgaeva1;4, Yakov Tsepilov1

1Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russian Federation; 2Priorov Central Scientific Research Institute of Traumatology and Orthopedics (CITO), Moscow, Russian Federation; 3Novosibirsk Research Institute of Traumatology and Orthopedics, Novosibirsk, Russian Federation; 4Novosibirsk State University, Novosibirsk, Russian Federation

Background/Objectives: Degeneration of intervertebral discs (IVDs) can lead to lumbar disc degeneration disease (LDDD), contributing to low back pain, the leading cause of disability worldwide. Despite extensive research, the precise molecular mechanisms underlying LDDD remain unclear. Here, we investigate gene expression differences between conditionally healthy and degenerative IVD from patients in the Russian disc degeneration study (RuDDS) cohort.

Methods: Intraoperative IVD fragments, including 14 conditional control samples (Pfirrmann grades I-III, M51.3, M51.8 ICD-10 codes) and 21 case samples (Pfirrmann grades IV-V, M51.1 ICD-10 code), were subjected to strand-specific paired-end RNA-sequencing using DNBSEQ platform. Read alignment and gene-level quantification were performed with STAR tool and summarizeOverlaps function from GenomicAlignments R package, respectively. Using DESeq2 R package, we performed quality control of count data and identified differentially expressed genes (DEGs) with an IHW-adjusted p-value threshold of 0.05. DAVID 2021 was utilized for GO annotation. BisqueRNA was used for cellular decomposition, based on scRNA-seq IVD atlas data. TPM values were calculated with RNA-SeQC, following GTEx V8 guidelines.

Results: Five case samples failed quality control. Cellular decomposition of remaining samples confirmed high percentage of chondrocytes (over 70%). We found 87 protein-coding DEGs (74 upregulated, 13 downregulated), including known LDDD-associated genes, such as TREM1, FOXC2. Enriched GO terms included skeletal system development and the Wnt signaling pathway.

Conclusion: We identified DEGs enriched in relevant GO terms, suggesting potential drug targets for LDDD treatment.

Grants: The study was supported by the Russian Science Foundation (RSF) grant No. 22-15-20037 and the Government of the Novosibirsk region.

Conflict of Interest: None declared

EP06.053 Whole genome sequencing identifies structural variants in familial osteoporosis

Azra Zejnelagic 1;1, Chanelle Cilia2, Jean Paul Ebejer1, Melissa Formosa2

1L-Università ta’ Malta, Centre for Molecular Medicine and Biobanking, Msida, Malta; 2L-Università ta’ Malta, Department of Applied Biomedical Science, Faculty of Health Sciences, Msida, Malta

Background: Osteoporosis is a metabolic bone disorder with a strong genetic influence, including structural variants (SVs). The study aimed to identify possible disease-causing SVs contributing to osteoporosis by comparing the and output of three SV detection tools following short-read whole genome sequencing (WGS).

Materials and Methods: A 2-generation family having multiple relatives with osteoporosis at the spine or hip was recruited. WGS was performed on 12 out of the 15 recruited relatives on the BGISEQ-500 platform, with sequencing data assessing using BreakDancer, Lumpy and Pindel for SV detection. Genotype calling was required for BreakDancer and Lumpy utilising BreakDown and SVtyper, respectively. Stepwise filtering was performed to prioritise SVs based on their population-based allele frequency, pathogenicity and relevance to bone physiology, whereas shortlisted SVs were confirmed by PCR sizing and Sanger sequencing.

Results: Approximately 400 of the called unique SVs were detected by each tool. Comprehensive variant filtering following a dominant inheritance pattern identified three validated SVs within ARHGEF3, TBX15, and ADAM9 in the Lumpy output, while four validated SVs in SOD2, KLF12, MCTP1, and PTPRM were identified by Pindel. No variants were shortlisted by BreakDancer. Lumpy exhibited superiority over Pindel and BreakDancer, showcasing faster runtime, smaller memory footprint for output files, and minimal system requirements.

Conclusion: Findings suggest that the identified SVs, alone or in combination, could be causal genetic determinants of osteoporosis that require further functional validation. These SVs may provide insights into the underlying genetic architecture of osteoporosis and uncover potential targets for therapeutic intervention.

Conflict of Interest: None declared

EP06.054 Identification of pathogenic mosaic variants on the opposite alleles of the FBN1 gene in a boy with a diagnosis of Marfan syndrome

Marlene Perez 1, Almudena Amor1, Laura Cazón1, Maria Sanchez1, Iria Gómez Díaz1, Rosalía Peteiro1, Ivonne Cárdenas Reyes1, María Valverde1, Soledad García Hernández1;2, Diego Cabrera Argaña1, Xusto Fernandez1, Martin Ortiz Genga1, Juan Pablo Ochoa1

1Health in Code, A Coruña, Spain; 2Hospital Universitario Clínico San Cecilio, Granada, Spain

Background/Objectives: Marfan syndrome (MFS) is an inherited connective tissue disorder with great clinical variability and estimated prevalence between 2-3/10,000, with approximately 25% cases with a de novo pathogenic variant in FBN1. The role of FBN1 mosaicism and its contribution to MFS remains a challenge, with an unknown frequency and information limited to case reports. We report a child with a positive systemic Marfan score, who exhibited two consecutive pathogenic mosaic FBN1 deletions on opposite alleles.

Methods: The patient’s genomic DNA was analyzed using a targeted 64-gene NGS panel which included FBN1. The informative variants in FBN1 were confirmed by Sanger and NextGeneDx® techniques. The frequencies of each of the variants were tested in different tissues (peripheral blood and saliva).

Results:Two consecutive null variants occurring on opposite FBN1 alleles were found at low variant allele frequencies (VAF), indicating a mosaic nature (see table 1). Neither variant was detected in unaffected parents by Sanger sequencing. The most plausible biological explanation is that both de novo mutations occurred in a single mutational event prior to gastrulation (since both variants are present in cells derived from different tissues), and a defective repair in the complementary strand caused that this individual was a carrier of two different populations of mutant cells carrying consecutive variants (two mutated cell populations).

Table 1: VAFs of the mosaic FBN1 mutations in both blood and saliva.

Variant

Blood

Saliva

NC_000015.9:g.48782170delG

NP_000129.3:p.(Trp988Glyfs*11)

18.78%

10.94%

NC_000015.9:g.48782171delC

NP_000129.3:p.(Ala987Profs*12)

24.91%

33.19%

Conclusion: This publication contributes to the biological understanding of Marfan syndrome.

Conflict of Interest: None declared

EP06.055 Novel variant in EDA gene causing mild hypohidrotic ectodermal dysplasia in males

Karolina Biel 1, Aleksander Jamsheer1

1Poznan University of Medical Sciences, Department of Medical Genetics, Poznań

Background/Objectives: Hypohydrotic ectodermal dysplasia (HED) is a group of functional and structural disorders of two or more ectodermal structures caused by mutations in a group of EDA/EDAR/NF-κB genes pathway. They are also involved in the pathogenesis of non-syndromic tooth agenesis (NSTA). It has been shown that NSTA and HED have overlapping clinical phenotype and genetic basis. For some, those two conditions are considered as one disease with different expressivity. This case report describes a family with variably expressed clinical manifestations of HED involving the EDA gene.

Methods: Patients A1 and A2 are 10 and 4-year-old brothers respectively, born to non-consanguineous parents at term. Patient A1 presented conical-shaped teeth, four missing teeth, abnormal hair and hypoplastic nipples. Patient A2 had conical-shaped deciduous teeth with missing two lateral incisors and dry, atopic skin. There was an absence of 12 permanent teeth in the pantomogram, predicting oligodontia. Their relatives presented features of NSTA or mild ectodermal dysplasia. Genetic testing, including EDA sequencing, was performed in patients A1 and A2.

Results: A novel likely pathogenic variant in EDA gene c.543_569del (p.Asn185_Pro193del) was identified in both male individuals. Pathogenic variants in this gene are associated with X-linked hypohydrotic ectodermal dysplasia and non-syndromic tooth agenesis.

Conclusion: These results support the hypothesis that NSTA and syndromic tooth agenesis represent the same disease entity with variable expressivity. These cases are a rare example of mild EDA-related hypohydrotic ectodermal dysplasia observed in males.

Grants: The public health system covered all genetic testing performed.

Conflict of Interest: None declared

EP06.057 Expanding the phenotypic spectrum of Osteogenesis Imperfecta

Diana Prepelita 1, Andreea Tutulan-Cunita1, Anca Pavel1, FLORINA MIHAELA NEDELEA2, VASILICA PLAIASU3, Ina Ofelia Focsa1, Danae Stampoulia1

1Cytogenomic Medical Laboratory, București, Romania; 2Filantropia Clinical Hospital, București, Romania; 3Institutul National pentru Sanatatea Mamei si Copilului “Alessandrescu-Rusescu”, București, Romania

Background. Osteogenesis imperfecta (OI) represents a clinical continuum, ranging from nearly asymptomatic disease with a mild predisposition to fractures to prenatal onset, perinatal lethal disease. Discovery of novel causative variants and further validation of known variants in OI genes continue to impact the clinical care, as a more precise genotype-phenotype correlation is warranted for improved care of these patients.

Methods. Six patients were analyzed using next-generation sequencing (NGS) via a skeletal dysplasia-targeted gene panel or whole exome sequencing (WES), utilizing Illumina technology. Bioinformatics pipelines and dedicated software processed the sequencing data, and variants were interpreted according to ACMG/ClinGen criteria. For prenatal cases, only pathogenic/likely pathogenic variants were considered.

Results. Sequencing revealed pathogenic variants in the COL1A1 gene (NM_000088.3) in all subjects. Two cases of OI type II had heterozygous variants c.1057G>A and c.2596G>A. Types 3 and 4 OI were identified with variants c.1678G>A and c.2775delT, respectively. Two cases with ambiguous phenotypes had heterozygous pathogenic variants c.658C>T and c.3505G>A. The genotype-phenotype correlations aligned with existing literature, with limited data for variants c.1057G>A and c.2775delT. The assessment distinguished between type II and other types, suggesting c.1057G>A is associated with type II, pending further evidence.

Conclusion. The increased heterogeneity of osteogenesis imperfecta complicates genetic counseling. Clarifying genomic variants’ phenotypic spectrum, especially distinguishing between mild and severe forms, significantly enhances medical care and genetic counseling for OI patients.

Conflict of Interest: None declared

EP06.059 Positive association between a common polymorphism within the ESR1 gene and the outcome of brace treatment in patients with idiopathic scoliosis

Svetla Nikolova1, Martin Pasev 1, Milka Dikova2, Alexandre Loukanov3

1Sofia University “St. Kliment Ohridski”, Faculty of Medicine, Biology, Medical Genetics and Microbiology, Sofia, Bulgaria; 2Medical University-Sofia, Faculty of Medicine, Department of Pediatrics, Sofia, Bulgaria; 3National Institute of Technology, Gunma College, Department of Materials Engineering, Gunma, Japan

Background/Objectives: The present study aims to investigate the possible association between 20 polymorphisms in 19 previously reported candidate-genes and the bracing outcome in patients with idiopathic scoliosis (IS).

Methods: The association study was conducted on 120 Bulgarian patients who have undergone brace treatment after being diagnosed with idiopathic scoliosis. The cases were divided into two subgroups based on curve progression. The mean Cobb angle was 60.0  ±  17.1° in the progressive group (n = 71), and 21.6  ±  6.2° in the non-progressive group (n = 49). The mean age was 11.2 ± 2.9, and 11.6 ± 2.0 years in the progressive and non-progressive group, respectively. The genotyping was carried out by TaqMan Real-Time PCR method. Differences of genotype and allele distribution between the groups were compared by Fisher Exact Probability Test with p-value less than 0.05 as statistically significant.

Results: In the progressive group (Cobb angle above 40°), the frequency of the variant pp genotype of PvuII (rs 2234693) in ESR1 was higher than that in the non-progressive one (p  <  0.05). No significant association was detected for the other 19 polymorphisms in ESR1, CHD7, MTNR1B, IL17RC, GPR126, TGFB1, LBX1, CHL1, IL-6, MMP3, TPH1, MATN1, ACE, ACTN3, AMPD1, VDR, BMP4, Lep, and IGF-1 genes (p  > 0.05).

Conclusion: The results suggest a positive association between a common variant of ESR1 gene (PvuII, rs 2234693) and the bracing outcome in Bulgarian patients with IS. Further population-based studies are needed to confirm these results.

Grants: MEXT/JSPS KAKENHI №T20K05260; Jikoshunyu Kyoinhaibun-keihi №T5452.

Conflict of Interest: None declared

EP06.060 Three variants in DYNC2H1 gene in a girl with Skeletal dysplasia

Maria Sredkova-Ruskova 1, Todor Ruskov1, Tsvetina Veleva1, Trayan Delchev1, Darina Kachakova-Yordanova2;3, Kalina Mihova2;3, Kunka Kamenarova2;3, Radka Kaneva2;3, Daniela Avdjieva-Tzavella1

1University Pediatrics Hospital, Medical University, Department of Clinical Genetics, Sofia, Bulgaria; 2Molecular Medicine Center, Department of Medical Chemistry and Biochemistry, Medical Faculty, Medical University, Sofia, Bulgaria; 3Laboratory of Genomic Diagnostics, Department of Medical Chemistry and Biochemistry, Medical Faculty, Medical University – Sofia, Bulgaria

Background/Objectives: Mutations in DYNC2H1 gene affect the function of primary cilia and cause a heterogeneous group of diseases such as skeletal dysplasia. DYNC2H1 gene encodes a protein called dynein-2, that is involved in intraflagellar transport within the cilia. Extensive genetic variation lead to phenotypic variability. Herein, we present a girl with isolated skeletal involvement and three variants in DYNC2H1 gene.

Case report: Girl presented at the age of 1 y 10 m with distorsion of the left lower leg. She was consulted with orthopedic surgeon and diagnosis of Morbus Blount was made. The child presented in our clinic at age of 11 years with short stature and obesity. Mesomelic shortening of upper and lower limbs, lateral deviation of forearms and lower legs, deformity of wrists were noticed. Non-skeletal involvement was not identified. Normal ultrasound of heart and abdomen.

Methods: Whole exome sequencing(WES) of skeletal dysplasia panel was performed.

Results: The following heterozygous variants with unknown significance in DYNC2H1 gene were identified:

  • c.10516G>T(p.Ala3506Ser) in exon 69, inherited from healthy mother

  • c.1326A>C(p.Glu442Asp) in exon 9, and

  • c.12745G>A(p.Val4249Met) in exon 88, both inherited from healthy father.

Conclusion: Variants c.1326A>C and c.12745G>A affect one allele and variant c.10516G>T affects the other allele of DYNC2H1 gene. This give us the reason to assume that the combination of the three variants is responsible for the observed phenotype despite classification of variants as VUS. Genotype-phenotype correlations need to be determined.

Grant references: none

Grants:

Conflict of Interest: None declared

EP06.061 Metaphyseal Dysplasia, Spahr Type: A Case Study with Exome Sequencing and Long-term Follow-up

Francesco Marco Parodo 1;2, Giulia Gori2, Annarita Giliberti2, Giovanna Traficante2, Elia Dirupo2, Elena Andreucci2, Giorgia Mancano2, Sara Bargiacchi2, Rosangela Artuso2, Stefano Stagi3;4, Laura Papi1;5, Angela Peron1;6

1University of Florence, Department of Clinical and Experimental Biomedical Sciences ‘Mario Serio’, Firenze, Italy; 2Meyer Children’s Hospital IRCCS, Medical Genetic, Firenze, Italy; 3Meyer Children’s Hospital IRCCS, Endocrinology Unit, Department of Pediatrics, Firenze, Italy; 4University of Florence, Department of Health Sciences, Firenze, Italy; 5Careggi University Hospital, Medical Genetics Unit, Firenze, Italy; 6Meyer Children’s Hospital IRCCS, Medical Genetics, Firenze, Italy

Background/Objectives: Metaphyseal dysplasia, Spahr type (MDST) is a rare skeletal dysplasia characterized by moderate postnatal short stature and progressive bowing of the legs, caused by biallelic pathogenic variants in the MMP13 gene. To date, only 12 affected individuals have been reported. We describe a 13-year-old boy with skeletal disproportion, decreased stature (10th percentile), lower limbs curvature, and joint pain. Initially suspected of rickets he has been monitored for 13 years since his growth trajectory was significantly below average (SD for height from -3.11 to -0.41), showing gradual improvement over time.

Methods: Trio exome sequencing was performed.

Results: Two compound heterozygous variants in MMP13 [NM_002427.4] were identified: c.619T>G;p.(Trp207Gly), and c.673G>A;p.(Gly225Ser) inherited from the mother and father, respectively. The c.673G>A variant is novel and classified as likely pathogenic according to the ACMG guidelines (PM3, PP3, PM1, PM2), while the c.619T>G variant is classified as pathogenic (PM3, PP5, PP3, PM1, PM2). The patient’s clinical manifestations and the molecular findings - including the specific location of the variants in exons 4 and 5 - are consistent with a diagnosis of MDST.

Conclusion: This case highlights the crucial role of exome sequencing in the early and precise diagnosis of skeletal dysplasias, leading to proper clinical management and avoiding inappropriate treatments. This is especially true for MDST, which mimics rickets, a primarily non-genetic disorder. The extended follow-up of the patient offers valuable data, enriching our understanding of the natural history and progression of MDST, and the previously unreported variant in MMP13 expands the mutational spectrum of MDST.

Grants: None received.

Conflict of Interest: None declared

EP06.062 Clinical exome sequencing (CES) identifies a novel homozygous variant in NECTIN1 causing CLPED1

Elif Yilmaz Gulec 1;2, Gulden Yorgancioglu Budak3

1Istanbul Medeniyet University, Medical School, Department of Medical Genetics, İstanbul, Türkyie; 2Istanbul Goztepe Prof. Dr. Suleyman Yalcin City Hospital, Department of Medical Genetics, Istanbul, Türkyie; 3İstanbul Health and Technology University, Faculty of Medicine, Department of Medical Biology, İstanbul, Türkyie

Background/Objectives: Cleft lip/palate-ectodermal dysplasia syndrome 1 (CLPED1; OMIM#225060), also referred as OFC7, is a rare genetic disorder characterized with several malformations including unique facial appearance with cleft lip/palate, hypotrichosis, ectodermal dysplasia and in some cases, intellectual disability. Autosomal recessive NECTIN1 (OMIM#600644) mutations are linked to CLPED1 etiology. We evaluated four-year-old-affected girl of Turkish origin born to a consanguineous parents. Among the physical examination findings of proband were cleft lip/high narrow palate, low weight and height. She also had mild pectus excavatum, hypotrichosis, ear and dental dysplasies, frontal bossing and no speech.

Methods: Clinical exome sequencing was performed to blood DNA sample of proband. In the NGS process, Illumina SBS (sequence by synthesis) technology was used and each nucleotide was read at a depth of at least 50x. ACMG 2015 guideline was taken as reference. 1000 genome project, dbSNP and Exac data were used as control population.

Results: Novel homozygous variant in NECTIN1 gene (NM_002855.5 c.1127_1139del6/6) was detected resulting in a frameshift (p.Val376GlyfsTer218) and an alternative stop codon at Exon6-3’UTR. Sanger sequencing was used for confirmation of suspected variant. Her parents and one of healthy sisters were heterozygous for NECTIN1, other sister was homozygous normal supporting autosomal recessive inheritance.

Conclusion: NECTIN1 encodes for a transmembrane cell-cell adhesion protein Nectin-1 that constructs adherens and tight junctions. Nectin-1 has a conserved C-terminal amino acid motif (Glu/Ala-X-Tyr-Val), where Afadin (AFDN; OMIM#159559) interacts via its PDZ domain. Our novel variant disrupted this motif which may affect Nectin-Afadin interaction and cause defective cytoskeletal organisation.

Conflict of Interest: None declared

EP06.063 NT5E-related calcification of joints and arteries in three Omani families due to a founder variant

Ghada A. Otaify 1;2, Katta M. Girisha3, Khalid S. Al Thihli1, Ahmed Alsariri4

1Genetic and Developmental Medicine Clinic, Sultan Qaboos University Hospital, Muscat, Oman, Genetics, Muscat, Oman; 2Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt, Clinical Genetics, Cairo, Egypt; 3Department of Genetics, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman, Genetics, Muscat, Oman; 4Armed Forces Hospital, Rheumatology department, Muscat, Oman

Background/Objectives: Calcification of joints and arteries (CALJA) is an ultra-rare, hereditary autosomal recessive disorder characterized by adult onset slowly progressive ectopic calcification that involves predominantly joint capsules and arteries of lower extremities leading to chronic arthritis and lower limb claudication. The disease is caused by mutations in the NT5E, which is responsible for pyrophosphate metabolism. Twenty-three patients from twelve families have been reported to date. Herein we report four patients from three unrelated Omani families.

Materials and Methods: Mutation analysis was performed using WES and sanger sequencing.

Results: the four patients ranging from 31 to 37 years old started their manifestations in the second decade with episodic attacks of seronegative arthritis that are intermittent and progressive. Their radiographs revealed periarticular calcification in different joints. Computed tomography of coronary arteries in two patients showed no calcifications and coronary calcium score was zero. There was no report of vascular calcification or intermittent claudication till now. Exome sequencing was conducted on the index cases of the three families and all harbored the same homozygous likely pathogenic variant c.1360G>A p.(Gly454Arg) in NT5E gene. The substitution is in close proximity to the highly conserved donor splice site. It was confirmed by Sanger sequencing and was identified in the affected sister in the second family.

Conclusion: This is the first report of three families with CALJA from the Middle East to shed light on this ultrarare disorder to consider in the differential diagnosis of patients with seronegative arthritis. We believe this represents a founder variant among Omani patients.

Conflict of Interest: None declared

EP06.064 From clinical observation to genetic confirmation: Somatic mosaic mutations in RHOA on ectodermal dysplasia with multi-system involvement

Enise AVCI DURMUŞALİOGLU 1, Yusuf Dogan1, Turkan Turkut Tan1, Dilsah Cogulu2, Esra Isik1, Ozgur Cogulu1, Tahir Atik1

1Ege University, Pediatric Genetics; 2Ege University, Pedodontics

Background/Objectives: Ectodermal Dysplasia with Facial Dysmorphism and Acral, Ocular, and Brain Anomalies (EDFAOB) is an exceedingly rare neuroectodermal syndrome with only 12 known cases. It arises from somatic mosaic mutations in RHOA gene and manifests with linear skin hypopigmentation, asymmetry of the face and limbs, dental and acral anomalies, and leukoencephalopathy, while typically preserving intellectual and neurological functions. In this report, we present two diagnosed cases of EDFAOB and aim to highlight hallmark clinical and genetic features of the syndrome.

Case Report: Two unrelated cases, aged 6 and 10, were presented with notable facial-body asymmetry, thin hair, epicanthus, dental issues, digital anomalies and Blaschko’s lines-aligned hypopigmentation. Both demonstrated normal neuromotor development without intellectual or neurological impairment. Case 1, 6-year-old girl, had esotropia and visual center atrophy; her MRI indicated bilateral white matter hyperintensities. Case 2, 10-year-old girl, MRI displayed unilateral hyperintense lesions in the left cerebral hemisphere. Prompted by their clinical signs, RHOA gene sequencing from hypopigmentated skin biopsies was conducted, confirming EDFAOB with the c.139G>A (p.Glu47Lys) mutation in allele fractions of 20% and 10%, respectively, absent in blood leukocytes and parental DNA.

Conclusion: These cases not only underscore the clinical and genetic features of EDFAOB but also stress the importance of thorough clinical evaluation. Such evaluation is critical in guiding the selection of precise genetic testing from the outset. The identification of the mutation exclusively in affected tissues corroborates a postzygotic mosaic distribution and refines the diagnostic process for this rare syndrome.

Conflict of Interest: None declared

EP07 Cardiovascular Disorders

EP07.002 Understanding the Genetic Architecture of Hypertrophic Cardiomyopathy in Pakistan - Experience from a Single Tertiary Healthcare Centre

Ameema Asad1, Fizza Akbar1, Asra Wahid2, Yawer Saeed3, Fateh Ali Tipoo3, Salman Kirmani 1

1Aga Khan University Hospital, Department of Women and Child Health; 2Jefferson Einstein Hospital-Philadelphia; 3Aga Khan University Hospital

Background/Objectives: Cardiomyopathies are broadly characterized by myocardial abnormalities. Hypertrophic cardiomyopathy (HCM) is the most common, caused by over 1500 pathogenic (P)/likely pathogenic (LP) variants in 26 genes. However, since most of this data is derived from individuals of European descent, we aimed to perform genetic analysis of HCM in Pakistani patients.

Methods: We conducted a retrospective review and prospective recruitment of patients from February 2021 to February 2023, who exhibited morphological findings consistent with HCM at the Aga Khan University Hospital Karachi, Pakistan. Our prospective patients underwent whole exome sequencing at 3billion, South Korea.

Results: Of the 67 patients (17 females, 50 males, 10 were under the age of 18), 25 (37%) had one or more (P/LP) variants in genes involved in HCM. Thirty percent harbored variants of uncertain significance (VUS), while 33% had negative results. Reverse phenotyping and variant analysis increased diagnostic yield from 37% to 46%. MYBPC3 was the most common gene (43%), followed by MYH7 and TNNI3. Pediatric patients with biallelic variants had autosomal recessive disease, leading to severe neonatal cardiomyopathy and subsequent deaths.

Conclusion: This study reports pathogenic or likely pathogenic variants in 31 patients across 10 genes, with a diagnostic yield of 46%, consistent with global literature. We also observed that consanguinity led to severe neonatal cardiomyopathy. Given this, access to genetic testing is crucial for earlier diagnosis, management, and genetic counseling as variant-specific therapies are developed, emphasizing the importance of addressing the genetic needs of under-represented populations.

Grants: Study was funded through intramural grant.

Conflict of Interest: None declared

EP07.003 The impact of longitudinal multidisciplinary care of early-diagnosed Alström syndrome

Dalma Ruzsics 1, Katalin Csonka2, Viktória Szabó3, Vera Goda4, Rita Bertalan1, Éva Pállinger5, Luca Kamilla Li1, Zoltán Liptai1, Csaba Vilmányi6, Csaba Bödör2, Árpád Ferenc Kovács1

1Semmelweis University, Pediatrics Center Tűzoltó Street Department, Budapest, Hungary; 2Semmelweis University, Department of Pathology and Experimental Cancer Research, Budapest, Hungary; 3Semmelweis University, Department of Opthalmology, Budapest, Hungary; 4South-Pest Central Hospital, National Institute of Hematology and Infectology, Budapest, Hungary; 5Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary; 6Gottsegen National Cardiovascular Center, Budapest, Hungary

Background: Alström syndrome is a multisystemic disorder characterized by congenital cardiomyopathy elicited by biallelic pathogenic variants in ALMS1. The diagnosis of the disease is often missed in early age due to the heterogenic symptom spectra. A significant proportion of symptoms may arise secondary from the progression of key organ involvement. The aim of our case report is the depiction of the early multidisciplinary care role in phenotype progression.

Methods: Seven-month-old male patient was evaluated for neonatal cardiomyopathy, delayed development and multiple minor anomalies. Genetic anamnesis, 5 generation pedigree, minor anomaly mapping were performed during pre-test counselling. Diagnostic testing was performed using (TruSight Cardio NGS Kit, 174 gene) cardiomyopathy gene panel sequencing. Segregation analysis of the parents was performed. The effect of 26 months of prospective multidisciplinary care has been evaluated.

Results: TruSight Cardio NGS sequencing revealed compound heterozygous pathogenic variants in ALMS1:c.8002C>T and ALMS1:c.11310_11313del. The parents are heterozygous carriers. A high phenotypic burden at diagnosis could be observed: vertical nystagmus with photoaversion, dilatated cardiomyopathy, obesity, recurrent infections and delayed psychomotor development. Evaluation at 33 months of age revealed stabilization of the cardiac status, visual and hearing impairment, amelioration of metabolic homeostasis, mild developmental delay and onset of pulmonary dysfunction.

Conclusions: Compared to literature data, the impact of multidisciplinary care seems to be pivotal in slowing down the progression of major organ involvement.

Grants: Hungarian National Research, Development and Innovation Office – NKFIH, OTKA PD_21 138521.

Conflict of Interest: None declared

EP07.004 Gene polymorphisms of angiopoietin-like protein 8 and plasminogen activator inhibitor-1 as potential effector of CAD

Aslıhan Gizem Bilgin 1;2, Berkay Ekici3, Aycan Fahri Erkan3, Neslihan Coban1;2

1Istanbul University Aziz Sancar Institute of Experimental Medicine, Genetics, Istanbul; 2Istanbul University Institute of Graduate Studies in Health Sciences, Genetics; 3Ufuk University Faculty of Medicine, Cardiology, Ankara, Türkyie

Background/Objectives: Atherosclerosis, which occurs through the accumulation of lipids in arterial walls, is the main reason for coronary artery disease (CAD), one of the leading causes of mortality worldwide. Angiopoietin-like protein 8 (ANGPTL8), which inhibits lipoprotein lipase, and plasminogen activator inhibitor-1 (PAI-1), which inhibits fibrinolysis, play key roles in atherosclerosis. In our study, we investigated the association between ANGPTL8 (rs2278426, C/T) and PAI-1 (rs1799762, 4G/5G) polymorphisms carriage and CAD in Turkey adults.

Methods: 528 unrelated individuals who underwent coronary angiography were included in this study and were grouped into non-CAD (<= 30% stenosis) and CAD (>=50% stenosis). Genomic DNA was isolated from peripheral blood samples of individuals and were genotyped for the polymorphisms using a real-time PCR. The presence of (CT + TT) genotypes for ANGPTL8, (4G/5G + 5G/5G) genotypes for PAI-1-5G and (4G/5G + 4G/4G) genotypes for PAI-1-4G are indicated by (+) and their absence are indicated by (-).

Results: CAD patients with ANGPTL8(+)/PAI-1-5G(+) had higher HDL levels than patients with ANGPTL8(-)/PAI-1-5G(-) (p = 0.036). The statistical significance of HDL level remained in the male-CAD group (p = 0.017). Moreover, non-CAD male individuals with ANGPTL8(+)/PAI-1-5G(-) had higher LDL levels than with ANGPTL8(-)/PAI-1-5G(+) (p = 0.030). Additionally, male-CAD patients with ANGPTL8(+)/PAI-1-4G(+) had higher LDL (p = 0.007) and HDL (p = 0.042) levels with ANGPTL8(-)/PAI-1-4G(-). On the other hand, female CAD patients with ANGPTL8(+)/PAI-1-4G(-) had lower HDL (p = 0.012) level than with ANGPTL8(+)/PAI-1-4G(+).

Conclusion: This study suggests that the carrying two polymorphisms together is at least as effective on CAD as alone and effect of carrying together two polymorphisms on CAD remains to be elucidated by functional experiments.

Conflict of Interest: None declared

EP07.007 Relationship of the species ratio of bacteria and the effectiveness of therapy for arterial hypertension in men and women with insulin resistance

Gulshara Abildinova 1;1, Maxsim Solomadin2, Anel Rakhimova3, Aida Yessentayeva3, Anna Borovikova3;4, Zhanna Zhabakova3

1Medical Center Hospital of the President’s Affairs Administration of the Republic of Kazakhstan, Laboratory genetics, Astana, Kazakhstan; 2Karaganda Medical University, Laboratory genetics, Karaganda, Kazakhstan; 3Medical Center Hospital of the President’s Affairs Administration of the Republic of Kazakhstan, Laboratory genetics, Astana; 4, Laboratory genetics, Kazakhstan

Background/Objectives: In aim to study predictors of the effectiveness of arterial hypertension therapy in 199 patients with insulin resistance and with a body mass index of 26.8 ± 5.1 and a waist circumference of 90 ± 13.5 cm, a study of the species ratio of the microbiome was conducted.

Methods: DNA isolated from a homogenized fecal sample in accordance with the manufacturer’s instructions.

General characteristics of the lifestyle of patients: 56% consumed vegetables at least 2 times a day, 41% consumed fruits daily, sleep duration of 6-8 hours was noted by 41% and a favorable lifestyle - 37%, optimal physical activity - 14%. 52% suffered from arterial hypertension, the duration of treatment was 4.6 ± 4.2. Ineffective treatment of arterial hypertension was noted in 7.5% of patients.

Results: The average age of the patients was: 49.2 ± 7.2 years, of which 73 were men, 126 - women. Predictors of ineffective treatment of arterial hypertension in men were the bacteria Firmicutes (p = 0.000000), Bacteroidetes (p = 0.0000009) and Prevotella (p = 0.000000). Among the phylum Bacteroidetes, B. caccae (p = 0.0000000) and B. cellulosilyticus (p = 0.0017) were dominant. The Bifidobacteriales fillet (p = 0.025) was dominated by B. longum (p = 0.00000000). In women, the phyla Actinobacteria (p = 0.035), Bacteroidetes (p = 0.000000) and Firmicutes (p = 0.0000000) were identified as predictors of ineffective treatment.

Conclusion: Thus, in patients with insulin resistance, the effectiveness of treatment for arterial hypertension correlated with the species ratio of the gut microbiome and depended on gender.

Conflict of Interest: None declared

EP07.008 Up-To-Date Analysis of Genetic Variants in Cardiovascular Disease-Associated Genes across a Diverse Cohort of Mendelian Conditions Patients and Electronic Health Records

Nadia Akawi 1, Fatma Aljasmi1

1United Arab Emirates University, Genetics & Genomics, United Arab Emirates

Background/Objectives: The genomic landscape of inherited cardiovascular disease (iCVD) is rapidly changing with new disease genes reported frequently. Clinical and genetic heterogeneity can be influenced by various factors, including differences in population demographics, variant interpretation methods, and reporting approaches.

Methods: We examine the most recent set of genes linked to iCVD forms. We performed enrichment analysis for associated pathways. We conducted a comprehensive analysis of iCVD variants in a cohort of 3,350 individuals from the United Arab Emirates (UAE).

Results: Here, we present an updated repository of 735 genes linked to the primary forms of iCVD, which encompass cardiomyopathy, cardiac arrhythmia, congenital heart disease, connective tissue disorders, pulmonary heart disease, and arteriopathies. We identified the enriched biological processes and pathways associated with these genes and have highlighted common pathways, including Apelin, FoxO, and Ras signalling, which are implicated in all forms of iCVD. Most of these variants were found to be rare and unique to the UAE population, distinguishing them from other populations. Within the Emirati Mendelian cohort, which includes families affected by rare genetic diseases, we identified a burden of pathogenic variants in families with CVD but did not observe a significant burden of variants of uncertain significance. Additionally, we re-evaluated the pathogenicity of variants reported in the electronic health records, which led to the discovery of more than 110 new iCVD-causing variants.

Conclusion: This study marks the first comprehensive profiling of iCVD variants in the UAE, a population that has been underrepresented in both public databases and clinical literature.

Grants: 12M106, 12M125, 31M491

Conflict of Interest: None declared

EP07.009 yield of gene panel-based genetic diagnostics in arrhythmia patients.

Nola Šiljić 1;2, Imke Christiaans1;2, Jan Jongbloed1;2

1University of Groningen, Groningen, Netherlands; 2University Medical Center Groningen, Department of genetics, Groningen, Netherlands

Background/Objectives: Because of genetic and phenotypic heterogeneity, patients suffering from inherited arrhythmias are provided with multigene panel testing using NGS. Detecting (likely) pathogenic variants in these patients enables gene-specific management for the risk of sudden arrhythmic death (SAD) as well as primary prevention of SAD in patients’ asymptomatic relatives. This study presents the yield of genetic diagnostics in primary arrhythmia syndrome patients at the University Medical Centre Groningen (UMCG).

Methods: We collected the data of probands in whom arrhythmia gene panels were analyzed between 2018, when the panels were introduced in patient care, and February 2023.

Results: An arrhythmia gene panel was performed in 197 patients. One or more variants classified as (likely) pathogenic ((L)P) or variant of uncertain significance (VUS) were identified in 59 patients (29.9%). Of these, 13 (6.6%) P and 4 (2%) LP variants were detected in 17 patients, in some cases together with an additional VUS. Moreover, in 42 patients (21.3%) only one or more VUS were detected, of which 16 (8.1%) had a “high VUS”. Multiple variants were detected in 10 patients (5%). Most (likely) pathogenic variants were identified in KCNQ1 (4), RYR2 (4) and KCNH2 (3).

Conclusion: Overall detection rate of variants classified as VUS or higher is 29.9%, with the majority classified as VUS. Most (L)P variants were detected in well-known arrhythmia genes, for which more evidence is available enabling the classification of these with higher confidence. Ongoing studies are focused on finding more proof for involvement of VUS identified in less studied genes.

Grants:

-

Conflict of Interest: None declared

EP07.010 Aspirin alleviates fibronectin-induced preeclampsia phenotypes in a mouse model and reverses fibronectin-mediated trophoblast invasiveness under hypoxia by regulating ciliogenesis and Akt and MAPK signaling

Meitsz Su 1, Hoi-Lam Tam2

1National Cheng Kung University Hospital, OB/Gyn, Tainan, Taiwan; 2National Cheng Kung University, OB/Gyn, Tainan, Taiwan

Background/Objectives: The placenta experiences a low-oxygen stage during early pregnancy. Aspirin is effective in preventating preeclampsia. Elevation of fibronectin (FN) level is associated with preeclampsia; however, the role of FN in the hypoxic phase and whether aspirin exerts its effect at this hypoxic stage remain unknown.

Methods: We determined pregnancy outcomes by injecting recombinant FN into C57BL/6 mice on GD 8 and GD12. One group of FN-injected mice was fed aspirin from GD3 to GD16. Maternal blood pressure, urine protein and fetal/placental weight were recorded. The effects of FN, the underlying pathways on trophoblast biology under hypoxia were investigated in FN-pretreated or FN-knockdown HTR-8/SVneo cells using cell viability, migration, invasion, Western blot and immunofluorescence assays in a hypoxic chamber. The impact of aspirin on FN-mediated trophoblast motility and pathways was evaluated.

Results: Preeclampsia-like phenotypes and fetal growth restriction developed in FN-injected pregnant mice. Trophoblast FN expression was upregulated under hypoxia, which could be suppressed by aspirin treatment. Moreover, FN inhibited trophoblast invasion and migration under hypoxia, which occurred through downregulating ZEB1/2, MMP 9 and the Akt and MAPK pathways. Aspirin was shown to reverse the FN-mediated inhibitory effect on trophoblast invasion/migration and ciliogenesis.

Conclusion: FN overexpression induces preeclampsia-like symptoms and impairs fetal growth in mice. Hypoxic conditions stimulate FN upregulation in trophoblasts. Aspirin exerts its suppressive effect on FN upregulation and FN-mediated cell function in the hypoxic stage of early pregnancy and therefore provides a preventative effect on preeclampsia development.

Grants: Ministry of Science and Technology, Taiwan (MOST 111-2314-B-006 -076 -MY3)

Conflict of Interest: Meitsz Su Ministry of Science and Technology, Taiwan, Hoi-Lam Tam: None declared

EP07.012 Next Generation Sequencing (NGS) as a useful tool for differentiating Athlete’s Heart and Cardiomyopathy in apparently healthy young athletes

Špela Stangler Herodež1, Tomaž Podlesnikar2, Lara Forstner1, Nadja Kokalj Vokač1, Matjaž Vogrin3, Danijela Krgović 1

1Maribor University Medical Centre, Clinical Institut of Genetic Diagnostics, Maribor, Slovenia; 2Maribor University Medical Centre, Division of Surgery, Department of Cardiac Surgery, Maribor, Slovenia; 3Maribor University Medical Centre, Division of Surgery, Department of Orthopaedics, Maribor, Slovenia

Background: Intensive, prolonged endurance and strength training in apparently healthy young athletes causes many physiologic and structural adjustments in heart, known as athlete’s heart. The correct distinction between physiological adaptations and pathological changes is crucial for the athlete, therefore screening programmes are recommended by medical and sports associations. Nevertheless, a small but not negligible proportion of young competitive athletes die by sudden deaths. In 25% the cause of sudden cardiac death is genetic and hereditary. Understanding the disease and determining the genetic component is of key importance for disease prognosis, therapy and the likely outcome of treatment. With NGS method it is possible to overcome these problems.

Methods: NGS analysis was performed for the sequencing of 41 athletes, currently without health problems, with the Illumina TruSight Cardio panel, targeting 174 genes involved in several cardiac disorders. Analysis and interpretation of the NGS data was done with Variant Studio (Illumina) software and free-access tools and databases.

Results: In 3 athletes pathogenic/probably pathogenic variants were found. The variants of unknown significance (VUS) were identified in 8 athletes. For all of them a more detailed follow-up with a cardiologist was advised.

Conclusion: We can conclude that NGS analysis is the key to fast and accurate clinical intervention, which enables correct differentiation between athlete’s heart and cardiomyopathy. In this way, we do not confuse the initial pathological changes with physiological ones, which could be fatal for the athlete with the continuation of competitive sports.

Grants: The work was supported by internal project IRP-2021/01.

Conflict of Interest: None declared

EP07.013 Chat-based physician consultation supports preventive actions in individuals at elevated risk of coronary heart disease

Eero Aaltonen 1, Nina Mars1;2, Sanni Ruotsalainen1, Roope Ripatti1, Johanna Aro1, Kristina Hotakainen3;4, Marianne Leinikka3, Iiro Heikkilä3, Elisabeth Widén1, Samuli Ripatti1;2;5

1Institute for Molecular Medicine Finland, Helsinki, Finland; 2Broad Institute of MIT and Harvard, Cambridge, United States; 3Mehiläinen Oy, Helsinki, Finland; 4University of Helsinki, Clinical Chemistry and Haematology, Helsinki, Finland; 5University of Helsinki, Public Health, Helsinki, Finland

Background/Objectives: A key challenge in coronary heart disease (CHD) prevention is motivating individuals at elevated risk to mitigate their risk. We evaluated how communicating CHD risk information based on clinical risk factors and polygenic risk score (PRS) through mobile device, coupled with an easily accessible chat-based consultation with a physician, would encourage risk-lowering action among individuals at elevated risk.

Methods: We studied 4808 Finnish individuals who had at least one of the following risk factors for CHD: BMI > 27, hypertension, or current smoking. 1050 of them (mean age 50.0, 59.8% women) received CHD risk information on their mobile device with a chat consultation opportunity. We sampled matched controls (N = 3758, mean age 53.3, 62.9% women) from an equivalent cohort study in which CHD risk was communicated online without chat consultation.

Results: Having the chat consultation available increased the likelihood of contacting a physician compared to matched controls (OR 1.81; 95%CI 1.42–2.31; p = 1.4×10-6). The effect size was similar in individuals at elevated risk (OR 1.84; 95%CI 1.33–2.55; p = 2.2×10-4). Almost all (99%) participants considered the clinical and genetic risk information useful when communicated through mobile device, with 91.1% believing that physicians are capable of utilizing genetic risk information in their care.

Conclusion: Allowing for a chat-based physician consultation both increased the likelihood of contacting a physician to discuss the risk information and supported preventative actions. Our findings support communication and consultation of genetic and clinical risk information through mobile app for primary prevention of CHD.

Grants:

Conflict of Interest: Eero Aaltonen: None declared, Nina Mars: None declared, Sanni Ruotsalainen: None declared, Roope Ripatti: None declared, Johanna Aro: None declared, Kristina Hotakainen Mehiläinen Oy, Marianne Leinikka Mehiläinen Oy, Iiro Heikkilä: None declared, Elisabeth Widén: None declared, Samuli Ripatti: None declared

EP07.014 Investigation of the Effect of miR-155, let-7c and miR-433 microRNA Expressions on Congenital Heart Diseases in Down Syndrome Individuals

Tuba Maras1, Sinem YALÇINTEPE 1, Hazal Sezginer Guler1, Murat Deveci2, Necdet Sut3

1Trakya University Faculty of Medicine, Medical Genetics, Edirne, Türkyie; 2Trakya University Faculty of Medicine, Department of Pediatry, Division of Pediatric Cardiology, Edirne, Türkyie; 3Trakya University Faculty of Medicine, Department of Biostatistics and Medical Informatics, Edirne, Türkyie

Background: Down syndrome (DS), a common autosomal chromosome aneuploidy, leads to various developmental disorders in individuals, including intellectual disability and congenital cardiac anomalies. The presence of an extra chromosome 21 (HSA21) is implicated in the pathological manifestations of DS. Congenital heart disease (CHD) is another common developmental disorder among individuals with DS, and there is a suspicion of an association between CHD and miRNAs expressed through chromosome 21. The aim of our study was to investigate the effect of miR-155, let-7c and miR-433 microRNAs on CHD in DS individuals with and without CHD.

Methods: Three different groups were included in the study: Group A (12 individuals - DS with congenital heart disease), Group B (15 individuals - DS without congenital heart disease) and Group C (15 individuals – control group). Peripheral venous blood was collected and plasma samples were manually isolated for RNA extraction. After conversion of RNA samples into cDNA, Real-Time PCR reactions were performed and Ct values were statistically analysed.

Results: The results showed that there was no statistically significant difference in miR-155, let-7c and miR-433 expression associated with congenital heart disease in individuals with DS (p: 0.392). The observation of insufficient expression of miR-433 in plasma indicated low levels of this miRNA in the peripheral blood.

Conclusion: Our study showed that miR-155, let-7c and miR-433 expression is not directly associated with congenital heart disease in individuals with DS. However, more comprehensive studies with larger sample sizes are required for the association of miRNA expression and congenital heart disease.

Conflict of Interest: None declared

EP07.015 Genetic testing in a representative Czech cohort of non-ischemic cardiac arrest survivors

Eva Kutílkova 1, Alice Krebsová1, Pavel Votýpka2, Petra Peldová2, Terezia Tavacova3, Veronika Zoubková2, Miloš Kubánek1, Petr Peichl1, Marketa Segetova1, Jana Haskova1, Markéta Adamová1, Jan Janousek4, Marek Sramko1, josef Kautzner1, Milan Macek2

1Institute of Clinical and Experimental Medicine, Department of Cardiology, Praha, Czech Republic; 22nd Faculty of Medicine, Charles University and Motol University Hospital, Department of Biology and Medical Genetics, Praha, Czech Republic; 32nd Faculty of Medicine, Charles University and Motol University Hospital, Children’s Cardiac Center, Praha, Czech Republic; 4Children’s Cardiac Center 2nd Medical Faculty of Charles University and University Hospital Motol, Praha, Czech Republic

Background: We aimed to determine the genetic aetiology of non-ischemic cardiac arrest in a representative Czech cohort of cardiac arrest survivors (CAS).

Methods: In total 218 CASs (135 males/83 females; the average age at CA 36 ± 16 years) referred from various cardiologic centres underwent genetic counselling and next-generation targeted/exom sequencing (SOPHiA GENETICS, Switzerland). Family cascade screening was performed in 503 relatives (2.3/case). Together with the genetic testing, additional non-invasive cardiology tests were performed including exercise ECG, ECG in higher leads and SaECG.

Results: The most frequent diagnosis in CAS was idiopathic ventricular fibrillation (IVF – 120 cases), before genetic examination. Other cases comprised inherited arrhythmias (LQT, CPVT, BrS) and cardiomyopathies (ARVC, DCM, HCM). Genetic testing identified the causative DNA variant in 58/218 (27 %) of CAS and led to the genetic stratification in 21/120 (18 %) of IVF patients mainly in the sense of arrhythmic syndrome (13/21, most frequent RYR2 – 6 cases). The most commonly mutated gene in CAS was PKP2 (11/218) followed by SCN5A, RYR2 and KCNH2 (7/218 each). Surprisingly, hypertrophic cardiomyopathy (HCM) is a minor part (7/218, 3%) of all clinical/genetic causes in our cohort.

Conclusion: Cardiogenetic examination reveals genetic cause of CA approx. 1/3 of all cases and leads to individualised therapy in CAS and assures identification of at-risk relatives. These results underscore the importance of complex care in individuals with life-threatening arrhythmias and cardiomyopathies, a system which is now well-implemented in our country.

Funding: LX22NPO5104, ZD-ZDOVA2-001, 00064203

Conflict of Interest: None declared

EP07.016 Mono and biallelic variants in TRIM63 are frequently associated with a unique form of hypertrophic cardiomyopathy

Noa Ruhrman Shahar 1;2, Lina Basel-Salmon1;2;3, Shay Ben-Shachar2;4, Dina Yagel5, sara hoss6, lily bazak1, Lena Sagi-Dain7, lilach Benyamini8, Sagi Josefsberg Ben-Yehoshua9;10, Rotem Greenberg4, Ofer Isakov2;4;5, elena friedman1, Michal Naftali5, daniel monakier6, moti haim11, amitai segev12, adel shalata13, nechama shalva14, alvit veber14

1Rabin Medical Center, genetic institute, Petah Tikva, Israel; 2Tel Aviv University, Faculty of Medicine, Tel Aviv-Yafo, Israel; 3Rabin Medical Center, Felsenstein Medical Research Center, Petah Tikva, Israel; 4Clalit Health Services, Clalit Research Institute, Ramat Gan, Israel; 5Clalit Health Services, Clalit sequencing center, Petah Tikva, Israel; 6Rabin Medical Center, Department of Cardiology, Petah Tikva, Israel; 7Carmel Medical Centre, Genetic Institute, Haifa, Israel; 8Shamir Medical Center (Assaf Harofeh), Genetic Institute, Be’er Ya’akov, Israel; 9Kaplan Medical Center, genetic institute, Rehovot, Israel; 10The Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem, Israel; 11Soroka Medical Center, department of cardiology, Be’er Sheva, Israel; 12Sheba medical center, The Leviev Heart Center, Ramat Gan, Israel; 13Bnai Zion Medical Center, Simon Winter Institute for Human Genetics, Haifa, Israel; 14Sheba, metabolic disease unit, Ramat Gan, Israel

Background/Objectives: Hypertrophic cardiomyopathy (HCM) is a prevalent hereditary heart disease, primarily affecting young adults with an incidence of 1:200 to 1:500. HCM may be a multifactorial or monogenic disorder and is often transmitted in an autosomal dominant manner. Variants affecting the TRIM63 gene have recently been identified as a rare cause for autosomal recessive HCM.

Methods: This study, conducted from June 2022 to October 2023, involved 107 patients diagnosed with HCM who underwent diagnostic gene-panel testing using a virtual, exome based panel for HCM, including 116 genes.

Results: Among the patients, three different biallelic pathogenic variants in TRIM63 were identified in 5 individuals (4.7%). Young age of onset, ventricular and supraventricular arrhythmias, and episodes of syncope/presyncope were common in all patients. Heterozygote variants in the gene with no additional pathogenic/likely pathogenic variants in other genes were found in additional six out of 80 with no clear monogenic diagnosis (8.6%). TRIM63 pathogenic variants were 8.2 times more prevalent among individuals with unexplained HCM than among individuals who underwent testing for other genetic panels (15/1584). Overall, TRIM63 pathogenic variants represent the third most common genetic cause for HCM (second only to MYBPC3 and MYH7 genes), and the most common autosomal recessive form of HCM in the Israeli population.

Conclusion: The study underscores the importance of bi and monoallelic variants TRIM63 in the genetic landscape of HCM and arrhythmias. We recommend including the TRIM63 gene in genetic testing panels for both conditions.

Grants: no grants

Conflict of Interest: None declared

EP07.017 HMGB1 mRNA expression in PBMCs of controls and patients six months after the first myocardial infarction, and correlations with left ventricular echocardiographic parameters

Jovana Kuveljic1, Ana Djordjevic 1, Milica Dekleva-Manojlovic2, Ana Kolakovic1, Ivan Zivotic1, Maja Zivkovic1, Tamara Djuric1

1“Vinca” Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia, Laboratory for Radiobiology and Molecular Genetics; 2Faculty of Medicine, University of Belgrade, Belgrade, Serbia

Background/Objectives: Myocardial infarction (MI) is followed by left ventricular (LV) remodeling, the process crutial in preserving heart structure and function. High mobility group box 1 (HMGB1) gene has been investigated in inflammatory reparative processes, whereas serum HMGB1 has been recognized as a candidate biomarker for acute MI. The aim of this preliminary study was to investigate the potential effect of HMGB1 mRNA expression on post-MI heart remodeling.

Methods: The study included 20 healthy controls and 60 patients with the first MI, prospectively followed-up for 6 months. The change (∆) in echocardiographic parameters, commonly used to assess LV remodeling, was calculated as the difference between the values at the 6-month follow-up and the baseline values. Relative HMGB1 mRNA expression in peripheral blood mononuclear cells (PBMCs) was detected using TaqMan® technology.

Results: The relative expression of HMGB1 mRNA was significantly higher in PBMCs of patients six months post-MI compared to controls (2-∆Ct HMGB1 mRNA: 0.037 ± 0.014 vs. 0.029 ± 0.009, p = 0.03). HMGB1 mRNA expression correlated positively with the ∆LV end-diastolic volume (R = 0.28, p = 0.03) and ∆LV end-sistolic volume (R = 0.27, p = 0.04), in the direction of an increase six months post-MI. There was no correlation of HMGB1 mRNA with LV ejection fraction or stroke volume.

Conclusion: Our results suggest that HMGB1 has detrimental impact on post-MI LV remodeling, correlating with changes in LV end-diastolic and end-sistolic volume. Further analysis on a larger sample size is required.

Grants: This study was funded by the Ministry of Science, Technological Development and Innovation of the Republic of Serbia (Agreement no. 451-03-47/2023-01/200017).

Conflict of Interest: None declared

EP07.018 The impact of MRAS on atherosclerosis-relevant phenotypes in smooth muscle cells

Pashmina Wiqar Shah 1, Tobias Reinberger1, Zouhair Aherrahrou1, Malte Spielmann2

1Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany; 2Institute for Human Genetics, University of Lübeck, Lübeck, Germany

A study by Erdmann et al. in 2009, revealed a region on 3q22.3, which encompasses the MRAS gene as a risk factor for CAD. MRAS encodes muscle Ras, a small GTPase that acts as a signal transducer in TNF and other related acute phase response signaling pathway. According to eQTL data, MRAS risk variants for CAD increase MRAS mRNA levels primarily in the arterial tissue and recent evidence depicted functional MRAS variants to be SMC-specific. The exact role of MRAS in atherogenesis and the therapeutic potential of targeting MRAS is still elusive. Therefore, we investigated the function of MRAS in vascular SMCs, one of the key cell types in the etiology of atherosclerosis and plaque stability. Human primary aortic SMCs transfected with MRAS-specific siRNA and murine aortic SMCs derived from our Mras/Apoe double knockout mouse model were subjected to functional assays including proliferation, migration and apoptosis. The absence of MRAS in murine SMCs leads to significant increase in proliferation, enhanced migration and reduced apoptotic activity compared to wild type SMCs when stimulated with TNF (n = 6, p < 0.001). Similarly, siRNA knockdown of MRAS in human SMCs increases migration and proliferation (n = 6, p < 0.01). Moreover, MRAS knockout leads to differential gene expression of contractile SMC marker genes such as Cnn1 and Tagln. Consistent with this, reduced MRAS levels in SMCs from 151 human donors also correlate with higher proliferation rates. In conclusion, MRAS deficiency modulates SMC biology in response to TNF. However, stimuli like IL6 and IL1ß will also be used to decipher its exact role in atherosclerosis.

Conflict of Interest: None declared

EP07.019 A novel epigenome discovery in Trisomy-21: parent origin of the non-disjunction influences the penetrance of congenital heart disease in Down syndrome

NAZIMAH NIK-MAHMOOD1;2, Thiloka Ratnaike 2;3, Timothy Hearn1

1University of Cambridge, Department of Medical Genetics, School of Clinical Medicine, Cambridge, United Kingdom; 2Colchester Hospital, ESNEFT, Children’s Department, Colchester, United Kingdom; 3University of Cambridge, Department of Paediatrics, Cambridge, United Kingdom

Background/Objectives: Congenital heart disease (CHD) is a major complication in people with Down Syndrome (DS). Although CHD prevalence is generally increased in individuals with DS, not all but 40%-50% are affected. We explore a novel hypothesis: the parent origin of duplication of chromosome-21 may influence the penetrance of the CHD phenotype in people with DS.

Methods: We obtained genomic data of 30 DS probands and both their parents through the Genomics England Research Environment (GEL RE) from the 100,000 genomes project and NHS Genomic Medicine Service patients. We performed a focussed genetic analysis within a region known as the Down syndrome critical region (DSCR) for CHD.

Parents’ diploid genotypes were compared with proband’s triploid genoytpe and matched with eight genotype ‘determinant’ scenarios that identify the origin of non-disjunction during meiosis.

Results: We saw Simpson’s paradox in our results. The majority of the total participants and the participants with CHD have duplication from their mother, (78%, of 30 and 60%, of 10 respectively). However, the likelihood of CHD is four times if duplication were from the father (OR = 4.7, p-value = 0.15). This result was not significant, possibly due to our small sample size

Conclusion: We propose parental origin of non-disjunction of chromosome-21 is an important epigenetic factor in disease prevalence for people with DS. Our findings suggest maternal origin of the duplication of chromosome 21 may have a lesser impact on phenotype and disease, compared to paternal origin- a ‘protective’ effect. Overall, increasing the life compatibility of people with DS.

Conflict of Interest: None declared

EP07.020 The identification of a large multi-exon deletion in RYR2

Claire Hopton 1;2, Glenda Beaman1;2, Rahat Perveen2, Kate Chandler1;2, William Newman1;2

1Manchester University NHS FT, Manchester Centre for Genomic Medicine, Manchester, United Kingdom; 2The University of Manchester, Division of Evolution, Infection and Genomics, Manchester, United Kingdom

Background/Objectives: Heterozygous missense variants of RYR2 are associated with catecholaminergic polymorphic ventricular tachycardia (CPVT). The majority of these variants result in a phenotype by a gain of function mechanism. However, variants which lead to a loss of function have been associated with a different arrhythmogenic phenotype, which has been termed the calcium release deficiency syndrome (CRDS). Recently, truncating variants of RYR2 have been identified in patients with arrhythmogenic phenotypes, however the mechanism of action of these remains unclear.

Methods: We identified a large intragenic deletion in RYR2 on array-CGH in a girl who was being investigated for autistic spectrum disorder. Following identification of this variant, we undertook cascade testing in the family and also cardiac screening in variant carriers. We used droplet digital PCR (ddPCR) and sanger sequencing to determine the breakpoints of this deletion.

Results: The deletion was found to be maternally inherited. Interestingly, the proband’s mother gave a history of palpitations and episodes of transient loss of consciousness. ddPCR and sequencing confirmed that the deletion spanned from intron 6 to intron 58 and included a small duplicated region.

Conclusion: Although exon 3 deletions of RYR2 are well described, large multi-exon deletions, as described in this case, are not frequently reported. This case adds to the variety and complexity of RYR2 variants which may lead to a cardiac phenotype. Further work needs to be undertaken to determine whether this variant results in a mutant RYR2 protein and whether there is an effect on calcium handling.

Grants: CH: NIHR ACL (2019-2023), WGN: Manchester NIHR BRC (IS-BRC-1215-20007).

Conflict of Interest: None declared

EP07.021 Relevance of Copy Number Variations for the genetic diagnosis of Long QT Syndrome.

Laura Cazón 1;1, Noël Brögger1, Luis De la Higuera Romero1, Marlene Perez1, Iria Gómez Díaz1, Rosalía Peteiro1, Maria Sanchez1, Emilia Maneiro1, Xusto Fernandez1, Diego Cabrera Argaña1, Almudena Amor1, Ivonne Cárdenas Reyes1, María Valverde1, Soledad García Hernández1, Martin Ortiz Genga1, Juan Pablo Ochoa1

1Health in Code, A Coruña, Spain

Background/Objectives: While the pathogenic role of Copy Number Variations (CNVs) in cardiovascular diseases has been previously described, their impact on entities such as Long QT syndrome (LQTS) remains unclear, with only a limited number of such variants reported thus far. This study aims to assess the prevalence of clinically relevant CNVs (likely pathogenic-LP or pathogenic-P) in a large cohort of index cases undergoing genetic testing for LQTS.

Methods: A retrospective study in a cohort of index cases with diagnosed or suspected LQTS referred to our laboratory for genetic testing with Next Generation Sequencing (NGS) was performed. The presence of LP/P CNVs was detected using a read depth approach and confirmed by orthogonal molecular techniques (Sanger or MLPA).

Results: A total of 1,183 index cases were examined. Approximately 27% (n = 322) had a positive result. CNV analysis was conducted on 1,071 (91%) of the cases, revealing 10 (0.9%) carriers of LP/P CNVs. Nine of these CNVs affected the KCNQ1 or KCNH2 genes (four deletions and one duplication in KCNQ1 and four deletions in KCNH2). The additional patient carried a deletion in RYR2, a gene associated with the development of catecholaminergic polymorphic ventricular tachycardia (CPVT), a phenotype that may be confused with LQTS.

Conclusion: In our cohort of patients referred for LQTS genetic testing, we identified clinically relevant CNVs in approximately 1% of cases. This represents a significant proportion of cases, underscoring the relevance of incorporating this analysis into routine genetic testing procedures.

Grants:

Conflict of Interest: Laura Cazón Full time, Noël Brögger part-time, Luis De la Higuera Romero full-time, Marlene Perez full-time, Iria Gómez Díaz full-time, Rosalía Peteiro full-time, Maria Sanchez full-time, Emilia Maneiro full-time, Xusto Fernandez part-time, Diego Cabrera Argaña full-time, Almudena Amor full-time, Ivonne Cárdenas Reyes full-time, María Valverde full-time, Soledad García Hernández full-time, Martin Ortiz Genga part-time, Juan Pablo Ochoa full-time

EP07.022 Construction of polygenic risk score prediction model for paroxysmal atrial fibrillation based on machine learning

Megumi Shiomi 1, Yuki Nagata1;2, Takeaki Sudo3, Takamasa Ichikawa2, Kensuke Ihara4, Ken Asada5, Yasuaki Tanaka6, Yasuteru Yamauchi7, Takeshi Sasaki8, Hitoshi Hachiya9, Yasushi Imai10, Hideo Fujita11, Tetsuo Sasano12, Tetsushi Furukawa4, Toshihiro Tanaka1;2

1Tokyo Medical and Dental University, Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo, Japan; 2Tokyo Medical and Dental University, Bioresource Research Center, Tokyo, Japan; 3Tokyo Medical and Dental University, Institute of Education, Tokyo, Japan; 4Tokyo Medical and Dental University, Department of Bio-informational Pharmacology, Medical Research Institute, Tokyo, Japan; 5RIKEN Center for Advanced Intelligence Project (AIP), Cancer Translational Research Team, Tokyo, Japan; 6Yokosuka Kyosai Hospital, Cardiovascular Center, Kanagawa, Japan; 7Yokohama City Minato Red Cross Hospital, Department of Cardiology, Kanagawa, Japan; 8National Disaster Medical Center, Department of Cardiology, Tokyo, Japan; 9Tsuchiura Kyodo Hospital, Cardiovascular Center, Ibaraki, Japan; 10Jichi Medical University, Division of Clinical Pharmacology, Department of Pharmacology, Tochigi, Japan; 11Jichi Medical University Saitama Medical Center, Division of Cardiovascular Medicine, Saitama, Japan; 12Tokyo Medical and Dental University, Department of Cardiovascular Medicine, Tokyo, Japan

Background/Objectives: Atrial fibrillation (AF) is a common heart rhythm disorder that often progresses from paroxysmal atrial fibrillation (PAF) to persistent AF. Diagnosing PAF is challenging due to its intermittent occurrence, and risk stratification methods are necessary. This study aimed to construct a polygenic risk score (PRS) -based model for predicting the risk of PAF.

Methods: A total of 1,798 subjects after undergoing quality control were divided into two groups: discovery and target sets. The target set had randomly 500 selected subjects, with the remaining 1,298 subjects as the discovery set. A genome-wide association study (GWAS) was conducted on the discovery set, and PRS was calculated using summary statistics of GWAS and the target set. The PRS-based model was constructed using the Light Gradient Boosting Machine framework, considering PRS and clinical factors. The model performance was evaluated using 5-fold cross-validation and area under the curve (AUC).

Results: The GWAS identified 62 SNPs exceeding the genome-wide significance threshold (p < 5 × 10-8). PRSs were calculated with varying p-value and r2 thresholds, selecting the model with an AUC greater than 0.7. Feature importance analysis ranked PRS, gender, and drinking history as the top three factors in the PRS-based model.

Conclusion: We constructed the PRS-based model that shows the highest AUC at the selected p-value and r2 thresholds. However, the AUC was lower than expected, and further studies are needed to construct a more accurate PRS-based model.

Grants: None

Conflict of Interest: Megumi Shiomi: None declared, Yuki Nagata: None declared, Takeaki Sudo: None declared, Takamasa Ichikawa: None declared, Kensuke Ihara: None declared, Ken Asada: None declared, Yasuaki Tanaka: None declared, Yasuteru Yamauchi: None declared, Takeshi Sasaki: None declared, Hitoshi Hachiya: None declared, Yasushi Imai Daiichi Sankyo Co., Ltd.,TOA EIYO Ltd., Hideo Fujita: None declared, Tetsuo Sasano Daiichi Sankyo Co., Ltd., Bayer Yakuhin Co., Ltd., Tetsushi Furukawa: None declared, Toshihiro Tanaka: None declared

EP07.023 PRKAG2 gene and Wolf-Parkinson-White-Syndrome: a diagnosis by chance? About a case report

Daniela González Fassrainer 1;1, Eva Wohlleber2, Martin Brauner2, Niels Vandendries2, Claudia Nevinny-Stickel-Hinzpeter1

1Synlab MVZ Humangenetik München, Munich, Germany; 2Synlab MVZ Humangenetik Freiburg, Freiburg, Germany

Background: We present a family (mother 59 y/o, son 26 y/o and daughter 29 y/o) who attended to genetic counselling for testing a familial pathogenic variant in SCN5A responsible for Brugada syndrome in the mother´s sister. The targeted analysis excluded the familial variant, therefore an analysis of the children was no longer indicated. Nevertheless, during anamnesis, it turned out that the daughter had a supraventricular tachycardia at the age of 16 (220/minute) and was diagnosed with Wolf-Parkinson-White (WPW) syndrome. In addition the son had two episodes of syncope at 7 and 15 years of age without a cause being found. WPW syndrome is a common arrhythmia affecting ~1-3/1000 individuals. Pathogenic variants in PRKAG2 have been described in some patients in association with cardiomyopathy and WPW. Because of the WPW we were driven to move forward with PRKAG2 analysis of the affected patient.

Methods: PCR amplification and direct sequencing of all coding exons and the relevant intron regions.

Results: PRKAG2-Gen: Variant c.997T>G, p.(Ser333Ala) heterozygous. Variant of unclear significance with a tendency towards a probable pathogenicity.

Conclusion: It is crucial to gather an extensive medical history and it is also important to establish if further genetic testing is required based on additional medical information. The exclusion of a known familial variant in a gene does not exempt a family from a further potential risk of a similar disorder.

Grants: --

Conflict of Interest: None declared

EP07.025 Homozygous p.Ser166Phe TNNI3 mutation in an adolescent with hypertrophic cardiomyopathy and cardiac arrest

Roman Praschberger 1, Johannes Zschocke1, Matthias Frick2, Sabine Rudnik1

1Medizinische Universität Innsbruck, Institute of Human Genetics, Innsbruck, Austria; 2Landeskrankenhaus Feldkirch, Internal Medicine I, Feldkirch, Austria

Background: Heterozygous mutations in TNNI3, encoding the critical cardiomyocyte regulator troponin I, are a rare cause (2-7%) of autosomal dominant hypertrophic cardiomyopathy (HCM). A large proportion of pathogenic TNNI3 mutations cluster in the functionally important exons 7-8, amongst them the c.497C>T, p.Ser166Phe mutation that is located in the troponin binding region. This mutation was reported as a founder mutation in the Netherlands resulting in a late onset HCM in the 4th decade of life (Van den Wijngaard 2011, PMID:21533915). It has an allele frequency of approx. 1:170.000 in Europeans (gnomAD)

Case Report: Here we report a 29-year-old woman who had a sudden cardiac arrest at age 16 and subsequently received a heart transplantation. Pathological examination of the explanted heart revealed hypertrophic cardiomyopathy with extensive fibrosis, connective and adipocyte tissue proliferation, necrosis and inflammatory infiltrates. She now underwent multigene panel analysis for HCM to determine the genetic risk to future children. Unexpectedly, a homozygous p.Ser166Phe mutation in TNNI3 was detected and confirmed heterozygous in her 74 year-old mother and 31 year-old sister. Her father was deceased at age 77 years of unrelated reasons. Family history did not reveal further cardiomyopathy cases.

Discussion: Biallelic TNNI3 loss of function mutations are rarely reported in neonatal and early onset dilated cardiomyopathy. The phenotype of our patient corresponds to a more classic juvenile onset HCM. Given that family history was negative, there should be a large number of non-penetrant maternal and paternal heterozygous mutation carriers.

Conclusion: Our pedigree provides evidence that p.Ser166Phe in TNNI3 might be a hypomorphic variant.

Conflict of Interest: None declared

EP07.026 TTN as the most prevalent cause of hereditary dilated cardiomyopathy

Marius Šukys 1, Eglė Ereminienė2;3, Karolina Mėlinytė-Ankudavičė3, Inga Nasvytienė4, Rimvydas Jonikas4, Kristina Aleknavičienė1, Darius Čereškevičius2, Sandra Žėkienė4, Zivile Zemeckiene1, Rasa Ugenskiene1

1Lithuanian University of Health Sciences, Department of Genetics and Molecular Medicine, Kaunas, Lithuania; 2Lithuanian University of Health Sciences, Institute of Cardiology, Kaunas, Lithuania; 3Lithuanian University of Health Sciences, Department of Cardiology, Kaunas, Lithuania; 4Hospital of Lithuanian University of Health Sciences Kauno Klinikos, Department of Genetics and Molecular Medicine, Kaunas, Lithuania

Background/Objectives: Dilated cardiomyopathy (DCM) is one of the most common genetic disorders with prevalence varying from 0.036% to 0.400%. Dilated cardiomyopathy is defined as increase of left ventricular end-diastolic diameter more than 117% and left ventricular ejection fraction (LVEF) lower than 45% and is not explained by conditions like coronary artery disease, hypertension, valvular pathology or congenital hear anomalies. Even though this disorder is known for many years, individual etiologic factor is rarely found. Many studies showed that causative genetic variant might be determined in 20-40% cases.

Methods: Retro/perspective analysis was performed for the adult patients consulted in 2019 – 2023 by clinical geneticist for suspected or confirmed DCM. Next generation sequencing (with Illumina NextSeq550) was conducted for genetic cardiac gene panel.

Results: 147 patient data was collected (average age 52.21, 69% males). 23 (16.3%) patients had causative genetic variant: 19 TTN, 2 BAG3, 1 FLNC, 1 LMNA. Patients with causative genetic variant tend to be younger (53.17 SD 11.92 vs. 47.04 SD 12.73, p < 0.05). The most prevalent TTN affected exon was 326th (10 out of 19 cases) and these patients had lower left ventricular ejection fraction (23.2 SD 9.67 vs. 38.2 SD 5.06, p < 0.05) despite the same average age at testing. We found 11 novel variants in TTN (NM_001267550): c.79568delT; c.46047_46048del; c.1389del; c.64554_64556delAGAinsTTTATGT; c.99672del; c.78246C>A; c.57165del; c.81679G>T; c.58859del; c.89743dup; c.82325dup.

Conclusion: Our cardiac gene panel diagnostic yield was 16.3%, what was lower than that mentioned in the literature. This indicates the need of stricter patient selection criteria for genetic testing.

Conflict of Interest: None declared

EP07.027 Contribution of four novel variants with inherited familial coronary artery disease

Mahsa Tahmasebivand 1, fatemeh ghodratpour1, Farzane Zare Ashrafi1, Marzieh Mohseni1, Sanaz Arzhangi1, Yasser Riazalhosseini2, Mark Lathrop2, Kaveh Hosseini3, Hossein Najmabadi1, Kimia Kahrizi1;2

1Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran; 2McGill Genome Centre, Montreal, Quebec, Canada; 3Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran

Background:Coronary artery disease (CAD) stands as a predominant cardiac complication and the leading global cause of mortality.Genetic predisposition plays a crucial role, with a more than twofold increase in CAD incidence observed in individuals with a family history of premature disease. This study, aimed to identify candidate rare genetic variants causing inherited CAD in individuals suffering from premature CAD.

Methods:Fifteen Iranian families, with two or more members with early-onset familial CAD/MI, were recruited for this ongoing cohort study at the Genetics Research Center, Tehran, Iran. The inclusion criteria required the presence of at least two affected family members with established premature CAD (<45 years in men and <55 years in women) in at least two generations. Whole-exome sequencing was conducted on one or two affected individuals, followed by stringent data filtering based on population frequency, in silico predictive algorithms, and phenotypic annotations. Mutations were validated through PCR and Sanger sequencing of the candidate gene region. Additional screening of both affected and unaffected family members is considered for the co-segregation study.

Results:The study identified novel variants potentially linked to CAD in four genes: LPA (c.3858C>A), PIKFYVE (c.5950C>T), PKP2 (c.1840-1G > A), and CAPN5 (c.893+1G > A), all of which co-segregated with familial premature CAD in four unrelated families. Notably, PKP2 (armadillo-related protein) and CAPN5 (calcium-dependent cysteine proteases) emerged as promising candidate CAD susceptibility genes.

Conclusion: The results suggest that PKP2 and CAPN5 variants may serve as putative causal factors in familial premature CAD.Further investigations, particularly functional studies, are warranted to elucidate the underlying mechanisms of pathogenicity.

GRANT: 801A/6/45329 to KK

Conflict of Interest: None declared

EP07.028 Uncommon pathogenic variant in Desmoplakin gene define a case of predominant left ventricular arrhythmogenic cardiomyopathy. Genetic testing usefulness.

Jesica Ramirez 1, Magalí Born2, Antonella Villagra1, Eliel Ramirez1, Nicolas Renna1

1Familial Cardiomyopathies Unit. Spanish Hospital from Mendoza. Argentina, Cardiology Department., Mendoza, Argentina; 2Central Hospital from Mendoza. Argentina, Clinical Department, Mendoza, Argentina

Background/Objectives: Arrhythmogenic cardiomyopathy (ACM) is a genetic heart muscle disease characterised by substitution of the ventricular myocardium by fibrofatty tissue which is a recognised arrhythmogenic myocardial substrate. In presence of arrhythmogenic left ventricular cardiomyopathy (ALVC) phenotypic features, demonstration of mutations in ACM related genes is mandatory for diagnosis. Our aim is to report the presence of an uncommon pathogenic variant in desmoplakin gene (DSP).

Methods: A 59-year-old female without familiar background presented with dysnea, sweating and palpitations. ECG showed ventricular tachycardia and she was admitted in coronary unit care. Echocardiogram revealed normal left ventricle (LV) sized with severely decreased systolic function (39%), grade 1 diastolic dysfunction and difuse hypokinesis, with normal right ventricle (RV). Cardiac catherization showed no coronary artery disease. Cardiac MRI demonstrated severely reduced global systolic function (34%), LV hypokinesis and akinetic segments with midwall presence of intramyocardial fat. Late gadolinium enhancement identified subepicardial fibrosis in 25% LV and lateral and septal fibrosis in RV. Considering Padua criterias for ALVC a blood sample was analyzed by multipanel sequencing (NGS) including genes for ALVC using ILLUMINA platform.

Results: Genetic testing reported one germline likely pathogenic, nonsense variant (c.2644G>T/p.Glu882Ter) in DSP gene.

Conclusion: DSP is one of the most common gene causing ALVC. However, this nonsense type variant has not been described to date in the population general (GnomAD) and has been reported in the ClinVar database as a pathogenic variant due to its two submitters. Cosegregation variant studies in offsprings was required. Implantable cardioverted defibrillator was neccesary due to high risk of sudden cardiac death.

Conflict of Interest: None declared

EP07.029 A RYR2 variant associated with catecholaminergic polymorphic ventricular tachycardia (CPVT) and neurodevelopmental disorder in two siblings.

Luisa Marsili 1;2, Melanie Rama3, Thomas Smol4, Guy Vaksmann5, Peter van Tintelen2, Jamal Ghoumid1

1Univ. Lille, ULR7364 RADEME, CHU Lille, Service de Génétique Clinique, Lille, France; 2University Medical Center Utrecht, Utrecht University, Department of Genetics, Utrecht, Netherlands; 3Institut de génétique Humaine, CHU LILLE, Lille, France; 4Univ. Lille, ULR7364 RADEME, Institut de génétique Humaine, CHU LILLE, Lille, France; 5Hôpital privé de La Louvière, Unité de cardiologie pédiatrique, Lille, France

Background/Objectives: Disease-causing variants in RYR2 cause catecholaminergic polymorphic ventricular tachycardia (CPVT), a rare inherited cardiac disease characterized by stress- and exercise-induced life-threatening ventricular arrythmias generally in the absence of cardiac structural abnormalities. Previous studies have suggested the association of RYR2-CPVT and neurodevelopmental disorders (NDDs). Here, we report two sisters harboring a RYR2 variant and presenting with CPVT associated with NDD.

Methods: Two sisters aged 25 and 23 years were referred to clinical genetics. They had undergone brain MRI, metabolic testing, and cardiac evaluation (echocardiography, electrocardiogram, and Holter monitoring). Genetic testing included karyotyping, FMR1 gene analysis, CGH-array, and whole-exome sequencing in the family quartet.

Results: The two sisters, the only children of healthy nonconsanguineous parents, presented with a non-progressive NDD marked by delayed developmental milestones, moderate ID, cerebellar ataxia, and anxiety disorder. Brain MRI and routine metabolic screening were normal. They also presented with a cardiac phenotype (age at onset one and 8 years), characterized by supraventricular tachycardia and non-sustained ventricular tachycardia despite receiving betablockers treatment, syncopal episodes occurring during acute emotion, and a structurally normal heart. Karyotype, FMR1 gene analysis, and CGH-array were normal. Exome sequencing identified the pathogenic RYR2 NM_001035.3: c.14864G>A p.(Gly4955Glu) variant in both sisters. No other (likely) pathogenic variant was identified. The RYR2 variant was not found in parental blood samples, but the identification of bidirectional ventricular tachycardia and supraventricular tachycardia at Holter monitoring in the mother could suggest maternal mosaicism.

Conclusion: We add further evidence on the association between RYR2-CPVT and NDDs.

Conflict of Interest: Luisa Marsili CHU Lille, Speaker at symposium, Sanofi, Melanie Rama: None declared, Thomas Smol: None declared, Guy Vaksmann: None declared, Peter van Tintelen Hartstichting; CardioVasculair

Onderzoek Nederland (CVON) projects: PREDICT2 2018-30,

DOSIS 2014-40, DOUBLE-DOSE 2020B005, Jamal Ghoumid: None declared

EP07.030 Whole exome sequencing findings in young people with sudden cardiac death

Elena Zaklyazminskaya 1, Mariam Sadekova1, Mikhaleva Ljudmila2, Sergey Dzemeshkevich3, Balashova Maria4;5

1Petrovsky National Research Centre of Surgery, Medical Genetics Laboratory, Moscow, Russian Federation; 2Petrovsky National Research Centre of Surgery, AVTSYN RESEARCH INSTITUTE OF HUMAN MORPHOLOGY, Moscow, Russian Federation; 3Petrovsky National Research Centre of Surgery, CardioSurgery, Moscow, Russian Federation; 4Sechenov 1st State Medical University, Moscow, Medical Genetics, Moscow, Russian Federation; 5Center of Genetics and Reproductive Medicine “Genetico”, Clinical Genetics, Moscow, Russian Federation

Background/Objectives: Sudden cardiac death (SCD) occurs in 1-35 years old with a frequency of 1.3-2.6 per 100,000. Our research aims to evaluate the effectiveness of whole exome sequencing (WES) in young SCD victims to reveal underlying heart disease.

Methods: Thirteen individuals affected by SCD, aged 1-22, underwent pathomorphological examination and WES of DNA from myocardial paraffin blocks and peripheral blood leukocytes. The data were analyzed using a domestic bioinformatics pipeline.

Results: There was no established diagnosis of primary heart disease before SCD in all patients. In 1 case, the cause of death was rupture of the ascending aortic aneurysm, in 12 cases the arrhythmic origin of the SCD was assumed. Autopsy shows that 2 patients had DCM, 5 had HCM, and 4 of the deceased had an apparently normal heart.

Genetic variants of the IV-V pathogenicity class were identified in 8 deceased (c.3697C>T in MYBPC3 (2 cases), c.4467G>C in SCN5A, c.688G>A in TPM1, c.1413_1414insT in DSC2, c.1403C>T in PTPN11, c.1588C>T in MIB1, c.3496C>T in COL3A1). In 2 cases, VUS (class III) c.2066G>A in SCN5A and c.641G>A in SCN1B genes were identified. There were no candidate genetic findings in the 3 individuals with SCD.

Conclusion: WES enabled to reveal the inherited disease in the majority of the young SCD cases (61.5%), and to perform family screening. Genetic approach in a powerful method improving post mortem evaluation of the SCD victims.

Grant References: The research was funded by the Russian Science Foundation (project №22-75-00134, https://rscf.ru/project/22-75-00134/) and FURG-2023-0009.

Conflict of Interest: Elena Zaklyazminskaya Research Project of Russian Academy of Science FURG-2023-0009, Mariam Sadekova: None declared, Mikhaleva Ljudmila: None declared, Sergey Dzemeshkevich: None declared, Balashova Maria PI, Russian Science Foundation grant No. 22-75-00134, https://rscf.ru/project/22-75-00134/

EP07.031 The homozygous missense p.(Ile172Thr) mutation in the TPM1 gene causes severe hypertrophic cardiomyopathy in a Consanguineous Family.

Mohammed Alghamdi 1, Patrice Bourgeois1, Emilie Consolino2, Pierre Jacques Ambrosi3, Karine NGUYEN PHONG2, Gilbert Habib3, Annachiara De Sandre-Giovannoli1

1Marseille, Molecular Genetics Laboratory, Biogénopôle M2GM, Marseille, France; 2Marseille, Medical Genetics unit, Marseille, France; 3Marseille, Cardiology and heart failure department, Marseille, France

Background/Objectives: Hypertrophic cardiomyopathy (HCM) is a common genetic cardiac disorder characterized by idiopathic left ventricular thickening, leading to heart failure and increased risk of arrhythmias and sudden cardiac death. It is mainly caused by mutations in genes encoding sarcomeric proteins. Alpha-tropomyosin, a thin filament protein encoded by the TPM1 gene, plays a crucial role in regulating cardiac muscle contraction.

We report a rare homozygous variant in the TPM1 gene associated with severe familial HCM.

Methods: Search for germline variants was performed by targeted diagnostic next-generation sequencing (NGS) in a 65 genes cardiomyopathy panel and confirmed by Sanger sequencing upon ACMG classification 4 or 5 (Richards et al. 2015, PMID 25741868).

Results: A homozygous missense pathogenic variant (class 5 ACMG) was identified in the TMP1 gene (NM_001018005.2: c.515T>C: p.(Ile172Thr)) in two siblings presenting with severe HCM and a family history of sudden cardiac death; one had cardiac transplantation at 42 years, the other presented with biventricular hypertrophy, atrial fibrillation and severely altered left ventricular relaxation.

Conclusion: The identification of this rare homozygous pathogenic variant in the TPM1 gene among members of a consanguineous family with severe HCM phenotypes allows for a discussion the genetic complexity underlying HCM as well as the role of TPM1 and of the p.Ile172Thr variant in cardiac manifestations. Our findings contribute to a broader understanding of HCM’s genetic bases and better defining genotype/phenotype relationships involving the TPM1 gene, for which homozygous missense variant carriers were rarely reported.

Grants: Assistance Publique Hôpitaux de Marseille, Marseille, France.

Conflict of Interest: None declared

EP07.032 Clinical utility of a multi-polygenic risk score in predicting cardiorenal complications in patients with type 2 diabetes

Johanne Tremblay 1, Redha Attaoua2, Mounsif Haloui2, Marie-Renée Guertin2, Edoh Kodji2, Pierre Dumas2, Janusz Kaczorowski1, Pavel Hamet1

1University of Montreal, CHUM Research Center, Montreal, Canada; 2CHUM, CHUM Research Center, Montreal, Canada

Background: We developed a multi-polygenic risk score (multiPRS) to predict the risk of micro- and macrovascular complications in T2D patients (Tremblay et al. Diabetologia 2021). Its clinical utility for individual risk prediction and population risk stratification of cardiorenal complications is being tested in a large (n = 2700 participants) pragmatic trial (GENOCARDIA).

Methods: We built a reference cohort of 20,000 T2D patients (64% men), aged 30 years and older (mean age, 62). The cohort was stratified into low, medium, and high-risk groups corresponding to <30th, 30-70th and >70th PRS percentiles. The percentile rank of the first 526 patients recruited in GENOCARDIA (53% men, mean age 64), with or without cardiorenal complications, was obtained by comparing their PRS values to the distribution of the reference cohort. A 5-year absolute risk estimate was derived from the incident rate of cardiorenal events per risk stratum.

Results: 5-year absolute risks were 1, 3 and 8% for low, medium, and high-risk strata in women <60 years and 5, 7 and 18% in older women. They were 5, 7 and 11% in younger and 8,12 and 20% in older men. Relative risks (high vs rest) were 3,7 and 2,8 in younger and older women, respectively, and 1.9 in men. The 30% high-risk threshold led to an 80% detection rate of previous cardiorenal complications among the 526 participants.

Conclusion: A multi-PRS helps identify T2D patients at high risk of cardiorenal complications who could benefit from an early intervention. Grants from Genome Quebec, FSISSS/MEIE and Genome Canada.

Grants: Genome Quebec, FSISSS/MEIE and Genome Canada.

Conflict of Interest: Johanne Tremblay OPTITHERA, Genome Quebec, Genome Canada, FSISSS/MEIE, Redha Attaoua: None declared, Mounsif Haloui: None declared, Marie-Renée Guertin: None declared, Edoh Kodji: None declared, Pierre Dumas: None declared, Janusz Kaczorowski FSISSS/MEIE, Pavel Hamet OPTITHERA, Genome Canada, Genome Quebec and FSISSS/MEIE

EP07.033 Genetic variants reveal cardiovascular disease genetic overlap between five common cardiovascular diseases: implications for biology-based partitioning polygenic risk scores.

Tigist Demssew Adane 1;1, Joyce Van Meurs1, André Uitterlinden1, Jeroen van Rooij1

1Erasmus MC, Rotterdam, Netherlands

Background/Objectives: Cardiovascular disease (CVD), the leading cause of death in Europe, is an umbrella term for several complex disorders involving gene-environment interactions with coronary artery disease (CAD), stroke, heart failure (HF), atrial fibrillation (AF), and peripheral arterial disease (PAD) as the most prevalent. They share common clinical risk factors, and overlapping etiologies have been recognized. Genome-wide association studies (GWAS) have identified hundreds of genetic risk factors for each complex disorder, representing underlying biological mechanisms. Our study investigates the genetic overlap and interconnection between these five common CVDs.

Methods: This study collected 468 genome-wide significant variants identified in recent GWAS of CVDs. We visualized the genetic network using Cytoscape with a linkage disequilibrium cutoff of > 0.1 and analyzed overlap and pathway enrichment using Metascape.

Results: More than half of the genetic variants associated with stroke, HF, and PAD were implicated in more than one CVD. In contrast, AF and CAD displayed distinct genetic patterns. About 29% of variants linked to stroke and PAD were also implicated with CAD. Additionally, 26% of HF variants and 12.9% of stroke variants showed associations with AF, which is a major risk factor for both conditions. Heart development, DNA regulation, PI3K-Akt signaling, collagen degradation, interleukin signaling, and muscle structure development are key shared pathways and GO enrichments.

Conclusion: This study found a shared genetic risk variant and an overlap between the common cardiovascular diseases. The findings suggest genetic risk estimations for CVD can be refined and clinically informed based on shared and distinctive biological pathways.

Conflict of Interest: None declared

EP07.034 А case-control study of association of FV HR2 6775A > G polymorphisms with thrombosis in Montenegrin patients; Haplotype analysis between FV 1691G > A and FV HR2 6775A > G polymorphisms

Sladjana Teofilov 1, Olivera Miljanovic1, Tatjana Ostojic1, Milena Bulatovic1, sasa perovic1, natasa djordjevic2

1Clinical Center of Montenegro, Podgorica, Montenegro., Center for Medical Genetic and Immunology, Podgorica, Montenegro; 2Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia, Department of Pharmacology and Toxicology, kragujevac, Serbia

Background Thrombosis is a pathological coagulation in blood vessels, where genetic risk factors play a significant role in its etiopathogenesis. The main objective of this study was to determine whether polymorphisms of FVHR26755A > G are associated with thrombosis. In addition, the association of thrombosis risk with polymorphisms in FV1691G > A was investigated. Given the presumed functional interconnection, alleles within the same gene may display linkage disequilibrium (LD). As a result, a thorough haplotype analysis was conducted for the investigated polymorphisms.

Methods The study comprised 103 patients, each confirmed to have at least one thrombosis based on valid diagnostic algorithms, along with 106 healthy subjects. Genotyping was preformed using the allele-specific PCR method.

Results No statistically significant difference was observed in the distribution of FVHR26755 polymorphisms between VTE patients and controls.The association and increased risk of thrombosis were confirmed for the variant genotypes of FV1691.Between the FV 1691 G > A and FVHR26775A > G polymorphisms, there is a moderate LD and the occurrence of all 4 haplotypes. Haplotypes 1691G > A-6775A > G associated with increased risk of thrombosis were A-A and A-G (χ2(3) = 24.03; p < 0.001). High-risk diplotypes were G-A/A-A and G-A/A-G (χ2 (3) = 27.57; p < 0.001), while haplotype A-G and diplotype G-A/A-G were present only in VTE patients.

Conclusion The exclusive presence of haplotype A-G and diplotype G-A/A-G in thrombotic patients suggests a potential association of the variant allele FVHR26775G with an increased risk of thrombosis.

Conflict of Interest: None declared

EP07.035 No difference was found in the spectrum of the rare causative variants in leucocytes and myocardium in patients operated for hypertrophic cardiomyopathy.

Mariam Sadekova 1, Balashova Maria2;3, Anna Motreva4, Sergey Dzemeshkevich5, Elena Zaklyazminskaya2;5

1Abrikosovskiy Pereulok, 2, laboratory of medical genetics, Moskva, Russian Federation; 2Genetico, Moskva, Russian Federation; 3Sechenovskiy Universitet, Moskva, Russian Federation; 4Federal Center for Cardiovascular Surgery, Astrakhan, Russian Federation; 5Abrikosovskiy Pereulok, 2, Moskva, Russian Federation

Background/Objectives: To elucidate the spectrum of causative genetic variants in DNA extracted from the leucocytes and myocardium in patients operated for hypertrophic cardiomyopathy (HCM).

Methods: The diagnosis of HCM and indications for myectomy were established by expert cardiosurgical centers for 69 (40 males) patients. The average age of patients was 37 years. DNA for whole exome sequencing (WES) were extracted from the leukocytes (69 patients) and myocardium samples (16/69 patients younger 20 y.o.) taken intra-operatively. WES was performed on the Illumina NovaSeq_6000 platform with the SureSelect_allExonsV7 kit. Pathogenicity was assessed in accordance with current recommendations.

Results: Familial inheritance of HCM was confirmed for 26 (38%) probands, sporadic in 27 (39%), for other probands (23%) familial data was incomplete

Unequivocal mutations were identified in 27 patients (39%). Majority of mutations were found in the MYBPC3 and MYH7 genes (8/8 equally), in 3 cases mutations were in the MYL3 gene. Noonan syndrome with mutations in PTPN11 and RIT1 genes was identified in 5 patients. VUS were detected in 26 cases (38%). De novo origin of the variants was confirmed in 4 patients.

Spectrum of causative variants in DNA from leukocytes and myocardium in 16 young patients was equal. Somatic mosaicism was not detected in this group.

Conclusion: Somatic myocardial mosaicism has a little if any effect as a factor modulating HCM severity, but further study is needed to confirm this conclusion.

Grant References: Routing investigations were supported by research project FURG-2024-0004, myocardial sequencing was performed with Russian Science Foundation project 22-75-00134, https://rscf.ru/project/22-75-00134/.

Conflict of Interest: Mariam Sadekova project FURG-2023-0009 collaborator, Balashova Maria PI Russian Science Foundation (project №22-75-00134, https://rscf.ru/project/22-75-00134/, Anna Motreva: None declared, Sergey Dzemeshkevich PI project FURG-2024-0004, Elena Zaklyazminskaya Collaborator project FURG-2024-0004

EP07.036 Loeys-Dietz syndrome associated with truncating variants in PMEPA1 gene: First description of the phenotypic spectrum in three Belgian families.

Claire Fouquet 1, Nicole Revencu2, Elisa Docampo1, Vincent Bours1, Nele Boeckx3, Bart Loeys3, Julie Harvengt1

1Centre Hospitalier Universitaire de Liège, Centre for Medical Genetics, Liège, Belgium; 2Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Centre for Medical Genetics, Brussels, Belgium; 3Antwerp University Hospital, University of Antwerp, Centre for Medical Genetics, Antwerp, Belgium

Background/Objectives: PMEPA1 has recently been identified as a predisposition gene for familial thoracic aortic aneurysm disease related to Loeys-Dietz syndrome (LDS). The PMEPA1 product is involved as an inhibitor regulator in the transforming growth factor beta pathway.

Methods: We report the genotype and phenotype of seven patients aged from 14 to 58 years from three different Belgian families.

Results: All three familial pathogenic variants are frameshifts, localised in the last exon of PMEPA1 and predicted to lead to a truncated protein.

Cardiovascular anomalies were present in six patients and included two aortic dissections, enlarged abdominal aorta, dilatation of the pulmonary artery and mitral valve prolapse. Congenital heart malformations, such as a bicuspid aortic valve or an atrial septal defect, were found in three patients. Thoracic scoliosis was described in three patients and in addition pes planus and pectus excavatum were found in two patients. The two youngest patients presented an umbilical hernia, of which the male also presented bilateral cryptorchidism. Myopia was reported in two patients. Craniofacial dysmorphisms included high palate and downslanting palpebral fissures as the most frequent features (n = 3), only one patient presented a hypertelorism and none had a bifid uvula. Other features included allergy and an autoimmune hypothyroidism.

Conclusion: Aortic and cardiovascular abnormalities are the most common features in our Belgian families with the PMEPA1 variants. Other non-vascular features are inconsistently associated. We suggest a highly individual and familial variable expressivity for this new LDS associated with PMEPA1. Further studies are required to investigate this genotype-phenotype correlation.

Conflict of Interest: None declared

EP07.037 TAB2-related syndromic cardiovascular heart disease with facial dysmorphism, skeletal and connective tissue defects in a three generation family

Ece Çepni 1, Serpil Eraslan2, Rukiye Eker Ömeroğlu3, Hülya Kayserili2;4

1Koc University, Institute of Health Sciences, Istanbul, Türkyie; 2Koç University Hospital, Genetic Diseases Evaluation Center, Istanbul, Türkyie; 3Istanbul Medical Faculty, Istanbul University, Department of Pediatric Cardiology, Istanbul, Türkyie; 4Koç University School of Medicine (KUSoM), Medical Genetics Department, Istanbul, Türkyie

Background/Objectives:Congenital heart defects (CHDs) are the leading cause of birth defect-associated infant mortality and morbidity worldwide. Autosomal dominant variants in TAB2 have been linked with non-syndromic congenital heart defects-2 (CHTD2,MIM#614980). Although CHTD2 is characterized by various structural and functional cardiac anomalies; extra-cardiac features including developmental delay, facial dysmorphism,skeletal and/or connective tissue findings have also been reported in an emerging number of studies1-9.

Case report:8 years 4 months old girl, secondborn to non-consanguineous couple, was referred to genetic outpatient clinics due to short stature and congenital heart anomalies (atrial septal defect, aortic regurgitation with right heart dominance).She had distinct facial dysmorphism comprising triangular face, monocular strabismus, low-set, protruding ears. Musculoskeletal findings, generalized joint hypermobility, cutaneous laxity with soft-velvety skin and pes planus, were considered suggestive for connective tissue disease. Pedigree analysis showed that her sister and father also had similar musculoskeletal findings along with mild facial dysmorphism.Her sister had severe mitral prolapsus necessitating surgery while her father and paternal grandmother were reported to have mitral insufficiency.

Methods-Results:Whole-exome sequencing (WES) revealed novel, likely pathogenic (class 2) TAB2 variant that creates a frameshift at codon 456 which is expected to undergo nonsense-mediated decay. Segregation analysis by Sanger sequencing confirmed the presence of the variant in the sister and father.

Conclusion:We herein report on a three-generation family with TAB2-related syndrome and add on the emerging syndromic phenotype of CHTD2 & provide further insight to the genotype-phenotype correlation.

References:1Thienpont et al., 2010;PMID:20493459;2Ackerman et al., 2016;PMID:27452334; 3Ritelli et al., 2018;PMID:28386937. 4Chen et al., 2020;PMID:31959127.5Morlino et al., 2019;PMID:31250519. 6Weiss et al., 2015;PMID:26139517. 7Westphal et al., 2022;PMID:34995729. 8Hanson et al., 2022;PMID:34741306. 9Woods et al.,2022;PMID:35971781.

Grants: None.

Conflict of Interest: None.

Conflict of Interest: None declared

EP07.038 Personalized management of complications development in heart failure patients

Madina Zhalbinova 1, Saule Rakhimova1, Ulan Kozhamkulov1, Kenes Akilzhanov2, Ayaulym Chamoieva1, Nazerke Satvaldina1, Zhanel Mirmanova1, Gulbanu Akilzhanova2, Makhabbat Bekbossynova3, Ainur Akilzhanova1

1National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan; 2Semey Medical University, Pavlodar Branch, Pavlodar, Pavlodar, Kazakhstan; 3National Research Cardiac Surgery Center, Astana, Kazakhstan

Background/Objectives: Heart transplantation is gold standard treatment for heart failure (HF) patients. Implantation of mechanical circulatory support (MCS) device could be performed instead of transplantation due to limited number of donors. However, implanted MCS causes complications development due to incorrect dosage of anticoagulants and platelet receptors dysfunction which is caused by device. The purpose of research was to identify influence of genetic factors on complications development.

Methods: Venous blood samples of HF samples (n = 98) were recruited. MCS were implanted: HMII, HM3, HVAD. Patients were prescribed with warfarin for prevention thrombosis complications. Two groups of HF were formed: 1. Without complications (n = 74); 2. With complications (n = 24). DNA samples were extracted and genotyped for gene polymorphisms encoding coagulation factors and platelet receptors.

Results: The distributions of allelic and genotype frequencies of rs5918 in ITGB3 gene were significantly different between patient groups (p < 0.05). TC genotype of SNP rs5918 in ITGB3 gene was significantly associated with patients’ complications according to regression analysis [(OR (95% CI): 5.37 (1.79–16.16), p = 0.0056)]. Moreover, distribution analysis showed that TC genotype was significantly higher in patients with thrombosis complications than without thrombosis (84.6 vs. 36.4%, p = 0.043). TT genotype was more prevalent in patients with HM3 device with absence of thrombosis events.

Conclusion: Our research found that rs5918 in ITGB3 gene could help to prevent/reduce complication development in HF patients with implanted MCS.

Grants: Supported by Committee of Science of the Ministry of Science and Higher Education of Republic of Kazakhstan (Grant No. AP14869903), (Grant No. BR21881970) and 211123CRP1608, A.A.

Conflict of Interest: None declared

EP07.039 Genomic and epigenetic insights into RASopathies: modifications in cardiovascular development

Botezatu Alina 1, Vlad Morhan2, Cecilia Lazea3, Florina-Victoria Nazarie4, Adina Chis4, Romana Vulturar4

1“Iuliu Hatieganu” University of Medicine and Pharmacy, Faculty of Medicine, Cluj-Napoca, Romania; 2“Ion Chiricuta” Institute of Oncology, Department of Radiation Oncology, Cluj-Napoca, Romania; 3“Iuliu Hatieganu” University of Medicine and Pharmacy, 1st Department of Pediatrics, Cluj-Napoca, Romania; 4“Iuliu Hatieganu” University of Medicine and Pharmacy, Department of Cell and Molecular Biology, Cluj-Napoca, Romania

Background/Objectives: RASopathies-patients, due to germline mutation of RAS-MAPK genes, display a broad spectrum of congenital heart disease (CHD). The importance of understanding the genetic and epigenetic mechanisms underlying the pathogenesis and clinical manifestations of CHD in these patients is obvious, and we are conducting a systematic review of these processes.

Methods: The review embraces an overview of key genomic elements implicated in cardiovascular function: susceptibility loci, genetic variants, and regulatory elements. We used PubMed, MEDLINE, SciELO, master’s and doctoral dissertations, using the keywords: “RASopathies”, “epigenetics”, “cardiac development”. By synthesizing all available information, we summarize how recent studies have advanced our understanding of cardiovascular genomics and epigenomics; we are highlighting less-explored epigenetic targets relevant to CHD in different RASopathies.

Results: Aberrant DNA methylation patterns at specific CpG sites within the regulatory regions of Ras/MAPK pathway genes may silence or activate these genes, contributing to the dysregulation observed in RASopathies. This review resumes also the current understanding of senescence in RASopathies and its impact on lifespan reduction. Notable, ex vivo cultured cardiac progenitor cells (CPCs) manifest senescence-like traits, intricately linked to the activation of ERK1/2-MAPK signalling. Furthermore, hypermethylation of several genes (as tumour suppressor genes, i.e. NF1) could lead to reduced expression, exacerbating the effects of mutations in these genes.

Conclusion: Rasopathies serve as a compelling model demonstrating the bidirectional influence between the genome and epigenome on susceptibility to epigenetic modifications, offering clinicians valuable insights to steer future clinical and laboratory research, targeted interventions, and personalized approaches for advancing cardiovascular health.

Conflict of Interest: None declared

EP07.040 More is not always better: the impact of gene selection in cardiomyopathies and long-QT syndrome

Clara Herrero-Forte1, Raluca Oancea-Ionescu 1, Carmen Cotarelo-Pérez1, Victoria Cañadas-Godoy2, M. Alejandra Restrepo-Córdoba3, María Fenollar-Cortés1

1Unidad de Genética Clínica. Servicio de Análisis Clínicos. Instituto de Medicina del Laboratorio.IdISSC. Hospital Clinico San Carlos, Madrid, Spain; 2Consulta de Cardiopatías Familiares/Unidad de Arritmias. Servicio de Cardiología. Instituto Cardiovascular. Hospital Clínico San Carlos; 3Unidad de Insuficiencia Cardiaca y Cardiopatías Familiares. Servicio de Cardiología. Instituto Cardiovascular. Hospital Clínico San Carlos, Madrid

Background/Objectives: Inherited Cardiac Disease represent a diagnostic challenge due to significant clinical and genetic heterogeneity. Next Generation Sequencing (NGS) improves the diagnosis, prognosis, and treatment of patients.

The objective is to analyze the diagnostic yield in our cohort, evaluating the impact of increasing number of genes and variant reevaluation.

Methods:

  • 300 patients (260 from 2016-2022; 40 from 2023) underwent NGS analysis with 37 genes for hypertrophic cardiomyopathy (HCM), 67 for dilated/arrhythmogenic cardiomyopathy (DCM), and 17 for long-QT syndrome (LQTS).

  • Increasing number of genes according to the 2022 Expert Consensus Statement of genetic testing for cardiac diseases, and reevaluation of previous variants detected in the 260 patients 2016-2022.

  • ACMG/AMP Standards and Guidelines were used to variant classification.

Results:

DCM

HCM

LQTS

Total

Total cases

167

116

17

300

P/LP cases

18 (11%)

32 (28%)

7 (4%)

57 (19%)

Cases with VUS

20 (12%)

7 (6%)

1 (6%)

28 (9%)

LP reclassified to VUS

4

1

5

VUS reclassified to LP

1

2

1

4

P/LP cases due to increased number of genes

VUS cases due to increased number of genes

2

2

  1. P/LP: Pathogenic/Likely Pathogenic; VUS: Variant of Uncertain Significance

Conclusion: Pathogenic variants are predominantly in a limited number of genes.

Increasing number of genes analyzed may not significantly improve diagnostic yield and could increase uncertainty in interpreting results, without improving clinical management.

Periodic variant reevaluations are crucial due to dynamic variant interpretation and classification, depending on current scientific knowledge.

Grants:

Conflict of Interest: None declared

EP07.042 Analysis of arrhythmia genes in severe and critical COVID-19 Kazakhstani patients

Nazerke Satvaldina 1, Saule Rakhimova1, Diana Samatkyzy1, Asset Daniyarov2, Ulykbek Kairov2, Ainur Akilzhanova1

1PI “National Laboratory Astana” Center for Life Science, Laboratory of Genomic and Personalized Medicine, Astana; 2PI “National Laboratory Astana” Center for Life Science, Laboratory of Bioinformatics and System Biology, Astana

Background/Objectives: Genetic predisposition is one of the risk factors in susceptibility to severe COVID-19 infection. The aim of this study was to analyse main arrhythmia genes among severe and critical COVID-19 infected group in comparison with healthy controls.

Methods: Genomic DNA was extracted from peripheral blood samples of patients. Whole-genome libraries were prepared using Illumina DNA Prep kit with IDT DNA/RNA UD Indexes. Whole-genome sequencing was done using S4 flow cell and 300 paired-end cycles on NovaSeq 6000 high-throughput platform.

Results: 52 patients with confirmed COVID-19 infection with severe or critical levels and 50 healthy controls was included to the study. Average age of case group was 59.8 ± 14, among them 40.4% males and 59.6% females. Case group has different CVD comorbidities such as arterial hypertension (57.7%), chronic heart failure (19.2%) and cardiac infarction (7.7%). Analysis of association included CASQ2, SCN5A, DSP, DSG2 and KCNE1 genes. Logistic regression analysis showed no difference between selected arrhythmia-related SNPs and disease risk (CASQ2 OR-0.87, p = 0.61; SCN5A OR-1.43, p = 0.26; DSP OR-0.70, p = 0.27; DSG2 OR-0.68, p = 0.18; KCNE1 OR-0.85, p = 0.56).

Conclusion: Our research showed that there is no statistically significant difference in arrhythmia SNPs between severely infected COVID-19 patients and healthy controls.

Grants: The research has been funded by the Science Committee of the Ministry of Science and Higher Education of the Republic of Kazakhstan (Grant No. AP19677442).

Conflict of Interest: None declared

EP07.043 Compliance and diagnostic yield of a Cardiogenetic clinic in Israeli patients

Sivan Koka 1;2, Moshe Rav Acha2;3;4, Tal Hasin2;3;4, Yoav Michowitz2;3;4, Bayya Feras2;3;4, Shemy Carasso2;3;4, Michael Glikson2;3;4, Ephrat Levy-Lahad1;2;4

1Medical Genetics Institute, Shaare Zedek Medical Center, Jerusalem, Israel; 2The Eisenberg R&D Authority, Shaare Zedek Medical Center, Jerusalem, Israel; 3Jesselson Integrated Heart Center, Shaare Zedek Medical Center, Jerusalem, Israel; 4Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel

Background/Objectives: With the advancement of sequencing technology and increase involvement of genomics approaches and the role of genetic variants in prognosis and clinical decisions, specialized clinics have become a necessity for good clinical practice. Commonly observed cardiac conditions in the Cardiogenetic clinic include cardiomyopathies, arrhythmias, aortopathies, dyslipidemias, and congenital heart defects. Their prevalence and genetic testing yield range from 1/100-1/20,000 and 10%–80%, respectively, depending on the condition. In Israel, gene-panel testing is covered by the Ministry-of-Health, for conditions with a diagnostic yield >50% in gene-panel.

Methods: Between 05/2019-01/2024, 158 patients were evaluated in the Cardiogenetics clinic. Most (79%) were referred for a personal diagnosis of cardiomyopathy or arrhythmia, with a wide range of ages (2-70 years). All underwent genetic counseling, personal and 3-generation family history was taken, and patients were offered genetic testing.

Results: A total of 87 patients (55%) underwent genetic testing, mostly by multi-gene panel testing. Overall, a positive result was found in 26% of cases. The highest diagnostic rate, 40% was found in hypertrophic cardiomyopathy patients. Variants of uncertain significance were found in 28% of cases. Compliance with testing was higher in those eligible for funding for testing, 60% vs. 43%.

Conclusion: : A multidisciplinary specialized Cardiogenetics clinic shows high compliance rates for genetic testing, which also depend on financial coverage. The yield varies depending on the condition, with overall molecular diagnosis of 1 out of 4 cases. Factors affecting diagnostic yield, which is lower than reported in the literature should be further investigated in a larger sample size.

Conflict of Interest: None declared

EP07.044 Genomic etiology and genotype-phenotype correlations of inherited aortopathy

Pawares Chitayanan 1

1Chulalongkorn University, Clinical Biochemistry, Thailand

Background/Objectives: This study aimed to investigate the genomic etiology of aortopathy in individuals under 60 years old and to discern genotype-phenotype correlations associated with this condition.

Methods: Patients presenting with aortic root dilatation or aortic dissection, referred to a clinical genetics service for evaluation, underwent whole-exome sequencing.

Results: Over the period from June 2014 to December 2023, 31 patients with aortopathy correlated with a genetic disorder were enrolled. The average age was 28.29 ± 11.66 years, predominantly male (64.50%). The mean z-score for aortic root size at the sinus of Valsalva was 4.98 ± 2.58. Aortic dissection was observed in 8 cases (25.81%). Genomic variants associated with aortopathy were identified in 26 cases (83.87%), with 16 pathogenic (51.60%) and 7 likely pathogenic variants (22.60%). FBN1 variants linked to Marfan syndrome were most prevalent, with null variants correlating with severity, e.g., early-onset aortic root surgery and the presence of lens subluxation. However, no genotype-phenotype correlation was found between types of FBN1 variants and systemic manifestations as per the Revised Ghent Nosology Criteria. Other contributing genes characterized in this study included ACTA2, COL3A1, DCHS1, FBN2, and TGFBR1

Conclusions: This study sheds light on the genomic underpinnings of inherited aortopathy and underscores the complexity of genotype-phenotype relationships in this condition. This data constitutes a pivotal factor for the development of precise management and the provision of genetic counseling to patients and their families.

Grants: Support for this research was provided by the Faculty of Allied Health Sciences, Chulalongkorn University.

Conflict of Interest: None declared

EP07.045 Clinical and functional characterization of MYBPC3 c.1458-1G > A and c.3331-1G > A founder mutations

Miryam Rosa Stella Foti1, Naomi Fanciullo2, Camilla Lucca3, Carlotta Pia Cristalli4, Chiara Diquigiovanni4, Agnese Scatigno5, Maria Schiavo2, Maddalena Graziosi2, Silvia Palmieri2, laura pezzoli3, Francesco Lai6, Vera Uliana7, Federico Barocelli7, Elia De Maria8, Alessandro Fucili9, Biagio Sassone10, Giulia Parmeggiani11, Enrica Perugini12, Rita Selvatici1, Alessandra Ferlini1, Maria Iascone3, Elena Biagini2, Cesare Rossi4, Francesca Gualandi 1

1Unit of Medical Genetics- University-Hospital Sant’Anna, Department of Medical Sciences and Department of Mother and Child, Ferrara; 2Cardiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Cardiac Thoracic and Vascular Department; 3ASST Papa Giovanni XXIII, Laboratory of Medical Genetics; 4IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna., UOC Genetica Medica; 5ASST Papa Giovanni XXIII, Paediatric Unit; 6Ospedale di Bentivoglio, U.O.C. Cardiologia; 7University Hospital of Parma, Unit of Cardiology; 8Ramazzini Hospital, Carpi, Modena, Cardiology Unit; 9University Hospital S. Anna Ferrara, Ferrara, Cardiology Department; 10SS.ma Annunziata Hospital, AUSL Ferrara, Cento, Ferrara., Cardiology Division, Department of Emergency; 11Medical Genetics Unit, AUSL Romagna, Cesena, Department of Clinical Pathology; 12Maggiore Hospital, Bologna, Cardiology Unit

Background/Objectives: Mutations in MYBPC3 gene, encoding the cardiac isoform of myosin-binding protein C, are associated with different cardiological phenotypes, mostly hypertrophic cardiomyopathy (HCM). Truncating mutations account for 72% of cases, followed by splicing (22%) and missense (6%) variants.

Methods: As regional reference centres for the clinical and genetic diagnosis of cardiomyopathies, we identified two recurrent MYBPC3 splicing mutations (c.1458-1G > A and c.3331-1G > A) occurring in 34 and 15 HCM probands, respectively. All analysed families originate from Emilia-Romagna Region, suggesting a founder effect and haplotype study is underway. Familial history, clinical presentation and follow-up findings were collected. RNA study was performed on endomyocardial biopsy tissue and peripheral blood.

Results: Both mutations induce aberrant splicing. In particular, c.1458-1G > A causes intron 16 retention while c.3331-1G > A determines the retention of 90bp of intron 30. The mutated alleles appear negatively selected, suggesting their degradation by nonsense-mediated mRNA decay. Preliminary clinical data attest a wide variability in age at diagnosis, generally made by chance or for positive familial history, and a low progression rate. Relevant associated findings are atrial fibrillation (20%), mostly parossistic (70%), apical involvement (15% of cases) and a restrictive pattern and/or outflow obstruction, detected in about 10% of the cohort.

Conclusion: The availability of a large cohort of patients carrying two recurrent MYBPC3 splicing mutations of founder origin allowed a detailed definition of genotype-phenotype correlations, with great impact on family counseling and on the evaluation of future therapeutic approaches.

Grants: RC-2022-2773270 project (Italian Ministry of Health), and the italian parents association “Voglio Volare -Davide Barbi”.

Conflict of Interest: None declared

EP07.046 Expanding the molecular spectrum of MCTP2 gene in patients with coarctation of the aorta.

Daniele Perrino 1, Monia Magliozzi1, Daniela D’Angelantonio1, Giovanni Parlapiano2, Anwar Baban2, Antonio Novelli1

1Bambino Gesù Children’s Hospital, IRCCS, Translational Cytogenomics Research Unit, Rome, Italy; 2Bambino Gesù Children’s Hospital, IRCCS, Pediatric Cardiology and Arrhythmia/Syncope Complex Unit, Rome, Italy

Backgroud: Left ventricular outflow tract obstructive defects (LOVTO) are a series of disorders that lead to a restricted blood flow from the left ventricle and express a variety of clinical manifestations. Coarctation of the aorta (CoA), a subtype of LOVTO, occupies 5–8% of all congenital heart defects. In 2013, MCTP2 gene has been identified as a possible genetic cause of CoA and related cardiac malformation. Currently, only few variants have been identified in this gene, mainly in patients diagnosed with coarctation of the aorta and suspicious bicuspid aortic valve.

Method: Genetic testing was performed at the genetic Laboratories of the Bambino Gesù Children’s hospital. The presence of rare variants (MAF < 0.01) involved “congenital heart defect”, “Aortic Coartaction” have been evaluated. Genetic testing was performed by a Next Generation Sequencing using an in silico panel, including MCTP2 and other cardiopathy associated genes

Results: We identified a group of patients with Coarctation of the aorta (CoA) harboring previously unreported variants in the MCTP2 gene. Here, we present clinical and molecular evidences supporting the likely pathogenic nature of this variants.

Discussion: The discovery of a new pathogenic variants in the MCTP2 gene expands the current variant spectrum in LOVTO patients and may lead to more informed clinical decisions for the timing and nature of interventions.

Conflict of Interest: None declared

EP07.047 Genetic diagnostic of hereditary heart diseases using next generation sequencing: experience from a tertiary hospital

Miriam Potrony 1;2;3, Irene Madrigal1;2, Maria Isabel Alvarez1;2;3, José Villanueva-Cañas4, Aina Montalban-Casafont4, Oscar Campuzano5;6, Aleksandra Mas-Stachurska7, Eduard Solé-González3;7, Aurora Sánchez Díaz1;2;3, Elena Arbelo3;6;7, Ana Garcia3;6;7, Celia Badenas1;2

1Biochemistry and Molecular Genetics Department, Hospital Clínic of Barcelona, IDIBAPS, Barcelona, Spain; 2CIBER of Rare Diseases (CIBERER), ISCIII, Barcelona, Spain; 3European Reference Network for rare, low prevalence and complex diseases of the heart - ERN GUARD-Heart, Barcelona, Spain; 4Molecular Biology CORE, Hospital Clínic of Barcelona, IDIBAPS, Barcelona, Spain; 5Medical Sciences Department, University of Girona, Girona, Spain; 6CIBER of Cardiovascular Diseases (CIBERCV), ISCIII, Madrid, Spain; 7Cardiovascular Institute, Hospital Clínic of Barcelona, IDIBAPS, Barcelona, Spain

Background/Objectives: Hereditary heart diseases are the first cause of sudden death in young people and a relevant source of heart failure. We aimed to assess the diagnostic performance of next generation sequencing (NGS) in patients with hereditary heart disease in clinical practice.

Methods: Exome sequencing was performed in 399 patients with cardiomyopathy, channelopathy or aortopathy, attended at Hospital Clinic of Barcelona (2019-2023). NGS was performed using Illumina technology and an in-house bioinformatic pipeline. Virtual gene panels were used according to genes associated with the referral reason. ACMG variant classification guidelines were used.

Results: Pathogenic/likely pathogenic (P/PP) variants were identified in 21% (85/399), variants of unknown significance (VUS) in 28% (113/399), and no variants were identified in 51% (201/399) of patients. The diagnostic yield varied according to the referral reason (Table). Most common mutated genes were MYBPC3 (29%), MYH7 (18%), TTN (7%), LMNA (6%), FLNC (6%), FBN1 (6%), SCN5A (5%) and DSP (4%).

Hereditary Heart Disease

P/PP

VUS

Negative

Hypertrophic cardiomyopathy(N = 166)

48 (29%)

48 (29%)

70 (42%)

Dilated cardiomyopathy(N = 43)

13 (30%)

20 (47%)

10 (23%)

Arrhythmogenic cardiomyopathy(N = 43)

7 (17%)

12 (28%)

23 (55%)

Brugada Syndrome(N = 86)

5 (6%)

19 (22%)

62 (72%)

Other cardiopathy(N = 41)

3 (7%)

10 (25%)

28 (68%)

Aortopathy(N = 21)

9 (43%)

4 (19%)

8 (38%)

Conclusion: The use of NGS for genetic diagnostic of hereditary heart diseases in clinical practice in a tertiary hospital resulted in a diagnostic yield of 21%. The high detection of VUS highlights the need to perform additional studies for reclassification and improvement of genetic testing.

Conflict of Interest: None declared

EP07.049 Genetic Testing of Paediatric Cardiomyopathies in Latvia: Confirming the Implication of CASZ1

Ludmila Voložonoka 1, Līvija Bārdiņa1, Egija Berga Svitina1, Irina Žiravecka1, Marija Jurcenko1, Ieva Mičule1, Ieva Grīnfelde1, Gita Taurina1, Dmitrijs Rots1

1Children’s clinical university hospital, Riga, Latvia

Background/Objectives: Paediatric cardiomyopathies (CMP) are rare yet serious conditions with high mortality rates. Genetic testing for CMP is necessary for accurate diagnosis, disease-specific prognosis, and management. Up to 40% of cases can be attributed to a genetic cause.

Methods: Newborns to 18-year-old patients were recruited at Children’s University Hospital in Riga, Latvia. Three cases were diagnosed with dilated cardiomyopathy (DCM), four with left ventricular non-compaction (LVNC), and eleven with hypertrophic CMP (HCM). Sixteen cases were non-syndromic, while two were syndromic. Whole-exome sequencing (WES) was performed for all, analysing 194 genes associated with CMP.

Results: Two patients had MYH7 variant explaining LVNC. A two-year-old patient with HCM had a pathogenic MYBPC3 variant, and a MYH7 VUS. In two patients Noonan syndrome due to PTPN11 and SOS1 variants was confirmed. In one patient NSD1-related Sotos syndrome was confirmed. One patient with LVNC had synonymous MYH7 variant predicted to affect splicing classified as a VUS. Additionally, in a 14-year-old female patient with LVNC, CMP panel testing was negative. Upon request to expand the analysis to open exome, LP loss-of-function variant NM_001079843.2:c.2583del in CASZ1 gene was identified. Notably, CASZ1 lacked a clinical association with any diagnosis in OMIM/PanelApp (January 2024), although CASZ1 is implicated in CMP in the literature.

Conclusion: Our study unveils the initial outcome of genetic testing for CMPs in Latvia. Seven patients (39%) received a definitive diagnosis for their condition, showcasing a significant diagnostic yield by using singleton WES. Consequently, the identification of a truncating CASZ1 variant in our cohort confirms the gene’s involvement in paediatric CMPs.

Conflict of Interest: None declared

EP07.050 Whole exome sequencing reanalysis unveils AKA9P frameshift variant associated with cardiac arrest and long QT intervals

Maria Soares Nobrega1, maria abreu2, rui baptista3, rita cerqueira 1, joaquim sá1;4, marisa teixeira1

1CGC Genetics, Unilabs, Porto, Portugal; 2Medical Genetics Unit, ULS de Santo António; 3Cardiology Unit, ULS Entre Douro e Vouga; 4Medical Genetics Unit, ULS de Coimbra

Background/Objectives: Cardiovascular diseases are often heritable, hence the importance of identifying new genetic variants associated with familial heart conditions. Here we report a 54 years old woman recovered from a cardiac arrest with ventricular fibrillation exhibiting prolonged QT intervals and ventricular fibrillation with a family history of arrhythmias and cardiovascular diseases. The patient’s cardiomyopathy panel was negative, therefore, whole exome sequencing (WES) was performed for further investigation.

Methods: WES was performed at Illumina Novaseq 6000® and the resulting data was processed and analyzed with an in-house bioinformatics pipeline and NGS analysis software.

Results: WES was performed and single nucleotide variant (SNV) analysis detected the frameshift variant NM_005751.5:c.2476dup p.(Ile826Asnfs*5), in heterozygosity, in exon 8 of 50 in the gene AKAP9. Variant presence was then confirmed through Sanger sequencing. According to ACMG criteria, the classification is a variant of unknown significance (PM2supporting/PVS1strong).

Conclusion: Here we report a woman diagnosed with long QT syndrome 11 [MIM 611820], presenting a loss of function variant in AKAP9 gene. Although clinical evidence for this gene is currently limited, a 47 years old patient with severe ventricular arrhythmia and cardiac arrest was recently documented with a frameshift variant in the same gene [PMID:36421840]. Therefore, this case supports the association of ventricular arrhythmia and long QT intervals to loss of function variants in AKAP9 gene and emphasizes the importance of WES reanalysis to broaden diagnostic possibilities for individuals who initially received a negative result in genetic panels.

Grants:

Conflict of Interest: None declared

EP07.051 Does the phenomenon of genetic anticipation exist in the hypertrophic cardiomyopathy?

Jesús Wagih Gómez 1, Lidia María Carrillo2, Cristina Gil3, Elisa Nicolas Rocamora1, David López2, María del Carmen Olmo2, Carmen Muñoz2, Marina Navarro2, Serena Evelina Margaretha Munteanu1, Juan Ramón Gimeno Blanes2, Maria Sabater Molina1;3

1Imib Instituto Murciano De Investigación Biosanitaria, El Palmar, Spain; 2Virgen of Arrixaca University Clinical Hospital, El Palmar, Spain; 3University of Murcia, Murcia, Spain

Background/Objectives: Genetic anticipation (GA) is a phenomenon whereby the age of onset and phenotypic severity increases over generations. This occurrence has been established in neurodegenerative disorders and in non-sense PALB2 cancer. Our aim was study GA in families with Hypertrophic cardiomyopathy (HCM) with non-sense mutations in MYBPC3.

Methods: We included 346 patients carrying mutations in MYBPC3 from 104 families. There were 210 (60.7%) affected and 136 (39.3%) unaffected carriers. Three generations (G): born between 1940-65 (137, 39.6%), 1966-90 (153, 44.2%) and 1991-2015 (53.15.3%). Age at diagnosis, phenotype severity and related cardiac events were studied.

Results: Age at diagnosis was lower for younger generations, symptoms being more prevalent in 1stG (46.4 vs 20.5%, p < 0.05). The echocardiographic phenotype was similar between 1stG vs 2ndG. Mean thickness (19.2 ± 4.9mm vs 18.9 ± 6.3mm, p = 0.75) and percentage of obstruction were similar (24.1vs 27.1%, p = 0.64). Left atrial size (42.4 ± 12.8mm vs 35.1 ± 13.9 mm p < 0.01), in addition, these patients were more symptomatic (16.7 vs 3.6%, p < 0.01) and had more atrial fibrillation (49.5vs 10.8%, p < 0.01). There were 2, 1.46% cases with sudden death (SD) or equivalent in 1stG (follow-up 79.5 months) and 4, 2.61% in 2ndG (follow-up 57.8 months). SD-free survival was significantly lower in the 2ndG (log rank p < 0.01).

Conclusion: The age of diagnosis is earlier in younger cases, probably due to the implementation of screening programmes. Phenotype is similar, with no significant differences in the hypertrophy and obstruction. The prognosis of major events could be worse in successive generations.

Conflict of Interest: None declared

EP07.052 Atrial amyloidosis caused by bialelic mutations in CORIN

Lenka Piherová 1, Petra Melenovská1, Viktor Stránecký1, Lenka Steiner-Mrazova1, Alena Vrbacká1, Helena Trešlová1, Alice Krebsová2, Miloš Kubánek2, Vojtech Melenovský2

1First Faculty of Medicine, Charles University Prague, Research Unit for Rare Disorders, Prague 2; 2Institute for Clinical and Experimental Medicine, Prague 4, Czech Republic

Background/Objectives: Middle-aged woman with hypertension, AFib and an incidental LA mass with a rim of late gadolinium-enhancing tissue. Upon surgery, an intraumal hematoma was found within entirely diffusely thickened LA wall, extensively infiltrated by ANP-containing amyloid (confirmed by immunohistochemistry and mass spectrometry). ANP gene sequence was normal, but western-blot of LA tissue showed predominance of proANP, consistent with defective CORIN processing. ANP precursor contains amyloid aggregation-prone segment.

Methods: ANP a CORIN exons were sequenced by Sanger method. By this method we found missense variant in exone 16. A large part of the gene was sequenced on a MinION sequencer. Large deletion was detected by genome sequencing on Illumina platform.

Results: We found bialelic variant in CORIN, which is serine protease cleaving proANP to ANP. Rare missense variant is located in scavenger receptor domain and second variant is a large deletion, found by genome sequencing. Deletion boundaries are localized in repetitive LINE/L1 segments. This variant leads to production of neo-protein, whereare scavenger receptor domain and peptidase S1 domain affected.

Conclusion: Due to biallelic variants in CORIN, protease enzymatic activity is affected. It is therefore likely that defects in proANP processing may lead to more extensive LA wall infiltration by ANP amyloid, mimicking tissue fibrosis on MR imaging, and contributing per se to arrhythmogenesis and LA dysfunction.

Grants: The project National Institute for Research of Metabolic and Cardiovascular Diseases (Programme EXCELES, Project No. LX22NPO5104) - Funded by the European Union – Next Generation EU.

Conflict of Interest: None declared

EP07.053 Variants in LPA are associated with mutation-negative Familial Hypercholesterolaemia: whole genome sequencing analysis in the 100,000 Genomes Project

Martin Bird 1, Antoine Rimbert2, Alan Pittman1, Steve Humphries3, Marta Futema1

1St George’s University of London, Cardiovascular and Genomics Research Institute, London, United Kingdom; 2Nantes Université, l’institut du thorax Unité, Nantes, France; 3University College London, Institute of Cardiovascular Science, London, United Kingdom

Background/Objectives: Familial Hypercholesterolaemia (FH) is an inherited disease of high LDL-cholesterol (LDL-C) caused by defects in LDLR, APOB, APOE and PCSK9 genes. A pathogenic variant cannot be found in ~60% of clinical FH patients. Using whole genome sequencing (WGS) we examined genetic determinants of FH phenotype.

Methods: WGS data generated by the 100,000 Genomes Project (100KGP) included 536 FH patients diagnosed using the FH Simon Broome criteria. Rare variants in known FH genes were analysed. Genome wide association study (GWAS) between FH variant-negative unrelated FH cases and 50,109 control participants of the 100KGP was run using high coverage WGS data. Polygenic risk scores for LDL-C (LDL PRS) and lipoprotein(a) (LPA PRS) were computed.

Results: An FH-causing rare variant was found in 17.4% of FH participants. GWAS of the FH variant-negative participants identified the LPA gene locus being significantly associated (p < 3.56x10-9). FH variant-negative participants had significantly higher LDL and LPA PRSs in comparison to the controls (p = 1.43×10-28 and p = 1.41×10-6, respectively). High LDL PRS was observed in 36.3% of FH variant-negative cases, whereas high LPA PRS in 18.5%, with 6.5% having both high LDL and LPA PRSs.

Conclusion: This genome-wide analysis of monogenic and polygenic FH causes confirms a complex and heterogenous genetic architecture of hypercholesterolaemia, with LPA playing a significant role. Both Lp(a) and LDL-C measurements should be included in the differential diagnosis of FH. Specific therapies to lower Lp(a) should be targeted to those who will benefit most.

Grants: St. George’s, University of London

Conflict of Interest: None declared

EP07.054 Evolution and diagnostic yield of an off-site outpatient cardiogenetics clinic

Vincent van der Pas1, Christian van der Werf2, Alexa Vermeer3, Eline Nannenberg3, Jurren van Opstal1, Arthur Wilde2, Saskia Van der Crabben 3

1MST, Cardiology, Enschede, Netherlands; 2Amsterdam UMC, locatie AMC, Cardiology, Amsterdam, Netherlands; 3Amsterdam University Medical Center, Human Genetics, Amsterdam, Netherlands

Background: With increasing demands for genetic testing in patients suspected of inherited cardiac conditions, 8 multidisciplinary outpatient cardiogenetics clinics from Amsterdam UMC in large, non-academic, hospital provides: on-site counselling by clinical geneticists/cardiologists, easier access to multidisciplinary cardiogenetics consultation and cross-disciplinary education on the appropriate use of genetic testing. Data of the largest outpatient clinic was evaluated for genetic yield.

Methods: Clinical phenotypes and genetic test results of unrelated probands (>18 years) referred to the multidisciplinary cardiogenetics outpatient clinic at Medisch Spectrum Twente (Enschede, Netherlands) between June 2018 to December 2023 were evaluated.

Results: Referrals increased from 2018 (n = 23), 2019 (n = 57), 2020 (n = 68), 2021 (n = 72), 2022 (n = 130) to 2023 (n = 139). Of 489 referred patients, 12 (2.5%) did not appear for counselling, 49 (10.0%) deferred from genetic testing and 17 (3.5%) did not meet clinical criteria for genetic testing after multidisciplinary discussion. Currently genetic test results of 335 patients (59.7% male; 51.6 ± 13.3 years) were available:

Genetic test result

Fenotype

N=

VUS

Class 4/5

cardiomyopathy

234 (69.9%)

110 (47.0%)

39 (16.6%)

Cardiac arrhythmia

83 (24.8%)

17 (20.4%)

15 (18.1%)

Aortic aneurysm

11 (3.3%)

1 (9.1%)

4 (36.4%)

Other

7 (2.1%)

1 (14.3%)

1 (14.3%)

Total

335 (100%)

129 (38.5%)

59 (17.6%)

Conclusion: These data show that close collaboration increased referrals, mainly of cardiomyopathy patients. Relatively high number of patients declining genetic testing could reflect local values. The comparatively high yield in arrhythmia might be related to a regional class 5 SCN5A variant.

Grants: Not applicable

Conflict of Interest: None declared

EP07.056 Deletion of the STS gene leads to isolated atrial fibrillation in a young male patient

Marie Arens1, Valerio Rhodio2, Elisabeth Graf2, Eimo Martens1, Juliane Winkelmann2, Dominik S. Westphal 1;2

1Klinikum rechts der Isar der Technischen Universität München, Department of Internal Medicine I, München, Germany; 2Klinikum rechts der Isar der Technischen Universität München, Institute of Human Genetics, München, Germany

Background/Objectives: Atrial fibrillation (AF) is mostly a polygenic inherited disease affecting elderly patients. However, juvenile AF may be the first symptom of a monogenic inherited cardiac disease. Recently, X-linked ichthyosis, often caused by deletions of the STS gene, has been associated with an increased risk for arrhythmias, including AF.

Methods: DNA was isolated from the patient’s peripheral blood lymphocytes. Exome sequencing (ES) was carried out using the Twist enrichment protocol and the Twist Human Exome 2.0 Plus Comprehensive Exome Spike-in (Twist Bioscience, San Francisco, California). Sequencing was performed on a NovaSeq 6000 as 100 bp paired end run (Illumina Inc., San Diego, California). Reads were aligned to the University of California Santa Cruz human reference assembly (hg19) using Burrows-Wheeler Aligner v.0.5.8.

Results: At the age of 26 years, a male patient presented to our emergency department with tachycardia and palpitations. The patient had no relevant medical history and family history was unremarkable. Tachycardic AF was diagnosed by ECG. There was a conversion to sinus rhythm under beta-blocker treatment. ES identified a hemizygous deletion in Xp22.31, comprising the STS gene. The patient, however, did not show clinical signs of ichthyosis.

Conclusion: To the best of our knowledge, we report the first male patient with AF, caused by deletion of Xp22.31 who is not affected by ichthyosis. Young male patients with AF should be screened for deletions of STS, even without skin lesions. By focusing only on cardiac disease genes in juvenile AF, other inherited non-cardiac diseases may be overlooked.

Grants: No funding was received for this study.

Conflict of Interest: None declared

EP07.057 Mitochondrial Complex I Deficiency: Unraveling the relevance of NDUFAF1 and WES in Pediatric Hypertrophic Cardiomyopathy

Silvia Kalantari 1, Daniele Veraldi2, Davide Politano3, Gaia Visani1, Antonia Apicella2, Elisa Giorgio1;4, Enza Maria Valente1;4, angela berardinelli5, Claudia Codazzi2, Fabio Sirchia1;6

1University of Pavia, Department of Molecular Medicine, Pavia, Italy; 2IRCCS Policlinico San Matteo Foundation, Pediatric Clinic, Pavia, Italy; 3University of Pavia, Department of Brain and Behavioral Sciences, Pavia, Italy; 4IRCCS Mondino Foundation, Medical Genetics Unit, Pavia, Italy; 5IRCCS Mondino Foundation, Department of Child Neurology and Psychiatry, Pavia, Italy; 6Fondazione IRCCS Policlinico San Matteo Foundation, Medical Genetics Unit, Pavia, Italy

Background/Objectives: Hypertrophic cardiomyopathy (HCM) is rare in childhood, but it is associated with significant morbidity and mortality. Genetic causes of HCM are mostly related to sarcomeric genes abnormalities, however syndromic, metabolic, and mitochondrial disorders play an important role in its etiopathogenesis in pediatric patients. Our objective is to describe a new case of HCM due to mitochondrial assembly factor gene NDUFAF1 biallelic variants. Alterations of this nuclear gene have been associated to Mitochondrial complex I deficiency, nuclear type 11 (OMIM *618234).

Methods: TRIO-based exome sequencing (ES) was performed, and the variants identified were then confirmed by Sanger sequencing and quantitative RT-PCR.

Results: Our case presented with HCM at 6 years of age, with unremarkable previous clinical and family history. TRIO ES identified a missense pathogenic variant [c.631C>T (p.Arg39Cys)] and an intragenic deletion encompassing exon 3 in the NDUFAF1 gene in compound heterozygous state in the patient. Following the diagnosis, brain imaging, EEG, electromyography, retinal and audiological exams were performed, and no abnormality was detected.

Conclusion: We here report the fourth case in the world of a child affected by complex I deficiency due to alterations in NDUFAF1 gene. His clinical picture appears milder when compared to the other cases described in the medical literature, increasing our knowledge regarding the highly heterogeneous clinical presentation associated with this disorder. Due to our patient’s mild clinical picture, a mitochondrial disease would never have been hypothesized prior to ES, underlying the importance of this analysis in pediatric HCM.

Grants:

Conflict of Interest: None declared

EP07.058 Analysis of the clinical and molecular characteristics of MYBPC3 and MYH7 related hypertrophic cardiomyopathy

Dovile Zebrauskiene 1;2, Egle Sadauskiene2;3, Ruta Masiuliene4, Jurate Barysiene2;3, Egle Preiksaitiene1

1Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania; 2Centre of Cardiology and Angiology, Vilnius University Hospital Santaros klinikos, Vilnius, Lithuania; 3Clinic of Cardiac and Vascular Diseases, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania; 4Faculty of Medicine, Vilnius University, Vilnius, Lithuania

Background/Objectives: Hypertrophic cardiomyopathy (HCM) is an inherited highly heterogeneous disease with variable penetrance and expression. MYBPC3 and MYH7 are the most affected genes causing sarcomeric HCM. The aim of the study was to analyse the phenotypic and molecular characteristics of individuals with MYBPC3 and MYH7 related HCM, examined and treated at Vilnius University Hospital Santaros klinikos (VUHSK).

Methods: We analysed data from clinical records of individuals with HCM treated at VUHSK between 2005 and 2023. Only unrelated individuals with pathogenic or likely pathogenic (P/LP) variant in MYBPC3 or MYH7 identified by next-generation sequencing were included for the final evaluation.

Results: Out of 65 unrelated individuals, 69% (n = 45) had the P/LP variant in the MYBPC3 (MYBPC3 group) and 31% (n = 20) had the P/LP variant in MYH7 (MYH7 group). In total 67 P/LP variants in MYBPC3 and MYH7 were identified (2 patients had 2 P/PL variants in MYBPC3 gene). Almost all individuals in MYH7 group had missense variants (18/20) while most patients in MYBPC3 group had truncating variants (27/47), p < 0.001. More individuals in MYH7 group had atrial fibrillation (AF) during primary evaluation than in MYBPC3 group (35.0% vs. 11.1%, p = 0.036). Whereas other clinical signs, instrumental and laboratory findings as well as follow-up data (interventional treatment and outcomes) were not statistically significantly different between the groups.

Conclusion: Individuals with MYH7 related HCM tend to have a higher incidence of AF. According to other clinical signs and findings of instrumental or laboratory tests MYH7 and MYBPC3 related HCM are phenotypically indistinguishable.

Grants: This research received no external funding.

Conflict of Interest: None declared

EP07.059 Diagnostic yield of chromosomal microarray analysis in patients with congenital heart disease

Tatjana Damnjanovic 1, Dijana Perovic1, Nela Maksimovic1, Biljana Jekic1, Marija Dusanovic Pjevic1, Milka Grk1, Ana Djuranovic1

1Institute of Human Genetics, Faculty of Medicine, UBFM, Human genetics, Belgrade

Objectives: Congenital heart disease (CHD) is the most common birth defect. According to published data, clinically significant copy number variants (csCNV) represent approximately 15% of genetic causes of CHD. We aimed to evaluate the diagnostic contribution of array comparative genomic hybridization (aCGH) in children with CHD and to analyze the distribution of different types of detected csCNV.

Methods: From patients who were referred to aCGH between 2018 and 2023, we singled out 196 children with CHD. aCGH analysis was performed by Agilent 8x60k slides.

Results:The study included 99 boys and 97 girls with a mean age of 3.17 ± 5.15. The detection rate of csCNV was 27.55%. Deletions were present in 61.1% of children. Recurrent CNVs were detected in 48.1% of cases. Among them most frequent are 22q11.21 (26.9%) and 7q11.23 deletion (15.4%). Majority of detected non-recurrent csCNV do not include genes directly associated with CHD. However, genes implicated in occurrence of CHD could be SOS1, SPAG1, NR2F2 and NFATC1, included in regions 2p22.2-p22.1, 8q22.2-q22.23, 15q26.2, and 18q21.33-q23, respectively. Among the children with seemingly isolated CHD (12.7%), 22q11 deletion was detected in two newborns without other clinical signs at the time of analysis (8%). Children with csCNV were statistically significantly more likely to have global developmental delay/ intellectual disabilities (p = 0.03), dysmorphia (p = 0.04), and epilepsy (p = 0.03) compared to children without csCNV.

Conclusion: aCGH is a significant diagnostic tool in patients with CHD, even in the case of isolated form since it enables early detection of syndromic CHD before the appearance of other symptoms.

Conflict of Interest: None declared

EP07.060 Genetic variants and clinical manifestations in familial atrial fibrillation: a systematic review

Maxime van Keulen1, Leonoor Wijdeveld 2, Dennis Dooijes1, Karin van Spaendonck-Zwarts3, Bianca Brundel2, Peter van Tintelen1

1Utrecht UMC, Clinical Genetics, Utrecht, Netherlands; 2Amsterdam UMC, Physiology, Amsterdam, Netherlands; 3Groningen University Medical Center, Clinical Genetics, Groningen, Netherlands

Consortium: CIRCULAR

Background/Objectives: Atrial fibrillation (AF) is a common arrhythmia associated with stroke and heart failure. In a subset of cases, AF manifests at a younger age (below 60 years), in the absence of known risk factors, and clusters within families. These familial AF cases are suspected to have a monogenetic substrate. Here, we systematically review published familial AF cases and delineate patterns in genetic testing results and symptomatology.

Methods: PubMed and Embase were searched for studies describing AF cases with 1+ affected family member(s) that included clinical data and genetic investigations. Articles describing secondary AF were excluded. For all affected relatives, data was collected on family history, age of diagnosis, symptoms, ECG and echo parameters, and genetic testing.

(Preliminary) results: The search resulted in 35 studies, containing 61 families and 194 affected individuals. Genetic testing ranged from single gene to whole exome sequencing. Studies focussed on genes known for overlap between AF, cardiomyopathies and arrhythmia syndromes. The selected studies reported 54 unique genetic variants. Most variants were in genes encoding ion channels (55.1%) and cytoskeletal proteins (20.4%). After reclassification, 15 variants are ostensibly pathogenic or likely pathogenic. Symptomatology varied widely, with 35 subjects (25.4%) harbouring predisposing variants being asymptomatic.

Conclusion: Most genetic variants were found in genes encoding ion channels. 194 subjects with familial AF demonstrated varied symptomatology with 25% being asymptomatic. Further studies, including segregation analysis and detailed clinical descriptions, are warranted to study the indications and implications for genetic testing in familial AF.

Grants: NWA-ORC project CIRCULAR NWO (NWA.1389.20.157)

Conflict of Interest: None declared

EP07.061 Assessment of arrhythmia-related genes in patients prediagnosed with long QT syndrome

Feyza Altunbas Yalabik 1, Aslı Toylu1, Ibrahim Basarici2, ercan mihci3, banu nur3, Fırat Kardelen4, Özden Altıok Clark1

1Akdeniz University Faculty of Medicine, Medical Genetics, Antalya, Türkyie; 2Akdeniz University Faculty of Medicine, Cardiology, Antalya, Türkyie; 3Akdeniz University Faculty of Medicine, Pediatric Genetics, Antalya, Türkyie; 4Akdeniz University Faculty of Medicine, Pediatric Cardiology, Antalya, Türkyie

Background/Objectives: Long QT Syndrome (LQTS) is an inherited channelopathy characterized by varying degrees of prolongation of the corrected QT interval on the electrocardiogram (>450 ms) and associated with an increased susceptibility to life-threatening ventricular arrhythmias. Most LQTS cases are associated with pathogenic variants in the KCNQ1, KCNH2, and SCN5A genes. Several other genes encoding either ion channel subunits or proteins that regulate channel function have also been implicated in LQTS causality.

Methods: We investigated 10 cases that underwent genetic testing for diagnosis of LQTS. LQTS-associated target genes were analyzed by next-generation sequencing. Variants were classified according to ACMG guidelines.

Results: Previously reported pathogenic variants in KCNQ1, KCNH2, SCN5A, and KCNE1 genes have been observed in seven patients. Variants with unknown clinical significance have been identified in KCNQ1, SCN5A, and AKAP9 genes in three cases. Pathogenic variants were most frequently observed in the KCNQ1 gene. Interestingly, we identified the c.1097G>A (p.Arg366Gln) KCNQ1 variant in 3 unrelated patients.

Conclusion: Our results include known pathogenic variants reported in the literature and various variants whose clinical significance is yet to be understood. Our study is expected to contribute to the literature by emphasizing the significance of genetic diagnosis in LQTS cases.

Grants: Akdeniz University Scientific Research Projects Coordination Unit, Project ID: 6541

Conflict of Interest: None declared

EP07.062 Association of C11orf58 gene, encoding chaperon “Hero”, with the increased risk of ischemic stroke

Irina Shilenok1;2, Ksenia Kobzeva 1;3, Olga Bushueva1

1Kursk State Medical University, Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk; 2Kursk Emergency Hospital, Division of Neurology, Kursk; 3Kursk State University, Department of Biology and Ecology, Kursk

Background/Objectives: Ischemic stroke (IS) holds a prominent position as a leading contributor to worldwide mortality and disability. While the involvement of chaperones in IS risk is well-established, the role of a recently discovered group of heat-resistant obscure proteins (“Hero” acronymically) with chaperone-like properties in the development of IS remains unexplored. This study aims to examine whether SNPs of C11orf58, a member of the “hero” proteins, are associated with the risk of IS development.

Methods: DNA samples from 2204 unrelated Russian subjects (917 IS patients, 1287 healthy controls) were genotyped for 14 common SNPs: rs6677, rs1846936, rs11024031, rs10766342, rs7928675, rs11024032, rs4757430, rs7951676, rs11826990, rs3203295, rs10832676, rs4757429, rs3802963, C11orf58 using probe-based PCR, and rs10734249 C11orf58 – using the MassARRAY-4 genetic analyzer.

Results: We observed an association between C11orf58 SNPs and increased IS risk across the entire group: rs10766342 (risk allele A, p = 0.02), rs11024032 (risk allele T, p = 0.014), rs11826990 (risk allele G, p = 0.0072), rs3203295 (risk allele C, p = 0.016), rs10832676 (risk allele G, p = 0.0056), rs4757429 (risk allele T, p = 0.023). Associations of these SNPs were modified by environmental factors, specifically smoking and fruit/vegetable consumption. Moreover, the SNP rs11024030 (risk allele C) is linked to an increased risk of IS exclusively in smokers (p = 0.032) and in patients with low fruit/vegetable intake (Pbonf = 0.032).

Conclusion: Thus, polymorphisms in the C11orf58 gene represent novel genetic markers of IS.

Grants: This research was funded by Russian Science Foundation (№ 22-15-00288, https://rscf.ru/en/project/22-15-00288/).

Conflict of Interest: None declared

EP07.063 Genetic findings of paediatric patients tested for long QT syndrome

Aurelija Kemezyte 1, Raimonda Meskiene2, Birutė Burnytė2

1Vilnius University, Faculty of Medicine, Vilnius, Lithuania; 2Institute of Biomedical Sciences, Vilnius University, Faculty of Medicine, Vilnius, Lithuania

Background/ Objectives: Long QT syndrome (LQTS) is a well renowned monogenic channelopathy, which clinical phenotype widely varies and largely depends on genetic penetrance and expressivity of phenomena. Study aim was to evaluate clinical and genetic data in patients referred for LQTS genetic diagnosis.

Methods: We performed a retrospective analysis of the virtual gene panel of hereditary cardiovascular diseases from whole exome sequencing data of the patients evaluated for LQTS between the years of 2021 and 2022.

Results: Clinical and genetic data from 92 unrelated patients (55%, 51/92 of females) were analysed. The mean age at time of testing was 10 ± 4.7SD years. Pathogenic or likely pathogenic variants were identified in 19% (17/92) of those patients. A total of 16 variants of unknown significance were identified in the study (7 absent in gnomAD). The most implicated gene was KCNQ1, with 65% (11/17) of the patients affected. Variants in KCNH2 gene were found in 12% (2/17) of patients, while 24% (4/17) were distributed in other genes. Nearly a quarter of patients (24%, 4/17) had a first degree relative with clinical LQTS. Among the patients, mean Schwarz score and longest measured QTc interval were 3.4 ± 1SD and 497ms ± 32SD, respectively. More than half (59%, 10/17) were asymptomatic, diagnosed accidentally based on ECG findings and/or family history.

Conclusion: This study confirms the importance of genetic testing for LQTS patients showing 19% of them to benefit from genetic result that will guide their clinical management.

Grants:

Conflict of Interest: None declared

EP07.064 New variants identified in low evidence genes associated with pulmonary arterial hypertension development

Natalia Gallego 1;2;3, Lucía Miranda-Alcaraz1;2;3, Mónica Mora1;2;3, Alejandro Cruz-Utrilla4;5, Dra. María Jesús del Cerro6, Manuel López Meseguer7, Amparo Moya Bonora8, Nuria Ochoa4;5, alejandro parra1;2;3, Patricia Pascual Vinagre1;2;3, Mario Cazalla1;2;3, cristina silvan1;2;3, Pedro Arias1;2;3, julian nevado1;2;3, Pablo LAPUNZINA1;2;3, Pilar Escribano-Subias4;5, Jair Tenorio1;2;3

1Instituto de Genética Médica y Molecular (INGEMM), Instituto de Investigación del Hospital Universitario La Paz (IdiPaz), Hospital Universitario La Paz, Madrid, Spain; 2Centro de Investigación Biomédica de Enfermedades Raras en Red, Instituto de Salud Carlos III (CIBERER), Madrid, Spain; 3ERN-ITHACA, European Reference Network on Rare Malformations Syndromes, Intellectual and other Neuro-developmental Disorders (ENR-ITHACA), Paris, France; 4Unidad multidisciplinar de Hipertensión Pulmonar, Servicio de Cardiología, Hospital Universitario 12 de Octubre, Madrid, Spain; 5European Reference Network on rare lung diseases (ERN-LUNG), Frankfurt, Germany; 6Unidad de Hipertensión Pulmonar Pediátrica, Servicio de Cardiología Pediátrica, Hospital Universitario Ramón y Cajal, Madrid, Spain; 7Departamento de Pneumología, ERN-Lung, Hospital Universitario Vall d’Hebron, Barcelona, Spain; 8Unidad de Cardiología Pediátrica, Departamento de Pediatría, Hospital Universitario La Fe, Valencia, Spain

Background: Pulmonary arterial hypertension (PAH) is a rare vasculopathy with significant morbidity and mortality. Variants in at least 27 genes have putative evidence for PAH causality. According to the ClinGen classification, there are several genes with moderate, limited and disputed relationship with PAH that require further study to definitively associate them with PAH.

Material and methods: We performed a retrospective study on 530 patients from the Spanish registries of adult and pediatric patients with PAH (REHAP and REHIPED). In all of them, whole exome sequencing was performed and a variant prioritization algorithm was designed to prioritize variants present in the 14 genes with moderate, limited and disrupted evidence of association with PAH.

Results: We have found 60 variants of unknown significance in the 14 studied genes, i.e., the 11% of the patients showed variants in these genes. It should be noted the variants detected in three of these genes: NOTCH1 (15 variants), ABCC8 (12 variants) and NOTCH3 (11 variants).

Conclusion: We add genetic evidence to the relationship of some genes associated with PAH, specially, NOTCH1, NOTCH3 and ABCC8. It would be recommended that genetic testing includes genes with moderate, limited and disrupted evidence although caution must be taken in the interpretation of the variants identified in this genes. It would be necessary more studies in higher cohorts and functional assays to improve our knowledge about the association of this variants with the disease.

Grants: PI21/01593, FCHP unrestricted grant.

Conflict of Interest: None declared

EP07.065 New candidate genes associated with the development of pulmonary arterial hypertension.

Lucía Miranda-Alcaraz1, Natalia Gallego1;2;3, Alejandro Cruz-Utrilla4;5, Dra. María Jesús del Cerro6, Manuel López Meseguer7, Amparo Moya Bonora8, Nuria Ochoa4, alejandro parra1;2, Patricia Pascual Vinagre1;2, mario cazalla1;2, cristina silvan1;2, Pedro Arias1;2, julian nevado1;2;3, Pablo LAPUNZINA1;2;3, Pilar Escribano-Subias4;5, Jair Tenorio 1;2;3

1La Paz University Hospital, Institute of Medical and Molecular Genetics (INGEMM), Madrid, Spain; 2CIBERER, Centro de Investigación Biomédica en Red de Enfermedades, Instituto de Salud Carlos III, Madrid, Spain; 3ERN-ITHACA, European Reference Network on Rare Malformations Syndromes, Intellectual and other Neuro-developmental Disorders, Belgium, Belgium; 4Hospital Universitario 12 de Octubre, Unidad multidisciplinar de Hipertensión Pulmonar, Servicio de Cardiología, Spain; 5ERN-LUNG, European Reference Network on rare lung diseases (pulmonary hypertension), Brussels, Belgium; 6Hospital Universitario Ramón y Cajal, Unidad de Hipertensión Pulmonar Pediátrica, Madrid, Spain; 7Hospital Universitario Vall d’Hebron, Departamento de Pneumología, Barcelona, Spain; 8Hospital Universitario La Fe, Unidad de Cardiología Pediátrica, Departamento de Pediatría, Valencia, Spain

Pulmonary Arterial Hypertension (PAH) is an infrequent disorder characterized by increased blood pressure in the pulmonary arteries, leading to progressive heart failure and premature death if not treated. This study aims to select and validate variants in genes previously associated with PAH and investigate new candidate genes potentially involved in the development of PAH. A total of 55 clinically diagnosed PAH patients and 21 unaffected family members were analyzed by whole exome sequencing (WES). Variant prioritization was conducted through a bioinformatic custom algorithm. Pathogenic and variants of uncertain significance (VUS) were identified in 30.9% of genes previously linked to PAH. Additionally, three VUS variant were identified in the following four new candidate genes: ATF2, HDAC5, VASH1, and UACA. Variants in ATF2, VASH1, and UACA may influence the hyperproliferation and resistance to apoptosis of pulmonary vascular cells, contributing to PAH development. Furthermore, haploinsufficiency in the post-translational protein acetylation regulatory mechanisms involving the HDAC5 gene may also contribute to PAH development, resulting in an aberrant epigenetic signature that exacerbates the characteristic vascular remodeling process. In summary, we have identified several variants in four candidate genes that can be potentially involve in the pathogenesis of PAH through different molecular mechanisms, although further research is needed to confirm the potential role of these genes in the development of PAH.

Grants: PI21/01593, FCHP unrestricted grant.

Conflict of Interest: None declared

EP07.066 Exploring the genetic landscape of transthyretin hereditary amyloidosis in Colombia: Unveiling phenotypic variations and dominant genotypes.

Marcela Galvez 1, jose geles1, Sandra Bello1

1Gencell Genética Avanzada, Genetic Research Unit, Bogotá, Colombia

Background/Objectives: The worldwide prevalence of Transthyretin Hereditary Amyloidosis (hATTR), an autosomal dominant disease, is 1 in 450,000 individuals. Mortality presents 10-15 years post-diagnosis. Phenotypic heterogeneity leads to diagnostic challenges, especially in non-endemic countries. This study aims to fill the knowledge gap of hATTR molecular profile in Colombia.

Methods: Retrospective observational study involving Colombian patients with suspected hATTR. Sequencing of TTR gene was performed at Gencell, a Colombian genetic laboratory.

Results: Data was collected from 601 patients. TTR-variants were found in 68 cases. From these, sequencing was indicated because of clinical suspicion (39.71%) or due to a positive a family history (60.29%); 34 positive cases belonged to 16 families. Median age at diagnosis was 52.5 years (IQR = 35-69), and 52.94% were female. Most of the variants found in TTR gene were pathogenic (94.12%) and only a small proportion were likely pathogenic (2.94%) and of unknown significance (2.94%). Predominant variants included p.Val142Ile (76.47%) and p.Val50Met (17.65%). Interestingly, two cases were homozygous.

Eleven of the index patients had autonomic dysfunction and 14 of the familial cases presented symptoms of hATTR. Among the p.Val142Ile group, neurological or cardiological signs or a mixture of both were documented. For the p.Val50Met group only neurologic or cardiologic features were recognized.

Conclusion: The predominant causative variant in Colombia is p.Val142Ile, which differs from endemic and other Latin-American countries were p.Val50Met is more prevalent. Also, we found cases with mixed phenotypes, which differs from other populations were only neurological or cardiological features are present in a patient.

Conflict of Interest: None declared

EP08 Metabolic and Mitochondrial Disorders

EP08.001 A case of mucolipidosis II alpha/beta (ı-cell disease) presenting with new findings.

Neslihan Cinkara 1, hacer ukba kına1, aslı durmuş2, Mustafa Yilmaz3

1Ministry of Health of Turkey, Trabzon Training and Research Hospital, Department of Medical Genetics, Trabzon, Türkyie; 2Ministry of Health of Turkey, Trabzon Training and Research Hospital, Department of Pediatrics Metabolic Disorders, Trabzon, Türkyie; 3Karadeniz Technical University, Faculty of Medicine, Department of Medical Genetics, Trabzon, Türkyie

Background/Objectives: Mucolipidosis II alpha/beta (I-cell disease; OMIM # 252500) is a rare autosomal recessive lysosomal storage disorder. Mucolipidosis II alpha/beta (MLII) is characterized by dysmorphic features, cardiac involvement, respiratory symptoms, dysostosis multiplex, severe growth abnormalities, mental and motor developmental abnormalities. MLII is caused by homozygous or compound heterozygous mutations in the GNPTAB gene. We report a case of a female newborn with MLII who presented with novel clinical features, including sialic aciduria, sacral dimple, and secundum ASD.

Methods: After obtaining informed consent from the family, biochemical and molecular studies were initiated. Karyotype analysis was performed, followed by targeted next-generation sequencing (NGS) using the ROCHE kit and MGI DNBSEQ-400 device. The results were interpreted using the Genomize Seq program.

Results: Biochemical findings indicated the following: (1) Increased urinary excretion of glycosaminoglycans, oligosaccharides and sialic acid. (2) Normal tandem mass spectrometry assay. (3) The results from enzyme assays are still pending. The proband showed clinical features of MLII, such as recurrent pneumonia, thickened skin, feeding problems, and coarsened facial features. The karyotype of the patient was 46, XX. NGS identified a nonsense likely pathogenic homozygous variant in the GNPTAB gene (NM_024312.5: c.3061 C > T; p.(Gln1021Ter)).

Conclusion: Here, we discussed a patient who was diagnosed with MLII based on molecular assays, clinical and biochemical examination. To our knowledge, the patient’s new findings such as sialic aciduria, sacral dimple and secundum ASD has not been previously described in the literature. Our findings contribute to the global pool of data, providing practical insights into the manifestations of I-cell disease.

Grants: None.

Conflict of Interest: None declared

EP08.002 Identification of two rare biallelic variants in SDHA in a case with non-syndromic optic atrophy and complex II deficiency

Petra Liskova 1;2, Marketa Tesarova1, Hana Štufková1, Jana Křížová1, Tomas Honzik1, Martin Meliska1, Marie Vajter1;2, Hana Hansikova1

1First Faculty of Medicine and General University Hospital in Prague, Department of Paediatrics and Inherited Metabolic Disorders, Prague; 2First Faculty of Medicine and General University Hospital in Prague, Department of Ophthalmology

Background/Objectives: Isolated defects in the mitochondrial respiratory chain complex II (CII), known to be caused by variants in SDHA, SDHB, SDHD and SDHAF1, are extremely rare. The resulting metabolic disorders are highly variable. Herein we report a case with isolated optic atrophy and CII deficiency.

Methods: Exome sequencing was performed in the proband using SureSelect Human All Exon V6 Kit. Single nucleotide and copy number variation analysis was performed by Franklin Genoox. Detected variants were confirmed by Sanger sequencing. Mitochondrial function was assessed by measuring oxygen consumption in lymphocytes and fibroblasts using Oxygraph.

Results: The 11.5-year-old patient manifested with nystagmus at 1.5 years, subsequently bilateral optic atrophy was documented. No other pathology was found including brain MRI. Two rare variants in SDHA (NM_004168.4) were identified c.476C>T p.(Pro159Leu) and c.1016C>T p.(Ser339Phe), both predicted deleterious by Revel. Compound heterozygosity was confirmed by Sanger sequencing in an unaffected mother. No other rare variants in genes associated with optic neuropathy (Panelapp Version 4.0) evaluated as possibly pathogenic were identified. Respirometry analysis in permeabilised lymphocytes showed that CII-dependent respiration covered only 27% of total respiratory capacity. In fibroblasts, it was 30%, which is only half of the respiration observed in controls. This finding is consistent with CII deficiency.

Conclusion: We described two novel likely pathogenic variants in SDHA and defined their impact on oxidative phosphorylation system function. Isolated optic atrophy in association with SDHA is reported for the first time.

Grants: NU22-07-00614, NU22-04-00499

Conflict of Interest: None declared

EP08.004 Molecular and biochemical findings in a cohort of 55 Moldovan pediatric patients suspected for mitochondrial disorders

Secu Doina 1, Daniela Blanita1, Natalia Usurelu1, Victoria Sacară1

1Institute of Mother and Child, National Center of Reproductive Health and Medical Genetics, Chisinau, Moldova

Background/Objectives: Mitochondrial disorders (MD) constitute a group of clinically heterogeneous conditions characterized by genetic abnormalities affecting the mitochondrial respiratory chain. This study aims to elucidate the biochemical and molecular genetic spectrum within a pediatric patient cohort exhibiting a phenotype indicative of MD.

Methods: A cohort of 55 patients exhibiting symptoms and signs suggestive of MD underwent screening for 11 prevalent pathogenic mitochondrial DNA (mtDNA) point mutations by the quantitative PCR high-resolution melting (qPCR-HRM) analysis, followed by Sanger sequencing. Additionally, quantitative plasma amino acids and urinary organic acids assay were performed.

Results: Upon enrollment our patients scored as possible MD according to the Nijmegen MD scoring system. Within the cohort, 12 patients scored as possible MD, 23 patients as probable MD, and 20 patients as definite MD. All patients with definite MD had elevated serum lactate. Alanine was elevated in 13 patients with definite MD. Abnormal urinary organic acid analysis was present in 8 patients. Following the testing of patients using the qPCR-HRM technique, a total of 6 patients were identified with mtDNA pathogenic mutations (m.3243 A > G in 2, m.8993 T > G in 2, m.8344 A > G, m.11778 G > A). Through the analysis of the mitochondrial genome via the Sanger sequencing technique, it was possible to determine pathogenic and potentially pathogenic mutations associated with the clinical presentation in a total of 12 patients.

Conclusion: In the cohort of 55 patients referred for the evaluation of clinically suspected MD, pathogenic and potentially pathogenic variants associated with the patients’ phenotype were identified in 18 patients (33%).

Grants: No grants received.

Conflict of Interest: None declared

EP08.005 A novel pathogenic variant leading to Fabry disease and the impact of Gb3 substrate accumulation in circulating lymphoctyes

Luca Kamilla Li 1, Nóra Fekete2, Dalma Ruzsics1, Péter Reismann3, Éva Pállinger2, Gyorgy Fekete1, Árpád Ferenc Kovács1

1Semmelweis University, Pediatric Center, Tűzoltó street Department, Budapest, Hungary; 2Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary; 3Semmelweis University, Department of Internal Medicine and Oncology, Budapest, Hungary

Background/Objectives: Fabry disease is an X-linked lysosomal storage disease caused by pathogenic variations of the GLA gene resulting in the lack or dysfunction of alpha galactosidase (AGAL) enzyme. The dysfunctional AGAL leads to accumulation of globotriaosylceramide (Gb3) substrate in key organs. The aim of our study was to assess the clinical impact of a novel GLA gene variant, whether it leads to classical or atypical Fabry disease.

Methods: Major organ involvement was assessed according to the national Fabry diagnostic and follow-up guidelines. Substrate accumulation of the affected male child and his mother was determined extra- and intracellularly in the circulating immunophenotyped lymphocytes as measured by flow cytometer. Additional Fabry patients (N = 7) were enrolled for Gb3 measurements. Lyso-Gb3 levels were measured by external diagnostic laboratory.

Results: The molecular diagnosis of Fabry disease was confirmed based on low AGAL activity (child: 0.9 umol/L/h, mother: 0.6 mol/L/h), high lyso-Gb3 concentration (child: 113.70 ng/mL, mother: 19.70 ng/mL) and GLA:c.142G>T. Signs of cardiac, kidney and neurological involvement were present both in the affected male child and his mother. Gb3 showed the highest accumulation intracellularly in CD19 + B cells (57.31 ± 16.98%) and CD3 + CD4 + T cells (41.04 ± 17.10%).

Conclusion: We described a novel GLA pathogenic variant leading to classical Fabry phenotype. Our results highlight the impact of different Gb3 accumulation patterns in different lymphocyte subpopulations, which may contribute to the systemic chronic inflammation observable in Fabry disease.

Grants: Hungarian National Research, Development and Innovation Office—NKFIH, OTKA PD_21 138521 grant

Conflict of Interest: None declared

EP08.006 The first year results of newborn screening for metabolic diseases in Western Ukraine

Olena Bahniuk 1, Nadiia Yusechko1, Inna Bahniuk1, Anna Yevstratyeva1, Maria Malaschynska2, Nataliia Olhovich3, Halyna Makukh1

1Regional center of neonatal screening of Lviv Regional Clinical Perinatal Centre”. Lviv, Ukraine, Lviv, Ukraine; 2Regional Clinical Perinatal Centre Lviv, Ukraine, Lviv, Ukraine; 3National Specialized Children’s Hospital Okhmatdyt, Kyiv, Ukraine, Kyiv, Ukraine

Background/Objectives: The analysis of organic acids and acylcarnitines in a dried blood spot is an important part of diagnosing inherited metabolic disorders and is a gold standard of NBS program. In October 2022, expanded newborn screening for 21 rare diseases, including 16 metabolic disorders, was implemented in Ukraine. The efficiency of NBS program for inherited metabolic disorders in Lviv Regional center of newborn screening is analyzed.

Methods: NBS for metabolic diseases is performed in four regional centers by the liquid chromatography-tandem mass spectrometry (LC-MS/MS) (Waters Xevo TQD) in DBS using RECIPE ClinSpot® reagents. Next step is clinical and molecular-genetic diagnostics of NBS positive cases.

Results: The entire neonatal screening process is monitored and documented in the National electronic healthcare system (e-health). 75,000 newborns were screened, resulting the early diagnostic of 11 children with metabolic disorders. The average turnout time for NBS is 5.0 + 1.8 days; the first blood spot re-call rate is 1.6%; 9.6% cases were interpreted as positive after re-call. Accordingly to diagnostic data, 7 patients were diagnosed with Phenylketonuria, PKU; 2 patients with Medium-chain acyl-CoA Dehydrogenase Deficiency, MCAD: АCADM с.532A > C/985A > C and p.Lys329Glu homozygous. The case of Maple Syrup Urine Disease, MSUD: BCK p.Thr211Met homozygous and one case patient with a HMG-CoA lyase deficiency (no clinically significant mutations were identified) were detected.

Conclusions: The newborn screening is free of charge for all babies born in Ukraine.

11 patients (0.019% of screened population) were diagnosed with metabolic disorders and started early treatment. The re-call rate should be considered to decrease.

Conflict of Interest: None declared

EP08.008 D-2-Hydroxyglutaric Aciduria Type I: An Over Looked Cause of Non-Accidental Injury

Thamer Alghamdi 1, Bader AlQahtani2, Majid Al Eissa2;3;4, Abdul Rafiq Khan5, Majid Alfadhel1;3;6, Norah Alsaleh1;6

1King Abdullah Specialized Children’s Hospital (KASCH), Genetics and Precision Medicine Department, Riyadh, Saudi Arabia; 2King Abdullah Specialized Children’s Hospital (KASCH), Department of Pediatrics, Riyadh, Saudi Arabia; 3King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia; 4King AbdulAziz Medical City, National Family Safety Program, Riyadh, Saudi Arabia; 5King AbdulAziz Medical City, Department of Pathology and Lab Medicine, Biochemical Metabolic Lab, Riyadh, Saudi Arabia; 6King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia

Background/Objectives: D-2-hydroxyglutaric aciduria (D2HGA) is a rare neurometabolic disorder characterized by the accumulation of D-2-hydroxyglutaric acid (D-2-HGA) and has two phenotypes, type I and II, each contributing to a distinct clinical manifestations. This condition is caused by mutations in the D2HGDH gene (Type I) or the IDH2 gene (Type II), impacting the tricarboxylic acid (TCA) cycle and leading to neurological manifestations. The clinical features might include subdural hematomas and retinal hemorrhages. The disorder typically follows an autosomal recessive inheritance pattern.

Here we report a case who presented initially with non-accidental injury, then was found to have D-2-hydroxyglutaric aciduria type I.

Methods/Case presentation: An 8-month-old girl, previously healthy, presented to the emergency room with a generalized tonic-clonic seizure following minor head trauma. Further evaluation revealed acute subdural hematoma and retinal hemorrhages, raising suspicion of non-accidental trauma. Biochemical testing sent as part of investigations in suspected non-accidental injury.

Results: Biochemical testing revealed elevated urinary levels of 2-Hydroxyglutaric acid. Targeted molecular genetic testing confirmed a homozygous likely pathogenic variant in D2HGDH gene (c.1421C>T, p.Ala474Val). Consistent with D-2-hydroxyglutaric aciduria type I (OMIM #600721).

Conclusion: Metabolic disorders, such as D-2-hydroxyglutaric aciduria type I, can present with clinical features that mimic non-accidental trauma. In cases of suspected child abuse, a thorough metabolic evaluation, including biochemical and molecular testing, is crucial to avoid falsely accusing caregivers and to ensure accurate diagnosis and appropriate management.

Grants: No grants.

Conflict of Interest: None declared

EP08.009 Navigating CPT II deficiency: Insights from neonatal screening to uniparental disomy exploration

Zafer Yüksel 1, Steffen Kühner1, Nahid Nahavandi1, Roschan Salimi Dafsari2, Mojgan Drasdo1

1Bioscientia Institut für Medizinische Diagnostik GmbH, Bioscientia Human Genetics, Ingelheim, Germany; 2University Hospital Düsseldorf, Department of General Paediatrics, Neonatology and Paediatric Cardiology, Düsseldorf, Germany

Background/Objectives: Carnitine palmitoyltransferase II (CPT II) deficiency, a disorder in long-chain fatty-acid oxidation, manifests in three clinical forms: lethal neonatal, severe infantile hepatocardiomuscular, and myopathic. The myopathic form, the most prevalent lipid metabolism disorder in skeletal muscle, presents with mild symptoms from infancy to adulthood. Neonatal and infantile forms exhibit severe multisystemic effects, including liver failure, hypoketotic hypoglycemia, cardiomyopathy, and early mortality. A 15-day-old baby girl was referred for gene panel testing due to elevated C12, C14, C16, and C18 in the acylcarnitine profile during newborn screening.

Methods: We conducted a comprehensive gene panel test, covering genes relevant to the elevated acylcarnitine levels observed in newborn screening (e.g., CACT, CPT2, ETFA, ETFB, and ETFDH).

Results: The identification of a homozygous missense variant, c.1393G>A p.(Ala465Thr), in the CPT2 gene located at chromosome 1p32.3 prompted further investigation. Segregation analysis revealed the father as heterozygous, while the mother displayed a wild-type genotype. After ruling out a deletion in the maternal allele, suspicion of uniparental disomy (UPD) arose. To substantiate this hypothesis, Optical Genome Mapping (OGM) and Whole-Exome Sequencing (WES) Trio analyses were conducted.

Conclusion: Due to paternal uniparental heterodisomy and partial isodisomy of chromosome 1, a heterozygous variant segregates biallelically within the choline/carnitine acyltransferase domain of the CPT2 gene, resulting in reduced enzyme activity of carnitine palmitoyltransferase II.

Conflict of Interest: Zafer Yüksel Full-time employer of Bioscientia, Steffen Kühner Full-time employer of Bioscientia, Nahid Nahavandi Full-time employer of Bioscientia, Roschan Salimi Dafsari: None declared, Mojgan Drasdo Full-time employer of Bioscientia

EP08.010 Whole exome sequencing reveals risk-conferring variants in a family having early-onset obese children

Hazal Banu Olgun Celebioglu 1;2, Ayse Pinar Ozturk3, Şükran Poyrazoğlu3, Feyza Tuncer1

1Istanbul University Aziz Sancar Institute of Experimental Medicine, Department of Genetics, Istanbul, Türkyie; 2Istanbul University Graduate School of Health Sciences, Istanbul; 3Istanbul University Faculty of Medicine, Department of Paediatric Endocrinology, Istanbul, Türkyie

Background/Objectives: Obesity is an ongoing health problem, which affects millions of individuals all over the world. This global epidemic continues to appear more particularly in children and is dramatically on the rise in low- and middle-income countries. Children with high body mass indexes (BMI) are considered at increased risk for mortality caused by T2DM, heart disease, and various cancer types. This study aimed to detect underlying genetic risk factors in a family having three early-onset obese children.

Methods: A family and three offspring were recruited to be evaluated for obesity. In BMI calculations, national growth chart and WHO standards were utilized for children and adults, respectively. Whole exome sequencing (WES) was performed on the youngest (age-of-10) morbidly obese child, who turned obese at five years old, utilizing Illumina NextSeq550. Bioinformatic analyses were performed on Genomize SEQ platform with variant filtering at MAF < 1% for all normal populations. Sanger sequencing was applied in variant confirmation and family segregation.

Results: Index case had early-onset severe obesity (+3,65 SD) on admission with grade-3 hepatosteatosis. Physical and serological examinations showed no other abnormalities. His mother, father, and sister were obese (BMIs: 33, 32, and 33 respectively), while his brother was overweight (+1,5 SD). WES revealed deleterious novel variants in more than 10 obesity and lipidosis-related genes segregated in the family.

Conclusion: Obesity inheritance has been detected in this family, where all individuals have elevated BMIs. All members should be under periodic follow-up due to shared genetic variants and potential comorbidities.

Grants: By Scientific Research Projects Coordination Unit of Istanbul University (TDP-2020-37103).

Conflict of Interest: None declared

EP08.011 Mitochondrial pathogenic variants in WES data: from screening to confirmation and follow-up

Sebastian Skoczylas 1, Tomasz Płoszaj1, Magdalena Traczyk-Borszyńska1, Karolina Gadzalska1, Paulina Jakiel1, Agata Pastorczak2, Ewa Juścińska1, Monika Gorządek1, Maciej Borowiec1, Agnieszka Zmysłowska1

1Medical University of Lodz, Department of Clinical Genetics, Lodz, Poland; 2Medical University of Lodz, Department of Genetic Predisposition to Cancer, Lodz, Poland

Background/Objectives: Mitochondrial diseases caused by mitochondrial DNA (mtDNA) mutations are diagnostically challenging due to factors such as heteroplasmy, age-related variant loss and evolving phenotypes. In this study, we analyzed exome sequencing data from 419 patients. The primary phenotypic features observed in this study group included neurodevelopmental and neurological disorders, as well as renal dysfunction.

Methods: The study material was DNA isolated from peripheral blood leukocytes using the Maxwell automated system (Promega). Whole exome sequencing was performed using the Twist Human Core Exome Kit (Twist Bioscience). mtDNA-Server was applied for mtDNA analysis and we developed a custom Python pipeline for variant prioritization.

Results: The screening process identified 1,000 unique variants with heteroplasmy greater than 10%, excluding HVR regions. Four confirmed pathogenic variants were detected (m.1555A > G, m.3243A > G, m.9035T > C, and m.11778G > A). Expanding our criteria revealed seven potentially deleterious variants at low levels, confirmed by next-generation sequencing and segregation in families and excluding NUMTs origin. Finally, two variants were identified as likely pathogenic (m.7392G > A and m.15897G > A).

Conclusion: Our study highlights the complexity of diagnosing mitochondrial diseases caused by mtDNA mutations and demonstrates the need for a comprehensive approach to identifying variants. The results also reveal the need to accurately identify phenotypes in patients and not limit clinical analysis to the leading symptom, but to extend the patient’s phenotypic evaluation to accompanying symptoms. Finally, this integrated approach aims to accelerate diagnosis, contributing to improved patient outcomes and quality of life.

Grant: No

Conflict of Interest: None declared

EP08.012 An ongoing collaboration: The Association for Creatine Deficiencies and the Clinical Genome Resource work together to facilitate variant classification.

Jennifer Goldstein 1, Heidi Wallis2, Emily Reinhardt2, Juliann Savatt3, Erin Rooney Riggs3, Amanda Thomas-Wilson4, Emily Groopman5, Vimla Aggarwal6, Simona Bianconi7, Raquel Fernandez8, Kim Hart9, Emily Kyle8, Nicole Liang10, Nicola Longo11, Christine Preston12, Daniel Reich13, Meredith Weaver8, Sarah Young14, Saadet Mercimek-Andrews15

1University of North Carolina at Chapel Hill, Department of Genetics, Chapel Hill, United States; 2Association for Creatine Deficiencies, Carlsbad, CA, United States; 3Geisinger, Danville, PA, United States; 4New York Genome Center, New York, NY, United States; 5Children’s National Hospital, Washington, DC, United States; 6Columbia University Irving Medical Center, New York, NY, United States; 7Kaiser Permanente, San Diego, CA, United States; 8American College of Genetics and Genomics, Bethesda, MD, United States; 9Utah Public Health Laboratory, Salt Lake City, UT, United States; 10The Hospital for Sick Children (SickKids), Toronto, Canada; 11University of Utah, Salt Lake City, UT, United States; 12Stanford University, Palo Alto, CA, United States; 13ARUP Laboratories, Salt Lake City, UT, United States; 14Duke University School of Medicine, Durham, NC, United States; 15University of Alberta, Edmonton, Canada

Consortium: The Clinical Genome Resource (ClinGen)

Background/Objectives: Understanding the clinical significance of variants within the genes causing cerebral creatine deficiency syndromes (CCDS) is important to ensure timely diagnosis and initiation of treatment for these neurological disorders. The Association for Creatine Deficiencies (ACD), a patient advocacy organization, collaborates with the NIH-funded Clinical Genome resource (ClinGen) to share data and assist in robust variant classification.

Methods: Via collaboration with ClinGen’s GenomeConnect, ACD registry participants provide their genetic testing reports for submission to the public variant database, ClinVar. Data is also shared with the ClinGen CCDS Variant Curation Expert Panel (VCEP), which has developed guidelines for classification of variants in the genes causing CCDS and submits those classifications to ClinVar as part of an FDA-approved process.

Results: Using published data, the ClinGen CCDS VCEP has classified 43 variants from ACD registry participants; 15 were classified by the VCEP as pathogenic (8 SLC6A8, 7 GAMT), 7 likely pathogenic (LP) (5 SLC6A8, 2 GAMT), 2 benign (1 SLC6A8, 1 GAMT), and 19 were variants of uncertain significance (VUS) (15 SLC6A8, 4 GAMT). 12 VUS were identified as tending towards LP by the VCEP based on the strength of evidence already available; ACD participants with these variants were recontacted to request additional case-level data (biochemical testing, magnetic resonance spectroscopy, family history, genetic testing of parents) with the goal of reclassifying these variants as LP or pathogenic.

Conclusions: Collaboration between the ACD and ClinGen increases understanding of the clinical significance of variants in the gene causing CCDS.

Grants: NIH U24HG009650, U24HG00683

Conflict of Interest: Jennifer Goldstein Full-time employee paid by a grant funding ClinGen., Heidi Wallis: None declared, Emily Reinhardt: None declared, Juliann Savatt My position at Geisinger is funded via multiple NIH grants: U24HG006834 and R01 HG009671 as well as a Helmsley Foundation Well grant 2305-06052., Employed full time at Geisinger, a health system in Pennsylvania, USA, I received travel support to present at the Jackson Laboratory Human and Mammalian Genetics and Genomics: The McKusick Short Course in 2023., Erin Rooney Riggs I am a PI on the NHGRI-funded grant U24HG006834 which supports the work of ClinGen., I am a full-time employee of Geisinger., I am a paid consultant for Illumina, Inc., Amanda Thomas-Wilson: None declared, Emily Groopman: None declared, Vimla Aggarwal: None declared, Simona Bianconi Consultant to Ultragenyx Pharmaceutical 2019-2022, Raquel Fernandez: None declared, Kim Hart Employed full time by the State of Utah Dept Health and Human Services, Emily Kyle: None declared, Nicole Liang: None declared, Nicola Longo: None declared, Christine Preston: None declared, Daniel Reich Employee of ARUP Laboratories, Meredith Weaver ClinGen key investigator liaison, salary paid by ClinGen grant, ClinGen key investigator liaison, salary paid by ClinGen grant, Sarah Young: None declared, Saadet Mercimek-Andrews: None declared

EP08.013 Novel homozygous nonsense variant in SERAC1 causes MEGDEL syndrome

Yasmina Elaribi 1, Sana Karoui2, Imen REJEB1, Alaa Ziadi1, Zohra Fitouri3, Manel Lajimi1, Syrine Hizem1, Houweyda Jilani1, Lamia BenJemaa1

1Mongi Slim hospital, Genetic, Tunis, Tunisia; 2Mongi Slim Hospital, Tunis, Tunisia; 3Children’s hospital of Tunis, Tunis, Tunisia

Background/Objectives: MEGDEL syndrome, also referred to as 3-methylglutaconic aciduria type VI, is a rare and severe neurometabolic disorder characterized by 3-methylglutaconic aciduria, deafness, encephalopathy, and Leigh-like lesions on brain magnetic resonance imaging. Patients with this syndrome carry homozygous variants in SERAC1 gene, encoding a protein which is essential for phospholipid exchange.

Methods: Here, we report clinical data of a patient who was referred to our department for encephalopathy and hepatic cholestasis investigation. The proband underwent a whole-exome sequencing. Segregation analysis was done by Sanger sequencing.

Results: The patient is a 2-year-old boy, born to healthy consanguineous parents after a normal pregnancy and delivery. He has a brother with autism spectrum disorder. At the age of 6 months, he presented a digestive hemorrhage, cholestasis and hepatomegaly. Since the age of 8 months, he developed recurrent hypertonic episodes of the upper limbs. Clinical examination revealed microcephaly (-3DS), axial hypotonia, psychomotor delay and quadripyramidal syndrome with choreic movements. Laboratory investigations showed high lactate and ammoniac levels in blood. Whole-exome sequencing analysis displayed a novel homozygous nonsense likely pathogenic variant in the exon 13 of SERAC1 gene (NM_032861.4):c.1379G>A (p.(Trp460Ter)). This variant has never been reported in the literature. Our case allows us to broaden the genotypic spectrum of the MEGDEL syndrome.

Conclusion: The present study highlights the importance of whole-exome sequencing in etiological diagnostic of such heterogeneous mitochondrial disorders where phenotypes are overlapping.

Conflict of Interest: None declared

EP08.015 Adipose Tissue Transcriptomic Analysis Reveals PDCD5 as a Hub Gene for Insulin Resistance

guanjie chen 1, Ayo Doumatey1, jie zhou1, Lin Lei1, Charles Rotimi1, Adebowale Adeyemo1

1National Human Genome Research Institute (NHGRI), CRGGH, Bethesda, United States

Objective: Insulin resistance (IR) is a key event in the development of type 2 diabetes (T2D). Numerous genetic variants have been identified for IR. However, studies of molecular alterations in relevant tissues are limited. This study conducted transcriptomic analyses of subcutaneous adipose tissue for two IR traits: fasting insulin (FI) and HOMA-IR.

Methods and results: Study participants are 30 Nigerians from the Africa America Diabetes Mellitus (AADM) Study. Adipose tissue was obtained from individuals without T2D via percutaneous needle biopsy of the vastus lateralis. RNA-seq of subcutaneous adipose tissue was done on a NovaSeq6000. DGE analysis was done with a linear mixed model (LMM), incorporating sample-sample correlation from GWAS and with adjustment for PEER factors. Weighted gene co-expression network analysis (WGCNA) was done to further explore gene expression relationships and identify gene modules.

We identified 30 unique DEGs (p-value < 0.0001) for the two traits (including 27 genes associated with HOMA-IR and 26 genes with FI), with 23 genes common to the two traits. Three of thirteen modules identified by WGCNA were associated with both HOMA-IR and FI. Notably, PDCD5 was a DGE (HOMA-IR β -2.5618 (SE 0.4778), p = 4.27×10-5); FI β -2.6366(SE 0.4834), p = 3.52×10-5) and also a hub gene for a significantly-associated gene module.

Discussion: Our analysis identified significant DGE and gene modules for FI and IR. PDCD5, an apoptosis-related protein that is involved in positive regulation of gene expression, is upregulated in early IR and has been suggested as a novel target for improving IR. Our findings provide functional insights into IR in an underrepresented population.

Conflict of Interest: None declared

EP08.016 A high diagnostic rate of inherited metabolic disorders by whole exome sequencing

Parnian Alagha 1;2, Fatemeh Ahangari1, Maryam Beheshtian1, Negar Molaei1;2, shima Dehdahsi1, Mahsa Fadaie1, Mehri Ashki1, Zohreh Elahi1, Maryam Mozaffarpour1, Parishad Saei1, Fatemeh Fatehi1, Khadijeh Noudehi1, Shima Zamanian Najafabadi1, Kimia Kahrizi2, Ariana Kariminejad1, Hossein Najmabadi1;2

1Kariminejad - Najmabadi Pathology & Genetics Center, Tehran, Iran; 2Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran

Background/Objectives: Metabolic disorders, as a global health concern, result from mutations in genes involving in various metabolic pathways and manifest with wide clinical spectrum including cognition, neurological, and muscular phenotype. Although tandem mass spectrometry and other inborn screening tests are widely used for diagnosis, they are mainly considered as screening tests. Since their specificity is low, Next generation sequencing (NGS), known for its reliability and accuracy, is one of the gold standards for identifying causative genes in metabolic disorders. Here, we report our experience in detecting genetic variants in 432 patients with suspected metabolic disorders subjected to whole exome sequencing (WES).

Methods: 432 individuals, suspected of having metabolic disorders, were referred to our genetics laboratory to be subjected to WES, between 2013 and 2023. All promising variants were confirmed by Sanger sequencing.

Results: Our investigated samples comprised 181 females and 251 males with 70% parental consanguinity rate. Disease onset mainly ranged from neonatal to adulthood, with the most commonly reported symptoms of intellectual disability/developmental delay (30%) hypotonia (27%) and muscle weakness (26%), and A diagnostic yield of 50% was obtained with 76% homozygosity. Notably PLA2G6, ETFDH and GMPPB genes emerged as the most frequently reported.

Conclusion: In nearly half of the patients, we achieved to diagnose the underlying molecular cause which is in consistency with previous studies. Our results emphasize on the importance of precise clinical diagnosis coupled with effective genetic tests like WES for identifying genetic causes of metabolic disorders.

Conflict of Interest: None declared

EP08.017 Characterization of individuals with fatty acid β-oxidation abnormalities identified through newborn screening program in a Spanish tertiary referral hospital

amanda herranz cecilia1;2, Carmen Rodríguez Jiménez1;2;3, Ana Carazo1;2, natividad gallego onís3, María Esther Rubio Martín3, Maria Victoria Del Pozo-Gomez3, Juan Manuel Montejo4, laura García Fernández4, Carlos Rodríguez-Antolín5, Ana Bergua Martínez6, Jose David Andrade Guerrero6, Ana Morais López6, Sonia Rodriguez Novoa 1;1;2;2

1Hospital Universitario La Paz, Metabolic Disease Laboratory, Genetic Department, Madrid, Spain; 2IdiPAZ, Hospital Universitario La Paz, Group of Dislipemias of genetic origin and metabolic diseases, Madrid, Spain; 3Hospital Universitario La Paz, Next-generation sequencing section, Genetic Department, Madrid, Spain; 4Hospital Universitario La Paz, Preanalytical section, Genetic Department, Madrid, Spain; 5Hospital Universitario La Paz, Bioinformatics section, Genetic Department, Madrid, Spain; 6Hospital Universitario La Paz, Nutrition and Metabolic Diseases Department, Madrid, Spain

Background/Objectives: Fatty acids β-oxidation disorders (FAODs) are a heterogeneous group of inherited metabolic disorders that occur with an overall estimated rate of incidence of 1:7914 in Spain. More than 25 genetically distinct disorders have already been described.

Under metabolic stress situations such as fasting, individuals with FAODs can present acute, life-threatening metabolic decompensations. However, the implementation of certain FAODs newborns screening programs (NBS) based on acylcarnitines measurement has allowed to improve clinical management and to reduce the risk of severe decompensations.

The aim of the present study is to analyze the results of the NBS program for FAODs in a single-center of Spain in terms of diagnosis, monitoring and genotypic spectrum.

Methods: Retrospective review of 95 newborns referred to a reference clinical unit to assess NBS biochemical abnormalities suggestive of FAODs between 2015-2023.

Results: Increased levels of very long-chain acylcarnitines (VLCAD) was the main cause of referral (65%). Molecular diagnosis was reached in 18% the cases, being medium-chain acylcarnitines deficiency (MCADD) the most frequent disorder (9.47%) followed by primary carnitine deficiency (CTD, 7.36%) and VLCAD deficiency (VLCADD, 5.26%). Diagnostic yield varies widely among biochemical suspicion groups (8.47% in the increased VLCAD levels group vs 70% in the increased MCAD levels group).

After the implementation of preventive measures, the majority of infants remained asymptomatic over the years. Nevertheless, symptoms were more common in the VLCADD group.

Conclusion: Molecular studies are a powerful tool to address FAODs, since they play a key role in diagnostic confirmation and reproductive counselling.

Grants: CA22/00002

Conflict of Interest: None declared

EP08.018 A case report of PEPCK-C deficiency in childhood: Metabolic laboratory findings affected by sudden acute liver failure

Andžela Lazdāne 1, Svetlana Vorslova1, Zanda Daneberga1;2, Ieva Grīnfelde1;2, Madara Auzenbaha1;2, Ieva Pukite1;2

1Children’s Clinical University Hospital, Riga, Latvia; 2Riga Stradins University, Riga, Latvia

Background/Objectives: Cytosolic phosphoenolpyruvate carboxykinase deficiency (PEPCK-C) is a rare autosomal recessive disorder caused by defect of PEPCK1 enzyme involved in metabolic pathway of gluconeogenesis. Clinically broad variability from severe neonatal hypoglycaemia, lactic acidosis, hepatomegaly and ketonuria to late-onset milder forms with mild seizures and liver disfunction, apnoea is observed.

Methods: We report a case of late-onset PEPCK-C deficiency where in an acute state abnormal and unusual metabolite pattern using GC-MS, UPLC-UV and TLC was observed.

Results: An 8-year-old girl with sudden, acute, unspecified liver failure and renal insufficiency after increased body temperature and prolonged vomiting was hospitalized. Some days before illness she ate enormous amount of sugar which might be a reason for grossly elevated sucrose level in carbohydrate spectra. Organic acid profile showed elevated adipic acid (about three times above the upper reference interval) and hydroxydicarboxylic aciduria with excretion of 3-hydroxyadipic lactone, 2-hydroxyadipic, 3-hydroxyadipic and 3-hydroxysebacic acids. Increased level of lactate, pyruvic acid, 3-methylglutaconic acid and abnormal amount of tricarboxylic acid cycle metabolites such as fumarate, malate, α-ketoglutarate were observed. Amino acid spectra showed vast glutamine level in plasma (3217.1 µM/L, normal <709.0 µM/L) and urine (2903.5 mmol/mol creatinine, normal <137.0 mmol/mol creatinine). None of amino acid concentrations showed decreased level, whereas almost all were detected as elevated.

Conclusion: The presentation of first PEPCK-C deficiency symptoms may occur in childhood, furthermore characteristic laboratory findings are not always unambiguous. Wide variability of metabolite abnormalities during metabolic crisis of cytosolic phosphoenolpyruvate carboxykinase deficiency can be observe.

Conflict of Interest: None declared

EP08.019 Case report: A deep intronic variant in PHEX in a family with hypophosphatemia

Karl Hackmann 1, Joseph Porrmann1, Sophie Henke2, Friederike Quitter2, Franziska Lange2, Evelin Schröck1;3, Doreen William1, Andreas Dahl4, Min Ae Lee-Kirsch5;6, Angela Hübner2, Katrin Koehler2

1Universitätsklinikum Dresden: Institut für Klinische Genetik, Dresden, Germany; 2Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes, Dresden, Germany; 3MPI-CBG, Dresden, Germany; 4DRESDEN-concept Genome Center, Dresden, Germany; 5Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Dresden; 6University Center for Rare Diseases, Faculty of Medicine and University Hospital Carl Gustav Carus, Dresden, Germany

Persistent hypophosphatemia may be acquired due to malnutrition, intestinal malabsorption, hyperparathyroidism, vitamin D deficiency, excessive alcohol consumption, some drugs, or organ transplantation. However, it may also have an underlying genetic cause due to mutations in several genes (CLCN5, CYP27B1, DMP1, FAM20C, FGFR1, FGF23, GNAS, PHEX, SLC34A3, and VDR).

Here we present a case of familial hypophosphatemic rickets diagnosed in five females and three males over four generations suggesting an autosomal dominant inheritance. Previous attempts to identify the causative variant were unsuccessful. Finally, genome sequencing revealed a rare and deep intronic variant (NM_000444.6:c.1646-1982T > G) in intron 15 (~22kb in size) in the PHEX (phosphate regulating endopeptidase X-linked) gene that segregated with the disease. SpliceAI, a deep learning based splice variant identification tool, predicted an increased likelihood of alternative splicing due to this variant with the creation of a new splice acceptor site. Using reverse transcription of the mRNA followed by PCR and sequencing, we show that the new splice site leads to an alternative splicing event, confirming the pathogenic character of this intronic variant

Pathogenic variants in PHEX cause X-linked hypophosphatemic rickets (XLHR; OMIM307800) with an unusually even sex distribution, mimicking autosomal dominant inheritance. This finding allows for evidence-based treatment of hypophosphatemia with burosumab in adults and adolescents in this family.

Conflict of Interest: None declared

EP08.020 A case of NBAS validated by Whole Genome Sequencing.

Frank Sleutels 1, S. Demirdas1, Esmee Oussoren2, Lies Hoefsloot1

1Erasmus MC, University Medical Center Rotterdam, Department of Clinical Genetics, Rotterdam, Netherlands; 2Erasmus University Medical Center, Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Rotterdam, Netherlands

Background/Objectives: We present a case as an example of how technological progress within the laboratory in combination with advances in understanding the genetic causes behind (metabolic) diseases will facilitate and accelerate future genetic diagnoses using Whole Genome Sequencing (WGS).

Methods/Results: The patient presented with severe cholestasis and acute liver failure (ALF) at the age of one year, receiving a liver transplantation at the age of two years. The genetic diagnosis followed fifteen years later after extensive genetic testing including Whole Exome Sequencing (WES) and the identification of the NBAS gene as a cause of autosomal recessive ‘Infantile liver failure syndrome 2“ by Haack et al, Am J Hum Genet. 2015. WES revealed compound heterozygosity of two previously undescribed mutations within the NBAS locus. A nonsense mutation (Single Nucleotide Variant; SNV) and a 3 kb duplication (Copy Number Variant; CNV). The challenges to detect compound heterozygosity of an SNV with a small CNV delayed the genetic diagnosis. To overcome these and other challenges, our laboratory has validated WGS for routine diagnostics for which this case was part of the validation cohort.

Conclusion: We implemented WGS with concurrent SNV and CNV detection which resolves many of our challenges to accurately and swiftly provide genetic diagnosis for many patients including the one described here.

Conflict of Interest: None declared

EP08.022 Cerebrotendinous xanthomatosis: a treatable hereditary cholesterol metabolism disorder

Mustafa Yilmaz1, AYBERK TURKYILMAZ1, KUBRA ADANUR SAGLAM1, nazime cebi2, Alperhan Cebi 1

1Karadeniz Technical University, Türkyie; 2Trabzon Legal Training and Research Hospital, Numune Campus, Türkyie

Background/Objectives: Cerebrotendinous xanthomatosis (CTX) is a rare autosomal ressesive disorder of cholesterol metabolism and characterized by abnormal deposition of cholestanol in tissues such as brain, spinal cord, eye and tendons. In this study, we aimed report clinical and genetic characteristics of 5 CTX patients.

Methods: All patients were evaluated with family history, neurological examination, brain magnetic resonance imaging (MRI) and electromyography. All cases were investigated with the hereditary ataxia gene panel using the next generation sequencing method.

Results: The median age of the patients at symptom onset was 21 years, and the median age at diagnosis was 35 years. In 4 out of 5 families, the parents were consanguineous. In terms of clinical findings, cataract was present in 3 (60%), polyneuropathy in 2 (40%), developmental delay/intellectual disability in all (100%), seizure in 2 (40%), osteoporosis in 1 (20%), and ataxia in 4 (80%). In brain MRI analysis, cerebral and cerebellar atrophy, hyperintensity in dentate nucleus were reported. Three different homozygous variants were detected in CYP27A1 gene (NM_000784.4: c.1476+2T > C, c.1_5delATGGC (p.Met1fs*178), c.646G>C (p.Ala216Pro) in these patients. The frameshift (c.1_5delATGGC, p.M1fs*178) variant revealed in 2 different cases was the novel variant. Colestanol levels of two patients are 38.1 ug/ml and 41.79 ug/ml.

Conclusions: Timely detection and treatment are key to prevent severe morbidity and mortality in CTX patients. Early initiation of oral chenodeoxy-colic-acid therapy is the treatment choice for neurological and non-neurological symptoms. The addition of the CYP27A1 gene to the hereditary ataxia, spastic paraplegia and cataract gene panels will contribute to the early diagnosis of CTX patients.

Conflict of Interest: None declared

EP08.023 Plasma microRNAs as biomarkers of cerebellar atrophy caused by phosphomannomutase deficiency (PMM2-CDG)

Florencia Epifani 1, Lluc Cabus2, Gregorio Nolasco1, Mercè Bolasell1, Adrián Alcalá San Martín1, Patricia Fernández1, Gisela Márquez1, Cristina Hernando-Davalillo1, Jennifer Pérez2, Esther Lizano2, Silvia Carbonell-Sala3, Joao Curado2, sonia belmonte2, Mercedes Serrano1;4

1Hospital Sant Joan de Déu, Barcelona, Spain; 2Flomics Biotech, Barcelona, Spain; 3Centre de Regulació Genòmica CRG, Barcelona, Spain; 4U703 CIBERER, Barcelona, Spain

Consortium: CDG Spanish Consortium

Background/Objectives: The most frequent congenital disorder of glycosylation is PMM2-CDG, caused by mutations in PMM2. Patients develop a cerebellar syndrome with ataxia, oculomotor disorders, and cognitive impairment, causing long-term disability, but also extraneurological symptoms. Despite early symptomatic, PMM2-CDG is normally diagnosed during childhood when the cerebellar atrophy is unstoppable and irreversible. Importantly, different strategies of treatment are under development. There is no known genotype-phenotype correlation or reliable biomarkers of evolution. The goal of this study is to find a non-invasive plasma-based miRNA PMM2-CDG signature and test its reliability as a biomarker.

Methods: Plasma was isolated from 32 PMM2-CDG patients and 76 controls and divided in two cohorts: a training cohort, and a validation cohort. The concentration of different miRNAs in plasma was assessed using NGS and the results were then mapped against the known miRNome. Using machine learning models two different signatures were built with miRNAs combinations.

Results: The signature obtained showed excellent performance in detecting PMM2-CDG, with a ROC higher than 0.9 in both the training and test cohorts. Moreover several genes present in the final signature were brain or cerebellum-specific, linking the dysregulated genes to the diseased tissue.

Conclusion: This is the first study proposing the detection of plasma miRNAs as a non-invasive method for the early detection of PMM2-CDG. If miRNA are useful biomarkers not only in the diagnosis but also in the monitoring and prognosis of PMM2-CDG deserves further studies.

Grants: This work was supported by FIS PI14/00021 and FI22/00218 from the national plan on I + D + I

Conflict of Interest: None declared

EP08.024 A novel deep intronic variant in CPT2 gene may be frequent in CPT2 deficient Arab patients.

Laura Gort 1;2;3, José Manuel Gonzalez de Aledo1, Abraham J. Paredes-Fuentes1, Ana Argudo-Ramirez1, Laura Pacheco1, Rosa Maria López-Galera1;2;3, José Antonio Arranz4, Clara Carnicer4, Ana Felipe4, Mireia Del Toro4, Antonia Ribes2;3, Judit Garcia-Villoria1;2;3

1Section of Inborn Errors of Metabolism - IBC. Department of Biochemical and Molecular Genetics. Hospital Clínic de Barcelona., Barcelona; 2August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona; 3Center for Biomedical Network Research on Rare Diseases (CIBERER), Madrid, Spain; 4Unit of Metabolic Diseases, Hospital Vall D’Hebrón, Barcelona, Spain

Background/Objectives: Carnitine palmitoyltransferase II (CPT2) and carnitine/acylcarnitine translocase (CACT) are enzymes involved in mitochondrial fatty acid beta-oxidation. Many Newborn Screening Programs (NBSPs) include the detection of both deficiencies. However, they exhibit the same biochemical profile, and a molecular test is necessary to achieve a definitive diagnosis.

The aim of this study was to diagnose a newborn from an Arab consanguineous couple, in whom CPT2 or CACT deficiency was suspected by the NBSP.

Methods: Whole Exome Sequencing (WES) revealed no variants. A skin biopsy was requested to perform cDNA analysis of the CPT2 and SLC25A20 genes. The cultured fibroblasts were incubated with cycloheximide to identify aberrant transcripts degraded by non-mediated decay system.

Results: CPT2 gene cDNA analysis revealed a homozygous 111bp insertion between exons 3 and 4 that corresponded to an intron 3 fragment (c.340_341ins111). Further analyses allowed the identification in homozygosity of the deep intronic change c.341-1694C > G as the cause of the exonization of this intronic fragment. This 111pb insertion is predicted to cause 38 amino acids insertion (p.Gly114delins38) in the protein. Parents were confirmed to be carriers of the intronic variant.

Afterwards, this intronic variant was also found in heterozigosity with a pathogenic variant in another Arab CPT2 deficient patient.

Conclusion: Here we demonstrate the eficacy of conducting cDNA studies in cases with inconclusive WES but with a clear biochemical profile. We report a new deep intronic variant in CPT2 gene. We recommend testing it when no variants are identified in this gene, particularly in Arab patients.

Conflict of Interest: None declared

EP08.025 Extreme clinical heterogeneity in a family with pyruvate kinase deficiency

Houyem Ouragini 1, Faten Fatnassi1, Ilhem BenFraj2, Dorra Chaouachi1, Monia Ouederni2, Samia Menif1

1Pasteur Institute of Tunis, Laboratory of Molecular and Cellular Hematology, Tunis-Belvedere, Tunisia; 2National Center of Bone Marrow Transplant, Department of Pediatrics- Immunohematology and Stem Cell Transplantation, Tunis-Bab Saadoun, Tunisia

Background/Objectives: Red cell pyruvate kinase (PK) deficiency is a rare congenital, nonspherocytic hemolytic anemia caused by a glycolytic defect due to mutations in the PKLR gene. PK catalyzes the conversion of phosphoenolpyruvate to pyruvate, creating red cell ATP. PK deficiency leads to a reduction in ATP and red cell lifespan. This recessive pathology is characterized by clinical heterogeneity resulting in a variable degree of hemolysis, significant morbidity and reduced health-related quality of life. Here, we report a consanguineous family with a child clinically affected by PK deficiency.

Methods: All members of the family underwent a complete blood count (CBC), PK assay and molecular analysis.

Results: The patient is a 13 years old boy who developed neonatal anemia. He presented with growth delay and receives transfusions every two weeks. His Hemoglobin was 7.8g/dL and the level of PK was reduced (2.45U/gHb). His parents and his brother are entirely healthy. Their hematological parameters were normal, whereas their PK activities were diminished. The patient was homozygous for the c.1010delG mutation, and his parents and his brother were heterozygous. This deletion results in a premature termination. The heterozygous status of this mutation does not explain the reduced level of PK in the healthy members of the family. Moreover, they showed a normal CBC. This suggests the existence of compensatory mechanisms.

Conclusion: This study highlights the complexity of establishing a phenotype/genotype correlation in a hereditary disease known as monogenic.

Conflict of Interest: None declared

EP08.026 Impact of mitochondrial disease in England: linking genetic diagnoses with routinely collected healthcare data on a national scale

Katherine Schon 1, Peter Stilwell2, Jeanette Aston2, Katrina Lavelle2, Margreet Luchtenborg2, Robert Pitceathly3, Michael Hanna3, Carl Fratter4, Rita Horvath5, Mary Bythell2, Steven Hardy2, Patrick Chinnery5

1University of Cambridge, Medical Genetics; 2NHS England; 3University College London, Neuromuscular Diseases; 4Oxford Genomic Laboratory Hub, Rare Mitochondrial Disease Service; 5University of Cambridge, Clinical Neurosciences

Background/Objectives: Mitochondrial disorders affect ~1 in 5000 people, but our understanding of their impact in the population is limited. We aimed to determine the lifespan, causes of death and risk of developing cancer in people with mitochondrial disorders in a population setting (English population, 60.2 million people).

Methods: Individuals with genetically confirmed mitochondrial disorders were identified by the NHS genetics laboratories and Genomics England. A curated registry was made within the National Congenital Anomaly and Rare Disease Registration Service. We performed individual-level linkage to nationally held data assets including vital status, death certification and cancer registration using unique NHS numbers. Kaplan Meier survival analysis and standardised cancer incidence ratios were calculated.

Results: 1134 individuals were included in the study (543 males, 591 females, median age 47 years, range 1 to 94). 92 were deceased. 15% were aged under 18 years or died during childhood. 73% had a mitochondrial DNA disorder and 27% had a nuclear mitochondrial disorder. Most children died due to the underlying mitochondrial disorder. Common causes of death in adults included cardiac deaths, cancers, and pneumonia. Survival varied significantly by genetic diagnosis. Median survival was 75 years in people with m.3243A > G and 80 years in Leber Hereditary Optic Neuropathy. Standardised cancer incidence ratios did not show a difference in cancer incidence compared to the general population.

Conclusion: There was no evidence of increased cancer risk. Median survival was longer than expected which has implications for counselling patients about prognosis and for planning healthcare services.

Grants: MRC ICGNMD MR/S005021/1

Conflict of Interest: None declared

EP08.027 A second case of Mitochondrial RNA Helicase SUPV3L1-Associated Neurodegenerative Disease: Ataxia, Spasticity, Optic Atrophy, and Skin Hypopigmentation (ASOASH).

Polina Tsygankova 1, Denis Chistol1, Tatiana Krylova1, Tatiana Markova2, Elena Dadali2, Ekaterina Zakharova1

1Research Centre for Medical Genetics, laboratory of inherited metabolic diseases, Moscow, Russian Federation; 2Research Centre for Medical Genetics, Moscow

Background: The mitochondrial degradosome complex (mtEXO) comprises polyribonucleotide nucleotidyltransferase 1 (PNPT1 or PNPase) and ATP-dependent RNA helicase (SUPV3L1 or SUV3). Previous reports by Van Esveld et al. (2022) identified a nonsense homozygous variant in the SUPV3L1 gene in siblings with mitochondrial disease. Our study presents the second documented case of SUPV3L1 pathology in humans.

Methods: Whole-genome sequencing was conducted on the DNBSEQ-400 platform using pair-end reading. Data analysis utilized an in-house developed pipeline and the NGSDATA-genome interface.

Results: The 17-year-old female, exhibited a diverse array of symptoms, including ataxia, spastic paraparesis, cognitive deficit, optic atrophy, and horizontal gaze-evoked nystagmus. Early onset of symptoms, such as ataxic gait and nystagmus, was noted, with subsequent progression of neurological manifestations. At the moment of the observation proband has vast regions of hypopigmented skin patches on the body and extremities which progresses in time. Whole-genome sequencing revealed compound heterozygous variants in the SUPV3L1 gene (NM_003171.5:c.272-2A > G (LP); c.1924A>C, p.(Ser642Arg) (VUS)). RNA analysis demonstrated splicing changes attributable to the c.272-2A > G transition. High-resolution respirometry showed low routine respiration but normal (borderline) respiration rates ratios using intact cells protocol.

Conclusion: This case underscores the phenotypic diversity associated with SUPV3L1 mutations, emphasizing the importance of considering mitochondrial RNA helicase dysfunction in the differential diagnosis of neurodegenerative disorders. Further elucidation of the molecular mechanisms underlying SUPV3L1-associated pathology may provide valuable insights into targeted therapeutic interventions.

Conflict of Interest: None declared

EP08.028 Clinically Suspected and Molecularly Diagnosed Mucoploysaccharidoses: Referred as Autism

Elif EROĞLU ŞİMŞEK 1, Ibrahim Akalin1, Murat Coskun2, Ayse Busra Akalin3, İlknur Sunar1, Nursel Elcioglu4

1Metagentech Center for Genetic Diseases, Istanbul, Türkyie; 2Istanbul University Faculty of Medicine, Child and Adolescent Psychiatry, Istanbul, Türkyie; 3Bahcesehir University, Faculty of Medicine, Istanbul, Türkyie; 4Marmara University Faculty of Medicine, Pediatric Genetics, Istanbul, Türkyie

Background/Objectives: Mucopolysaccharidosis type 3A (MPS 3A), also known as Sanfilippo syndrome, is a rare genetic disease. MPS 3A arises from the improper breakdown of glycosaminoglycans (GAGs) and typically manifests symptoms during childhood. Key symptoms include mental retardation, hyperactivity, speech impairments, vision and hearing loss, skeletal abnormalities, and reduced motor skills.

Methods: A 4-year-old male patient who is not able to talk was referred to genetic testing with suspicion of autism. He had plump cheeks, prominent eyebrows, distinct philtrum; mucopolysaccharidosis was suspected immediately at first glance during physical examination at genetic center. Enlarged costae was detected on the X-ray graph. A whole exome sequencing analysis was performed for molecular diagnoses.

Results: WES analysis revealed a pathogenic c.1298G>A (p.Arg433Gln) homozygous mutation in sulfamidase (SGSH) gene, which is related with autosomal recessive MPS 3A (Sanfilippo) Syndrome.

Conclusion: A homozygous mutation in SGSH gene causing MPS 3A which is one of the lysosomal storage diseases was detected with WES and in silico analysis. As in mental retardation and autism spectrum disorder (ASD), Sanfilippo syndrome causes progressive nervous system damage that leads to clinical findings such as decline in speech skills, hearing loss, behavioral and sleep disorders. This is why a multidisciplinary approach should be embraced and mucopolysaccharidosis should be considered when examining children with ASD symptoms.

Grants:

Conflict of Interest: None declared

EP08.029 investigations reveal new insights into ISCA2 variants: update on multiple mitochondrial dysfunctions syndrome 4

Mazhor Aldosary 1, Zuhair Alhassnan2, Dilek Colak3, Mohammad Al Muhaizea4, Namik Kaya1

1Centre for Genomic Medicine, Translational Genomics Departmen, Riyadh, Saudi Arabia; 2Centre for Genomic Medicine, Medical Genomics Department, Riyadh, Saudi Arabia; 3King Faisal Specialist Hospital and Research Centre, Molecular Oncology Department, Riyadh, Saudi Arabia; 4King Faisal Specialist Hospital and Research Centre, Neuroscience Center, Riyadh, Saudi Arabia

Background/Objectives: Iron Sulfur Clusters (ISC or Fe-S clusters) are conserved cofactors of iron and sulfur associated to the cysteine sulfurs of proteins [1]. ISCA2, one of the essential ISC assembly members, is important for multiple versatile cellular mechanisms such as DNA replication, RNA transcription, oxygen sensitization, and electron transport [2, 3].

Methods: Autozygome analysis and whole exome sequencing to identify the genetic cause of the disease. We utilized Sanger sequencing for variant confirmation. Multiple sequence aignment, protein modeling, qPCR, RT-PCR, dipstick assay, and mitochondrial respiration measurements for further characterization.

Results: We identified a Saudi patient with a novel and pathogenic transition variant (NM_194279.3:c.70A>G:p.Arg24Gly) in ISCA2. RTPCR on total RNA revealed a faulty splicing due to the transition. The splicing error caused a retention of DNA from the intronic region resulting in larger transcripts as compared to control transcripts. This novel mutation caused increased levels of mtDNA copy numbers and complex I activity. Moreover, assessment of the mitochondrial respiration revealed a decrease in the mitochondrial respiration parameters in the patients’ cells as compared to those of the control.

Conclusion: We reported the first Saudi case having a different variant. This study expands the phenotypic and genotypic spectrum of MMDS4 even in a highly consanguineous population in which a founder variant is frequently encountered in medical genetics clinics considering that the disease being a rare disorder.

References:

1.Lill, R., et al., 2012. 1823(9): p. 1491-508.

2.Ajit Bolar, N., et al., 2013. 22(13): p. 2590-602.

3.Al-Hassnan, Z.N., et al., 2015. 52(3): p. 186-94.

Grants: KSCDR/RAC#2180004

Conflict of Interest: None declared

EP08.030 Other possible genetic causes of low ceruloplasmin levels in patients with clinically suspected Wilson’s disease

Agnese Zarina 1;2;3, Agrita Puzuka1;2, Madara Auzenbaha1;2, Ieva Tolmane4;5, Zita Krumina6, Linda Gailite1, Dmitrijs Rots1

1Rīga Stradiņš University, Scientific Laboratory of Molecular Genetics, Riga, Latvia; 21Rīga Stradiņš University, Department of Biology and Microbiology, Riga, Latvia; 3, Riga, Latvia; 4Riga East Clinical University Hospital, Stationary “Infectology Center of Latvia”, Riga, Latvia; 5University of Latvia, Faculty of Medicine, Riga, Latvia; 6Rīga Stradiņš University, Department of Biology and Microbiology, Riga, Latvia

Background/Objectives: Wilson’s disease (WD) is a rare genetic disorder of impaired copper metabolism due to pathogenic variants in the ATP7B. The diagnosis is based on the Leipzig criteria, clinical signs, laboratory features and genetic testing. This study aims to investigate additional genetic explanations for WD symptoms and abnormal laboratory tests in patients without a confirmed ATP7B genotype.

Methods: Ten patients with clinically confirmed WD and one or none pathogenic ATP7B gene variant underwent whole-genome sequencing. The ATP7B was analyzed for coding and non-coding and structural variants and, if no candidate variant was identified, coding variants in “Mendeliome” gene panel (n = 4296) was analyzed for an alternative molecular cause.

Results: Two patients had the most common pathogenic variant in the ATP7B, NM_000053.3:c.3207C>A p.(His1069Gln), in a heterozygous state, one of which carried also VUS in CP – NM_000096.4:c.1209-3C > A (PM2; PP3). Two patients were carriers of likely pathogenic variants in the CP gene: NM_000096.4:c.2527_2528del p.(Ser843PhefsTer10) (PVS1; PM2) and NM_000096.4:c.2426-1G > C (PVS1; PM2). Three patients with CP gene variants had lower ceruloplasmin levels (0.0993 ± 0.0269 mg/dl) than those without the variants (0.1567 ± 0.0172 mg/dl). Six individuals had no ATP7B variants and no additional candidate variants identified using WGS.

Conclusion. Low ceruloplasmin levels, a diagnostic criterion for WD, can be caused by various factors, including CP gene variants in a heterozygous state. It highlights the importance of reviewing diagnostic criteria for WD and shows that CP gene variants can mislead the WD diagnosis.

Conflict of Interest: None declared

EP08.031 Single mitochondrial DNA deletions in the NGS era

Ligia Almeida 1, Catarina Pereira1, Mariana Ferreira1, Lia Abbasi Moheb1, Jorge Pinto Basto1, Omid Paknia1, Peter Bauer1

1CENTOGENE GmbH, Rostock, Germany

Background/Objectives: Single large-scale mtDNA deletions manifest as overlapping clinical phenotypes: Kearns-Sayre syndrome (KSS), Pearson syndrome (PS), chronic progressive external ophthalmoplegia (CPEO). They vary in size and can be identified in DNA from blood, buccal cells, and urine in affected children, and skeletal muscle tissue in affected adults. The present work shows our experience in a clinical diagnostic setup highlighting the value of the NGS in the diagnosis of mitochondrial disease, namely mtDNA deletions.

Methods: Patients for whom a targeted mitochondrial panel or exome sequencing was performed in the last 2 years were included. Reasons for referral included: seizures, stroke, CPEO, lactic acidosis. DNA was extracted from blood and NGS was performed.

Results: In 15 cases a single large-scale mtDNA deletion was detected with heteroplasmy levels ranging from 21% to 81%. Age at diagnosis varied from eight months, youngest patient diagnosed with PS, to a 23 year-old patient with KSS spectrum. Considering that in the same group of patients, we identified in 67 cases, pathogenic/likely pathogenic mtDNA variants; hence, single large-scale mtDNA deletions represent 18% of mtDNA disease-causing variations.

Conclusion: Although MDs are increasingly recognized, their diagnosis remains a challenge due to their genetic and clinical heterogeneity as well as complexity of mitochondrial genomics and often limited coverage of quantitative mtDNA changes in standard NGS assays and related bioinformatic pipelines. However, NGS technologies allows for the identification of single large-scale mtDNA deletions in a single platform and workflow, providing highly accurate and cost-effective diagnostic tests as our data demonstrate.

Conflict of Interest: None declared

EP08.032 Chanarin-Dorfman syndrome case in Lithuania

Inga Poceviciene 1, Inga Nasvytienė1, Rasa Ugenskiene1;2

1Hospital of Lithuanian University of Health Sciences Kauno klinikos, Department of Genetics and Molecular Medicine, Kaunas, Lithuania; 2Lithuanian University of Health Sciences, Department of Genetics and Molecular Medicine, Kaunas

Background/Objectives: Chanarin-Dorfman syndrome (CDS) is a very rare autosomal recessive nonlysosomal inborn error of neutral lipid metabolism. It is characterized by nonbullous erythrodermic form of ichthyosis, with variable involvement of other organs, such as liver, central nervous system, eyes, and ears.

Methods: 56 years old woman was reffered to geneticist because of suspicion of Ichthyosis congenita. From 5 years of age she had ichthyosis, from 10 years of age she had bilateral hearing impairment. She had a hematologist consultation several years ago for anemia: intracytoplasmic vacuoles were observed within peripheral blood neutrophils. Four months ago patient suffered an ischemic stroke, which left hypoesthesia on the left. In this time analyses showed liver dysfunction. The findings of computed tomography showed liver is enlarged, with an uneven, wavy contour, unevenly dysmetabolic, multiple hypodense foci of various sizes in the parenchyma, splenomegaly. The Child-Pugh score is B, and the Model for End-stage Liver Disease score is 17. It is planned to add the patient to the waiting list for liver transplantation.

Results: Homozygous likely pathogenic variant in ABHD5 (NM_016006.6) gene were found after whole exome sequencing: c.133+3A > C. Variant were classified based on the ACMG guidelines.

Conclusion: If the patient has ichthyosis along with damage to other organs, and there is no family history, one should think about rare syndromes. Genetic testing is important in the diagnosis of rare diseases, how in this case.

Grants: -

Conflict of Interest: None declared

EP08.034 Population-based cohort of patients with Gaucher disease in southwestern Colombia: sociodemographic, clinical, enzymatic and molecular differences by disease severity

Daniela Arturo-Terranova1;2, Lina Moreno-Giraldo3, Jose Maria Satizabal Soto 1

1Universidad del Valle, Colombia; 2Universidad Santiago de Cali, Colombia; 3Universidad Libre, Colombia

Background/Objectives: Gaucher disease (GD) is the most prevalent lysosomal storage disease, is characterized by beta-glucocerebrosidase deficiency. Inadequate function of this enzyme results in accumulation of glucocerebroside in cells of the macrophage monocyte system. Accumulation occurs in the spleen, liver, bone marrow, lungs, and central nervous system.

Methods: A retrospective observational cross-sectional study was performed using a prospectively collected database of 30 patients diagnosed with GD from southwestern Colombia.

Results: A total of 30 patients (16 women and 14 men) were studied. The mean age was 35 years (IQR 9–74). The initial symptom prior to the diagnosis of the patients in 87% included visceromegaly and cytopenias. 23% with a family history of GD, 6% with Ashkenazi Jewish ancestry. GBA gene analysis reported homozygosity in 1/30 patients (3.3%), compound heterozygosity in 12/30 (40%), heterozygous patients in 10/30 (33.3%), and 23% of patients still didn’t have with molecular test; the most frequently genetic variant was p.Asn409Ser (42%), followed by p.Lys237Glu (18%) and p.Leu483Pro (10%). All the variants presented a pathogenic clinical significance. After treatment 20% presented hepatosplenomegaly, 10% with previous splenectomy. Regarding bone disease, 20% reported bone pain. 79.5 % patients received specific therapy [ERT Alpha : 63.3% Beta : No report ; TRS : 16.2%]

Conclusion: In Colombia there is no similar case follow-up study in patients with DG, so advancing in the understanding of clinical manifestations will contribute to an early and targeted diagnosis and treatment, providing adequate genetic counseling, thus reducing morbidity and mortality.

Conflict of Interest: None declared

EP08.036 Novel deep intronic variant in GALC gene in the patient with infantile Krabbe disease

Nina Maric 1, Bozica Kecman2, Slavica Ostojic2

1University Clinical Centre of the Republika Srpska, Clinic for Children’s Diseases, Banja Luka, Bosnia and Herzegovina; 2Mother and Child Health Care Institute of Serbia “Dr Vukan Cupic”, Beograd, Serbia

Background/Objectives: Krabbe disease (KD) is a rare autosomal recessive neurodegenerative disorder caused by a deficiency of lysosomal enzyme galactocerebrosidase due to mutations in GALC gene (14q31). The majority of patients have an infantile form of disease, presenting within the first 6 months as extreme irritability, spasticity, and developmental delay. The most common patogenic variant in GALC gene is 30 kb deletion. Other variants are less frequently found and deep intronic variants have been described only in a few cases.

Methods: We report a novel deep intronic variant in GALC gene in an 8-month-old patient with an infantile form of KD who has been suffering from irritability and psychomotor regression since the age of 5 months. Clinical, neuroimaging and EEG findings were typical for KD and galactocerebrosidase enzyme activity in leukocytes was very low (0% of normal activity). Genetic testing was done using whole exome and genome sequencing (WES and WGS) and chromosomal microarray analysis (aCGH).

Results: WES revealed, and aCGH confirmed, heterozygous interstitial pathogenic 800 kb long deletion in 14q31.3 cytoband including GALC gene (paternal origin). Following WGS detected the novel hemizygous deep intronic variant c.329-420C > G in GALC gene (maternal origin).

Conclusion: This case might expand the spectrum of GALC mutations and highlight the importance of studying variants in a deep intronic sequence in patients with KD. Confirmation of genetic data regarding the pathogenicity of this novel variant could assist in genetic counseling for family, prediction of clinical outcomes and in decision about clinical intervention for positive cases during newborn screenings.

Grants:

Conflict of Interest: None declared

EP09 Immunology and Hematopoietic System

EP09.002 Maternal cell engraftment in two patients with severe combined immunodeficiency

Rungroj Thangpong 1;2, Phawin Kor-anantakul1;2, Wuttichart Kamolvisit1;2, Chupong Ittiwut1;2, Rungnapa Ittiwut1;2, Narissara Suratannon3, Vorasuk Shotelersuk1;2, Kanya Suphapeetiporn1;2

1Faculty of Medicine, Chulalongkorn University, Center of Excellence for Medical Genomics, Department of Pediatrics, Bangkok, Thailand; 2King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Excellence Center for Genomics and Precision Medicine, Bangkok, Thailand; 3Department of Pediatrics, Faculty of Medicine, Chulalongkorn Universityv, Center of Excellence for Allergy and Clinical Immunology, Division of Allergy, Immunology and Rheumatology, Bangkok, Thailand

Background/Objectives: IL2RG is one of the most common genes responsible for X-linked severe combined immunodeficiency (SCID) (OMIM: 300400). Maternal T cell engraftment has been reported in several cases of IL2RG SCID, identified through clinical phenotype and surface immunophenotype. Exome sequencing (ES), a widely used platform, emerges as a robust genetic testing method for inborn errors of immunity (IEI). Given a sufficient read depth in ES, successful detection of maternal cell engraftment becomes feasible.

Methods: Two patients with IL2RG SCID underwent exome sequencing followed by Sanger sequencing. Additionally, fluorescent in situ hybridization (FISH) was employed to validate the maternal cell engraftment in case 2.

Results: We identified the pathogenic variant c.202G>A, [p.(Glu68Lys)] in IL2RG for case 1 and c.694G>A, [p.(Gly232Arg)] in IL2RG for case 2. Additionally, both patients exhibited a low read depth of reference base pairs in the variants, along with certain single nucleotide polymorphisms (SNPs) on the X chromosome. Further exploration of case 2 confirmed low levels of XX cells through XY-Probe FISH, substantiating the evidence of maternal cell engraftment.

Conclusion: ES serves as a valuable tool for genetically diagnosing patients with IEI and detecting maternal cell engraftment. It is recommended to initiate early and comprehensive molecular genetic testing in conjunction with immunological assessment.

Grants: Health Systems Research Institute (#65-089 and #66-122), and the Care-For-Rare Foundation

Conflict of Interest: None declared

EP09.003 The JAK2 exon 12 mutations in V617F negative patients with erythrocytosis

Biljana Todorić-Živanović 1, Milica Strnad1, Lavinika Atanaskovic2, Marija Elez2

1Military Medical Academy, Molecular Pathology, Belgrade, Serbia; 2Military Medical Academy, Haematology, Belgrade, Serbia

Background/Objectives: The JAK2 V617F mutation is present in more than 95% of polycythemia vera (PV) patients (pts). However, there is still a spectrum of unclear erythrocytosis pts. JAK2 exon 12 mutations are detected in 2-5% of V617F negative PV pts.

Methods: We selected a group of 25 JAK2V617F negative pts, with erythrocytosis, previously analyzed, in our department. We analyzed them for JAK2 exon 12 mutations. DNA was extracted from peripheral blood samples, according to standard procedures. We used RQ-PCR mutations screening based methodology for detecting a group (I540-E543Del, R541-E543Del, F537-K539Del, H538-K539Del, K539L, N542-E543Del) of exon 12 mutations.

Results: We detected one positive patient for exon 12 mutation, in this group. Spontaneous erythroid progenitor cells (BFU-E) and lower level of erythropoietin were also found in this patient.

Conclusion: This group of mutations are not so frequent, but they helps in distinguishing between PV and secondary erythrocytosis pts. Detecting of a novel mutations in signaling pathways represents a potential new targets for molecularly targeted therapies and serve as the potential prognostic factors, in corelation with clinical course of the disease.

Conflict of Interest: None declared

EP09.004 Mutation of Human disabled homolog 2-interacting protein (DAB2IP) gene associates with a new type of hereditary angioedema

Rosa Santacroce 1, Giorgia Cordisco1, Alessandra Ranaldi1, Daniel Vazquez2, Anna Laura Colia1, Angelica Leccese1, Maria Francesca D’Ambrosio1, Giovanna D’Andrea1, Dario Josviack3, Maurizio Margaglione1, Maria d’Apolito1

1Medical Genetics, Department of Clinical and Experimental Medicine, Foggia, Italy; 2Clinica Privada Monte Grande, Servicio de Alergia, Buenos Aires, Argentina; 3Instituto de Medicina Respiratoria, Santa Fe, Argentina

Background/Objectives: Hereditary angioedema is a rare inherited disorder characterized by recurrent episodes of the accumulation of fluids outside of the blood vessels, causing rapid swelling of tissues in the hands, feet, limbs, face, intestinal tract, or airway We conducted genetic studies in Argentine patients with HAE-unknown to identify novel causative genes.

Methods: Whole exome sequencing (WES) was performed on affected family members to identify potential genetic variants associated with nC1-INH-HAE.

Results: A missense variant in the Human disabled homolog 2-interacting protein (DAB2IP) gene, specifically the D239N variant, was identified. In silico analysis using various tools indicated that the D239N variant could have a detrimental effect on the functionality of the DAB2IP protein. The D239N gene variant was found to be rare and not reported in existing databases. Conservation analysis revealed a high conservation of the DAB2IP protein across different species. Protein structure modeling predicted changes in the mutant D239N protein structure, impacting protein stability. Mutagenesis experiment was conducted and Confocal microscopy analysis shown reduced co-localization of the mutant protein with VEGFR2, suggesting a diminished ability of VEGFR2 binding.

Conclusion: The study identified a novel missense variant (D239N) in the DAB2IP gene in a family with HAE-unknown. The findings highlight the role of dysregulated VEGF-mediated signaling in altered endothelial permeability, contributing to recurrent angioedema in affected individuals.

Grants: Progetto “HEAL ITALIA – Health Extended Alliance for Innovative Therapies, Advanced Lab-research and Integrated Approaches of Precision Medicine” Cod. PE_00000019:

Conflict of Interest: None declared

EP09.005 A Homozygous IFNGR1 Mutation Revealed Rare Immunodeficiency Syndrome For A Patient Having Common Symptoms

İlknur Sunar1, Ayse Busra Akalin2, Ibrahim Akalin1, Fatih Demircioğlu 3, Mohammed Alchalabi4, Zeynep Derin5, Betül Tavil6

1Metagentech Center for Genetic Diseases, Istanbul, Türkyie; 2Bahcesehir University, Faculty of Medicine, Istanbul, Türkyie; 3Medistate Hospital, Pediatric Hematology, Istanbul, Türkyie; 4Albatool Teaching Hospital for Maternity and Children, Pediatrics, Diyala, Iraq; 5Bilgi University, Genetics and Bioengineering, Istanbul, Türkyie; 6Medistate Hospital, Pediatric Hematology, Türkyie

Background/Objectives: Fever, anemia, lymphadenopathy and hepatosplenomegaly are common clinical symptoms in malignancy, immunodeficiency and/or metabolic and infectious diseases. Genetic analysis using whole exome sequencing (WES) is becoming essential to unveil diagnostic dilemmas.

Methods: Here we report a 1.5-year-old girl born to a third-degree consanguineous marriage who was admitted first at 7th month with fever. The symptoms were sequentially fever, weight loss, lymphadenopathy and hepatosplenomegaly. Anemia, chronic infection and leukocytosis were encountered at her next admission. Her family history was remarkable for four siblings who had passed because of the same clinical evidence. WES and in silico research were carried out in rush.

Results: A homozygous c.105dup:p.T36Yfs*3 (NM_000416.3) frameshift variant was identified on Interferon Gamma Receptor 1 (IFNGR1) gene causing susceptibility to mycobacterial infections. Then, bone marrow transplantation was planned for the patient, but the patient passed away due to her worsening clinical condition.

Conclusion: Here we reported a third case of Immunodeficiency 27A with c.105dup homozygous variant up to date. Although the variant has not been reported yet at in silico databases like GnomAD and ClinVar, which led us to suppose it as novel. But comprehensive literature search revealed that it had been reported previously by Dorman et al. in 2004 just once in a table -causing difficulty for apparent detection- for two Turkish patients with similar clinical findings. In conclusion, development of novel techniques is needed and crucial to establish immediate genotype-phenotype relationship of the mutations when the time is a matter of life.

Grants:

Conflict of Interest: None declared

EP09.006 Causal DNA variants identified in SCID-associated genes in Slovak population.

Gabriela Krasnanska 1;2, Lenka Wachsmannova1, Marian Baldovič1;3, Gabriela Bľandová1;4, Vladimír Eliáš1, Michal Konecny1;2

1GHC GENETICS SK, Laboratory of genomic medicine, Bratislava, Slovakia; 2Institute of Biology and Biotechnology, Department of Biology, Trnava, Slovakia; 3Faculty of Natural Sciences Comenius University in Bratislava, Department of molecular biology, Bratislava, Slovakia; 4Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Bratislava, Slovakia

Causal DNA variants identified in SCID-associated genes in Slovak population.

Background: Severe combined immunodeficiencies (SCID) belong to the inborn errors of immunity (IEI), represent a rare inherited disorder, generally characterised by recurrent infections and manifestations of immune dysregulation in early childhood. The International Union of Immunological Societies has overall described 19 SCID-associated genes, representing by ADA, AK2, CD3D, CD3E, CD3Z, CORO1A, DCLRE1C, IL2RG, IL7R, JAK3, LAT, LCP2, LIG4, NHEJ1, PRKDC, PTPRC, RAC2, RAG1 and RAG2.

Methods: Our study has been focused on the complex genomic analysis of IEI associated virtual panel of genes based massive parallel sequencing on platforms CES and WES, in Slovak population. Further analysis was aimed for identification of causal DNA variants in mentioned SCID associated genes. Samples were collected from Ambulances of Clinical Genetics from different regions of Slovakia in the period 2019-2023.

Results: Altogether, we have analysed the cohort of 135 samples from patients with suspected IEI. We have identified 14 unique DNA variants with potential pathogenic and uncertain significance clinical effect in 9 SCID genes. Concretely, we have identified two causal DNA variants in the IL2RG gene and one VUS DNA variant with potentially pathogenic predictions in the RAC2 gene.

Conclusion: The presented study, focused on the comprehensive genomic analysis of SCID patients, represents a pilot study in the Slovak population. Presented results confirm that the performance of massive parallel sequencing in patients with IEI, represents a practical complex approach, which leads to fast and effective diagnostics and further treatment.

Conflict of Interest: None declared

EP09.007 Identification of genetic biomarkers for enhanced anaphylaxis diagnosis

Manca Svetina 1, Mitja Košnik1;2, Renato Eržen1;3, Peter Kopač1;2, Peter Korošec1;4;5, Matija Rijavec1;6

1University Clinic of Respiratory and Allergic Diseases Golnik, Golnik; 2Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; 3Topolšica Hospital, Topolšica, Slovenia; 4Faculty of Pharmacy, University of Ljubljana, Ljubljana; 5Faculty of Medicine, University of Maribor, Maribor, Slovenia; 6Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia

Background: Anaphylaxis, a severe hypersensitivity reaction, poses a life-threatening risk and manifests through diverse symptoms, making diagnosis challenging. Although serum tryptase levels are measured to confirm anaphylaxis, the absence of tryptase dynamics is often encountered, indicating the potential involvement of unidentified biomarkers and pathways.

Methods: Bulk RNA-sequencing in 15 patients with anaphylaxis revealed that IL1R2, MMP9, and OSM could play an important role. Subsequent validation of these candidate genes was conducted in an independent cohort comprising 32 anaphylaxis patients with paired acute and convalescent samplings and 13 healthy control individuals. Total serum tryptase levels and mRNA levels of IL1R2, MMP9 and OSM were determined using FEIA-CAP and RT-qPCR, respectively.

Results: Gene expression of IL1R2, MMP9 and OSM significantly increased during acute anaphylaxis compared to convalescent samples (all p < 0.001) and healthy subjects (all p < 0.001). The ROC area under the curve (AUC) was the highest for the Gene mean, calculated as the mean of standardized gene expression variables of IL1R2, MMP9 and OSM (AUC = 0.96), followed by OSM expression (AUC = 0.90), IL1R2 expression (AUC = 0.89) and MMP9 expression (AUC = 0.86). As indicated by estimated AUCs, Gene mean, OSM and IL1R2 expression showed higher accuracy in discriminating between patients with anaphylaxis and the controls than tryptase (AUC = 0.88).

Conclusion: IL1R2, MMP9, and OSM could serve as biomarkers for anaphylaxis, confirming clinical diagnosis and offering insight into novel pathways for potential therapeutic strategies.

Conflict of Interest: None declared

EP09.008 A new case of Ghosal syndrome: a rare but treatable cause of anemia and developmental delay

Willemijn Kuper 1, Eva Rettenbacher2, Annet Van Hagen1

1Amsterdam UMC, human genetics, Amsterdam, Netherlands; 2Amsterdam UMC, pediatrics, Amsterdam, Netherlands

Background/objectives: We present a case of a 2 year old girl who visited our pediatric hematology and clinical genetics clinic with unexplained anemia and a motor developmental delay.

Methods: The following diagnostics were performed: laboratory analysis of the hematological parameters, bone marrow biopsy, mitomycin c (MMC) test for differentiation from Fanconi anemia and trio-whole exome sequencing (WES) focused on genes associated with bone marrow failure. Following the WES results, also X-rays were made focused on the bones of the legs.

Results: Laboratory analyses showed anemia and mild thrombocytopenia (Hb - 6.5 g/dl, platelets – 106 x 109E/l). MMC test was negative for Fanconi anemia. Trio-WES however showed bi-allelic variants in the TBXAS1 gene: c.245T>C p.(Leu82Pro) and c.1016T>C p.(Leu339Pro) both considered pathogenic after the X-rays confirmed abnormalities associated with Ghosal hematodiaphyseal dysplasia (Ghosal syndrome).

Conclusion: Ghosal syndrome is a rare autosomal recessive syndrome characterized by metadiaphyseal dysplasia of long bones and defective hematopoiesis caused by pathogenic variants in TBXAS1.

Patients with Ghosal syndrome have been traditionally treated with repeated courses of corticosteroids leading to improvement of cytopenia and bony changes. Recently, patients were shown to also respond well to high dose NSAIDs by reducing COX products. Because of the more favorable side-effect profile of the latter, we recently started treatment with NSAIDs in our patient. Follow-up is needed to assess efficacy and side-effects.

Thus, early diagnosis of Ghosal syndrome allows treatment of patients with corticosteroids or, as shown by recent research, NSAIDs.

Conflict of Interest: None declared

EP09.009 CTLA-4 insufficiency: interdisciplinary care is required for correct diagnosis and adaptation of therapeutic management

Victoria Paul 1, Nadia Korthals1, Judit Horvath1, Frank Tüttelmann1, Bodo Grimbacher2;3;4, Eva Eßeling5

1University Münster, Department of Medical Genetics, Münster, Germany; 2Albert-Ludwigs-University of Freiburg, Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Freiburg, Germany; 3Albert-Ludwigs-University of Freiburg, CIBSS – Centre for Integrative Biological Signalling Studies, Freiburg, Germany; 4Satellite Center Freiburg, RESIST – Cluster of Excellence 2155 to Hanover Medical School, Freiburg, Germany; 5University Hospital Münster, Department of Medicine A, Haematology, Oncology, and Pneumology, Germany

Background/Objectives: CTLA4 encodes the CTLA-4 costimulatory molecule expressed on activated T-cells. Monoallelic pathogenic variants in CTLA4 cause CTLA-4 insufficiency, an immune-dysregulatory syndrome with incomplete penetrance and highly variable expressivity. CTLA-4 insufficiency is an ultra-rare disorder affecting both proliferation and function of T-cells. Its symptoms may include immunodeficiency, autoimmunity, and autoinflammation of virtually any organ, rendering clinical diagnosis difficult.

The index patient was referred to our outpatient genetics clinic with suspected dyskeratosis congenita because short telomere length was found in lympho- and granulocytes. He presented at age 20 with severe anaemia and thrombocytopenia with spontaneous bleeding, and recurrent infections (perianal abscess, mastoiditis). Bone marrow transplantation was considered, but postponed due to suspected aspergillosis of the lungs.

Methods: We obtained a detailed individual and family history. Exome sequencing, literature research, and interdisciplinary consultations were conducted. Additionally, we discussed the case in a virtual conference with clinicians specialised in inborn errors of immunity.

Results: Genetic work-up yielded a likely pathogenic (ACMG class 4: PVS1, PM2) heterozygous frameshift variant in CTLA4. We detected no additional potentially causative variant in other relevant genes.

Conclusion: Exome sequencing and interdisciplinary exchange facilitated correct and timely diagnosis in the severely ill patient. Once diagnosis was established, adaptations to therapeutic approach were made and the patient underwent successful bone marrow transplantation. Testing of family members can now be offered, with implications for therapy and management.

Grants: B.G. is funded by the Deutsche Forschungsgemeinschaft (SFB1160/2_B5; RESIST–EXC 2155–Project ID 390874280; and the BMBF (GAIN 01GM2206A).

Conflict of Interest: None declared

EP09.010 First report of KRAS gene gain of function somatic variant p.Gln22Lys as a cause of RAS-associated autoimmune leukoproliferative disorder (RALD).

Rasa Insberga 1;2, Ieva Mičule1, Ludmila Voložonoka1, Dmitrijs Rots1, Dace Enkure1, Anna Valaine3

1Children’s Clinical University Hospital, Medical Genetics and Prenatal Diagnostics Clinic, Rīga, Latvia; 2Riga Stradins University, Rīga, Latvia; 3Children’s Clinical University Hospital, Department of hematooncology, Rīga, Latvia

Background/Objectives: Differential diagnosis of autoimmune lymphoproliferative syndrome (ALPS) is broad and includes RAS-associated autoimmune leukoproliferative disorder (RALD). KRAS gene (MIM*190070) is a protooncogene that is involved in cell division regulation. Somatic gain of function mutations of the KRAS and NRAS have been described in association with RALD. However, so far only mutations affecting two hotspot codons have been described: p.Gly12 and p.Gly13. Codon Gln22 variant has been described as an oncogenic variant in tumours, but not as a somatic variant causing RALD. Here, we present a case with p.Gln22Lys somatic variant as a cause of RALD.

Methods: Individual was consulted in Children’s Clinical University Hospital, Riga, Latvia. Proband, a 6-year-old female, has had neck, axillary and inguinal lymphadenopathy since the age of two years and also has bilateral nodules in thyroid gland (TI-RADS 4b-c). She has increased number of double-negative cells, so based on her presentation ALPS was suspected.

Results: Proband underwent diagnostic exome sequencing, with analysis focused on immunodeficiencies, which identified a gain of function variant in the KRAS gene c.64C>A, p.Gln22Lys with allele frequency that convincingly corresponds with somatic event (VAF = 33%, 43/131 reads). This variant has been described in cancers and proven functionally to result in gain-of-function, but currently the child does not have haematological malignancy. Therefore, the final diagnosis of RALD was established.

Conclusion: We describe novel oncogenic KRAS variant as a cause of RALD and highlight the importance of detecting also somatic variants in the diagnostic process for patients with suspected primary immunodeficiencies.

Grants: None.

Conflict of Interest: None declared

EP09.011 Plasma exosomal miR-15a-5p regulate Smad7/TGF-β1 signalling in systemic lupus erythematosus-associated renal damage

Ana Flores-Chova 1, Olga Martinez-Arroyo1, Marta Mendez-Debaets1, Laia Garcia-Ferran1, Lorena Adan1, Liria Terrádez2, Josep Redon1;3, Maria J Forner1;3;4, Ana Ortega1, Raquel Cortés1

1Biomedical Research Institute Hospital Clinico – INCLIVA, Cardiometabolic and Renal Risk Research Group, Valencia, Spain; 2Hospital Clinico Valencia, Anatomical Pathology, Valencia, Spain; 3Hospital Clinico Valencia, Internal Medicine, Valencia, Spain; 4Universitat de Valencia, Medicine, Valencia, Spain

Background and objectives: A previous specific miRNA profile in plasma exosomes from systemic lupus erythematosus patients (SLE) was identified by our group. Exosomal miR-15a-5p had significant increased levels in patients with albuminuria and bioinformatic analysis showed that miR-15a-5p could be regulating TGF-β1 signalling. Our objective was analysed exosomal miR-15a-5p association with lupus nephritis (LN) and its role regulating TGF-β1 pathway.

Methods: Plasma samples were obtained from 115 SLE patients (30 with LN) and 30 controls. Plasma-exosomal miR-15a-5p was quantified by qPCR. Its role in TFG-β1 pathway was evaluated with mimics-15a-5p transfection experiments in podocytes and Smads molecules were assessed.

Results: Plasma exosomal miR-15a-5p was increased in LN compared to controls or SLE without LN. For discriminating renal damage between patient groups, miR-15a-5p obtained a significant AUC (0.81, p < 0.0001). Next, we analyzed the association between the ISN/RPS 2003 classification of LN in 18 renal biopsies and exosomal miR-15a-5p levels were significantly increased in proliferative forms (Class III + IV). In vitro experiments showed increased miR-15a-5p in exosomes from podocytes treated with TGF-β1. After transfection experiments SMAD7 mRNA levels were significantly decrease with or without treatment (p < 0.05) and immunofluorescence experiments showed a significant increase in Smad7 protein nuclear/cytoplasmatic ratio.

Conclusion: Plasma exosomal miR-15a-5p was increased in LN and regulate Smad7/TGF-β1 signalling, a critical pathway in SLE-associated renal damage progression. This finding could contribute to advances in SLE diagnosis and as promising therapeutic targets.

Grants: Health Institute Carlos III [PI18/01405, PI21/00249; PI23/01179; FI20/00096 FI22/00032; Miguel Servert Contract CP22/00069] IJC2020-045308-I and CNS2022-136-175 by MCIN/AEI. European Regional Development Fund (ERDF).

Conflict of Interest: Ana Flores-Chova Health Institute Carlos III, FI22/00032, PI21/00249; CNS2022-136-175 by MCIN/AEI, Olga Martinez-Arroyo Health Institute Carlos III, FI20/00096, PI21/00249; CNS2022-136-175 by MCIN/AEI, Marta Mendez-Debaets: None declared, Laia Garcia-Ferran: None declared, Lorena Adan: None declared, Liria Terrádez Health Institute Carlos III, PI21/00249, Josep Redon: None declared, Maria J Forner Health Institute Carlos III [PI18/01405, PI21/00249, Ana Ortega Health Institute Carlos III [PI23/01179] IJC2020-045308-I and CNS2022-136-175 by MCIN/AEI., Raquel Cortés Health Institute Carlos III [PI18/01405, PI21/00249; FI22/00032; Miguel Servert Contract CP22/00069] CNS2022-136-175 by MCIN/AEI.

EP09.012 A survey study to investigate for the presence of a variant of the X-chromosome Interleukin-1 Receptor-Associated Kinase 1 (IRAK1) gene suspected to produce a hyperactive IRAK1 protein in synovial fibroblasts in rheumatoid arthritis, instead resulted in revealing a predominance of men with very high cyclic citrullinated antibody titers suggesting environmental exposure as cause of Veteran population’s inflammatory arthritis

Lane Scheiber II1, Neha Gupta 2, Sarah Ford1, Evan Dombrosky1

1Richmond Veterans Administration Medical Center, Rheumatology, Richmond, United States; 2Virgina Commonwealth University, Division of Rheumatology, Allergy and Immunology, Richmond, United States

Background/Objectives: Rheumatoid arthritis (RA) is postulated to be the result of a combination of genetic and environmental influences. Root cause analysis of genetic processes of seropositive RA (SPRA) demonstrated that toll-like receptor 4 on synovial fibroblasts is triggered due to Escherichia coli Group D lipopolysaccharides utilizing Interleukin-1 Receptor-Associated Kinase 1 (IRAK1) protein; this gene located on the X-chromosome, with SPRA being highly recognized to demonstrate a variable penetrance in women. Typically, 7.7% of RA patients should be rheumatoid factor (RF) positive, cyclic citrullinated antibody (CCP-Ab) negative. CCP-Ab is associated with the development of RA in smokers.

Methods: A six-month survey designed to investigate for maternal family history of RA in RF positive, CCP-Ab negative male patients was conducted in the Richmond Veterans Administration Medical Center Rheumatology outpatient clinic.

Results: Three patients (1.19%) of 252 were RF positive, CCP-Ab negative - one with paternal RA history, one adopted, one uncertain regarding RA family history.

Conclusion: Survey results are inconclusive for IRAK1 gene influence. Excluded patients demonstrated most RA patients had a high titer CCP-Ab. Prior studies have shown RA predominantly in women, with a CCP-Ab positive prevalence of 64-96%. Our survey showed a majority of men with very high CCP-Ab and low percent of RF positive CCP-Ab negative. Theoretically, onset of RA in the veteran population may be related to genetic predisposition and exposure to selective smoke antigens. This suggests early screening for CCP-Ab and anti-citrullinated protein antibody FAB glycans somatic hypermutation in high-risk positions may avoid occurrence of RA in this population.

Grants: Not applicable

Conflict of Interest: None declared

EP09.013 The zebra among horses - COPA syndrome as a potentially underdiagnosed autoimmune disorder

Jasmin Blatterer 1, Sabrina Hammer1, Sophie Bierbaumer1, Rusmir Husic2, Johannes Schmid3, Elisabeth Schreiner1, Heidelis Tichy1, Klaus Wagner1, Sarah Verheyen1

1Medical University of Graz, D&F Institute of Human Genetics, Graz, Austria; 2Medical University of Graz, Department of Rheumatology and Immunology, Graz, Austria; 3Medical University of Graz, Division of General Radiology, Department of Radiology, Graz, Austria

Background/Objectives: COPA encodes a subunit of coat protein complex I (COPI), which is involved in vesicular protein transport between Golgi and ER. COPI dysfunction can cause an autosomal dominant autoimmune disorder with lung, joint and kidney involvement. Here, we report on a 30-year-old patient with COPA syndrome (CS) who was initially diagnosed with juvenile systemic lupus erythematosus (SLE) and discuss CS as a potentially underdiagnosed autoimmune disorder.

Methods: Trio Whole exome sequencing was performed during our patient’s pregnancy due to abnormal fetal ultrasound findings. Reverse phenotyping of the patient and a literature review were conducted subsequent to the genetic diagnosis.

Results: A previously reported heterozygous missense variant NM_004371.4:c.718T>C, p.(Trp240Arg) in COPA was identified as an incidental finding in our patient and her unborn child. At age three she presented with pulmonary hemosiderosis. Arthritis and neurodermatitis manifested in adulthood. Arthritis was seropositive for antinuclear antibody (ANA), rheumatoid factor and antineutrophil cytoplasmatic antibodies (ANCA). A literature review revealed different initial clinically suspected diagnosis before CS was genetically diagnosed, including hypersensitivity pneumonitis, lymphangioleiomyomatosis, rheumatoid arthritis (RA) and SLE.

Conclusion: CS is a rare genetic disorder with variable phenotypic expression, of which less than 100 individuals have been described in the literature. The clinical presentation of CS overlaps with better-known autoimmune disorders such as SLE or RA. CS is a potentially underdiagnosed hereditary autoimmune disease. Especially in patients with infantile disease onset, genetic testing should be considered for diagnostic and therapeutic purposes as well as risk assessment in affected families.

Conflict of Interest: None declared

EP09.014 Expression of the immune related genes TGFB1, CCL5, IL1B, OAS1, IRF9, and IFI6 genes in COVID-19 pathogenesis

Malena Gajate-Arenas 1, Ingrid Fricke-Galindo2, Omar García-Pérez1, Angélica Domínguez-de-Barros1, Gloria Pérez-Rubio2, Roberto Dorta Guerra1;3, Ivette Buendia Roldán4, Leslie Chavez-Galán5, Jacob Lorenzo-Morales1;6;7, Ramces Falfán Valencia2, Elizabeth Córdoba-Lanús1;6;7

1Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, La Laguna, Tenerife, Spain; 2Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, HLA Laboratory, Ciudad de México, Mexico; 3Universidad de La Laguna, Departamento de Matemáticas, Estadística e Investigación Operativa. Facultad de Ciencias. Sección de Matemáticas., La Laguna, Tenerife, Spain; 4Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Translational Research Laboratory on Aging and Pulmonary Fibrosis, Ciudad de México, Mexico; 5Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Laboratory of Integrative Immunology, Ciudad de México, Mexico; 6Consorcio Centro de Investigación Biomédica (CIBER) de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain; 7Universidad de La Laguna, Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, La Laguna, Tenerife, Spain

Background/Objectives: COVID-19, a disease cause by SARS-CoV-2 infection, is characterized by a wide range of clinical manifestations. Aging, underlying diseases and the genetic background has been related to worse outcomes. In the present multicentric study, the differential expression of the immunity related genes: IRF9, CCL5, IFI6, TGFB1, IL1B and OAS1 was analyzed in moderates and severe cases of COVID-19.

Methods: The relative gene expression in whole-blood samples from the 160 hospitalized individuals was determined by a two-step RT-qPCR. Clinical variables were recorded.

Results: The multivariate analysis revealed that the expression of OAS1 (p < 0.05) and IFI6 (p < 0.05) was higher in moderate hospitalized cases than in severe ones. A lower risk of requiring invasive mechanical ventilation (IMV) was associated with the expression of OAS1 (OR = 0.64, CI = 0.52-0.79; p = .001), IRF9 (OR = 0.581, CI = 0.43-0.79; p = 0.001), and IFI6 (OR = 0.544, CI = 0.39-0.69; p < 0.001). The patient survival was associated with the expression levels of IRF9 (OR = 0.800, CI = 0.653-0.979; p = 0.030), IFI6 (OR = 0.827, CI = 0.690-0.991; p = 0.039), TGFB1 (OR = 0.646, CI = 0.50-0.83; p = 0.001) and CCL5 (OR = 0.57, CI = 0.39-0.83; p = 0.003).

Conclusion: In conclusion, we found differences in the immune related-gene expression between moderate and severe cases of COVID-19. Moreover, our findings confirm the relevance of OAS1, IRF9, and IFI6 in the outcomes of patients infected with SARS-CoV-2, as they associated with a lower risk of death and IMV suggesting a protective effect against SARS-CoV-2 infection.

Grants: Consorcio Centro de Investigación Biomédica (CIBER) de Enfermedades Infecciosas (CIBERINFEC) (CB21/13/00100), Instituto de Salud Carlos III, 28006 Madrid, Spain; Cabildo Insular de Tenerife 2023–2028 (PI-CC202302222, Cabildo.23); and Ministerio de Sanidad, Spain.

Conflict of Interest: None declared

EP09.015 “Characterization of pathogenic and likely pathogenic variants in Romanian children with inborn errors of immunity”

Cristina-Loredana Pantea 1;2, Adela Chirita-Emandi1;2, Mihaela Bataneant3, Maria Puiu1;2

1Genetics Discipline, Center of Genomic Medicine, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Genetics, Timișoara, Romania; 2Regional Center of Medical Genetics Timiș, Clinical Emergency Hospital for Children “Louis Țurcanu” Timișoara, Romania, part of ERN ITHACA, Genetics, Timișoara, Romania; 3Emergency Hospital for Children Louis Ţurcanu, Genetics, Timișoara, Romania

Background/Objectives: Inborn errors of immunity (IEI) are monogenic disorders with a wide spectrum of clinical phenotypes including immune dysregulation, autoimmunity, autoinflammation, and malignancy. Regarding the life-threatening consequences of some IEI, genetic analysis has improved diagnosis and outcome for many affected patients.

We aim to investigate the genotypic findings in children with IEI.

Methods: Clinical and genetic investigations were performed in 85 children with diagnostic suspicion of IEI. Genetic analysis used next generation sequencing panels that included genes associated with IEI (ranging between 1-471 genes) or Whole Exome Sequencing (WES)/ Whole-Genome Sequencing (WGS).

Results: Genetic investigations identified pathogenic or likely pathogenic variants for IEI in 34/85 (40%) participants, in 26 genes. 15/85 (18%) participants had variants of uncertain significance. Mean age at IEI diagnosis was 7,2 (±5.6) years. 4/34 (11,7 %) of patients with positive finding had a family history of IEI. Most frequent genes identified were JAGN1 (5 patients), ATM (2), CFTR (2), CYBB (2), FAS (2), and TRNT1 (2).

Conclusion: A great variety of genes were identified as causative for IEI in our cohort. Considering the highly variable and unspecific phenotype, together with no family history in most cases, a genotyping-first approach constitutes the best option for timely diagnosis and treatment.

Conflict of Interest: None declared

EP09.016 FYN as a potential gene associated with hemophagocytic lymphohistiocytosis

Katharina Schau-Römer 1, Simon Vieth2, Almuth Caliebe2, Malte Spielmann1;3, Inga Nagel1

1University Hospital Schleswig-Holstein, Campus Kiel, Institute of Human Genetics, Kiel, Germany; 2University Hospital Schleswig-Holstein, Campus Kiel, Department of Pediatrics, Kiel, Germany; 3Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Institute of Human Genetics, Lübeck, Germany

Background/Objectives: Hemophagocytic lymphohistiocytosis is a rare and severe immunological disorder characterized by the hyperactivation of immune cells, notably macrophages and T-cells. In adults, the secondary (acquired) form is predominant, whereas in children, the primary (inherited) form is more common.

Methods: Trio-exome sequencing was performed on the index person and her parents. The index person, a 2-year-old girl suspected of having hemophagocytic lymphohistiocytosis. Clinically, the patient presented with recurrent fever episodes, pancytopenia, hepatosplenomegaly, and NK cell depletion and EBV + T-Zell lymphoproliferative disease.

Results: We identified a potentially protein damaging de novo variant in the FYN gene (NM_002037.5:c.526C>T p.(Arg176Cys)). FYN is a tyrosine kinase involved in immunomodulation and interacts, among others, with LYN in the autoimmune response. While FYN has not been described as an OMIM morbid gene yet, pathogenic variants in LYN are associated with systemic autoinflammatory disease with vasculitis (SAIDV).

Conclusion: Since LYN is a known OMIM morbid gene involved in the regulation of immunological processes and FYN is known as an interacting partner, we believe that FYN represents a promising candidate gene associated with our patient’s disease.

Conflict of Interest: None declared

EP09.017 ACTN1-related thrombocytopenia: homozygosity for an ACTN1 variant results in a more severe phenotype

Melania Eva Zanchetta 1, Serena Barozzi2, Federica Isidori3, Caterina Marconi3, Loredana Farinasso4;5, Roberta Bottega1, Anna Savoia6, Alessandro Pecci2;7, Michela Faleschini1

1Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy; 2IRCCS Policlinico San Matteo Foundation, Medical Department, Pavia, Italy; 3IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; 4Regina Margherita Children Hospital, Turin, Italy; 5University of Turin, Turin, Italy; 6University of Verona, Department of Engineering for Innovation Medicine, Verona, Italy; 7University of Pavia, Department of Internal Medicine, Pavia, Italy

Background/Objectives: ACTN1-related thrombocytopenia (ACTN1-RT; (OMIM: 615193)) is a rare autosomal dominant disorder characterized by thrombocytopenia, platelet macrocytosis and mild bleeding tendency, caused by mutations in the ACTN1 gene which encodes a conserved F-actin binding and crosslinking protein. Overexpression of mutant ACTN1 causes disorganization of the actin cytoskeleton in cells. To date, approximately 50 heterozygous mutations spanning the entire gene have been reported.

Methods: Mutation screening was performed by whole exome sequencing (WES) in the proband of a Moroccan family, followed by Sanger sequencing for segregation analysis. Measurements of platelet diameter on peripheral blood smears was performed by software-assisted image analysis. F-actin staining was performed by immunofluorescence in cells ectopically expressing WT or mutant ACTN1.

Results: We describe for the first time two patients affected with ACTN1-RT caused by a homozygous variant in ACTN1. They presented with thrombocytopenia of moderate degree and marked platelet macrocytosis with giant platelets, whereas their heterozygous relatives had normal or slightly decreased platelet counts, and a lower degree of platelet macrocytosis. Both homozygous patients have mitral and aortic valve defects that cannot be explained by mutations in other genes. Immunofluorescence analysis of U20S cells overexpressing mutant ACTN1 shows an abnormal organization of the actin cytoskeleton compared to control cells.

Conclusion: Our study suggests that allelic burden modulates the phenotypic impact of ACTN1 mutation affecting the severity of platelet macrocytosis. Although the involvement of the mutation in the pathogenesis of the cardiac defect remains to be proven, we suggest that echocardiography may be appropriate in patients with ACTN1-RT.

Conflict of Interest: None declared

EP09.018 A t(1;2)(p36;p21) translocation in a compound heterozygous beta thalassemia patient needs weekly blood transfusions.

Sude sultan Özdemir 1, Ibrahim Akalin2;3, Fatih Demircioğlu4, İlknur Sunar5, AHMED AL-OMAR6, Ayşe Akalın7, Betül Tavil8

1Istanbul university, Molecular biology and genetics, Istanbul, Türkyie; 2Metagentech Center, Genetic Disease, Istanbul, Türkyie; 3, Istanbul, Türkyie; 4Medistate Hospital, Pediatric Hematology, Istanbul, Türkyie; 5Metagentech center, Genetic disease, Istanbul, Türkyie; 6Metagentech center, Genetic disease, Istanbul, Türkyie; 7Bahçeşehir University, Faculty of medicine, Istanbul, Türkyie; 8Medistate Hospital, Pediatric Hematology, Istanbul, Türkyie

Introduction: Beta thalassemia is an autosomal recessive blood disorder that leads to abnormal hemoglobin production due to genetic mutations in the globin chain. Addressing the pathogenesis, clinical features, and recent advances in diagnosis and treatment of beta thalassemia is essential for understanding of disease progression.

Materials and Methods: Here, we report a 20-year-old female born to non-consanguineous parents. She was suffering from triggered recurrent blood transfusions need as weekly Chromosomal analysis and whole exome sequencing were done for mutation analyses.

Results: WES analyses revealed compound heterozygous pathogenic HBB: c.251delG and HBB: c.112delT mutations that was confirmed by parental mutation analyses who carried one of the mutations. Chromosome analyses revealed a t(1;2)(p36;p21) translocation in one metaphase.

Conclusions: The t(1;2)(p36;p21) translocation was attributed with generation of PDRM16/THADA fusion transcript that responsible for development of myelodysplastic syndrome (MDS). The major questions arose whether the translocation was a consequence of recurrent transfusions or a co-expresssion or was the result of beta thalassemia progression itself which may end up with MDS development. Hence, for the first time, we report t(1;2)(p36;p21) translocation in a molecularly confirmed beta thalassemia patient, up to the literature.

Conflict of Interest: None declared

EP09.019 From hysteria to targeted therapy and complex genetic counselling in half-sisters with Germline mosaicism in STAT3 in hyper-IgE syndrome

Quentin Sabbagh1, Jean-David Cohen2, Jeremie Mortreux3, Laure Raymond3, Guilaine Boursier4, Vanna geromel3, eric jeziorski5, Vincent Le Moing6, Didier Bessis7, David Geneviève 1

1Montpellier University, INSERM U1183, Service de Génétique Clinique, Centre Hospitalier Universitaire de Montpellier, Montpellier, France; 2Montpellier University, Département de Rhumatologie, Montpellier, France; 3Laboratoire Eurofins Biomnis, Lyon, France; 4Montpellier University, Department of Molecular Genetics and Cytogenomics, Rare and Autoinflammatory Diseases Unit, Centre Hospitalier Universitaire de Montpellier, Montpellier, France; 5Montpellier University, Département de Pédiatrie Générale, Infectiologie et Immunologie Clinique, Centre de Référence des Maladies Auto-Inflammatoires Rares et des Amyloses, Centre Hospitalier Universitaire de Montpellier, Montpellier, France; 6Montpellier University,r, Département de Maladies Infectieuses et Tropicales, Centre Hospitalier Universitaire de Montpellie, Montpellier, France; 7Montpellier University, Département de Dermatologie, Centre Hospitalier Universitaire de Montpellier, Montpellier, France

Hyperimmunoglobulin E syndrome (HIES, 5 different monogenic diseases) is a rare immunological congenital disease. The major autosomal dominant form of HIES is caused by loss-of-function variations in the signal transducer and activator of transcription 3 gene (STAT3 OMIM: 102582). The main clinical features are recurrent skin abscesses, severe atopic dermatitis, recurrent sino-pulmonary infections and, less frequently, arthritis, and high IgE levels, with onset in early childhood and autoimmune manifestations from the 2nd decade onwards.

We perform exome sequencings in a girls and her unaffected parents presenting eczema, recurrent skin infection, episodes of pneumonia and kidney infection and arthritis with previous diagnosis of hysteria. We also perform exome sequencing in the half-sister (same mother) with similar but slight features. In addition, she had dental malposition.

We report for the first time a germline mosaic in STAT3 responsible for HIES in two maternal half-sisters with a variable clinical presentation, particularly moderate in one of the half-sisters. Both presented with moderate elevated HIE but at lower lever than the classical criterion and without hyperoesinophilia.

This clinical presentation reminds us that we must take into account the fact that even healthy parents of individuals with a “de novo” genetic disease can be carriers of a germline mosaic, but also that two parents with a variable phenotypic expression can present similar hereditary disease. Finally, we to propose targeted immunotherapy (Dupilumab), which has resolved the infectious and cutaneous symptoms but not the arthritis. Bi-immunotherapy could be the key to recovery for these 2 half-sisters.

Conflict of Interest: None declared

EP09.020 Association study of a STAT3 Genetic polymorphism rs1053004 with susceptibility to Behçet’s Disease in Tunisian population

Ahmed Adel Gereisha 1, Rajaa Lahmar1, Rasene Gereisha1, Elyes Chabchoub1, Ramzi Zemni1, Anis Mzabi2, Neirouz Ghannouchi3, Foued Ben Hadj Slama1

1Ibn El Jazzar Medical Faculty of Sousse, Immunogenetics Unit, Sousse, Tunisia; 2UNIVERSITY HOSPITAL SAHLOUL, sousse, Tunisia, Internal medicine Department, Sousse, Tunisia; 3Hospital F. Hached, Internal medicine Department, Sousse, Tunisia

Background/Objectives: Behçet’s disease (BD) is a chronic multisystemic vasculitis of unknown etiology, involving complex interactions between genetic, immunological, and environmental factors. STAT3 gene, a crucial transcription factor in immune responses, has been incriminated in different autoimmune and autoinflammatory diseases. Herein, we aimed to investigate the association between STAT3 rs1053004 polymorphism and BD in a cohort of Tunisian patients.

Methods: STAT3 rs1053004 was investigated in a case-control study involving 110 BD patients and 116 healthy controls. We performed genomic DNA extraction and a Mutagenically separated PCR (MS-PCR) SNP genotyping with newly designed primers.

Results: STAT3 rs1053004 was significantly associated with BD susceptibility in our study. GG genotype was significantly more frequent in BD patients than in healthy controls (27.3% vs. 12.1%, Pc = 0.005, OR 2.83, 95% CI 1.37-5.81). The minor G allele of STAT3 rs1053004 was more prevalent in BD patients than in healthy controls (36% vs. 21%). G allele was associated with an increased BD risk (pc = 0.028, OR = 2.11, 95% CI = 1.12–3.97), a risk factor for BD development in both dominant and recessive models (pc = 0.020, OR = 1.91, 95% CI = 1.10–3.30; pc = 0.006, OR = 2.67, 95% CI = 1.32–5.38).

Conclusion: In our study, we found for the first Time the association between STAT3 rs1053004 and BD. Further validation through prospective studies is crucial to reinforce these findings.

Grants: This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Conflict of Interest: None declared

EP09.021 A homozygous splicing variant c.591+2T > G in F3 causing a novel bleeding disorder

Vivekananda Bhat 1, Namanpreet Kaur1, Amit Jairaman1, Jeevan Kumar1, Radhakrishnan Periyasamy1, Anju Shukla1

1Kasturba Medical College, Manipal, Department of Medical Genetics, Manipal, India

Background: F3 (OMIM #134390) encodes tissue factor (TF) which initiates the coagulation cascade by binding to factor VII and catalyses the proteolytic activation of factors IX and X. Currently, only one family carrying a heterozygous frameshift variant in F3 is reported.

Methods: We ascertained a 13-year-old female with epistaxis, gum bleeding, anemia, and menorrhagia with normal coagulation work up. Singleton exome sequencing (ES) and Sanger validation and segregation was performed in the family. Splicing assay and nonsense-mediated decay (NMD) assay was performed to assess the pathogenicity of the variant.

Results: Solo ES revealed a homozygous canonical splicing variant, g.94533088A > C (NM_001993.5:c.591+2T > G) in F3. Her parents are carriers. This variant is absent in gnomAD and in our in-house data. Splice AI predicts the variant to cause aberrant splicing. In splicing assay, patient cDNA was not detected as compared to the control, thereby possibly undergoing NMD. Further, NMD assay revealed that there are two alternative transcripts with partial exon 4 skipping. Gene and protein expression experiments are currently under process. Functional work carried out on the previously reported heterozygous loss-of-function variant suggested haploinsufficiency as the likely mechanism of disease. We report biallelic loss-of-function splice variant which might lead to the reduction in the TF levels leading to severe bleeding manifestations.

Conclusion: This report provides further evidence for a novel bleeding disorder caused by biallelic loss-of-function variants in F3.

Grants: India Alliance/DBT Wellcome Trust funded project, Centre for rare disease diagnosis, research and training (Grant ID number: IA/CRC/20/1/600002).

Conflict of Interest: None declared

EP09.022 Pioneering diagnostic effect of whole exome sequencing for diverse rare diseases in three patients with common symptoms of anemia and hepatosplenomegaly

Betül Tavil 1, Motusheva Raushan Kimashbekovna2, İlknur Sunar3, Ayse Busra Akalin4, Ibrahim Akalin5

1Medistate Hospital, Pediatric Hematology, Istanbul, Türkyie; 2, Kyrgyzstan; 3Metagentech Center for Genetic Diseases, Istanbul, Türkyie; 4Bahcesehir University Faculty of Medicine, Medicine, Istanbul, Türkyie; 5Metagentech Center for Genetic Diseases, Genetics, Istanbul, Türkyie

Background/Objectives: Anemia and hepatosplenomegaly are common symptoms for various diseases including hematology. Immediate diagnosis and effective treatment would be essential for life threatening conditions.

Methods: We report three unrelated patients aged one- to three-year-old with common symptoms of anemia and hepatosplenomegaly. Whole exome analysis was performed for molecular diagnosis.

Results: WES analysis revealed 3 different underlying conditions for each patient. Diagnosis of Gaucher’s disease, spherocytosis type 1 and hypochromic macrocytic anemia with Iron overload 2 were put due to homozygous GBA c.754T>A:p.Phe252Ile (NM_0001567.4), heterozygous ANK1 c.4096delT:p.C1366Afs*40 (NM_000037.4) and heterozygous STEAP3 c.121C>T:p.R41C (NM_182915.3) mutations were detected, particularly.

Conclusion: Molecular diagnosis using WES would be promising instant approach to unveil differential diagnosis of genetic diseases even having common symptoms. Hence clinical treatment would begin immediately for applicable diseases

Grants:

.

Conflict of Interest: None declared

EP09.023 SBDS Gene Mutations in Shwachman-Diamond Syndrome: A Prime Genetic Diagnostics for the Ukrainian Population

Volodymyr Kravets1;2, Maria Dushar1, Ivanna Shymanska1, Olena Boldyreva3, Liliya Nazarenko4, Halyna Makukh 1;2

1Scientific Medical Genetic Center LeoGENE, Lviv, Ukraine; 2The Ivan Franko National University of Lviv, Genetics and Biotechnology, Lviv, Ukraine; 3KNP “City Clinical Children’s Hospital N16” of the Kharkiv City Council, Hematology, Kharkiv, Ukraine; 4KNP “Cherkasy Regional Children’s Hospital”, Cherkasy, Ukraine

Background/Objectives: More than 90% Shwachman-Diamond syndrome (SDS) cases are caused by mutations in the Shwachman-Bodian-Diamond syndrome (SBDS) gene. SBDS protein coding region shares 96.8% homology with its pseudogene (SBDSP), which makes it challenging to analyse with NGS. SDS complicating diagnosis is resulting in a lack of data on the prevalence of SBDS gene mutations.

Methods: DNA samples of patients with clinical symptoms (pancreatic insufficiency, marrow dysfunction and/or skeletal abnormalities) and their relatives were extracted from blood leucocytes and analysis of SBDS gene was performed by Sanger Sequencing using QuantumDye sequencing kit (Quantum Seq), DreamTaq PCR MM (Thermo Scientific) and a tailored designed primer on SeqStudio Capillary electrophoresis (Applied Biosystems).

Results: We designed primers for Sanger sequencing of 5 exons of SBDS gene, considering similarity of SBDS to SBDSP, and tested on several healthy individuals, amplification of SBDS happened, but not SBDSP. We have confirmed SDS diagnosis in five children and SBDS carrier status in patient’s relatives. Notably, four patients presented mutations c.258+2T > C and c.183_184delinsCT in exon 2 of the SBDS gene. In one case - deletion in exon 5 (c.673_677del) with c.258+2T > C mutation have been detected. The targeted therapeutic interventions including bone marrow transplantation was started for diagnosed cases.

Conclusion: The direct sequencing of exon 2 of SBDS gene is the first tier of genetic testing for Shwachman-Bodian-Diamond syndrome suspected cases. Identifying the most frequent mutations for the Ukrainian population may simplify and reduce the cost for the SDS genetic diagnosis.

Grants:

Conflict of Interest: None declared

EP09.024 Retrospective evaluation of multiplex ligation dependent probe amplification analysis in alpha thalassemia patients

Emine Göktaş 1, sümeyye şanal1, hüseyin tokgöz2, Ayse Gul Zamani1, selman yıldırım1

1Necmettin Erbakan University, Medical Faculty, Medical Genetics; 2Necmettin Erbakan University, Medical Faculty, Haematology

Objectives: Alpha thalassemia is an autosomal recessive disease characterized by hypochromic microcytic anemia. Eighty-five percent of cases arise from deletions in the HBA1 or HBA2 genes, while the remaining 15 percent result from sequence changes. We aimed to present the results of deletion-duplication analysis performed using the Multiplex Ligation-dependent Probe Amplification (MLPA) method in patients diagnosed with alpha thalassemia.

Methods: We examined variations in alpha-globin gene copy numbers determined by MLPA in 29 alpha thalassemia patients(16 females, 13 males). We correlated the genotypes and phenotypes by analyzing the hemogram parameters and hemoglobin electrophoresis findings of patients with detected deletions in MLPA.

Results: Deletions were detected in 15 (52%) of the patients presenting with a preliminary diagnosis of alpha thalassemia. Among cases with detected deletions, 11 (7 females, 4 males) showed a deletion in one α-globin gene copy, while 4 (3 females, 1 male) exhibited deletions in both copies of the α-globin gene. The most common deletion observed in patients was -α3.7 (60%), followed by -α20.5 (33%) and –αMED-1 (13%). Biallelic observation of different forms of the -α3.7 deletion [-a3.7(A)/-a3.7(D), -a3.7(D)/-a3.7(F)] was noted in two cases. Additionally, one case showed biallelic -a3.7(D) deletion along with (-a)20.5 biallelic deletion, and in another case, besides -a3.7(D) deletion, a pathogenic variant p.Gly60Asp was detected in the HBA1 gene through sequence analysis.

Conclusion: MLPA is very effective in the diagnosis of alpha thalassemia. However, in cases where MLPA analysis is normal, sequence analysis of the HBA1 and HBA2 genes should be considered.

Grants: No

Conflict of Interest: None declared

EP09.025 Beyond the Recessive Model: A Look at Heterozygous MEFV Mutations in Familial Mediterranean Fever

Antonia Vardeva 1, Hristo Ivanov1, Hassan Burnusuzov1, Vili Stoyanova1

1Medical University of Plovdiv, Paediatrics and Medical Genetics, Plovdiv, Bulgaria

Background/Objectives: Familial Mediterranean fever (FMF) is a common autoinflammatory genetic disorder characterized by recurrent self-limiting episodes or attacks of fever along with serosal inflammation, with amyloidosis being the most fatal complication. We report four cases with similar symptoms, three children and a 35-year-old woman who presented with additional symptoms of high IgM levels, hair loss, and anemia.

Methods: Target panel of 642 genes was performed on all patients

Results: All patients were found to be heterozygous for different pathogenic variants of MEFV: c.2177T>C, c.2230G>T, c.2082G>A, and c.2080A>G.

Conclusion: Familial Mediterranean Fever (FMF) is primarily caused by recessive mutations in the MEFV gene. These are typically gain-of-function mutations that result in reduced affinity to regulatory proteins and continuous activation of the pyrin inflammasome. Diagnosis of FMF largely relies on clinical findings, but the detection of two pathogenic MEFV mutations is considered confirmatory. However, recent studies have found only one pathogenic MEFV variant in approximately 25% of clinically diagnosed FMF cases. To understand the correlation between phenotype and genotype, factors such as complex alleles, modifier loci, genetic heterogeneity, and potential epigenetic influences should be considered. Therefore, future management of patients and their families should be based on both clinical evaluation and genetic counseling. In conclusion, the presence of heterozygous MEFV mutations can also lead to FMF symptoms. This highlights the importance of genetic counseling in managing such cases.

Conflict of Interest: None declared

EP09.026 Immunogenomics solves missing heritability and ends the diagnostic odyssey in RAG1-SCID

Marieke De Bruyne 1;2, Lore Pottie1;2, Stijn Van de Sompele1;2, Nancy De Cabooter1;2, Delfien Bogaert3;4, Levi Hoste4, Toon Rosseel1;2, Kim De Leeneer1;2;5, Victoria Bordon3, Jana Neirinck6;7, Mattias Hofmans7, Carolien Bonroy6;7, Simon J. Tavernier1;2;4, Kathleen Claes1;2;5, Filomeen Haerynck4;8, Elfride De Baere1;2

1Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium; 2Ghent University, Department of Biomolecular Medicine, Ghent, Belgium; 3Ghent University Hospital, Department of Pediatrics, Division of Pediatric Hemato-Oncology and Stem Cell Transplantation, Ghent, Belgium; 4Ghent University, Department of Internal Medicine and Pediatrics, Primary Immunodeficiency (PID) Research Laboratory, Center for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Center, Ghent, Belgium; 5Cancer Research Institute Ghent (CRIG), Ghent, Belgium; 6Ghent University, Department of Diagnostic Sciences, Ghent, Belgium; 7Ghent University Hospital, Department of Laboratory Medicine, Ghent, Belgium; 8Ghent University Hospital, Department of Pediatrics, Division of Pediatric Pulmonology, Immunology and Infectious Diseases, Ghent, Belgium

Background/Objectives: Severe combined immunodeficiency (SCID) is a life-threatening Inborn Error of Immunity (IEI) due to defective T-cell maturation. The diagnosis is usually established by rapid and highly sensitive immunophenotyping, directing downstream genetic testing. A genetic diagnosis is needed for personalized treatment and genetic counseling. Despite genomic advances, 15% of SCID remains unsolved. We report a consanguineous family with three relatives displaying T-B-NK + SCID, supporting RAG1/RAG2 deficiency but molecularly undiagnosed for several years.

Methods: Flow cytometric immunophenotyping, targeted next-generation sequencing (tNGS) of RAG1 and RAG2 and IEI gene panel-based clinical exome sequencing (CES) was performed in two affected cousins. AutoMap revealed regions of homozygosity (ROH) in the exome data. Copy number variants were evaluated using ExomeDepth. Reads of RAG1 and RAG2 were manually inspected in Integrative Genomics Viewer. Segregation analysis in parents was done using tNGS.

Results: Immunophenotyping showed absence of naïve T-cells and recent thymic emigrants. tNGS and IEI-CES did not reveal pathogenic variants in known SCID genes nor in other IEI genes. AutoMap detected several ROHs, containing the RAG1 and RAG2 genes. Manual inspection of their respective reads in IGV showed a putative complex structural variant (SV) impairing RAG1, homozygous in the cousins and heterozygous in the parents. Long-read sequencing using adaptive sampling and Optical Genome Mapping are pending to precisely delineate and characterize the SV.

Conclusion: Identification of a complex SV disrupting RAG1 ends the diagnostic odyssey in a SCID family, expands the genetic architecture of RAG1-SCID and emphasizes the value of immunogenomics combining flow cytometric immunophenotyping and genomics in standard-of-care diagnostics of SCID.

Grants:

Conflict of Interest: None declared

EP09.027 Phenotype expansion of SAMD9L-associated Ataxia-Pancytopenia Syndrome

Elisabeth Schreiner 1, Sophie Bierbaumer1, Jasmin Blatterer1, Sabrina Hammer1, Stefan Kuehberger1, Heidelis Tichy1, Klaus Wagner1, Sarah Verheyen1

1Medical University of Graz, Diagnostic and Research Institute of Human Genetics, Graz, Austria

Background/Objectives: SAMD9L-associated Ataxia-Pancytopenia Syndrome is a hematologic disorder characterized by cerebellar ataxia, nystagmus, predisposition to bone marrow failure, myelodysplasia and myeloid leukemia. Monosomy 7 can occur in blood cells. To date, less than 100 individuals with a pathogenic variant in SAMD9L have been reported. Dysmorphic facial features have been mentioned (Davidsson et al. 2018), but not described in the literature so far.

Methods: Phenotype based trio exome analysis was performed in a 16 month old girl using the HPO-terms decreased body weight, myelodysplasia and short philtrum. Deep sequencing was used to determine the variant allele frequency (VAF) in blood and buccal mucosa cells. Reverse phenotyping and review of the SAMD9L-associated phenotype in literature was performed.

Results: We present a case of SAMD9L-associated Ataxia-Pancytopenia Syndrome with striking facial features. Our patient carries a de novo pathogenic missense variant [NM_152703.3:c.3100G>T, p.(Asp1034Tyr)] with a VAF of 0.27 in blood and 0.49 in buccal mucosa. Based on our findings, it is a germline variant with reversion mutation in the blood. Failure to thrive was noted in the first months of life. Cytopenia occurred at age 11 months and progressed to refractory pancytopenia accompanied by monosomy 7 mosaicism. Facial features included prominent, deep-set eyes, up-slanting palpebral fissures, straight eyebrows, a short philtrum, down-turned corners of the mouth and a pointed chin.

Conclusion: Our findings suggest a SAMD9L-associated phenotype apart from myelodysplasia. Facial features should be documented in patients with SAMD9L variants to possibly facilitate a clinical diagnosis.

Grants: None.

Conflict of Interest: None declared

EP09.028 Thrombomodulin gene polymorphism differentially influence the development of acute graft versus host disease and outcome of hematopoietic stem cell transplantation

Livia Varga 1;2, Nora Meggyesi1, Andras Bors1, Katalin Balassa2, Viktor Lakatos1, Melinda Paksi1, Laszlo Gopcsa1, Attila Tordai2, Masszi Tamás2, Peter Remenyi1, Hajnalka Andrikovics1

1Central Hospital of Southern Pest (DPC); 2Semmelweis University (SE)

Background/Objectives: Single-nucleotide polymorphisms (SNPs) within the thrombomodulin gene (THBD) of the recipient might contribute to endothelial dysfunction, abnormal reactivity during graft versus host disease (GvHD), influencing mortality after allogeneic hematopoietic stem cell transplantation (HSCT).

Methods: Three SNPs (rs1962, rs1042579, rs1042580) of THBD were genotyped in two cohorts. Cohort “A” consisted of 400 patients [207 sibling/193 HLA-matched donor; 256 myeloablative (MAC)/144 reduced intensity conditioning (RIC); HSCT between 2007-2013] and cohort “B” [n = 320; 88 sibling/164 HLA-matched/68 haploidentical donor; 252 MAC/68 RIC; HSCT between 2015-2019]. SNPs were genotyped by melting curve analysis (LightCycler480II) from DNA taken before HSCT.

Results: In MAC conditioning, THBD homozygosity for any SNP affected overall survival (OS) differently in the two studied cohorts. In cohort “A”, homozygosity for either THBD SNP influenced acute GvHD grades III-IV [8.4% (8/95) in homozygotes vs. 16.7% (51/305) in non-homozygotes, p = 0.048]. Consequently, favourable OS was observed in homozygous patients (60-month-OS: 58.5 ± 5.4% vs. 42.8 ± 3.1%, p = 0.017). In cohort “B”, recipient homozygosity for either THBD SNP did not affect risk of acute GvHD grades III-IV [7.9% (5/63) in homozygotes vs. 8.4% (16/189) in non-homozygotes, p = 0.567]. In cohort “B”, patients homozygous for any THBD SNP had unfavourable outcome (60-month-OS: 51.9 ± 4.1% vs. 20.1 ± 6.0%, p = 0.000). In both cohorts THBD homozygosity did not influenced survival in RIC.

Conclusion: Our results suggest that recipient’s THBD homozygous genotypes have altered effect in different HSCT settings. Our finding might be explained by different GvHD prophylaxis in myeloablative conditioning. THBD homozygosity may be a predictive biomarker for HSCT outcome.

Conflict of Interest: None declared

EP09.029 miR-423-5p and miR-4508 are downregulated in asymptomatic and mild symptomatic COVID-19 cases

Omar García-Pérez1, Malena Gajate-Arenas1, Angélica Domínguez-de-Barros1, Candela Sirvent-Blanco1, Alma García-Ramos1, José E Piñero1;2;3, Jacob Lorenzo-Morales1;2;3, Elizabeth Córdoba-Lanús 1;2

1Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC). Universidad de La Laguna, San Cristóbal de La Laguna; 2Consorcio Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas (CIBERINFEC). Instituto de Salud Carlos III, Madrid, Spain; 3Departamento de Obstetricia y Ginecología. Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología. Facultad de Ciencias de la Salud. Universidad de La Laguna, San Cristóbal de La Laguna, Spain

Background/Objetives: Host-encoded miRNAs plays an important role in the pathogenesis of viral infections. These miRNAs can activate the cytokine storm associated SARS-CoV-2 infection and might be used as biomarkers for COVID-19 disease. The advance of new therapies makes the study of miRNAs involved in the development of COVID-19 of crucial importance. The aim of the present study was to determine a potential circulating miRNAs profile to be altered in asymptomatic/mild COVID-19 cases in serum biological samples.

Methods: Circulating miRNAs from serum belonging to 40 asymptomatic or mild symptomatic COVID-19 patients and 29 age and gender-matched healthy controls were determined by NGS and then validated by qPCR. miRNAs were isolated and sequenced by NGS technology. In the validation step miRNAs expression was determined by qPCR. and calculated by ΔΔCt method. In silico analysis were performed for target prediction.

Results: Eighteen miRNAs resulted dysregulated in the screening step (p < 0.001; FDR < 0.0001). Among these, four miRNAs were validated as downregulated in asymptomatic/mild COVID-19 cases with respect to controls: miR-423-5p and miR-4508 (Log2FC = -2.59 and -2.7; p < 0.001, respectively) followed by miR-21-5p and miR-1246 (Log2FC = -1.94 and -1.1; p = 0.011 and p < 0.001, respectively). In silico analysis indicated miR-1246 to target ACE2 via TMPRSS2-binding; miR-4508 and miR-21 to target MAPK in JAK/STAT central inflammation via, while miR-423-5p in the immune response may act on type I IFN signalling.

Conclusion: miR-423-5p and miR-4508 were found to be downregulated in the asymptomatic or mild cases infected with SARS-CoV-2. Functional analyses are needed to confirm these findings.

Grants: CIBERINFEC-CB21/13/00100 and PI-CC202302222, Cabildo.23.

Conflict of Interest: Omar García-Pérez Financiado por el Cabildo Insular de Tenerife 2023-2028, PROYECTO CC20230222, CABILDO.23, Malena Gajate-Arenas: None declared, Angélica Domínguez-de-Barros: None declared, Candela Sirvent-Blanco: None declared, Alma García-Ramos: None declared, José E Piñero: None declared, Jacob Lorenzo-Morales: None declared, Elizabeth Córdoba-Lanús CIBERINFEC-CB21/13/00100

EP09.030 Assesment of Chromosome fragility test in southern Tunisian patients with fanconi anemia : Experience of our Medical Genetics department

Fatma Turki 1, hassen kamoun1, Rim Frikha1

1Hedi Chaker Hospital, Medical Genetic Department, Sfax

Fanconi anemia (FA) is a rare autosomal recessive disease characterized by bone marrow failure and increased predisposition to malignancy. Its frequency is important due to the high rate of consanguineous unions in our country. The complexity of FA’s clinical profile can lead to a diagnosis’ delay, which has a direct impact on the therapeutic management of these patients. Chromosome fragility test was the main reference test which reveals chromosomal breaks.

The aim of our study is to report the cytogenetic analysis’results of patients referred to our department for FA suspicion.

Karyotyping with Mitomycin C (MMC) was performed in 13 patients. MMC treated and MMC-untreated peripheral lymphocyte cultures from patients and controls were prepared in the same conditions. 50 and 25 metaphases selected randomly were analyzed from respectively the culture with and without MMC. For the controls, 50 metaphases were analysed from the culture with MMC. FA was confirmed when the percentage of aberrant cells is >65%. The diagnosis of FA is eliminated if this percentage is less than 30%.

5 out of 13 patients were tested positive for FA with a mean pourcentage of MMC induced aberrant cells equal to 81%. These patients presented hematological abnormalities, physical and or skeletal deformities, Fanconi-like facies and delayed physical development. The FA’s diagnosis was exluded in the remaining patients, with an average percentage of chromosomal breaks equal to 22%.

Our results emphasize the importance of conducting an early diagnostic test in FA patients and their potential sibling donors and an appropriate monitoring for physical abnormalities and related cancers.

Conflict of Interest: None declared

EP09.031 Unusual presentation of DCLRE1C gene-related SCID: A case report of challenging molecular diagnosis

Marija Rozevska 1;2, Viktorija Arnite2, Gita Taurina2, Ineta Grantina1;2, Inga Nartisa1;2, Dmitrijs Rots1;2, Natalja Kurjane1;2

1Riga Stradins University, Rīga, Latvia; 2Children’s Clinical University Hospital, Rīga, Latvia

Background. Severe combined immunodeficiency (SCID) is a critical genetic condition that hampers immune system development, often fatal if not diagnosed and treated promptly. Infants with SCID require urgent hematopoietic stem cell transplantation, with the best outcomes seen when diagnosis and treatment occur before 3.5 months of age.

This report outlines the case of a 1.5-year-old boy with SCID who exhibited unusual symptoms after receiving the BCG vaccine at 2 months. Despite an atypical immunological profile with the presence of CD3+ and CD4+ cells, the absence of CD8+ cells hinted at a potential primary immunodeficiency.

Methods. Next-generation sequencing of Primary Immunodeficiency gene panel revealed a deletion of DCLRE1C gene exons 1-3 was identified. The deletion has 0.2 reads ratio, which is borderline value between homozygous and heterozygous deletion. However, QC revealed ~15% cell contamination (with female sample). The contamination was identified also from freshly extracted blood sample on chromosomal microarray and confirmed to be of maternal origin by QF-PCR. The deletion was confirmed to be homozygous in child, confirming the SCID diagnosis. Karyotype culture failed to grow, precluding karyotyping - a phenomenon often observed in ‘null’ variants of DCLRE1C gene.

Results: These findings confirmed the diagnosis of DCLRE1C gene-related SCID. The homozygous deletion could be missed or misinterpreted as heterozygous as the sample was “contaminated” with maternal cell line.

Conclusion: This case underscores critical importance of newborn screening for such conditions, a practice initiated in Latvia as of April 1, 2023. Understanding SCID biology and genetics is important for correct diagnosis.

Conflict of Interest: None declared

EP09.032 Detection of CALR gene mutations in JAK2V617F and MPL unmutated chronic myeloproliferative neoplasms

Milica Strnad 1, Biljana Todorić-Živanović1, Lavinika Atanaskovic2, Marija Elez2

1Military Medical Academy, Institute of Pathology and Forensic Medicine, Belgrade, Serbia; 2Military Medical Academy, Clinic of Hematology

Background/Objectives: Mutations in the CALR gene, which encodes the calcium-regulating protein calreticulin, are the second most common mutations (after JAK2V617F) associate with myeloproliferative neoplasms (MPNs), essential thrombocythaemia (ET) and primary myelofibrosis (PMF). CALR mutations are detected in about 48-68 % of patients with ET or PMF with wild-type alleles of JAK2V617F (Janus kinase 2) and MPL (myeloproliferative leukemia virus oncogene).

Methods: In this study, we selected a group of 20 JAK2V617F and MPL unmutated patients with MPNs (15 ET, 5 PMF) previously analyzed, in our laboratory. Genomic DNA was isolated from Na-citrate-treated blood using DNA extraction kits. Mutations of CALR (Type I, Type II, Del I,Del II and Del lII) were determined by Real-Time PCR analysis (mutation screening kit)

Results: CALR mutations were found to be positive in 10 (50%) cases.The most frequent mutations were Type I and Del I (60%). Del II mutation was not detected. In eight patients (80% of patients with mutations) were detected more than one mutation.

Conclusion: The World Health organization (WHO) embedded CALR and MPL genes mutation, combined with previously described mutations in JAK2V617F gene, in diagnostic criteria for PMF and ET, since 2016. In addition to helping diagnose MPNs, CALR mutation testing can provide information about a prognosis. Also, expression of the mutant protein on the cell surface have a great potential as targets for molecular-targeted drugs and immunotherapy.

Grants:

Conflict of Interest: None declared

EP10 Intellectual Disability

EP10.001 De novo deep intronic variant in the GATAD2B gene in two brothers with GAND syndrome

Olga Levchenko 1

1Research Centre for Medical Genetics, Moscow, Russian Federation

Background/Objectives: GATAD2B-associated neurodevelopmental disorder (GAND syndrome, MIM:615074) results from pathogenic variants in the GATAD2B gene, an important player in transcription regulation. This study focuses on a non-consanguineous family with two affected brothers presenting a heterozygous variant deep within an intronic region of the GATAD2B.

Methods: Whole genome sequencing (WGS) was performed on the trio. FastQ files was obtained from LLC «Biotech Campus».

Results: Two male siblings, aged 10 and 5 years, exhibited developmental delays with delayed motor skills and limited verbal communication. Despite the absence of complications during pregnancy and childbirth, both brothers displayed a distinct clinical phenotype, including an asthenic build, scoliosis, broad forehead, broad nasal tip, thin upper vermilion, and a cleft chin. The older brother additionally exhibited wing-shaped shoulder blades and protruding front incisors, while the younger brother presented with pectus excavatum. They are friendly and communicate with each other in their own language. WGS analysis identified a de novo variant (NM_020699.4:c.1217-399C > T) in the GATAD2B gene, subsequently confirmed by Sanger sequencing in both siblings, with the absence of the variant in parents. The SpliceAI tool predicted a high likelihood of forming a new splice acceptor site (Δ score 0.74). Face2Gene showed a medium level of agreement with GAND syndrome. Variant classified as likely pathogenic (PS2, PM2, PP3). Furthermore, the gonadal mosaicism is the most likely for this family.

Conclusion: This study expands the spectrum of GAND syndrome to include cases associated with deep intronic variant. This finding underscores the importance of WGS in elucidating the genetic basis of complex neurodevelopmental disorders.

Conflict of Interest: None declared

EP10.002 The end of a 20-year genetic odyssey: identification and characterization of a near coding variant associated with leaky mis-splicing inNAA10

Elsa Leitão 1, Sabine Kaya,1, Anne Faudet2, Frédéric Tran Mau-Them3;4, Isabelle An5, Perrine Charles2, Delphine Heron2, Christel Depienne1

1Institute of Human Genetics, University Hospital Essen, University Duisburg-Essen, Essen, Germany; 2Department of Clinical Genetics, Reference Center for Rare Diseases « Intellectual disabilities of rare causes » Déficiences, APHP Sorbonne Université, University Hospital Pitié Salpêtrière, Paris, France; 3UF “Innovation diagnostique dans les maladies rares”, CHU Dijon, Dijon, France; 4Inserm-UB-UMR1231 GAD, Dijon, France; 5Epileptology Unit and Reference Center of Rare Epilepsies, APHP Sorbonne Université, University Hospital Pitié Salpêtrière, Paris, France

Background/Objectives: Approximately half of patients with neurodevelopmental disorders are currently left without a genetic diagnosis after multiple genetic tests, including exome sequencing. Several reasons may explain the failure to detect pathogenic variants, including the presence of variants with atypical effects in known genes.

Methods: Here, we reanalyzed the data of a family with two male siblings who both had developmental and epileptic encephalopathy, intellectual disability, autism spectrum disorders and movement disorders. Previous inconclusive genetic analyses included quartet exome sequencing and a trio genome analysis combined with RNA sequencing in blood.

Results: We identified a hemizygous cytosine-to-adenine substitution in the polypyrimidine tract in position -9 of the acceptor splice site of exon 6 in NAA10. This variant was prioritised because of possible phenotypic match and X-linked inheritance. We provide evidence that this substitution results in leaky mis-splicing, i.e. both a normal and an aberrantly spliced isoform lacking the in-frame exon 6 are transcribed from the mutant allele. The variant was inherited from the heterozygous mother but was absent from the maternal grandmother. As the grandfather was deceased, we analysed available maternal sisters and showed that the variant arose de novo on the grandmother’s haplotype.

Conclusion: In conclusion, we have identified and characterised a variant with atypical effects in NAA10 that we were able to classify as likely pathogenic. The identification of this variant finally ended a 20-year genetic odyssey in this family. This example illustrates the need to consider intronic variants that may alter splicing in known genes that fit the patient’s phenotype.

Grants: UK Essen

Conflict of Interest: None declared

EP10.003 Novel OGT gene variant leading to OGT-associated congenital disorder of glycosylation syndrome

Júlia Krisztina Erhardt 1, Krisztina Nemeth1, Éva Pállinger2, Gyorgy Fekete1, Árpád Ferenc Kovács1

1Semmelweis University, Pediatric Center Tűzoltó street Department, Budapest, Hungary; 2Semmelweis University, Department of Genetics, Cell- and Immunobiology, Budapest, Hungary

Background/Objectives: OGT gene encodes the UDP-N-acetylglucosamine-peptide N-acetylglucosaminyltransferase (OGT) protein, which is responsible for the O-linked glycosylation of several proteins in a plethora of cells, including neuronal cells. In recent years pathogenic OGT variants were linked with intellectual, motor and language developmental disorders, resulting in OGT-associated intellectual developmental disorder (MIM: 300255). The reported pathogenic variants in OGT are mostly missense variants affecting the TPR and CAT domains.

Methods: Two-year-old male patient was evaluated for delayed development and multiple minor anomalies. Genetic anamnesis, 5 generation pedigree and minor anomaly mapping were performed during pre-test counselling. Diagnostic testing included arrayCGH and exome sequencing. Segregation analysis was performed for parents.

Results: Genetic anamnesis revealed neonatal hypotonia, delayed motor and speech development. Minor anomaly mapping revealed convergent strabismus, high palate, widely spaced teeth, low set ears and microcephaly. ArrayCGH did not reveal any pathogenic CNV. Exome sequencing resulted in two variants of unknown significance: heterozygous KAT6A:c.302A>G and hemizygous OGT:c.1900C>T. KAT6A variant in silico analysis yielded a benign prediction. The identified missense variant in OGT was pathogenic according to in silico analysis and healthy parents were not carrying this variant. The variant alters the CAT domain of OGT, and variants affecting CAT domain have been classified as pathogenic. In silico OGT elicited T-cell dysfunction was evaluated.

Conclusion: Based on the molecular testing, patient phenotype and in silico analysis we propose, that OGT:c.1900C>T may be a pathogenic variant in OGT-associated congenital disorder of glycosylation.

Grants: Hungarian National Research, Development and Innovation Office – NKFIH, OTKA PD_21 138521.

Conflict of Interest: None declared

EP10.005 Co-occurrence of Temple-Baraitser/Zimmerman-Labland syndrome and Myhre syndrome in one patient

Katarzyna Połatyńska 1, Tadeusz Kałużewski2, Agnieszka Gach2, Łukasz Kępczyński2

1Polish Mother’s Memorial Hospital Research Institute, Department of Child Neurology and Epileptology, Łódź, Poland; 2Polish Mother’s Memorial Hospital Research Institute, Department of Genetics, Łódź, Poland

Background/Objectives: KCNH1 gene encodes a voltage-gated potassium channel Kv10.1 expressed in central nervous system and is essential for brain development. De novo missense gain-of-function variants in KCNH1 gene were associated with two distinct syndromic encephalopathies – Temple-Baraitser syndrome (TMBTS) and Zimmerman-Labland syndrome type 1 (ZLS1), as well as non-syndromic epileptic encephalopathy or isolated epilepsy. SMAD4 gene encodes a member of signaling cascade; de novo gain-of-function missense variants affecting Ile500 residue, were associated with Myhre syndrome.

Methods: A whole exome sequencing was performed for a 3 year-old female proband presenting epilepsy with generalized seizures of early onset, microsomia, global developmental delay, and facial dysmorphism, resembling Myhre syndrome.

Results: A heterozygous, previously reported, pathogenic variant of SMAD4 c.1498A>G, affecting Ile500 residue was identified. This was consistent with suspected Myhre syndrome. Additionally, a heterozygous, previously reported variant of KCNH1 c.1070G>A, affecting the hot-spot residue Arg357 was identified. This was consistent with KCNH1-dependant encephalopathy diagnosis.

Conclusion: ZLS1 and TMBTS represent recognizable syndromes, KCNH1-activating mutation carriers may also present phenotypes which cannot be attributed to both entities. ZLS1 and TMBTS can be regarded as specific examples of wider clinical spectrum. Although our patient presented indistinct features of KCNH1 disorder, it was not suspected because of the clear of Myhre-like appearance. Nevertheless epilepsy is highly uncommon observation in SMAD4-Myhre syndrome. Despite the complexity of clinical picture, whole exome sequencing enabled simultaneously diagnosis of the two entities co-occurring in one patient.

Grants: Internal grant no 18/GW

Conflict of Interest: None declared

EP10.006 New truncating variant in WDFY3 gene associated with neurodevelopmental disorder and macrocephaly

María Juliana Ballesta Martínez 1, María José Sánchez Soler1, Ana Teresa Serrano Antón1, Marta Domínguez Jiménez1, Vanessa Lopez1, Encarna Guillén-Navarro1

1Hospital Clinico Universitario Virgen de la Arrixaca, Sección Genética Médica, Murcia, Spain

Background/Objectives: WDFY3 gene has been associated with neurodevelopmental disorders (NDD). In 2016 a missense variant located in the PH-domain of WDFY3 gene was linked to moderate intellectual disability and microcephaly due to a dominant negative effect and upregulation of canonical Wnt-pathway. Posteriorly in 2019 truncating variants in WDFY3 gene were associated to neurodevelopmental disorders and macrocephaly suggesting that loss-of-function variants lead to macrocephaly via downregulation of the Wnt pathway. We report on a new case with a previously not described pathogenic truncating variant in WDFY3 which further supports its implication in NDD, and delineate the phenotype.

Methods / Clinical description: 8 year-old boy, first child of healthy non-consanguineous parents. Normal pregnancy and delivery, no feeding or medical issues. Patient had mild intellectual disability (IQ 63), mild language delay, relative macrocephaly (OFC 81th centile, +0.9 SD), coarse facial features, tall forehead, arched, wide and sparse eyebrows, hipertelorism with wide palpebral fissures. Long neck with downsloping shoulders, wide hands with redundant skin, brachydactyly and persistent fetal pads. Clinical diagnostic suspicion was Kabuki-like syndrome (KMT2D, KDM6A, CDK13 genes). Abdominal and cardiac sonogram were normal.

Results: Array-CGH and fragile X were normal, and clinical exome sequencing detected a previously not reported de novo variant c.1906C>T; p.Arg636* in exon 14 of WDFY3 gene (NM_014991.4).

Conclusion: 1- We report on a new case of truncating variant in WDFY3 gene supporting its implication in NDD and macrocephaly. 2- Previous published cases don’t describe patient dysmorphism which is evident in our patient. 4-Achieving molecular diagnosis with NGS permits adequate genetic counseling in families.

Conflict of Interest: None declared

EP10.007 A novel splicing variant of KDM6A in a family with Kabuki Syndrome 2

Bilgen Bilge Geçkinli 1, aysenur ersoy1, onur hanoglu1, Vedat Yuce1, mehmet ali soylemez1, Ahmet Arman1

1Marmara University Faculty of Medicine, Department of Medical genetics, Istanbul, Türkyie

Background: Kabuki Syndrome 2 (#300867) is an X- linked dominant inherited disorder caused by mutations in the KDM6A gene. Intellectual disability is accompanied by distinct dysmorphic features. KDM6A encodes a tetratricopeptide repeat (TPR) protein which catalyzes the demethylation of tri/dimethylated histone H31.

Methods: DNA was isolated from the patient’s peripheral blood and Clinical Exome Sequencing (CES) was performed via next-generation sequencing (Illumina NextSeq 500).

Results:The patient is a 2-year- old male who was referred to our clinic for dysmorphism, epilepsy, hydrocephalus and hypothyroidism. He was born after an IVF pregnancy via cesarean section at 28th weeks due to decreased fetal movements. His neuromotor development was behind his age. He had eye slippage on his left eye. He had long palpebral fissures, sparse lateral eyebrows, short columella, broad tip of the nose, high arched palate, abnormal dentition and persistent fetal fingertip pads. His height was 25-50P, weight was <3P and head circumference was 10-25P. He was treated with double antiepileptics due to EEG abnormalities and subtle seizures. He was hemizygous for a novel KDM6A (NM_001291415) c.974_974+2del variant in the donor splice site of exon 11. According to the ACMG criterias it was likely pathogenic. The patient inherited the variant from his mother. His sister also had suggestive phenotype and segregation analysis is planned.

Conclusion:The majority of patients with Kabuki syndrome have heterozygous mutations in KMT2D gene. In our patient, functional studies are planned for a probable exon skipping leading to a frameshift.

Conflict of Interest: None declared

EP10.008 De novo loss of function variants of TCF20 gene in two individuals with postnatal overgrowth and intellectual disability.

Klaudia Berk 1, Renata Posmyk1

1Medical University of Białystok, Clinical Genetics, Białystok

Background: Overgrowth syndromes are a genetically heterogeneous group of rare disorders manifested by excessive tissue development accompanied by neurological anomalies such as intellectual disability (ID). A few de novo variants potentially disrupting TCF20 function in patients with overgrowth and ID have been reported.

Methods: Presented patients (P1, P2) were referred to the clinical genetics unit due to mild ID. Both were born at term with normal measurements without congenital anomalies and had muscular hypotonia in early childhood. At the age of 14 years, P1 displayed postnatal tall stature (height: 183 cm, >97 pc, weight: 63 kg, 75c, OFC: 54 cm,75c), scoliosis, and minor findings such as thick lips, and single transverse palmar crease. Physical examination of female P2 at the age of 13 years revealed a macrocephalic girl of tall stature (height: 169 cm, 97 pc, weight 54 kg: 75 pc; OFC: 57 cm >97c) with small anomalies included subtle facial dysmorphism, scoliosis, joint hypermobility, and finger and toe contractures.

Results: WES tests revealed a de novo stop gain NM_001378418.1, c.2594C>G variant (P1) and frameshift NM_005650.4 c.3119delC (P2) variant in TCF20 gene. Moreover, aCGH identified benign duplication arr[hg19]7p14.3 (31960509_32604726)x3 in P2.

Conclusion: WES is a powerful tool for detecting causative variants in patients with „overgrowth” without a characteristic dysmorphic pattern. Presented cases expand the clinical phenotype of patients with de novo variants in TCF20 gene, particularly regarding growth disturbances.

Grants:This work was funded by the National Science Center [grant number NCN/1/DA/21/001/1106].

Conflict of Interest: None declared

EP10.009 A novel case with biallelic pathogenetic PTRHD1 variants leading to intellectual disability

Özlem Baysal 1, Jikke-Mien Niermeijer2, Rolph Pfundt1;3, Bert de Vries1;3

1Radboud University Medical Center, Human Genetics, Nijmegen, Netherlands; 2St. Elisabeth Hospital, Neurology, Tilburg, Netherlands; 3Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, Netherlands

Background/Objectives: Biallelic pathogenetic variants in PTRHD1 (OMIM*617342) have been described as causative for autosomal recessive intellectual disability and parkinsonism. To date only a limited number of patients have been reported so far. We aim to extend our clinical knowledge about the consequences of variants in this gene by presenting a novel patient.

Methods: We describe the clinical and genetic findings of a novel patient with biallelic variants in PTRHD1.

Results: Our patient is a female, born to consanguineous parents of Syrian descent. She was diagnosed with a developmental delay at the age of 4 years old. An MRI scan showed no brain abnormalities. She was referred to our genetics clinic at the age of 8 years old. During physical examination, some mild facial dysmorphisms were noticed such as arched eyebrows with synophrys, an upward slant of the palpebral fissures, a short philtrum and prominent teeth. There was joint hypermobility but no further signs of neurological abnormalities.

By using trio whole exome sequencing, a homozygous frameshift variant was found (NM_001013663.1:c.280del p.(Leu94fs). This variant was novel, but likely causative for her developmental delay. No other possible causative SNVs or CNVs were detected.

Conclusion: This case report provides further evidence for PTRHD1 being a novel intellectual disability gene. Furthermore, it provides clinical data which can be used to delineate the PTRHD1 related phenotype. Further international collaboration is needed to collect clinical data of the novel cases in order to establish the clinical spectrum.

Grants: None

Conflict of Interest: None declared

EP10.010 Diagnostic yield of WES in different groups of patients with intellectual disability

Dmytro Mykytenko 1;2

1Mother and Child Genetic Center, Clinical dept, Kyiv, Ukraine; 2Academician Yury Bugai International Scientific and Technical University, Kyiv, Ukraine

Background/Objectives: The diagnostic strategy for Intellectual disability (ID) remains imperfect and has internal contradictions. Many doctors and patients mistakenly believe that NGS-based tests are a reliable universal approach. The purpose of the study was to determine the diagnostic yield of the WES-SOLO for ID case evaluation.

Methods: Patients (3-6 yrs), with ID, who underwent diagnostics by WES + CNV+Mitochondrial DNA-Solo (2019-2023) (after karyotyping) were retrospectively divided into 2 groups without (353 cases) and with (163 cases) markers of genetic pathology (congenital malformations, congenital metabolic defects, multiple minor developmental anomalies). Statistical evaluation was carried out by Chi-square method.

Results:Dependence of the diagnostic yield on the clinical situation is shown in the table.

Parameter

Markers of genetic pathology

P-index

present

absent

abs

%

abs

%

Negative

84

51,5

219

62,2

0.02

VUS

28

17,2

51

14,4

0,42

Positive:

51

31,3

83

23,4

0,06

including:

- CNV

10

12

- monogenic

41

67

(2+ genes)

2

4

- mitochondrial

2

TOTAL

163

100

353

100

-

Positive+VUS

79

48,5

134

37,8

0,02

missed

1 FMR1 case

0,6

2 FMR1 cases

0,9

-

Conclusion: NGS-based methods are characterized by a significantly reduced diagnostic yield in the absence of markers of genetic pathology. Given the missed cases, the NGS-based methods cannot be considered an independent diagnostic approach. The necessity of genetic diagnostics for ID case evaluation should be determined individually, considering the likelihood of the impact of the diagnostic result on the treatment scheme modification, possible inclusion into clinical trials, and the planning of the next pregnancy.

Grants: -

Conflict of Interest: None declared

EP10.011 NGS data interpretation gap: about a deep intronic variant in MBD5 gene

Salwa Ben Yahia 1, Ahlem Achour1, Syrine Adhoum1, Amelie PITON2, faouzi maazoul1, jamel chelly2, ridha mrad1, Mediha Trabelsi1

1Charles Nicolle Hospital, Department of Congenital and Hereditary Diseases, Tunis, Tunisia; 2New Civil Hospital, Genetic Diagnostic Laboratory (UF1420), Strasbourg, France

Background: Syndromic Intellectual Disability (SID) is defined by the association of an intellectual disability (ID) with other anomalies such as facial dysmorphia, neurological signs or behavioral disorders. NGS is now part of the arsenal of available diagnostic techniques of SID.

We aim to illustrate NGS data interpretation difficulties by reporting the case of a new variant in MBD5 gene.

Methods: Gene panel sequencing (GPS) including 556 ID genes was performed in a patient referred to our department for psychomotor retardation (PMR) and ID. We interpreted variants using ACMG criteria

Observation: A 3.5 years old girl was referred for PMR and ID. She was born to unrelated parents and had no particular family history. She had facial dysmorphia, autism spectrum disorders and a severe ID.

GPS revealed the (NM_018328.4) c.2845+234A > G variant in heterozygous state in MBD5 gene that is linked to a neurodevelopmental disorder. This deep intronic variant has never been reported and was not found in the gnomAD database (PM2). It is predicted to create a new cryptic accepting splice site although it is far from the canonical splice sites (SliceSiteFinder-like, MaxEntScan, NNSPLICE, GeneSplicer) (PP3). As MBD5 gene has two different transcripts, this variant can either be deep intronic (c.2845+234A > G) (NM_018328.4) or exonic (c.3079A>G) (NM_001378120.1). In both cases, this variant is classified as of uncertain significance.

Conclusion: As MBD5 haploinsufficiency is characterized by ID, speech impairment, seizures, sleep disturbances, and abnormal behavior, our patient’s phenotype is non-specific. Thus, a complementary WES remains necessary to exclude the implication of other genes.

Conflict of Interest: None declared

EP10.012 A rare type of genetic intellectual developmental disorder overlapping with Noonan syndrome type 5 diagnosed by WES

Delia Sabau 1;2, Antoanela Curici2;3, Mihai Mitroi2

1Carol Davila University of Medicine and Pharmacy, Discipline of Medical Genetics, Bucharest, Romania; 2Synevo, Department of Medical Genetics, Chiajna, Romania; 3Carol Davila University of Medicine and Pharmacy, Discipline - Cellular, Molecular Biology and Histology, Bucharest, Romania

Background/Objectives: This study presents a 25-year-old male patient referred to our clinic for a genetic consultation due to his clinical manifestations such as facial dysmorphism, moderate intellectual developmental disorder with psychiatric components and social difficulties, left cerebellar hygroma with left cerebellar hypoplasia and insomnia.

Methods: Throughout the genetic consultation, we examined the patient’s phenotypical traits and symptoms, meticulously constructed the family tree, and delved into the intricacies of his personal medical history. The assessment of the patient’s symptoms guided the recommendation for Whole Exome Sequencing (WES) testing to uncover a potential underlying genetic cause.

Results: WES testing returned positive for two heterozygous variants: a pathogenic variant, ZMYND11: NM_006624.7:c:744_745del p.(Cys249Serfs*2) and a likely pathogenic variant, RAF1: NM_002880.4:c.1426C>T p.(Leu476Phe). The coexistence of these variants is correlated with the patient’s distinct phenotype, thus making possible the diagnosis of both Intellectual developmental disorder, autosomal dominant 30 (MIM# 616083) and Noonan syndrome 5 (MIM# 611553).

Conclusion: The association between the two variants discovered is considered to have resulted in this patient’s particular phenotype with intellectual development disorder and particular Noonan phenotype. Overall, this study emphasizes the importance of genetic consultations and advanced genomic technologies in elucidating the genetic basis of complex disorders, offering crucial information for accurate diagnosis and personalised management of the patients. This case also highlights the utility of genetic testing, specifically WES, in unraveling complex phenotypes and facilitating a precise diagnosis.

Grants: None

Conflict of Interest: None declared

EP10.013 Identification of a new variant in KIDINS220 gene for SINO diagnosis

Paula Muñiz Sevilla1, alba navarro romero 1, Marián Lázaro1, Laura Rausell1, Pablo Gargallo1, Susana García-Linares2

1Health in Code, Valencia, Spain; 2Hospital Universitario Clínico San Cecilio, Granada, Spain

Background/Objectives: Spastic paraplegia, intellectual disability, nystagmus, and obesity (SINO) (OMIM#617296) is an autosomal dominant neurologic disorder characterized by rapid growth, global developmental delay, spastic paraplegia, ophthalmologic defects, and dysmorphic facial features. Pathogenic variants in the KIDINS220 gene are associated with SINO. To date, it has not been possible to define the phenotypic spectrum related to the gene because there are a reduced number of patients described. In this study we identified a novel nonsense mutation in KIDINS220 that has allowed us to diagnose SINO syndrome.

Methods: We carried out a whole exome of a 5-year-old female patient with corpus callosum aplasia, strabismus, paraplegia and tall stature. For the variant analysis, a filtering according to phenotypic characteristics was carried out. The classification of variants was performed according to the guidelines of the American College of Medical Genetics (ACMG).

Results: We detected the variant NM_020738.2: c.4347del (p.L1451*) in heterozygosis in KIDINS220, that lead to premature stop codon in the last exon. This variant does not appear in population or clinical databases. We performed segregation in parents, confirming it was a de novo variant. The ACMG classification of the variant was Likely pathogenic (PVS1 strong, PM6 supporting and PM2 supporting).

Conclusion: We have identified a likely pathogenic variant within the KIDINS220 gene. Because the clinical phenotype observed in the patient was compatible with that previously described in the literature, the diagnosis of SINO was established. To the best of our knowledge, this would be the fifth patient described with this rare entity.

Conflict of Interest: None declared

EP10.014 Incomplete penetrance and highly variable X-linked neurodevelopmental phenotype associated to a truncating variant in CNKSR2

Virginia Garcia-Solaesa 1, aranzazu perez-juana1, Mercè Artigas López1, Maria Ordóñez-Marina1, amaya bengoa alonso1, Fermin Garcia-Amigot1, Sara Pasalodos-Sanchez1, Lourdes Morales-Garófalo2, Eva Barbon-Alonso3, Maria Antonia Ramos-Arroyo1

1Hospital Universitario de Navarra, Medical Genetics Department, Pamplona; 2Hospital Reina Sofia, Clinical Laboratory, Tudela, Spain; 3Hospital Universitario de Navarra, Clinical Laboratory, Pamplona, Spain

Background: CNKSR2 is a causative gene of an X-linked neurodevelopmental syndrome, characterized by intellectual disability, early-onset seizures, speech delay and attention deficit/ hyperactivity. Among 20 published families with truncating mutations, all hemizygous males presented a severe/moderate phenotype and some heterozygous females showed moderate/mild symptoms (1). We present the first large family segregation analysis of a CNKSR2 truncating variant, showing incomplete penetrance.

Case report: The proband was a 21-week gestation male fetus, carrier of a maternally inherited nonsense variant (c.886C>T) in CNKSR2, identified as an incidental finding of a trio exome sequencing study, done for other diagnostic purpose. We performed familial segregation analysis to assess its pathogenicity.

Results: We studied nine maternal family members, seven males and two females. Variant c.886C>T was identified in two males: the mother´s father, a 72-year old healthy male, and a 16-year old boy, diagnosed of attention deficit disorder and hyperactivity. Female carriers (the mother and two relatives) were asymptomatic. However, one of the mother´s paternal aunts had a history of chronic refractory epilepsy (onset at 15 years) and behavioral difficulties, who died at 83 years. Her carrier status could not be assessed.

Conclusion: Limited knowledge on the pathogenic effect of variants in the CNKSR2 gene represents a great challenge for genetic counselling. Truncating changes may exhibit incomplete penetrance and a highly variable clinical severity in males and females. Additional familiar and gene functional studies should clarify phenotype-genotype correlation.

1. Ito, H.; Nagata, K.-i. Functions of CNKSR2 and Its Association with Neurodevelopmental Disorders. Cells 2022, 11,303.

Conflict of Interest: None declared

EP10.015 Mutation analysis of PAH gene in phenylketonuria patients from North of Iran: Identification of three novel pathogenic variants

Mohammad Reza Mahdavi 1, Atefeh Khoshaein2, Hossein Jalali1, Mohammad Amirzadegan2, Mahan Mahdavi2

1Thalassemia Research Center, Hemoglobinopathies Institute, Mazandaran University of Medical Sciences, Sari, Iran; 2Sinaye Mehr Research Center, Sari,Iran

Background/Objectives: There are more than 1100 different Pathogenic variants in the phenylalanine hydroxylase (PAH) gene that is responsible for phenylketonuria (PKU) diseases and the spectrum of these mutations is varied in different ethnic groups. The aim of the present study was to identify the frequency of pathogenic variants in all 13 exons of PAH gene among patients with PKU in Mazandaran and Golestan provinces in north of Iran.

Methods: Forty unrelated PKU patients from Mazandaran and Golestan provinces were enrolled in the study. Genomic DNA was extracted from leukocytes using Qiagen DNA extraction kit and polymerase chain reaction - restriction fragment length polymorphism, and Sanger sequencing methods were applied to detect the variants.

Results: Twenty one different pathogenic variants were observed among 40 investigated patients. The c.1066-11G > A variant had the highest frequency (27.5%) and c.168+5G > C, c.473G>A, and c.782 G > A variants were other most frequent mutations with the allelic frequencies of 7.5, 5, and 5% respectively. Three novel pathogenic variants including c.773T>G, c.878 T > C, and c.1245del variants were observed among the investigated patients.

Conclusion: The introduction of pathogenic variants in the PAH gene in each ethnic group provide a valuable data regarding the understanding of the disease pathogenesis and can be helpful for the prenatal diagnosis programs.

Grants: Mazandaran University of Meical Sciences

Conflict of Interest: None declared

EP10.016 Hao-Fountain Syndrome: first reports of inherited variants

Diogo Fernandes da Rocha 1, Vera Santos1, Sofia Dória2, Ana Grangeia1;2

1Unidade Local de Saúde São João, Serviço de Genética Médica, Oporto, Portugal; 2Faculdade de Medicina da Universidade do Porto, Departamento de Genética, Oporto, Portugal

Background/Objectives: Hao-Fountain syndrome (HAFOUS) is a rare neurodevelopmental disorder linked to USP7 haploinsufficiency, crucial for endosomal recycling. This syndrome is characterized by global developmental delay, intellectual disability with significant speech delay, behavior abnormalities and dysmorphic facial features.

Methods: First case: A 10-year-old first-born boy presenting with global developmental delay, hypotonia, clubfoot, macrocephaly, increased postnatal weight and height, and aggressive behavior. Additionally, his mother had a speech delay during infancy and later faced learning difficulties. Second case: A 34-year-old pregnant woman with a fetus displaying elevated nuchal translucency that underwent spontaneous abortion at 15 gestational weeks. This woman presented with dysmorphic features, small hands and feet, and history of hypotonia and global developmental delay during infancy and learning difficulties during school years.

Results: In the first case, array-CGH identified a duplication at the Xq11.2 region involving the ZC4H2 gene. While Wieacker-Wolff syndrome was initially presumed, the child’s overgrowth raised doubts as well as the lack of recognition of triplosensitivity as a disease mechanism. Subsequently, trio exome sequencing revealed a maternally inherited pathogenic splicing variant in the USP7 gene (NM_003470.3):c.2047+1G > T. In the second case, array-CGH performed on fetal DNA extracted from chorionic villous sampling revealed a 59kb-deletion at the 16p13.2 region, spanning the USP7 gene, later confirmed as maternally inherited.

Conclusion: For the first time ever, we documented the possibility of inherited USP7 variants within families, challenging the prevailing de novo assumption. We also broadened the spectrum of this syndrome by delineating a subgroup of patients exhibiting an overgrowth phenotype.

Grants: None.

Conflict of Interest: None declared

EP10.017 The first pathogenic variant in the RREB1 gene causing Noonan-like Rasopathy

Olga Shatokhina 1, Fatima Bostanova1, Maria Bulakh1, Oksana Ryzhkova1

1Federal State Budgetary Institution “Research Centre For Medical Genetics”, The Shared Resource Centre (SRC) “Genome”, Moscow, Russian Federation

Objectives: Noonan syndrome, a multisystem disorder within the RASopathies, is caused by dysregulation of the RAS–MAPK pathway. The RREB1 gene, a zinc finger transcription factor in this pathway, has been implicated in 6p terminal deletions, but no direct association with disease had been established. This study reports the first point pathogenic variant in RREB1 linked to Noonan-like RASopathy.

Methods: Genetic testing encompassed whole-genome sequencing (WGS) and Sanger sequencing of the proband, his parents and sibling.

Results: The proband exhibited a novel de novo c.2677del, p.(Ala893Argfs*20) variant in the RREB1 gene, suggestive of haploinsufficiency. Clinical assessment of the 3-year-old proband demonstrated alignment with Noonan-spectrum disorder traits. Comparative analysis with microdeletion cases, including those involving RREB1, elucidated shared clinical traits, such as delayed motor skills, intellectual disability, and facial dysmorphisms and distinctive features, such as blue sclerae and the absence of cardiac anomalies.

Conclusion: This study conclusively establishes RREB1 haploinsufficiency as a primary contributor to a novel Noonan-like RASopathy, emphasizing key clinical features. The findings underscore the imperative for further investigations into the functional consequences of RREB1 mutations. Additionally, they contribute to a comprehensive understanding of Noonan-like RASopathies, providing potential avenues for future advancements in diagnostic and therapeutic approaches.

Conflict of Interest: None declared

EP10.018 a novel variant in asns gene responsible for syndromic intellectual disability and microcephaly: case report and literature review

Behzad Davarnia 1, farzad ahmadabadi2, mohammad jahanpanh1, diana mokhtari1, Sara Arish1

1Iran, Department of Genetics and Pathology, Ardabil University of Medical Sciences, Ardabil, Iran., Iran; 2Iran, Pediatric Neurology Research Center, Pediatric Neurology Department, Mofid Children Hospital, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran., Iran

Background/Objectives: The ASNS (ASNS, MIM 108370) gene variations are responsible for the asparagine synthetase deficiency (ASNSD, MIM 615574), a very rare autosomal recessive disease characterized by some cerebral anomalies. These patients have congenital microcephaly, progressive encephalopathy, severe intellectual disability, and intractable seizures.

Methods: Clinical characteristics of the patient were collected. Exome sequencing was used for the identification of variants. Sanger sequencing was used to confirm the mutation in the target region. The structure of the protein was checked using the DynaMut2 web server.

Results: The proband is an 11-year-old Iranian-Azeri girl with primary microcephaly and severe intellectual disability in a family with consanguineous marriage. The onset of symptoms emerged at her 10-20 th days of life, while refractory epileptic gaze and unilateral tonic-clonic seizures initiated without any provoking factor such as fever. Brain MRI revealed no abnormalities except for brain atrophy. The karyotype was normal. Using exome sequencing, we identified a novel homozygous mutation of thymine to adenine (NM_001673.5:c.538T&gt;A) in the ASNS gene. Both parents had a heterozygous mutation in this location. Subsequently, Sanger sequencing confirmed this variant. We also reviewed clinical manifestations and MRI findings of the previously reported patients.

Conclusion: In the present study, a novel homozygous variant was recognized in the ASNS gene in an Iranian-Azeri girl manifesting typical ASNSD symptoms, particularly intellectual disability, and microcephaly. This study expands the mutation spectrum of ASNSD, and reviews previously reported patients.

Grants: This research received no specific grant, funding, equipment, or supplies from any funding agency in the public, commercial, or not-for-profit sectors.

Conflict of Interest: None declared

EP10.019 Cost of genome analysis in patients with intellectual disabilities: a micro-costing study in a French setting

Charley Robert-Viard 1, Anne-Laure Soilly2, Celine Besse3, anne boland3, Delphine Bacq3, Alexia Benoit4, Hamza Hadj-Abdallah5;6, Benedicte Gerard4, Charlotte Poe7, Ange-Line Bruel7, Patrick Nitschké8, Alban Simon9, Stanislas Lyonnet5;6, Laurence Faivre7;10, Christel Thauvin-Robinet7;10, Sylvie Odent11, Delphine Heron12, Damien Sanlaville13, Thierry Frebourg14;15, Jean Muller4;9;16, Yannis Duffourd7, Marion Bouctot1, Niki Sabour1, Marie-Laure Humbert1, Christelle Delmas17, Jean-François Deleuze3, Francis Guillemin18, Valerie Seror19, Hamza Achit18, Helene Esperou17, Christine Binquet1, Hélène Dollfus9, Catherine Lejeune1

1CHU Dijon Bourgogne, Inserm, Université de Bourgogne, CIC 1432, Module Épidémiologie Clinique, F-21000 Dijon, France; 2CHU Dijon Bourgogne, Délégation à la Recherche Clinique et à l’Innovation, USMR, F-21000 Dijon, France; 3Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), Evry, France; 4Laboratoires de Diagnostic Génétique, Hôpitaux Universitaires de Strasbourg, Institut de Génétique Médicale d’Alsace (IGMA), 67000 Strasbourg, France; 5Fédération de Génétique et Médecine Génomique, Hôpital Necker-Enfants Malades, GHU APHP. Centre-Université Paris Cité, Paris, France; 6Inserm, IHU Imagine—Institut des Maladies Génétiques, Université Paris Cité, Paris, France; 7Inserm, Université Bourgogne-Franche-Comté, UMR1231, équipe GAD, Dijon, France; 8Plateforme Bio-Informatique, Inserm UMR 1163, Institut Imagine, Université Paris Cité, Paris, France; 9Inserm UMRS_1112, Institut de Génétique Médicale d’Alsace, Université de Strasbourg, France et Service de Génétique Médicale Hôpitaux Universitaires de Strasbourg, Strasbourg, France; 10CHU Dijon-Bourgogne, Centres de Référence Maladies Rares « Anomalies du Développement et syndromes malformatif de l’Est », Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Dijon, France; 11Service de Génétique Clinique, Centre de Référence Anomalies du Dévelopment CLAD- Ouest, CNRS, IGDR UMR6290 (Institut de Génétique et Dévelopment de Rennes), ERN ITHACA, Université de Rennes, Rennes, France; 12Unité Fonctionnelle de Génétique Médicale et Centre de Référence « Déficiences Intellectuelles de Causes Rares », APHP Sorbonne Université, Groupe Hospitalier Pitié-Salpêtrière et Hôpital Trousseau, Paris, France; 13Hospices Civils de Lyon, GHE, Service de Génétique, Université Claude Bernard Lyon 1, Lyon, France; 14CHU de Rouen, Service de Génétique, Rouen, France; 15Inserm, UMR1245, Centre de Génomique et de Médecine Personnalisée, Université de Normandie, Rouen, France; 16Unité Fonctionnelle de Bioinformatique Médicale Appliquée au Diagnostic (UF7363), Hôpitaux Universitaires de Strasbourg, Strasbourg, France; 17Inserm, Pôle de Recherche Clinique, Paris, France; 18CIC1433-Epidémiologie Clinique, Centre Hospitalier Régional et Universitaire, Inserm, Université de Lorraine, Nancy, France; 19Aix Marseille Univ, IRD, APHM, SSA, VITROME, IHU-Méditerranée Infection, Marseille, France

Consortium: DEFIDIAG group

Background/Objectives: Intellectual disability (ID) is the most frequent reason for referral to a paediatric genetics centre. In order to help the French health authorities determine tariff for genome sequencing (GS) using solo or trio analysis (GSS/GST), we have estimated the cost per unit for GS diagnostic tests for patients with ID.

Methods: A bottom-up micro-costing approach was used for the year 2021 as part of the French DEFIDIAG pilot study (NCT04154891) from the “Plan France Médecine Génomique 2025”. Data collection and monetary valuation of all the required resources (labour, equipment, disposable materials and reagents, reusable materials) were carried out in collaboration with the GAD team (Inserm UMR 12131), the IGMA (Institut de Génétique Médicale d’Alsace), the Necker Hospital laboratory (AP-HP, Paris), the BIPD (Plateforme de Bioinformatique, Paris) and the Centre National de Recherche en Génomique Humaine (CNRGH). Several sensitivity analyses were performed.

Results: The unit cost per diagnostic analysis was estimated at €2,454 for GSS and €4,908 for GST. The analytical step represented 76.5% of the total cost for GSS and 82.5% for GST. The simultaneous decrease in the price of capture kits and of flowcells (30% and 50% respectively) was associated with reduced unit costs of €2,024 for GSS and €3,619 for GST.

Conclusion: This is the first estimate of the cost of GS in the French context for ID. Cost estimates are heavily influenced by the price of consumables. More generally, the organisation and size of the centres involved and the study period also have a significant impact on the results.

Grants: PFMG 2025

Conflict of Interest: None declared

EP10.021 Identification of causal DNA predispositions in patients with ASD in Slovak population

Michal Konecny 1;2, Gabriela Krasnanska1;2, Silvia Lakatosova3, Lenka Wachsmannova1, Marian Baldovič1;4, Vladimír Eliáš1, Gabriela Bľandová1;5, Gabriela Repiska6

1GHC GENETICS SK ltd., Laboratory of Genomic Medicine, Bratislava, Slovakia; 2Faculty of Natural Sciences, University of St. Cyril and Methodius, Department of Biology, Institute of Biology and Biotechnology, Trnava, Slovakia; 3Comenius University in Bratislava, Faculty of Medicine, Institute of Physiology, Academic Research Centre for Autism, Bratislava; 4Faculty of Natural Sciences, Comenius University, Department of molecular biology, Bratislava, Slovakia; 5Faculty of Medicine, Comenius University, Institute of Medical Biology, Genetics and Clinical Genetics, Bratislava, Slovakia; 6Faculty of Medicine, Comenius University, Institute of Physiology, Academic Research Centre for Autism, Bratislava, Slovakia

Identification of causal DNA variants in patients with ASD in Slovak population

Background: Autism spectrum disorder (ASD) represents a group of etiologically and symptomatologic heterogeneous developmental disorders manifesting with deficits in communication, social interaction, and presence of repetitive stereotype behaviours. The current overall population prevalence in the developed countries is estimated at 1.5%. Several dozens of ASD predisposing genes have been identified, which together represent 10-20% of cases. As with many other psychiatric disorders, advances in DNA technologies that allow genome-wide testing confirmed presence of heterogeneous inheritance.

Methods: In our study, we have analysed 35 samples (21 females, 14 males) with ASD using CES/WES massive parallel sequencing approaches and analysed by a virtual panel of genes, based on the HPO phenotype intellectual disability.

Results: Overall, we have detected the presence of causal DNA variants in 17 patients, representing 48.6% of analysed cohort. In detail, we have identified 17 causal DNA variants (pathogenicity class 4 or 5) in 16 genes. We have also focused on the presence of borderline class 3/4 variants (VUS with potentially pathogenic predictions), which have been identified in 13 patients. Further, we aimed to the association of pathogenic DNA variants with presence of somato-facial and neuro-behavioural features in analysed patients.

Conclusion: The presented study, focused on the comprehensive genomic analysis of ASD patients confirm that the performance of WES/CES sequencing represents a practical complex approach, which leads to the more effective clarification of the role of genetic background in ASD etiopathogenesis.

Grants: The study supported by APVV-20-0070.

Conflict of Interest: None declared

EP10.022 First report of familial cases of DeSanto-Shinawi syndrome

Bashair Magadmi 1, Thomas Courtin1, Sophie Rondeau1, marion lesieur-sebellin1, Giulia Barcia1, Marlene Rio1

1Necker Hospital, Service de Médecine génomique des maladies rares, Paris, France

Background/Objectives: DeSanto-Shinawi syndrome (DESSH, OMIM #616708) is a rare autosomal dominant neurodevelopmental disorder caused by loss-of-function variants in the WAC gene. Affected individuals are characterized by developmental delay, intellectual disability, behavioral problems, and dysmorphism.

The WAC gene, mapping at the 10p12.1 region, encodes the WW domain-containing adapter with coiled-coil region (WAC) protein which plays a crucial role in various biological processes.

With the exception of one family with presumed parental germline mosaicism, all individuals diagnosed to date have the disorder as the result of a de novo pathogenic variant.

Results: Here, we present familial cases of DESSH in three siblings and their father caused by a heterozygous pathogenic variant, NM_016628 c.84_106dup (p.Pro36Leufs164), in exon 3 of the WAC gene.

The three siblings displayed a spectrum of neurodevelopmental disorders, including: Intellectual disability ranging from severe to moderate, speech and language impairments and cognitive impairment. Brain abnormalities, irncluding heterotopia and gyration anomalies, were observed in two of them. Distinctive facial features, consistent with DESSH, were present in three siblings.

Their father also experienced learning and speech difficulties. He also had distinctive facial features and MRI revealed cerebral atrophy.

Conclusion: To the best of our knowledge, this is the first inherited case of DESSH. The variable severity of neurodevelopmental difficulties highlight clinical variability in this syndrome even within the same family. Brain abnormalities present in 3 cases may expand the spectrum of this condition.

Grants: Varvagiannis, K., de Vries, B. B. A., & Vissers, L. E. L. M. (2017, November 30). WAC-Related Intellectual Disability. GeneReviews®. Retrieved from https://www.ncbi.nlm.nih.gov/books/NBK547098/

Conflict of Interest: None declared

EP10.023 Novel mutation in KARS1 identified in a family with intellectual disability and hearing impairment by whole exome sequencing

Asem Alkhateeb 1

1Jordan University of Science and Technology, Biotechnology and Genetics, Irbid, Jordan

Background/Objectives: Pathogenic mutations in Lysyl-tRNA synthetase 1 (KARS1) have been reported recently in patients with early-onset complex neurodevelopmental phenotypes and hearing impairment. We sought to delineate the cause of a neurologic phenotype in 3 affected sibs in one family.

Methods: We carried out whole exome sequencing in a proband with intellectual disability and hearing problems and in her consanguineous parents, followed by Sanger sequencing in two additional affected sibs and three normal sibs within the same family. Various in silico tools were implemented to further analyze effect of mutation found.

Results: We found a novel missense mutation in KARS1, c.989T>C, p.Leu330Pro (NM_001130089.2). The mutation segregated with the phenotype in the family. The parents are heterozygous, 3 affected sibs are homozygous for the mutation and 3 normal sibs are heterozygous. Various in silico tools supported a deleterious effect of the mutation on protein structure and function.

Conclusion: Trio whole exome sequencing represents a fast and easy tool to analyze candidate genes in families affected with neurodevelopmental phenotypes. Here we found a novel mutation in a gene already associated with disease. Further functional analysis would be needed to dissect the exact effect of this mutation on KARS1 protein function and disease mechanism.

Grants: Jordan University of Science and Technology (#20190464)

Conflict of Interest: None declared

EP10.024 Wiedemann-Steiner syndrome, a rare cause of intellectual disability

Nuray Ozturk 1, Mert Coşkun2, Duygu Gamze Aracı2, sezin yakut uzuner3, Aslı Toylu2, Gokcen Karamik1, Öznur YILMAZ BAYER1, guldeniz oner1, banu nur1, ercan mihci1

1Akdeniz University School of Medicine, Pediatric Genetics, Antalya, Türkyie; 2Akdeniz University School of Medicine, Medical Genetics, Antalya, Türkyie; 3Akdeniz University School of Medicine, Medical Biology and Genetics, Antalya, Türkyie

Background/Objectives: Wiedemann-Steiner syndrome (WDSTS, OMIM #605130) is a rare autosomal dominant genetic disorder characterized by intellectual disability, developmental delay, dysmorphic facial features, growth retardation, hypertrichosis and congenital anomalies. The facial features include thick eyebrows, long eyelashes, hypertelorism, narrow downslanting palpebral fissures, and a broad nasal bridge. WDSTS is caused by heterozygous pathogenic variants in KMT2A, a gene encoding a histone methyltransferase. The exact prevalence of WSS is not known, almost 250 patients have been reported in the literature. Herein, we describe three patients affected with WDSTS.

Methods: Patient 1 (P1,15-month-old,female), Patient 2 (P2,16y,male), and Patient 3 (P3,3y,male) were referred due to intellectual disability, developmental delay, dysmorphic facial features. Their prenatal screening and natal history were normal. There was parental consanguanity in P1. Their cranial MRI and chromosome analysis were normal. Abdominal ultrasonography revealed right renal agenesis in P2 and hydronephrosis in P3.

On physical examination, P1 and P2 had short stature, microcephaly, synophrys, ptosis, hirsutism, wide nasal bridge, broad nasal tip, thin upper lip. P3 had short stature, prominent glabella, epicanthus, broad nasal bridge, hypertelorism.

Results: Clinical-exome sequencing analysis displayed a likely pathogenic heterozygous missense variant of c.3581G>A, p.(Cys1194Tyr) for P1, a heterozygous pathogenic nonsense variant of c.3241C>T p.(Arg1081*) for P2 and a likely pathogenic heterozygous novel variant of c.4032_4033del, p.(Lys1346SerfsTer24) for P3 in the KMT2A gene.

Conclusion: We emphasized that WDSTS should be considered in the differential diagnosis with its typical dysmorphic findings in patients presenting with intellectual disability and developmental delay.

Grants: None

References: https://doi.org/10.1002/ajmg.a.62124., https://doi.org/10.3390/children9101545, PMID: 35617449

Conflict of Interest: None declared

EP10.025 A Novel Variant in CUL4B associated with X-linked Syndromic Intellectual Developmental Disorder, Cabezas Type

Norah Alsaleh 1;2

1King Abdullah Specialized Children’s Hospital, King Abdulaziz Medical City, Ministry of National Guard-Health Affairs (NGHA), Department of Genetics and Precision Medicine, Riyadh, Saudi Arabia; 2King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia

Background/Objectives: The CUL4B gene encodes a protein of the Cullin 4B-RING ubiquitin ligase complex, which regulates the degradation of cellular proteins, signals nucleotide excision repair, and is involved in DNA damage response. Variants in this gene have been linked to an X-linked syndromic intellectual developmental disorder, Cabezas (MRXSC) (MIM#300304). Clinical features include facial dysmorphism, intellectual disability (ID), speech delay, abnormal gait, and short stature. Here, we present a case with MRXSC with a novel copy number variant in the CUL4B gene that was missed by clinical exome and chromosomal microarray.

Methods/Case: A 40-month-old male presented with dysmorphic features, global developmental delay, mainly speech, strabismus, failure to thrive, short stature, and relative macrocephaly. Pregnancy and antenatal history were unremarkable. The parents are non-consanguineous, with one older daughter who is healthy. There is no similar presentation in the family.

Results: Basic metabolic work-up, including urine organic acids, acylcarnitine profile, and plasma amino acids, were unremarkable. Chromosomal microarray revealed a heterozygous maternally inherited duplication of 871 kb in the short arm of chromosome 3. Clinical exome sequencing was reported as negative. We then followed up with genome sequencing duo, which detected a maternally inherited hemizygous copy number loss involving exon 1 of the CUL4B gene; it was confirmed by real-time quantitative PCR.

Conclusion: MRXSC has been previously described to be caused by variants in the CUL4B gene. Here, we report a case with a novel copy number loss involving exon 1 of CUL4B. Most sequence and copy number variants were reported upstream of exon 2 of CUL4B.

Grants: None

Conflict of Interest: None declared

EP10.026 Molecular profile of Zhu-Tokita-Takenouchi-Kim syndrome (ZTTK) in Brazilian patients: case series and new SON variants

RAYANA MAIA 1, Matheus Lira2, Erika Santos2

1Federal University of Paraíba, Departament of Pediatrics and Genetics, João Pessoa, Brazil; 2Federal University of Campina Grande, Campina Grande, Brazil

Background/Objectives: Zhu-Tokita-Takenouchi-Kim (ZTTK) syndrome is a neurodevelopmental disorder first described in 2015, caused by heterozygous loss-of-function variants in SON gene, with multisystem involvement and a broad clinical phenotype which includes short stature and various malformations. The objective of this work is to describe the clinical profile of the largest group of Brazilian patients with the condition.

Methods: Patients with molecular confirmation of pathogenic variants in the SON gene by whole exome sequencing, gathered in a Brazilian group of patients with the syndrome.

Results: Patients come from different regions of the country and the age at diagnosis varies from prenatal to 12 years old. Of the 9 patients, 78% (n = 7) are male and 78% (n = 7) have frameshift variants and the remaining cases are nonsense variants. There are 7 new variants, the others corresponding to the most common reported variant c.5753_5756delTTAG.

Conclusion: Define the molecular etiologies of intellectual disability and neurodevelopmental delay contributes to understand the underlying mechanisms. The molecular profile of this group corroborates the literature of haploinsufficiency of the SON gene. It will enriches genetic data and favors knowledge about genotype-phenotype correlation.

Table 1. Mutations reported at Brazilian patients.

Patient number

Variant

Consequence

1

c.2847_2854del

p.(Tyr949*)

2

c.1134dupC

p.Ser379Leufs*48

3

c.5730del

p.(Ser1911Profs*95)

4

c.2919dupA

p.Pro974Thrfs*5

5

c.1134dupC

p.Arg1112*

6

c.3306dup

p.(Met1103Tyrfs*9)

7

c.5753_5756delTTAG

p.(Val1918Glufs*87)

8

c.371_375delAAAAG

p. (Glu124Glyfs*5)

9

c.5753_5756delTTAG

p. (Va11918Glufs*87)

Conflict of Interest: None declared

EP10.027 Novel variant in RFX7 gene : a neurodevelopmental disorder associated short stature.

Teresa CARRION 1;2, Maria Antonia Grimalt3, Victor Asencio4, MARIA CAIMARI JAUME5, Jordi Roldan6

1Hospital Universitario Son Espases, Pediatrics, Palma de Mallorca; 2Hospital Universitario Son Espases, Unidad de Diagnostico Molecular y Genética Clínica, Palma de Mallorca; 3Hospital Universitario Son Espases, neuropediatrics, Palma de Mallorca; 4Fundació Institut d’Investigació Sanitària Illes Balears (IdISBa), Unidad de Diagnostico Molecular y Genética Clínica, Palma de Mallorca; 5Hospital Universitario Son Espases, pediatrics endocrinology, PALMA DE MALLORCA; 6Hospital Universitario Son Espases, neuroradiology pediatrics, Palma de Mallorca

Case presentation

We report a 11 year old girl followed since infancy for genetics, neurologist and psychiatry due to intellectual disability, dysmorphic features, autism spectrum and attention deficit hyperactivity disorder.

She was born of non-consanguineous parents. No history of intellectual disability.

Physical Examination: Size: 120 cm (-3.8 SD). She showed peculiar fascies with broad and flat forehead, low and small set ears, arched eyebrows, bulbous nose tip. Thin upper lip, high-arched palate, macrodontia, clinodactyly of 5th fingers of both hands, fetal pads.

Investigations and Results: Neurometabolical studies were normal.

Brain MRI showed decreased volume of the corpus callosum and the pons. Pituitary gland of normal size.

Karyotype 46 XX. gen SHOX, Array-CGH 60 Kb and exome for autism and intelectual disability were normal.

Trio exome sequencing, done on the patient and both parents, revealed a de novo heterozygous frameshift variant in RFX7 NM_022841.7:c.2504dupT, p. His836fs*2.

The variant found is not found in the general population databases consulted.The variant, by doubling the T nucleotide at position 2504, generates an alteration in the reading pattern during protein synthesis from the histidine 836 residue which, in turn, causes premature termination. It is assumed, in the absence of functional studies, that a truncated protein is synthesized.

Associated disease include intellectual developmental disorder, autosomal dominant 71, with behavioral abnormalities.

Conclusion: Here we emphasize the importance of imminent and repeated expanded genetic testing to ensure early diagnosis and triage for rare pediatric disorders.

Conflict of Interest: None declared

EP10.028 Deep-intronic de novo variant in HNRNPK as likely cause for Au-Kline syndrome

Zarah Kowalzyk 1, Marion Bermudez1, Joseph Porrmann1, Nataliya DiDonato1;2, Christoph Hübner2, Andreas Dahl3, PY Billie Au4, Lindsay Phillips4, Sanaa Choufani5, Rosanna Weksberg5;6, Doreen William1, Evelin Schröck1;7, Arne Jahn1

1Institute for Clinical Genetics, University Hospital Carl Gustav Carus at TUD Dresden University of Technology, Dresden; 2Department of Neuropediatrics, Faculty of Medicine of TUD Dresden University of Technology and University Hospital Carl Gustav Carus at TUD Dresden University of Technology, Dresden, Germany; 3DRESDEN-concept Genome Center, Center for Molecular and Cellular Bioengineering, TUD Dresden University of Technology, Dresden, Germany; 4Department of Medical Genetics and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, Calgary, Canada; 5Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Canada; 6Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Canada; 7Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany

Background/Objectives: Pathogenic variants in the HNRNPK gene are associated with autosomal dominant Au-Kline syndrome (OMIM #616580), which is characterized by developmental delay and intellectual disability, hypotonia and typical facial features. We report a case with overlapping phenotypic features for which facial abnormalities led to the differential diagnosis of Au-Kline syndrome.

Methods: We report on a 6-year-old girl with suspicion of a syndromic disease for whom whole genome sequencing and HNRNPK variant segregation was performed after unremarkable initial diagnostics. Face2Gene helped to identify differential diagnoses. HNRNPK-specific DNA methylation signature was undertaken to help with variant classification using a well-established HNRNPK signature.

Results: For a 6 year old-girl with developmental delay, short stature, heart defect, omphalocele, reflux nephropathy, suspicion of optic disc coloboma and interstitial lung disease, initial genetic diagnostics were unremarkable (karyotyping, trio-exome, methylation analysis for Beckwith-Wiedemann syndrome). Face2Gene yielded a high gestalt match for Au-Kline Syndrome. However, we note that interstitial lung disease is not a phenotype linked to HNRNPK before. Whole Genome Sequencing of the index detected a heterozygous deep-intronic variant in HNRNPK (NM_031263.4:c.214-55T > A). Segregation analysis showed that this variant was de novo. Analysis of a HNRNPK-specific DNA methylation signature classified this variant as positive on the signature, further supporting the clinical diagnosis of Au-Kline syndrome (PMID: 36130591).

Conclusion: Face2Gene and genome sequencing were useful to identify a potential causative intronic HNRNPK- variant in a girl with a complex phenotype. A DNA methylation signature supported variant interpretation, particularly in individuals with health issues not previously reported in association with the syndrome.

Grants:

Conflict of Interest: None declared

EP10.029 Two new cases of CTCF-related Intellectual Disability

Ana Capela 1, Cláudia Falcão Reis1;2;3;4

1Centro Hospitalar Universitário de Santo António, Medical Genetics Department, Porto, Portugal; 2School of Medicine, University of Minho, Life and Health Sciences Research Institute (ICVS), Braga, Portugal; 33ICVS/3B’s – PT Government Associate Laboratory, Braga/Guimarães, Portugal; 4Abel Salazar Biomedical Sciences Institute, Porto University, Unit for Multidisciplinary Research in Biomedicine, Porto, Portugal

Background/Objectives: CTCF-related intellectual disability (MIM #615502) was first described in 2013 and manifests with variable degrees of intellectual disability (ID), microcephaly, and growth retardation. We report two patients diagnosed with CFCT-related ID.

Methods: Review of clinical files.

Results: Patient 1 is an 18 yo male, born to non-consanguineous parents. Pregnancy was uneventful. He presented with congenital microcephaly and feeding difficulties and later developed behavioral issues and developmental delays. At 17 yo he presented with moderate ID (Global IQ of 62 on WISC-III scale), microcephaly, small ears, telecanthus, retrognathia, and short webbed neck. WES detected a novel likely pathogenic variant in CTCF c.1485_1486dup p.(Lys496Ilefs*16), in heterozygosity. Parental studies determined likely de novo origin.

Patient 2 is a 4 yo male, born to non-consanguineous parents. Both parents and older sister reported learning difficulties. Pregnancy and labor were uneventful. He presented with language delay at 18 months and was diagnosed with autism spectrum disorder at 3 yo. Array-CGH showed a 15q11.2 BP1-BP2 microdeletion. He was then referred to medical genetics where small, cupped ears, hypertelorism and thin upper lip were noted, raising suspicion for a concomitant second diagnosis. WES identified an heterozygous likely pathogenic variant in CTCF c.1025G>A p.(Arg342His). Parental segregation studies are ongoing with mother sharing a similar facial gestalt.

Conclusion: Both cases exhibit distinct phenotypic profiles, ranging from mild cognitive impairment to autism spectrum disorder, reflecting the heterogeneous nature of CTCF-related ID. Patient 2 highlights the need for further investigation when clinical features are more complex, especially in a dysmorphology setting.

Grants:

Conflict of Interest: None declared

EP10.030 Rare manifestations in a patient with Vulto-van Silfhout-de Vries Syndrome carrying a novel mutation in the DEAF1 gene

Gergely Büki 1, Agnes Till1, Tímea Bujtás1, Anna Zsigmond1, Krisztina Galimurka1, Attila Gyenesei2, Kinga Hadzsiev1, Judit Bene1

1University of Pécs, Clinical Centre, Medical School, Medical Genetics, Pécs, Hungary; 2University of Pécs, Szentágothai Research Centre, Pécs, Hungary

Background/Objectives: Vulto-van Silfout-de Vries syndrome (VSVS; OMIM615828) is an extremely rare, autosomal dominant, intellectual developmental disorder caused by de novo mutations in the DEAF1 (OMIM602635) gene. VSVS is mainly characterized by delayed psychomotor development, impaired speech, and behavioral abnormalities, including autistic features and poor eye contact. Additional nonspecific manifestations consist of seizures, hypotonia, increased pain threshold and sleep disturbances. Although previous comprehensive studies established clinical heterogeneity among patients, clinodactyly and duplex kidney have not been presented so far. Hereby we demonstrate a patient with a novel mutation who shows the aforementioned symptoms.

Methods: Whole-exome sequencing (WES) was performed on a 7-year-old male patient to identify the disease-causing mutation. Libraries were prepared using Illumina DNA Prep With Enrichment Kit and sequenced on a NovaSeq 6000 platform (Illumina) with 100bp paired-end chemistry. Sanger sequencing was applied for confirmation, followed by targeted sequencing of the parents.

Results: WES data analysis revealed a novel heterozygous missense mutation (c.648G>C, p.Lys216Asn; NM_021008) in the DEAF1 gene. De novo origin was determined. Our patient was diagnosed with autism and presented delayed psychomotor development, speech impairment, joint laxity, hypotonia and minor dysmorphic features. Besides characteristic manifestations, duplex kidney and bilateral clinodactyly were observed.

Conclusion: Intra- and interfamilial phenotypic variability is a well-known phenomenon in several Mendelian disorders. Although our patient shares common hallmarks of the syndrome with the previously reported cases, according to our current knowledge no patient has been presented with duplex kidney or bilateral clinodactyly. Hereby we report a patient with such manifestations, which further contribute to the clinical heterogeneity.

Grants:

Conflict of Interest: None declared

EP10.031 Copy number variants of uncertain significance in chromosome 22

Zivile Zemeckiene 1;2, Rasa Traberg1;2, Inga Nasvytienė1, Marius Šukys1;2, Rasa Ugenskiene1;2

1Hospital of Lithuanian University of Health Sciences Kauno klinikos, Department of Genetics and Molecular Medicine, Kaunas, Lithuania; 2Lithuanian University of Health Sciences, Department of Genetics and Molecular Medicine, Kaunas, Lithuania

Background/Objectives: Deletions and duplications of chromosome 22 are among the most common copy number variants (CNVs) associated with neurodevelopmental disorders, including well described syndromes like 22q11.2 microdeletion (velocardiofacial/DiGeorge), 22q11.2 microduplication, Phelan-McDermid and others. There is still little information about rare atypical variants.

Methods: The testing was performed in the Hospital of Lithuanian University of Heath Sciences Kauno Klinikos during the year 2021-2023. SNP-based human microarray technology (Illumina) was used. All reports were reviewed and patients with variants of uncertain significance (VUS) in chromosome 22 were selected for further analysis.

Results: Ten patients had VUS variants in chromosome 22 (two deletions and eight duplications). All variants were smaller than 300 Kb. One patient had duplication within 22q13.2 band (encompassing XPNPEP3 gene), also VUS duplication within 16p13.11 band. This was the only patient with microcephaly and Pierre-Robin sequence. Other nine patients had CNVs within 22q11.2 band. CNVs encompassed either PRODH (2 patients) or TOP3B gene (6 patients), one patient had two separate CNVs involving both genes. All patients had neurodevelopment or psychomotor development delay, older patients had problems with language development. Four patients with variants involving TOP3B and one with PRODH had various organ anomalies (heart, kidneys, eyes, brain). Patient phenotypes were variable. Hypertelorism (5 patients), ear abnormalities (5 patients) and micrognathia (3 patients) were the most common dysmorphic features.

Conclusion: 9 of 10 patients had VUS variants of chromosome 22 involving TOP3B or PRODH genes. This suggest potential importance of those genes in neurodevelopmental disorders.

Grants: -

Conflict of Interest: None declared

EP10.032 Refining genotype-phenotype correlation in complex chromosomal rearrangements using optical genome mapping - case report

Vladimira Vallova 1;2, Dominik Režný1, Jan Smetana1, Renata Gaillyova3, Petr Kuglik1;2

1Faculty of Science Masaryk University, Dept. of Experimental Biology, Brno, Czech Republic; 2University Hospital Brno, Centre of Molecular Biology and Genetics, Brno, Czech Republic; 3University Hospital Brno, Institute of Medical Genetics and Genomics, Brno, Czech Republic

Background/Objectives: Detection of chromosomal structural abnormalities based on the routine examination of the genome has some shortcomings - low resolution and laboriousness (karyotype) and lack of detection of balanced genetic changes (array-CGH). These shortcomings seem to be crucial especially in the determination of precise breakpoints in translocations, inversions and in the detection of complex cytogenetic rearrangements. Recently, technologies are emerging that could overcome these shortcomings. These are based on the analysis of long DNA molecules - optical genome mapping and long-read whole-genome sequencing.

Methods: We present a case report of a patient with neurodevelopmental disorder and complex rearrangement involving chromosomes 3 and 7, who underwent traditional genetic tests (karyotyping, aCGH, FISH) followed by optical genome mapping (OGM).

Results: The analysis using OGM was able to detect multiple rearrangements and refine the coordinates of individual structural variants. Based on relationships between aberrations, we were able to reconstruct events on affected chromosomes. Additionally, a potential disruption of the CNTN4 gene located at the translocation breakpoint was discovered. This disruption may describe the patient’s phenotype, as the gene’s product plays a crucial role in the development of the nervous system.

Conclusion: Technologies based on the analysis of long DNA molecules could not only detect novel causal rearrangements, but also refine breakpoints and thus contribute to the discovery of specific affected genes or mechanisms leading to the pathological phenotype of patients.

Grants: Supported by Ministry of Health, Czech Republic - Conceptual development of research organization (FNBr, 65269705).

Conflict of Interest: None declared

EP10.033 ANKDR11-related disorder: A chromatinopathy associated with a large phenotypic spectrum.

Fatma Majdoub 1, Imene Boujelbene2, Manel Guirat2, Alaa Ziadi2;3, Amal Souissi4, Ines Hsairi5, ridha mrad3, Hassen Kamoun2, Sabeur Masmoudi4, Mediha Trabelsi3, Ikhlas Ben Ayed2;4

1Hedi Chaker Hospital, Medical Genetic Department, Sfax; 2Hedi Chaker Hospital, Medical Genetic Department, Sfax, Tunisia; 3Charles Nicolle Hospital, Department of Hereditary and Congenital Disorders, Tunis, Tunisia; 4Center of Biotechnology of Sfax, University of Sfax, Laboratory of Molecular and Cellular Screening Processes (LPCMC), LR15CBS07, Sfax, Tunisia; 5Hedi Chaker Hospital, Pediatric Neurology department, Sfax, Tunisia

Background/Objectives: Chromatinopathies are a group of more than 100 syndromes associated with neurodevelopmental disorders (NDD) characterised by an imbalance in the state of chromatin. ANDRD11, involved in chromatin regulation via histone acetylation, is the major gene in KBG syndrome. However, it has also been reported in association with different clinical presentations ranging from a Cornelia de Lange syndrome (CDLS) phenotype to non-syndromic intellectual disability.

Methods: In this study, we report two unrelated patients referred for KBG syndrome (P1) and CDLS-like syndrome (P2) phenotypes. The genetic study of this patient was performed using the FGDYS-HaloPlex-HS panel, a designed gene panel, through the HaloPlex HS Target Enrichment System provided by Agilent Technologies (Agilent, Santa Clara, CA, USA). Targeted gene sequencing was used to investigate CDLS-associated genes (NIPBL, SMC1A, RAD21, SMC3, HDAC8) and CDLS-like associated genes (ANKRD11, EP300, SETD5, BRD4, CREBBP, ZMYND11, MED13L, PHIP, EHMT1).

Results: We identified two class 5 heterozygous variants in the ANKRD11 gene (NM_001256182.1) gene associated with KBG syndrome (OMIM : 148050): a frameshift variant (p.Glu800AsnfsTer62) in patient P1 and a nonsense variant in patient P2 (p.Tyr1406Ter).

Conclusion: The two truncating variants of the ANKRD11 gene identified in this study were previously reported in the literature and are located in exon 9, which represents a mutational hotspot for this gene. Our study highlights the overlap between the two KBG/CDLS entities. It also confirms the variable phenotypic expressivity of the ANKRD11 gene.

Grants: FGDYS-PFR 2019- D3P3. KAFSS- PAQ-Collabora 3-8.

Conflict of Interest: None declared

EP10.034 Comprehensive clinical and genetic characterization of a Spanish cohort of Bainbridge Ropers syndrome

Laura Trujillano 1, Irene Valenzuela Palafoll1, Mar Costa-Roger1, Ivon Cuscó2, Anna Maria Cueto-González1, Amaia Lasa-Aranzasti1, Barbara Masotto1, Anna Abulí1, Marta Codina1, Ana Maria Cordero1, M Luisa Carcas Espinosa1, Miguel del Campo3, Juan Antonio Moreno Ruiz4, Cristina Pardo Dominguez4, Carmen Palma Milla5;6, Rubén Pérez de la Fuente5;6, Juan Francisco Quesada Espinosa5;6, Blanca Gener-Querol7, María Juliana Ballesta Martínez8, Alejandro J Brea-Fernández9;10, Montse Fernández-Prieto9;10, Elisabet Gabau11, Anna Ruiz11, Juan Pablo Trujillo Quintero11, Fernando Santos-Simarro12, Mónica Roselló Piera13, Carmen Orellana13, Francisco Martinez13, Didac Casas-Alba14, Mercedes Serrano15, María Palomares-Bralo16, Emi Rikeros Orozco16, María de los Ángeles Gómez Cano16, Pilar Tirado-Requero17, Elena García-Arumí1, Eduardo Tizzano1

1Barcelona, Clinical and Molecular Genetics Area, Vall d’Hebron Hospital; Medicine Genetics Group, Vall d’Hebron Research Institute (VHIR), Barcelona, Spain; 2Hospital de la Santa Creu i Sant Pau, Genetics Department, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona; 3University of California, San Diego School of Medicine, Clinical Pediatrics, San Diego, United States; 4Hospital Costa del Sol, Department of Pediatrics, Marbella, Spain; 5Hospital Universitario 12 de Octubre, Department of Genetics, Madrid, Spain; 6Hospital Universitario 12 de Octubre, UDISGEN (Unidad de Dismorfología y Genética), Madrid, Spain; 7Cruces University Hospital. Biobizkaia Health Research Institute, Department of Genetics., Vizcaya, Spain; 8Hospital Clinico Universitario Virgen de la Arrixaca, Sección Genética Médica. Servicio de Pediatría., Murcia, Spain; 9Universidade de Santiago de Compostela, Grupo de Genómica y Bioinformática, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Centro de Investigación Biomédica en Red de Enfermedades Raras del Instituto de Salud Carlos III (CIBERER-ISCIII), Santiago de Compostela, Spain; 10Universidade de Santiago de Compostela, Grupo de Medicina Xenómica, Santiago de Compostela, Spain; 11Parc Taulí Hospital Universitari, Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA), Universitat Autònoma de Barcelona, Unidad de Genética Clínica, Centro de Medicina Genómica, Sabadell, Spain; 12Hospital Universitario Son Espases, Unidad de Diagnóstico Molecular y Genética Clínica, Palma de Mallorca, Spain; 13Hospital Universitario y Politecnico La Fe, Genetics Unit, Valencia, Spain; 14Hospital Sant Joan de Déu, Department of Genetic and Molecular Medicine/IPER, Institut de Recerca, Esplugues de Llobregat, Spain; 15Institut de Recerca, Hospital Sant Joan de Déu, Pediatric Neurology Department, Esplugues de Llobregat, Spain; 16Hospital Universitario La Paz, Clinical Genetics Section, Medical and Molecular Genetics Institute (INGEMM) IdiPaz, CIBERER, Madrid, Spain; 17University Hospital La Paz, Neuropediatric Unit, Madrid, Spain

Background/Objectives: Bainbridge-Ropers Syndrome (BRPS) is a genetic condition resulting from truncating variants in the Additional Sex Combs-Like 3 (ASXL3) gene. The primary clinical features include neurodevelopmental and language impairments, behavioral issues, hypotonia, feeding difficulties, and distinctive facial features.

Methods: We conducted a comprehensive analysis of 22 Spanish patients with BRPS, aiming to enhance our understanding of clinical and molecular aspects and establish a genotype-phenotype correlation.

Results: We identified nineteen ASXL3 variants, with ten previously unreported and three recurrent ones. Furthermore, we documented a case of germinal mosaicism. The predominant prenatal finding was intrauterine growth restriction (35%). Following birth, intellectual disability and language issues consistently prevailed, accompanied by feeding difficulties (90.5%), hypotonia (85.7%), gastroesophageal reflux disease (82.4%), autistic spectrum disorder (75%), and joint laxity (73.7%). Upon stratifying the sample based on the ASXL3 variant exon location, individuals with variants in the 3′ mutational cluster region (MCR) of exon 12 exhibited a heightened incidence of perinatal feeding problems (p = 0.035). Remarkably, those individuals with variants in the 5′ MCR of exon 11 displayed reduced percentiles in height (p = 0.008) and head circumference (p = 0.007) postnatally, combined with a notable prevalence of arched eyebrows (p = 0.038).

Conclusion: This study represents the first detailed characterization of a Spanish cohort with BRPS. Our research establishes a correlation between genotype and phenotype based on the ASXL3 variant location.

Grants: Part of this study is funded by the Health Department of the Catalan Government (PERIS SLT002/16/00174).

Conflict of Interest: None declared

EP10.035 Novel loss-of-function variant in the YY1 gene in a child with global developmental delay and movement disorder

Teodora Barbarii 1, Diana Barca2;3

1University of Medicine and Pharmacy “Carol Davila”, Medical Genetics Department, Bucharest, Romania; 2Clinical Hospital ”Prof. Dr. Alexandru Obregia”, Expertise Center for Rare Pediatric Neurology Disorders, Pediatric Neurology Department, Bucharest, Romania; 3University of Medicine and Pharmacy “Carol Davila”, Pediatric Neurology, Bucharest, Romania

Background/Objectives: Loss-of-function variants in the YY1 gene are known to cause Gabriele-de Vries syndrome (GADEVS) characterized by global developmental delay (GDD), craniofacial dysmorphism, multiple congenital anomalies and various comorbidities. To date, less than 30 patients with GADEVS have been reported in the literature and only a few of them are associating movement disorders (MD). Moreover, recent publications described patients who initially presented MD or MD with mild GDD and their phenotype eventually progressed to severe forms of generalized dystonia or other complex MD until their adulthood.

Methods: We describe a 2 year old boy with mild global developmental delay, facial dysmorphism and recurrent infections. At 9 months old he presented tremor, choreothetosis, orofacial dyskinesia and developed a chronic ataxic syndrome. Brain MRI was negative. Genetic analysis was performed by whole exome sequencing (WES).

Results: A heterozygous variant NM_003403.4:c.844del, p.(Met282fs*) in the YY1 gene was detected which is predicted to result in a loss-of-function effect. The mutation was classified as likely pathogenic and has never been reported before. The child underwent speech and kinetotherapy sessions and his current development is in the broadly normal range for his age.

Conclusion: The causality between YY1 haploinsufficiency and MD evolution is still being studied. YY1 gene could be considered both a MD and an intellectual disability gene since the MD can be the prominent feature of the phenotype as well as the first symptom of the disease. Patients carrying YY1 variants need periodic follow-up since their phenotype can progress into debilitating forms of MD.

Conflict of Interest: None declared

EP10.036 Single nucleotide variants in the ABCA2 transmembrane domains cause a severe and complex neurodevelopmental phenotype

Karit Reinson 1;2, Kaisa Teele Oja1;2, Mihkel Ilisson2, Eleanor G. Seaby3, Monica H Wojcik3;4, Anne O’Donnell-Luria3;4, Bryan M Wittmann5, Adam Kennedy5, Karen DeBalsi5, Christiane Zweier6;7, Georgia Vasileiou6;7, Issam A. Alkhawaja8, Thomas Wirth9, Steffen Syrbe10, Suad Alyamani11, Fowzan Alkuraya11, David Ros-Pardo12, Iñigo Marcos-Alcalde12, Paulino Gomez-Puertas12, Ke Su13;14, Shaohua Fan13, Yu Ma15, Yi Wang15;16, Marie Vidailhet17, Alessandra Renieri18;19;20, Anna Maria Pinto20, Farah Almadhoun18, Andrea Petersen21, Kari Magnussen21, Olaf Bodamer21, Nicole Fleischer22, Amber Begtrup23, Sander Pajusalu1;2, Katrin Ounap1;2

1University of Tartu, Department of Clinical Genetics, Institute of Clinical Medicine, Tartu, Estonia; 2Tartu University Hospital, Genetics and Personalized Medicine Clinic, Tartu, Estonia; 3Broad Institute of MIT and Harvard, Cambridge, MA, United States; 4Boston Children’s Hospital, Boston, MA, United States; 5Metabolon, Morrisville, NC, United States; 6Inselspital Bern, University of Bern, Department of Human Genetics, Bern, Switzerland; 7Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; 8Albashir Hospital, Amman, Jordan; 9Strasbourg University Hospital, Strasbourg, France; 10Centre for Paediatrics and Adolescent Medicine, University Hospital Heidelberg, Division of Pediatric Epileptology, Heidelberg, Germany; 11King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia; 12Centro de Biología Molecular Severo Ochoa, CBMSO (CSIC-UAM), Molecular Modeling Group, Madrid, Spain; 13State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, China; 14MOE Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, China; 15Division of Neurology, Children’s Hospital of Fudan University, No. 399 Wanyuan Road, Shanghai, China; 16Institute of Brain Science, Fudan University, Shanghai, China; 17Pitié-Salpêtrière University Hospital Paris, Paris, France; 18Medical Genetics, University of Siena, Siena, Italy; 19Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy; 20Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy; 21Randall Children’s Hospital at Legacy Emanuel Medical Center, Portland, United States; 22FDNA Inc., Boston, Massachusetts, United States; 23GeneDx, 207 Perry Parkway, Gaithersburg, United States

Background/Objectives: The ATP-binding cassette subfamily A member 2 (ABCA2), encoded by ABCA2, is an endolysosomal membrane protein that mediates the movement of sphingolipids within cellular compartments. ABCA2 has two transmembrane (TM) domains and two nucleotide-binding domains with an intervening loop domain. Variants in this gene have been previously associated with a neurodevelopmental disorder, and we further delineate the phenotypic spectrum.

Methods: We describe 14 patients with single nucleotide variants (except frameshift variants) in ABCA2: eight patients with TM variants (4 mono- and 4 biallelic) and six with non-TM (lumen/cytosol/C-terminus) variants (3 mono- and 3 biallelic). Clinical and molecular information was obtained by a questionnaire.

Results: Patients with TM variants have more severe and complex phenotypes. The prevalence of childhood onset epilepsy was high in individuals with TM variants [6/8 (75%)] versus 1/6 (17%) in the non-TM variants (p = 0.049; Fisher’s exact test). The severity of intellectual disability (ID) was statistically significantly more severe in the TM group (p = 0.046, Mann-Whitney U-test). Similar trends were observed with autistic features [TM 3/7 (43%), non-TM 0/5 (0%)], behavioral abnormalities [TM 4/8 (50%), non-TM 1/5 (20%)] and neurodevelopmental problems [TM 7/8 (88%), non-TM 4/6 (67%)]. However, due to small sample sizes, these trends did not reach statistical significance.

Conclusion: These findings indicate that the variants occurring in the TM region have a more disruptive effect on protein function and ultimately cause abnormalities in the affected patient’s phenotypes.

Grants: PUT355, PRG471, PRG2040, PSG774, UM1HG008900, K23HD102589, RTI2018-094434-B-I00, PID2021-126625OB-I00, DTS20-00024

Conflict of Interest: None declared

EP10.038 Identification of de novo variation in patients with neurodevelopmental disorder using trio whole genome sequencing.

Sarah Baer 1;2, Merol Jeanne1, Lamouche Jean-Baptiste1, Geoffroy Véronique3, Tarek Alouane1, Salima EL CHEHADEH1;4, Valérie Skory1, sophie scheidecker3;5, Benjamin Durand4, Elise Schaefer4, Jean Muller3;5, Amelie Piton1;5

1Institute for Genetics and Molecular and Cellular Biology (IGBMC), University of Strasbourg, CNRS UMR7104, INSERM U1258, Illkirch, France; 2Hopitaux Universitaires de Strasbourg, Pediatric neurology, Strasbourg, France; 3Institut de génétique médicale d’Alsace IGMA, Laboratoire de Génétique Médicale, INSERM U1112, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg UMRS_1112, Strasbourg, France; 4Institut de Génétique Médicale d’Alsace (IGMA), Service de génétique clinique, Strasbourg, France; 5Hopitaux Universitaires de Strasbourg, Laboratoires de diagnostique génétique, Strasbourg, France

Background: Half of the patients with neurodevelopmental disorders remain with negative genetic analyses. This could be due to variants occurring outside of coding sequences having functional consequences difficult to predict, structural variations or non-classical mutational effects like expansion, mobile element, or variants in genes currently not known to be involved in pathology.

Methods/results: We performed trio-WGS in a cohort of 18 individuals negative after simplex WGS. After filtering with quality and frequency criteria, we found 114 de novo variants on average per patient with de novoCNN and 128 with Strelka. The IGV visualization showed that 83% and 74% of the variants detected appear truly de novo. Finally, with the two programs, 97 de novo variants were identified, 89% confirmed by IGV. We identified between 73 and 130 variants per trio including 1 to 3 in coding regions. Three nonsynonymous variants occurred in genes with high intolerance to loss-of-function (DOP1A, PNMA5, and TM9SF3), with ongoing collaborations. De novo variants located outside coding regions are distributed as: 1% in untranscribed regions, 45% in introns, 4% downstream or upstream of protein-coding genes, and 50% in intergenic regions. Regarding intronic variations, between 6 and 13 variations are predicted to affect splicing. Further annotations of uORF, long non-coding RNA, and other regulatory sequences have been collected to prioritize candidate variants.

Conclusion: In this project, we developed a strategy to identify de novo variations in patients with sporadic NDD, through trio whole genome trio sequencing. This highlighted novel candidate genes and variants located both in and outside coding regions.

Conflict of Interest: None declared

EP10.039 Identification of a novel homozygous missense variant in TMEM147 in two brothers with severe intellectual disability

Linda Sofan 1, Rebecca Buchert1, Wael Al-Ameri2, Olaf Riess1, Tobias Haack1, Tawfiq Froukh3

1University of Tübingen, Institute of Medical Genetics and Applied Genomics, Tübingen, Germany; 2Ibn Alhytham Hospital, Amman, Jordan; 3Philadelphia University, Department of Biotechnology and Genetic Engineering, Amman, Jordan

Background/Objectives: Intellectual disability (ID) is a neurodevelopmental disorder with high phenotypic and genetic heterogenicity leading to new ID-associated genes being identified. Recently, TMEM147, a transmembrane protein that functions in the nuclear envelope and endoplasmic reticulum, has been identified as an ID gene. Here we report on two cases of intellectual disability with a variant in TMEM147 which was found through whole exome analysis.

Methods: Via exome sequencing, a homozygous missense variant in TMEM147 was identified in a case with intellectual disability. Variant segregation in the family was performed using Sanger sequencing.

Results: We examined two brothers with severe intellectual disability, hyperactivity, hypotonia, coarse facial features, exophoria, prominent nasal root, and down-slanting palpebral fissures. Exome sequencing identified a homozygous highly conserved missense variant in TMEM147 (NM_032635.4: c.137A>C (p.Gln46Pro)) that segregates with the phenotype in the family. The amino acid exchange to proline is predicted to break the α-helix structure of the second transmembrane domain and impair protein function.

Conclusion: TMEM147 has been linked recently to ID of variable severity with facial dysmorphism. We report on a new variant in this gene in two affected siblings who share phenotypic similarity with reported cases such as hyperactivity, hypotonia, coarse facial features, exophoria, prominent nasal root, thin upper lip, everted lower lip, down-slanting palpebral fissures, large mouth, and thick curly hair. In addition to the phenotypic resemblance, the predicted impact of the identified variant on protein function provides evidence of the pathogenicity of this variant.

Grants: German Academic Exchange Service (DAAD, Project-ID 57166498)

Conflict of Interest: None declared

EP10.040 Impact of reanalysis of clinical exome data on diagnostic yield

Louisa Sanchez 1, Evelyn Douglas1, Sin Lay Kang1, Kathy Cox1, Linda Burrows1, Abhi Kulkarni2, Karin Kassahn2, Lesley McGregor3, Jan Liebelt3, Suzanne Sallevelt3, Shannon LeBlanc3, Christopher P. Barnett3, Song Gao2, Evelyn Collen2, Kathryn Friend1, Sui Yu1

1SA Pathology, Genetics and Molecular Pathology, North Adelaide, Australia; 2SA Pathology, Genetics and Molecular Pathology, Adelaide, Australia; 3Women’s and Children’s Hospital, Paediatric and Reproductive Genetics Unit, North Adelaide, Australia

Background/Objectives: The introduction of Next-Generation Sequencing (NGS) into routine clinical practice has greatly improved diagnostic yield for patients. Cases assessed by our laboratory predominantly have intellectual disability and/or congenital abnormalities (including facial dysmorphism). Although a single analysis of exome data resolves many cases (30% likely pathogenic/pathogenic, 14% variants of uncertain significance), a genetic cause in many others (55%) remains unidentified.

Methods: From the initiation of diagnostic whole exome sequencing in our laboratory (2020) to the end of 2023, approximately 1100 exomes were reported: trio exomes (mother, father and proband) (873); singleton exomes (228). During this same period, 61 requests (for 49 patients) to reanalyse existing patient NGS data were completed. 8 cases had multiple requests for reanalysis (range 2-3 reanalyses).

Results: The initial NGS analysis for cases where reanalysis was subsequently requested, identified no reportable variants in approximately 69% of cases (34/49), with variants of uncertain significance and pathogenic/likely pathogenic variants reported in the remainder of cases. Reasons given for the reanalysis requests included the availability of additional phenotypic information, requests for specific gene or panel analysis not undertaken on the original analysis, and expansion of the analysis from a targeted panel to a whole exome analysis. Reanalysis yielded reportable variants in 9 (18%) cases. 27 cases (55%) remained without any reportable variants.

Conclusion: Reanalysis of NGS data over time has improved diagnostic yield, with reportable variants identified in approximately 20% of cases.

Conflict of Interest: None declared

EP10.041 Identification of novel mutation in DYRK1A gene associated with white matter abnormality as a rare feature in Intellectual Developmental Disorder 7

Judit Bene 1, Anna Zsigmond1, Gergely Büki1, Attila Gyenesei2, Zsuzsa Siegler3, Kinga Hadzsiev1

1University of Pécs, Medical School, Department of Medical Genetics, Pécs, Hungary; 2University of Pécs, Szentágothai Research Centre, Pécs, Hungary; 3Bethesda Children’s Hospital, Department of Neurology, Budapest, Hungary

Consortium: -

Background: DYRK1A (dual-specificity tyrosine-phosphorylation-regulated kinase) gene has an important role in neuronal development, particularly in regulating neuronal proliferation, differentiation, plasticity and death. De novo mutations of the gene is associated with a rare autosomal dominant Intellectual Developmental Disorder 7 (IDD7) (OMIM 614104) characterized by intellectual disability, speech and language delays, microcephaly, facial dysmorphism, seizure and feeding difficulties. Various brain abnormalities (i.e. enlarged ventricles, corpus callosum atrophy, cerebellar atrophy) have been described so far, however, white matter abnormality was not observed among patients with IDD7. Here we report a patient with DYRK1A-associated IDD who displayed periventricular leukomalacia on MRI.

Methods: Whole-exome sequencing (WES) was performed on a 7-year-old male patient using Illumina DNA Prep With Enrichment Kit for library preparation and sequenced on a NovaSeq 6000 platform (Illumina) with 100bp paired-end chemistry. For validation purposes and segregation analysis Sanger sequencing was applied.

Results: WES data analysis revealed a novel heterozygous frameshift mutation (c.1524delC, p.Thr508Profs*84; NM_001396) in the DYRK1A gene. Genetic tests of the parents confirmed the de novo origin of the mutation. Our patient was diagnosed with primary microcephaly, psychomotor and language development delay, hypotonia, febrile seizures and minor dysmorphic features. Besides the characteristic hallmarks, periventricular leukomalacia and clinodactyly were observed.

Conclusion: Although patients with DYRK1A-associated IDD7 display recurrent pattern of clinical manifestations, some unique feature can be observed like periventricular leukomalacia in our patient. This study expands the knowledge of mutation spectrum and clinical features associated with IDD7.

Conflict of Interest: None declared

EP10.042 De novo variants in ATP6V1B2 cause a developmental and epileptic encephalopathy: a case report and review of the literature

Eva-Maria Kraus 1, Robin-Tobias Jauss1, Konrad Platzer1, Franziska Schnabel1

1University Leipzig Medical Center, Institute of Human Genetics, Leipzig

Background/Objectives: ATP6V1B2 encodes a subunit of the vacuolar ATPase, a multisubunit enzyme that facilitates acidification of intracellular organelles. Heterozygous (de novo) variants are causative for the syndromic disorders Zimmermann-Laband syndrome and deafness-onchodystrophy syndrome. In four single-case reports, de novo variants associated with epileptic encephalopathy, profound developmental delay and microcephaly have been described, but a detailed description is lacking.

Methods: We performed trio-genome sequencing using standard wet-lab techniques and the emedgene software for data analysis.

Results: We identified the heterozygous de novo variant NM_001693.4:c.1316_1318del, p.Val439del in ATP6V1B2 in a 13-year-old girl with therapy-resistant epilepsy, severe global developmental delay with absent speech and inability to walk, microcephaly, facial dysmorphism, minor cerebral MRI abnormalities, hypotonia, enamel hypoplasia, feeding difficulties, myopia and hypercholesterolemia. Our case is the 2nd individual described with a de novo inframe deletion and a severe neurodevelopmental phenotype.

Conclusion: This case report further solidifies the emerging epileptic neurodevelopmental phenotype due to de novo variants in ATP6V1B2 and provides detailed clinical data that is essential for comprehensive patient care.

Grants: None

Conflict of Interest: None declared

EP10.043 A recurrent CREBBP variant in a patient with Menke-Hennekam syndrome and normal intelligence

Juliane Hoyer 1, Cornelia Kraus1, André Reis1;2

1Friedrich-Alexander Universität Erlangen-Nürnberg, Institute of Human Genetics, Erlangen, Germany; 2Friedrich-Alexander Universität Erlangen-Nürnberg, Centre for Rare Diseases, Erlangen, Germany

Background/Objectives: Pathogenic variants in CREBBP cause Rubinstein–Taybi syndrome (RTS). Since 2016, variants affecting specific regions of exons 30 and 31 have been reported in individuals lacking the characteristic facial and limb dysmorphism associated with classical RTS. In 2019, the name of Menke–Hennekam syndrome (MHS) was proposed to differentiate the distinct phenotypes.

Methods: Here, we report a female individual with feeding difficulties shortly after birth. She presented at age 17 years with mild facial dysmorphism, microcephaly and dystrophy. Due to a severe ADHD she needed a personal classroom aide. However, IQ testing revealed repeatedly normal results and she successfully graduated after 5 years of secondary school education.

Whole exome sequencing identified a de novo in-frame deletion of three bp (p.Phe1633del) in CREBBP.

Results: The previously reported cluster of variants associated with MHS overlaps with the ZNF2 and ZNF3 domains. However, the variant Phe1633del affects a highly conserved residue in the HAT domain (histone acetyl transferase domain) and was previously reported in a 2-year-7-month old Chinese girl. Both individuals shared the common symptoms of MHS, including facial dysmorphism, microcephaly and feeding problems, but did not show other characteristics, such as autistic behaviour, hearing impairments and epilepsy. It is remarkable, that both patients presented with ADHD, which was not described previously. Most strikingly, our patient is the only MHS individual reported to date with normal intelligence.

Conclusion: Our findings contribute to genotype-phenotype correlation in CREBBP-associated disorders and highlight further genetic variability in MHS.

Grants: none

Conflict of Interest: None declared

EP10.044 A case of maternal uniparental isodisomy for chromosome 6 (UPD(6)mat)

Maria Franaszczyk 1;1, Aleksandra Świeca1, Agnieszka Pollak1, Piotr Gasperowicz1, Grażyna Kostrzewa1, Piotr Stawiński1, Karolina Rutkowska1, Małgorzata Rydzanicz1, Krzysztof Szczałuba1, Rafał Płoski1

1Medical University of Warsaw, Warszawa

Background/Objectives: The imprinting-dependent phenotypic effects of maternal uniparental disomy (UPD) for chromosome 6 remain under debate. Despite only a few reported cases, no specific phenotype is prevalent, except for intrauterine growth restriction (IUGR) and low birth weight.

Methods: In our proband, pregnancy was complicated by polyhydramnios, but no significant intrauterine growth complications were noted during serial ultrasound examinations. She was born preterm at 35 weeks via caesarean delivery, with birth weight, length, and head circumference below average. At 8 months of age, she was treated for poor weight gain and anaemia. Growth parameters at 4 years and 10 months of age were below the 3rd centile. After consultation with a neurologist, she was diagnosed with autism and pervasive developmental disorders and subsequently referred for genetic testing to investigate the cause of the developmental delay and regression.

The proband and her parents underwent trio exome sequencing (ES), including the analysis of regions of homozygosity (ROH).

Results: The analysis of ROH in the proband in relation to her parents led to the diagnosis of uniparental isodisomy of chromosome 6. Verification and assessment of the parental origin of chromosome 6 isodisomy, performed by testing selected single nucleotide variants (SNVs) located within both arms of chromosome 6, allowed us to determine the maternal origin of chromosome 6.

Conclusion: Given the rarity of upd(6)mat, our case exhibits characteristic features consistent with previous descriptions, thereby facilitating a better understanding of its clinical presentation.

Grants: N/A

Conflict of Interest: None declared

EP10.045 Detection of Copy-Number Variants in patients with overgrowth syndromes using 850K SNP-arrays

alejandro parra 1;2;3, Jair Tenorio1;2;3, julian nevado1;2;3, Mario Cazalla1;2;3, Patricia Pascual Vinagre1;2;3, Lucía Miranda-Alcaraz1;2;3, Natalia Gallego1;2;3, cristina silvan2, Pedro Arias3, Jesús Pozo-Román4;5, María Juliana Ballesta Martínez6;7, Encarna Guillén-Navarro1;7, ignacio arroyo8, Pablo LAPUNZINA1;2;3

1CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain; 2INGEMM-Idipaz, Institute of Medical and Molecular Genetics, Madrid; 3ITHACA, European Reference Network, Hospital Universitario La Paz, Madrid; 4Unit of Paediatric Endocrinology. Department of Pediatrics. Hospital Universitario Infantil Niño Jesús, Madrid, Spain; 5Department of Pediatrics. Medical School. Autonomous University of Madrid, Madrid, Spain; 6Sección de Genética Médica, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain; 7Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain; 8Pediatrics Department, San Pedro de Alcántara Hospital, Cáceres, Spain

Background/Objectives: Overgrowth syndromes (OGS) comprise a heterogeneous group of disorders whose main characteristic is that either the weight, height or head circumference are above the 97th centile or 2–3 standard deviations above the mean for age, gender, and ethnic group. Several CNVs have been associated with the development of OGS throughout history.

Methods: This study suppose the application of 850K SNP-arrays to 150 OGS patients from the Spanish OverGrowth Registry Initiative. A Whole Genome Sequencing analysis was performed on patients with CNVs in order to obtain more information and better understand the molecular basis of the CNVs.

Results: We identified CNVs associated or probably associated with this type of disorders in 13 individuals, obtaining a diagnosis yield of 8.67%.

Conclusion: New regions have been found with deletions or duplications probably associated with alterations of growth, which, in addition, could include other types of alterations such as intellectual disability.

SNPs microarrays are a great tool for the detection of dose alterations in patients with overgrowth without known molecular defect.

The high phenotypic overlap between the different overgrowth syndromes makes a molecular study essential to identify the associated genetic defect, which will allow better detection, monitoring and management of patients.

Grants: This research was funded by the FIS PI20/01053, from the ISCIII with funding from FEDER, Europe; by PMP21/00063 from the ISCIII with funding from the FEDER, Europe and PMP22/00049 from the ISCIII with funding from the FEDER, Europe.

Conflict of Interest: None declared

EP10.047 Molecular and Clinical data of two patients with Kabuki syndrome: reporting a novel pathogenic variant

Nada Amllal 1;2, maria zerkaoui3, Wafaa Jdioui3, abdelaziz sefiani1;2, siham chafai elalaoui3, lyahyai jaber1

1Faculty of Medicine and Pharmacy of Rabat, University Mohammed V, Research Team in Genomics and Molecular Epidemiology of Genetic Diseases, Genopath Centre, Rabat, Morocco; 2National Institute of Health, Medical Genetics, Rabat, Morocco; 3Ibn Sina CHU, Medical Genetics Unit, Rabat, Morocco

Background: Kabuki syndrome is a rare genetic disorder characterized by distinct facial features, developmental delay, intellectual disability, and several congenital anomalies. It was named after the traditional Japanese theater due to facial resemblance between the patients’ facial features and the kabuki make up. They often present with long palpebral fissures, arched eyebrows, long eyelashes, and a flat nasal tip. This syndrome is caused by pathogenic variants in genes related to chromatin regulation, mainly the KMT2D gene and less frequently KDM6A gene. Its diagnosis is based on clinical evaluation followed by genetic testing to confirm the diagnosis by identifying the pathogenic variant while its management mainly relies on symptom-specific interventions. In this work, we report molecular and clinical data of two Moroccan children with typical Kabuki syndrome.

Methods: The two patients have typical Kabuki syndrome phenotype. Next Generation Sequencing was performed on both. Conventional Sanger sequencing was performed to confirm the presence of the detected variants.

Results: The first patient harbors the monoallelic missense mutation KMT2D(NM_003482.4):c.4093G>T(p.Val1365Phe) while the second one harbors the novel monoallelic frameshift mutation KMT2D(NM_003482.4):c.15108_15110delp.(His5036_Glu5037delinsGln).

Conclusion: These findings expand phenotypic and molecular spectrums of Kabuki syndrome and emphasize the importance of combining clinical and molecular data for more precise and personalized management of patients with rare disorders.

Conflict of Interest: None declared

EP10.048 Ιdentification of a new variant in PAK3 and its association with intellectual disability type 30

IRATXE HUERTA 1, Mertxe Télez1, maría garcía barcina1

1IMQ Analíticas, Genetics Department, IMQ Analíticas, Bilbao, Spain, Erandio, Spain

Background: The presence of variants in PAK3 gene is associated with intellectual developmental disorder type 30 (OMIM#300558), with X-linked recessive inheritance pattern (XLID30). This pathology is characterized by intellectual disability, aggressive behaviour and self-harm, global developmental delay, language delay, truncal and limb hypotonia, facial hypotonia, microcephaly, elongated face, cerebral atrophy and hypoplasia of the corpus callosum, as the most frequent events.

Methods: An 11-year-old patient was referred to the Clinical Genetics Department for presenting global developmental delay. No neurological history of interest in first-degree relatives. Normal gestation, delivery and neonatal examination. With difficulties in gross and fine psychomotor skills, language disorders, impulsive behaviours and difficulties in relationships with peers. Physical examination revealed, among others, a longilinear habitus, pectum excavatum, slightly winged scapulae, elongated face with large, protruding ears, slight retrognathia and 5th finger bilateral clinodactyly. aCGH and study of FMR1 gene with no significant findings. Clinical exome analysis was recommended to determine the cause of the clinical picture and to offer appropriate genetic counselling.

Results: Variant c.442_445delCATG; p.(His148Aspfs*3) in PAK3 is identified in haemozygosis. This is a loss-of-function variant which generates a premature stop codon. It is categorized as a probably pathogenic variant and it has not been previously described in the literature consulted. Its characteristics and its de novo origin support a probable association with the patient’s phenotype based on current knowledge.

Conclusión: This results supports the causality of the new variant p.(His148Aspfs*3) in PAK3 gene in the patient’s phenotype associated with XLID30.

Conflict of Interest: None declared

EP10.049 Case report: Clinical and genetic analysis of a family with ZFR-related neurodevelopmental disorder

Barbara Golob 1, Mihael Rogac1, Aleš Maver1, Borut Peterlin1

1University Medical Centre Ljubljana, Clinical Institute of Genomic Medicine, Ljubljana, Slovenia

Background/Objectives: ZFR gene has been recently reported in association with a novel neurodevelopmental disorder. Here we present a family of four with 3 affected family members with intellectual disability, dysmorphic features, and autosomal dominant inheritance pattern of frameshift variant in ZFR gene. This family is a part of cohort in international ZFR matchmaking consortium (manuscript in preparation).

Methods: The index case, a 7-year-old girl, presented with mild intellectual disability characterized by motor restlessness, aberrant speech development, short attention span, reduced motivation, low tolerance, and gross motor deficits with dysmorphic facial features (high forehead, wide nasal base, short eyebrows, bulbous nose, short philtrum, and low-set ears). Her mother had learning disabilities (an adjusted primary school curriculum), short stature, and overlapping dysmorphic features. A 9-year-old full brother shared some phenotypic characteristics, including academic struggles marked by reading deficits and retention in the first grade. A 15-year-old half-brother, born of the mother’s first marriage, demonstrated excellent academic performance. Metabolic screening, microarray analyses, whole exome sequencing - trio (WES) and Sanger sequencing were employed.

Results: While metabolic screening and microarrays returned normal results, WES indicated a presence of maternally inherited heterozygous frameshift variant c.157_182dup, p.Ala63fs in ZFR gene. Segregational analysis revealed that the variant has been present in the patient’s mother and one brother, with overlapping clinical presentation. The variant was absent in an asymptomatic brother.

Conclusion: Here we report a variant in ZFR gene, which segregates with a phenotype in a family with three affected members with intellectual disability and dysmorphic features.

Grants: /

Conflict of Interest: None declared

EP10.050 Uncommon 19.p13.2 deletion unmasked ultra-rare autosomal recessive condition – mucolipidosis type IV

Urszula Wysocka 1, Łukasz Kępczyński1, Agata Kucińska1, Katarzyna Połatyńska2, Tadeusz Kałużewski1, Agnieszka Gach1

1Polish Mother’s Memorial Hospital Research Institute, Department of Genetics, Łódź, Poland; 2Polish Mother’s Memorial Hospital Research Institute, Department of Developmental Neurology and Epileptology, Łódź, Poland

Background: Mucolipidosis type IV (OMIM #252650) is an ultra-rare autosomal recessive lysosomal storage disorder with psychomotor developmental delay, visual impairment and achlorhydria. A mutation in the MCOLN1 gene causes an alteration of the protein mucolipin-1 that results in the accumulation of lipids and proteins in cytoplasmic vacuoles derived from lysosomes.

Methods: We present the case of a 6-year-old girl who showed severe intellectual disability, mild dysmorphic features and stereotypic behavior. Prior genetic testing excluded Rett syndrome and Angelman syndrome. Previous aCGH testing was normal.

Results: A hemizygous loss-of-function variant MCOLN1: c.920del (p.Leu307ProfsTer65) was identified in whole exome sequencing. This variant has been reported in ClinVar database as likely pathogenic, and in LOVD database as pathogenic, associated with mucolipidosis type IV. The variant results in frameshift, premature protein synthesis termination and nonsense mediated mRNA decay. Additionally, the uncommon deletion of 180.54 kb in the 19p13.2 region, overlapping the whole MCOLN1 gene coding region was found. The deletion results in the complete absence of the second allele of the MCOLN1 gene, unmasking the autosomal recessive disease. Prior aCGH testing did not reveal this deletion, due to poor representation of the deleted region.

Conclusion: Whole exome sequencing with copy number variation analysis is an important tool for genotype-phenotype correlation, precision in diagnosis, and in-depth understanding of the disease mechanisms.

Conflict of Interest: None declared

EP10.051 Genetic counselling of patients with intellectual disability and autism spectrum disorders.

Riina Žordania 1;2, Kati Kuuse3, Ülle Murumets3, Edgar Tarelkin4, Laura Roht2

1Tartu University Hospital, Genetics and Personalized Medicine Dept, Tallinn, Estonia; 2Tartu University Hospital, Genetics and Personalized Medicine Clinic, Tallinn, Estonia; 3Tartu, Genetics and Personalized Medicine Clinic, Tartu, Estonia; 4Tallinn Children’s Hospital, Children’s Psychiatry, Tallinn, Estonia

Introduction

Intellectual disability (ID) and autism spectrum disorders (ASD) forms a large part of patients in genetic counselling. ID and ASD are both genetically highly heterogeneous and may be inherited or de novo. ASD diagnosis often co-occurs with ohter conditions.

Aim

We gathered 72 patients aged 1-38 years consulted by geneticist due to ID and ASD. Different diagnostic methods (chromosomal microarray, NGS and metabolic workup) were used.

Results

Data were analysed in different age groups (0-3; 3-7; 7-18 years).

Complicated fertility and/or pedigree were in 60% of families. Pathogenic/likely pathogenic/variant of unknown significance (VOUS) copy number alterations were detected in 12%. NGS panel detected inherited syndromes in 40% of patients: Cornelia de Lange, Kleefstra, Smith-Kingsmore, SCA21, etc. Trio exome analysis was performed in eleven families and two patients got a final diagnosis.

Conclusions

1. Etiological diagnosis was found in half of the cases (47,2%).

2. Patients with ID, ASD and normal chromosomal microarray, NGS is indicated.

3. Final etiological diagnosis allowed correct genetic counselling in those families.

Conflict of Interest: None declared

EP10.052 First report of a de novo missense variant in SRCAP in a child with neurodevelopmental delay and musculoskeletal abnormalities

Zianka Meyer 1, Stefan Weiß1, Douglas Friday1, Sophia Kummerow1, Irina Hüning2

1Diagenom GmbH, Rostock, Germany; 2MVZ für Humangenetik & Molekularpathologie, Rostock, Germany

Background/Objectives: Heterozygous truncating de novo variants in the SRCAP gene and distinct methylation signatures have been associated with Floating-Harbor syndrome (FLHS) and with developmental delay, hypotonia, musculoskeletal defects, and behavioral abnormalities (DEHMBA).

Methods: Clinical examination and whole-exome sequencing (WES) of a patient with neurodevelopmental delay, dysmorphic facial features, and congenital anomalies was performed.

Results: The 15-year-old boy was found to be affected by global developmental delay as well as behavioural abnormalities. Additional features included hypotonia, mandibular prognathia, narrow jaw, pectus excavatum, pes planus, arachnodactyly, and abnormality of connective tissue with striae distensae. WES revealed a heterozygous likely de novo missense variant in the SRCAP gene, NM_006662.3: c.3670C>A (p.Arg1224Ser).

Conclusion: So far only truncating variants in the SRCAP gene have been described as pathogenic. Here we report on the first patient with clinical features consistent with DEHMBA and a likely de novo missense variant in the SRCAP gene, classified as likely pathogenic (class 4) according to the ACMG standards and guidelines, raising the question of the role of missense variants in the expression of SRCAP-associated phenotypes.

Grants: The authors received no financial support for the research, authorship and publication of this report.

Conflict of Interest: None declared

EP10.054 A heterozygous de novo variant in CAPRIN1 potentially causing high-output chylothoracis with severe respiratory adjustment disorder and status epilepticus

Anna-Sophie Liegmann 1, Kirstin Hoff2, Britta Hanker1, Malte Spielmann1;2, Inga Nagel2

1Institute of Human Genetics, University of Lübeck, Lübeck; 2Institute of Human Genetics, University of Kiel, Kiel

Background/Objectives: The cytoplasmic activation/ proliferation-associated protein 1 is encoded by CAPRIN1 and plays a crucial role in the gene regulation of cell cycle control-associated genes. In a recent study, 12 patients with CAPRIN1 variants were described with language impairment in all cases, and some showing respiratory problems as well as seizures.

Methods: Trio-exome sequencing was conducted on a patient and her unaffected parents, uncovering a heterozygous de novo variant in CAPRIN1. Subsequently, a re-analysis of 2441 exomes from the Institute of Human Genetics at the University of Kiel focused exclusively on variants in the CAPRIN1 gene was performed.

Results: In the exome of the initial patient, a five month old girl, the variant NM_005898.5:c.879+3A > G in CAPRIN1 was identified as de novo. The girl showed a severe respiratory adjustment disorder, high-output chylothoracis, persistent foramen ovale, late-onset-sepsis, and status epilepticus. The re-analysis of 2441 exomes revealed two promising variants in CAPRIN1. The first variant, NM_005898.5:c.494G>A, was found in the exome of a 13-year-old boy with neurodevelopmental delay, seizures, microcephaly, abnormality of movement, and brain atrophy. The second variant, NM_005898.5:c.1506T>G, was carried by a 9-year-old boy with neurodevelopmental delay, behavioral abnormality, ventricular septal defect, and microcephaly.

Conclusion: The phenotypes of two of the three presented patients with potentially protein damaging CAPRIN1 variants overlap with the published CAPRIN1-associated phenotypes. Interestingly, the phenotype of the third patient additionally contains chylothoracis, persistent foramen ovale and late-onset-sepsis, which has not been described in association with CAPRIN1 in the literature yet.

Conflict of Interest: None declared

EP10.055 Clinical features and genetic findings in 15 unrelated families with KBG syndrome

Juan Pablo Trujillo-Quintero 1, Carmen Manso2, Elisabeth Gabau Vila1, Neus Baena1, Núria Capdevila1, Anna Brunet-Vega1, Victor Martinez-Glez1, Nino Spataro1, Anna Ruiz1

1Center for Genomic Medicine, Parc Taulí Hospital Universitari. Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA). Universitat Autònoma de Barcelona. Sabadell, Spain.; 2Servei de Medicina Pediàtrica, Parc Taulí Hospital Universitari. Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA). Universitat Autònoma de Barcelona. Sabadell, Spain

Background/Objectives: KBG syndrome is caused by loss-of-function variants in the ANKRD11 gene and is characterized by dysmorphic features, macrodontia, developmental delay/intellectual disability (ID), skeletal anomalies, short stature, and behavioral disorders.

Methods: Array comparative genomic hybridization followed by exome sequencing was used in patients with neurodevelopmental disorders/intellectual disability. Methylation episignature testing was performed to confirm KBG diagnosis in one patient.

Results: We present a clinical and molecular characterisation of 15 patients affected by KBG syndrome. Among genetic variants identified, 13 were de novo, 11 nonsense or frameshift variants, and 2 intronic variants. One patient was carrier of a partial gene duplication. All variants were classified as likely pathogenic/pathogenic following ACMG guidelines.

Clinical description of patient’s features showed that all patients presented dental, craniofacial, skeletal and behavioural anomalies (autism spectrum disorder, attention deficit hyperactivity disorder, others). Only 4 patients presented short stature. Mild to moderate ID was present in 3 patients while 7 patients were affected by borderline intellectual disability. Only one patient presented severe ID comorbid with a severe psychiatric disorder.

Conclusion: We describe 15 additional KBG patients. Among them one presents severe intellectual disability comorbid with a severe psychiatric disorder. Further studies are needed to determine whether there is an additional genetic diagnosis.

The episignature analysis highlights its utility to determine the pathogenicity of uncertain variants on the ANKRD11 gene.

Conflict of Interest: None declared

EP10.056 Identification and functional characterization of a novel variant in SYNRG gene causing intellectual disability

Ramya Sukrutha 1, ananthapadmanabha kotambail1, GAUTHAM ARUNACHAL UDUPI1, CHETAN GHATI1

1National Institute Of Mental Health & Neuro Sciences (NIMHANS), Department of Human Genetics, Bengaluru, India

Background/Objectives: Synergin, Gamma (SYNRG) gene is associated with rare phenotypes of intellectual disability and hereditary spastic paraplegia. Only a few cases have been reported worldwide. mRNA harbouring premature termination codons (PTCs) are known to be rapidly degraded by nonsense-mediated mRNA decay (NMD), which limits the production of truncated proteins. PTCs that escape from NMD can produce defective protein products that have detrimental effects. Here we report a case of intellectual disability with a PTC that escapes NMD contributing to the phenotype.

Methods: Exome sequencing had revealed a novel homozygous stop gained variant NM_007247.6:c.3491C>A (NP_009178.3:p.Ser1164Ter) in SYNRG gene with predicted protein truncation. RNA was extracted from peripheral blood along with an age matched control. Transcriptome sequencing was performed to ascertain the effect of the rare variant on NMD after appropriate consenting process. Western blot analysis for the quantification of protein is in progress.

Results: The variant is predicted to cause loss of normal protein function through protein truncation. This variant is a stop gained variant which occurs in exon 18 of SYNRG upstream of where NMD is predicted to occur. Analysis of transcriptome sequencing did not show transcript differential expression indicating a possible escape of NMD.

Conclusion: Our study identifies PTCs where NMD is predicted to occur yet can lead to mendelian phenotypes due to a truncated protein with more than 10% removal of the C terminal domain. It emphasises the critical role of functional analysis such as transcriptome and protein-based analysis in understanding the molecular pathogenesis of ultra-rare variants.

Grants:

Conflict of Interest: None declared

EP10.057 Understanding the role of FAM120A Gene in a Neurodevelopmental Disorder

ABDULLAH SEZER1, Damla Puçak 2, Sebiha Cevik2, Oktay I. Kaplan2

1Ankara Etlik City Hospital, Department of Medical Genetics, Ankara, Türkyie; 2Abdullah Gul University, School of Life and Natural Sciences, Kayseri, Türkyie

Background/Objectives: The family with sequence similarity 120 member A (FAM120A) gene encodes a protein with RNA-binding activity and is predicted to participate in mRNA transport in the cytoplasm. The protein has been associated with the activation of multiple kinases.

However, while FAM120A has been previously linked to schizophrenia and found to be expressed in the central nervous system, its functional characterization in neural tissues and its possible relation to neurodevelopmental disorders remain unexplored. Here, we describe a patient carrying a potentially null variant in the FAM120A gene, exhibiting a neurodevelopmental disease phenotype.

Methods: The patient was a 7-year-old male with mild intellectual-motor delay, behavioral abnormalities, epilepsy, optic disc hypoplasia, and strabismus. In the WES analysis, a homozygous nonsense variant (NM_014612, c.1824delT, p.Tyr608Ter) was identified in the FAM120A gene. To elucidate the effects of FAM120A loss, especially in neurons, we employed CRISPR/Cas9 to create a null mutant in the nematode Caenorhabditis elegans.

Results: The genotypic data were uploaded to several genetic networks (GeneMatcher and Matchmaker Exchange). Unfortunately, a second case could not be identified through this process. Currently, we are investigating whether FAM-120A null mutants exhibit functional and structural defects in neurons.

Conclusion: Neurodevelopmental disorders, affecting people of all ages, exhibit varied symptoms, including intellectual disability and disruptions in behavior, communication, and social interaction. Despite identifying many associated genes, our understanding of their genetic basis is incomplete. In this context, we suggest that the FAM120A gene may potentially act as a causal gene in the etiology of neurodevelopmental disorders.

Grants: None

Conflict of Interest: None declared

EP10.058 Broadening the spectrum of TAF4 related neurodevelopmental disorder (T4NDD)

Noel Macmanus1, Asunción Díaz1, Alícia Artigas Baleri2, Lucia Dougherty de Miguel1, Eulàlia Turón-Viñas1, Susana Boronat1, Ivon Cuscó 2;3

1Paediatric Neurology, Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain., Spain; 2Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), Genetics Department. Hospital de la Santa Creu i Sant Pau, Barcelona, Spain., Barcelona, Spain; 3Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain

Background/Objectives: TAF4 variants have been related to Autosomal dominant intellectual developmental disorder-73 (MRD73) (MIM# 620450)- This new association was made possible thanks to the identification of de novo putative loss-of-function variants in 10 patients with neurodevelopmental disorder (NDD). The aim of this case report is to broaden the knowledge of this disorder by presenting a new case.

Methods: We performed a detailed review of clinical manifestations of a patient in whom we identified a de novo putative loss-of-function variant in TAF4 by whole exome sequencing. Family and personal history, psychomotor development, phenotypic features, MRI images and genetic studies were collected.

Results: We have identified a de novo TAF4 variant (c.3157C>T:p.Q1053X) in a 3-year-old patient with global developmental delay, especially in speech, facial dysmorphisms (arched eyebrows, synophrys, upslating palpebral fissures, prominent nose, full lips and retrognathia) and joint laxity. An intrauterine growth retardation were detected during pregnancy. The birth was premature (34 weeks) and with low weight for gestational age (1550 grams). A brain MRI showed a diffuse cerebral atrophy with mild supraventricular dilatation.

Conclusion: Use of whole exome sequencing in the diagnosis of NDD has allowed the identification of new genes associated with this pathology. The identification of a new case with a de novo putative loss of function variant in the TAF4 gene and his similar phenotype as other patients reported, reinforces the role of this gene in the recently described syndrome.

Grants:

Conflict of Interest: None declared

EP10.059 Clinical and molecular perspectives of Pitt Hopkins syndrome: case series.

Marta Domínguez Jiménez 1, Ana Teresa Serrano Antón1, María José Sánchez Soler1, Julia Araújo De Castro2, Sonia Martínez Peñas2, Vanessa Lopez1;3, María Juliana Ballesta Martínez1;3, Encarna Guillén-Navarro1;3

1Virgen of Arrixaca University Clinical Hospital, Medical Genetics, El Palmar, Spain; 2Virgen of Arrixaca University Clinical Hospital, Pediatrics, El Palmar, Spain; 3Carlos III Health Institute, Madrid, Spain

Background/Objectives: Pitt-Hopkins syndrome (PTHS) is characterized by developmental delay with moderate to severe intellectual disability, behavioural and sleep disturbances, altered breathing pattern and specific facial features, among others.

It is due to haploinsufficiency of the TCF4 gene, and typically results from heterozygous de novo pathogenic variants or deletions.

We propose the clinical and molecular characterization of patients diagnosed with PTHS.

Methods: Retrospective descriptive study of patients diagnosed with PTHS in the Genetics section of a tertiary hospital between 2015 and 2023.

Results:

Table 1. Epidemiology and molecular diagnosis.

Individual-1

Individual-2

Individual-3

Individual-4

Age at diagnosis (years)

5

3

3

2

Sex

Heterozygous TCF4 variant

p.G340fs

(de novo)

18q21.2 (0.11Mb) deletion

(de novo)

18q21.2 (5.7Mb)

deletion

(de novo)

p.(Ala614Val)

Table 2. Clinical characteristics.

Indivi-dual-1

Indivi-dual-2

Indivi-dual-3

Indivi-dual-4

Total

Craniofacial features

Microcephaly

+

-

+

-

2/4

Characteristic facial features

+

+

+

+

4/4

Neurological

Hypotonia

-

+

+

+

3/4

Developmental delay

+

+

+

+

4/4

Speech delay

+

+

+

+

4/4

Seizures

-

-

-

-

0/4

Behavioral

+

+

+

+

4/4

Breathing abnormalities

+

-

-

-

1/4

Gastrointestinal

-

-

+

-

1/4

Musculoeskeletal

+

+

+

+

4/4

Ophthalmological

-

+

+

+

3/4

Conclusion: This series reaffirms key features of PTHS, such as distinct facial characteristics and developmental delays. Low breathing abnormality prevalence may be age-related. No seizures and a lower microcephaly rate were observed, contrasting literature.

Comprehensive clinical and molecular data improve the understanding of PTHS, facilitating early recognition and diagnosis.

Conflict of Interest: None declared

EP10.060 De novo variants in SP9 cause a novel form of interneuronopathy characterized by intellectual disability, autism spectrum disorder, and epilepsy with variable expressivity

Marine Tessarech 1;2, Gaelle Friocourt3, Florent Marguet4, Maryline Lecointre4, Le Mao Morgane2, Cyril Mignot5, Boris Keren5, Benedicte Heron6, Charlotte De Bie7, Koen Van Gassen7, Didier Loisel8, Benoit Delorme8, Steffen Syrbe9, Rodrigo Muñoz Díaz2, Klabunde-Cherwon Annick9, Rami Abou Jamra10, Meret Wegler10, Bert Callewaert11, Annelies Dheedene11, Merzouka Zidane-Marinnes12, Agnes Guichet1;2, Céline Bris1;2, Patrick Van Bogaert13, Florence Biquard14, guy lenaers2, Pascale Marcorelles15, Claude Férec3, Bruno Gonzales4, Vincent Procaccio1;2, Antonio Vitobello16, Dominique Bonneau1;2, Annie Laquerriere4, Salim Khiati2, Estelle Colin1;2

1Angers University Hospital, Department of Medical Genetics, Angers, France; 2University of Angers, Mitovasc Unit, UMR CNRS 6015 INSERM 1083, Angers, France; 3Brest University, Inserm UMR 1078, Brest, France; 4Rouen University Hospital, Department of Pathology, Rouen, France; 5Assistance Publique-Hôpitaux de Paris, Department of Genetics, Paris, France; 6Trousseau Hospital, Department of Pediatric Neurology, Paris, France; 7University Medical Center Utrecht, Department of Genetics, Utrecht, Netherlands; 8Angers University Hospital, Department of Radiology, Angers, France; 9Medical Faculty of Heidelberg, Division of Pediatric Epileptology, Heidelberg, Germany; 10University of Leipzig Hospitals and Clinics, Institute of Human Genetics, Leipzig, Germany; 11Center for Medical Genetics, Department of Biomolecular Medicine, Gent, Belgium; 12Angers University Hospital, Department of Pathology, Angers, France; 13Angers University Hospital, Department of Neuropediatrics, Angers, France; 14Angers University Hospital, Department of Gynecology, Angers, France; 15Brest University Hospital, Department of Pathology, Brest, France; 16CHU Dijon Bourgogne, Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, Dijon, France

Background/Objectives: Interneuronopathies are a group of neurodevelopmental disorders characterized by deficient migration and differentiation of GABAergic interneurons resulting in a broad clinical spectrum, including autism spectrum disorders, early-onset epileptic encephalopathy, intellectual disability, and schizophrenic disorders. SP9 is a transcription factor belonging to the Krüppel-like factor and specificity protein family, the members of which harbor highly conserved DNA binding domains. SP9 plays a central role in interneuron development and tangential migration, but it has not yet been implicated in a human neurodevelopmental disorder.

Methods: Cases with SP9 variants were collected through international data-sharing networks. To address the specific impact of SP9 variants in silico and in vitro assays were carried out.

Results: De novo heterozygous variants in SP9 cause a novel form of interneuronopathy. SP9 missense variants affecting the Glutamate 378 amino acid result in severe epileptic encephalopathy due to hypomorphic and neomorphic DNA-binding effects, whereas SP9 loss-of-function variants result in a milder phenotype with epilepsy, developmental delay, and autism spectrum disorder.

Conclusion: De novo heterozygous SP9 variants are responsible for a neurodevelopmental disease. Interestingly, variants located in conserved DNA-binding domains of KLF/SP family transcription factors may lead to neomorphic DNA-binding functions resulting in a combination of loss- and gain-of-function effects.

Grants: None

Conflict of Interest: None declared

EP10.061 Catatonia in Kleefstra syndrome requires multidisciplinary management

Solveig Heide1, Adrien Legrand2, Cristina Alina Birzu3, Daphné Lehalle1, Romain Duquet1, Anna Gerasimenko1, Delphine Heron1, Perrine Charles 1

1Salpêtriere Hospital, Genetic department, reference center for intellectual disabilities, Paris, France; 2Ste Anne Hospital, Psychiatric department, reference center for rare diseases with psychiatric manifestations, Paris, France; 3Salpêtriere Hospital, Neurologic department, ICM Paris Brain Institute, Paris, France

Catatonia in Kleefstra syndrome requires multidisciplinary management.

Kleefstra syndrome is a rare genetic disorder caused by haploinsufficiency of the EHMT1 gene, characterized by the core clinical phenotype of intellectual disability, hypotonia, severe speech delay, and distinct facial characteristics with additional clinical features including sleep disturbance, overweight, psychiatric disorders, and autism spectrum disorder. To date, a limited number of case reports of Kleefstra syndrome with psychiatric manifestations including psychosis have been reported, often accompanied by functional regression.

We report three young adults including two twin sisters who develop psychiatric catatonia-like manifestations associated with regression. For one case diagnosis of dysimmun encephalitis was evoked. Therapeutic treatment has shown lack of response to traditional treatments for catatonia and strategy was ultimately based on anti-psychotic medications and encephalitis management.

These cases illustrate the importance of psychiatric comorbidities in Kleefstra syndrome, difficulties in therapeutic care with a need for multidisciplinary management and the importance of carrying out an exhaustive etiological assessment before linking psychiatric features to Kleefstra syndrome.

Conflict of Interest: None declared

EP10.062 Two North African siblings with MEF2C-related disorders due to parental Germline Mosaicism

Aïcha Boughalem 1, aziza belkhayate2, Laurence Lohmann1, Mylene Valduga1, Pascale Kleinfinger1, Aline Receveur1, stéphane serero3, Detlef Trost1

1CERBA, Department of Human Genetics, Frépillon, France; 2Laboratoire D’Analyses Medicales Biolab, Génétique, Rabat, Morocco; 3Cerba HealthCare, Issy-les-Moulineaux, France

Aim: MEF2C haploinsufficiency syndrome is a severe neurodevelopmental disorder that is not easily recognized clinically. We report Two north African siblings with MEF2C-related disorders due to parental mosaicism.

Methods: We present the case of two siblings from a non-consanguineous North African family, both exhibiting MEF2C-related disorders. The older sister, aged 8 at the first consultation, displayed symptoms from the age of 3, including global developmental delay, lack of speech, intellectual disability, and strabismus. Notably, there was an absence of epilepsy. A milder phenotype was observed in the younger sister.

Results: Exome sequencing in 2022 identified a heterozygous splice variant in the MEF2C gene in the older sister, classified as probably pathogenic according to the American College of Medical Genetics.

The identified variant, not documented in Clinvar or HGMD pro databases, is novel in the current literature. Family segregation analysis disclosed parental mosaicism, indicating the presence of the variant in an estimated 20% of the asymptomatic father’s blood cells. This finding was further validated through targeted Sanger analysis.

The affected younger sister also displayed the splice variant in a heterozygous state with a less severe intellectual disabilities.

Conclusion: This additional reported case contributes to expanding the cohort of patients with MEF2C point variations, highlighting the significance of such variations, particularly among underrepresented North African patients. It represents only the second reported instance of parental mosaicism (PMID: 34055696), emphasizing the crucial role of genetic counseling within this family.

Keywords: MEF2C haploinsufficiency syndrome, Exome sequencing, Germinal mosaicism, MEF2C gene mutations, North African variant.

Conflict of Interest: None declared

EP10.063 Novel hemizygous variant in NONO gene identified in two stepbrothers

Ilona Jaszczuk 1, Monika Lejman2, Borys Styka2, Maciej Geremek3, Beata Nowakowska3, Izabela Winkler4, Agata Filip1

1Medical University, Department of Cancer Genetics with Cytogenetic Laboratory, Lublin, Poland; 2Medical University, Laboratory of Genetic Diagnostics II Chair of Paediatrics, Lublin; 3Institute of Mother and Child, Department of Medical Genetics, Warsaw, Poland; 4St. John’s Center of Oncology of the Lublin Region, Second Department of Gynecological Oncology, Lublin

Background/Objectives: The X-linked syndromic intellectual developmental disorder-34 (MRXS34, OMIM # 300967) is a rare genetic developmental disorder, firstly described in 2015. MRXS34 is characterized by delayed psychomotor and speech development, intellectual disability, dysmorphic facial features and congenital brain defects. The cause of this syndrome are constitutive hemizygous variants of the NONO gene (Xq13.1). We present two 3-year and 17-year old stepbrothers with recognized MRXS34 and confirmed variant in the NONO gene.

Methods: In the diagnostic process of the younger brother, microarray SNP 750K and whole-exome sequencing (WES) were performed. In the case of the older brother, only WES was performed.

Results: In the case of the younger brother, molecular analysis using microarray method did not reveal any genomic imbalances in the examined regions. WES analysis allowed the identification of a new hemizygous variant c.1131+3A > G in the NONO gene, which was also detected in the case of the older brother. This variant has not been previously reported in the ClinVar Database. The mutation was confirmed by Sanger sequencing. The patients’ mother was confirmed to be a carrier of the c.1131+3A > G variant of the NONO gene.

Conclusion: In the case of patients with intellectual disability and characteristic dysmorphic features, MRXS34 should be taken into account in the differential diagnosis, especially in the case of X-linked disease. Tests using the WES technique facilitate diagnosis and enable the detection of new pathogenic variants in genes. Patients with MRXS34 require multidirectional therapy with individualized education.

Grants:

Conflict of Interest: None declared

EP10.064 Clinical-genetic approach to syndromic conditions with macrocephaly and ASD: variants in PTEN and PPP2R5D are the most recurrent gene mutations in a patient-oriented diagnostic strategy

Marina Carapelle1, Silvia Frangella1, Anna Gloria Renzi1, Domizia Pasquetti1, Elena Sonnini1, Annalisa Gazzellone1, Clarissa Modafferi1, Pino D’Ambrosio1, VALENTINA TREVISAN1;2, Arianna Panfili3, Paolo Niccolò Doronzio1, Giuseppe Marangi1, Daniela Orteschi1;4, Emanuela Lucci Cordisco4, Pietro Chiurazzi4, Ilaria Contaldo5, Chiara Veredice6, Federica Francesca L’Erario 1, Marcella Zollino4;7

1Section of Genomic Medicine, Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Roma; 2Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; 3Scientific Directorate, Policlinico Universitario “A.Gemelli” Foundation IRCCS, Rome, Italy; 4Medical Genetics Unit, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; 5Child Neurology and Psychiatric Unit, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Roma; 6Pediatric Neuropsychiatry Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; 7Department of Life Sciences and Public Health, Catholic University, Rome, Italy

Macrocephaly can be an isolated, benign trait or a clinical feature of several monofactorial conditions, often in association with generalized or segmental overgrowth, intellectual disability (ID), or autism spectrum disorder (ASD). On the other hand, primary ASD can present with developmental delay and relative macrocephaly.

We tested by means of next-generation sequencing (NGS) of a panel of 27 genes (ABCC9, AKT1, AKT2, AKT3, BRWD3, DIS3L2, DNMT3A, EZH2, GPC3, GPC4, HERC1, MED12, MTOR, NFIA,NFIX, NSD1, PDGFRB, PIK3CA, PIK3R1, PIK3R2, PPP2R1A, PPP2R5D, PTEN, RAB39B, RNF135, SETD2 and TBC1D7) a cohort of 75 patients (62 males and 13 females), with ID/ASD and a nonspecific clinical presentation including accelerated growth/overgrowth, relative or true macrocephaly and additional few clinical manifestations.

Two patient’s categories were selected: A) with true macrocephaly and ID with or without ASD (tot 38) and B) with relative macrocephaly and ASD (tot 37). A search for PTEN variants was mainly planned in group B). Pathogenic variants were only identified in the first group, with 12 patients/38 (32%) (6 males and 6 females), and included variants in PTEN (4 cases), PPP2R5D (4 cases), NSD1 (3 cases), MTOR (1 case).

Conclusions. Variants in PTEN and PPP2R5D accounts for a high percentage of gene mutations (66% in our cohort) in a selected patient’s series with ASD/ID, true macrocephaly, and few additional, nonspecific features. The diagnostic yield of the present highly focused gene panel is similar to the WES. Suggestions for clinical applicability of the present gene panel and the clinical spectrum of the PTEN and PPP2R5D associated disorders are discussed.

Conflict of Interest: None declared

EP10.065 A large four-generation family with neurodevelopmental disorders related to a synonymous variant in FRMPD4

stéphanie moortgat 1, Anne Destree1, Deniz Karadurmus1, Olivier Monestier1

1Institut de Pathologie et de Génétique, Centre de Génétique Humaine, Gosselies

Variants in FRMPD4 are associated with mental retardation, X-linked 104 (MRX104, OMIM 300983). The gene encodes the FERM and PDZ domains containing 4 (FRMPD4) protein. The protein, highly expressed in the brain, regulates the formation of excitatory synapses and dendritic spines. To date, 15 variants in FRMPD4 have been identified in 21 patients. Various neurodevelopmental disorders were described in > 50% of affected male patients including developmental delay, intellectual disability, epilepsy and/or behavioral abnormalities.

We describe a four-generation Belgian family in which 8 affected male patients and 5 affected females present with developmental delay, intellectual disability and behavioral anomalies. Whole exome sequencing in two affected brothers revealed the hemizygous c.1470G>A, p.(Lys490 = ) variant in FRMPD4. It was absent from gnomAD database, implicated the last nucleotide of exon 13/17 but was not definitely predicted to impact splicing of the FRMPD4 transcript. We performed splicing analysis with RNA extracted from proband’s fibroblasts cells culture and showed a loss of exon 13 leading to the in-frame loss of several amino acids in the FERM functional domain of the protein (p.Gln430_Lys490del). We confirmed the segregation of the variant with the phenotype in all affected siblings (absent in 3 healthy males, present in 2 healthy female carriers).

In conclusion, we report the first likely pathogenic synonymous variant and its functional impact in FRMPD4-related disorders. We discuss the phenotypic spectrum of the disease and compare the patients with those previously reported. We highlight intrafamilial clinical variability, in particular in females ranging from asymptomatic to moderate intellectual disability.

Conflict of Interest: None declared

EP10.066 First report of EBF3 neurodevelopment disorder from Southeast Asia: A case report and review of literature

chitra ramalingam1, Jiin Ying Lim 2;3;4, sylvia kam2;3;4, Hai Yang Law2, nur afiqah binte mohd mislan2, weng khong lim3;5, ivy ng2, terrence thomas6, sandra sylvia mascarenhas1, saumya shekhar jamuar2;3;4;5

1KK Women’s and Children’s Hospital, Child Development, Singapore; 2KK Women’s and Children’s Hospital, Genetics service, Paediatrics, Singapore, Singapore; 3SingHealth Duke-NUS Genomic Medicine Centre, Singapore, Singapore; 4Duke-NUS Medical School, Paediatrics Academic Clinical Programme, Singapore, Singapore; 5SingHealth Duke-NUS Institute of Precision Medicine, Singapore; 6KK Women’s and Children’s Hospital, Neurology, Paediatrics, Singapore

Background/Objectives: Neurodevelopmental disorders (NDD) are a group of conditions that are heterogenous in their clinical presentation, and identifying a genetic cause can be difficult. The use of next generation sequencing has helped in identifying disease-gene associations in patients presenting with nonspecific clinical features. EBF3 neurodevelopmental disorder (EBF3-NDD) is a recently described NDD characterized by hypotonia, global developmental delay (GDD), intellectual disability (ID), gait or truncal ataxia, seizures, behavioural problems and facial dysmorphism1.

Case presentation: We report a 12-year-old Malay female, who is the second child born to non-consanguineous parents. During initial review at 2 months of age, she was noted to have hypotonia, soft dysmorphic features and heart murmur. Echocardiograms identified moderate peri membranous ventricular septal defect (VSD), small atrial septal defect (ASD), and patent foramen ovale (PFO). On further reviews, she had significant motor delay with hypotonia, GDD, ataxia, pes planus, articulation difficulty, behavioural difficulty with short attention span, mild ID, and short stature with failure to thrive.

Initial investigations include normal Brain MRI and normal karyotype with negative FISH test for 22q11.2 deletion syndrome. Chromosomal microarray analysis (CMA) detected a gain of 1.608 Mb at Xp22.31, which is of uncertain significance, likely benign. Whole exome sequencing identified a heterozygous likely pathogenic variant c.196A>G(p.Asn66Asp) in EBF3 (NM_001375391.1). This variant is absent in population and has been observed in another individual with EBF3-NDD1.

Conclusion: EBF3-NDD and its associated phenotype continues to evolve. Identification of this variant is beneficial in further evaluation, surveillance, and management of our patient.

Conflict of Interest: None declared

EP10.068 New frameshift variant in METTL5 gene in a case of microcephaly and neurodevelopment delay: a possible founder effect in a spanish southeast village.

ANGEL ZUNIGA 1, Graciela Pi2, ANTONIO MARTINEZ CARRASCAL3

1HU La Ribera, GENÉTICA, Alzira (Valencia), Spain; 2HU La Ribera, PEDIATRÍA, Alzira (Valencia), Spain; 3HG de Requena, PEDIATRÍA, Requena (Valencia), Spain

Background: Intellectual disability (ID) is a highly heterogeneous disorder, affecting 1–3% of the world’s population, which is associated with a significant alteration in cognitive development, adaptive functioning and behavioral problems. The list of monogenic forms of ID has increased rapidly in recent years thanks to the implementation of NGS (more than 2500–3000 genes associated with different forms of ID). Recently, N6-methyladenosine (m6A) has been found abundantly in human messenger RNAs (mRNAs) and it has been shown to regulate most stages of mRNA metabolism. METTL5 is one of the members of the methyltransferase-like protein family and it may have a global epigenetic regulatory role in the brain in addition to its synapse-dependent role.

Results: We report a 13-month-old girl with primary microcephaly, intrauterine growth retardation and dysmorphic traits, with a novel frameshift variant in METTL5 gene using whole-exome sequencing (WES).

In MRI a delay in myelination was observed for the patient’s age, with less myelination than expected in the anterior arm of the internal capsules and in semioval centers. There are no other morphological or signal intensity alterations in the brain, cerebellar or brainstem parenchyma. The morphology and size of the ventricular system are appropriate for the patient’s age.

Non-consanguineous healthy parents were heterozygous for this variant and both from a village in southeast Spain.

Conclusion: This frameshift variant has not been previously reported in METTL5 gene. More members of the respective families need to be analyzed to see if it could be a variant with a founder effect.

Grants:

Conflict of Interest: None declared

EP10.069 A comparative analysis of diagnostic effectiveness between aCGH and New Generation Sequencing in cases with neurodevelopmental disorders and/or congenital anomalies

Arzu Guliyeva 1;2, Elif Yilmaz Gulec1;2, Filiz Ozen2

1Istanbul Medeniyet University, Faculty of Medicine, Medical Genetics Department, Istanbul, Türkyie; 2Goztepe Prof.Dr. Suleyman Yalchin City Hospital, Medical Genetics, Istanbul, Türkyie

Background/Objectives: The integration of genomic tests, Microarray-based Comparative Genomic Hybridization (aCGH) and Next Generation Sequencing (NGS), has significantly advanced the diagnosis of neurodevelopmental disorders (NDD). aCGH, now a primary assessment tool, particularly excels in cases of intellectual disability, multiple malformations, and autism, owing to its heightened yield and resolution. In parallel, NGS, encompassing targeted panel testing, exome sequencing, and whole genome sequencing, successfully concludes diagnostic odysseys for 25-30% of unselected cases with rare monogenic syndromes, especially in genetically heterogeneous conditions.

Methods: 150 clinically diagnosed patients,within the age range of 5 months to 24 years, with NDD’s and/or congenital anomalies were referred to us. They were categorized into global developmental delay/intellectual disability (GDD/ID), autism spectrum disorders (ASD), other NDD’s and congenital anomalies (CA). Initially, aCGH was performed for the patients. For cases where results were inconclusive or showed variations of uncertain significance (VUS), we proceeded with further testing using NGS methods.

Results: Through the aCGH method, pathogenic copy number variations were identified in 6% of the patients (n = 9). The chromosome locations were 16p11.2, 11q24.2q25, 2p16.3(n = 2), 15q13.2q13.3, 7q36.3, 6q27, 1q21.1q21.2, Xq28. The remaining patients were analyzed using the NGS method, revealing pathogenic and likely pathogenic variants in 19% of the cases (n = 27). Overall, pathogenic and clinically related VUS variants were detected 27,3% by aCGH versus 38,7% by NGS.

Conclusion: Exome sequencing outperformed aCGH in all instances. According to these results, in addition to aCGH, the use of exome sequencing methods is also deemed necessary as a first-tier test.

Grants: None

Conflict of Interest: None declared

EP10.070 From uncertainty to clarity in diagnosing autism and developmental delay: a tale of two cases

Ivan Tourtourikov1;2, Malina Stancheva3, Tanya Kadiyska 2;4

1Medical University of Sofia, Department of Medical Chemistry and Biochemistry, Sofia, Bulgaria; 2Genetic Medico-Diagnostic Laboratory Genica and Genome Center Bulgaria, Sofia, Bulgaria, Sofia, Bulgaria; 3Mediva Medical Centre, Sofia, Bulgaria; 4Medical University of Sofia, Department of Physiology and Pathophysiology, Sofia, Bulgaria

Background/Objectives: Autism spectrum disorder (ASD) and intellectual disability (ID) with or without developmental delay (DD) represent the most common chronic mental conditions encountered in pediatric care. Identification of an underlying genetic cause has the potential to direct patients and families to personalized management and condition-specific resources and supports. Here we present our findings in two unrelated cases with suspected ASD, in which genetic testing was able to clarify the diagnosis.

Methods: Whole exome sequencing (WES) was carried out on both patients. Segregation analysis and variant confirmation was performed via Sanger sequencing.

Results: Case 1: 13-year old male proband with developmental delay, delayed speech and language development. MRI showed nonspecific gliosis, with abnormal slow activity from EEG. WES demonstrated a homozygous missense variant – FOLR1:c.427T>A, p.W143R (OMIM #P 613068). Both parents were revealed to be heterozygous asymptomatic carriers.

Case 2: 12-year old female proband with ASD, presenting with mild to moderate developmental delay. Array CGH was normal. Quantitative EEG showed dysregulation in the left superior and middle temporal gyrus. WES revealed a heterozygous missense variant in the KCNJ11 gene, c.808C>A, p.L270M (OMIM #P 618856). Segregation analysis showed that the variant was inherited from the mother, who did not display neurological symptoms.

Conclusion: Accurate molecular diagnosis is crucial in patients with ASD/ID. WES was able to clarify the diagnosis of cerebral folate transport deficiency and neonatal diabetes, providing clinically actionable findings for both cases.

Grants:

Conflict of Interest: None declared

EP10.071 The second described case of a de novo ATXN2L missense variant in a child with global developmental delay

Susanne Ledig 1, Ann-Christin Tewes1, Frank Tüttelmann1, Oliver Schwartz2, Judit Horvath1

1Department of Medical Genetics, University Hospital Münster, Münster, Germany; 2Department of Paediatric Neurology, University Hospital Münster, Münster, Germany

Background/Objectives: The role of de novo mutations in developmental delay is well established, but the set of associated genes is still continuously increasing. Trio-exome sequencing in non-consanguineous families facilitate the detection of the individual causative genetic variant.

The patient, a premature born (34th week) and at the time of investigation 4 years old boy was referred because of global developmental delay with significantly impaired non-linguistic cognitive performance, and severe impairment of expressive and receptive language skills, retarded fine motor skills.

Methods: Chromosomal analysis of the patient revealed a normal male karyotype (46,XY). Trinucleotide repeat expansion in exon 1 of the FMR1 gene turned out normal. SNP-array analysis identified no pathogenic imbalances. Therefore, we performed in the boy and his unaffected parents exome-based trio analysis.

Results: Explorative exome analysis from the patient and his parents revealed a heterozygous de novo missense variant c.1342C>T p.(Arg448Cys) in ATXN2L (NM_148414.2). The CADD score of 29.4 and the different in silico prediction tools assume the variant as disease causing. It was classified as likely pathogenic (class 4 according to the ACMG guidelines: PS2, PM2, PP3).

Conclusion: To the best of our knowledge, this is only the second published case of a child with developmental delay and a de novo missense variant in ATXN2L. With this report, we want to raise awareness of ATXN2L as a relevant gene implicated in this highly heterogeneous condition. Detailed clinical descriptions of further patients is necessary for a precise genotype-phenotype correlation.

Grants: None.

Conflict of Interest: None declared

EP10.072 A maternally derived complex small supernumerary marker chromosome involving chromosomes 8 and 14: case report and review of the literature

fatima ouboukss 1, ZHOUR EL AMRANI1, hicham bouchahta1, ilham ratbi2, Thomas Liehr3, abdelaziz sefiani1, Abdelhafid Natiq2

1National Institute of Health, Department of Medical Genetics, rabat, Morocco; 2a.Research team in genomics and molecular epidemiology of genetic diseases, Genomics Center of Human Pathologies, Faculty of Medicine and Pharmacy, University Mohammed V in Rabat, Morocco, rabat, Morocco; 3c. Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, Am Klinikum 1, D-07747 Jena, Germany

Background/Objectives: The majority of small supernumerary marker chromosomes (sSMCs) originate from a single chromosome. However, complex sSMCs consist of genetic material from more than one chromosome, typically two. Complex sSMCs involving chromosomes 8 and 14 are exceedingly rare.

Methods: We report the case of a 14-month-old boy born to an unrelated couple. At birth, the child was hypotonic, with a cleft lip and palate, and ocular involvement. Throughout his development, he experienced feeding difficulties, stunted growth, and delayed psychomotor development. Classical and molecular cytogenetics were utilized to establish a genotype-phenotype correlation.

Results: A balanced maternal translocation was identified as: t(8;14)(p22.3;q21)mat. This led to a partial trisomy of chromosomes 8 and 14 in the affected boy due to a meiotic 3:1 segregation.

Conclusion: This report underscores the significance of cytogenetics in the diagnosis of rare genetic disorders and its impact on the genetic counseling of patients and their families. There are three other reported cases involving both chromosomes 8 and 14 with different breakpoints; the complex sSMC in this case is characterized as der(14)t(8;14)(p22.3;q21)mat.

Conflict of Interest: None declared

EP10.073 Novel PUS7 mutations in patients presenting with intellectual deficiency and growth retardation

Camille Bergès 1, Clément SAUVESTRE2, Sophie Naudion2, Catherine Vincent-Delorme3, Thomas Smol3, melanie rama3, Muhammad Umair4, Reza Maroofian5, Tobias Haack6, Mona Grimmel6, Anne Slavotinek7, Stephanie Efthymiou5, Faisal Zafar8, Erum Afzal8, Tracy Dudding-Byth9, Vincent MIchaud1

1Hospital Center University De Bordeaux, Génétique moléculaire, Bordeaux, France; 2Hospital Center University De Bordeaux, Génétique clinique, Bordeaux, France; 3Hospital Center University De Lille, Génétique clinique, Lille, France; 4King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia; 5University College London, United Kingdom; 6Universitätsklinikum Tübingen – Institut für Medizinische Genetik und angewandte Genomik, Tübingen, Germany; 7University of California San Francisco, San Francisco, United States; 8Multan Hospital, Hyderabad, Pakistan; 9Hunter Genetics, Waratah, Australia

Background/Objectives: Pseudourdylation is a frequent post-transcriptional modification resulting in uridine isomerization in 5-ribosyluracil also called pseudouridine. This mechanism leads to RNA stability with an increase in base-stacking and the creation of hydrogen bonds. Recently, papers reported different variants in PUS7 that were involved in marked growth retardation with microcephaly, associated with intellectual deficiency and behavioral issues such as self- and other-aggressivity.

In total, 13 cases were described in the literature presenting with homozygous or heterozygous composite variants.

Methods: Using WES or WGS, we identified new variants in PUS7 in patients harbouring intellectual deficiency, important failure to thrive and aggressive behaviour.

Results: In total, we report 17 new cases harboring new variants with a phenotype similar to the previously described one. This cohort was made possible through a Genematcher international collaboration.

Conclusion: These results bring new insights in PUS7 driven syndrome, with new clinical features reported. It aims to increase the knowledge regarding this kind of syndrome and may help geneticist with diagnosis in a next generation sequencing era.

Conflict of Interest: None declared

EP11 Neurogenetic and Psychiatric Disorders

EP11.003 Targeted screening of TARDBP in Bulgarian Amyotrophic Lateral Sclerosis patients

Slavko Ormandzhiev 1;2, Tihomir Todorov2, Teodor Angelov3, Teodora Chamova3, Vanyo Mitev1, Albena Todorova1;2, Ivaylo Tarnev3;4

1Medical University of Sofia, Department of Medical Chemistry and Biochemistry, Sofia, Bulgaria; 2Genetic Medico-Diagnostic Laboratory “Genica”, Sofia, Bulgaria; 3University Hospital Aleksandrovska, Clinic of Neurology, Sofia, Bulgaria; 4New Bulgarian University, Department of Cognitive Science and Psychology, Sofia

Background/Objectives: Amyotrophic lateral sclerosis (ALS) is a rare progressive neurodegenerative disease, characterized by upper and lower motor neuron impairment, leading to hypotrophy, muscle and respiratory failure.

The cause of ALS is not yet determined, but there are more than 40 associated genes to date, most of which account only for rare cases. Most common affected genes are C9orf72, SOD1, FUS and TARDBP. It has been established that aggregates of the transactive response DNA-binding protein (TDP-43), encoded by the TARDBP gene, in the neurons in the brain, lead to degeneration and death of affected neurons in ~4% of ALS cases.

Methods: TARDBP mutation screening was performed by Sanger sequencing on a cohort of 200 Bulgarian ALS patients.

Results: Mutation frequency for our cohort was 1.5% (3/200), with one missense variant found in two of our patients (c.881G>T, p.Gly294Val) and another missense variant, located in proximity to the first one (c.883G>A, p.Gly295Ser). Both variants are categorized as pathogenic. One of the patients with the p.G294V variant has a deceased father, who suffered from ALS. Two of the patient’s relatives were also screened and turned out to be carriers of the variant.

Conclusion: Taking into account the lack of genetic data for Bulgarian ALS patients, this study enriches the worldwide database by confirming the presence of pathogenic variants in TARDBP in Bulgaria. This indicates the significant role of screening for the TARDBP gene in order to genetically characterize the Bulgarian ALS patients.

Grants:

Conflict of Interest: None declared

EP11.004 A de novo deletion in the upstream region of ZIC2 in a patient with Holoprosencephaly

Erkan Koparir 1, Thomas König2, Erdmute Kunstmann1, Juliane Spiegler2, Thomas Haaf1, Eva Klopocki1

1Julius Maximilians University Würzburg, Institute of Human Genetics, Germany; 2University Hospital Würzburg, Department of Pediatrics, Würzburg, Germany

Background: Holoprosencephaly (HPE) is the most common congenital cerebral malformation in humans, characterized by impaired forebrain cleavage and midline facial anomalies. HPE is genetically and clinically heterogeneous. The ZIC2 (ZIC Family Member 2, MIM#603073) is one of the genes most commonly associated with HPE. ZIC2 encodes a transcription factor that plays several roles in neurological development.

Methods: In this study, we evaluated a patient with clinical features including HPE, coloboma and central diabetes insipidus. We performed molecular karyotyping and whole-exome sequencing to identify the underlying molecular pathology.

Results: A de novo heterozygous microdeletion of 1.09 Mb which encompasses five OMIM genes not associated with an HPE phenotype was detected in the long arm of chromosome 13 (13q32.3; hg19 chr13:99,489,272-100,577,947) by molecular karyotyping. The distal breakpoint is located in a non-coding region upstream of ZIC2. Additional clinically relevant variants were not identified by whole-exome sequencing.

Conclusion: An increasing number of studies emphasize the role of non-coding variants in such as enhancer, silencer or promoter sequences in the development of hereditary diseases. In this report, we present the identification of a de novo deletion which was not detected in population databases involving upstream of ZIC2 in a patient with HPE. We hypothesize that this deletion might have affected ZIC2 expression and contributed to our patient’s phenotype. We plan to perform whole-genome sequencing and functional studies to characterize the deletion more precisely.

Conflict of Interest: None declared

EP11.005 Advancing Clinical Diagnosis of Early childhood epilepsies: A Whole Exome Sequencing Approach

Christina Stoikova 1;1;1, Nevyana Ivanova1, Kalina Mihova1, Sashka Zhelyazkova2, MARIA PETROVA3, Genoveva Tacheva4, Ilyana Pacheva5, Iliyana Aleksandrova6, Daniela Deneva6, Ivan Litvinenko4, Ivan Ivanov5, Veneta Bojinova-Tchamova6, Ivaylo Tarnev2, Albena Jordanova7, Ivanka Dimova1, Radka Kaneva1

1Medical University - Sofia, Medical Chemistry and Biochemistry, Sofia, Bulgaria; 2Medical University - Sofia/ MBAL “Aleksandrovska”, Department of Neurology and Psychiatry/ Neurologic Clinic, Sofia, Bulgaria; 3Medical University - Sofia/ MHATNP“St.Naum”, Neurological Clinic for Children, Sofia, Bulgaria; 4Medical University - Sofia/ SBAL “Prof.Ivan Mitev”, Child Neurology Clinic, Sofia, Bulgaria; 5UMBAL “St.Georgi”, Pediatric Clinic, Plovdiv, Bulgaria; 6Medical University - Sofia/ MHATNP “St.Naum”, Neurological Clinic for Children, Sofia, Bulgaria; 7VIB-UAntwerp, Center for Molecular Neurology, Antwerpen, Belgium

Background: Epilepsy in children manifests across a spectrum, ranging from mild to most severe cases of treatment-resistant epileptic encephalopathies. Accurate and timely diagnosis is crucial for effective management and improved outcomes. This study explores the application of Whole Exome Sequencing (WES) in clinical diagnostics of epilepsy in children.

Methods: Six pediatric patients with epilepsy, spanning mild to severe cases, with negative results of microarray analysis and screening of the SCN1A gene, underwent WES to identify potential genetic factors contributing to their condition. The identified mutations were subsequently verified using Sanger sequencing.

Results: Our results revealed mutations not only in well-known genes associated with epilepsy but also in rare ones. The identified variants included both known pathogenic and likely-pathogenic mutations, alongside novel findings. The identification of mutations in less common genes highlights the complexity of the genetic basis of epilepsy.

Conclusion: Genotype-phenotype correlations provide valuable insights into the diverse genetic mechanisms underlying epilepsy in children. The integration of WES into clinical diagnostics allows comprehensive assessment, helping to identify both common and rare genetic variants. The therapeutic impact of identifying the specific variants in each patient extends beyond the scope of conventional treatments, potentially paving the way for targeted therapies. This personalized approach holds promise for more effective and tailored treatment strategies, thereby improving the therapeutic interventions in pediatric neurology.

Grants: NextGenerationEU/National Recovery and Resilience Plan of the Republic of Bulgaria, project № BG-RRP-2.004-0004-C01, D01-302/17.12.2021, D01-165/28.07.2022

Conflict of Interest: None declared

EP11.006 Molecular diagnostics of autistic spectrum disorders empowered by whole exome sequencing reveals rare monogenic forms

Nevyana Ivanova 1, Kalina Mihova1, Christina Stoikova2, Maria Sredkova-Ruskova3, Trayan Delchev3, Veneta Bojinova-Tchamova4, Daniela Avdjieva-Tzavella3, Vanyo Mitev2, Albena Jordanova1;5, Ivanka Dimova6, Radka Kaneva1

1Medical University - Sofia, Medical Chemistry and Biochemistry, Sofia, Bulgaria; 2Medical University - Sofia, Department of Medical Chemistry and Biochemistry, Molecular Medicine Center, Sofia, Bulgaria; 3SBAL Children’s Hospital „Prof. Dr. Ivan Mitev“, Clincal Genetics, Sofia, Bulgaria; 4University Hospital for Neurology and Psychiatry “Sveti Naum”, Neurological Clinic for Children, Sofia, Bulgaria; 5Vlaams Instituut voor Biotechnologie - University of Antwerp, Center for Molecular Neurology, Antwerpen, Belgium; 6Medical University - Sofia, Department of Medical Genetics, Molecular Medicine Center, Sofia, Bulgaria

Background/Objectives: Autism spectrum disorder (ASD) is among the most common neurodevelopmental disorders in children worldwide with a proved genetic background that affected social interactions, language communication and learning. Recently, the implementation of whole-exome sequencing (WES) led to the identification of de novo variants in many genes suggesting that autism could be present as a monogenic disorder caused by single nucleotide polymorphisms (SNPs). Here, we discuss our findings from WES in five children with ASD.

Methods: WES-analysis was performed to look for potentially pathogenic SNP-variants and Sanger sequencing was used to confirm them and to check segregation in the families.

Results: In three patients, disease-causing variants affected genes for known genetic syndromes (ADNP, HNRNPK and ZC4H2). De novo disruptive variant was identified in the DSCAM gene known to cause ASD by dosage mechanism. In one patient, two biallelic missense variants were found in trans-position in the SGSM3 gene, which was associated with autosomal recessive intellectual disability so far.

Conclusion: WES significantly increases the diagnostic power of the molecular genetic testing for ASD revealing novel genes and phenotype associations. Among them are genes that participate in the nervous system development playing a role of transcription factors, cell adhesion and signaling molecules. In many cases these are genes previously associated with intellectual disability. Autism could be the only featured clinical presentation of rare genetic syndromes that are difficult to suspect based on the clinical picture due to phenotype variability.

Grants: NextGenerationEU/National Recovery and Resilience Plan of the Republic of Bulgaria, project № BG-RRP-2.004-0004-C01; D01-302/17.12.2021; D01-165/28.07.2022.

Conflict of Interest: None declared

EP11.007 CAPRIN1-linked neurodevelopmental disorder: understanding the role of CAPRIN1 loss on neuronal differentiation, neurogenesis, and proliferation.

Lisa Pavinato 1;2;3, Antonietta Verrillo1;2, Chiara Leso4, Erika Ortolan3, Eleonora Campus5, Elena Rita Vecchi5, Andrea Angius6, Vincenzo Rallo6, Enza Ferrero3, Giovanni Battista Ferrero7, Luciano Conti5, Arianna Baggiolini1;2, Alfredo Brusco8;9

1Institute for Oncology Research, BIOS + , Bellinzona, Switzerland; 2Università della Svizzera Italiana, Lugano, Switzerland; 3Molecular Biotechnology Center “Guido Tarone” - University of Turin, Medical Sciences, Torino, Italy; 4Molecular Biotechnology Center “Guido Tarone”, Medical Sciences, Torino, Italy; 5CIBIO - University of Trento, Cellular, Computational and Integrative Biology, Trento, Italy; 6Institute of Genetic and Biomedical Research, National Research Council, Monserrato (CA), Italy; 7University of Turin, Department of Clinical and Biological Sciences, Orbassano (TO), Italy; 8Univerity of Turin, Neuroscience “Rita Levi Montalcini”, Turin, Italy; 9Città della Salute e della Scienza University Hospital, Medical Genetics Unit, Turin, Italy

Background/Objectives: We recently discovered that CAPRIN1 loss-of-function variants are associated with an autosomal dominant neurodevelopmental disorder. In neurons derived from hiPSCs haploinsufficient for CAPRIN1, we observed both structural and functional abnormalities. Transcriptome analysis revealed impairments in several cellular processes, including cell cycle control and proliferation, DNA replication, translation, oxidative phosphorylation, neurogenesis and neuronal outgrowth.

We aimed to further investigate the role of CAPRIN1 in determining the fate of neurons, and to uncover the underlying pathogenic mechanisms.

Methods: We generated forebrain cortical organoids (fBOs) using CAPRIN1 patient-derived hiPSCs. We analysed fBO cellular architecture and composition, and neuronal activity using immunofluorescence analysis, multicolor immunophenotyping panels and metabolic studies.

Results: During the development of CAPRIN1 fBOs, we observed a significant decrease in their overall size and abnormal morphology. Our preliminary findings show that the loss of CAPRIN1 leads to impaired cell proliferation and cell cycle regulation, as well as an imbalance in the composition of fBOs. Specifically, we observed an overrepresentation of immature neurons and glial cells. Since these cells primarily rely on glycolysis, β-oxidation, and redox signalling for their metabolism, we are performing metabolic analyses. Initial results suggest an increase in glycolysis and an increase in reactive oxygen species.

Conclusion: The development and investigation of CAPRIN1 fBOs are beginning to provide insights into the involvement of CAPRIN1 in neuronal differentiation, neurogenesis and proliferation, with a focus on its role in regulating protein translation. Additionally, this study will emphasize the similarities between CAPRIN1 and its main interactor, FMRP, associated with the Fragile-X-syndrome.

Grants:

Conflict of Interest: None declared

EP11.008 Methylation landscape in fragile X-associated tremor/ataxia syndrome

Laia Rodriguez-Revenga 1, Judit Garcia-Guallarte2, Germán Casabó-Vallés2, Trinitat Alberola-Pons2, Irene Madrigal1, Maria Isabel Alvarez1

1Hospital Clinic Barcelona, Biochemistry and Molecular Genetics Department, Barcelona, Spain; 2EpiDisease S.L. (Spin‑Off CIBER‑ ISCIII), Valencia, Spain

Background/Objectives: Fragile X-associated tremor/ataxia syndrome (FXTAS) is a late-onset neurodegenerative disorder with reduced penetrance that appears in adult FMR1 premutation carriers (55-200 CGGs). Epigenetics, especially the contribution of DNA methylation has been described as a mechanism mediating different clinical phenotypes and as a potential biomarker for disease prediction. This study aimed to investigate the role of DNA methylation in FXTAS using whole blood of FMR1 premutation carriers.

Methods: In total, 54 FMR1 premutation carriers were enrolled, meeting the following criteria: 20 FXTAS patients (FMR1 premutation carriers with tremor or ataxia symptoms before age 65) and 34 non-FXTAS patients (FMR1 premutation carriers with no clinical evidence of any main neurodegenerative diseases over the age of 68 years). DNA methylation profiles were generated using the Illumina MethylationEPIC v2.0 BeadChip.

Results: We identified 21 significant differentially methylated regions (DMRs) in whole blood from FXTAS patients, compared with non-FXTAS patients. Of these DMRs, 7 were hypomethylated and 15 were hypermethylated. Moreover, 17 DMRs overlapped with promoters and 4 were identified to be related with genes: PCDHGA2, PCDHGA1, RUNX1 and PDXK. For the DMRs, functional enrichment analysis revealed 62 GO biological processes and 7 KEGG pathways significantly deregulated in FXTAS patients (FDR < 0.05).

Conclusion: We found aberrant methylation in DMR-related genes implying important roles in FXTAS etiology.

Grants: Work supported by the Instituto de Salud Carlos III (ISCIII) (through the project PI21/01085), co-funded by the European Union, and AGAUR (2021-SGR-01492). The CIBER de Enfermedades Raras is an initiative of the Instituto de Salud Carlos III.

Conflict of Interest: None declared

EP11.009 Modern genetic classifications of malformations of cortical brain development, identification of causative genes, and radiological findings in the pediatric Egyptian population

Hasnaa Elbendary 1, maha zaki1, Karima Rafat1, Mohamad Abdelhamid2

1National Research centre, clinical genetics, Cairo, Egypt; 2National Research centre, Molecular genetics, Cairo, Egypt

Background/Objectives: Malformations of cortical development (MCDs) are neurodevelopmental disorders that result from abnormal development of the cerebral cortex in utero. This abstract aims to provide an overview of the advancements in genetic classification systems through diagnosis of 94 Egyptian families with MCDs.

Methods: 30% of patients underwent targeted gene sequencing; next-generation sequencing was applied to 70% of patients. Patients were recruited from neurogenetic clinic of the National Research Centre of Cairo.

Results: Here in, we reported the genetic findings of a cohort of MCDs patients (n = 100) from 94 Egyptian families, list of sorted-out genes (RELN, VLDRL, ASPM, WDR82, WDR62, CIT, CENP, GPR56, and Rttn 10). The most common disorder was microcephaly with simplified gyral pattern, which was detected in 40% (n = 40), with many novel mutations detected; the majority of them were missense mutations followed by frame shift mutations. The second common disorder with higher prevalence was the lissencephaly spectrum in 30% of patients (n = 30), followed by the least prevalent hemimegalencephaly (n = 6), periventricular nodular heterotopia (PVNH) (n = 5), and tublinopathies and other MCD (n = 19).

Conclusion: To the best of our knowledge, this is the first and largest cohort of patients with MCDs from Egypt and Africa. We present prevalence, clinical findings, and neuroimaging findings confirmed by genetic diagnosis. We report up to 15 variants that have not been reported before.

We present a diagnostic flow chart that can be applied to patients with MCDs and a list of MCDs genes with their phenotypes.

Grants: SDTF 33492

Conflict of Interest: Hasnaa Elbendary full time, maha zaki principal investigator, full time, Karima Rafat full time, Mohamad Abdelhamid full time

EP11.010 Broadening the phenotypic spectrum of biallelic SHQ1-variants

Sabrina Stefanelli 1, Stephanie Jünemann2, Pascal Joset1, Isabel Filges1

1Universitätsspital Basel, Medical Genetics, 4031; 2Universitäts-Kinderspital beider Basel, Basel, Switzerland

Background/Objectives: To date, only few patients with an ultrarare neurodevelopmental disorder due to biallelic variants in the SHQ1 gene are described. Children present with global developmental delay and neurological symptoms such as hypotonia and childhood-onset dystonia. Affected patients all show low levels of homovanillic acid (HVA) in cerebrospinal fluid (CSF) suggesting a dysfunction in dopamine metabolism and proposed as a diagnostic criterion. Parkinsonian drugs show some clinical response. We describe a one-year-old girl with biallelic SHQ1 variants, developmental delay, neurological symptoms but normal HVA values in CSF analysis.

Methods: Our patient underwent full clinical evaluation by a multidisciplinary team. We performed Trio-Exome sequencing (ES) and genotype-phenotype comparisons based on existing literature and database search.

Results: Clinical examination showed marked truncal hypotonia and increased muscle tone with pathological reflexes in the lower limbs. There were no dystonic movements. EEG and brain MRI were unremarkable. HVA in CSF was normal. Paternal family history is significant for adult early onset Parkinson disease. Trio-ES evidenced the novel paternally inherited SHQ1-variant c.460C>T and the known maternally inherited SHQ1-variant c.523G>T. Both variants were classified as likely pathogenic (ACMG criteria).

Conclusion: This is the first case of a child with biallelic SHQ1 variants and compatible clinical signs but normal CSF HVA levels. Several exams are ongoing. We describe commonalities and differences of patients with this ultrarare condition. Moreover, the family history of early-onset Parkinson’s disease opens up further discussion on the role of SHQ1 in dopamine metabolism.

Grants: none

Conflict of Interest: None declared

EP11.011 Homozygous loss-of-function variant of LBX1 - confirmation of the existence of congenital central hypoventilation syndrome type 3

Łukasz Kępczyński 1, Anna Łupińska2;3, Paula Smalczewska2, Katarzyna Połatyńska4, Jordan Sałamunia5, Tadeusz Kałużewski1, Renata Stawerska2;3, Arkadiusz Zygmunt2;3, Andrzej Lewiński2;3, Agnieszka Gach1

1Polish Mother’s Memorial Hospital Research Institute, Department of Genetics, Łódź, Poland; 2Polish Mother’s Memorial Hospital Research Institute, Department of Endocrinology and Metabolic Diseases, Łódź, Poland; 3Medical University of Łódź, Department of Pediatric Endocrinology, Łódź, Poland; 4Polish Mother’s Memorial Hospital Research Institute, Department of Child Neurology and Epileptology, Łódź, Poland; 5Laboratory of Medical Genetics of the “Genos” Partnership R&D Division, Łódź, Poland

Background/Objectives: LBX1 encodes a homeodomain containing transcription factors, involved in neural cell fate control. LBX1 gene was putatively associated with congenital central hypoventilation syndrome type 3 (CCHS3). Only one familial case of such a disease was described in literature – in two consanguineous brothers, presenting congenital hypoventilation syndrome. Here, we describe second, unrelated case of homozygous LBX1 loss-of-function variant, resulting in similar phenotype.

Methods: A whole exome sequencing was performed for a 6 year-old female proband of Romani ancestry, presenting obesity, global developmental delay, stereotypic behavior, and congenital central hypoventilation, requiring continuous positive airway pressure therapy.

Results: A homozygous, likely pathogenic loss-of-function variant LBX1: c.707del was identified. This variant results in frameshift (Val236Alafs). The mRNA product does not undergo nonsense mediated decay, but probably undergo non-stop mediated decay. Variant was located in 8.86 Mb region of homozygosity, overlapping the whole LBX1 coding region. 3 additional synthenic homozygous variants (c.709C>T, c.711C>T and c.717A>T) were identified in LBX1 gene below the frameshift variant.

Conclusion: Identified variant is located in the same nucleotide as in the first description of patients with congenital central hypoventilation syndrome type 3. Our observation confirms existence of this disorders and widens the phenotypic characteristics of CCHS3.

Grants:

Conflict of Interest: None declared

EP11.012 MECP2 duplication syndrome: phenotypic characterization, AI-based diagnosis, and severity scale development

Antonio Federico Martínez-Monseny 1, Lourdes Vega Hanna1, didac casas-alba1, francesc palau martinez1, merce bolasell1, patricia rubio1, mar o’callaghan1, Ainhoa Pascual-Alonso1, sol balsells1, Judith Armstrong1

1

Consortium: MDS Study Group

Background/Objectives: MECP2 duplication syndrome (MDS) (MIM#300260) is a rare X-linked neurodevelopmental disorder. This study aimed to (1) expand the clinical and molecular description of MDS patients, (2) automate diagnosis using the Face2gene platform, (3) develop a specific clinical severity scale, and (4) analyse correlations between the scale and variables.

Methods: descriptive and retrospective study, including genetically confirmed MDS patients evaluated at a tertiary pediatric hospital between January 2012 and December 2022. Epidemiological, clinical, and molecular data including CGH-array, FISH, maternal origin and X-inactivation were included. Patient photographs were collected for training the Face2Gene algorithm. The MECPDup scale was developed incorporating elements from the Motor Behavior Assessment scale (Rett syndrome) and the main clinical features of MDS.

Results: Thirty-five patients (0-24 years, 30 males), were included. Six male patients died from respiratory complications. Clinical characteristics in males: ID (100%), hypotonia (93%), ASD (77%), seizures (53%, 33% treatment-refractory), and developmental regression (52%), recurrent respiratory infections (79%), swallowing difficulties (73%), constipation (73%), and gastroesophageal reflux (57%). The Face2Gene algorithm was successfully trained to recognize MDS patients. The MECPDup scale was validated and correlated with the MBA scale. The MECPDup score was significantly higher in males (P < 0.001) and showed significant positive correlations with age (R = 0.720, P < 0.001) and duplication size (Rho = 0.370, P = 0.044). Males with early death had significantly higher MECPDup scores (P = 0.009).

Conclusion: The integration of AI-based facial recognition technology and the development of the MECPDup severity scale hold promise for enhancing diagnostic accuracy, monitoring disease progression, and evaluating treatment responses in individuals affected by MDS.

Conflict of Interest: None declared

EP11.013 Ataxia-oculomotor apraxia type 4 and hereditary iron overload - case report

Nuno Ferreira 1

1Hospital do Divino Espírito Santo, Clinical Pathology Department, Portugal

Background/Objectives: Ataxia-oculomotor apraxia type 4 (AOA4) is a rare autosomal recessive cerebellar ataxia, due to mutations in PNKP gene. Hereditary iron overload (HIO) is a genetically inherited disorder characterized by excess iron stores, related to mutations in HFE gene. We aim to describe a case of simultaneous AOA4 and HIO.

Methods: Case report of a 29-year-old man simultaneously with AOA4 and HIO.

Results: He started progressive neurological manifestations when 9 year-old. Presently, he has cognitive impairment, dysarthria, oculomotor apraxia, sensorimotor polyneuropathy, tremor and cerebellar ataxia; he’s wheelchair-bound and dependent. Genetic testing found a mutation of PNKP gene, which, allied to clinical features and obesity, established diagnosis of AOA4. When 15 year-old, he started increased levels of serum ferritin (>300ng/ml), high transferrin saturation (>45%) and iron deficient anemia, without symptoms. Genetic testing revealed heterozygosity for p.Cys282Tyr (C282Y) mutation of HFE gene and for p.His63Asp (H63D) mutation, thus HIO. He started regular therapeutic phlebotomies till serum ferritin was within acceptable levels. Currently, he has normal serum ferritin (99,2 ng/mL) and transferrin saturation (22%).

Conclusion: AOA4 is the most common ataxia-oculomotor apraxia in Portugal. It is progressive, debilitating and has no specific treatment. HIO is characterized by increased serum ferritin (>300ng/ml in men) with or without high transferrin saturation, with or without functional iron deficient anemia. Most patients are asymptomatic. Heterozygotes for p.Cys282Tyr (C282Y) or p.His63Asp (H63D) mutations of HFE gene usually don’t express hemochromatosis phenotype; instead, they have HIO. HIO has a positive prognosis if diagnosed early and effectively treated.

Grants: None.

Conflict of Interest: None declared

EP11.014 New player in lipopolysaccharide-induced neuroinflammation: programmed cell death ligand 1 (PDL-1)

Ayfer Pazarbasi 1, Gulsevinc Aksoy1, İnayet Nur Uslu1

1Cukurova University Faculty of Medicine, Medical Biology, Adana, Türkyie

Background: Microglial cells which are brain-resident immune cells are responsible for innate immunity and have vital importance initiation of neuroimmune responses to various conditions such as brain injury, infection and neurodegeneration. The programmed death (PD)-1/PD-L1 pathway results in functional inhibition of T cells and it is a known negative immune checkpoint. PD-1/PD-L1 pathway reduces excessive immune reaction and prevents cellular toxicity. In this study, we aimed to investigate changes in expression of PD-1 and PD-L1 genes after activation of N9 microglial cells with Lipopolysaccharide.

Methods: N9 microglial cells were treated with 100 ng/mL LPS for 24 hours. After RNA isolation and cDNA synthesis, Real-time PCR reactions were performed for PD-1 and PD-L1 genes. Relative mRNA levels of the genes was calculated by 2-ΔΔCt method. Independent samples t-test was used to compaire differences in expression levels of the genes between control and LPS-treated cells.

Results: In this study, we determined that the expression level of PD-L1 gene was significantly increased in N9 microglial cells that were treated with 100 ng/mL LPS. However, we did not find any statistically differences in expression level of PD-1 gene between control and LPS-treated cells.

Conclusıon: In this study, we showed that PD-L1 expression was increased in LPS-activated microglia cells. Therefore our results suggest that PD-1/PD-L1 pathway, a negative immune regulatory system of T cells, can play an important role in LPS-induced neuroinflammation.

Grant: This work was supported by “Cukurova University Research Projects Funding Unit” with project number:TSA-2022-14801.

Conflict of Interest: Ayfer Pazarbasi This work was supported by “Cukurova University Research

Projects Funding Unit” with project number:TSA-2022-14801, Gulsevinc Aksoy This work was supported by “Cukurova University Research

Projects Funding Unit” with project number:TSA-2022-14801, İnayet Nur Uslu This work was supported by “Cukurova University Research

Projects Funding Unit” with project number:TSA-2022-14801

EP11.015 Level of molecules involved in Aβ-induced inflammatory response in human neuroblastoma cells

Lütfiye Özpak 1

1Kahramanmaraş Sütçü İmam University, Medical Biology, Kahramanmaraş, Türkyie

Background/Objectives: One of the pathophysiological findings of Alzheimer’s disease is neuroinflammation. In Alzheimer’s, a neurodegenerative disease, chronic inflammation and the continuous, excessive release of cytotoxic factors adversely affect brain functions. The synthetic peptide Aβ25-35 retains many of the physical and biological properties of Aβ. In vitro studies involving Aβ stimulation, it has been demonstrated that inflammation and apoptosis are induced. In this study, we aimed to determine the levels of neuroinflammatory markers in human neuroblastoma cells (SH-SY5Y) where we induced neurotoxicity by applying Aβ25-35.

Methods: Human neuroblastoma cells were exposed to Aβ25-35 a dose of 20 micromolars for 24 hours, as determined by MTT results. The mRNA of inflammatory cytokines (IL-6, TNF-α, IL-1β, IL-10) were measured using real-time PCR.

Results: Our results show that the expression levels of the cytokines involved in the inflammatory response were increased in the Aβ induced group compared with the control. The results are statistically significant (p < 0.05).

Conclusion: While it is still unclear whether inflammation is a cause or a consequence of Alzheimer’s disease targeting the reduction of these inflammatory markers can be a potent way to control and prevent neurodegenerative diseases.

Conflict of Interest: None declared

EP11.017 A new case of Li-Campeau syndrome: Homozygous exonic deletion of the UBR7 gene

Tarik Duzenli 1, gulsum kayhan1

1Gazi University, Department of Medical Genetics, Ankara, Türkyie

Background/Objectives: Biallelic pathogenic variants in the ubiquitin protein ligase E3 component n-recognin 7 gene (UBR7) cause Li-Campeau syndrome (OMIM #619189, LICAS), which is characterized by developmental delay/intellectual disability, epilepsy, hypothyroidism, and ptosis. To date, only nine patients have been reported.

Methods: A 16-year-old girl from a consanguineous marriage was referred to our clinic with learning difficulties, attention-deficit / hyperactivity disorder, and congenital hypothyroidism. She also had dysmorphic facial features, ptosis, and hypoplasia of the toenails. Chromosomal microarray (CMA) and whole exome sequencing (WES) were performed on leukocyte-derived DNA samples. Conventional PCR followed by gel-electrophoresis was hired to confirm the variant.

Results: CMA analysis were normal, and the WES analysis revealed a homozygous deletion of exon 7 of the UBR7 gene (NM_175748) (hg19:chr14:93684622-93685319). The deletion was confirmed with conventional PCR.

Conclusion: The variant in our patient is significant because it is the first reported single exon deletion in this syndrome. Additionally, our patient exhibited unique findings that have not been previously reported, such as ADHD and hypoplasia of the toenails. The absence of developmental delay, cardiac anomaly, and epilepsy in our patient, commonly observed in the majority of previously reported patients, may reflect the variable phenotype of the syndrome. This report expands on both the phenotype and genotype of LICAS, an ultra-rare disease.

Grants: none

Conflict of Interest: None declared

EP11.018 Two recently identified unrelated cases of NEDAUS

Friedrich Stock 1, Jasmin Beygo1, Ilaria Parenti1, Harald Surowy1, Christopher Schröder1, Elsa Leitão1, Antje Kampmeier1, Christel Depienne1, Frank J. Kaiser1;2, Alma Küchler1

1Universitätsklinikum Essen, Institut für Humangenetik, Essen, Germany; 2Essener Zentrum für Seltene Erkrankungen (EZSE), Essen, Germany

Background/Objectives: Variants in CUL3 (OMIM *603136) were associated with neurodevelopmental disorders for the first time in 2020. In 2023, a clinical entity was described based on a study containing 35 individuals carrying heterozygous (mainly predicted loss-of-function) variants in CUL3. According to its main features, this entity was named NEDAUS (NEurodevelopmental Disorder with or without AUtism or Seizures, OMIM #619239). NEDAUS is characterized by early onset global developmental delay, growth restriction, and various neurologic anomalies (autism spectrum disorder, seizures, encephalopathy, brain malformation, behavioural anomalies).

Methods: By genome and exome sequencing respectively,

Results: we identified two novel variants in CUL3 in two unrelated children, a five-year-old boy of German origin and a six-year-old girl of Moroccan Berber origin. Both variants, one splice-site mutation and one nonsense mutation, occurred de novo and were classified as disease causing (class 5) due to their predicted truncating effect. The clinical phenotype of the affected children with short stature, global developmental delay, and microcephaly is compatible with the previously noted cases of NEDAUS. Additionally, the boy features several malformations (patent ductus arteriosus, patent foramen ovale, unilateral cryptorchidism, ectopic kidney) and unilateral hearing loss. In the girl, no morphologic anomalies were noted; she experienced a single fever-related seizure.

Conclusion: To our knowledge, 40 persons with CUL3-associated neurodevelopmental delay have been described in recent years. Our two novel cases contribute to the clinical delineation of the rather new clinical entity NEDAUS and broaden the mutational spectrum.

Grants:

Conflict of Interest: None declared

EP11.019 Identification of copy number variants in a family with high prevalence of psychosis

Fernanda Francis-Cartin 1;2, Esteban J. Rodriguez1;3;4, Gabriela Chavarria-Soley1;5, Alejandro Sanchez-Flores6;7, Henriette Raventós1;5

1Centro de Investigacion en Biologia Celular y Molecular, Universidad de Costa Rica, San Jose, Costa Rica; 2Programa de Posgrado en Biología, Universidad de Costa Rica, San Jose, Costa Rica; 3Instituto de Investigaciones en Salud, Universidad de Costa Rica, San Jose, Costa Rica; 4Programa de Posgrado en Microbiologia, Universidad de Costa Rica, San Jose, Costa Rica; 5Escuela de Biologia, Universidad de Costa Rica, San Jose, Costa Rica; 6Unidad Universitaria de Secuenciacion Masiva y Bioinformatica, Universidad Nacional Autonoma de Mexico, Morelos, Mexico; 7Instituto de Biotecnologıa, Universidad Nacional Autonoma de Mexico, Morelos, Mexico

Background/Objectives: We used two phenotypes for the association of CNVs, psychosis in the course of the disease (as a syndromic phenotype) and mood disorders (as a categorical phenotype). The objectives were to analyze whether CNVs associated with the presence of the phenotypes in a family, as well as to determine which pathways or genes are compromised by the gain or loss of these variants.

Methods: We developed a workflow to obtain the calling and filtering of CNVs for association testing. For CNV calling, CNVpytor software was used; the filter was performed using DGV, overlap was evaluated with intersect tool. Multiinter tool was used to determine the breakpoint. Filters by phenotype were applied, in which CNVs that were absent in affected were eliminated, as well as variants with a frequency greater than 95% and less than 5%.

Results: There is at least one variant, cnvdup137, associated with mood disorders. However, there are other good candidates to explain part of the genetic architecture of the family (cnvdel221 and cnvdup175). The associated variant contains genes that code for immunoglobulins and overlaps with the immunoglobulin kappa locus.

Conclusion: These results support the line of research that demonstrates the involvement of the immune system in the central nervous system. This study provides the first evidence of CNVs associated with psychiatric conditions in the Costa Rican population, and also lays the foundations to begin in-depth research on alterations in the immune function of this family that are increasing the risk of psychiatric conditions.

Grants: Fondo de Becas CENAT-CONARE

Conflict of Interest: None declared

EP11.020 Association study between the DISC1 gene and psychosis in a multigenerational Costa Rican family with a high prevalence of psychiatric disorders

Esteban J. Rodriguez 1;2;3, Fernanda Francis-Cartin1;4, Gabriela Chavarria-Soley1;5, Henriette Raventós1;5

1Universidad de Costa Rica, Centro de Investigación en Biología Celular y Molecular, San José, Costa Rica; 2Universidad de Costa Rica, Instituto de Investigaciones en Salud, San José, Costa Rica; 3Universidad de Costa Rica, Programa de Posgrado en Microbiología, San José, Costa Rica; 4Universidad de Costa Rica, Programa de Posgrado en Biología, San José, Costa Rica; 5Universidad de Costa Rica, Escuela de Biología, San José, Costa Rica

Background/Objectives: DISC1 is a gene originally identified as affected by a chromosomal translocation in a Scottish family with prevalence of psychiatric disorders. Multiple DISC1 variants have been reported associated with psyactric diosrders in different populations, although few have been repeated between studies. Nevertheless, molecular analyses agree on the participation of the protein in neurologically relevant pathways. Alterations on cell lines and mouse models has also shown involvement of the protein with presence of neurodevelopmental deficits, neurodegenerative processes, and psychiatric disorder-like phenotypes. Therefore, we sought to determine if there is an association between the DISC1 gene and psychosis in a Costa Rican family with high prevalence of psychiatric disorders.

Methods: Association test was performed between psychosis and DISC1 variants obtained from a variant calling pipeline for the complete sequence of the gene from 110 subjects belonging to a multigenerational Costa Rican family with a high prevalence of psychiatric disorders.

Results: We found 10 variants with nominal significance values (without correction) associated with psychosis, nine of them within the same region of the gene (intron 9-10) being rs5781671 the most associated with a nominal value p = 0.0088.

Conclusion: Association between DISC1 and psychosis was detected for nine variants in the same intronic region of DISC1. Different studies have reported different variants within the same region as being associated with multiple psychiatric disorders but the possible impacts of the variants on alternative splicing processes, modification of miRNA regulation or regulations in transcriptional processes as well as post-transcriptional assembly and/or regulation needs to be further explored.

Grants: Fondo de Becas CeNAT-CONARE

Conflict of Interest: None declared

EP11.021 Genetic variability in the expression of primary emotions in five relevant genes: Differences across sexes

Timotej Glavač 1, Maruša Barbo2, Sara Redenšek Trampuž2, Metka Ravnik-Glavač2, Maja Zupančič1, Vita Dolzan2

1University of Ljubljana, Faculty of Arts, Department of Psychology, Ljubljana, Slovenia; 2Faculty of Medicine, University of Ljubljana, Institute of Biochemistry and Molecular Genetics, Ljubljana, Slovenia

Background/Objective: The investigation of genetic variability associated with the expression of primary emotions (SEEKING, FEAR, ANGER, SADNESS, CARE and PLAY) in humans is still in its early phases.

Methods: We genotyped 334 young adults (56.8% female) who completed an online form of a short measure assessing primary emotions and provided a buccal swab for 14 SNPs across the following genes: BDNF, TPH2, COMT, OPRM1, and OXTR.

Results: We found sex specific differences in the influence of the included polymorphisms. In females, BDNF rs6265 GG homozygotes had higher FEAR (p = 0.05) and ANGER (p = 0.035) in the dominant model, FEAR (p = 0.017) and SANDESS (p = 0.006) in recessive models of BDNF rs28722151 was higher in GG homozygotes and BDNF rs11030101 TT homozygotes had higher levels of FEAR (p = 0.008) and ANGER (p = 0.001) in recessive models. In males, AA homozygotes for COMT rs4680 had lower levels of ANGER (p = 0.031) and SADNESS (p = 0.002) in the recessive model. TPH2 rs1843809 TT homozygotes reported lower levels of PLAY in the recessive model (p = 0.019). In the additive model TPH2 rs4290270 AA homozygotes had lower ANGER (p = 0.043). In GG homozygotes for TPH2 rs4570625 ANGER was higher (p = 0.030), while CARE was lower (p = 0.038) in the dominant model.

Conclusion: This study shows the importance of sex differences in the influence of selected gene polymorphisms on the primary emotions.

Grants: Slovenian research agency No. P1-0170 and P5-0062

Conflict of Interest: None declared

EP11.022 A bi-allelic variant c.6577C>T in NAV3 is associated with global developmental delay, dysmorphism and cerebellar abnormalities

Selinda Mascarenhas 1, Vaishnavi Badiger1, Sheeba Farooqui1, Anju Shukla1, Radhakrishnan Periyasamy1

1Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India

Background/Objectives: Neuron navigator 3 (NAV3) gene encodes for Nav3, a cytoskeletal-associated protein belonging to the neuron navigator family (NAV1-3), that is involved in developmental processes like neuritogenesis and cellular migration.

Methods: Singleton exome sequencing (ES) was performed for the proband, followed by Sanger segregation and validation.

Results: A five-year-old female presented with global developmental delay (GDD), intellectual disability, facial dysmorphism, and behavioral abnormalities. Computed tomography of the brain showed cerebellar vermian hypoplasia. Singleton ES revealed a novel homozygous variant, c.6577C>T p.(Arg2193Ter) in NAV3 (NM_001024383.2). Sanger validation and segregation confirmed carrier status in the parents. This variant is absent in gnomAD or our in-house database and is predicted to introduce a premature termination codon, potentially initiating nonsense-mediated mRNA decay or producing a truncated protein product. NAV3 is predominantly expressed in the brain, mainly in the cerebellar hemispheres. Nav3 and Nav2 share identical protein domains and together with Nav1, constitute a complex that interacts with Trio (Rho guanine exchange factor) to activate Rac1 (Rho GTPase), which initiates downstream signalling pathways for various neurodevelopmental processes. Recently, bi-allelic variants in NAV2 were reported in a seven-year-old female with GDD, cardiac malformations, and brain abnormalities predominantly in the cerebellum, and was associated with neurodevelopmental disorder (NDD).

Conclusion: We herein report a family with a bi-allelic variant in NAV3 and propose it as a novel candidate gene for NDDs.

Grants: IA/CRC/20/1/600002 (DBT/Wellcome Trust India Alliance)

Conflict of Interest: None declared

EP11.023 Novel variants in FZR1 consolidate its role in developmental and epileptic encephalopathies type 109

Yasaman Pakdaman 1, Somayeh Bakhtiari2;3, Roseline Caumes4, paskal cullufi5, Yiran Xu6, Changlian Zhu6;7, Yangong Wang8, Thomas Smol9, Michael C. Kruer2;3, Aboulfazl Rad1, Shivendra Kishore1, Gabriela Oprea1

1Arcensus GmBH, Rostock, Germany; 2Barrow Neurological Institute, Phoenix Children’s Hospital, Phoenix, United States; 3Departments of Child Health, Cellular and Molecular Medicine, Genetics, and Neurology, University of Arizona College of Medicine-Phoenix, Phoenix, United States; 4Clinique de Génétique, CHU Lille, Hospital Center University De Lille, Lille, France; 5Mother Theresa Hospital Center Tirana, Tirana, Albania; 6Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China; 7Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Goteborg, Sweden; 8Children’s hospital of Fudan University, Institutes of Biomedical Sciences of Fudan University, Shanghai, China; 9Institut de Génétique Médicale, Hospital Center University De Lille, Lille, France

Background/Objectives: To date, pathogenic variants in over 100 genes are reported in association with Developmental and Epileptic Encephalopathies (DEE). De novo loss-of-function FZR1 (fizzy and cell division cycle 20-related protein 1 gene) variants were recently associated with DEE type109, with only four reported patients. In this study, we report novel FZR1 variants in three unrelated patients with similar phenotypes.

Methods: Three patients were analysed by whole exome sequencing: patient 1, who manifested developmental delay and tonic-clonic seizures at 2.5 months of age; patient 2, a 6 years-old boy with developmental delay, ataxia and seizures starting at 5 months of age; and patient 3, a 17 months-old boy with developmental delay, hypotonia and feeding difficulties. Confirmation of variants origin has been performed by Sanger analysis and SNP array.

Results: Two de novo heterozygous variants were identified in the FZR1 gene (NM_016263.4): a missense c.868G>A, p.Asp290Asn variant in patient 1, and a multi-exon duplication c.824-2_*29dup in patient 2 encompassing exons 10-14. Patient 1 developed more severe phenotypes including dysmorphic features, hypotonia and recurrent infections, and was deceased at 6 months old. Patient 3 was found to carry a heterozygous missense variant, c.1126G>A; p.Gly376Ser, which is assumed to arise de novo. This patient had not developed any types of seizure by the last examination.

Conclusion: We report three additional cases of DEE type 109. Our observations, confirm previous studies that de novo heterozygous variants in FZR1 cause global developmental delay and various types of seizures in the first months or years of life.

Conflict of Interest: None declared

EP11.024 Precision medicine: Lessons from a designated clinic for patients with Phelan McDermid Syndrome

Odelia Chorin 1;2, Ben Pode Shakhed1, annick REIN ROTHSCHILD1, Lior Greenbaum2, Shelly Lev-Hochberg1

1Institute of Rare Diseases, Lily and Edmond Safra Hospital for Children, Ramat Gan, Israel; 2Danek Gertner Institue of Human Genetics, Sheba Medical Center, Ramat Gan, Israel

Background/Objectives: Designated clinics focused on a specific rare genetic disorder may improve patient outcomes through specialized, multidisciplinary teams gaining experience with various aspects of the disease, expertise in treatment and creating a social support network for families.

Phelan-McDermid Syndrome (PMS) is caused by haplo-insufficiency of SHANK3 and is characterized by global developmental delay (GDD), autism, and additional multisystem manifestations.

Methods: We aimed to establish a specialized clinic with nationwide referrals, for individuals with PMS. Demographic, clinical, and molecular data was collected for each patient.

Results: Eighteen patients (mean age 12.5 years; range, 4-36y) of which 11 females (61%) were examined. Half of cases were caused by chromosomal deletions: mosaicism for the chromosomal aberrations (n = 2); parental balanced translocation (n = 1); or ring chromosome (n = 2).

Patients presented with autism (11/18), severe intellectual disability (11/18), with multiple episodes of regression of acquired skills from infancy (11/18). Additional manifestations included sleeping difficulties (13/18), behavioral problems (10/18), seizures (8/18), feeding difficulties (5/18), cardiac involvement (4/18), orthopedic (5/18) and endocrine abnormalities (5/18).

No clear correlation was observed between mutation type and severity of disease, with significant phenotypic variability. Patients with mosaic disease presented with similar severity to heterozygous germline carriers.

Treatment modalities included Cannabis both as an anti-seizure and as a relaxing medication. Antipsychotic medications can cause severe regression and should be avoided.

Conclusions: Our experience with a designated Phelan-McDermid clinic adds to current knowledge regarding genotypic and phenotypic spectrum of this disorder in Israel. Such clinics may enable to establish patient registries, creating a hub for research and possibilities for treatment.

Conflict of Interest: None declared

EP11.025 Clinical characterisation of patients with pathogenic variants in SHANK family members

Carmen Manso1, Nino Spataro1, Lidia Torrent2, Laura Plans3, Mercè Casadesus3, Victor Martinez-Glez1, montserrat pamias2;4, Anna Ruiz 1

1Parc Taulí Hospital Universitari, Institut d’Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Center for Genomic Medicine, Sabadell, Spain; 2Parc Taulí Hospital Universitari, Institut d’Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Salut Mental Infantojuvenil, Sabadell, Spain; 3Althaia. Xarxa Assistencial Universitària de Manresa, SEMSDI-Catalunya Central, Manresa, Spain; 4Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM)

Background/Objectives: Pathogenic variants in the SHANK family genes have been linked to autism spectrum disorder, as well as to other neuropsychiatric and neurodevelopmental disorders.

We aim to deeply characterise the neurodevelopmental, neuropsychiatric and dysmorphic features of patients with SHANK pathogenic variants in order to identify common features.

Methods: Whole exome sequencing was performed in 115 patients (aged 6-18 years) with mild intellectual disability/borderline intellectual functioning and another psychiatric comorbidity

Clinical, dysmorphlogical, neurodevelopmental and psychopathological features were retrospectively reviewed.

Results: Intragenic pathogenic variants in SHANK1 (2/5), SHANK2 (2/5), and SHANK3 (1/5) were identified in 5 out of 115 patients (5/115, 4.5%). All five patients presented significant language and motor delay. The intellectual quoficient ranged from 63 to 75. All patients were in special education and had a recognised disability of >50%. Two of them presented EEG alterations but with no seizure manifestation. All five patients presented an anxiety disorder, requiring antidepressant treatment (SSRI). All five patients show common dysmorphic features: deep-set eyes, hypotelorism, synofridia, high palate and discrete hypermobility

Conclusions: We have identified common dysmorphic and neuropsychiatric features in patients with SHANK pathogenic variants. The inclusion of encephalographic recordings may allow a preventive epilepsy treatment. The clinical description of patients with SHANK pathogenic variants may facilitate a more precise clinical management and possible use of targeted therapies.

Grants: This work is supported by Instituto de Salud Carlos III-FEDER, (PI19/01902).

Conflict of Interest: None declared

EP11.026 Clinical and neuroradiological spectrum of biallelic variants in NOTCH3

Pablo Iruzubieta 1;2, Cesar Augusto Pinheiro Ferreira Alves3, Aisha Mohamed Saeed Mohamed Al Shamsi4, Gehad ElGhazali5, Lorenzo Pinelli6, Diego Lopergolo7, Bernard P H Cho8, Amna Mohammed Al-Futaisi9, Fatema Al-Amrani9, Jessica Galli10, Katarina Vulin11, Elisa Fazzi10, Francisco Barajas-Olmos12, Holger Hengel13, Amy Jolly8, Maha Zaki14, Bayan Mohammed Aljamal15, Fowzan Alkuraya15, Michele Ragno16, Luigi Trojano17, Silvia Bianchi7, Francesca Pescini7, Amal Al Tenalji18, Majid Aziz18, Miriam Schmidts19;20, Giovanni Zifarelli21, Ludger Schöls22, Tobias Haack23, Adriana Rebelo24, Stephan Zuchner24, Lyn Griffiths25, Arman Cakar1, Lorena Sofía Orozco Orozco26, Karla García Helmes27, Meisam Babaei28, Peter Bauer21, Won Chan Jeong29, Ehsan Ghayoor Karimiani30, Wendy Chung31, Vahideh Nasr32, farhad assarzadegan32;33, Bita Shalbafan33, Hugh Markus8, Henry Houlden1, Reza Maroofian1

1UCL Queen Square Institute of Neurology, London, United Kingdom; 2Donostia University Hospital, San Sebastian, Spain; 3Children’s Hospital of Philadelphia, Department of Radiology, Division of Neuroradiology, Philadelphia, United States; 4Tawam Hospital, Al-Ain, United Arab Emirates; 5Sheikh Khalifa Medical City- Union71- Purehealth, Abu Dhabi and College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates; 6Neuroradiology Unit, Pediatric Neuroradiology Section, ASST Spedali Civili, Brescia, Italy; 7Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy. UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Siena, Italy; 8Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom; 9Department of Child Health. College of Medicine and Health Sciences. Sultan Qaboos University, Oman; 10Child Neurology and Psychiatry Unit, ASST Spedali Civili of Brescia, Brescia, Italy; 1114Department of Medical and Laboratory Genetics, ERN-Ithaca Zagreb Center, Children’s Hospital Zagreb, Zagreb, Croatia; 12Immunogenomics and Metabolic Disease Laboratory, Instituto Nacional de Medicina Genómica, SS, Mexico, Mexico City, Mexico; 13, Tübingen, Germany; 14Human Genetics and Genome Research Division, Clinical Genetics Department, National Research Centre, Cairo, Egypt; 15Department of Translational Genomics, Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia; 16Pianeta Salute, Viale Assisi, 88, 63084 Villa Pigna, Ascoli Piceno, Italy; 17Department of Psychology, University of Campania ‘Luigi Vanvitelli’, Viale Ellittico 31, Caserta, Italy; 18Sheikh Khalifa Medical City- Union71- Purehealth, Abu Dhabi and College of Medicine and Health Sciences, United Arab Emirates University, United Arab Emirates; 19Pediatrics Genetics Division, Center for Pediatrics and Adolescent Medicine, Faculty of Medicine, Freiburg University, Mathildenstrasse 1, Freiburg, Germany; 20CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany; 21Centogene, Germany; 22Department of Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany; 23Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany; 24Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, United States; 25Centre for Genomics and Personalised Health. Bridge and BridgeTech Programs. Queensland University of Technology., Queensland, Australia; 26Department of Genetics, General Hospital Dr. Aurelio Valdivieso, Oaxaca de Juárez, Oaxaca, Mexico; 27Department of Genetics, General Hospital Dr. Aurelio Valdivieso, Oaxaca de Juárez, Oaxaca, Oaxaca, Mexico; 28Department of Pediatrics, North Khorasan university of medical sciences, Bojnurd, Iran; 293billion, Seoul, Korea, Rep. of South; 30Genetics Section, Molecular and Clinical Sciences Research Institute, St. George’s, University of London, London, United Kingdom; 31Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, United States; 32Department of Neurology - Kermanshah Imam Reza (AS) Hospital Complex, Kermanshah University of Medical Sciences., Kermanshah, Iran; 33Clinical Research Development Center of Labbafinejad Hospital, Shahid Beheshti University of Medical Sciences. Tehran, Iran., Teheran, Iran

Background/Objectives: NOTCH3 variants are the most frequent cause of hereditary cerebral small-vessel disease (SVD). Although cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is well-studied, autosomal recessive (AR) forms of NOTCH3-SVD are ultra-rare and poorly characterised.

Methods: Here we delineate the genotype-phenotype correlation of AR-NOTCH3-SVD. We describe 25 new patients with biallelic NOTCH3 variants and review the literature to identify additional cases.

Results: We show that biallelic truncating variants lead to a neurodevelopmental disorder with spasticity, seizures, childhood-onset stroke and periatrial white matter volume loss (periventricular leukomalacia-like appearance), while patients with biallelic cysteine missense variants exhibit a “CADASIL-like” phenotype with early adulthood onset, stroke, dementia and deep white matter lesions without significant volume loss. Unlike carriers of missense variants, carriers of truncating variants are mainly asymptomatic.

Conclusion: NOTCH3-SVD are more complex than previously appreciated. Here we focus on biallelic NOTCH3-SVD to further elucidate the genotype-phenotype variability in these patients who show two distinct phenotypes likely related to two different disease mechanisms.

Grants: The Wellcome Trust, The MRC, The MSA Trust, The National Institute for Health Research University College London Hospitals Biomedical Research Centre, The Michael J Fox Foundation (MJFF), BBSRC, The Fidelity Trust, Rosetrees Trust, Ataxia UK, Brain Research UK, Sparks GOSH Charity, Alzheimer’s Research UK (ARUK), CureDRPLA. PI was supported by a European Academy of Neurology Clinical Fellowship and a Fellowship from the Movement Disorders Group from the Spanish Society of Neurology.

Conflict of Interest: None declared

EP11.027 RFC gene variant as a potential marker of remitting-relapsing course of multiple sclerosis

Zoia Rossokha 1, Maya Lafarenko2, Nazar Negrych3, Liliia Fishchuk1, Tetyana Nehrych4

1Reference-centre for molecular diagnostic of Public Health Ministry of Ukraine, Molecular genetics lab, Kyiv, Ukraine; 2Lviv regional clinical hospital, Department of Neurology, Lviv, Ukraine; 3Danylo Halytsky Lviv National Medical University, Department of Social Medicine, Economics and Organization of Health Care, Lviv, Ukraine; 4Danylo Halytsky Lviv National Medical University, Department of Neurology, Lviv, Ukraine

Background: Multiple sclerosis (MS) is a chronic, progressive autoimmune disease of the central nervous system, that is one of the leading causes of disability for young people worldwide. The issues of the etiology and pathogenesis of this disease and remitting-relapsing course are not fully understood, and therefore it is extremely important to determine new genetic mechanisms. The gene RFC encodes the solute carrier family 19 member 1 – transmembrane protein that is involved in intracellular folate transport. The current study aimed to examine the association of variant rs1051266 of the RFC gene with the risk of remitting-relapsing MS.

Methods: There were conducted a case-control study including 88 patients with remitting-relapsing MS (moderate and severe course) and 70 healthy volunteers (comparative group). In all patients, the diagnosis of MS was confirmed according to the 2017 McDonald criteria. Genotyping of the RFC gene was performed by the PCR-RFLP method.

Results: The genotype frequencies were GG – 28.4%, GA – 46.6% and AA – 25.0% in patients with MS, versus GG – 48.6%, GA – 32.9% and AA – 18.6% in comparative group. The GG genotype was significantly higher in comparative group (χ2 = 6.77, p = 0.009, OR = 0.50 (0.26-0.97)). Also was found differences between investigated groups in allele frequencies. Allele A was significantly more common in the patients with MS (χ2 = 5.64, p = 0.02, OR = 1.73 (1.10-2.74)).

Conclusion: There is an association of the variant rs1051266 of the RFC gene and the risk of remitting-relapsing MS development, but large-scale studies are still needed to support our findings.

Conflict of Interest: None declared

EP11.028 The phenotypic spectra of Hungarian patients with late-onset Tay-Sachs disease: presenting symptoms, patient routes, disease progression and therapeutic experience

Idris Janos Jimoh 1, Agnes Palasti1, Viktor Molnár1, Zsuzsanna Aranyi2, Zoltan Grosz1, Maria Judit Molnar1

1Semmelweis University, Institute of Genomic Medicine and Rare Disorders, Budapest, Hungary; 2Semmelweis University, Department of Neurology, Budapest, Hungary

Background/Objectives: Tay-Sachs disease is a rare neurogenetic disease caused by pathogenic variants of the HEXA gene. The disease can be divided into juvenile, infantile, and late-onset (LOTS) forms that are associated with different residual enzyme activities. The identification of LOTS patients still remains a diagnostic challenge due to the wide range of presenting symptoms and different patient routes. Thus, despite the long history of the disease, characterization of late-onset patients is still lacking. We aimed to demonstrate the phenotypic spectra of Hungarian LOTS patients.

Methods: We demonstrate a case series of 3 patients. Deep-phenotyping by physical examination, MRI, EMG, myopathology, enzyme activity, and detailed follow-up were performed.

Results: The presenting symptoms varied. Patient1 with homozygous p.Gly269Ser variant had initially psychotic symptoms from the age of 18, followed by muscle weakness twelve years later, whereas Patient2 with compound heterozygous p.Gly269Ser and p.Ser215Asn variants had presenting symptoms of ataxia, mild paraparesis, and dysthymia since the age of 30 years, Patient3 had compound heterozygous p.Gly269Ser and p.Tyr427Ilefs*5 variants causing muscle hypotrophy and paraparesis since the age of 18 years. One of the patients has been treated with Miglustat for 1.5 years resulting in stabilization of his symptoms.

Conclusion: With the patient’s different phenotypes, we can demonstrate, the wide phenotypic spectra in which neurogenic muscle weakness and psychiatry symptoms can be the red flags. The p.Gly269Ser rare variant is common in the Ashkenazi population but it is commonly identifiable in Hungary as well in LOTS.

Grants: TKP2021-NVA-15, OTKA139010

Conflict of Interest: None declared

EP11.029 A novel nucleotide deletion variant in the KCNT1 gene in a patient diagnosed with epilepsy.

Duygu Gamze Aracı 1, Özden Altıok Clark1, Öznur YILMAZ BAYER2, banu nur2, Senay Haspolat3, Aslı Toylu1, ercan mihci2

1Akdeniz University Faculty of Medicine, Medical Genetics, Antalya, Türkyie; 2Akdeniz University Faculty of Medicine, Pediatric Genetics, Antalya, Türkyie; 3Akdeniz University Faculty of Medicine, Pediatric Neurology, Antalya, Türkyie

Background/Objectives: The KCNT1 gene encodes a sodium-activated potassium channel, crucial for regulating neuronal excitability and highly expressed in the nervous system. Variants in KCNT1 have been associated with several epilepsy syndromes, including epilepsy of infancy with migrating focal seizures (EIMFS) and autosomal dominant nocturnal frontal lobe epilepsy (ADNFLE). This study aims to evaluate the clinical and genetic characteristics of a patient presenting with seizures suspected to be related to a novel KCNT1 variant, c.1116_1123del.

Methods: The patient’s genomic DNA was extracted from a peripheral blood sample. Exome sequencing was performed. Variants were analyzed using the Sophia DDM program. Variant information servers dbSNP, ClinVar, Ensembl, and ACMG 2015 criteria were used for evaluations.

Results: In the KCNT1 gene, c.1116_1123del (p.Lys372AsnfsTer121) was identified in the patient. This deletion is predicted to result in a frameshift and premature truncation of the protein, potentially disrupting the normal function of the sodium-activated potassium channel. This variant is not present in ClinVar or any population/disease-specific databases.

Conclusion: Detecting the novel KCNT1 variant c.1116_1123del suggests a potential genetic cause for our patient’s syndrome. However, further functional studies are needed to directly assess this specific variant’s impact on channel function. Our findings show that changes in the KCNT1 gene can underlie a broad spectrum of epilepsy phenotypes and highlight the importance of genetic testing in the diagnosis and management of epilepsy.

Grants: None.

Conflict of Interest: None declared

EP11.030 A case of Perrault syndrome with a novel variant diagnosed at a late stage with predominance of neurological findings.

hacer ukba kına 1, Neslihan Cinkara1, Momen Kanjee2

1Ministry of Health of Turkey, Trabzon Training and Research Hospital, Department of Medical Genetics, Trabzon, Türkyie; 2Ataturk University, Faculty of Medicine, Department of Medical Genetics, Erzurum, Türkyie

Background/Objectives: Perrault syndrome (OMIM # 233400) is a rare autosomal recessive disorder characterized by hearing loss of variable severity and ovarian dysgenesis. It is divided into subtypes according to the presence or absence of neurological components. So far, 8 genes have been identified in the etiology of the disease. In this report, we present a case with a compound heterozygous novel mutation in the HSD17B4 gene who presented to our clinic with gait disturbance and loss of balance.

Methods: Informed consent was obtained from the patient and DNA was extracted from peripheral blood. Next-generation sequencing, using a ROCHE kit, was performed with MGI DNBSEQ-400 device. The results were analyzed with the Genomize Seq program

Results: A 44-year-old woman was referred to our clinic with gait and balance disorders. It was learned that the patient’s complaints started at the age of 22, she had mild hearing loss since childhood, and she entered menopause at the age of 30. Brain MR imaging revealed cerebellar atrophy. The NGS analysis of the patient, whose parents were not related, revealed compound heterozygous variants in the HSD17B4 gene: a 5’ UTR c.-133G > A and a novel missense c.425A>G p.(D142G)

Conclusion: Perrault syndrome is a rare disease with clinical and genetic heterogeneity with approximately 100 cases reported to date. Patients are mostly diagnosed in childhood due to hearing loss. In this report, we presented a patient with a novel variant, exhibiting predominant neurological findings, diagnosed at a late stage compared to the literature.

Grants: None.

Conflict of Interest: None declared

EP11.031 Molecular findings and phenotypic comparison of Hungarian patients with PRRT2-associated epilepsy

András Szabó 1, Márta Czakó1, Gergely Büki1, Zsolt Bánfai1, Anita Maasz1, Agnes Till1, Barbara Patócs2, Judit Bene1, Kinga Hadzsiev1

1University of Pécs, Medical School and Clinical Centre, Department of Medical Genetics, Pécs, Hungary; 2Bethesda Children’s Hospital, Department of Pediatric Neurology, Budapest, Hungary

Background/Objectives: PRRT2 point mutations and deletions have been identified as a causal factor behind the development of paroxysmal movement disorders including benign infantile epilepsy, paroxysmal kinesigenic dyskinesia, and infantile convulsions with paroxysmal choreoathetosis providing the core features of the PRRT2-related phenotypic spectrum. Hereby, we report phenotypic comparison of the first Hungarian patients with PRRT2 point mutations and a deletion affecting the entire gene, supplementing the previously described phenotypic picture.

Methods: aCGH, WES or various next-generation panel investigations were applied for the examination of Hungarian patients with seizures, paroxysmal movement disorders or complex phenotypic picture involving these features. Detailed genotype-phenotype analysis was performed.

Results: aCGH examination of the investigated patients revealed a 115.8 kb copy number loss of the 16p11.2 chromosomal region affecting eight genes including PRRT2. WES and next-generation panel investigations revealed a PRRT2 c.649_650dupC heterozygous frameshift mutation in six patients. Infantile convulsions and/or choreathetosis were observed in all patients with point mutation. Our patient with PRRT2 deletion showed more complex phenotype including infantile convulsions, extreme obesity, renal agenesis, patent ductus arteriosus, neurodevelopmental delay, intellectual disability and dysmorphic features.

Conclusion: Our results contribute to the expansion of the clinical spectrum of patients with PRRT2-associated paroxysmal movement disorders described so far and highlights the importance of application of aCGH and next-generation sequencing and detailed phenotyping of such patients. Furthermore, the complex phenotypic picture of our patient with the microdeletion emphasize the possible role of the additional affected genes in the development of the 16p11.2 deletion-related phenotype.

Grants:

Conflict of Interest: None declared

EP11.032 Novel pathogenic variant in TSC2 gene in association with rhabdomyoma administered with intrauterine mTOR inhibitor

Anita Maasz 1, Tímea Bodó2, Agnes Till1, Gábor Molnár3, György Masszi4, Gusztáv Labossa3, Zsuzsanna Herbert5, Judit Bene1, Kinga Hadzsiev1

1University of Pécs, Medical School and Clinical Centre, Department of Medical Genetics, Pécs, Hungary; 2Bethesda Children’s Hospital, Budapest, Hungary; 3University of Pécs, Medical School and Clinical Centre, Department of Obstetrics and Gynaecology, Pécs, Hungary; 4University of Pécs, Medical School and Clinical Centre, Department of Paediatrics, Pécs, Hungary; 5University of Pécs, Medical School and Clinical Centre, Department of Medical Imaging, Pécs, Hungary

Background/Objectives: Tuberous sclerosis complex (TSC) is a rare multisystem genetic disorder characterized by tumours of the heart, brain, skin, lungs, and kidneys, seizures, and neuropsychiatric disorders. Variants of TSC2 and TSC1 genes have been associated with the disease.

Methods: A pregnant woman in her 30th weeks of gestation was referred for examination. In her fetus, left ventricular rhabdomyoma were detected. Umbilical cord blood samples from the fetus were taken and analysed by multiplex ligation-dependent probe amplification (MLPA) method and by Sanger sequencing.

Results: A de novo heterozygous frameshift variant in TSC2 gene (c.3037delG variant, p.Asp1013IlefsTer3, rs137854301) was detected. The classification of this TSC2 variant provides strong evidence of pathogenicity. A unique five-week oral immunosuppressant therapy (everolimus) given to the mother reduced the size of the rhabdomyoma. The child’s status has been monitored for four years. The patient had good vitals and was under regular cardiological control, showed balanced circulation. The symptoms were well-manageable, however temporarily. Therapy-resistant focal seizures were frequent. Subependymal giant cell astrocytoma (SEGA) identified by regular neuroimaging examinations remained unchanged, which may be a consequence of early intrauterine treatment.

Conclusion: Our study broadens the genetic spectrum of TSC and emphasizes the importance of timing of genetic tests and interventions and close monitoring, lifelong multidisciplinary follow-up, and management of patients with TSC.

Grant: This study was supported by the Medical School of University of Pécs, “ÁOK-KA” Research Program; KA-2023-22.

Conflict of Interest: None declared

EP11.033 Correlation between DNA methylation of buccal cells and brain activity (fNIRS) in pre-schooler females with ASD: a proof-of-concept observational clinical study

Andrea Stoccoro 1, Eugenia Conti2, Elena Scaffei2, Laura Baroncelli3, Marianna Giangreco1, Sara Calderoni2, Fabio Coppedè1, Lucia Migliore1, Roberta Battini2

1University of Pisa, Department of Translational Research and of New Surgical and Medical Technologies, Pisa; 2IRCCS Stella Maris Foundation, Department of Developmental Neuroscience, Pisa; 3National Research Council, Institute of Neuroscience, Pisa

Background/Objectives: Diagnosis of autism spectrum disorder (ASD) remains challenging, particularly in young females, due to the heterogeneity of the behavioral phenotype and the capacity of camouflage. Epigenetic alterations, including DNA methylation, and brain activity could help in ASD neurobiological characterization, as well as in providing non-invasive biomarkers for ASD, even before full-blown expression of symptoms. The current study aimed to investigate the feasibility of combining DNA methylation and brain activity to identify and characterize female ASD.

Methods: The study population included 12 ASD and 13 typically developing (TD) pre-schooler females. DNA methylation levels of 15 genes involved in ASD were evaluated in buccal swab cells. Functional Near-infrared Spectroscopy (fNIRS) was used to evaluate visually-evoked cortical hemodynamic responses as a proxy of brain activity.

Results: Methylation levels of miR28 and MTHFR genes were lower in ASD compared to TD subjects. Moreover, methylation of five genes, including MTHFR, miR23/27, miR30e, BCL2, BDNF and EN2 correlated with different fNIRS signal features. Among them, MTHFR methylation positively correlated with oxygenated hemoglobin (OHb). Interestingly, we previously showed that the average amplitude of cortical changes of OHb concentrations was significantly lower in ASD vs TD group.

Conclusion: Our preliminary data show that peripheral DNA methylation combined with fNIRS analyses could provide useful non-invasive biomarkers for young ASD females, as well as new insights of ASD etiology. Of note are the associations among MTHFR methylation, oxygenated OHb concentrations and ASD status, which deserve to be further investigated.

Grants: Progetti di Ricerca di Ateneo (PRA_50_2020), University of Pisa

Conflict of Interest: None declared

EP11.034 Rab11-binding protein RELCH/KIAA1468 is a potential candidate gene for global developmental delay, Intellectual disability with microcephaly and seizures

Lubaba Bintee Khalid1, Bilal Ahmad Mian1, Asmat Ali1, Mina Shahid2, Mathias Toft3;4, Zafar Iqbal4, Ambrin Fatima 1

1Aga Khan University, Department of Biological and Biomedical Sciences, Karachi, Pakistan; 2Aga Khan University, Department of Radiology, Karachi, Pakistan; 3University of Oslo, Institute of Clinical Medicine, Oslo, Norway; 4Oslo University Hospital, Department of Neurology, Oslo, Norway

RELCH/KIAA1468 is a Rab11-GTP binding protein involved in a novel nonvesicular mechanism of cholesterol transport from recycling endosomes (REs) to the trans-Golgi network (TGN). So far variants in RELCH/KIAA1468 have never been linked with any human disease. We identified two patients from a highly consanguineous Pakistani family, presenting with global developmental delay, intellectual disability, microcephaly, generalized spasticity with hand dystonia, no response to visual stimuli or auditory cues, and Spontaneous movement in all limbs. The patients are not able to sit, stand or hold their neck. Whole-exome sequencing of the affected child identified a frameshift variant in KIAA1468:c.2988_2989del (NM_001346231);p.N996fs which segregated with the phenotype. The patient’s MRI displayed severe thinning of corpus callosum, Gyral dysmorphism in both occipital lobes with Encephalomalacia in left occipital lobe and Ventriculomegaly. Moreover, we initiated functional experiments in patient-derived cells and zebra fish to unravel the pathogenicity of novel variant identified in our patients.

Conflict of Interest: None declared

EP11.035 Exome re-analysis after deep phenotyping and clinical updating: an example of effective dynamic workflow to reach a diagnosis

Luciana Musante 1, Maria Teresa Bonati1, Giulia Maria Di Marzio1, Martina La Bianca1, Giorgia Girotto1;2, Caterina Zanus1

1Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, Italy; 2Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy

Background/Objectives: Neurodevelopmental disorders (NDDs) are estimated to have a prevalence of 5-10% in Europe. Whole Exome Sequencing (WES) is considered the first-tier genetic test for patients with NDDs. Although a diagnosis can be made in about 30-53%, many patients remain undiagnosed. The aim of the present work is to highlight the utility of specific HPO terms to finalize the WES data reanalysis after a non-diagnostic result and the molecular outcome.

Methods: Systematic patient clinical evaluation; HPOs reviewed in a multidisciplinary team. WES-trio data re-analysis. In silico and in vitro functional studies.

Results: We report on a girl referred at 8 months for postnatal microcephaly and motor delay. Brain MRI showed increased periencephalic subarachnoid liquor spaces, under-representation of the white matter, thinning of the corpus callosum. At that time WES analysis resulted negative. However, at 14 months she developed epilepsy with nocturnal tonic seizures. At 18 months, she presented with global developmental delay, absent speech, lack of attention and eye contact, brachymicrocephaly, minor dysmorphisms, drooling and poor use of hands. WES re-analysis identified a novel compound heterozygous genotype of AP3B2 gene (MIM: #617276, DEE48): a missense variant (NM_001278512: c.2728G>A, p.Val910Met) predicted to result in decreased protein stability and the c.2497+11G > C expected to generate a novel donor splice-site leading to a premature stop codon. Functional studies on patient’s fibroblasts are in progress.

Conclusion: Our results illustrate how a dynamic workflow revisiting patient’s genetic data over time together with a systematic clinical re-assessment, represents an efficient strategy to improve pathogenic variants identification.

Conflict of Interest: None declared

EP11.037 Genetic Risk Factors for Developing Wernicke-Korsakoff Syndrome (WKS) in an Alcohol-Dependence Cohort: A Polygenic Risk Score Analysis of Serum Vitamin Concentrations and Dementia Subtype

Yiwen Huang1, Irene Guerrini2;3, Iain Smith4, Mathis Heydtmann5, Marsha Morgan6, Allan Thomson1;3, Nicholas Bass1, Andrew McQuillin 1

1University College London, Division of Psychiatry, London, United Kingdom; 2South London and Maudsley NHS Foundation Trust, London, United Kingdom; 3King’s College London, Institute of Psychiatry, Psychology and Neuroscience, London, United Kingdom; 4Alcohol Related Brain InjuryTeam, Stirling, United Kingdom; 5Dumfries & Galloway Royal Infirmary, Department of Gastroenterology, Cargenbridge, United Kingdom; 6University College London, UCL Institute for Liver & Digestive Health, London

Background/Objectives: Wernicke-Korsakoff syndrome (WKS) is a severe neurological disorder most commonly found in individuals with severe alcohol use disorder; it is associated with deficiencies of the B vitamins, particularly thiamine (vitamin B1). However, the genetic susceptibility to WKS among alcohol-dependent individuals remains underexplored. This study employs a polygenic risk score (PRS) analysis to investigate the genetic risk factors underlying WKS development, emphasizing the interplay between circulating vitamin B concentrations and dementia subtypes.

Methods: The study includes participant selection, genotyping, and statistical analysis, encompassing a genome-wide association study (GWAS) for WKS development (cases 214; controls 2733) and PRS analysis of serum vitamin B concentrations and dementia subtypes on WKS development within an alcohol dependence cohort.

Results: No genome-wide significant SNPs were found in the GWAS for WKS development among alcohol-dependent individuals. Gene based tests and pathways enrichment analyses of the WKS GWAS data did not identify likely risk mechanisms. The genetic risk associated with serum vitamin B3 (R2 0.013; P 0.00094) and vitamin B12 (R2 0.005; P 0.047) concentrations were correlated in genetic dimensions with WKS development within this cohort.

Conclusion: This research advances the understanding of the genetic risk factors associated with WKS in alcohol dependent individuals and highlights the role of the B vitamins, especially vitamin B3.

Grants: This work was supported by the Brain Damage Research Trust

Conflict of Interest: None declared

EP11.038 Phenotypical diversity in a family with EIF2AK2-associated disease.

Joseph Porrmann 1, Sylvia Hütter1, Jens Schallner2, Andreas Dahl3, Karl Hackmann1, Marcus Franke1, Evelin Schröck1;4

1University Hospital Carl Gustav Carus at TUD Dresden University of Technology and Faculty of Medicine of TUD Dresden University of Technology, Dresden, Institute for Clinical Genetics, Dresden, Germany; 2Faculty of Medicine of TUD Dresden University of Technology and University Hospital Carl Gustav Carus at TUD Dresden University of Technology, Dresden, Germany, Department of Pediatrics, Dresden, Germany; 3TUD Dresden University of Technology Germany, DRESDEN-concept Genome Center, Center for Molecular and Cellular Bioengineering, Dresden, Germany; 4Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany, Dresden, Germany

Background: EIF2AK2 encodes the protein kinase R which plays a role in the cellular integrated stress response. Heterozygous variants in EIF2AK2 have been associated with LEUDEN syndrome (LEUkoencephalopathy, Developmental delay, and Episodic Neurological regression syndrome, MIM 618877). Additional symptoms can include motor abnormalities, cognitive impairment, corpus callosum hypoplasia and microcephaly. Notably, further patients with heterozygous EIF2AK variants have been described with mostly isolated dystonias (MIM 619687) as well as with Pelizaeus-Merzbacher-like disease. Functional studies have suggested a loss-of-function effect of variants causing LEUDEN and a gain-of-function effect of variants leading to dystonia.

Methods: We report on a 10-year-old boy with fever induced developmental regression, hypomyelination, corpus callosum hypoplasia, microcephaly, ataxia, spasticity and dystonia for whom whole genome sequencing and EIF2AK2 variant segregation was performed.

Results: Sequencing of the index patient detected a heterozygous variant in EIF2AK2, that had previously been described in a patient who likewise showed features of LEUDEN. Segregation showed that his brother and mother also carried the variant. The 6-year-old brother had developmental delay, hypomyelination, corpus callosum hypoplasia, microcephaly but no episodes of regression and the mother presented with isolated hemidystonia and choreoathetosis that was not compatible with LEUDEN syndrome.

Conclusion: While all previously reported patients with LEUDEN carried (likely) de novo variants, we report the first family with an inherited EIF2AK2 variant and a variable phenotype. Even though maternal mosaicism cannot be fully excluded, the different phenotypes within a single family indicate variability that questions the strict genotype-phenotype association and functional dichotomy of gain-of-function versus loss-of-function variants.

Conflict of Interest: None declared

EP11.039 Association of RANBP2 and CHRNB1 mutations and Acute Necrotizing Encephalopathy

Mustafa Bakırtaş 1, Alara Akdeniz2, Hazel Delal Dara Kar3, Zerrin Çelik1, Yunus Kasım Terzi1, Halil İbrahim Aydın4

1Baskent University, Medical Genetics, Ankara, Türkyie; 2Gaziantep Children’s Hospital, Pediatrics, Gaziantep, Türkyie; 3Polatli Duatepe State Hospital, Pediatrics, Ankara, Türkyie; 4Baskent University, Inborn Errors of Metabolism, Ankara, Türkyie

Background/Objectives: Acute Necrotizing Encephalopathy(ANE) is a rapidly progressive encephalopathy associated with viral pathogens. Most ANEs occur sporadic and non-recurrent. RANBP2 pathogenic variants predispose individuals to recurrent ANE(ANE1), described as Infection-induced acute encephalopathy 3 (IIAE3)[MIM:608033]. It is inherited in an autosomal dominant manner with reduced penetrance. In this study, we aimed to discuss a patient with ANE in whom we found a pathogenic variant in RANBP2 and a variation of unknown clinical significance in CHRNB1.

Methods: The genomic DNA was extracted from the peripheral blood sample. Whole-exome sequencing (WES) was performed using the MGI-DNBSEQ-G400 instrument. Confirmation of the results and segregation analysis were done by the Sanger sequencing.

Results: A 2-year 5-month-old boy born to unrelated healthy parents was consulted to our clinic for developmental delay and hypotonia. He developed varicella at 3 months and had a febrile convulsion at 8 months. Metabolic tests were unremarkable. MRI and clinical presentation suggested ANE. WES revealed a novel maternally inherited mutation of RANBP2[c.1430A>G] and paternally inherited CHRNB1[c.610+4A > G] mutation.

Conclusion: CHRNB1 encodes the beta subunit of Nicotinic Acetylcholine Receptor(nAChR). Some viruses like Rabies virus(RABV) and SARS-CoV-2 show their pathogenic effects via nAChRs. Viral pathogens play a crucial role in ANE1 pathophysiology. Thus, CHRNB1 could act as another susceptibility factor for ANE1 when inherited with a RANBP2 variant. We report the first case of ANE1 with a novel RANBP2 mutation. Our patient is the only family member who harbors both mutations and fulfills the diagnostic criteria of ANE. RANBP2 and CHRNB1 mutations could lead to ANE1 when inherited together.

Grants: No

Conflict of Interest: None declared

EP11.040 Hypomyelinating leukodystrophy-10 with identified homozygous mutation in the PYCR2 gene: A new Moroccan case

Houda Jelti 1;2, Sabrine Bouressas1;2, Abdelhaq Lamaibdel1;2, Oussama Kettani1, Jaouhara MAAMAR1;2, Ouafae Kaissi1, Badreddine Nouadi1, Hasna Hamdaoui1, Afaf Lamzouri1;2

1Department of Medical Genetics and Oncogenetics, Mohammed VI University Hospital, Tangier, Morocco; 2Life and Health Sciences Laboratory, Faculty of Medicine and Pharmacy of Tangier, Abdelmalek Essaâdi University, Morocco

Background: Hypomyelinating leukodystrophy-10 (OMIM #616420), is a rare inherited neurologic disorder characterized by progressive postnatal microcephaly, severely delayed psychomotor development, and observable hypomyelination in brain imaging. This disorder follows an autosomal recessive inheritance pattern attributed to mutations in the PYCR2 gene which is mapped to chromosome 1q42 and encodes an enzyme involved in the proline biosynthesis pathway. Initially described in 2015, only 24 cases have been reported worldwide. In this report, we present an additional case to expand our understanding of this recently identified syndrome.

Method: A five-years-old girl, first-born child of a healthy non-consanguineous couple in northern Morocco, was referred to genetics clinical counseling for evaluation of a microcephaly, chronic diarrhea, excessive vomiting and failure to thrive. Clinical examination revealed a global development delay with spasticity, muscular amyotrophy, hyperkinetic movements and dysmorphic features. Brain MRI revealed a thin corpus callosum and cortical atrophy.

Result: Whole-exome sequencing identified a homozygous pathogenic variant in PYCR2, confirming hypomyelinating leukodystrophy-10. Segregation analysis revealed that both parents were heterozygous carriers of the same variant. Genetic counseling was provided to the family, who opted for either preimplantation or prenatal diagnosis for future pregnancies.

Conclusion: The identification of the PYCR2 variant underscores the crucial role of the geneticist in diagnosing this condition and providing genetic counseling to families, aiming to minimize the risk of recurrence in future pregnancies. Although the disease is exceptionally rare and recently identified, the number of cases is expected to increase with expanded use of exome/genome-sequencing, highlighting the need for ongoing research to explore potential therapeutic strategies.

Conflict of Interest: None declared

EP11.041 Association of miRNA expression in plasma and cerebrospinal fluid with Alzheimer’s disease biomarkers

David Vogrinc 1, Milica Gregorič Kramberger2;3;4, Andreja Emeršič2, Saša Čučnik2;5;6, Katja Goricar1, Vita Dolzan1

1Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Pharmacogenetics Laboratory, Ljubljana; 2University Medical Centre Ljubljana, Department of Neurology, Ljubljana; 3Faculty of Medicine, University of Ljubljana, Ljubljana; 4Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, Huddinge, Sweden; 5University Medical Centre Ljubljana, Department of Rheumatology, Ljubljana; 6Faculty of Pharmacy, University of Ljubljana, Ljubljana

Background/Objectives: Circulating miRNAs recently emerged as novel potential biomarker of Alzheimer’s disease (AD) and mild cognitive impairment (MCI) as a support to cerebrospinal fluid (CSF) biomarkers in earlier diagnosis of the disease. Our aim was to assess the association of target miRNA expression in plasma and CSF with biomarker levels (amyloid beta (Aβ) and tau protein from CSF) or cognitive test (MMSE) score.

Methods: Our study included 62 AD patients, 24 MCI patients with pathological CSF biomarker levels and 31 MCI patients with normal CSF biomarker levels. The expression of target miRNAs (hsa-miR-146a-5p, hsa-miR-451a, hsa-miR-30c-5p, hsa-miR-375-3p, hsa-miR-107, hsa-miR-193b-3p and hsa-miR-29c) in plasma and CSF samples was determined with qPCR, using TaqMan™ Advanced miRNA Assays.

Results: In the entire cohort, higher expression of miR-30c-5p in plasma was associated with higher CSF total tau (p = 0.035). In CSF samples, higher expression of miR-146a-5p was associated with higher p-tau181 (p = 0.042), while higher expression of miR-107 was associated with higher total tau (p = 0.025). In the AD group, higher expression of miR-146a-5p in plasma was associated with lower Aβ42/40 (p = 0.027), while higher expression of miR-29c in CSF was associated with lower Aβ42/40 (p = 0.023). Higher expression of miR-146a-5p in CSF was associated with lower MMSE scores in the entire cohort (p = 0.039).

Conclusion: We showed important associations of miR-146a-5p, miR-30c-5p, miR-375-3p, miR-107 and miR-29c with AD CSF biomarkers and MMSE scores that support their potential as circulating AD biomarkers.

Grants: ARRS P1-0170.

Conflict of Interest: None declared

EP11.042 Pure parkinsonism as possible phenotype expansion of THAP1-related disorders

Enrico Ambrosini 1, Rita Cancilla2, Jefri Jeya Paul3, Peter Bauer3, Barbara Garavaglia4, Valeria Barili5, Antonio Percesepe1;5, Anna Negrotti6

1University of Parma - Hospital, Medical Genetics Unit, Parma, Italy; 2University of Parma - Department of Medicine and Surgery, Neurology, Parma, Italy; 3CENTOGENE GmbH, Rostock, Germany; 4Istituto Neurologico “Carlo Besta” | Fondazione IRCCS, Medical Genetics and Neurogenetics Unit, Milano, Italy; 5University of Parma - Department of Medicine and Surgery, Medical Genetics, Parma, Italy; 6University of Parma - Hospital, Neurology Unit, Parma, Italy

Background/Objectives: Parkinson’s disease (PD) and dystonia show clinical and genetic heterogeneity and a certain degree of overlap. A particular form of autosomal dominant dystonia, mainly characterized by focal involvement, is caused by variants in THAP1. Parkinsonism is not a typical symptom of the disease, but a gene-based burden analysis and a recent case report indicate a possible role of this gene in PD.

We describe a patient with parkinsonism, no signs of dystonia and a likely pathogenic variant in THAP1.

Methods: Clinical diagnosis was made according to the criteria of the “UK Parkinson’s disease Brain Bank”. Patient underwent brain MRI and SPECT. She was involved in the ROPAD (Rostock International Parkinson’s Disease) study, including a Next Generation Sequencing panel targeting 68 defined or suspected PD-relevance genes.

Results: A 61-year-old female patient, referred to the Movement Disorders outpatient clinic for resting tremor of the right upper limb, hyposmia and micrographia, was diagnosed with bilateral idiopathic parkinsonism, prevailing on the right side. Family history was unremarkable.

Genetic testing did not identify a clear pathogenic variant in PD-related genes, while reporting a heterozygous variant in THAP1 (c.70_71+8del). This ten-nucleotide deletion can be interpreted as “Likely Pathogenic” according to ACMG Guidelines. The same variant had already been reported in a patient with cervical dystonia.

Conclusion: Given the reduced penetrance of THAP1 variants, it cannot be excluded that the variant reported in our patient is just an incidental finding. Nevertheless, evidence of the involvement of this gene in PD is increasing, suggesting a phenotype expansion of THAP1-related diseases.

Grants: -

Conflict of Interest: None declared

EP11.044 A Clinical Case of targeted memantine treatment in a patient with GRIN2A Mutation and Epileptic Encephalopathy.

Shokhijakhon Shokhimardonov 1, Nodira Tuychibaeva1

1Tashkent Medical Academy, Neurology, Tashkent, Uzbekistan

Background/Objectives: Epileptic encephalopathy with continuous spike and wave during sleep (CSWS) is a syndrome marked by progressive cognitive decline, epileptic seizures, specific EEG patterns, and a complex/polygenic inheritance. Given the elevated incidence of drug resistance, the development of more effective and targeted treatment approaches for patients is relevant.

Methods: In our study, we intentionally selected 88 patients diagnosed with epileptic encephalopathy with continuous spike and wave during sleep. All participants were tested with: neuropsychological, clinical, neurophysiological, neuroimaging, and genetic assessments, including whole exome sequencing (WES). Among these individuals, we identified one with a GRIN2A mutation showing a gain of function variant (GoF), for further targeted treatment with memantine with daily dose 0.5 mg/kg per day."

Results: At a memantine dosage of 0.5 mg/kg, with no adjustments to antiepileptic drug doses, we observed a notable 65% reduction in average seizure frequency. This decline manifested within several weeks of reaching the full memantine dose and persisted throughout the 3-month follow-up period. We also observed an improvement in interictal EEG recordings, with no epileptiform discharges during both wakefulness and sleep, contrasting with the EEG prior to memantine initiation. While there were slight changes in cognitive skills, overall improvement was noted.

Conclusion: This clinical case exemplifies the significant potential of targeted treatment for epilepsy. Further clinical research and continuous follow-up are necessary to gather additional insights and information.

Grants:

Conflict of Interest: None declared

EP11.045 Role of circulating miRNAs expression in different biofluids in patients with intracranial aneurysms

sara khadem ansari1, Ebru Erzurumluoglu Gokalp1, emre ozkara2, Ozlem Aykac3, Atilla Ozcan Ozdemir3, Sevilhan Artan 1

1Eskisehir Osmangazi University, Faculty of Medicine, Medical Genetics, Eskisehir, Türkyie; 2Eskisehir Osmangazi University, Faculty of Medicine, Neurosurgery, Eskisehir, Türkyie; 3Eskisehir Osmangazi University, Faculty of Medicine, Neurology, Eskisehir, Türkyie

Background/Objectives: Intracranial aneurysm (IA) is a cerebrovascular disease with a significant morbidity and mortality rate. Therefore, it is urgent to identify diagnostic and prognostic biomarkers of IA and its subsequent rupture. In this study, we aimed to analyze the miRNA expression profiles to identify potential biomarkers of SAH and evaluate the usability of blood for a non-invasive approach.

Methods: The expression levels of eight miRNAs has been investigated in blood and CSF samples obtained on day five after SAH from 24 patients (CSF and blood specimens from 12 ruptured IA patients, blood specimens from 12 unruptured IA patients) and 14 controls using qRT-PCR.

Results: miR-29a, miR-200a-3p, miR-451a, miR-1297, and miR-502-5p in blood and miR-29a, miR-200a-3p, miR-451a, miR-126, miR-146a-5p and miR-27b-3p in CSF were found to be differentially expressed in ruptured IA patients compared to controls. miR-29a, miR-200a-3p, and miR-451a were significantly altered in both biofluids (AUCs>0,8). miR-126, miR-200a-3p, miR-451a, and miR-502-5p showed a higher expression in the ruptured IA group compared to unruptured IA. We also found that lower blood miR-451a levels (AUC = 0,960) were associated with an increased risk for poor clinical outcomes post-SAH.

Conclusion: miR-29a, miR-200a-3p, and miR-451 could be candidate non-invasive biomarkers for SAH. Also, miR-126, miR-200a-3p, miR-451a, and miR-502-5p might influence the risk of aneurysmal rupture, and highly expressed miR-451a might be considered as a potential biomarker in the diagnosis, severity, and prognosis of SAH.

Grants: This work was supported by the Scientific Research Projects Fund of Eskisehir Osmangazi University [TCD-2021-1737, TCD-2021-1710].

Conflict of Interest: None declared

EP11.046 Exploring Molecular Insights into Cortical Brain Malformations or Microcephaly in Turkey Through Whole-Exome Sequencing

Buşra Kasap 1, Cengiz Yalçınkaya2, Dilek Uludağ Alkaya1, Filiz Geyik3, Nilay Güneş1, Sema Saltık4, Ahmet Okay Çağlayan5, Beyhan Tüysüz1

1Istanbul University-Cerrahpaşa, Cerrahpaşa Medical Faculty, Department of Pediatric Genetics, Istanbul, Türkyie; 2Istanbul University-Cerrahpaşa, Cerrahpaşa Medical Faculty, Department of Neurology, Istanbul, Türkyie; 3Istanbul University, Institute of Graduate Studies in Health Sciences, Department of Genetics, Istanbul, Türkyie; 4Istanbul University-Cerrahpaşa, Cerrahpaşa Medical Faculty, Department of Pediatric Neurology, Istanbul, Türkyie; 5Dokuz Eylül University, Institute of Health Sciences, Department of Molecular Medicine, Izmir, Türkyie

Background/Objectives: Microcephaly and/or cortical malformations encompass a spectrum of neurogenetic disorders marked by intricate diagnostic complexities arising from the genetic and clinical diversity within this group of conditions. The aim of this study was to identify the molecular etiology in patients with microcephaly or cortical malformations.

Methods: Whole exome sequencing was performed on 85 patients with cortical dysplasia or microcephaly, following the exclusion of recognized neurological and metabolic disorders. The patients divided into three groups: cortical malformation (n = 28), microcephaly (n = 27), and microcephaly and cortical malformation (n = 30).

Results: In the entire cohort, the rate of molecular diagnosis was 54.1%. Within the cortical malformation subgroup, the diagnostic rate was 50%, with biallelic variants identified in LAMC3, EMC1,PCLO, WRN, GPR56 and ARV1 while monoallelic variants were identified in LIS1, WDFY3, PIK3R2 and EBP. In microcephaly subgroup, the diagnostic rate was 44.4% with biallelic variants identified in UNC80, ASPM, LIG4, PCNT, CEP135, MTMR1, WWOX and TRMT10A while hemizygous variants were identified in KDM5C, PQBP1 and RS1. In microcephaly and cortical malformation subgroup, the diagnostic rate was 66.7% with biallelic variants identified in PCNT, MCPH1, STIL, PNKP, ERCC6, CEP152, WDR62,CEP135, WDR62, SLC1A4, ASNS, OXR1, MOCS2, MFSD2A, KIF7,OCLN, AP4M1 and RFWD3, while hemizygous variants were identified in CASK and RLIM. Novel candidate genes, GIT1, ING1,ASTN1 and ZNF385B were identified in four families. No mutation was detected in 41.2% of the patients.

Conclusion: Our study provides valuable insights into the underlying molecular mechanisms of cortical brain malformations and microcephaly.

Grants: This study was funded by Scientific-Research-Projects-Coordination-Unit of Istanbul University-Cerrahpaşa. Project number: TOA-2019-33466.

Conflict of Interest: None declared

EP11.047 Whole exome sequencing results in autism spectrum disorders patients living in the Trakya Region of Türkiye: Trakya University experience

Engin Atli 1, Sinem YALÇINTEPE1, Veysel Oz2, Drenushe Zhuri1, Yasemin Ozen1, Emine Ikbal Atli1, Selma Demir1, Işık Görker3, HAKAN GURKAN1

1Türkiye, Medical Genetics, Edirne, Türkyie; 2Türkiye, Child Neurology, Edirne, Türkyie; 3Türkiye, Child and adolescent psychiatry, Edirne, Türkyie

Background/Objectives: Autism spectrum disorders (ASD) is a neurodevelopmental disorder characterized by three core symptoms with impaired reciprocal social interaction and communication, a pattern of repetitive behavior and/or restricted interests in early childhood. Despite studies suggesting that autism spectrum disorders (ASDs) are highly heritable, the basis for this heritability remains largely unexplained.

Methods: 50 patients who applied to our clinic between August 2021 and June 2023 were included in the study. 10 of the patients were referred with a diagnosis of atypical autism, 20 with clinical findings accompanying ASD, and 20 with a diagnosis of ASD only. 16 of the patients were woman and 34 were men. Age ranges were between 4 and 24. The patients karyotype analysis, aCGH and fragile X analysis results were found to be normal. Whole exome sequencing was performed on the Illumina NovaSeq 6000 instrument with the QIAseq Human Exome Kit.

Results: Previously reported Pathogenic/Likely pathogenic variations were detected in a total of 9 patients. While no clinically relevant pathogenic variation was detected in 8 patients, unknown significance (VUS) variations was detected in a total of 32 patients. Among the Likely pathogenic/Pathogenic variations, 4 were evaluated as de novo according to the results of the segregation analysis.

Conclusion: The pathogenic variation detection rate (18%) in all patients was consistent with the literature, but was slightly higher. Additionally, our findings support the suggestion that WES analysis can be used as an effective primary diagnostic method in clinical practice in ASD patients.

Grants: No

Conflict of Interest: None declared

EP11.048 Identification of a two novel SCN1A gene mutations in a patient with therapy-resistant epilepsy

Krisztina Galimurka 1, Renata Szalai1, Agnes Till1, Judit Bene1, Attila Gyenesei2, Kinga Hadzsiev1

1University of Pecs, Medical School, Department of Medical Genetics, Pecs, Hungary; 2University of Pecs, Szentágothai Research Centre, Pecs, Hungary

Background/Objectives: Voltage-gated sodium channel (VGSC) plays an important role in neuronal excitability. SCN1A gene encodes the type 1 alpha subunit of neuronal VGSC. SCN1A gene mutations are the most common cause of genetic epilepsy with febrile seizures plus (GEFS + ) and Dravet syndrome (DS). DS belongs to group of severe drug resistant epilepsies with frequent seizures, that usually begin in the first year of life.

Methods: Here we report a 21 months old Hungarian patient with focal and generalized epilepsy. Sequencing analysis was performed using Illumina DNA Preparation With Enrichment Kit and Illumina NovaSeq 6000 sequencing technology. Sanger sequencing confirmed the presence of the pathogenic variants.

Results: NGS analysis of epilepsy gene panel containing 473 genes identified two novel missense c.426T>G (p.Cys142Trp) and c.5012 T > G (p. Phe1671Cys) variants in SCNA1 gene (NM_006920) in a heterozygous state. The classification of the detected variants are likely pathogenic based on the American College of Medical Genetics and Genomics (ACMG) guideline.

Conclusion: The molecular genetic analysis is an effective diagnostic tool to identify the cause of epilepsy. In turn, an accurate diagnosis is highly important for selection of the most effective therapy especially in early childhood to prevent future complications. The SCN1A missense mutations can be associated with both mild and severe clinical presentations and typical clustering of symptoms become apparent only during follow-up which make the diagnosis challenging. In our case, two novel missense mutations together can affect the patient phenotype more severely and be the cause of DS.

Grants:

Conflict of Interest: None declared

EP11.049 Bi-allelic pathogenic variants in SNX14 gene in two adult brothers affected by intellectual disability and movement disorder.

Nino Spataro 1, Núria Capdevila1, Carmen Manso1, Viviana P Beltran-Salazar2, Victor Martinez-Glez1, Anna Ruiz1

1Parc Taulí Hospital Universitari, Institut d’Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Center for Genomic Medicine, Sabadell; 2Consorci Corporació Sanitària Parc Taulí, UDIAT Diagnostic Center, Sabadell

Bi-allelic pathogenic variants in SNX14 gene in two adult brothers affected by intellectual disability and movement disorder.

Background/Objectives: Biallelic pathogenic variants in the SNX14 gene cause intellectual disability-coarse face-macrocephaly-cerebellar hypotrophy syndrome, also known as autosomal recessive spinocerebellar ataxia 20 (SCAR20). SNX14 loss-of-function variants account for approximately 9.9% of early-onset cerebellar atrophy and ataxia cases.

We aim to describe the clinical features and trajectories of two adult patients harboring bi-allelic pathogenic variants in SNX14 gene.

Methods: aCGH, 5’ UTR CGG repeats expansion of FMR1 and whole exome sequencing (WES) analysis were performed in two brothers (currently aged 48 and 52 years old) affected by intellectual disability, ataxia and sensorineural deafness (only 1 patient) attended by our unit since childhood.

Results: Through WES, two compound heterozygous variants in SNX14 gene were identified in both affected brothers and not observed in unaffected siblings. According to ACMG/AMP criteria, the frameshift c.1883delC variant was classified as likely pathogenic. RNA analysis of the intronic variant c.1811-8A > G shows an alteration in the splicing acceptor site causeing an insertion of 7 nucleotides, altering the reading frame and generating a premature stop codon.

Conclusion: To our knowledge, most of the reported cases are in paediatric age and very few adults patients have been reported. To date, no patients aged more than 40 years old and affected by SCAR20 syndrome have been reported. The clinical characterization of our patients could shed light on the prognosis and the clinical trajectories of patients affected by SCAR20 syndrome.

Conflict of Interest: None declared

EP11.051 Clinical and molecular characterization of 28 Tunisian patients with Rett syndrome

Wissal Tergui 1, Ahlem Achour1;2, Nesrine Kerkeni1;2, Amira Zanati1, Wiem Essalah1, Zouhour Miladi3, Ichraf Kraoua3, Ilhem Turki3, ridha mrad1;2, Mediha Trabelsi1;2

1Charles Nicolle Hospital, Department of Congenital and Hereditary Diseases, Tunis, Tunisia; 2Faculty of Medicine of Tunis, University of Tunis El Manar, Laboratory of Human Genetics (LR99ES10), Tunis, Tunisia; 3National Institute Mongi Ben Hmida of Neurology, Department of pediatric neurology, Tunis, Tunisia

Background/Objectives: Rett syndrome (RTT, OMIM #312750) is a severe neurodevelopmental disorder primarily affecting females, with an incidence ranging from 1:10000 to 1:15000. It’s characterized by developmental regression and stereotypic hand movements. More than 95% of classical and approximately 75% of atypical RTT cases arise from a pathogenic variant in MECP2 gene.

Methods: We carried out a retrospective descriptive study on a cohort of Tunisian female patients diagnosed with RTT by Sanger sequencing of MECP2 gene and followed up at the Department of Congenital and Hereditary Diseases of Charles Nicolle Hospital over a period of 20 years from June 2003 to December 2023.

Results: Our patients presented classical (17/28) and atypical (11/28) RTT according to the 2010 diagnostic criteria. The mean age at first examination was 4 years old. They were mainly referred for developmental delay/regression (19/28), autistic features (13/28) and stereotypic hand movements (8/28).

Molecular analysis revealed 15 different MECP2 variants (six missense, five nonsense and four frameshift), all of them, except the p.(Pro384Hisfs*25), have been previously described. 87% of variants were located in exon 4 (13/15). The most common variants, detected in 12/28 patients, were p.(Arg133Cys), p.(Arg255Ter) and p.(Arg168Ter).

Conclusion: This study emphasizes the clinical and genetic heterogeneity of the RTT already described in literature. Despite the development of NGS technologies, Sanger sequencing of MECP2 gene remain a reliable method to confirm the diagnosis of RTT and provide an appropriate genetic counselling for the patients’ families.

Grants:

Conflict of Interest: None declared

EP11.052 Complex neurological phenotype in female carrier of TLR7 frameshift deletion

Maya Atanasoska 1;2, Lubomir Balabanski1, Slavyana Yaneva Staykova1, Irena Bradinova1;3, Draga Toncheva1;4, Radoslava Vazharova1;5

1CellGenetics Laboratory, Genomic laboratory, Sofia, Bulgaria; 2Sofia University “St Kliment Ohridski”, Faculty of Biology, Department of Genetics, Sofia, Bulgaria; 3National Genetic Laboratory, UHOG “Maichin dom”, Sofia, Bulgaria; 4Bulgarian Academy of Science, Sofia, Bulgaria; 5Sofia University “St Kliment Ohridski”, Faculty of Medicine, Department of Biology, Medical genetics and Microbiology, Sofia, Bulgaria

Background/Objectives: The toll-like receptors (TLRs) have a key role in activation of innate and adaptive immunity. TLR7 is an endosome receptor, located in plasmacytoid dendritic cells and B cells. The receptor controls viral immune response, through recognition of virus-associated ssRNA and dsDNA containing guanosine and uridine-rich sequences.

Methods: We examined a 35-year-old female with suspected multiple sclerosis and a clinical feature with non-progressive neurological symptoms manifesting in paralysis of the lower limbs. Her symptoms started after vaccination for hepatitis B. Whole genome sequencing on a BGISEQ-500 platform, was performed followed by segregation analysis through Nanopore sequencing on MiniOn platform.

Results: Novel frameshift deletion TLR7:c.2441del, was identified. The variant was inherited from our patient’s father, who developed severe pneumonia due to the SARS-CoV-2 coronavirus, our patient had mild COVID-19 symptoms. The variant is not present in the gnomAD database. So far, no functional analysis was performed.

Conclusion: The association of TLR7 in systemic lupus development, was finally confirmed in 2022 when Brown et al. identified a gain-of-function variant in the gene and associated it with neurolupus in humans. It opened a whole new era of genetic mechanism of this disorder and possibility for targeted therapy. Our patient immunological tests did not confirm systemic lupus. We assume that TLR7:c.2441del leads to abnormal interferon production, which is protective for viral infections, but may cause complex neurological phenotype.

Grants: BNSF, KP-06-N43/8.

Conflict of Interest: None declared

EP11.053 A novel intragenic deletion in NRXN3 in a boy with autistic trait disorder

Neus Baena 1, Montserrat Garcia-Puig2, Anna Brunet-Vega1, Laura Capel1, Carmen Manso1, Juan Pablo Trujillo Quintero1, Núria Capdevila1, Victor Martinez-Glez1

1Corporació Sanitaria Parc Tauli, Centre de Medina Genòmica, Sabadell, Spain; 2Corporació Sanitaria Parc Tauli, Pediatria, Sabadell, Spain

Background/Objectives: The human neurexin family consists of three unrelated genes (NRXN1, NRXN2 and NRXN3), they are highly expressed in presynaptic nerve terminals and have important roles in synaptic cell adhesion and neurotransmitter release. Heterozygous deletions of chromosomal region 14q24.3-31.1 involving NRXN3 have been associated with various neuropsychiatric conditions and autistic features with/without developmental delay/intellectual disability.

Methods: An 180k aCGH (Oxford Gene Technology, OGT) was used as a first tier test for neurodevelopmental disorders. Customized MLPA analyses enable to validate and to determine the inheritance of pathogenic copy number variants.

Results: The patient is a boy two-years-old with autism (ASD), no language, he presents a stereotyped behaviour consisted in hand movements, flapping movements and turn around on himself, abnormal sleep and behavioral problems.

A novel intragenic deletion from exon 9 to 12 in NRXN3 gene was identified. The deletion was classified as likely pathogenic following ACMG classification. To our knowledge, this is the first patient described in the literature with this deletion.

Conclusion: Loss of function variants in the NRXN3 gene have been shown to cause ASD. We described a boy with ASD and abnormal behaviour caused by a novel deletion in the NRXN3 gene. Genotype-phenotype correlation will be presented and segregation analysis.

Increasing aCGH resolution to single exons led to detection of small CNVs in known disease genes. Our findings highlight the importance of high-resolution aCGH and the critical analysis of aCGH data surrounding genes that are involved by genomic variation.

.

Conflict of Interest: None declared

EP11.054 Exploring the genetic background of hereditary ataxias in Hungary with special emphasis on repeat expansion-related ataxias

Lilla Buzai-Kiss 1;2, Barbara Trombitas1, Péter Balicza1;2, Judit C. Sági1, szabolcs udvari1, Maria Judit Molnar1;2

1Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Hungary; 2HUN-REN Multiomics Neurodegeneration Research Group

Background: Repeat expansions are linked to many ataxias, including Friedreich ataxia (FRDA), spinocerebellar ataxias (SCA), and Cerebellar Ataxia with Neuropathy and Vestibular Areflexia Syndrome (CANVAS). The exact percentage and phenotypic spectrum of ataxias associated with repeat expansion are currently unavailable in Hungary.

Patients and Methods: Our NEPSYBANK holds records for 1014 ataxia patients: 840 for SCA, 244 for FRDA, and 78 for CANVAS. Zygosity and repeat sizes were determined using TP-PCR, fragment analysis, and Southern blot. Additionally, 95 cases underwent targeted ataxia NGS panel or whole-exome sequencing.

Results: In our cohort, polyQ ataxias were most common (N = 110): SCA1 (N = 40), SCA2 (N = 21), SCA3 (N = 6), SCA6 (N = 2), SCA7 (N = 1), FRDA (N = 10). CANVAS affected 8 families. In the FRDA group, 5 patients had heterozygous pathogenic GAA expansions. One case had a rare damaging variant on one allele, and in four cases, the other variant was not found, yet patients showed the typical FRDA phenotype. In the CANVAS cohort, one pathogenic allele was detected in 4 samples, both in 2 samples, and uncertain expansion in one allele in 1 samples. NGS sequencing found rare deleterious variants in 12% of tested patients, with POLG1, SPG7, SETX, ATM, CYP27A, SACS, and mtDNA most frequently affected.

Conclusion: Hereditary ataxias exhibit diverse clinical features, demanding a blend of clinical assessments and molecular genetic testing for precise identification. PolyQ ataxias dominate our cohort, emphasizing the need for thorough genetic tests, including NGS sequencing. CANVAS is a common cause of adult-onset ataxias, but its molecular diagnosis is intricate and often requires cascade testing.

Grants: TKP2021-NVA-15, TKP2021-EGA-25,OTKA139330

Conflict of Interest: None declared

EP11.055 Phenotypic diversity of WFS1-related diseases

Eva Pinti 1, Anna Lengyel1, Agnes Sallai1, Andras Seres2, Gyorgy Fekete1, Iren Haltrich1, Kálmán Tory3

1Gyermekgyógyászati Klinika - Tűzoltó utcai Részleg, Semmelweis Egyetem, Budapest, Hungary; 2Budapest Retina Associates, Budapest, Hungary; 3Pediatrics Clinic no. I. of Semmelweis University, Budapest, Hungary

Background: WFS1 is a dosage sensitive gene, which plays a role in cellular apoptosis, therefore its loss of function variations cause progressive neurodegenerative syndromes with different inheritance pattern. In relation to this disease spektrum we examined three patients with initially different, then more and more overlapping phenotypes. The symptoms of a 47-year-old female in sequence of appearence were congenital bilateral sensorineural hearing loss, primary hypogonadism (11 years), insulin-dependent diabetes (23 years) and visual impairment (in her 30s). In her history she had negative GJB2 sequencing, conventional karyotyping and FMR1 trinucleotide repeat analysis. The other two patients were siblings. The 16-year-old girl had rapidly progressive vision loss in early childhood, which was followed by insulin-dependent diabetes. Her 11-year-old brother was diagnosed with insulin-dependent diabetes at the age of 6 years, vision loss and central diabetes insipidus appeared later.

Methods: According to the various features of the adult patient we indicated whole exom sequencing. The more typical phenotype of the siblings allowed for tartgeted testing: the older child had a Wolfram-syndrome panel, followed by the variation specific Sanger-sequencing of her brother.

Results: The patients were diagnosed with WFS1 pathogenic variants. The adult female had a heterozygous alteration causing autosomal dominant Wolfram-like syndrome, and the siblings had compound heterozygosity causing autosomal recessive Wolfram-1 syndrome.

Conclusion: These cases were examples for the high-diversity of WFS1 related phenotypes, as the disease courses of the siblings were different, and as the heterozygous female patient had primary hypogonadism, which is more characteristic of the recessive form.

Grants: No.

Conflict of Interest: None declared

EP11.056 Association of TIRAP gene polymorphisms with neuroinflammatory diseases

ASLI KARACAN 1;2, Ayça Dilruba aslanger1, Guven Toksoy1, Zehra Oya Uyguner1, Ayse Evrim Bayrak1

1Istanbul University, Istanbul Faculty of Medical, Department of Medical Genetics, Istanbul, Turkey; 2Istanbul University, Institute of Graduate Studies in Health Sciences, Istanbul, Turkey

Background/Objectives: The Toll-like Receptor (TLR) signaling pathway plays a crucial role in various brain disorders like epilepsy, autism spectrum disorder (ASD), and neurometabolic diseases. Polymorphic variants in this gene play a role in the susceptibility to inflammatory diseases. This study aims to investigate the association between single nucleotide variations (SNVs) of TIRAP and neuroinflammatory diseases in patients receiving a genetic diagnosis at our clinic.

Methods: SNVs in TIRAP were analyzed in-house whole exome sequencing (WES) data of 200 individuals (mean age 10.93 years; 105 males and 95 females) including epilepsy-ASD (n = 40), neurometabolic disorders (n = 24). Allele frequencies of SNVs were determined and associations with neuroinflammation were evaluated using SPSS 16.0, p < 0.05 considered significant. Haplotype association studies are ongoing.

Results: Among 25 identified SNPs, we calculated allele frequencies of 11 most frequent SNPs (MAF > 0.05) in exonic, intronic and 3’-UTR regions of TIRAP according to WES data and statistically evaluated allele and genotype frequencies between subjects. The variant rs611953 (c.*109G > A) was found to be statistically significant, with a MAF = 0.35 in the control group and MAF = 0.50 in the neurometabolic diseases (n = 24). For the rs609634 (c.646+174C > T) variant, MAF = 0.304 in the control group and MAF = 0.425 in epilepsy-ASD (n = 40).

Conclusion: This study highlights the potential role of TIRAP SNVs in neuroinflammation-related diseases, suggesting their impact across different developmental stages. However, further research is needed to understand how these variations functionally contribute to such diseases.

Grants: This study was supported by IUSRP /TDK-2023-39951) and TUSEB (2022-A9-28202.

Conflict of Interest: None declared

EP11.057 Aggregation of variants in genes associated with late onset Alzheimer’s disease polygenic pathways detected in Unspecified Dementia Bulgarian patients

Dimitar Serbezov 1, Sena Karachanak-Yankova1;2, Dragomira Nikolova1, Marta Mihaylova1, Savina Hadjidekova1, Diana Belejanska3, Shima Mehrabian3, Mariya Petrova3, Latchezar Traykov3;4, Draga Toncheva1;4

1Medical University-Sofia, Medical genetics, SOFIA, Bulgaria; 2Faculty of Biology, Department of Genetics, Sofia, Bulgaria; 3Medical University-Sofia, Department of Neurology, Sofia, Bulgaria; 4Bulgarian Academy of Sciences, Sofia, Bulgaria

Background/Objectives: Genetic studies have given insights into the etiology of early onset neurodegenerative disorders, such as early onset Alzheimer’s disease (AD) and frontotemporal dementia (FTD), but atypical dementia cases exist, designated under the umbrella term Unspecified Dementia (UD), that do not fit these patterns and their causes remain elusive.

Methods: Whole-genome sequencing was performed on a DNA pool, set up with DNA isolated from 50 Unspecified Dementia (UD) Bulgarian patients. The presence of variants in genes associated with late onset AD was investigated.

Results: Our analysis establishes aggregation of variants associated with late onset AD in genes constituent of established polygenic AD pathways. These are the Sortilin Related Receptor 1gene (SORL1), the Catenin alpha-3 gene (CTNNA3), the Complement receptor 1 gene (CR1), the GRB2-associated-binding protein 2 gene (GAB2) and the Insulin Degrading Enzyme gene (IDE).

Conclusion: In addition to its well established cause for the development of AD, our results indicate that the rs429358 variant of the APOE gene might also play role in the etiology of UD in Bulgarian patients. These findings should be further studied in individual DNA samples of patients with unspecified dementia.

Grants: KP-06-N33/5 from 13.12.2019 - National Science Fund of Bulgaria"

Conflict of Interest: None declared

EP11.059 Co-occurance of rare variants in lysosomal storage disease associated genes and other rare neurodegenerative disase associated genes

Tamás Szlepák 1;2, Bernadette Kálmán3, Róbert Sepp4, Judit Mária Molnár1;2

1Institute of Genomic Medicine and Rare Disorders, Semmelweis University; 2HUN-REN, Multiomic Neurodegeneration Research Group; 3Department of Laboratory Medicine, University of Pécs; 4Second Department of Internal Medicine and Cardiology Center, University of Szeged

Background: Lysosomal storage diseases (LSDs) are monogenic disorders with AR inheritence, but many monoallelic variants of these genes can have clinical relevance in other phenotypes and diseases, such as GBA1 or SMPD1 heterozygous rare variants (RVs) in Parkinson disease, PSAP gene causing Gaucher-like phenotype, or other biochemical pathways of LSD associated genes. Due to widespread usage of the high-throughput genomic testings, more and more RVs are identified with a presumably significant role or modifying effect in complex phenotypes.

Methods: In the registries of our Institute, we did the systematic analysis of 30 GBA-associated PD, 9 Gaucher, 72 Fabry, 24 Pompe and 7 ASMD patients’ genetic architecture to identify patients harbouring multiple heterozygous damaging RVs. Thirty percent of the cases had WES.

Results: We found coexisting RVs in 7 cases. Four GBA-associated PD patients carried SMPD1, SPG11, SNCA or a second GBA1 rare variants. We identified a male patient with mild Gaucher phenotype carrying GAA pesudodeficiency allele beside the GBA1 RV. We also found two patients with Fabry-like symptoms carrying the controversial D313Y GLA variant and a second pathogenic variants either in MYBPC3 or SPG7 genes. In one child GBA1 and NPC1 heterozgyous pathogenic variants were coexisting.

Conclusion: In the era of expansive NGS testings, the identification and evaluation of clinical effect of co-existing pathogenic variants are not just a major challange, but an important factor to understand the patomechanism of diseases and to offer more suitable genetic counseling and treatments.

Grants: TKP2021-EGA.25, TKP2021-NVA-15, OTKA 139010

Conflict of Interest: None declared

EP11.060 Investigation of depression and fatigue levels of multiple sclerosis patients

Hülya Şenol1, Tahmina Chalabi Kurt1, Serdal Işıktaş1, Şükrü Tüzmen 2;3

1Cyprus Health and Social Sciences University, Health and Social Sciences, Morphou, Mersin 10 via Türkiye, Cyprus; 2Eastern Mediterranean University (EMU), Faculty of Dentistry, Famagusta, Mersin 10 via Türkiye, Cyprus; 3GenBiomics R&D, Techno Park, Eastern Mediterranean University (EMU), GenBiomics R&D, Famagusta, Mersin 10 via Türkiye, Cyprus

Background/Objectives: Multiple sclerosis (MS) is an autoimmune disease that affects central nervous system cells. Although the exact cause of MS is unknown, factors such as genetic factors, environmental factors and immune system disorders are thought to play a role. The main purpose of this study is to examine depression and fatigue in MS patients and to investigate the effects of these conditions on the patients’ quality of life.

Methods: A total of 226 MS patients’ survey was prepared. The first part of the survey includes questions about the sociodemographic information, the second part includes Beck Depression Inventory (BDI) items, and the third part includes Chalder Fatigue Scale (CFA) items. Data were analyzed using SPSS 28 program. Data were collected from MS family groups from Azerbaijan, MS and psychology groups from Turkey and Northern Cyprus.

Results: MS errors increase as the level of depression increases, as does the level of fatigue. Regression analysis results show that depression has a significant effect on fatigue and that depression explains a large part of the variation in fatigue levels. According to the results of the analysis, Cronbach’s Alpha value for the Beck Depression Scale was 0.86; Cronbach’s Alpha value for the Chalder Fatigue Scale was determined as 0.930.

Conclusion: To the best of our knowledge, this study is the first to examine depression and fatigue in MS patients in Azerbaijan, Türkiye and North Cyprus. The reliability analysis results show that both scales have high internal consistency and that these scales can be used reliably.

Conflict of Interest: None declared

EP11.061 Detection of frontotemporal dementia - associated variants in a pooled sample of Bulgarian patients with unspecified dementia by whole genome sequencing

Dragomira Nikolova 1, Sena Karachanak-Yankova1;2, Dimitar Serbezov1, Mikaela Stancheva2, Desislava Nesheva1, Marta Mihaylova2, Diana Belejanska3, Mariya Petrova3, Shima Mehrabian3, Latchezar Traykov3, Savina Hadjidekova1, Draga Toncheva4

1Medical University - Sofia, Medical Faculty, Department of medical genetics, Sofia, Bulgaria; 2Sofia University “St.Kl.Ohridski”, Department of genetics, Sofia, Bulgaria; 3University Hospital “Alexandrovska”, Department of Neurology, Sofia, Bulgaria; 4Bulgarian Academy of Sciences, Sofia, Bulgaria

Background: Dementia is a broad category of brain diseases that cause a long term decrease in the ability to think and remember. It is a serious social problem as it affects person’s daily functioning. Its pathogenesis comprises malfunctioning of genes involved in the development of polygenic diseases as Alzheimer’s (AD) and frontotemporal dementia (FTD). This study aims to unveil common genes and variants between FTD and unspecified dementia (UNSPD).

Methods: Equimolar amounts of DNA samples of 50 Bulgarian patients with UNSPD have been pooled and whole genome sequenced with 100x coverage. Our UNSPD SNP data were screened for 215 FTD-associated variants taken from DisGeNet database.

Results: Twenty-seven variants associated with FTD were discovered in genomic data of patients with UNSPD. Twenty-one variants were intergenic, intronic or synonymous. Of the rest, three variants were located either in the upstream regulatory gene region (rs2255166 in TEPSIN, rs6111609 in RRBP1, rs111463574 in NDUFA12) or 3’-UTR region (rs1048775 in TEPSIN); one variant was missense (KIF24 rs17350674) and one was downstream (TMEM106B rs7791726). KIF24 rs17350674 and FUS rs1052352 create a stop gain site and were reported to be associated with frontotemporal lobar degeneration (FTLD). Altogether five SNP variants were reported in ClinVar as benign, twenty-two were not reported.

Conclusion: In order to clarify the etiology of UNSPD, we report twenty-seven variants in 17 genes which are associated with FTD and FTLD. We suggest they play a role in the development of UNSPD as well which identifies common pathways between those two related conditions.

KP-06-N33/5 from 13.12.2019 - NSFB

Conflict of Interest: None declared

EP11.065 A founder variant in ZNF335 causes a novel phenotype: Autosomal recessive distal motor neuropathy

Mohammad Al Muhaizea1, Ali Alshehri1, Hanan AlQudairy2, Sarah Alruways2, Nouf Almutairi2, Rawan Almas3, Albandary Al-Bakheet2, Aljouhra AlHargan2, Anoud Albader2, Ghadeer Alhamid2, Dilek Colak4, NAMIK KAYA 2

1King Faisal Specialist Hospital and Research Center (KFSHRC), Neuroscience Center, Riyadh, Saudi Arabia; 2King Faisal Specialist Hospital and Research Center (KFSHRC), Translational Genomics Department, Riyadh, Saudi Arabia; 3King Faisal Specialist Hospital and Research Center (KFSHRC), Medical Genomics Department, Riyadh, Saudi Arabia; 4King Faisal Specialist Hospital and Research Center (KFSHRC), Department of Molecular Oncology, Riyadh, Saudi Arabia

Introduction: ZNF335 plays a crucial role in methylating and regulating neuronal gene expression and harbors mutations linked to several neurological phenotypes, mainly microcephaly (OMIM:615095).

Methods: We ascertained six patients from three Saudi families. After informed consents were obtained from the patients and parents, peripheral blood were collected from the participants. Whole Exome Sequencing (WES) was done using the index patient’s DNA extracted from the blood sample. ZNF335’s expression was measured on total RNA extracted from the index patient’s fibroblasts. The quantification was based on delta delta CT method. Immunoblotting experiments were done according to standard procedures using enhanced chemiluminescence substrate (Thermo Scientific) and visualized via Gel Doc XR system (Bio‐Rad Laboratories).

Results: We have identified a novel variant (NM_022095: exon14:c.1897A>G; p.K633E) using WES, Sanger sequence verification in six patients from three Saudi families. The variant has been found to cause autosomal recessive distal motor neuropathy, deterioration to motor difficulties, gait abnormalities, and speech problems. We comprehensively investigated the clinical characteristics of the patients and compared their phenotypes to the previously reported patients. We observed disturbed expression patterns of ZNF335 in the index patient’s cells.

Conclusion: This study expands the phenotypic and genotypic spectrum of pathogenic ZNF355 variants. Our study provides the first evidence linking the genetic defects of the gene to a novel phenotype of distal motor neuropathy. We suggest that those patients present with muscle weakness, motor difficulties, steppage gait, and areflexia should be screened for ZNF335 mutations for differential diagnoses.

Grants: KSCDR/RAC#2180004

Conflict of Interest: None declared

EP11.066 A further case of heterozygous de novo nonsense variant in RNF13 gene

Andrea Pietra 1;2, Mina Grippa2, Elisabetta Spezia3, Patrizia Bergonzini3, Alberta Patanè4, Alberta Leon4, Lorenzo Iughetti3, Olga Calabrese2

1IRCCS Azienda Ospedaliero-Universitaria di Bologna, Medical Genetics Unit, Bologna, Italy; 2AOU Modena, SSD Genetica Medica, Modena, Italy; 3AOU Modena, UO Pediatria, Modena, Italy; 4Research&Innovation S.R.L., R&I Genetics, Padova, Italy

Background/Objectives: RNF13 variants are involved in a complex neurodevelopmental disorder hitherto only rarely reported.

Methods: We performed trio WES analysis in our patient after a negative epilepsy NGS panel.

Results: Our patient is a 15 years-old male, born at term, with SGA and microcephaly, after a pregnancy complicated by oligohydramnios. At birth axial hypotonia and hypertonia of arms and legs bilateral club feet, adducted thumbs, dysmorphisms, cryptorchidism were noted. An early infantile epileptic encephalopathy with suppression burst pattern become evident and brain MRI showed myelination delay, abnormal cortical gyration, vermian hypoplasia, enlargement of lateral ventricle, thin corpus callosum. A spastic-dystonic tetraparesis with microcephaly and cranio-facial dysmorphisms was diagnosed and from 10y of age an aortic root enlargement was observed.

The trio whole-exome sequencing observed a heterozygous de novo nonsense variant in RNF13 gene, located between residues 293 and 311. The laboratory performing the analysis classified the variant as VoUS. Nevertheless, due to bioinformatic tools, variant characteristics and the significant phenotypic overlap between our patient and the individuals reported in the literature, the variant is considered diagnostic.

Conclusion: To our knowledge, our patient is the seventh described so far. The nonsense variants observed in our patient, as well as others similarly affected patients, are expected to escape nonsense-mediated decay, leading to a truncated protein which lacks the putative regulatory region between amino acids 309 and 319. The current evidence suggests that the 292–312 region might have a negative regulatory (suppressive) on downstream targets such that its removal or modification could lead to a gain-of-function effect.

Grants:

Conflict of Interest: None declared

EP11.067 Screening for genes with disease risk variants in early-onset Alzheimer’s disease patients from pooled whole genome sequencing data

Sena Karachanak-Yankova 1;2, Dimitar Serbezov1, Dragomira Nikolova1, Desislava Nesheva1, Marta Mihaylova1, Mikaela Stancheva2, Dimitrina Georgieva2, Slavica Josifovska3, Diana Belejanska4, Shima Mehrabian4, Mariya Petrova4, Latchezar Traykov4, Savina Hadjidekova1, Draga Toncheva5

1Medical University-Sofia, Department of Medical Genetics, Sofia, Bulgaria; 2Sofia University “St. Kliment Ohridski”, Department of Genetics, Sofia, Bulgaria; 3Faculty of Natural Sciences and Mathematics, “Ss.Cyril and Methodius” University, Laboratory of Molecular Biology, Skopje, Macedonia; 4Medical University-Sofia, Depatment of Neurology, UH “Alexandrovska”, Sofia, Bulgaria; 5Bulgarian Academy of Sciences, Sofia, Bulgaria

Background/Objectives: Alzheimer’s disease (AD) is the most common form of dementia. According to age of onset, it can be early-onset: EOAD (diagnosed before age of 65) and late-onset (diagnosed after age of 65). EOAD accounts for about 5% of all AD cases and is thought to be caused by rare highly penetrant mutations in single genes (mainly APP,PSEN1/2), known in 10-15% of cases. The remainder may be due to enrichment of common AD risk genetic factors. In this context, we have searched for genes with AD risk variants in pooled whole genome sequencing data of EOAD patients.

Methods: A pooled DNA sample was constructed by mixing equimolar amounts of DNA samples of 49 EOAD patients, which was then analyzed by whole genome sequencing, 100 x using BGISEQ-500 system. The detected SNPs were screened for AD associated variants from DisGeNET Database.

Results: We have detected 911 AD associated variants. Among them 178 are intergenic and the remainder are found in 401 genes. The genes which contain more than 10 variants are as follows: NECTIN2 (adhesion molecule), SORL1 (Sortilin Related Receptor 1), CR1 (member of the receptors of complement activation), TOMM40 (for protein localized in the outer mitochondrial membrane) and MS4A4E (member of the membrane-spanning 4-domains subfamily A).

Conclusion: The studies of the genetic etiology of EOAD should focus also on common risk variants. For the purpose we propose five genes (NECTIN2, SORL1, CR1, TOMM40, MS4A4E) which should be further investigated in individual EOAD samples.

Grants: KP-06-N33/5 from 13.12.2019 - National Science Fund of Bulgaria

Conflict of Interest: None declared

EP11.068 Rare copy number variants involving genes associated with Parkinson disease in a cohort of individuals with autism spectrum disorders

Aurora Arghir 1, Sorina Mihaela Papuc1, erbescu alina1, Maria Dobre2, Budisteanu Magdalena1;3

1Victor Babes National Institute of Pathology, Medical Genetics Laboratory, Bucharest, Romania; 2Victor Babes National Institute of Pathology, Bucharest, Romania; 3Prof. Dr. Alexandru Obregia Clinical Hospital of Psychiatry, Bucharest, Romania

Background: Autism spectrum disorders (ASD) designate a group of neurodevelopmental conditions characterized by important clinical and genetic heterogeneity. Recent studies suggested an overlap between ASD and Parkinson disease (PD) in terms of clinical manifestation and underlying genetic defects (1,2). Objective: to assess the frequency of rare copy number variants (CNVs) which intersect PD associated genes in the chromosomal microarray results obtained from a pediatric ASD group.

Methods: 305 children diagnosed with ASD were enrolled in our study between September 2019 and February 2022. Array-CGH was performed on whole blood gDNA using 4x180K SurePrint G3 Human CGH microarray (Agilent Technologies). CNVs were classified according to ACMG guidelines.

Results: Array-CGH analysis detected clinically relevant CNVs in 27 out of 305 ASD individuals. Genomic imbalances of uncertain significance were detected in 108 patients. Among the latter, three deletions encompassed genes involved in Mendelian forms of PD or acting as risk factors in PD, namely parkin RBR E3 ubiquitin protein ligase (PRKN), synuclein alpha interacting protein (SNCAIP), and Rab9 effector protein with kelch motifs (RABEPK).

Conclusion: Our study found three deletions involving genes associated with PD in a group of 305 children with ASD. Our data adds to the previous reports of genomic imbalances of PD genes in ASD, further supporting the hypothesis that these conditions might share molecular mechanisms of pathogenesis.

Grants: The research leading to these results has received funding from the EEA Grant 2014-2021, under the project contract No 6/2019.

Ref:

1. Mai et al. https://doi.org/10.1002/acn3.51736; 2. Torres et al. https://doi.org/10.3390/ijms21165724

Conflict of Interest: None declared

EP11.069 Long-read sequencing with Cas9-targeted enrichment approach to identify the GGC expansion in the NOTCH2NLC gene

Marinella Corbetta 1, Ettore Salsano2, Chiara Pisciotta2, Carla Zanferrari3, Alberto Albanese4, Giorgio Giaccone5, Stefania Magri1, Franco Taroni1

1Fondazione IRCCS Istituto Neurologico Carlo Besta, Unit of Medical Genetics and Neurogenetics, Milan, Italy; 2Fondazione IRCCS Istituto Neurologico Carlo Besta, Unit of Rare Neurodegenerative and Neurometabolic Diseases, Milan, Italy; 3ASST Melegnano-Martesana, Unit of Neurology-Stroke Unit, Vizzolo Predabissi, Italy; 4IRCCS Humanitas Research Hospital, Department of Neurology, Rozzano, Italy; 5Fondazione IRCCS Istituto Neurologico Carlo Besta, Unit of Neurology 5 and Neuropathology, Milan, Italy

Background/Objectives: Neuronal Intranuclear Inclusion Disease (NIID) (OMIM #603472) is a neurodegenerative disorder caused by ≥60 GGC repeats in the 5’UTR of NOTCH2NLC gene. It is one of the most frequent causes of adult-onset nonvascular leukoencephalopathy in Asian population, with only three European patients with this mutation described to date.

Methods: We selected 13 patients from 6 families according to their clinical and neuroradiological phenotype. To overcome the limitations of repeat-primed PCR (RP-PCR) amplification of a GC-rich region within a cluster of 5 highly homologous genes, we used a nanopore Long-Read Sequencing (LRseq) approach (Oxford Nanopore Technologies) based on Cas9-mediated enrichment of the 5’UTR NOTCH2NLC region.

Results: We obtained a mean of 76,662 reads with a mean depth of 183X at the target region. Repeat sizing revealed the presence of one expanded allele (range 85-328) in all cases. RP-PCR assay was consistent with LRseq data in all families but one, in which sequence data revealed a deletion of ~80 bp in the repeat-flanking region targeted by the RP-PCR forward primer. Repeat contraction was observed in one family, in which a higher disease severity was associated with the shorter expansion.

Conclusion: We identified a NOTCH2NLC expanded allele in 13 European subjects from 6 families with sporadic, apparently recessive, or dominant pattern of inheritance. Our results demonstrate the essential contribution that sequencing approaches make to the understanding of the variability and structure of NOTCH2NLC repeat regions with crucial implications for diagnosis and counselling.

Grants: MoH PNRR-MR1-2022-12375648 to S.M., and FRRB CP_20/2018 to F.T.

Conflict of Interest: None declared

EP11.070 Secondary symptomatic dystonia with a genetic cause

Geir Julius Braathen 1, Kristian Tveten1, Sarka Øygarden2, Elin Dahl2

1Telemark Hospital, Department of Medical Genetics, Skien, Norway; 2Telemark Hospital, Department of Neurology, Skien, Norway

Background/Objectives: Dystonia is a movement disorder with involuntarily muscle contractions that can lead to repetitive or twisting movements. Causes can be either primary, i.e idiopathic / genetic, or secondary, i.e symptomatic dystonia.

Methods: We describe a 24 year old female. She had anomalies – pyloric stenosis, foramen ovale, progressive myopia and deformed auditory ossicles. Later she had hypermobile joints and recurrent bone fractures. She had an inconclusive genetic evaluation at 8 years of age.

Recurrent infections, pneumonias, sinusitis, breast abscess and hidrosadenitis, occurred in her teens.

At age 17 she got athetotic and choreatic movements of right arm and leg and slight oral dyskinesias. Brain MRI revealed bilateral white matter changes while PET revealed slight asymmetrical uptake in basal ganglias. Methylprednisolone for five days had clinical effect. Autoimmune cause was probable. She was treated with intravenous immunoglobulin for a year. Her dystonia disappeared.

Dizziness and headache at age 23 was primarily considered to be hypertensive in origin. Her symptoms persisted. A genetic follow-up was done.

Results: Trio whole exome sequencing of the lady and her parents revealed a de novo novel heterozygous sequence variant in the NFE2L2 gene, and the result was NM_006164.5:c.88C>T p.(Leu30Phe).

Conclusion: The NFE2L2 gene encodes the transcription factor NRF2 which is a stress regulator in mammalian cells. Sequence variants in NFE2L2 gene can cause NFE2L2-associated immunodeficiency and leukoencephalopathy. Her intermittent right sided dystonia was regarded to be caused by the simultaneously detected asymmetrical changes In her basal ganglia.

Grants: None

Conflict of Interest: None declared

EP11.071 A novel mutation in a patient with Creutzfeldt-Jakob disease.

Nataliia Andreeva 1, Roman Bikanov1

1Skolkovo Innovation Center, First genetics, Moscow, Russian Federation

Background/Objectives: A 39-year-old man came to the consultation, due to severe impairment of cognitive functions he could not independently make complaints. According to his wife until 2023 the patient was relatively healthy. In March - increased fatigue and drowsiness. Since April - episodes of disorientation and forgetfulness. In May - gait unsteadiness and time disorientation. Over the course of six months, the disease progressed rapidly and at the time of consultation the patient had severe impairment of cognitive and motor functions. A diagnosis of Creutzfeldt-Jakob disease was suspected.

Methods: The patient’s venous blood was sampled. The coding sequence and adjacent intronic regions of the PRNP gene were studied using direct automatic Sanger sequencing.

Results: Sanger sequencing of the PRNP gene: an undescribed before insertion of 144 nucleotide pairs was identified in exon 2 c.249_250ins144, p.Gln83_Pro84ins48, leading to the insertion of six additional octapeptide repeats with no frameshift in the heterozygous state. Insertions similar to this have previously been described in patients with Creutzfeldt-Jakob disease.

Conclusion: Based on complaints, medical history and examination data, the diagnosis of Creutzfeldt-Jakob disease was confirmed. It is a fatal degenerative brain disorder also known as neurocognitive disorder due to prion disease. The cause of the patient’s disease was an insertion of 144 nucleotide pairs, which had not previously been described in the literature. The prognosis for life is unfavorable. The inheritance risk is high: 50% regardless of gender. By the family’s decision two of his children were tested: the variant was not inherited.

Grants:

Conflict of Interest: None declared

EP11.073 De Novo IMMP2L Gene Deletion in a Child with Complex Motor Tics and Developmental Delay: Contributing to Susceptibility to Neurodevelopmental Disorders?

Amal Alhashem 1;2, Inesse Abdallah3, Hamoud alonazi3, Mishal Alsulami3, Hatem Elghezal3

1Prince Sultan medical military city, pediatric, Riyadh, Saudi Arabia; 2Al Faisal University, collage of medicine, Riyadh, Saudi Arabia; 3Prince Sultan Military Medical City, central lab, Riyadh, Saudi Arabia

Background/Objectives: The inner mitochondrial membrane peptidase subunit 2-like gene (IMMP2L) located in 7q31.1 has been linked to various neurodevelopmental disorders, including autism spectrum disorders, attention-deficit/hyperactivity disorder, and Gilles de la Tourette’s syndrome. With the advent of comparative genomic hybridization (CGH) as a pivotal diagnostic tool for neurodevelopmental disorders, recurrent copy number variations (CNVs) have been identified in the IMMP2L gene (MIM 605977), likely due to breakpoint clustering. This study presents clinical and molecular data from a patient with a de novo heterozygous intragenic IMMP2L microdeletion and discusses the significant association between 7q31.1 disruption by deletion and susceptibility to neurodevelopmental disorders.

Methods:

Case report

We describe a 3-year-old girl who has exhibited complex motor tics, developmental delay, and various cognitive/behavioral disturbances since early life. Chromosomal analysis and comparative genomic hybridization were performed using Agilent Technologies® oligonucleotide arrays for the patient and both parents.

Results: CGH analysis revealed a 331 Kb de novo pathogenic heterozygous deletion at 7q31.1 encompassing exons 1 to 3 of the IMMP2L gene.

Conclusion: The application of CGH in the evaluation of intellectual disability and congenital malformations has unveiled new recurrent CNVs and novel microdeletion/microduplication syndromes. Array-CGH technologies have facilitated the identification of an increased number of patients carrying CNVs. This study discusses the functions of the IMMP2L gene, suggesting that its disruption may serve as a high-risk factor for neurodevelopmental and movement disorders.

Grants: Non

Conflict of Interest: None declared

EP11.074 A new, deep intronic VUS in VPS13A gene as a culprit of AR Choreoacanthocytosis in a Saudi family with interfamilial variability.

Hajar AlAkeel 1, Mohammed Almannai1, Ahmad Abulaban2

1Riyadh, Genetic and Precision Medicine, Riyadh, Saudi Arabia; 2Riyadh, Neurology., Riyadh, Saudi Arabia

Background/Objectives: A 37 year old previously healthy lady, experienced her first symptoms at age of 24 when she developed tics (vocal + motor) and seizure activity, progressed over 2 years to chorea, dystonia, dysarthria, dysphagia, has lost ability to walk, along with cognitive slowness.

Labs showed normal lipoprotein level, acanthocytes were present in blood smear. MRI showed cudate and putamen atrophy with supratentorial white matter T2 hyperintesnities.

Family history is significant for a brother, 46 years, with milder presentation of controlled seizure, mild tongue dystonia, and similar MRI findings, 2 sisters who passed away with the same phenotype as hers, and 2 affected maternal cousin.

Methods: VPS13A sequencing sent initially showed VUSs, followed by WGS revealing a new VUS, segregation of 9 family members performed, further verification using Chorein Western blot conducted on research basis with the kind curtesy of Dr.Kevin Peikert through DZNE.

Results: Gene sequencing showed 3 VUSs, which were all excluded by family segregation, then WGS revealed a new, deep intronic VUS, C.3339 + 902A > G in VPS13A, that segregates well; homozygous in all 4 affected members. Chorein Western blot revealed no VPS13A protein for our patient nor her affected brother, it was done as well for 2 other family members with a heterozugous state, came unremarkable.

Conclusion: We here by report a novel, deep intronic variant as culprit for AR Choreoacanthocytosis in this family that was detected by WGS following -ve gene sequencing, verified by Western Blot, we would also like to address noticeable interfamilial variability in clinical phenotype.

Grants: Non.

Conflict of Interest: None declared

EP11.075 Autosomal recessive face of a dominant disease: Neuronal intranuclear inclusion disease and a novel NOTCH2NLC variant in a family.

Gokalp Celik1, Fatma Dereli1, Hasan Baş1, Gulay Ceylaner 1, Serdar Ceylaner1

1Intergen Genetics and Rare Diseases Diagnosis Center, Medical Genetics, Ankara, Türkyie

Background/Objectives: Neuronal intranuclear inclusion disease is an inherited condition, passed down with an increased chance in each generation due to heterozygous expanded GCC repeats in the 5-prime untranslated region of the NOTCH2NLC gene. Here, we present a rare homozygous frameshift variant of this gene, which we believe explains the clinical findings in two siblings with severe neurodevelopmental delay and epilepsy.

Methods: Whole-exome sequencing was conducted on both siblings and their parents to identify sequence and copy number variants. Following the absence of significant results in the analysis of disease-associated genes, we proceeded to examine rare homozygous variants through homozygosity mapping. Variants with potential pathogenic consequence were confirmed by next-generation sequencing.

Results: A girl and a boy born from a consanguineous marriage were assessed with severe developmental delay, epilepsy, and cerebral atrophy. The initial exome sequencing of the family did not reveal suitable variants to account for the clinical presentation in the affected siblings. Subsequent investigations in candidate genes identified a homozygous frameshift variant, NM_001364013.2: c.670del; p.Gln224Serfs*48 in the NOTCH2NLC genes of both siblings. The evaluation of other variants in the locus demonstrated that the variant is present in the actual gene rather than in pseudogenes. The parents were found to be heterozygous for the variant.

Conclusion: We believe we have identified the first instance of the autosomal recessive form of this disease, resulting from a total loss of function in the NOTCH2NLC. Identifying comparable family cases will aid in substantiating our hypothesis and devising additional experimental investigations.

Grants: None.

Conflict of Interest: None declared

EP11.076 Characterization of a cell model to investigate hereditary spastic paraplegia SPG9: insights into cell phenotype and amino acid supplementation as a potential treatment.

Cecilia Evangelisti1, Sherin Ramadan2, Natalia Fava2, Marco Seri1, emanuele panza 1;2

1IRCCS S.Orsola-Malpighi, Bologna, Italy; 2University of Bologna, Department of Medical and Surgical Sciences, Bologna, Italy

Background/Objectives: Spastic Paraplegia type 9 (SPG9) is a hereditary neurodegenerative disorder caused by mutations in the ALDH18A1 gene. In this study, we present a characterization of a cellular SPG9 model based on a fibroblast cell line derived from a patient harbouring a confirmed ALDH18A1-related hereditary spastic paraplegia pathogenetic variant (NM_002860.4 c.727G>C, p.Val243Leu). Our investigation focuses on elucidating fundamental cellular properties, including cell dimensions, growth dynamics, and mitochondrial features, with the aim of gaining insights into the pathological mechanisms underlying SPG9.

Methods: Initial analyses by of the SPG9 cell model revealed distinct alterations in cell dimensions, growth pattern, and mitochondrial features such as ROS production, ATP synthesis, by phalloidin staining, MTT analysis, galactose growth compared to WT cells. Furthermore, we explored a therapeutic avenue by investigating the effects of amino acid supplementation on our in vitro model.

Results: Preliminary results suggest that it is possible to identify specific cell phenotypes and that specific amino acid supplementation may hold promise as a potential treatment strategy for SPG9, offering a targeted approach to mitigate cellular dysfunction associated with ALDH18A1 gene mutations.

Conclusion: Our study provides a comprehensive understanding of the cellular characteristics of SPG9, emphasizing the potential utility of amino acid supplementation as a therapeutic intervention. Moreover, our findings contribute to unravel the molecular mechanisms underlying SPG9 and pave the way for the development of targeted therapies for this rare debilitating neurodegenerative disorder.

Grants: not applicable.

Conflict of Interest: Cecilia Evangelisti IRCCS S.Orsola-Malpighi, Sherin Ramadan PhD student University of Bologna, Dept. of Medical and Surgical Sciences, Natalia Fava Student of the Medical Genetics program, Marco Seri Director of the IRCCS S.Orsola-Malpighi, emanuele panza Assistant Professor University of Bologna, Department of Medical and Surgical Sciences

EP11.077 Prevalence of CADASIL in a consecutive series of 3,274 patients with acute cerebral stroke admitted to a single-center Stroke Unit

Silvia Baratta 1, Davide Curti2, Salvatore D’Asero2, Marcello Tognozzi2, Luca Moschini3, Massimo Camerlingo2, Franco Taroni1

1Fondazione IRCCS Istituto Neurologico Carlo Besta, Unit of Medical Genetics and Neurogenetics, Milano, Italy; 2Policlinico San Marco, Stroke Unit, Zingonia, Italy; 3Policlinico San Marco, Unit of Neuroradiology, Zingonia, Italy

Background/Objectives: Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is the most common monogenic disorder causing lacunar stroke and cerebral small vessel disease. It is caused by mutations in the NOTCH3 gene resulting in the loss or gain of a cysteine. Varying prevalence of CADASIL in stroke has been reported in multicentric studies and nonconsecutive series of patients, suggesting that the disease is likely underestimated. We determined the prevalence of NOTCH3 pathogenic variants in an MRI-verified cohort of apparently sporadic patients with lacunar infarct consecutively admitted to a single Stroke Unit serving a large population area in Northern Italy.

Methods: 3,274 consecutive patients were treated because of a first acute cerebral infarction. All patients underwent MRI evaluation. Deep next-generation sequencing of the entire NOTCH3 gene was performed in the patients showing MRI lesions compatible with CADASIL.

Results: Twenty percent of patients (n = 654) had a lacunar stroke. In 22 of them, MRI evaluation showed lesions compatible with CADASIL (confluent leukoaraiosis with Fazekas grade ≥2 and the characteristic CADASIL positive white matter hyperintensities in the anterior temporal pole and the external capsule). NOTCH3 pathogenic or likely pathogenic variants (ACMG class 4 and 5) were identified in 11 patients (50%, aged 42-81yrs).

Conclusion: Our results indicate a prevalence of 1.7% among patients with lacunar infarction, which is higher than reported in previous series (~0.5%). This might be due to (a) the monocentric nature of this study, (b) the homogeneity of the population, and/or (c) the completeness of NOTCH3 sequencing.

Grants: FRRB CP_20/2018 to F.T.

Conflict of Interest: None declared

EP11.078 Sleep problems in female carriers of premutation in the FMR1 gene

Milica Pešić 1, Milena Stevanović2, Nikola Andrejić3, Jovan Pesovic4, Sanja Cirkovic5, Sanja Dimitrijevic6, Danijela Bascarevic6, Natasa Dragasevic Miskovic7, Ivana Novakovic1;7, Dragana Protic6;8

1Faculty of Medicine, University of Belgrade, Institute of human genetics, Beograde, Serbia; 2Clinic of Psychiatry, University Clinical Centre of Serbia, Belgrade, Serbia; 3Faculty of Medicine, University of Belgrade, Belgrade, Serbia; 4Faculty of Biology, University of Belgrade, Center for Human Molecular Genetics, Belgrade, Serbia; 5Mother and Child Health Care Institute of Serbia “Dr Vukan Čupić”, Faculty of Medicine, University of Belgrade, Belgrade, Serbia; 6Fragile X Clinic, Special Hospital for Cerebral Palsy and Developmental Neurology, Belgrade, Serbia; 7Neurology Clinic, University Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia; 8Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia

Background/Objectives: Female carriers of premutation (PM) in the FMR1 gene are at risk of developing Fragile X-associated Primary Ovarian Insufficiency (FXPOI) and Fragile X-associated Tremor Ataxia Syndrome (FXTAS). However, the most prevalent problems of PM carriers are Fragile X-associated Neuropsychiatric Disorders (FXAND). Specifically, anxiety, depression, chronic fatigue, fibromyalgia, and some other problems are common. However, there are not enough data related to sleep problems and quality of sleep in PM carriers. The object of this study was to investigate sleep problems among female PM carriers.

Methods: The study included female PM carriers identified through cascade genetic testing using an AmplideX PCR kit. The sleep problem was investigated using the Pittsburgh Sleep Quality Index (PSQI).

Results: The statistical analysis was done using TIBCO Statistica. The study included 28 participants aged 48.6 ± 14.7 y. (range 21-77 y.). Participants were mostly younger than 50 y. (n = 18, 64.3%). The number of CGG repeats in the PM allele ranged from 60 to 160 (mean 93.8 ± 22.8), but a majority (n = 20, 71.4%) had greater than 80 CGGs. Most participants (n = 17, 60.7%) did not fulfill the criteria neither for FXPOI nor FXTAS. The mean PSQI score was 4.9 ± 4.6. A positive correlation was found between sleep problems and levels of anxiety which was present among 42,8% of participants. Higher PSQI scores were observed among participants with higher levels of anxiety (r = 0,6971; r2 = 4859; p = 0.00002).

Conclusion: Our study showed the high frequency of sleep problems in female PM carriers and the correlation between sleep problems and anxiety levels.

Grant: 2020 — 2024 FX2020-DP-BG-001

Conflict of Interest: None declared

EP11.079 Identification of Neurotransmission and Synaptic Biological Processes Disrupted in Autism Spectrum Disorder Using Interaction Networks and Community Detection Analysis

Joana Vilela 1;2, Hugo Martiniano1;2, Ana Rita Marques1;2, João Xavier Santos1;2, Muhammad Asif1;2;3, Célia Rasga1;2, Guiomar Oliveira4;5, Astrid Vicente1;2

1Instituto Nacional de Saúde Doutor Ricardo Jorge, Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Lisboa, Portugal; 2BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Lisboa, Portugal; 3Government College University Faisalabad, Department of Bioinformatics and Biotechnology, Faisalabad, Pakistan; 4Centro de Investigação e Formação Clínica, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra (CHUC), Unidade de Neurodesenvolvimento e Autismo, Serviço do Centro de Desenvolvimento da Criança, Coimbra, Portugal; 5Coimbra Institute for Biomedical Imaging and Translational Research, University Clinic of Pediatrics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal

Background/Objectives: Hundreds of genes have been implicated in Autism Spectrum Disorder (ASD), including neurotransmission and synaptic (NS) genes; however, the genetic architecture of ASD is unclear. We analysed the biological processes affected by NS gene variants identified in individuals with ASD, and the networks connecting those processes.

Methods: We searched for ultra-rare (UR) loss-of-function (LoF) variants in 1216 NS genes in the whole-exome sequencing dataset from the Autism Sequencing Consortium (N = 3938 cases). NS genes with UR LoF variants were used to build a network of protein–protein interactions, and the network’s protein communities were identified using the Leiden algorithm.

Results: We identified 356 variants in 208 genes, with a preponderance of UR LoF variants in the PDE11A and SYTL3 genes. These genes are important candidates for ASD since PDE11A may be implicated in the formation of social interactions and SYTL3 regulates neuronal migration and neurotransmitter release. We defined seven network communities clustering synaptic and neurotransmitter pathways, as well as mitochondrial metabolism, with biological pathways not usually associated with ASD, such as cytochromes P450 (CYPs). CYPs are important for the modulation of the metabolism of neurochemicals such as neurosteroids, dopamine or serotonin.

Conclusion: We identified novel candidate genes and defined new biological pathways for ASD. These results provide further evidence for the role of NS genes and the pathways with which these genes interact in ASD.

Grants: Fundação para a Ciência e a Tecnologia (FCT):UIDB/04046/2020 (https://doi.org/10.54499/UIDB/04046/2020) and UIDP/04046/2020 (https://doi.org/10.54499/UIDP/04046/2020) Center grants FCT to BioISI; PAC-POCI-01-0145-FEDER-016428-MEDPERSYST and DeePer-(EXPL/CCI-BIO/0126/2021).J.V.,A.R.M.,J.X.S.fellowships:BioSys PD65-2012: J.V.Ref:PD/BD/131390/2017;A.R.M.Ref:PD/BD/113773/2015;J.X.S.Ref:PD/BD/114386/2016

Conflict of Interest: None declared

EP11.080 A noval heterozygous DPF2 gene mutation associated with Coffin Siris syndrome type 7

Beril Beray Soydemir 1, Ayse Busra Akalin1, Mehmet Ersahin2, Ibrahim Akalin3

1Bahcesehir University Faculty of Medicine, Medicine, Istanbul, Türkyie; 2Medistate Hospital, Neurosurgery, Istanbul, Türkyie; 3Metagentech Center for Genetic Disorders, Medical Genetics, Istanbul, Türkyie

Background/Objectives: Coffin Siris Syndrome is a genetically heterogenous disorder compromised of lots of phenotypic series. The syndrome is characterized by mental retardation associated with coarse facial features, hypertrichosis, sparse scalp hair, absent fifth fingernails or toenails.

Methods: Here we present 12-year-old female patient born to 3rd degree consanguineous marriage and referred to our clinic with dysmorphic face, microcephaly and epilepsy. Her initial symptoms started at 6th year. She had absence type seizures. Her physical examination revealed synophrys, deviated ears, hypertrichosis, laterally deviated toes, intellectual disability, absence of reading or writing, thin upper lips, thick lower lips. Her family history revealed blind cousins born to consanguineous marriage. Cytogenetic and whole exome sequencing was done.

Results: WES analysis revealed heterozygous c.374C>G (p.Thr125Ser) variant of unknown significance in DPF2 gene.

Conclusion: The noval variant in DPF2 gene was not defined in Insilco databases before and here we, for the first time, declare the association of heterozygous c.374C>G (p.Thr125Ser) mutation with autosomal dominantly inherited Coffin Siris Syndrome type 7.

Grants:

Conflict of Interest: None declared

EP11.081 Gene panels and mode of inheritance – What not to do

Cecilie Pape Madsen 1, Allan Thomas Højland1;2, Thomas Blauenfeldt3

1Aalborg University Hospital, Department of Clinical Genetics, Aalborg, Denmark; 2Aalborg University, Department of Clinical Medicin, Aalborg, Denmark; 3Aalborg University Hospital, Department of Molecular Diagnostics, Aalborg, Denmark

Background/Objectives: Primary coenzyme Q10 deficiency type 4 (COQ10D4) is an autosomal recessive disorder caused by mitochondrial respiratory chain dysfunction due to pathogenic variants in the COQ8A gene. The main clinical manifestations are childhood-onset cerebellar ataxia, exercise intolerance, and, sometimes, varying degrees of learning disorders and seizures. A potential treatment is oral administration of high-dose oral coenzyme Q10 supplements. We present the case of a woman who has suffered from mild ataxia and learning difficulties since childhood as well as muscle weakness over the past decade. Both the woman and her sister, who also exhibits ataxia, are offspring of unaffected consanguineous parents. The sister’s son has suffered from lifelong ataxia with cerebral atrophy.

Methods/Results: The index patient was analyzed using whole genome sequencing with an ataxia-specific gene panel. We found the genotype: NM_020247.5(COQ8A):c.[1826T > C];[1826T > C] p.[(Leu609Pro)];[(Leu609Pro)]. The index’s sister and nephew were analyzed using whole exome sequencing of the COQ8A gene only. The sister genotype: NM_020247.5(COQ8A):c.[1826T > C];[1826T > C] p.[(Leu609Pro)];[(Leu609Pro)]. The nephews genotype: NM_020247.5(COQ8A):c.[1826T > C];[499_500delCA] p.[(Leu609Pro)];[(Gln167Ilefs*12)].

Conclusion: Our research revealed two sisters born to consanguineous parents who were both homozygous for COQ8A:c.1826T>C p.(Leu609Pro) and the son of one of the sisters who were compound heterozygous for COQ8A:c.1826T>C p.(Leu609Pro) and COQ8A:c.499_500delCA p(Gln167Ilefs*12). The variants have to our knowledge not previously been reported in the literature. This case illustrates the importance of including genes in gene panels with other modes of inheritance than suggested by the pedigree.

Conflicts/Grants: none.

Conflict of Interest: None declared

EP11.082 Clinical features in Ukrainian patients with Wiedemann-Steiner syndrome caused by mutations in KMT2A gene

Vira Galagan 1, Nikita Pozhar1, Maryna Tsygankova1, Olga Zhurakhovska1, Valentyna Kurakova1, Viktoriia Serafymovych1

1Okhmatdyt, The Center of Medical Genetics, Kyiv, Ukraine

Background/Objectives: Three children of different ages with various clinical symptoms were diagnosed Wiedemann-Steiner syndrome (WSS) at Specialized Centre of Medical Genetics of children’s hospital “Okhmatdyt” during 2023.

Methods: Genetic counseling of 3 children, aged 1 months to 9 years was undergone. Cytogenetic examination included G-banding karyotyping according to the standard protocol has determined normal male karyotypes. All the cases were confirmed by next-generation sequencing (NGS) based molecular testing.

Results: All children were born from physiological pregnancies, via physiological deliveries, in term, with normal body weight. All the families had normal family history. Congenital developmental defects (CDD) take place in 2 of 3 patients – hypoplasia of hippocampus, asymmetry of brain hemispheres, platybasia, atrophy of optiс nerves, L-shaped renal dystopia, cryptorchidism, inguinal hernia. Prenatally diagnosed CDD was only in 1 patient – renal CDD. Phenotypes of 2 children were similar to Rubinstein-Taybi syndrome phenotype. Hypertrichosis cubiti and broad terminal phalanges of the first fingers and toes were observed in all patients. Short stature and developmental delay were identified in 2 of 3 children, seizures – in 1 of 3. With regard to findings of the molecular genetic testing: 2 patients had heterozygous pathogenic variants c.3790C>T (p.Arg1264*) and с.553C > T (p.Arg185*) in gene KMT2A respectively, one heterozygous pathogenic variant c.10498C>T (p.Gln3500*) and one variant of uncertain significance c.9269C>T (p.Pro3090Leu) in gene KMT2A were found in 1 case.

Conclusion: With the purpose of making a final diagnosis, differential diagnosis of WSS with Rubinstein-Taybi syndrome and Cornelia de Lange syndrome using NGS based molecular testing is recommended.

Conflict of Interest: None declared

EP11.083 Novel inherited CHD3 gene variants in patients with drug-resistant epilepsy

Lucie Sedlackova 1, Markéta Vlčková2, Petra Lassuthova1, Markéta Havlovicová2

1Second Faculty of Medicine, Charles University and University Hospital Motol, Neurogenetic Laboratory, Department of Pediatric Neurology, Prague 5, Czech Republic; 2Second Faculty of Medicine, Charles University and University Hospital Motol, Department of Biology and Medical Genetics, Prague 5, Czech Republic

Background/Objectives: Variants in CHD3 gene were described to be a cause of Snijders-Blok-Campeau syndrome (SNIBCPS), the rare autosomal dominant neurodevelopmental disease characterized by a global developmental delay, impaired intellectual and speech development, behavioral disorder and distinctive facial phenotype. The severity of the neurological symptoms and the presence of other features is variable; however, drug-resistant epilepsy has not been described as one of the common feature in these patients so far. Reduced penetrance and/or variable expressivity has been reported recently.

Methods: We report three patients with drug-resistant epilepsy from unrelated families. Carrier parents without epilepsy had a milder phenotype (psychiatric diagnosis) or were healthy. Patients were analyzed by exome sequencing and the variants were confirmed by Sanger sequencing in the whole trios.

Results: We described patients carrying novel inherited heterozygous missense CHD3 variants. These variants were not previously published, were classified as variants of uncertain significance and were reported in population database gnomAD v4 with zero or one allele frequency. Patients‘ phenotypes were found to overlap with those in SNIBCPS patients including global developmental delay, speech disorder/delay, intellectual disability and different dysmorphic features, except for drug-resistant epilepsy.

Conclusion: Considering the fact that many neurodevelopmental disorders have similar clinical features it is necessary to carefully correlate genotypes with phenotypes already described in literature. As drug-resistant epilepsy has not been described as common feature in SNIBCPS patients, it remains to answer if CHD3 variants could be causal in our patients despite the reduced penetrance and/or variable expressivity.

Grants: LX22NPO5107 (MEYS)

Conflict of Interest: None declared

EP11.084 Clinical and molecular description of four families with dentatorubropallidoluysian atrophy diagnosis

Barbara Masotto 1;2;3, Eulàlia Rovira-Moreno1;2;3, Anna Esteve-García4, Ariadna Padró Miquel5, Cristina Sau-Puig4, Cinthia Aguilera5, Nuria Llecha4;5, Laura Trujillano1;2;3, Marta Codina1;2;3, Amaia Lasa-Aranzasti1;2;3, Berta Alemany6, Pedro Alía4, leticia Iranzo Nuez1;2;3, Elena García-Arumí1;2;3, Eduardo Tizzano1;2;3

1Hospital Universitari Vall d´Hebron, Área de Genética Clínica y Molecular, Barcelona, Spain; 2European Reference Network on Rare Congenital Malformations and Rare Intellectual Disability ERN -ITHACA; 3Vall d´Hebron Institut de Recerca (VHIR), Medicine Genetics Group, Barcelona, Spain; 4Hospital Universitari de Bellvitge-IDIBELL. Universitat de Barcelona, Clinical Genetics Unit, Barcelona, Spain; 5Laboratori Clínic Territorial Metropolitana Sud, Hospital Universitari de Bellvitge-IDIBELL. Universitat de Barcelona, Genetics Laboratory, Barcelona, Spain; 6Servei de Neurologia ICS/IAS, Hospital Josep Trueta/Hospital Santa Caterina, Unitat d’Atàxies, Paraparèsies Espàstiques i Malalties Rares, Girona, Spain

Background/Objectives: Dentatorubropallidoluysian atrophy (DRPLA) is a neurodegenerative disease characterized by ataxia, myoclonus, chorea, and dementia in adults, and epilepsy, intellectual disability (ID), and cognitive impairment in children. It results from pathogenic expansions of CAG triplets in the ATN1 gene, with correlation between repeat number and onset, phenotype, and prognosis. The aim is to provide a clinical-molecular description of four families diagnosed with DRPLA through a retrospective genetic analysis.

Methods: Families underwent PCR amplification of the repetitive region in exon 5 of ATN1 and capillary electrophoresis. In the pediatric patient from Family 2, repeat number was retrospectively reviewed in whole-exome sequencing (WES) with Expansion Hunter (EH).

Results: Family 1: 69-year-old woman with ataxia (22/61 repeats). Mother and grandmother with ataxia. Family 2: 48-year-old man with chorea and ataxia (22/59 repeats). His 13-year-old daughter had epilepsy and ID (21/69 repeats via PCR and 73 via EH). Grandmother with ataxia. Family 3: 30-year-old man with ADHD, epilepsy, and ID (19/67 repeats). Father with dementia and abnormal gait (17/62 repeats). Unaffected sister (19/64 repeats). Family 4: 43-year-old woman with ataxia (20/66 repeats).

Conclusion: In children with ID and/or epilepsy and family history of adult-onset ataxia, a DRPLA diagnosis should be considered. For cases involving cognitive-neurological impairment, ATN1 expansion analysis should follow array-CGH and WES. Expansion detection methods can enhance the diagnostic yield in cases of neurodevelopmental abnormalities. This study emphasizes the complexity of presymptomatic testing and recommends incorporation of tailored genetic counselling protocols.

Conflict of Interest: None declared

EP11.085 Association of polygenic liabilities for schizophrenia and bipolar disorder with educational attainment and cognitive aging

Chi-Shin Wu1, Chia-Lin Hsu1, Mei-Chen Lin1, Mei-Hsin Su2, Yen-Fen Lin1, Chia-Yen Chen3, Po-Chang Hsiao4, Yi-Jiun Pan2, Pei-Chun Chen1, Yen-Tsung Huang5, Shi-Heng Wang 1

1National Health Research Institutes; 2China Medical University; 3Biogen; 4National Taiwan University; 5Academia Sinica

Background/Objectives: To elucidate the specific and shared genetic background of schizophrenia (SCZ) and bipolar disorder (BPD), this study explored the association of polygenic liabilities for SCZ and BPD with educational attainment and cognitive aging.

Methods: Among 106,806 unrelated community participants from the Taiwan Biobank, we calculated the polygenic risk score (PRS) for SCZ (PRSSCZ) and BPD (PRSBPD), shared PRS between SCZ and BPD (PRSSCZ+BPD), and SCZ-specific PRS (PRSSCZvsBPD). Based on the sign-concordance of the susceptibility variants with SCZ/BPD, PRSSCZ was split into PRSSCZ_concordant/PRSSCZ_discordant, and PRSBPD was split into PRSBPD_concordant/PRSBPD_discordant. Ordinal logistic regression models were used to estimate the association with educational attainment. Linear regression models were used to estimate the associations with cognitive aging (n = 27,005), measured by the Mini-Mental State Examination (MMSE), and with MMSE change (n = 6,194 with mean follow-up duration of 3.9y) in individuals aged≥ 60 years.

Results: PRSSCZ, PRSBPD, and PRSSCZ+BPD were positively associated with educational attainment, whereas PRSSCZvsBPD was negatively associated with educational attainment. PRSSCZ was negatively associated with MMSE, while PRSBPD was positively associated with MMSE. The concordant and discordant parts of polygenic liabilities have contrasting association, PRSSCZ_concordant and PRSBPD_concordant mainly determined these effects mentioned above. PRSSCZvsBPD predicted decreases in the MMSE scores.

Conclusion: Using a large collection of community samples, this study provided evidence for the contrasting effects of polygenic architecture in SCZ and BPD on educational attainment and cognitive aging and suggested that SCZ and BPD were not genetically homogeneous.

Grants: National Health Research Institutes (CG-112-GP-10, CG-113-GP-10, NHRI-EX109-10931PI, NHRI-EX110-10931PI, NHRI-EX111-10931PI).

Conflict of Interest: None declared

EP11.087 Peculiarities of phenotypic signs in inverted duplication of the long arm of chromosome 2 in the q35q37.3 area. Clinical case.

Iryna Lastivka 1, Vita Antsupova2, Ludmila Khlunovska1, Larysa Sheiko3, Ljudmila Brisevac3, Anastasiya Babintseva4, Iana Ushko2

1Bukovinian State Medical University, Department of Pediatrics and Medical Genetics, Chernivtsi, Ukraine; 2Bohomolets National Medical University, Department of Pathophysiology, Kyiv, Ukraine; 3Shupyk National Healthcare University of Ukraine, Department of medical and laboratory genetics, Kyiv, Ukraine; 4Bukovinian State Medical University, Department of Pediatrics, Neonatology and Perinatal Medicine, Chernivtsi, Ukraine

Background/Objectives: Inverted duplication of the distal long arm of chromosome 2 is a rare aberration. Features of phenotypic manifestations depend on the size and localization of the duplication. We present the phenotype of a girl with an inverted duplication of 2q in the region q35q37.3.

Methods: Clinical genealogical, cytogenetic and molecular genetics methods.

Results: A 5-year-old girl with a diagnosis: "Organic disorder of the CNS with cognitive impairment and general language underdevelopment of the 1st level. Inferior flaccid paraparesis, ataxic syndrome." Phenotype: Mongoloid incision, hypertelorism of the eyes, thin upper lip, Cupid’s Bow-shaped mouth, long philtrum, short neck, wide feet, valgus deformity of the lower extremities, bilateral clubfoot, kyphoscolytic posture. The child was born at the 41st week of pregnancy from the 1st planned pregnancy against the background of anemia and oligohydramnios. Short umbilical cord. Anthropometric parameters at birth: weight 3600 g (+1σ), body length 55 cm (+2σ). The Apgar scale – 7/8 points. Delay in stato-kinetic development was detected in the 4th month after birth. Family history: the mother’s age was 30 years; the father’s age – 32 years. Heredity on the mother’s side is burdened with diabetes. The maternal cousin has hemihypertrophy.

The chromosomal analysis using GTG staining revealed an inverted duplication of the long arm of chromosome 2 in the q35q37 region. The type of rearrangement was confirmed by the FISH method: 46,XX,dup(2)(pter→q37.3::q37.3::q35→qter).ish dup(2)(wcp2 + ).

Conclusion: Awareness of the clinical features of inverted duplication (2)(q35q37) is important for the detection of chromosomal aberrations in children with impaired psycholinguistic development.

Conflict of Interest: None declared

EP11.088 Transcriptional regulatory role of SKOR1 in restless legs syndrome

Faezeh Sarayloo 1;2, Dan Spiegelman2, Daniel Rochefort2, Fulya Akcimen1, Rachel De Barros Oliveira2, Patrick Dion2, Guy Rouleau1;2

1McGill University, Human Genetics, Canada; 2McGill University, Montreal Neurological Institute, Canada

Background/Objectives: Restless legs syndrome (RLS) is a common sleep-related sensory-motor disorder. RLS is characterized by uncomfortable sensations in the legs during the evening or at night. The symptoms can be partially relieved by movement, so affected individual needs to walk during rest time; this interferes with sleep. Among the GWAS identified loci for RLS, variants in the SKOR1 noncoding regions were among the most significant and robustly replicated reports.

Methods: SKOR1 is highly expressed in the CNS of humans and mice. Skor1 acts as a corepressor of Lbx1 transcription factor in mice and these two genes act together to regulate the cell fate of interneurons in the dorsal horn of the spinal cord. We investigated the regulatory role of SKOR1 using a global RNA-sequencing approach in human cell lines where SKOR1 was either overexpressed or silenced. For this work we generated and validated a novel poly-clonal anti-SKOR1. This was followed by pathway and gene set enrichment analyses of the differentially expressed genes.

Results: We observed enrichment of genes involved in neurodevelopment and iron metabolism, two RLS relevant pathways. Analysis of our different datasets further supports and highlights the regulatory role of SKOR1, which when dysregulated might represent a key pathogenic element of RLS.

Conclusion: This study highlights a transcriptional regulatory role for SKOR1 on pathways involved in RLS. A better understanding of SKOR1 and its activity could open new avenues of investigation for the development of a much-needed therapy.

Grants: RN254517-332736/Canadian Institute of Health Research/International

Conflict of Interest: None declared

EP11.089 Genetic causes of epilepsy in children

Karim Yanin 1, Olga Gumeniuk1, Yuriy Chernenkov1, Anna Fisun1, Olga Groznova2

1Saratov State Medical University, Saratov, Russian Federation; 2Veltischev Research and Clinical Institute for Pediatrics and Pediatric Surgery of the Pirogov Russian National Research Medical University, Moscow, Russian Federation

Background/Objectives: Epilepsy is a common neurological disorder characterized by recurrent epileptic seizures. In approximately 30% of cases there is a clear extraneous acquired cause (eg, head trauma, stroke) but in the remainder genetic factors have a major role (M S Hildebrand et al, 2013). Objective: to Study the genetic causes of epilepsy in children.

Methods: 24 patients with epilepsy aged 1-17 years were examined. General examination, neurological examination, electroencephalography, including monitoring, whole-genome sequencing and сonfirmatory analysis sequencing by Sanger were performed.

Results: There were no pathogenic variants in the genome in 2 patients. The following pathogenic variants in genes were identified in 22 (92%) patients with epilepsy: ARIDIB, ARIDIA, ARID2 (Coffin-Siris type 1, 2 and 6 accordingly, OMIM 135900), KCNMA1, WDR26 (Skraban-Deardorff syndrome, 617616), PHF6 (Börjeson-Forssman-Lehmann syndrome, 301900), ANKRDII (KBG-syndrome, 148050), MAGEL2 (Schaaf-Yang syndrome, 615547), ACTA2, CDH19 (618916), MECP2 (n = 2)(Rett syndrome, 312750), KMT2D (Kabuki syndrome, 147920), DYNCIHI (614563), CREBBP (Menke-Hennekam syndrome, 618332), NONO (300967), DYNC1H1 (614563) and UBE2A (300860), MED13L (616789), CDK19 (618916), PP3CA (617711), NBEA (619157), MOSPD2. All variants have been confirmed by Sanger sequencing. In 81% cases had of the mutations de novo.

Conclusion: Pathogenic variants in the genome have been identified in most patients with epilepsy. All variants are associated with syndromes or conditions characterized by the development of seizures or epilepsy. Epilepsy in children is an absolute indication for conducting a molecular genetic study.

Grants: No

Conflict of Interest: None declared

EP11.090 A unique Neuronal Ceroid Lipofuscinosis Type 1 case due to paternal Uniparental Disomy

Simge Tuana Ay 1;2, Sena Cetin1;2, Filiz Ozen1, Elif Yilmaz Gulec1;2

1Goztepe Prof. Dr. Suleyman Yalcın City Hospital, Medical Genetics, Istanbul, Türkyie; 2Istanbul Medeniyet University, Faculty of Medicine, Department of Medical Genetics, Istanbul, Türkyie

Background/Objectives: Neuronal Ceroid Lipofuscinosis-Type 1 (NCL1) (MIM # 256730), is a rare and devastating neurodegenerative disorder primarily affecting the central nervous system. Biallelic pathogenic variants in the PPT1 gene result in NCL1 which manifests with progressive cognitive decline, motor impairment, seizures and vision loss starting in infancy. We report a unique case of NCL1 with a rare mechanism of occurrence for autosomal recessive disorders.

Methods: A 1-year-old boy with neuromotor developmental delay and seizures was evaluated in our clinic and he underwent chromosome analysis, SNP-based microarray, NGS-based panel testing respectively. The variant identified through the panel test was confirmed by Sanger sequencing.

Results: Multigene panel testing identified a novel homozygous missense PPT1(NM_000310.4) gene variant: c.559C>T (p.His187Tyr). The proband’s homozygosity and his father’s heterozygous carrier status were confirmed by Sanger sequencing, while the mother displayed the wild-type sequence. Considering the possibility of uniparental disomy (UPD) as a rare mechanism of disease, father-mother-proband trio SNP-microarray analysis were evaluated. A 9 Mb region of loss of heterozygosity on chromosome 1 including the PPT1 gene was identified in the proband carrying paternal markers proving the paternal-UPD resulting in homozygosity for the paternal variant.

Conclusion: This study highlights the importance of integrating multiple genetic analyses to uncover the complex genetic basis of NCL-Type 1 and sheds light on the potential contribution of UPD mechanisms in autosomal recessive disorders. Consequently, the family’s recurrence risk for the disorder in subsequent pregnancies dropped from anticipated 25 % risk to population risk. Detailed genetic counseling was provided.

Grants: None

Conflict of Interest: None declared

EP11.091 FRRS1L-related developmental epileptic dyskinetic encephalopathy (FRRS1L-DEDE): 24 cases demonstrating the facial phenotype.

Mohnish Suri 1, Matthias De Wachter2, Sarah Weckhuysen3, Michael C. Kruer4, Somayeh Bakhtiari4, Henry Houlden5, Reza Maroofian5

1Nottingham Clinical Genetics Service, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom; 2Department of Paediatric Neurology, Antwerp University Hospital, Antwerp, Belgium; 3Department of Neurology, Antwerp University Hospital, Applied and Translational Neurogenomics Group, VIB Centre for Molecular Neurology, VIB, Antwerp, Belgium; 4Department of Child Health, Phoenix Children’s Hospital, Phoenix, Arizona, United States; 5Department of Neuromuscular Disorders, Institute of Neurology, University College London, London, United Kingdom

Consortium: FRRS1L-DEDE Study Group

Background/Objectives: FRRS1L-related developmental epileptic dyskinetic encephalopathy (FRRS1L-DEDE) is a severe, autosomal recessive neurodevelopmental disorder that was first reported in 2016. Although a total of 50 individuals from 16 families have been reported to date, no information is available regarding the facial dysmorphology of this condition.

Methods: We reviewed the clinical and molecular findings in 100 individuals from 58 families. The facial dysmorphology of FRRS1L-DEDE was assessed using photographs and videos from 24 individuals from 21 families including 18 new cases from 18 families and 6 previously published cases from 3 families.

Results: The facial dysmorphology cohort included 8 females and 14 males (2 patients’ sex unknown) with ages ranging from 1-26 years. FRRS1L variants (homozygous in 18 families) included missense, frameshift, and stop-gain variants and a recurrent in-frame deletion (c.584_586del p.(Gly195del)).

Analysis of the full cohort confirmed that the key clinical findings were severe to profound developmental delay, absent or limited speech, drug-resistant epilepsy with onset usually in the first 2 years of life, movement disorder, and facial dysmorphism. Regression was seen in 76% of cases, where this information was available, with an age range of 3-39 months.

The most frequently seen facial dysmorphic features were tall forehead, high anterior hairline, narrow forehead, hypertelorism, low-set ears, full or broad nasal tip, long philtrum, and broad or tall chin. There was a recognisable face in about half of the younger children.

Conclusion: This series helps to confirm the clinical phenotype of FRRS1L-DEE with emphasis on its facial dysmorphology, which can help with its clinical recognition.

Grants:

Conflict of Interest: None declared

EP11.092 Attenuated huntingtin gene CAG nucleotide repeat size in individuals with Lynch syndrome

Karin Skarping1, Larissa Larissa2, Åsa Petersén3, Huu Phuc Nguyen2, Samuel Gebre-Medhin 1

1Division of Clinical Genetics, Department of Clinical Genetics and Pathology, Lund; 2Ruhr University, Department of Human Genetics, Bochum, Germany; 3Lund University, Translational Neuroendocrine Research Unit, Lund

Background/Objectives: DNA mismatch repair (MMR) is thought to contribute to the onset and progression of Huntington disease by promoting somatic expansion of the pathogenic CAG nucleotide repeat in the huntingtin gene (HTT). Individuals with Lynch syndrome (LS) are prone to cancer due to heterozygous constitutional pathogenic variants (PV) in any of the MMR genes (MLH1, MSH2, MSH6 or PMS2). To investigate if PVs in MMR genes affect the number of constitutional CAG repeats in HTT we have studied HTT CAG repeat size in two cohorts of individuals with LS.

Methods: Number of CAG repeats in HTT was determined using clinical standard PCR amplification and capillary electrophoresis fragment analysis in lymphocyte DNA from individuals (Lund cohort/Bochum cohort) with constitutional PV in MLH1 (n = 18/82), MSH2 (n = 19/92), MSH6 (n = 21/24) and controls (n = 19/559). The sum of CAG repeats for both HTT alleles in each individual was calculated due to unknown segregation with the LS allele.

Results: In the larger Bochum cohort, the sum of CAG repeats was lower in the MLH1 subgroup compared to controls (MLH1 35.40 CAG repeats ± 3.6 vs. controls 36.89 CAG repeats ± 4.5; p = 0.014). All LS genetic subgroups in the Bochum cohort displayed lower frequencies of unstable HTT intermediate alleles and lower HTT somatic CAG repeat expansion index values compared to controls.

Conclusion: Collectively, our results indicate that MMR gene haploinsufficiency could have a restraining impact on constitutional HTT CAG repeat size and support the notion that the MMR pathway is a driver of nucleotide repeat expansion diseases.

Grants: ALF-agreement.

Conflict of Interest: None declared

EP11.093 The contribution of genome sequencing to the diagnosis of early-onset drug-resistant epilepsies

Giulia Barcia 1;2, Jean-Madeleine de Sainte Agathe2;3, Lionel Arnaud2;3, Marie Faoucher2;4, Anne-Sophie Lèbre5;6, Nicolas Chatron6;7, Christele Dubourg4, Cyril Mignot8, Laurence Perrin9, Patrick Van Bogaert10, Silvia Napuri4, Anne de Saint Martin11, Marie Therese Abi Warde11, Sarah Baer11, Mathieu MILH12, Nathalie Villeneuve12, Anne Lepine12, Dorothée Ville7, Anne-Lise Poulat7, Matthildi Athina Papathanasiou Terzi7, Eleni Panagiotakaki7, Claire Bar13, Frederic Villega13, Caroline Hachon-LeCamus14, Jennesson Mélanie15, Julien Thevenon16, Henri Margot17, Marine Legendre17, Elise Boucher18, Juliette Piard18, Olivier Patat14, Valentin Ruault19, Marine Lebrun20, Anaïs Lharidon2, Virginie Bernard6, Rima Nabbout21, Laurent Villard22, Gaetan Lesca23

1Hopital Necker Enfants Malades, Médecine Génomique, Paris, France; 2Laboratoire Seqoia, Paris, France; 3Hopital Pitié Salpetrière, Génétique, Paris, France; 4CHU Rennes, Laboratoire de Génétique Moléculaire et Génomique, Rennes, France; 5CHU Reims, Génétique, Reims, France; 6Plateforme Auragen, Lyon, France; 7CHU Lyon, Génétique, Lyon, France; 8Hopital Pitié Salpetrière, Paris, France; 9Hopital Universitaire Robert Debré, Génétique, Paris, France; 10CHU Reims, Pediatric Neurology, Angers, France; 11CHU Strasbourg, Pediatric Neurology, Strasbourg, France; 12CHU Marseille, Pediatric Neurology, Marseille, France; 13CHU Bordeaux, Pediatric Neurology, Bordeaux, France; 14Hospital Center University De Toulouse, Pediatrics, Toulouse, France; 15CHU Reims, Pediatrics, Reims, France; 16CHU Grenoble, Genetics, Grenoble, France; 17CHU Bordeaux, Genetics, Bordeaux, France; 18CHU Besancon, Genetics, Besançon, France; 19CHU Montpellier, Genetics, Montpellier, France; 20CHU Saint-Etienne, Genetics, Saint-Etienne, France; 21Hopital Necker Enfants Malades, Pediatric Neurology, Paris, France; 22CHU Marseille, Genetics, Marseille, France; 23CHU Lyon, Genetics, Lyon, France

Consortium: Reference Centers for Rare Epilepsies

Background/objectives: The EPIGENE network was set up in 2014 to coordinate the development of high-throughput sequencing for the genetic diagnosis of epileptic disorders. We defined a gene panel (GP) that was regularly upgraded. Since the introduction of the French Genomic Medicine plan, in 2020, genome sequencing (GS) is available as a second-line genetic analysis in patients with early-onset drug-resistant epilepsies.

Methods. Between January 2021 and September 2023, 284 GS results were returned to the patients.

Results. GS provided an additional diagnosis compared with the GP in 83 patients (29%). Pathogenic or likely pathogenic variants were found in 64 different genes: 39 genes absent from any version of the GP, 15 genes present only in the latest versions, and 10 genes included in the GP since the first version. Two or more variants were found for 12 genes. For several genes, a clinical presentation with epilepsy as a prominent feature has recently been described (ANKRD11, SMC1A). False-negative results of the GP were related to deep intronic variants (SCN1A, SCN8A), partial exon deletions or deletions of promoter regions, duplication of a repeat motif (ARX), and balanced translocation (CDKL5). A de novo variant of SCN1A or PCDH19 were found in more 8/13 patients with Dravet syndrome or other fever-sensitive epilepsies.

Conclusion. GS, now included in the clinical workup of patients with a negative GP, provides a significant additional yield in patients with early-onset drug-resistant epilepsy. GS also increases the diagnostic rate in Dravet syndrome through global exploration of the SCN1A and PCDH19 genes.

Conflict of Interest: None declared

EP12 Neuromuscular Disorders

EP12.001 Another novel patient with pathogenic variant in MAPK8IP3 and clinical features of the MAPK8IP3-related neurodevelopmental disorder

Kumiko Yanagi 1, Hiroshi Matsumoto2, Masahiko Yamamori1, Hajime Wakamatsu3, Tomomi Hidai1, Arisa Igarashi1, Yoichi Matsubara4, Tadashi Kaname1

1National Center for Child Health and Development, Department of Genome Medicine, Tokyo, Japan; 2Saitama Medical University, Department of Pediatrics, Saitama, Japan; 3National Defense Medical College, Department of Pediatrics, Saitama, Japan; 4National Center for Child Health and Development, Tokyo, Japan

Background: We previously reported five Japanese patients with neurodevelopmental disorder (OMIM #618443) having two heterozygous variants in MAPK8IP3 (NM_001318852.2), c.1735C>T:p.(Arg579Cys) or c.3439C>T:p.(Arg1147Cys). The patients had delayed motor development, intellectual disability, and hypoplasia of the corpus callosum. Some patients had spastic diplegia, no walking or hypotonia in infancy. However, the clinical features of patients with pathogenic variants of MAPK8IP3 are not yet clear. Here we report another novel patient with the c.1735C>T variant. We summarise clinical features of the MAPK8IP3-related neurodevelopmental disorder by reviewing previous and present patients.

Patient: The proband is a boy born to nonconsanguineous parents. He is followed up due to intellectual disability and motor retardation. He has nonverbal. Spastic diplegia was presented at the age of 1 year. He was able to crawl but not walk at 18 months. Brain MRI showed hypoplasia of the corpus callosum and enlarged ventricles due to reduced white matter volume.

Methods: After obtaining written informed consent from his parents, trio-based whole exome sequencing analysis was performed using a Human All Exon V6 Kit (Agilent Technology, CA) and NovaSeq 6000 (Illumina, CA). Variants were called and annotated using CompStor NOVOS and InSight (OmniTier, CA).

Results: After filtering analysis of the variants, a heterozygous de novo variant c.1735C>T was identified. Patients with variant c.1735C>T, including previous patients, shared spastic diplegia and walking difficulties.

Conclusion: Spastic diplegia and walking difficulty may be characteristic features of the patients with the variant c.1735C>T. The accumulation of more patient information should be important.

Grants: Japan Agency for Medical Research and Development (23ek0109549s0203, 23ek0109549s0203)

Conflict of Interest: None declared

EP12.002 Genetic screening for carriage of hereditary motor and sensory neuropathy type VIC in the yakut population

Anastasia Maksimova 1, Polina Golikova1, Aitalina Sukhomyasova1;2, Nadezhda Maksimova1

1Medical Institute, M.K. Ammosov North-Eastern Federal University, Research Laboratory “Molecular Medicine and Human Genetics”, Yakutsk, Russian Federation; 2Republican Hospital №1 – “National Medical Center”, Medical Genetic Center, Yakutsk, Russian Federation

Background/Objectives: Hereditary motor and sensory neuropathy type VIC with optic atrophy (HMSN6C) is an autosomal recessive axonal sensorimotor peripheral neuropathy characterized by progressive distal muscle weakness and atrophy primarily affecting the lower limbs. Individuals usually have pes cavus, hammertoes, and atrophy of the intrinsic hand muscles. In addition, progressive optic atrophy and visual impairment occur during adulthood. Previously, we found a mutation in the PDXK gene among patients with Charcot-Marie-Tooth disease with optic nerve atrophy in 48 people. The purpose of this study is to determine heterozygous carriage of hereditary motor-sensory neuropathy type 6C in the yakut population.

Methods: The population sample of healthy individuals consisted of 96 people (46 men, 50 women). We used the Real-time PCR method with fluorescent probes based on the Scientific Laboratory of Molecular Medicine and Human Genetics.

Results: DNA samples (n = 96) of healthy people from unrelated families of the Yakut ethnic group were studied to determine heterozygous carriage. As a result of the analysis, heterozygous carriage was identified in 3 people out of 96, which amounted to 2.88%.

Conclusion: The results obtained demonstrate a high frequency of heterozygous carriage (2.88%), which indicates the need for prevention through the introduction of molecular genetic screening of the population, primarily prenatal screening in affected families.

Grants: Research is part of the project FSRG-2020-0014 “Arctic Genomics: epidemiology, heredity and pathology”.

Conflict of Interest: None declared

EP12.003 Lack of GFPT1 mimicking congenital myasthenic syndrome facilitates aggregation of misfolded proteins and triggers ER stress

Ruchen Zhang 1, Bisei Ohkawara1, Mikako Ito1, Akio Masuda1, Kinji Ohno1

1Nagoya University Graduate School of Medicine, Center for Neurological Diseases and Cancer, Division of Neurogenetics, Nagoya, Japan

Background/Objectives: Pathogenic variants in human GFPT1, encoding glutamine fructose-6-phosphate amidotransferase 1 (GFPT1), cause limb-girdle congenital myasthenic syndromes (LG-CMS). GFPT1 is the rate limiting enzyme in the hexosamine biosynthesis pathway (HBP) to produce UDP-GlcNAc for glycosylations of proteins and lipids. Dysregulation of glycosylation generates misfolded/unfolded proteins, which subsequently causes endoplasmic reticulum (ER) stress and activates unfolded protein response (UPR). The objectives of this study are to analyze a knock-in (KI) mice carrying a pathogenic variant identified in a GFPT1-CMS patient, and to explore the mechanisms by which GFPT1 deficiency causes skeletal muscle dysfunctions.

Methods: We established a homozygous KI mouse line carrying c.716dupG in Gfpt1, which is equivalent to NM_001244710.2:c.722dupG identified in a LG-CMS patient, by CRISPR/Cas9 technique. Behavioral, morphological, and biochemical analyses were performed on the Gfpt1-KI mouse.

Results: In the skeletal muscles of Gfpt1-KI mice, Gfpt1 expression and the amount of UDP-GlcNAc were markedly reduced. Gfpt1-KI mice showed muscle weakness, smaller cross section area of myofibers, fragmented AChR clusters, abnormal neuromuscular junction (NMJ) ultrastructures, and tubular aggregates. Gfpt1-KI mice also showed increased expressions of ER stress markers. Denervation exacerbated misfolded protein aggregates, ER stress, and attenuated autophagy more in Gfpt1-KI mice than in wild-type mice.

Conclusion: Lack of Gfpt1 impairs glycosylation, which triggers misfolded protein aggregates and maladaptive UPR, and eventually causes CMS-like phenotypes including muscle atrophy and NMJ dysfunction.

Grants: Japan Society for the Promotion of Science (JSPS), and THERS Interdisciplinary Frontier Next Generation Researcher (Bioscience)

Conflict of Interest: None declared

EP12.004 Loss-of-function variants in MYL1 are associated with a severe congenital myopathy characterized by a selective involvement of type II myofibres

Irene Madrigal 1;2, Andrés Nascimento3, Laia Rodriguez-Revenga1;2, Cristina Jou3, Daniel Natera-de Benito3, Giorgia Sebastiani4, Gemma Arca4

1Hospital Clinic of Barcelona and IDIBAPS, Biochemistry and Molecular Genetics, Barcelona, Spain; 2Instituto de Salud Carlos III, CIBER of Rare Diseases (CIBERER), Spain; 3Hospital Sant Joan de Déu, Neuromuscular Unit, Barcelona, Spain; 4Hospital Clinic Barcelona, Neonatology, BCNatal, Barcelona

Background/Objectives: Congenital myopathies are a clinically and genetically heterogeneous group of disorders characterized by early-onset muscle weakness and distinctive structural abnormalities in muscle biopsy. Massive sequencing technology is being applied to newborns to diagnose unknown conditions and it has been increasingly used for the genetic diagnosis of diseases with heterogeneous clinical phenotypes, or with multiple genes involved. The MYL1-related congenital myopathy is a very rare entity that has been diagnosed in very few individuals.

Methods: Exome sequencing, histological, immunohistochemical analysis, immunofluorescence, and ultrastructural studies were used to achieve a correct diagnosis.

Results: The patient was a premature newborn in which two compound heterozygosis variants were identified in the MYL1 gene. The maternally inherited variant (p.Gln112Ter) produces a premature stop codon in the protein, and the paternally inherited variant (c.478+1G > A) is a splicing variant predicted to produce the loss of function for authentic splice site and promoting an exome skipping. Clinical manifestations included severe generalized weakness, generalized areflexia, thoracic scoliosis, bilateral cryptorchidism and long and thin bones. The patient died at 24 days and was diagnosed with a very severe congenital myopathy with selective involvement of type II myofibres.

Conclusion: The clinical presentation of MYL1-related congenital myopathy can be very variable and an appropriate diagnostic approach requires the integration of clinical revisions, muscle magnetic resonances and genetic and histological analysis. In addition, our work reinforces the benefits of implementing exome sequencing in neonates with clinical suspicion of congenital myopathy

Grants: AGAUR (2021-SGR-01492). CIBER-Rare Diseases is an initiative of the ISCIII.

Conflict of Interest: None declared

EP12.005 Detection of known pathogenic variants in whole genome sequencing data

Ann-Kathrin Zaum 1, Natalie Pluta1, Anna Forster1, Astrid Pechmann2, Delia Lorenz3;4, Juliane Lippert1, Yvonne Jelting1, Simone Rost1;5

1Institute of Human Genetics at the University of Würzburg, Würzburg, Germany; 2University of Freiburg - pediatric neurology and muscle disorders, Freiburg im Breisgau, Germany; 3Center for Rare Diseases, University Hospital Würzburg, Würzburg, Germany; 4University Children’s Hospital Würzburg, University of Würzburg, Würzburg; 5Medizinisch Genetisches Zentrum München, München, Germany

Background/Objectives: Whole genome sequencing (WGS) is an effective technique that is becoming increasingly important in the field of molecular genetic diagnostic. As more variants in non-coding regions such as introns and promoter regions are reported as pathogenic, the interpretation of WGS data becomes easier.

Methods: We present two cases of hitherto unsolved cases of muscle weakness, one case with elevated creatine kinase levels and one with congenital hypotonia/myasthenia. Preceding molecular diagnosis including panel and whole exome sequencing (WES) revealed no pathological findings. We performed WGS on the patients as part of a WGS study. In one case we additionally performed segregation analysis.

Results: In both cases we were able to identify pathogenic variants. The homozygous variant c.914+6T > C in RXYLT1 was previously published also in homozygous state in a different patient and proved to impact splicing [1]. In the second case, a heterozygous pathogenic variant (c.264C>A, p.(Asn88Lys)) in RAPSN had already been detected by WES. We could identify the second variant (c.-199C > G, p.?) in RAPSN by WGS. This variant is a known pathogenic promoter variant [2]. The segregation analysis confirmed the compound heterozygosity of the variant.

Conclusion: In summary, we successfully used WGS to establish the genetic diagnosis of two patients and showed that WGS is already a useful diagnostic tool.

Grants: Funded by Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) – 444748124.

1. Zaum, A.K., et al. Neuromuscul Disord, 2018. 28(8): p. 671-674.

2. Ohno, K., et al. Hum Mol Genet, 2003. 12(7): p. 739-48.

Conflict of Interest: None declared

EP12.006 Exome or panel in paediatric patients with spastic paraplegia?

Dana Safka Brozkova 1, Anna Uhrova Meszarosova1

12nd Faculty of Medicine, Charles University, Department of Paediatric Neurology

Background/Objectives: Hereditary spastic paraplegia (SPG) is a heterogeneous disorder with almost 150 genes implicated in the disease. The onset is usually in adulthood, but clinical sings of SPG like the lower limb spasticity or gait on tips displays already in childhood.

Methods: During years 2014 – 2023 were enrolled 107 paediatric patients with the onset of the disease before the 20 years of age. In patients the virtual gene panel included 144 genes was examined from WES data. In patients without genetic diagnosis after the panel examination, the exome data were evaluated.

Results: The diagnostic yield in paediatric patients with virtual gene panel for SPG was 21 %. However, extended exome evaluation increased the diagnostic yield to 31 %. The biggest difference in the clarified patients according to the used method was between the groups of patients 0 - 5 and 15 - 20 years of disease onset. Among the 15 clarified youngest patients (0 – 5) were 9 genetically diagnosed with virtual gene panel and 6 with exome sequencing. On the contrary, all clarified adolescent patients (15 – 20) were genetically diagnosed with panel evaluation.

Conclusion: Our large cohort of 107 paediatric patients clearly point out the importance of exome evaluation in young children to state the genetic diagnosis. The spastic gait in small children frequently mimics symptoms of the more complex disease not covered in the virtual gene panel.

Grant reference: Supported by the MHCR AZV NU22-04-00097.

Conflict of Interest: None declared

EP12.007 A founder mutation in the COQ7 gene, may result in a pure form of Hereditary Spastic Paraplegia (HSP)

Parsa Soleimani 1, Aida Ghasemi2, Davood Zare-Abdollahi1, afagh alavi1;2

1University of Social Welfare and Rehabilitation Sciences, Genetics Research Center, Tehran, Iran; 2Tehran University of Medical Sciences, Neuromuscular Research Center, Tehran, Iran

Background/Objectives: Hereditary spastic paraplegia; HSP is a heterogeneous group of neurological disorders characterized by lower limbs spasticity and weakness. To date, ~85 HSP-causing genes have been identified; thus, many HSP-causing genes have remained unknown. Here, we suggest the COQ7 gene, previously linked to primary COQ10-deficiency, may be a novel putative HSP-causing gene.

Methods and Results: During whole-exome sequencing of a large cohort of Iranian HSP cases (>90 families), a homozygous variant, NM_016138.5:c.332T>C:p.(Leu111Pro), in the COQ7 gene was identified in three probands and co-segregated with the disease status in their families, establishing it as a disease-causing variant. Haplotype analysis using six intragenic single-nucleotide polymorphisms/SNPs in COQ7 revealed an identical haplotype in the probands.

Conclusion: To date, five mutations have been reported in COQ7: three in a primary COQ10-deficiency, one in a distal hereditary motor neuropathy, and one in a HSP-like patient. Interestingly, the last patient was a Canadian girl of Iranian descent who carried p.(Leu111Pro) variant. Although the haplotype of the Canadian girl is not available, we suppose she has the same haplotype and suggest the variant may be a founder mutation in the Iranian population that results in a distinct phenotype such as HSP. Therefore, it appears that mutation in COQ7, similar to various other HSP-causing genes such as SPG11, is linked to a spectrum of phenotypes.

Such phenotypic heterogeneities have also been recently reported in another gene involved in the COQ10-synthesis pathway, COQ4. Building on this precedent, we propose that COQ7 may also represent a novel putative HSP-causing gene.

Grants: 2592

Conflict of Interest: None declared

EP12.008 Suspected Novel Founder Neurogenetic Disorder in Slovak and Czech Roma Populations

Maria Giertlova 1;2;3, Petra Drenčáková1, Lenka Nosková4, Jana Šaligová5, Ľudmila Potočňáková5, Simona Drobňáková5, Miriam Kolníková6, Katarína Kušíková6, Patrícia Balážová6, Angela Baranová7, Ladislav Gurčík8, Zuzana Mažeriková9, Martin Mistrík10, Paula Stretavská11, Marie Zikanova4, Viktor Stránecký4, Jakub Sikora4, Tomas Honzik4, Stanislav Kmoch4

1Unilabs Slovakia LtD, Department of Medical Genetics, Košice, Slovakia; 2Faculty of Medicine, University of P.J. Šafárik in Košice, Department of Neurology, Kosice, Slovakia; 3Children’s Faculty Hospital Banská Bystrica, Department of Medical Genetics, Banska Bystrica, Slovakia; 4First Faculty of Medicine, Charles University and General University Hospital in Prague, Department of Paediatrics and Inherited Metabolic Disorders, Prague, Czech Republic; 5Children’s Faculty Hospital in Košice, Department of Inherited Metabolic Disorders, Kosice, Slovakia; 6National Institute of Children’s Diseases, Department of Peadiatric Neurology, Bratislava, Slovakia; 7Children’s Faculty Hospital in Košice, Department of Peadiatric Neurology, Kosice, Slovakia; 8Hospital AGEL Levoča, Department of Neurology, Levoča, Slovakia; 9Unilabs Slovakia LtD, Department of Medical Genetics, Prešov, Slovakia; 10Unilabs Slovakia LtD, Department of Medical Genetics, Spišská Nová Ves, Slovakia; 11Unilabs Slovakia LtD, Department of Medical Genetics, Banská Bystrica, Slovakia

Background/Objectives: The different genetic background and specificities of the rare diseases results in need of the scientific, population-specific genomic approach in the Roma population. We present the preliminary data of genomic analysis of a cohort of 13 patients with a childhood onset neuromuscular phenotype of unknown genetic cause.

Methods: Exome sequencing and whole genome sequencing were performed in affected patients and healthy family members.

Results: 5 males and 7 females (5 unrelated families) of Roma ethnicity from Slovakia and Czech Republic presented with motor development delay, short stature, failure to thrive, hypotony, hypermobility, areflexia, facial stigmatization, severe scoliosis, progressive proximal muscle weakness from approximately 5 to 6 years of age with loss of independent walking from 9 to 18 years of age. No specific biochemical abnormality was present. Facial weakness, hypomimia, ptosis, ophthalmoplegia, and dysphagia have also been observed. All affected individuals shared a 2.5 Mb common region of homozygosity on chromosome 17. In this region, three rare homozygous variants of unknown significance segregated with above mentioned phenotype: an intronic variant in CHRNB1 (OMIM #616314, #616313) and two variants in OMIM genes not associated with the disease - FGF11, belonging to the fibroblast growth factor family, and TMEM102, involved in the mitochondria-dependent apoptosis pathway.

Conclusion: A probable new founder disease with autosomal recessive inheritance is suggested. Further analysis is required to elucidate the underlying molecular pathogenesis.

Grants: The work was supported by the grants NU23-07-00281 and LX22NPO5107. We thank to the National Center for Medical Genomics (LM2023067) for WES analyses.

Conflict of Interest: None declared

EP12.009 ATXN1 intermediate repeats and clinical progression in ALS patients

Francesca Luisa Conforti 1;1, Martina Pecoraro2, Benedetta Perrone1, Viviana Mosca2, Paola Ruffo1, Vincenzo La Bella2

1University of Calabria, Department of Pharmacy and Health and Nutritional Sciences, Medical Genetics Laboratory, Rende, Italy; 2University of Palermo, Department of Experimental Biomedicine and Clinical Neurosciences, Palermo, Italy

Background: Repeat expansions involving the ATXN-1 and ATXN-2 genes have been shown to be part of the complex genetic architecture of amyotrophic lateral sclerosis (ALS). In 2012, the association between ATXN1 intermediate repeats and ALS was first described by our research group [1]. We further showed that ATXN1 is a disease modifier that predisposes C9orf72 repeat expansion carriers to develop ALS [2].

Methods: In this study, we analyzed the frequency and clinical characteristics of 328 ALS patients with ATXN1 intermediate repeats (32-33 glutamines, polyQ) in a cohort of Italian patients.

Results: Patients with an increased number of polyQ repeats ≥ 32 had a trend of survival shorter than those with <31 repeats (median survival: polyQ ≥32, 25 months [IQR = 18-31; polyQ ≤32, 30 months [IQR = 26-33], log-rank = 2.11 [DF = 1], p = 0.14). Interestingly, though not significant, there was a trend towards a more aggressive disease, in terms of clinical progression, as measured by DFS [3] in the group with polyQ ≥32, 0.82, IQR: 0.4-1.9 vs polyQ ≤32, 0.58, IQR: 0.3-1.13, p = 0.10.

Conclusion: In our study, ALS patients with intermediate polyQ ≥32 ATXN-1 repeat show a non-significant but suggestive trend toward faster disease progression and shorter survival. These findings will need to be confirmed in a larger population and, if confirmed, it could pave the way for refining individual prognostic predictions and improving ALS clinical trial design, as is already happening with ATXN-2.

1.Neurology. 2012;79:2315–20.

2.Neurobiol Aging. 2021;99:99.e7–99. e14

3.Neurology. 2006; 66:265–267.

Conflict of Interest: Francesca Luisa Conforti Associate professor, University of Calabria, Martina Pecoraro PhD fellowship, Benedetta Perrone post PhD fellowship, Viviana Mosca fellowship, Paola Ruffo PhD fellowship, Vincenzo La Bella Full Professor, University of Palermo

EP12.012 Genetic variability in oxidative stress pathway: Impact on clinical course and treatment response in patients with spinal muscular atrophy

Maruša Barbo 1, Tanja Blagus1, Vita Dolzan1, Blaž Koritnik2;3, Metka Ravnik-Glavač1

1Faculty of Medicine, University of Ljubljana, Institute of Biochemistry and Molecular Genetics, Ljubljana, Slovenia; 2University Medical Centre Ljubljana, Institute of Clinical Neurophysiology, Ljubljana, Slovenia; 3Faculty of Medicine, University of Ljubljana, Department of Neurology, Ljubljana, Slovenia

Background: The severity of spinal muscular atrophy (SMA) varies among affected individuals, as does their response to treatment with nusinersen/risdiplam, emphasizing the need to explore contributing factors. We hypothesized that these factors are influenced by oxidative stress processes, given their emerging role in neurological disorders, aiming to gain insight into SMA etiopathogenesis.

Methods: We genotyped 54 SMA patients and 163 healthy controls for 12 polymorphisms in the SOD2, CAT, GPX1, NFE2L2, KEAP1, HMOX1, and HMOX2 genes using competitive allele-specific PCR. Genetic data were analysed for associations with SMA type, age at symptom onset, and treatment outcome, defined as longitudinal change in motor function. Statistical analyses included logistic regression, Fisher’s exact, Mann-Whitney, and Kruskal-Wallis tests.

Results: AA homozygotes for the HMOX2 rs1051308 polymorphism (p = 0.042) were associated with more severe SMA (type 2) compared to AG heterozygotes who were more likely to have milder SMA (type 3). Additionally, HMOX2 rs1051308 AA homozygotes showed earlier onset of disease symptoms (p = 0.031). In terms of response to treatment, heterozygotes (OR = 5.538; 95% CI = 1.273–24.104; p = 0.023) and carriers of at least one polymorphic GPX1 rs1050450 A allele (OR = 4.500; 95% CI = 1.054–19.217; p = 0.042) had higher odds of a favourable response to treatment than GG homozygotes.

Conclusion: Genetic variations in the HMOX2 and GPX1 genes influence the course of SMA and reveal potential biomarkers for predicting treatment response.

Grants: Slovenian research agency (No. P1-0170, M.B. PhD thesis grant).

Conflict of Interest: None declared

EP12.013 Clinical Manifestations and Genetic Analysis of Huntington’s Disease in a Ukrainian Family

Danyal Nassar 1;1, Khrystyna Kachurak1, Oleksandr Kornutii1, Anastasiia Dutchak1

1Ivano-Frankivsk, Student, Ivano-Frankivsk, Ukraine

Background/Objectives: Huntington’s Disease (HD) is a rare autosomal dominant neurodegenerative disorder characterized clinically by involuntary movements, neuropsychiatric symptoms, and dementia. The cause is an increase in CAG repeats in the IT15 gene, which encodes the Huntington protein. The aim of this study was to investigate clinical manifestations and analyse DNA changes in a family with Huntington’s chorea.

Methods: Syndromological, clinical-genealogical, molecular-genetic (Institute of Molecular Biology and Genetics, Kyiv, Ukraine).

Results: Our 43-year-old woman proband complained of involuntary movements of the trunk and limbs, worsening of speech and memory, slowed reactions, headaches, swallowing difficulties, starting five years ago. Admitted to Ivano-Frankivsk’s regional clinical hospital due to worsening health, she shares these symptoms with her mother, aunt, uncle, grandfather, and biological sister. The proband has two children: 25 and 14 years old. The daughter shows no clinical symptoms, while the son has been diagnosed with cryptogenic epilepsy and complex focal seizures. Neurological assessment revealed slow reactions, involuntary movements, facial twitching, Romberg’s sign instability, and coordination errors. The proband receives clonazepam and phenibut. Genetic analysis of the HTT gene showed 45 CAG repeats, while the son, daughter, and sister revealed 22, 42, and 45 CAG repeats, respectively.

Conclusion: Huntington’s Disease, a chronic neurodegenerative disease, is characterized by focal neuronal loss and brain atrophy. This patient with Huntington’s Disease exhibited typical symptoms and a positive family history. There is a correlation between the CAG repeat number and symptom. Current treatments cannot halt disease progression, necessitating breakthroughs to promptly prevent it.

Grants: No.

Conflict of Interest: None declared

EP12.014 Comprehensive clinical and molecular characterization of Neurofibromatosis Type 1: Unveiling the diversity of pathogenic variants and overlapping phenotypes

Woori Jang 1, Jiyeon Kim2, Kyoung-Jin Park3, Joonhong Park4

1College of Medicine, Inha University, Department of Laboratory Medicine, Incheon, Korea, Rep. of South; 2Inha University Hospital, Department of Laboratory Medicine, Incheon, Korea, Rep. of South; 3Sungkyunkwan University Samsung Changwon Hospital, Department of Laboratory Medicine, Changwon, Korea, Rep. of South; 4Jeonbuk National University Medical School and Hospital, Department of Laboratory Medicine, Jeonju, Korea, Rep. of South

Background/Objectives: Neurofibromatosis type 1 (NF1) stands out as one of the prevailing Mendelian disorders, exhibiting a broad spectrum of manifestations. The 2021 NF1 diagnostic criteria revision highlights the role of comprehensive molecular characterization in achieving early diagnosis. Our objective was to present the clinical and mutation spectrum observed in individuals primarily diagnosed with NF1.

Methods: Clinical exome sequencing or Sanger sequencing, utilizing gDNA and cDNA, was employed, and MLPA or CMA was conducted to identify large deletions/duplications within the NF1 gene.

Results: Out of 69 individuals screened for NF1, 43 patients (62.3%) exhibited pathogenic or likely pathogenic variants (PVs/LPVs). Among these, 7 were novel and 33 were known variants. Notably, 15 variants were frameshift (37.5%), 11 were nonsense (27.5%), 8 were splice-site (20.0%), 4 were missense (10.0%), and 2 (5.0%) manifested as contiguous deletions. These contiguous deletions included one multi-exon deletion (exons 32-36) and a 1.4 Mb type-1 NF1 deletion (arr[GRCh37] 17q11.2(28,996,679_30,391,813)x1). Utilizing NGS, a novel LPV in SPRED1 was found in a child presenting multiple café-au-lait macules (CALMs) and developmental delay. A CBL PV was detected in a child displaying CALMs, cryptorchidism, pectus excavatum, and developmental delay. Additionally, a female exhibiting the coexistence of two germline PVs in NF1 and BRCA2 genes presented with typical features of NF1 and early-onset ovarian serous carcinomas.

Conclusion: Our findings contribute to expanding the molecular spectrum associated with NF1 and disorders exhibiting overlapping phenotypes.

Grants: This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT) (No. 2022R1G1A1013497).

Conflict of Interest: None declared

EP12.015 An unexpected transmission of ACTA1-related myopathy due to a somatic mosaic variant heterozygous in blood: a pitfall for molecular analysis

Benjamin Durand 1, Aurélie Gouronc2, Aleksandra Nadaj Pakleza3;4, Nicolas Dondaine2, Claire Feger2, Maria Cristina Antal5;6, Béatrice Lannes7;8, sophie scheidecker2, Valerie Biancalana2

1Hôpitaux Universitaires de Strasbourg, Service de Génétique Médicale, Institut de Génétique Médicale d’Alsace, Strasbourg, France; 2Hôpitaux Universitaires de Strasbourg, Laboratoires de Diagnostic Génétique, Strasbourg, France; 3Hôpitaux Universitaires de Strasbourg, Centre de Référence des Maladies Neuromusculaires Nord-Est-Ile-de- France, Service de Neurologie, Strasbourg, France; 4ERN Euro-NMD, Paris, France; 5Hôpitaux Universitaires de Strasbourg, Service de Pathologie - UF6349 Foetopathologie, Strasbourg; 6Université de Strasbourg - Faculté de médecine, Institut d’Histologie ICube UMR7357 - équipe IMIS, Strasbourg; 7Hôpitaux Universitaires de Strasbourg, Service de Pathologie, Strasbourg; 8Université de Strasbourg, Strasbourg

We report on a family in which a pregnancy was marked by a first trimester therapeutic abortion for fetal immobility. Exome sequencing performed on chorionic villi DNA identified a heterozygous ACTA1 pathogenic missense variant, previously described in a severe antenatal form of myopathy.

Parental segregation by Sanger sequencing identified the ACTA1 variant at heterozygous state in the father who had no significant clinical signs of myopathy. However, he presents with left peripheral facial paralysis and paresis of the right brachial plexus put down to a difficult delivery. The variant was not inherited from his parents and subsequent Sanger sequencing of other tissues confirmed the somatic mosaicism ranging from 50% in saliva to 15 % in skin fibroblasts and 10% in the muscle.

De novo ACTA1-variants can occur during embryonic development and may result in a mosaic pattern. Rare healthy parents of affected offspring have been described with very-low-grade somatic mosaicism in blood. In addition, some patients presenting with a variable severity of symptoms have been described with somatic mosaicism in blood and muscle. However, this the first case of a paucisymptomatic individual carrying a pathogenic ACTA1 variant at the heterozygous state in blood.

The present study points out that mosaic variants might represent a pitfall when detectable heterozygous on blood DNA and not in lane with clinical data. Indeed, the discrepancy between the severity of the fetal phenotype and the absence of neuromuscular symptoms in the father of the fetus due to this somatic mosaicism represented a molecular diagnosis challenge.

Conflict of Interest: None declared

EP12.016 A homozygous missense variant in COL12A1 causes Ulrich congenital muscular dystrophy-2 in a consanguineous family

Hijab Zahra 1, Ghazala Zafar2, Haq Nawaz Khan2, lubaba khalid2, omar mahmud3, Mathias Toft4, Zafar Iqbal5, Ambrin Fatima2

1Aga Khan University, centre for regenerative medicine and stem cell research, Karachi, Pakistan; 2Aga Khan University, biological and biomedical sciences, Karachi, Pakistan; 3Aga Khan University, medical college, Karachi, Pakistan; 4University of Oslo, institute of clinical medicine, Norway; 5University of Oslo, department of neurology, Norway

Background: The COL12A1 gene encodes a protein of the fibril-associated collagens with interrupted triple helices (FACIT) family. Mutations in COL12A1 are associated with two distinct clinical phenotypes: a single autosomal dominant mutation in heterozygotes causes Bethlem muscular dystrophy, while biallelic combinations of autosomal recessive variants lead to Ullrich congenital muscular dystrophy-2 (UCMD2).

Objectives/ Methodology: Here, we studied a five-generation consanguineous Pakistani family with three affected members who presented with variable degrees of impaired motor function, including muscle weakness and hypotonia. The plausible candidate variant for the phenotype was determined by whole exome sequencing.

Results: We identified a novel homozygous missense variant in exon 6 of the COL12A1 gene (c.3151C>G: p.Val1051Leu) which segregated with the phenotype in an autosomal recessive manner. This variant was classified as VUS according to the ACMG classification. The consequences of the candidate variant were explored at a cellular level by immunofluorescence and immunoblotting using patient-derived fibroblasts. These primary cells exhibited significant reductions in COL12A1 protein expression versus healthy controls as measured by immunofluorescence and western blotting. However, the levels of COL12A1 mRNA remained unchanged across patient and control fibroblasts.

Conclusion: Considering previous literature together with our findings, the role of COL12A1 mediated autosomal recessive UCMD-2 is strengthened. We add to the clinical and genetic spectrum of COL12A1 related recessive disease, raising the total number of patients from four to seven.

Conflict of Interest: None declared

EP12.017 Mosaic genotypes in Duchenne/Becker muscular dystrophy (DMD/BMD) as a biological model for the manifestation of DMD symptoms in female carriers

Elena Zinina 1, Vyacheslav Tabakov1, Irina Panchuk1, Inna Sharkova1, Olga Shchagina1, Aleksander Polyakov1

1Research Centre for Medical Genetics, Moscow

Background/Objectives: The aim of our study was to investigate the mechanism of development of Duchenne/Becker muscular dystrophy in patients with variants in the DMD gene in a mosaic state.

Methods: Two patients were included in our study. Potentially pathogenic variants were searched for by MLPA MRC-Holland and massively parallel sequencing.

Results: Patient 1: 24 years old, complaints of painful spasms in leg and back muscles, dilated cardiomyopathy, elevated CPK level up to 4000 U/L. Multiplex ligase-dependent sample amplification (MLPA) revealed deletion of exon 3 of DMD gene in mosaic state in blood cells (88% of clones), this deletion was also recorded in cells of other embryonic origin: seminal fluid (90%), fibroblasts (87%) and myoblasts (66%). Patient 2: 6 years old, complaints of recurrent muscle pain, elevation of CPK up to 3500 U/L, pathogenic variant - 61.5% in blood. We searched for exon 3 deletion detected in patient 1 in genomic DNA and cDNA isolated from passages 1, 3, 5, 7 of the patient’s myoblast culture obtained during muscle biopsy. In passage 1 culture, the ratio of normal to mutant clones was 20%:80%. In subsequent passages, the proportion of mutant clones gradually decreased to a ratio of 80%:20% in passage 7, indicating elimination of myoblasts carrying the deletion.

Conclusion: Muscle tissue is a syncytium that requires 10% of normal dystrophin for normal function. Studying and finding the mechanism of action of the defective gene is important for understanding the risks of muscle pathology in female carriers, as well as the prospects for gene replacement therapy.

Grants:

Conflict of Interest: None declared

EP12.018 Translating the Effects of CMT2S Variants with Human-on-a-Chip Neuromuscular Junction Systems: Personalized Medicine Rescue

Christina Tyner 1, Sandra Smieszek1, Bart Przychodzen1, Caroline Johnson1, Christos Polymeropoulos1, Gunther Birznieks1, Walker Hagan2, Leticia Lenkiu2, Caitlyn Niccum2, Heather Cannon2, Rocky Brighton2, Xiufang Guo3, Kenneth Hawkins3, Nesar Akanda3, James Hesperos2;3, Mihael Polymeropoulos1

1Vanda Pharmaceuticals Inc., Washington, DC, United States; 2Hesperos, Inc, Orlando, United States; 3UCF NanoScience Technology Center, Orlando, United States

Background/Objectives: Immunoglobulin mu‐binding protein 2 (IGHMBP2) variants can cause Charcot-Marie-Tooth disease Type 2S (CMT2S), a rare neuromuscular disorder, by resulting in abnormal RNA processing leading to alpha‐motor neuron degeneration.

A patient was reported with IGHMBP2 variants. Whole genome sequencing revealed a cryptic splice site variant resulting in IGHMBP2 haploinsufficiency. We examined the resulting phenotype by analyzing patient motor neurons (MNs) and neuromuscular junctions (NMJs).

Methods: In collaboration with Hesperos Inc., MNs and skeletal myofibers were generated from the patient’s fibroblasts. CMT2S-MNs and wild-type MNs (WT-MNs) were integrated into a dual-chamber NMJ platform. NMJ functional defects were analyzed by NMJ number per chamber, fidelity, and fatigue index (FI). MNs were then incubated with an antisense oligonucleotide (ASO) designed to target and restore IGHMBP2, and treatment effects were assessed.

Results: Patch-clamp electrophysiology revealed hyperexcitability and spontaneous firing of CMT2S-MNs and reduced resting membrane potential, capacitance, and cell body area compared to WT-MNs. CMT2S-NMJ analysis revealed no fidelity or NMJ number differences but a higher FI than WT-NMJ. CMT2S-NMJs showed quick fatigue, characterized as tetanus followed by decay, simulating muscle weakness. We demonstrate rescue of NMJ functioning following ASO treatment, captured by an FI decrease and reduction in decay and sporadic responses, likely to lead to clinical motor control restoration.

Conclusion: The genetic diversity and limited natural history of CMT2S have limited precise models for therapeutic investigation. This N-of-1 case exemplifies the research capabilities allowing for the design of personalized in vitro models of rare diseases, serving as a framework for CMT2S research.

Conflict of Interest: Christina Tyner Vanda Pharmaceuticals, Vanda Pharmaceuticals, Sandra Smieszek Vanda Pharmaceuticals, Vanda Pharmaceuticals, Bart Przychodzen Vanda Pharmaceuticals, Vanda Pharmaceuticals, Caroline Johnson Vanda Pharmaceuticals, Vanda Pharmaceuticals, Christos Polymeropoulos Vanda Pharmaceuticals, Vanda Pharmaceuticals, Gunther Birznieks Vanda Pharmaceuticals, Vanda Pharmaceuticals, Walker Hagan: None declared, Leticia Lenkiu: None declared, Caitlyn Niccum: None declared, Heather Cannon: None declared, Rocky Brighton: None declared, Xiufang Guo: None declared, Kenneth Hawkins: None declared, Nesar Akanda: None declared, James Hesperos: None declared, Mihael Polymeropoulos Vanda Pharmaceuticals, Vanda Pharmaceuticals

EP12.019 First in class ASO targeting IGHMBP2 cryptic splice variant: efficacy and safety

Caroline Johnson 1, Sandra Smieszek1, Bart Przychodzen1, Christina Tyner1, Christos Polymeropoulos1, Gunther Birznieks1, Mihael Polymeropoulos1

1Vanda Pharmaceuticals Inc., Washington, DC, United States

Background/Objectives: Charcot-Marie-Tooth disease Type 2S (CMT2S) is a rare autosomal recessive Charcot-Marie-Tooth disease subtype. Variants in immunoglobulin mu‐binding protein 2 (IGHMBP2) may result in abnormal RNA processing leading to alpha‐motor neuron degeneration, causing CMT2S. A patient was reported with pathogenic variants within IGHMBP2. Whole genome sequencing (WGS) revealed a paternally inherited cryptic splice site variant (non‐coding variant (c.1235+894 C > A)) deep in intron 8. The resulting transcript undergoes nonsense‐mediated decay, resulting in IGHMBP2 haploinsufficiency. Our objective was to target this cryptic splice site, rescuing IGHMBP2 with a novel antisense oligonucleotide (ASO).

Methods: WGS confirmed this pathogenic variant and a 19-mer ASO was designed targeting deep in intron 8 (c.1235+894 C > A), around CACTTCCAC(A)GGGGGAAGA. Fibroblasts underwent ASO treatment (1µM) and 48-hour incubation. Flow cytometry and fluorescein-labelled ASO (GFP + 99.8%) confirmed cellular entry.

Results: IGHMBP2 increased (~50-70%) in treated samples (WB antibody Sigma SAB2106426). qPCR confirmed an increased ratio of restored wild-type transcript to cryptic exon-containing transcript (~1.8-fold). RNA sequencing further confirmed an increase of IGHMBP2 expression of 1.3log2-fold (adjusted p-value < 0.002). Data support this ASO as a potential treatment for restoring IGHMBP2. Potential ASO toxicity was evaluated in a 3-month rat study. Transient clinical observations were noted and resolved within 24 hours, with no neurobehavioral changes. The no-adverse-event-level was the highest dosage.

Conclusion: CMT2S cases caused by IGHMBP2 variants are increasingly reported. N-of-1 precision medicine approach is instrumental in designing treatments for this diverse genetic disorder. This case exemplifies how WGS-based clinical diagnoses and research capabilities allow for the design of personalized ASO-based treatments.

Conflict of Interest: Caroline Johnson Employee and stock holder of Vanda Pharmaceuticals, Inc., Employee and stock holder of Vanda Pharmaceuticals, Inc., Sandra Smieszek Employee and stock holder of Vanda Pharmaceuticals, Inc., Employee and stock holder of Vanda Pharmaceuticals, Inc., Bart Przychodzen Employee and stock holder of Vanda Pharmaceuticals, Inc., Employee and stock holder of Vanda Pharmaceuticals, Inc., Christina Tyner Employee and stock holder of Vanda Pharmaceuticals, Inc., Employee and stock holder of Vanda Pharmaceuticals, Inc., Christos Polymeropoulos Employee and stock holder of Vanda Pharmaceuticals, Inc., Employee and stock holder of Vanda Pharmaceuticals, Inc., Gunther Birznieks Employee and stock holder of Vanda Pharmaceuticals, Inc., Employee and stock holder of Vanda Pharmaceuticals, Inc., Mihael Polymeropoulos CEO and stock holder of Vanda Pharmaceuticals, Inc., CEO and stock holder of Vanda Pharmaceuticals, Inc.

EP12.020 Biallelic variants in the neuronal isoform of the DST gene as a cause of hereditary sensory neuropathy

Petra Lassuthova 1, Dagmar Grecmalova2, Annette Lischka3, Katja Eggermann3, Ingo Kurth3, Radim Mazanec4

1University Hospital Motol, Department of Paediatric Neurology, Prague; 2University Hospital Ostrava, Department of MEdical Genetics, Ostrava; 3RWTH Aachen, Institut für Humangenetik und Genommedizin, Aachen; 4University Hospital Motol, Neurology department, Prague

Consortium: the ENISNIP Network

Background/Objectives: The DST gene has many isoforms that differ in tissue expression - isoform 1 is expressed in myoblasts, isoform 3 in skin, isoform 6 in brain. Biallelic variants in the neuronal isoform of the DST gene cause hereditary sensory and autonomic neuropathy 6 (HSAN6) – a form of peripheral neuropathy with distal weakness and severe sensory loss, symptoms of dysautonomia might also be present.

Methods: We have analysed exome sequencing data from 1200 patients in the Czech Republic. Of these, more than 600 patients were diagnosed with peripheral neuropathy. All variants in the DST gene from our in-house database were evaluated and we searched for biallelic variants in the neuronal form of DST.

Results: Among our patients, we detected 759 rare variants in the DST gene, of which 489 were in non-coding regions. The residual 270 were evaluated. Interesting biallelic variants were detected in six families and segregation studies were performed for characterization of these variants.

After all analyses, causal pathogenic variants in the DST gene were found in one family. In the other families, variants did not segregate with the disease and/or the phenotype of the patients was different.

Conclusion: Assessing DST variants is challenging. Finding biallelic variants in a patient is quite common. However, the variants need to be carefully evaluated based on isoform and phenotype, and segregation studies are required. DST is a very rare cause of HSAN. In our cohort, pathogenic variants in the DST gene are the cause of HSAN in only one family, so far.

Grants: AZV NW24-04-00349

Conflict of Interest: None declared

EP12.021 Emery-Dreifuss muscular dystrophy in Two Moroccan families and report of a novel LMNA mutation

Yasmina Rahmuni 1;2, Youssef El Kadiri1;2, lyahyai jaber1, birouk nazha3, nesnassi mounir3, abdelaziz sefiani1;2, ilham ratbi1;4

1Faculty Of Medicine And Pharmacy Of Rabat, Research team in genomics and molecular epidemiology of genetic diseases, Faculty of Medicine and Pharmacy, Mohammed V University - Rabat, Morocco., Rabat, Morocco; 2National Institute of Health, Medical Genetics, Rabat, Morocco; 3Ibn Sina CHU, Rabat, Morocco; 4Ibn Sina CHU, Medical Genetics Unit, Rabat, Morocco

Background: Emery-Dreifuss muscular dystrophy (EDMD) is a neuromuscular disorder characterized by muscle weakness and atrophy associated with early tendon retractions and late cardiomyopathy. Several genes are associated with EDMD of which the two major ones EMD (EDMD1 X-linked recessive) and LMNA (EDMD2 and EDMD3 autosomal dominant and recessive respectively) are involved in 55% of patients. Diagnosis is based on the clinical triad but due to intra- and inter-familial heterogeneity, only molecular diagnosis by NGS allows to confirm with certainty EDMD by identifying the mutation in the causal gene.

Methods: We report clinical and molecular data of two unrelated Moroccan patients with EDMD phenotype by Next-Generation Sequencing.

Results: NGS sequencing of a custom gene panel lead to the identification of a deleterious hemizygous splicing mutation, NM_000117.3(EMD): c.399+1G > T in the EMD gene in one patient and a novel frameshift mutation in exon 9 of the LMNA gene, NM_170707.4(LMNA): c.1549_1550del in the other patient. Carrier status of the EMD mutation was investigated in several relatives at risk.

Conclusion: We emphasize the value of NGS in EDMD for a precise molecular diagnosis, a proper clinical management of patients and appropriate genetic counseling of families.

Conflict of Interest: None declared

EP12.022 Homozygous variant in NRDC gene in two siblings with developmental delay and seizure

Fatemeh Fatehi 1, Zeinab Ghorbanoghli1, Mahdieh Kooshki1, Shima Zamanian Najafabadi1, Khadijeh Noudehi1, Mina Makvand1, Hossein Najmabadi1;2, Ariana Kariminejad1

1Kariminejad-Najmabadi Pathology & Genetics Center, Tehran Iran; 2Genetics Research Center, University of Social Welfare & Rehabilitation Sciences, Tehran, Iran

Background: NRDC gene encodes a protein which interacts with heparin-binding EGF-like growth factor and plays a part in cell migration and proliferation. Animal studies showed progressive neurodegenerative disorder with motor impairment and neuronal defects.

Phenotype related to NRDC gene mutation has only been reported in a 15-year-old boy. The patient had developmental delay in the first year of life with hypotonia, absent speech and seizures. Optic atrophy, microcephaly and progressive cortical and cerebellar atrophy on brain imaging was reported. Similar phenotype was reported in the sibling who died at 16 months of age. However, the contribution of this phenotype with NRDC gene is yet to be confirmed.

Methods: Whole-exome sequencing (WES) was performed in one patient with neurodevelopmental delay and seizure. Thereafter, we performed segregation study on the entire family.

Results: We identified a homozygous likely pathogenic frameshift deletion in exon 15 of NRDC gene. The older sister with similar phenotype was homozygous, and the parents and healthy sister were heterozygous for this variant. The two sisters aged 17 and 12, showed developmental delay, microcephaly, hypotonia, speech impairment and seizure.

Conclusion: This is the second report of a family of two siblings with neurodevelopmental delay and mutation in NRDC gene. Further studies are needed to confirm the contribution of this phenotype with NRDC gene.

Grants: There are no grants to disclose

Conflict of Interest: None declared

EP12.023 Patients with idiopathic dry cough: suitability of incluiding CANVAS disease in the differential diagnosis

María Fenollar-Cortés 1, Vicente Gajate-García2, Clara Herrero-Forte1, Raluca Oancea-Ionescu1, Carmen Cotarelo-Pérez1, Laura García-Reguero1, Juan Luis Rodríguez-Hermosa3, alejandro horga2

1Hospital Clínico San Carlos, Unidad de Genética Clínica. Instituto de Medicina del Laboratorio., Madrid, Spain; 2Hospital Clínico San Carlos, Servicio de Neurología, Madrid, Spain; 3Hospital Clínico San Carlos, Servicio de Neumología, Madrid, Spain

Background/Objectives: CANVAS (Cerebellar Ataxia, Neuropathy, Vestibular Areflexia Syndrome) is a slowly progressive neurological disorder that begins in adulthood, clinically characterized as cerebellar ataxia, sensory neuropathy (PNP), bilateral vestibulopathy and autonomic dysfunction, caused by the biallelic expansion of the pentanucleotide AAGGG in intron 2 of the RFC1 gene. Chronic spasmodic dry cough appears in 80% of patients as the first symptom, years (up to decades) before the rest of the cardinal symptoms. We present a study of the RFC1 gene in 13 patients undergoing follow-up in Pneumology for chronic idiopathic cough.

Methods: The 13 patients were selected by Pneumology and examined by Neurology to study the presence/absence of peripheral polyneuropathy. The study of the number of AAGGG, AAAGG, and AAAAG expansions in intron 2 of the RFC1 gene was carried out using PCR and TP-PCR.

Results: Three (23%) of the 13 patients were diagnosed with CANVAS disease, two of whom had mild PNP without symptoms noticed by patients.

Conclusion: Despite the limited series of patients studied, this work seems to indicate that, within the differential diagnosis of idiopathic dry cough, the study of the RFC1 gene should be considered to rule out CANVAS disease.

Age at diagnosis

Cough (years)

PNP

Results

56

12

no

Positive

80

4-5

no

Negative

40

10

no

Negative

60

>20

no

Negative

48

8

Mild

Positive

79

>50

no

Negative, carrier

64

4-5

no

Negative

74

7

no

Negative, carriesr

64

5

no

Negative

60

20

No

Negative

61

>20

Mild

Positive

59

>40

No

Negative

58

3

No

Negative

Conflict of Interest: None declared

EP12.024 Tracing The Genetic Basis of Hypotonia in Whole Exome Sequencing Data: Experinces of Trakya University Genetic Diagnosis Center

Selma Demir 1, Sinem YALÇINTEPE1, Veysel Öz2, Yasemin Karal2, Engin Atli1, Drenusha Zhuri1, Hazal Sezginer Guler1, Yelda Yalçın3, HAKAN GURKAN1

1Trakya University Faculty of Medicine, Department of Medical Genetics, Edirne, Türkyie; 2Trakya University Faculty of Medicine, Department of Pediatrics, Edirne, Türkyie; 3Van Education and Research Hospital, Department of Pediatrics, Van, Türkyie

Background/Objectives: Central and peripheral hypotonia is a common phenotype seen among the neonatal and pediatric patients. Hypotonia may be presented as isolated or it may be a part of a wide range of neurological or metabolic disorders, necessiating a multidisciplinary approach including genetic screening. In this study, we aimed to present the genetic analysis results of Whole Exome Sequencing (WES) of the patients directed to our center for genetic evaluation of hypotonia.

Methods: Our internal database records including 450 cases who underwent whole exome sequencing analysis for various reasons in our center between May 2021 and November 2023 were retrospectively searched. WES had been performed on the Illumina NovaSeq 6000 with the QIAseq Human Exome Kit, using the DNA isolated from peripheral blood samples. WES results of 16 patients (8 males and 8 females aged between newborn and 12 with hypotonia/neuromuscular developmental delay/muscle weakness were reviewed in the context of this study.

Results: In five cases, pathogenic/likely pathogenic (P/LP) variations were identified in PRUNE1, DPYD, SPAST, RYR1 and MECP2 genes. Variations of unknown clinical significance were identified in various genes, also including genes whose anomalies may be associated with hypotonia, were identified in in nine cases.

Conclusion: P/LP variations were determined in 31,25 % of the patients. The relative high frequnecy of P/LP variations and their distribution among different genes supports the importance of WES as a feasible tool in the assesment of newborn and pediatric patients with hypotonia.

Grants:None

Conflict of Interest: None declared

EP12.025 2024 update of the National French consensus on gene lists for the diagnosis of muscular diseases using high-throughput sequencing

Emmanuelle Pion1, Mireille Cossee2;3, Valerie Biancalana4, Cecile Acquaviva5, Céline Bouchet6, Julien Fauré7, Roseline Froissart5, France Leturcq8, Rita Menassa9, Corinne Metay10, Laurence Michel-Calemard9, Emmanuelle Campana-Salort11, Juliette Nectoux8, François Petit12, John Rendu7, Pascale Richard10, Damien Sternberg13, Sandrine Vuillaumier-Barrot6, Shahram Attarian11, Svetlana Gorokhova14, Martin Krahn 14

1FILNEMUS, Laboratoire de génétique moléculaire, CHU de Montpellier, France; 2Laboratoire de Génétique Moléculaire, CHU de Montpellier, Montpellier, France; 3PhyMedExp, Physiologie et Médecine Expérimentale du cœur et des muscles, Université de Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France; 4Laboratoire de Diagnostic Génétique, CHRU de Strasbourg, Strasbourg; 5Service de Biochimie et Biologie Moléculaire, UM Maladies Héréditaires du Métabolisme, Centre de Biologie et de Pathologie Est, CHU de Lyon, Bron, France; 6Biochimie et département de génétique, Hopital Bichat-Claude Bernard CHU Paris Nord-Val de Seine, Paris, France; 7Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, Grenoble, France; 8Service de médecine génomique des maladies de système et d’organe, DMU Biophygen, Hopital Cochin, APHP, Université de Paris, Paris, France; 9Service de biochimie et biologie moléculaire, UM Pathologies Neurologiques, Musculaires et Cardiaques, Centre de biologie et pathologie Est, CHU de Lyon HCL, Bron, France; 10UF Cardiogénétique et Myogénétique Moléculaire et Cellulaire, Centre de Génétique Moléculaire et Chromosomique, Hopitaux Universitaire Pitié-Salpétrière, APHP, Paris, France; 11Service de Maladies neuromusculaires et SLA, Hôpital la Timone, APHM, Marseille, France; 12Laboratoire de Génétique Moléculaire Service de Biochimie et Hormonologie, Hôpital Antoine Béclère, APHP, Clamart, France; 13Service de Biochimie Métabolique, Hopitaux Universitaires Pitié Salpétrière, APHP, Paris, France; 14Marseille Medical Genetics, U1251, Département de génétique médicale, Hopital Timone Enfants, INSERM, Aix-Marseille Université, APHM, Marseille, France

Background/Objectives: In 2018, the French National Network for Rare Neuromuscular Diseases (FILNEMUS) established a high-throughput sequencing (HTS) diagnostic strategy for muscle-diseases, based on 13 consensus gene lists according to phenotypic and/or histological entry-diagnosis groups, implemented at the national level.

Following evolutions in gene-disease associations, and the discovery of novel genes, an updating process has been conducted in 2023.

Methods: In consultation with all FILNEMUS molecular diagnosis laboratories, we carried out a literature search to identify newly reported genes. Gene-disease association were assessed/updated for all new and previously included genes among the national FILNEMUS gene lists, using data from the GeneCC consortium. Data on most frequently implicated genes were collected at the national level.

Results: The FILNEMUS gene lists were updated based on gene-disease associations. Using data collected from all FILNEMUS molecular diagnosis laboratories, we set up an additional gene list ("major genes list"), to enable a rapid response excluding/confirming the most frequently implicated muscle-disease genes in France. This strategy is combined with the option of continuing with the analysis of an extended list (grouping all genes from the different FILNEMUS muscles-diseases gene lists), or proceeding directly to genome analysis on the French national whole-genome sequencing platforms (Plan France Médecine Génomique 2025 - PFMG2025).

Conclusion: Through its 2024 updated version, the National French consensus on gene lists for the diagnosis of muscular diseases using HTS will help avoiding unnecessary analysis of variants of uncertain significance, and reduce the turn-around time for genetic results and diagnostic odyssey, in coordination with the French national whole-genome sequencing platforms (PFMG2025).

Conflict of Interest: None declared

EP12.027 A previous unreported deleterious variant in FAM111B in a four-generation kindred reveals diverse degree of musculotendinous, pulmonary and pancreatic manifestations

Mar Costa-Roger 1;2, Ana Maria Cordero1;2, Ivon Cuscó1;2;3, Elena García-Arumí1;2, Eduardo Tizzano1;2

1Vall d’Hebron Hospital Universitari, Department of Clinical and Molecular Genetics, Barcelona, Spain; 2Vall d’Hebron Institut de Recerca (VHIR), Medicine Genetics Group, Barcelona, Spain; 3Present address: Hospital de la Santa Creu i Sant Pau, Genetics Department, Barcelona, Spain

Background/Objectives: Pathogenic variants in the FAM111B gene (OMIM *615584) have been described in patients with POIKTMP (Poikiloderma, hereditary fibrosis, tendon contracture, myopathy and pulmonary fibrosis) (OMIM #615704) with an autosomal dominant inheritance pattern. We report a four-generation kindred from Italian-Argentinian origin with 9 affected patients who showed no dermatological manifestations and a diverse degree of musculotendinous, pulmonary and pancreatic manifestations.

Methods: Patients have been followed by years by several specialists without a clear diagnosis. After genetic consultation, DNA from peripheral blood was obtained and exome analysis was performed.

Results: The main features found in the members of this family consist of 1) distal tendon contractures in extension, particularly in biceps brachii and carpal extensors; 2) progressive proximal and distal muscle weakness; 3) progressive pulmonary fibrosis; 4) pancreatic insufficiency; 5) pancreatic carcinoma. Dermatologic features, such as poikiloderma, were not observed or referred.

The variant c.1887C>A: (p.Phe629Leu) in FAM111B was identified predicting a change from Phenylalanine to Leucine in amino acid 629. This variant is not present in population databases (gnomAD) but is reported in the database related to clinical phenotype as probably pathogenic in a single case (ClinVar). The in silico pathogenicity predictor algorithm (AlphaMissense), which considers the conservation and structure parameters of the protein, predicts it as a deleterious variant.

Conclusion: This family showed diverse multisystemic fibrotic manifestations due to a previous unreported FAM111B dominant likely pathogenic variant. Long-term follow up to document evolution and prevention of pulmonary and pancreatic complications should be provided.

Grants:

Conflict of Interest: None declared

EP12.028 The newborn screening results for SMA 5q in the Russian Federation for 2023

Maria Akhkiamova 1, Olga Shchagina1, Aleksander Polyakov1, Sergey Kutsev1, Sergey Voronin1, Andrey Marakhonov1

1Research Centre for Medical Genetics, Moscow, Russian Federation

Consortium: none.

Background/Objectives: Newborn screening for SMA 5q in Russia: the first-year results.

Methods: EONIS ™ – PerkinElmer* system, TK-SMA JSC Generium, nescreen SMA/TREC/KREC, DNA technology, SALSA MLPA Probemix P060-B2, Sanger sequencing.

Results: The Reference Center received 250 blood samples of newborns with a positive result of the SMA screening test from 1,100,000 newborns born in 2023. 0 copies of the SMN1 gene were found in 48% of 250 people. At the same time, 2 copies of SMN2 were found in 35.3% of newborns, 3 copies in 26%, 4 copies in 26%, 5 copies were found in one newborn (0.9%), chimeric genotypes were observed in 11.8%. The incidence of the disease is 1 in 11 000 people. The incidence of this disease according to NBS results in different regions of the Russian Federation ranges from 2.95 to 7.99 per 100,000 people. Currently, we have information about 4 newborns who had a negative screening result and showed symptoms of SMA 5q. Their diagnosis was confirmed by molecular genetic methods. All had 0 copies of SMN1 and 2 copies of SMN2. In addition, accidental findings were found in the field of extraction of diagnostic oligonucleotides. To date, 94% of newborns with SMA 5q detected during NBS have received pathogenetic or etiotropic therapy.

Conclusion: NBS has shown its high effectiveness for early detection of SMA 5q in the Russian Federation.

Grants: none.

Conflict of Interest: None declared

EP12.029 Rare genomic structural variants identified through extensive Next Generation Sequencing approaches

Alice Margutti1, Martina Mietto1, Vittoria Nagliati1, Maria Sofia Falzarano1, Gaetano De Feo1, Giusy Cavarretta1, Fernanda Fortunato1, Marcella Neri1, Francesca Gualandi1, Alessandra Ferlini1, Rita Selvatici 1

1University Hospital S. Anna Ferrara, Unit of Medical Genetics, Department of Medical Sciences and Department of Mother and Child, Ferrara, Italy

Background: Dystrophinopathies are a group of X-linked genetic disorders caused by DMD gene mutations. Next-generation sequencing (NGS) has greatly improved our ability to identify rare cases with complex rearrangements. Here we report 3 patients with DMD mutations identified exclusively by NGS.

Methods: The following NGS approaches were used: i) targeted NGS-panel for Patient-1; ii) Whole Exome Sequencing (WES) for Patient-2; iii) Long-Read WGS (LR-WGS) for Patient-3.

Results: Targeted NGS-panel in Patient-1 identified the deletion c.6439-59019_6459del of approximately 59 Kb extending between intron 44 and the first 21 nucleotides of exon 45. RNA analysis highlighted skipping of exon 45. WES analysis in Patient-2 identified the total absence of coverage of the entire DMD gene and several contiguous genes. The extensive deletion of about 10Mb in the chrXp21.3p21.1 region was confirmed by array-CGH. LR-WGS performed in Patient-3 identified a large inversion of almost 15Mb involving intron 1 of the NHS gene and intron 44 of the DMD gene, thus justifying the complex phenotype of DMD and cataract. RNA analysis using FluiDMD-card addressed the identification of the breakpoint in intron 44 of the DMD gene.

Conclusion: NGS has revolutionized the field of genetic diagnostics by allowing the simultaneous analysis of multiple genes or even the entire exome or genome. The benefits of NGS in DMD diagnosis not only include the identification of common mutations but also complex mutations that are often difficult to detect using traditional methods.

Grants: EU project Solve-RD Grant Agreement number: 779257. Several authors are members of the ERN-NMD-Project-ID N°870177 and ERN-ITHACA [EU F.P.A. ID:3HP-HP-FPA ERN-01-2016/739516].

Conflict of Interest: None declared

EP12.030 Clinical characteristics of patients with oculopharyngeal muscular dystrophy (OPMD) in Israel

Lior Greenbaum 1;2, merav ben-david1;2, alex zvulunov1;3;4, amir dori1;2

1Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; 2Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; 3The non-profit organization for promotion of health and cure of OPMD, Israel; 4Reichman University, Herzliya, Israel

Background/Objectives: Oculopharyngeal muscular dystrophy (OPMD) is a late-onset autosomal dominant muscular dystrophy, caused by (GCN) repeat expansion in the PABPN1 gene. OPMD has an increased frequency in Israel, due to founder mutation in Bukharian Jews.

Methods: We describe the genetic, clinical and electrodiagnostic characteristics of OPMD patients in Israel. Since January 2022, patients with genetic confirmation of OPMD were assessed at our neuromuscular clinic in a tertiary referral center, and enrolled to the national Israeli OPMD registry (IsrO-PRD).

Results: We included 73 Jewish patients (37 males, 50.7%), and 66 (90.4%) were of Jewish Bukhara descent. Sixty-eight (93.2%) were heterozygotes and 2 (2.7%) were homozygotes for 13 (GCN) repeats, 1 (1.4%) homozygote for 11 (GCN) repeats and 2 (2.7%) were heterozygotes for longer repeat expansions. Symptoms onset was at the age of 51.5 ± 7.7 years (range 35-69) years. The first presenting symptom was dysphagia in 28 patients (38.6%), ptosis in 27 (37%) and both appeared in parallel in 15 (20.5%). At the time of clinical evaluation, proximal muscle weakness was detected in 47 patients (64.4%), which presented at age 58.6 ± 6.9 (range 35-76). Electrodiagnostic studies showed early recruitment or small motor units in 14/24 (41.6%) of patients and fibrillation potentials in 9/24 (37.5%).

Conclusion: The majority of OPMD patients in Israel are from Bukharian descent, commonly heterozygous for the 13 (GCN) repeats allele. Our study provides relevant data about the clinical and electrophysiological features, which is comparable to samples of OPMD patients from other populations.

Grants: The non-profit organization for promotion of health and cure of OPMD, Israel.

Conflict of Interest: None declared

EP12.031 A noval KIF1A mutation associated with NESCAV syndrome

KADRİYE FARYAP 1, Ayse Busra Akalin2;3, Elif EROĞLU ŞİMŞEK4, Ibrahim Akalin4

1Yıldız Technical University, Molecular Biology and Genetics, İstanbul, Türkyie; 2Bahcesehir University, Medicine, Türkyie; 3, İstanbul; 4Metagentech Center for Genetic Diseases, İstanbul, Türkyie

Background/Objectives: Muscular dystrophies are hereditary myogenic growths that are associated with progressive muscle wasting and weakness of variable distribution and severity. It begins in childhood and causes symptoms such as muscle weakness, deformities in the joints and spine, and difficulty in movements. NESCAV Syndrome is an autosomal dominant neurodegenerative disease with symptoms starting in infancy or early childhood. Its major features are intellectual disability, speech delay and learning disability.

Methods: A 6-year-old male patient was born to third degree consanguinity with normal route without cyanosis. His initial symptoms were inability to walk and talk. He had eye contact. Upon clinical examination contracture in the knees, strabismus, hyperactivity, long philtrum, bulbous nose and wide thumb with short nails were detected. When he tried to stand trembling in the legs were observed. He was operated for cataract on his right eye. Whole exom sequencing was performed for molecular diagnosis.

Results: WES analyses revealed pathogenic/likely pathogenic c.223C>T(p.Arg75Trp) heterozygous mutation in KIF1A gene, which is responsible for autosomal dominantly inherited “NESCAV [MIM#614255]” syndrome. Differential diagnosis were skeletal dysplasia, neurodevelopmental defects and mental retardation.

Conclusion: KIF1A gene mutations were responsible for autosomal dominantly inherited “NESCAV [MIM#614255]” syndrome. Though the mutation previosly defined in the ClinVar database and it is very rare and only reported in autism spectrum disorder, ADHD with intelligence deficit. We report this mutation for the first time to be associated with NESCAV syndrome.

Grants:

Conflict of Interest: None declared

EP12.032 Prune syndrome diagnosed in a newborn with congenital hypotonia: homozygous missense variant in PRUNE1 gene responsible of clinical phenotype.

Graciela Pi 1, ANGEL ZUNIGA2, MLuisa Bueno-Sanchez1, Joana Ortega Sanz1

1HU La Ribera, PEDIATRÍA, Alzira (Valencia), Spain; 2HU La Ribera, GENÉTICA, Alzira (Valencia), Spain

Background: PRUNE syndrome, or neurodevelopmental disorder with microcephaly, hypotonia, and variable brain anomalies (OMIM#617481), is a new rare autosomal recessive neurodevelopmental disease that is caused by homozygous or compound heterozygous mutation in the PRUNE1 gene. As this is a newly syndrome, there are scarce data in the literature about clinical and radiological phenotypes.

Results: In this case report, we describe clinical, radiological, and molecular findings of a 4-month-old girl with severe hypotonia, low weight at birth and dysmorphic traits. First suspicion was a metabolic vs neuromuscular disease by deterioration and low increase in body weight. Subsequent studies rule out underlying metabolic disease.

MRI result: isolated diffuse cerebellar involvement as a marker of infectious/parainfectious process, rather than metabolic disease. At five months severe neurodevelopmental delay: axial hypotonia, no head support, no eye contact, no social smile. Distal joint contractures. West syndrome.

Targeted exome sequencing using genomic DNA from the patient’s specimen, the exonic regions, and flanking splice junctions of the genome were sequenced by NGS. Patient was homozygous for a missense variant of PRUNE1 gene classified as likely pathogenic following ACMG criteria. Consanguineous healthy parents were heterozygous for this variant. Most of the previously reported cases have homozygous missense variants (90%).The high rate of homozygous mutations can be explained by the elevated rate of consanguinity in the published cases.

Conclusions: Clinicians must consider PRUNE syndrome in any child who presents with dysmorphic features, profound developmental delay, progressive microcephaly, central hypotonia, peripheral spasticity, delayed myelination, brain atrophy, and a thin corpus callosum.

Conflict of Interest: None declared

EP12.033 Determination of the length of the CAG repeat in the androgen receptor gene among Ukrainian patients with suspected Kennedy’s disease.

Bohdan Tretiak 1, Danuta Zastavna1, Veronika Hamkalo1, Khrystyna Bakum1, Hayane Akopyan1

1State Institution «Institute of Hereditary Pathology of the National Academy of Medical Sciences of Ukraine», Diagnostics of hereditary pathology, Lviv, Ukraine

Background/Objectives: Spinobulbar muscular atrophy (SBMA) Kennedy syndrome is an X-linked recessive disease characterized by progressive muscle weakness. The diagnosis of SBMA is confirmed by molecular genetic testing in men with >39 CAG repeats in the AR gene. The aim of the work was to study the expansion of CAG repeats in the AR androgen receptor gene in individuals with suspected SBMA.

Methods: Clinical and genealogical, method of differential diagnosis, DNA isolation and purification, molecular genetic: polymerase chain reaction, electrophoresis in agarose gel.

Results: A molecular genetic study of trinucleotide CAG-repeats expansion in androgen receptor gene in 32 people with suspected Kennedy’s syndrome was performed. In 5 probands of the study group, 38 CAG repeats (the upper limit of the norm) were established and in 27 examined patients, the number of CAG repeats did not exceed 37 (the norm). Among the examined group of patients, was found a family in which three men had 49 CAG repeats in the AR gene, which confirmed the presence of Kennedy’s syndrome. Also, two isolated cases with 60 CAG repeats were found in the study group of men, which proved the presence of SBMА.

Conclusion: Kennedy’s syndrome is a rare X-linked recessive disease that requires the development of specific biomarkers to clarify the pathogenic process and facilitate early diagnosis.

Grants:

Conflict of Interest: None declared

EP12.035 The utility of whole exome sequencing in accurate genetic diagnosis of neuromuscular disorders in Iran

Negar Molaei 1;2, Fatemeh Ahangari1, Maryam Beheshtian1, Parnian Alagha1;2, shima Dehdahsi1, Mahsa Fadaie1, Mehri Ashki1, Zohreh Elahi1, Maryam Mozaffarpour1, Parishad Saei1, Fatemeh Fatehi1, Khadijeh Noudehi1, Shima Zamanian Najafabadi1, Kimia Kahrizi2, Ariana Kariminejad1, Hossein Najmabadi1;2

1Kariminejad - Najmabadi Pathology & Genetics Center, Tehran, Iran; 2Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran

Background/Objectives: Neuromuscular disorders (NMDs) recognized as clinically and genetically heterogeneous diseases include multiple conditions such as myopathy, motor-neuron disorder, neuromuscular-junction disorders, and neuropathy. Involvement of more than 600 genes associated with this condition, complicates the process of applicable genetic diagnosis. However, the advent of next generation sequencing (NGS) paved the way for identifying disease-associated genes. Thus, This study aimed to investigate the genetic causative variants among 1190 patients diagnosed with NMDs.

Methods: Whole exome sequencing (WES) was conducted on 1190 NMD patients who were referred to our genetic diagnosis center in the past 10 years. Clinical features and diagnoses were validated by clinicians and candidate disease-causing variants were confirmed by Sanger sequencing

Results: A genetic diagnosis was established for 572 patients (~48%), with CAPN3, DYSF, DMD, RYR1, LAMA2, SGCA for myopathies, SPG11, SOD1 for motor-neuron disorders, CHRNE, COLQ for neuromuscular-junction disorders and GDAP1, MME, SH3TC2 for neuropathies being the most frequently reported genes, which have been shown to be related to either appropriate nervous system or skeletal muscle function. Disease onset varied from neonatal to adulthood and the parental consanguinity rate in our studied samples was 66%. Notably the diagnostic yield for inbred families were 53% compared with outbred families with diagnostic rate of 38%.

Conclusion: Utilizing WES, was helpful to facilitate rapid and accurate diagnosis nearly half of the investigated cases in line with diagnostic yields of NGS reported in previous literatures. It supports the efficiency of NGS for identifying the underlying genetic etiology of heterogeneous inherited conditions.

Conflict of Interest: None declared

EP12.036 B3GALNT2-Related Dystroglycanopathy: Expanding the Phenotype

Zeinab Ghorbanoghli 1, Fatemeh Ahangari1, Fariba Afroozan1, Mahtab Ramezani2, Hossein Najmabadi1;3, Ariana Kariminejad1

1Kariminejad-Najmabadi Pathology & Genetics Center, Tehran Iran; 2Neuromuscular Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran; 3Genetics Research Center, University of Social Welfare & Rehabilitation Sciences, Tehran,Iran

Background: Muscular dystrophy-dystroglycanopathies (MDDGs) are a well-recognized subtype of congenital muscular dystrophy. MDDGA11 is caused by biallelic pathogenic variants in B3GALNT2 and is characterized by wide range of symptoms including muscle weakness, intellectual disability, and abnormalities in the brain and eyes. To date, 16 patients have been reported with biallelic mutation in B3GALNT2 gene and clinical features of MDDG. Remarkably, two recent studies reported 7 individuals with psychomotor and speech delay, epilepsy, and behavioural problems, but no signs of muscular dystrophy and eye problems.

Methods: Using whole-exome sequencing in one patient with an unexplained neurodevelopmental delay and bilateral hearing loss, we identified a homozygous mutation in B3GALNT2 gene. Thereafter, we performed segregation study by sanger sequencing on the family.

Results: Three siblings with mild-to-severe intellectual disability and bilateral hearing loss were homozygote in the B3GALNT2 gene for a variant defined as chr1:235643447G > A; c.574C>T (p.Arg192Cys) in exon 5. Two unaffected siblings and parents were heterozygous for this variant. Physical examination of the affected sisters revealed abnormality of gait with various severity and therefore, they were referred to a neurologist and ophthalmologist for further evaluation. Electromyography and brain MRI were performed on one sister which showed muscular dystrophy (predominantly distal) and abnormal foci in cerebral hemispheres and mild atrophic changes in cerebellar hemispheres, respectively. Eye examination was unremarkable.

Conclusion: We report three sisters with homozygous mutation of B3GALNT2 gene with mild to severe intellectual disability, distal muscle weakness and hearing loss. This study further expands on the heterogenous phenotype of MDDGs.

Grants: There are no grants to disclose.

Conflict of Interest: None declared

EP12.037 Genetic variability in inflammation pathway influences clinical course and treatment response in patients with spinal muscular atrophy

Maruša Barbo1, Tanja Blagus1, Vita Dolzan1, Blaž Koritnik2;3, Metka Ravnik-Glavač 4

1Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana; 2Institute of Clinical Neurophysiology, University Medical Centre Ljubljana, Slovenia; 3Department of Neurology, Faculty of Medicine, University of Ljubljana, Slovenia; 4Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia

Background: Several studies indicated that neuro-inflammation as well as systemic inflammation of varying severity are present in patients with spinal muscular atrophy (SMA).

Methods: We used allele-specific PCR to genotype 54 SMA patients and 163 healthy controls for polymorphisms in nine inflammatory process genes: GSTP1, IL1B, IL6, MIR146A, TNF, NLRP3, CARD8, and NOS1. Genetic data were statistically analysed for associations with the risk and treatment response of SMA disease.

Results: Our study showed that GC heterozygotes (OR = 2.813; 95% CI = 1.270–6.234; p = 0.011) and CC homozygotes (OR = 3.145; 95% CI = 1.269–7.793; p = 0.013) for the IL6 polymorphism rs1800795 had almost threefold and more than threefold greater odds of SMA, respectively, compared to GG homozygotes. In contrast, heterozygotes (GA) and carriers of at least one polymorphic A allele (GA + AA) for TNF polymorphism rs1800629 (p = 0.031 and p = 0.048, respectively) have reduced odds of developing SMA. In addition, TNF rs1800629 heterozygotes (GA) were found to be associated with an older age at symptom onset (p = 0.033). Concerning treatment respond, IL6 rs1800795 heterozygotes (GC) had lower odds for responding to treatment (p = 0.036). In contrast, CC homozygotes for MIR146A rs2910164 had more than 16 times greater odds for responding to treatment than GG homozygotes (p = 0.028).

Conclusion: Genetic variations in IL6, TNF and MIR146A genes influence the SMA course and reveal potential biomarkers for predicting treatment response.

Grants: Slovenian research agency (No. P1-0170, M.B. PhD thesis grant).

Conflict of Interest: None declared

EP12.038 A closer look at FSHD molecular diagnosis via molecular combing data

Manar Kaptan 1, Beyza Yavuzcan1, Serpil Eraslan2, Şahin Avcı2, İlker Eren3;4, gulshan yunisova3;5, Mehmet Demirhan3;4, Piraye Oflazer3;5, Hülya Kayserili1;2;3;6

1Koç University, Graduate School of Health Sciences, Istanbul, Türkyie; 2Koç University Hospital, Genetic Diseases Evaluation Center, Istanbul; 3Koç University Hospital, Muscle Diseases Center, Türkyie; 4Koç University Hospital, Orthopedics and Traumatology Department, Istanbul, Türkyie; 5Koç University School of Medicine, Neurology Department, Istanbul, Türkyie; 6Koç University School of Medicine, Medical Genetics Department, Istanbul, Türkyie

Background/Objectives: FSHD1 is caused by the contraction of the D4Z4 repeat array on 4q35. In healthy individuals the array has 11 to 150 repeats, in patients, it is reduced to 1-10 repeats. There is also a highly homologous array on 10q26. Molecular diagnosis is very challenging, relying on the identification of exact repeat number, polymorphic constitution, and homologous region distinction, as well as determining mosaic cases and those with unusual rearrangements. We aimed to perform molecular diagnosis of FSHD and interpretation of results in the context of the phenotype using Molecular combing (MC) as a very sensitive hybridization-based technique.

Methods: HMW-DNA isolation, MC slide preparation, and hybridization-wash-stain procedures were performed in 86 Turkish index patients. Scanned slide images were analyzed by a specific software.

Results: FSHD1 diagnosis was confirmed in 82.5% of patients (n = 71/86) having 1-10 RUs. 15 patients did not reveal contracted alleles. 3% of cases were mosaics and two patients (2.3%) had complex rearrangements. Comparison of RUs size and the age of onset, and disease severity amongst patients showed that most of our data is in concordance with currently published data.

Conclusion: More detailed molecular analysis results will indeed enable us to approach genotype-phenotype correlation on a more solid ground. This is the largest molecular diagnosis cohort presented from Turkey’s first FSHD Molecular Research & Diagnostics Unit so our expanding FSHD cohort will yield more precise results in the future, enhancing our understanding of the disease manifestation and progression within our population.

Conflict of Interest: None declared

EP13 Multiple Malformation/Anomalies Syndromes

EP13.001 Whole-exome sequencing for diagnostic investigation of 249 children from the Brazilian public health system (SUS)

Bruno Stephan1, Juliana Leao1, Debora Pegos1, Julia Abrahão1, Vitoria do Val1, Matheus Castro1, Rachel Honjo1, Debora Bertola1, Hiromi Aoi2, Saida Ken2, Rie Seyama2, Yuri Uchiyama2, Naomichi MATSUMOTO2, Chong Kim 1

1Instituto da Criança e do Adolescente - ICr HCFMUSP, Genetics Unit, São Paulo, Brazil; 2Yokohama City University School of Medicine, Department of Human Genetics, Yokohama, Japan

Background/Objectives: Despite being a broad-range test and key tool for diagnosis and genetic counseling of rare diseases, whole-exome sequencing (WES) remains overall absent inside the Brazilian Public Health System (SUS). Our study describes the major clinical and molecular findings within a sample of Brazilian children with intellectual disability or congenital anomalies.

Methods: Selected patients with suspicion of unknown genetic etiology were evaluated and followed-up at our Genetics service in São Paulo. Blood samples from the probands and their parents were submitted to WES, analyzed at Yokohama University in Japan.

Results: Between February 2017 and April 2022, from a total of 249 probands, we detected 118 positive cases (47,3%) of genetic rare disorders. The majority of variants were found in over a hundred different genes, with de novo occurrence confirmed in at least 38 families; some of these had never been described before, such as the mutations in BRF1 and SLC35A3. However, recurrence in rare genes did occur, with 4 unrelated patients presenting pathogenic variants in SCN1A. Though less frequent, autosomal recessive and X-linked disorders were present in 27 and 6 families, respectively; another 8 cases were secondary to copy-number variants (CNVs). Additionally, relevant variants of uncertain significance (VUS) were present in 5 families. Nevertheless, 126 children (50,6%) received negative results and will be considered for reanalysis.

Conclusion: This is the largest cohort of trio-analysis WES in children without previous diagnostic confirmation exclusively from SUS; positivity rate was near 50%, aligned with diagnostic yield observed around the world.

Grants: AMED (JP23ek0109674+JP23ek0109549+JP23ek0109617)

Conflict of Interest: None declared

EP13.003 A novel EHMT1 variant in a Turkish case with Kleefstra Syndrome

aysenur ersoy 1, Bilgen Bilge Geçkinli1, Zeynep Baser1, onur hanoglu1, cekdar kapazan1

1Marmara University Pendik Research And Education Hospital, medical genetics, istanbul, Türkyie

Background: Kleefstra syndrome 1 (#610253) is an autosomal dominant syndrome of epilepsy, developmental delay and dysmorphism. Half of the cases is caused by chromosome 9q34.3 deletions and half by point mutations in EHMT1 gene which encodes for a histone methyltransferase.

Methods: DNA was isolated from the patient’s peripheral blood and Clinical Exome Sequencing (CES) was performed via next-generation sequencing (Illumina Nextseq 500).

Results: The patient is a 13-year-old male who was referred to our clinic for dysmorphism, strabismus, mental retardation and epilepsy. Strabismus was detected when he was 40-days-old. He had congenital hip dislocation. He had been treated with valproic acid due to attacks of screaming, audio or visual hallucinations,urinary incontinence and an abnormal EEG. His neurodevelopmental milestones were behind his age. Height and weight parameters were 3rd percentile and head circumference was 50-75 percentile. He had coarse face, hypertelorism, long palpebral fissures, bulbous nasal tip, carp-shaped mouth, thin upper lip, wide spaced teeth, low-set posteriorly rotated ears. Ecocardiography showed dilation in aortic root and ascending aorta. He was heterozygous for EHMT1 (NM_024757) c.2908 A > G (p.Lys970Glu) variant. The variant is not reported on ClinVar database and in silico tools unanimously agree on the disease causing property of the variant. Segregation analysis of the parents is planned.

Conclusion: Kleefstra syndrome is known to be a deletion syndrome, but half of the cases are the result of point mutations. More research is needed to establish whether there are genotype phenotype correlations of these two distinct mechanisms.

Grants: None

Conflict of Interest: None declared

EP13.004 Copy number variation analysis of exome sequencing data readily discriminates distal 11p13 deletion syndrome from WAGRO in a girl with aniridia

Georgia Christopoulou 1, Stavros Bournazos1, Alexandra Fountoulaki2, maria kassotaki2, Ioannis Goniotakis2, ILIANNA MANIADAKI2, Pantelis Constantoulakis1, Eleftheria Papadopoulou2

1Genotypos M.S.A., Genetics and Genomics, Athens, Greece; 2University Hospital of Heraklion, Pediatrics, Heraklion, Greece

Background/Objectives: Congenital aniridia is a rare disease, with estimated incidence 1:64,000. Aniridia can occur either as an isolated malformation or as a part of a syndrome. The best-known syndromic form is WAGR (Wilms tumor, Aniridia, Genitourinary anomalies, Retardation) or WAGRO (plus Obesity), a contiguous genes deletion syndrome of the 11p13 region (encompassing the PAX6, WT1 and BDNF genes). We present a case of syndromic aniridia which etiology was deciphered by exome sequencing (ES) copy number variation (CNV) analysis.

Methods: A 5.5-year-old female was referred to a pediatrics hospital unit for the investigation of aniridia detected during routine ophthalmology examination. Besides aniridia, the patient presented developmental delay and increased body mass index for age and gender. Genitourinary ultrasound was normal. ES was applied on DNA extracted from peripheral blood: library preparation was performed with Twist Human Core Exome EF Multiplex Complete kit (TWIST Bioscience), sequenced on NextSeq-550/550 (Illumina). Bioinformatics analyses were conducted by SOPHiA DDM® bioinformatics pipelines. Confirmatory testing was performed by Cytogenetics Whole-Genome CytoScan 750K array (Affimetrix).

Results: ES revealed a ≈5Mbp 11p13 interstitial deletion including 17 OMIM genes among which, PAX6, ELP4 and BDNF but not WT1, thus leading to distal 11p13 deletion syndrome with low risk of Wilms tumor. The deletion was confirmed by arrayCGH.

Conclusion: In children with aniridia associated with mental retardation and obesity, it’s essential to exclude PAX6 deletions that extend to the WT1 gene, which put the patients at risk of developing Wilms tumor. ES consists a dynamic tool for this purpose.

Grants:

Conflict of Interest: None declared

EP13.005 Complex phenotype of 47,XXX/45,X mosaicism, Sotos syndrome and neurofibromatosis type 1

Ana-Maria Meašić 1;2;3, Katarina Vulin1;2;3, Mijana Kero4, Leona Morožin Pohovski1;2;3, Lara Samadan1, Adriana Bobinec1;2;3, Sanja Pejić Rosko5, Silvija Pušeljić6, Ljubica Odak1;2;3

1Children’s Hospital Zagreb, University of Zagreb, School of Medicine, Department of Medical and Laboratory Genetics, Endocrinology and Diabetology with daily care unit, Zagreb, Croatia; 2University of Zagreb School of Medicine, Scientific Centre of Excellence for Reproductive and Regenerative Medicine (CERRM), Zagreb, Croatia; 3European Reference Network – Intellectual disability, Tele Health, Autism and Congenital Anomalies (ERN-ITHACA); 4University Hospital of Split, Paediatric Diseases Department, Split, Croatia; 5Children’s Hospital Zagreb, University of Zagreb, School of Medicine, Department of Paediatric Neurology with daily care unit, Zagreb, Croatia; 6Osijek University Hospital, Department of Paediatric Neurology, Genetics, Endocrinology, Metabolic Diseases and Rheumatology, Osijek, Croatia

Background/Objectives: A complex clinical phenotypes are not so rare in the era of molecular genetics and represent a diagnostic challenge. We present a 5-year-old girl with neurodevelopmental delay, facial dysmorphia, macrocrania, overgrowth, multiple cafe-au-lait spots, axillary freckles, multiple neurofibromas, Lisch nodules, FASI lesions and optic gliomas. Her mother has multiple cafe-au-lait spots, axillary freckles, multiple neurofibromas and borderline intellectual functioning. Her father has frontal bossing, overgrowth and mild intellectual disability.

Methods: Cytogenomic analyses included chromosomal microarray (CMA), performed with Agilent SurePrint G3 Unrestricted CGH ISCA v2, 8x60K platform, and peripheral blood karyotype. Clinical exome sequencing (CES) was performed using TruSight One Sequencing panel.

Results: CMA results show X chromosome trisomy in 50% of the sample. Classic cytogenetic analysis revealed mosaic female karyotype: 47,XXX[86]/45,X[14]. CES analysis detected heterozygous missense variant in NSD1 gene c.6614A>G, p.(His2205Arg) classified as pathogenic.

Conclusion: Our patient shows a complex phenotype of mosaic chromosomopathy 47,XXX/45,X, Sotos syndrome and probably neurofibromatosis type 1. She inherited Sotos syndrome from her father. She and her mother fulfill clinical criteria for neurofibromatosis type 1, but we couldn’t provide genetic evidence for it yet.

The possibility of being affected with more than one genetic condition should be considered when clinical findings are incoherent with the primary found genetic diagnosis. A complete diagnosis improves clinical management and, of course, better explains complex phenotype.

Grants: This study was supported by CERRM, Republic of Croatia, and by the EU through ERDF, under grant agreement No. KK.01.1.1.01.0008, project „Reproductive and Regenerative Medicine - Exploring New Platforms and Potentials”.

Conflict of Interest: None declared

EP13.006 Diagnosis of Wolf-Hirschhorn syndrome combining CNV detection based on Whole Exome Sequencing (WES) data and conventional karyotype

Faidon-Nikolaos Tilemis 1, Nikolaos Marinakis1;2, Nikoletta Selenti1, Korina Karachristou3, Konstantina Katechi3, Konstantina Kosma1, Aphrodite Kampouraki1, Jan Traeger-Synodinos1

1Laboratory of Medical Genetics, St. Sophia’s Children’s Hospital, National and Kapodistrian University of Athens, Athens, Greece; 2Research University Institute for the Study and Prevention of Genetic and Malignant Disease of Childhood, St. Sophia’s Children’s Hospital, National and Kapodistrian University of Athens, Athens, Greece; 3A’ Neonatal Intensive Care Unit, St. Sophia’s Children’s Hospital, 11528 Athens, Athens, Greece

Background/Objectives: Wolf-Hirschhorn syndrome (WHS) is a rare genetic disorder characterized by distinctive craniofacial features (‘Greek warrior helmet’), severe prenatal/postnatal growth retardation, microcephaly, and developmental delay. In most cases WHS is caused by deletions on the short arm of chromosome 4 (from 1.4 to 30 Mb), while in the remaining cases it results from inherited (often maternal) unbalanced translocations.

Methods: A 1-month-old male was referred with severe intrauterine growth retardation (IUGR), abnormal facial shape, hypospadias and cryptorchidism. Following clinical evaluation, pre-test counselling and signed consent, WES and karyotype were ordered simultaneously. WES was performed with IDT xGen Exome Research v2 kit on Illumina NextSeq-500 system and bioinformatic analysis with VarSome Clinical platform, including CNV detection by ExomeDepth. Variants were classified following ACMG and ClinGen recommendations. Further investigation included conventional karyotype on the patient and his parents.

Results: A 20.8Mb heterozygous pathogenic deletion of the region 4p16.3-p15.31 (DEL:chr4:85622_20884332, hg19) and a 868.1Kb likely pathogenic duplication of the region 19p13.3 (DUP:chr19:281388_1149441, hg19) were identified by ExomeDepth. Subsequent conventional karyotype confirmed the deletion in the patient, and detected an unbalanced translocation between chromosomes 4 and 19 in his mother (46,XX,t(4;19)(p15.2;p13.3)).

Conclusion: Identification of a deletion in 4p16.3-p15.31 and a duplication in 19p13.3 region, resulting from unbalanced segregation of a reciprocal 4;19 translocation in the mother supported the WHS diagnosis. The implementation of WES reduces time to a definitive diagnosis of cases with severe phenotypes, as it enables detection of different types of causative genetic variants, which can be confirmed by other orthogonal methods.

Grants: None

Conflict of Interest: None declared

EP13.007 The first study of cardiac defects in italian Kleefstra syndrome cohort

federica gaudioso 1, Camilla Meossi1, Cristina Gervasini2;3, Valentina Massa2;3, Romano Tenconi4, giulietta scuvera1, Paola Marchisio5;6, camilla lucca7, Maria Iascone7, laura pezzoli7, Donatella Milani5

1Policlinico of Milan, Medical Genetics Unit, Milano, Italy; 2Università degli Studi di Milano, Department of Biomedical Sciences for Health, Milano, Italy; 3University of Milan, “Aldo Ravelli” Center for Neurotechnology and Experimental Brain Therapeutics, Milano, Italy; 4Università di Padova, Clinica Ostetrica e Ginecologica Padova, Unità di Genetica Clinica, Padova, Italy; 5Policlinico of Milan, Milano, Italy; 6University of Milan, Department of Pathophysiology and Transplantation, Milano, Italy; 7ASST Papa Giovanni XXIII Hospital, Laboratory of Medical Genetics, Bergamo, Italy

Background/Objectives: Kleefstra syndrome type 1 (KS1) is a rare genetic disorder caused by a chromosome 9q34.3 deletion or a pathogenetic variant in EHMT1 gene. The main clinical features are neurodevelopmental delay and dysmorphisms, possibly associated with seizures, sleep disorders, skeletal and behavioural anomalies. Cardiac defects are also reported, including structural heart diseases and/or arrhythmias. Deepening the knowledge on the diagnosis and prevalence of cardiovascular issues we aim to improve the management of KS1.

Methods: 34 patients, belonging to the first KS1 Italian cohort, were admitted to our Hospital. Clinical data and EKG/cardiac ultrasounds were collected, splitting structural anomalies from arrhythmias. A patient with long QT syndrome underwent a NGS (Next Generation Sequencing) panel test.

Results: 21 patients showed a structural cardiac disease, 3 a frequency/rhythm anomaly and 4 both the conditions, including mild and non-specific structural and electrical anomalies. The patient with long QT syndrome tested negative to a specific NGS panel test.

Conclusion: Our results are consistent with recent studies (Vasireddi et al. 2024) underlying the underestimated prevalence of rhythm anomaly even in the absence of structural heart disease. Arrhythmias are alarming emerging signs in these patients, in which clinical investigations are further hindered by the language defect, so specific diagnosis of KS1 and further studies of the cardiac rhythm are a necessary steps to assure a correct care.

Grants: This study is funded by biomedical research regional foundation(FRRB, Fondazione Regionale Ricerca Biomedica) of Lombardia district(Drop by Drop, ID 3441133).

Conflict of Interest: None declared

EP13.008 Using the NGS method to diagnose CASGID syndrome

Vita Antsupova 1, Iryna Lastivka2, Ludmila Khlunovska2, Anastasiya Babintseva3, Ljudmila Brisevac4, Larysa Sheiko4, Iana Ushko1

1Bohomolets National Medical University, Department of Pathophysiology, Kyiv, Ukraine; 2Bukovinian State Medical University, Department of Pediatrics and Medical Genetics, Chernivtsi, Ukraine; 3Bukovinian State Medical University, Department of Pediatrics, Neonatology and Perinatal Medicine, Chernivtsi, Ukraine; 4Shupyk National Healthcare University of Ukraine, Department of medical and laboratory genetics, Kyiv, Ukraine

Background/Objectives: Syndrome’s Infantile cataract, skin abnormalities, glutamate excess, and impaired intellectual development (CASGID) is a rare disease. The presence of multiple malformations causes difficulties in making a diagnosis. The use of the NGS method helps to detect a genetic defect in a short time and clarify the diagnosis.

Methods: Clinical genealogical, instrumental, whole-exome sequencing.

Results: A 5-year-old girl with a diagnosis of cerebral palsy, spastic tetraparesis, gross delay in stato-kinetic and psycho-speech development, symptomatic epilepsy. The child was born from the first planned pregnancy, which ran with the threat of miscarriage at 13-14 and 33-34 weeks of gestation. Anthropometric indicators at birth corresponded to the norm. Apgar score 8/8 points. At the 3rd month of life, visual impairment and delay in stato-kinetic development were detected. At the time of the examination: child’s weight 8 kg (-3DS), height 87 cm (-3DS), BMI 10.6 (-3DS), microcephaly, high palate, cataract, coloboma, enophthalmia, amaurosis, nystagmus, skin with purple-bluish color of the fingers and toes, erythematous subcutaneous nodules on the limbs. Muscle hypotrophy, muscle hypertonus. Kyphoscoliosis. MRI: incomplete myelination of brain substances. Hypotrophy of the brain convolutions. Signs of significant liquor discirculation.

Due to the multiplicity of lesions, the child underwent whole exome sequencing. A pathogenic denovo mutation was identified in the heterozygous state of the GLS gene NM_014905.5:c.1445C>G (NP_055720.3:p.Ser482Cys), which made it possible to diagnose CASGID syndrome.

Conclusion: the discovered missense variant of the GLS gene causes the development of CASGID syndrome. The combination of phenotyping and genetic testing is essential for the verification of CASGID syndrome.

Conflict of Interest: None declared

EP13.009 Tatton-Brown-Rahman Syndrome due to 2p23.3 microdeletion

Alfredo Repáraz-Andrade 1, Cristina Torreira Banzas1, Milagros Blanco Pérez1, Begoña Durán Fernández-Feijoo2, Arturo Fernández Nogueira1

1Álvaro Cunqueiro Hospital, Genetics and Molecular Pathology, Vigo, Spain; 2Álvaro Cunqueiro Hospital, Neuropediatrics, Vigo, Spain

Background: Tatton-Brown-Rahman Syndrome (TBRS, MIM #615879, ORPHA:404443) is a rare autosomal dominant multiple congenital anomalies syndrome described in 2014. It is characterized by tall stature due to postnatal overgrowth, mild to moderate intellectual disability, seizures and subtle distinctive facial features. TBRS may be associated with an increased risk of developing acute myeloid leukemia.

TBRS results from mutations in DNMT3A and findings suggest haploinsufficiency as the pathogenetic mechanism, even though ClinGen Haploinsufficiency Score for DNMT3A is 1 (Little Evidence for Haploinsufficiency), requiring that more information is necessary.

Methods: We present a 3 years old female patient with delayed language development, obesity and seizures.

Automatic DNA extraction was performed following blood sample venipuncture (QIAcube, Qiagen) and Comparative Genomic Hybridization array (aCGH) was performed with Agilent GenetiSure Cyto CGH 4x180 K.

Bioinformatic interpretation was done with Agilent Cytogenomics 4 and Alissa Interpret CNV software. Genome Build GRCh37 and ISCN 2020 recommendations were followed.

Results: arr[GRCh37] 2p23.3(24605334_25676487)x1

aCGH: 1,0 Mb interstitial heterozygous deletion in 2p23.3 (chr2:24605334_25676487), encompassing 3 genes: DNMT3A (MIM *602769), ADCY3 (MIM *600291) and POMC (MIM *176830).

Classified as PATHOGENIC.

Parental studies: pending.

Conclusion: Tatton-Brown-Rahman Syndrome (TBRS, MIM #615879, ORPHA:404443) is a recent described disorder. Most of the cases reported are due to mutations in DNMT3A and few whole gene deletions have been described. ClinGen Haploinsufficiency Score for DNMT3A is 1.

This finding supports the DNMT3A haploinsufficiency role in the pathogenic mechanism of TBRS.

Conflict of Interest: None declared

EP13.010 Cases diagnosed with Temple syndrome in the last 10 years and main differential diagnoses: Silver-Russell and Prader-Willi syndromes.

Carmen María Dolores Sánchez1, Daniel Doval Calvo2, María José Sánchez Soler1;3, Ana Teresa Serrano Antón 1;3, María Juliana Ballesta Martínez1;3, Vanesa López González1;3, Marta Domínguez Jiménez1;3, Marta Roldán Montero1, Elena Campillo Antón1, Encarna Guillén-Navarro1;3

1Hospital Universitario Virgen de la Arrixaca; 2Hospital General Universitario Santa Lucía; 3Instituto Murciano de Investigación Biosanitaria

Background/Objectives: Temple syndrome is an imprinting disorder involving genes within the imprinted region of chromosome 14q32.

Fetal growth restriction (FGR), feeding issues, developmental delay/intellectual disability (DD/ID), hypotonia and short stature are common to Temple (TS), Silver-Russell (SRS) and Prader-Willi (PWS) syndromes. The similar phenotype makes differential diagnosis difficult, especially during neonatal period.

Objective: clinical and molecular characterization of patients with these disorders.

Methods: Retrospective descriptive study of patients diagnosed with TS, SRS and PWS in the Genetic Section of a tertiary Hospital in the last 10 years.

Results: 23 cases: 10-SRS, 6-TS and 7-PWS. No patients diagnosed prenatally but 16/21(76%) with ultrasound abnormalities, the most frequent FGR and polyhydramnios.

Clinical manifestations

Positive cases/

TS

PWS

SRS

Total

Feeding issues

6/6

6/7

8/10

20/23(87%)

DD/ID

6/6

5/7

5/10

16/23(70%)

Short stature

3/6

4/7

9/10

16/23(70%)

Hypotonia

5/6

7/7

2/10

14/23(61%)

Prominent forehead

1/6

1/7

6/10

8/23(35%)

Syndrome

Molecular diagnosis

Positive cases/total

TS

Maternal UDP14

3/6(50%)

Hypomethylation 14q32

3/6(50%)

SRS

IC1 hypomethylation

4/10(40%)

Mutation in HMGA2 gene

3/10(30%)

Maternal UDP11p15

1/10(10%)

Maternal UDP7

1/10(10%)

Deletion 8q12 de novo (including PLAG1 gene)

1/10(10%)

PWS

Deletion 15q11.2-q13.1 de novo

4/7(57%)

Maternal methylation pattern on both chromosomes 15

2/7(29%)

Maternal UDP15

1/7(14%)

Conclusion: Our results highlight the clinical overlap between these entities, and the importance of assessment by clinical genetics for their proper characterization.

It is advised to request arrayCGH and MS-MLPA in patients with FGR, hypotonia, feeding difficulties, short stature and neurodevelopmental disorder.

The distribution of the molecular defects identified for each syndrome overlaps with the literature.

Grants: No.

Conflict of Interest: None declared

EP13.012 Challenging diagnostics of low level der(17) mosaicism: case report

Andreja Zagorac 1, Peter Gradišnik2, Faris Mujezinović3, Nadja Kokalj Vokač1

1University Medical Centre Maribor, Clinical Institute for Genetic Diagnostics, Maribor, Slovenia; 2University Medical Centre Maribor, Division of Paediatrics, Maribor, Slovenia; 3University Medical Centre Maribor, Department of Perinatology, Maribor, Slovenia

Background/Objectives: Identifying low-level mosaics remains a diagnostic challenge and their detection is extremely important for an accurate diagnosis. We report on an eight-month-old girl who exhibits profound generalized hypotonia, no head control, dysmorphic appearance, hypertelorism and congenital entropy of the right lower eyelid. MRI showed a tethered cord with lumbosacral hydromyelia, reduced white matter volume with hypoplastic callosum, focal pachygyria and polymicrogyria, and thinned optical nerves. She has butterfly deformations of the vertebral bodies.

Prenatal and postnatal karyotyping and FISH revealed a low level der(17) mosaicism (~10%) but a normal aCGH result.

Methods: aCGH, karyotyping and FISH analysis with DNA probes along the chromosome 17, were performed. Samples of chorionic villi and peripheral blood, were tested.

Results: Analyzing native chorionic villi sample and peripheral blood normal aCGH female profiles and mosaic karyotypes with a der(17) cell line, were obtained. FISH confirmed the mosaic der(17). Only aCGH on cultured chorionic villi revealed terminal gain of 17q21.33 to 17q25.3 and terminal loss of 17p13.3 in approximately 20% of the sample. According to all tests the girl´s karyotype is: 46,XX,der(17)(qter→q21.33::p13.3→qter)[10]/46,XX[90].

Conclusions: Detection of low-level mosaicism involves many factors, like the type and number of tissues analyzed, the number of cells studied and the sensitivity of the techniques applied. We report on a girl with mosaic duplication 17q21.33qter and deletion of 17pter. Low grade mosaicism was obtained after karyotyping and FISH. Similar duplications were reported with congenital developmental abnormalities and neurodevelopmental symptoms but clinical significance of the observed low-level mosaicism is not clear.

Conflict of Interest: None declared

EP13.013 Non-canonical mis-splicing variant in SIPA1L2 identified in individuals with developmental delay, congenital heart defect and dysmorphic features

adina fuchs 1;2, Yonatan Rips2, shira Yanovsky-Dagan2, Tamar Harel1;2

1Hebrew University of Jerusalem, Faculty of Medicine, jerusalem, Israel; 2Hadassah Medical Center, Department of Genetics, jerusalem, Israel

Background/Objectives: The SIPA1L2 gene encodes a member of the SIPA1L family of neuronal RapGAPs. Regulation of Rap1/2 GTPase activity by SIPA1L2 is a mechanism to control TrkB signaling in endosomes at presynaptic terminals and activation of the ERK pathway. SIPA1L2 knockout mice show impaired BDNF-dependent presynaptic plasticity. A homozygous variant (NM_020808.5: c.1806G>T; p.(Gly602Gly)) in the SIPA1L2 gene was identified in two siblings with developmental delay, congenital heart defect and distinctive craniofacial features.

Methods: A non-canonical mis-splicing variant was identified in two affected siblings by exome sequencing. The effect of the variant was assessed in patient-derived EBV-transformed lymphoblastoid cell lines at the cDNA and protein levels, by reverse transcriptase (RT)-PCR and ELISA assay.

Results: cDNA analysis of the identified variant revealed deletion of 2bp leading to a frameshift and premature termination codon. This would be expected to lead to nonsense mediated decay and/or truncation of the protein before the RapGAP domain. Quantitative PCR revealed lower expression of RNA in the patient. In addition, patient vs. control cells showed an increase in the phosphorylation of ERK indicating increased activity of the ERK pathway.

Conclusion: Overall, our results suggest that the non-canonical variant leads to mis-splicing, reduced RNA levels and a probable loss-of-function mechanism leading to an over-activation of the ERK pathway. Further studies are underway to gain a mechanistic understanding into the disease pathogenesis. Continued work may be able to offer the possibility of treatment using splice-switching oligonucleotides, an effective and specific way to target and alter splicing in a therapeutic manner.

Conflict of Interest: None declared

EP13.014 10.06 Mb microduplication in 1p36.13p36.11 in a girl with developmental delay, short stature, microcephaly, and submucosal cleft palate

Rosanna H.E. Krakowsky 1;1, Tobias Bartolomaeus1, Anne-Christin Teichmann1, Helene Faust1, Vincent Strehlow1, Franziska Schnabel1

1Institute of Human Genetics, University Medical Center, Leipzig, Germany

Introduction: Microduplications are common causes of intellectual disabilities, facial dysmorphic features and microcephaly, while the observed phenotypical effects often root in changes of dose-sensitive genes. Hence, further research is necessary to elucidate the link between changes in gene dosage and resulting phenotype.

Results: We report a case of a girl born at 38 + 4 weeks of gestation with intrauterine growth retardation and placental insufficiency. At initial consultation she was four months of age presenting with facial dysmorphic features, short stature, microcephaly, accentuated lateral ventricles, postaxial hexadactyly, hearing impairment, strabismus as well as submucosal cleft palate. Re-evaluation at the age of 18 months additionally showed muscular hypotonia, global developmental delay and visual impairment. Copy number variation analysis based on whole exome sequencing revealed a heterozygous de novo 10.06 Mb microduplication in 1p36.13p36.11 (chr1:17281179-27339211) spanning a region of 148 protein-coding genes, 37 of which are disease-associated including DHDDS, RPL11, CDC42, ARlD1A, GRHL3 and KDM1A.

Discussion: Until today, there is no other patient known with a similar genotype. In the literature, only one patient with a considerably smaller microduplication involving ARID1A and PIGV1 is reported featuring intellectual disability, microcephaly, facial dysmorphism and hexadactyly, too (Coutton et al., 2013). Interestingly, 1p36 microdeletion syndrome leads to symptoms comparable to our patient; thereunder developmental delay, facial dysmorphic features, muscular hypotonia, structural brain abnormality and hearing impairment. The clinical course of patients with the detected microduplication is unknown; therefore, collection of clinical data to further decode effects of this alteration on the phenotype is crucial.

Conflict of Interest: None declared

EP13.015 The diagnostic prospects of Dup15q syndrome

Oksana Tyshchenko 1;2, Maria Dushar1, Nataliya Huleyuk1;3, Sofiia Levandivska1, Erika Patskun4, Halyna Makukh1;5

1Scientific Medical Genetic Center LeoGENE, Lviv, Ukraine; 2Ivan Franko National University of Lviv, Department of Genetics and Biotechnology, Lviv, Ukraine; 3Institute of Hereditary Pathology, National Academy of Medical Sciences of Ukraine, Lviv, Ukraine; 4West Ukrainian Specialised Children Hospital, Lviv, Ukraine; 5Lviv regional clinical perinatal center, Regional center of neonatal screening, Lviv, Ukraine

Background/Objectives: Dup15q is a clinically identifiable syndrome that results from the duplication (or multiplication) of the 15q11.2-q13.1 region. Most reported cases concern de novo mutations. In family cases, the development of symptoms occurs when the duplication is inherited from the healthy mother due to imprinting differences in the region. The diagnostic prospects of interstitial 15q duplication are analyzed.

Methods: Genomic DNA was isolated from peripheral blood by an automated method on a MagCore® PLUS. MLPA (Probemix P245, MRS, Holland), conventional cytogenetic analysis of cultured lymphocytes from peripheral blood, and FISH analysis were carried out. The results of NGS sequencing of target gene panel using Illumina technology were analyzed.

Results: It was established that among six independent cases of Dup15q syndrome, four patients inherited interstitial 15q duplication from a phenotypically healthy mother. 15q duplication occurred de novo in rest cases. No abnormalities were detected with standard karyotyping in four patients, as chromosomal changes are below the resolution of this method. The clinical presentation was not different for inherited and de novo cases of Dup15q syndrome and manifested with facial differences, epilepsy, developmental speech delay, autistic spectrum disorders, hyperactivity, and intellectual disability. We suggest MLPA as a quicker and more cost-effective diagnostic method for Dup15q syndrome.

Conclusion: Family history and parental testing are critical for medical-genetic consulting and risk assessment for Dup15q syndrome. These insights contribute to a better understanding and treatment of Dup15q syndrome, advocating for further research to enhance diagnostic approaches and therapeutic strategies.

Grants:

Conflict of Interest: None declared

EP13.016 Exploring the spectrum of Tricho-rhino-phalangeal syndrome, type I in Korean patients: Clinical and genetic characteristics

Jung Min Ko 1, A Young Park1, Naye Choi1

1Seoul National University Children’s Hospital, Pediatrics, Seoul, Korea, Rep. of South

Tricho-rhino-phalangeal syndrome I (TRPS I, OMIM#190350) is a rare autosomal dominant disorder with variable expressivity, often leading to underdiagnosis. This study aimed to characterize the clinical features and genotypes of 19 Korean TRPS I patients. The retrospective analysis included demographic, clinical, and radiologic data, and genetic confirmation was achieved through various genetic analysis methods.

Results revealed a median age at diagnosis of 8.9 years, with hypotrichosis (37%), short stature (21%), and hand deformity (16%) as common presenting signs. Hip dysplasia was observed in 11% of patients. Most (95%) had heights below the 50th percentile of the age-and sex-matched controls, with a median height of -1.32 SDS. Sparse hair and pear-shaped noses were universal, while skeletal abnormalities included small hands, cone-shaped epiphyses and occasional hip dysplasia, pes planus, and scoliosis. Treatment included Minoxidil for hypotrichosis (63%) and surgical interventions for hip dysplasia and knee metaphyseal dysplasia.

Except for four (21%) patients with exonic deletions, each of 15 patients had different pathogenic sequence variants in TRPS1. The identified variants were distributed through exon 3 to 7, with 47% being novel. We could not find any meaningful differences of clinical and radiological manifestations according to exonic locations of variants

In conclusion, TRPS I should be considered in cases of early-onset alopecia, short stature, and skeletal dysplasia particularly involving the hands, hips, and knees. While individualized variants were prevalent, there was no mutational hotspot in TRPS1. Notably, 21% had exonic deletions in our cohort, emphasizing the need of copy number variant analysis in cases without sequence alterations.

Conflict of Interest: None declared

EP13.017 Can CNVs cause a KCNK9 imprinting syndrome?

Marta Wójcik 1, Magdalena Badura-Stronka1;2, Aleksandra Wnuk-Kłosińska1;2, Dorota Simon3, Aleksandra Jakubczak3, Michał Piechota3, Aleksander Jamsheer1;2

1Diagnostyka Genesis, Genetic clinic, Poznań, Poland; 2Poznan University of Medical Sciences, Department of Medical Genetics, Poznań, Poland; 3Diagnostyka Genesis, Laboratory for medical genetics, Poznań, Poland

Background/Objectives: Pathogenic variants on the maternal copy of the KCNK9 gene give rise to a well-defined syndrome characterized by muscular hypotonia and developmental delay, known as KCNK9 imprinting syndrome (KIS) or Birk-Barel syndrome (MIM612292). Until now, only point mutations have been identified as causal factors for this syndrome. This case report highlights a patient exhibiting pertinent symptoms and a duplication involving the KCNK9 gene.

Methods: The patient, a 3-year-old male, was born prematurely at 36 weeks of pregnancy. From the early months, he displayed feeding difficulties, muscular hypotonia, and significant motor delays, while speech and social interactions remained unaffected. CPK and lactate yielded normal results, and imaging studies (brain MRI, heart, and abdomen ultrasound) revealed no abnormalities. Genetic testing included karyotyping, array comparative genomic hybridization (aCGH), multiplex ligation-dependent probe amplification (MLPA) of the SMN1 gene, methylation-specific polymerase chain reaction (MS-PCR) of the SNRPN locus, triplet-primed polymerase chain reaction (TP-PCR) of the DMPK gene, and whole exome sequencing (WES) encompassing the mitochondrial genome.

Results: aCGH identified a de novo duplication involving the KCNK9 gene and partially the TRAPPC9 gene: arr[GRCh37] 8q24.3(140,375,243-140,821,425)x3. Parents of the boy are undergoing testing for a specific SNP found on the duplicated allele to determine its parental origin.

Conclusion: The identified CNV may represent a novel cause of KCNK9 imprinting syndrome. Further investigation, including functional tests, is imperative to confirm the pathogenicity of this duplication.

Grants: The cost of all tests was covered by the public health insurance or the patient’s parents.

Conflict of Interest: None declared

EP13.018 Features of multisystem developmental pathology in an 8-month-old girl with terminal 4q deletion syndrome (de novo) from Ivano-Frankivsk (Ukraine)

Nataliya Kitsera1, Maya Bondarenko 2

1Lviv Oncology Regional Treatment and Diagnostic Center, Lviv, Ukraine; 2Ivano-Frankivsk National Medical University, Ivano-Frankivsk, Ukraine

Consortium: Dear colleagues!

We will be grateful to you for the exemption from payment of the registration fee for the conference in connection with the war with the aggressor country Russia.

Background/Objectives: Terminal 4q deletion syndrome is a rare disease with a variety of clinical polymorphisms.

The purpose of this study - diagnose the patient’s chromosomal changes and clinical manifestations.

Methods: clinical, chromosomal analysis (Ukraine).

Results: The girl was born with microcephaly from the 2nd complicated pregnancy of healthy unrelated Ukrainian parents aged 31, after spontaneous delivery (36 weeks). Birth weight - 2050, height - 46 cm, head circumference -27 cm, chest circumference -30 cm, Apgar score of 7-8 points. The 1st pregnancy - miscarriage at 6 weeks.There are no hereditary diseases and birth defects in the pedigree.

Parents turned to a geneticist with complaints of delayed physical, mental retardation, epileptic seizures in a 8-month-old daughter to clarify the diagnosis

Physical examination revealed microcephaly, dysplastic auricles: left elf-like ear; flat back of the head; short lip filter, thin upper lip; micrognathia of the upper jaw; macroglossia; hypertelorism; saddle nose; long fingers and toes; four-finger folds of both palms; clinodactyly of the little fingers; partial atrophy of the optic nerve of the right eye, complex astigmatism. Serum biochemistry and blood test, echocardiography, ultrasonography of abdomen were normal. MRI of skull - Blake’s pocket cyst.

Chromosomal analysis - 47,XX,del(4)(q32),+mar. Normal cytogenetic analysis of the parents showed that this chromosomal rearrangement arose de novo. The girl is now 4 years old and is under the supervision of specialists.

Conclusion: The rarity of terminal 4q deletion syndrome complicates genotype-phenotype correlation with dysmorphic features, behavioral disorders, developmental delay at birth or in early childhood.

Grants:No.

Conflict of Interest: None declared

EP13.019 Smith-Magenis syndrome with RAI1 gene deletion - case report in a patient from Romania

Miruna Gug 1, Diana Miclea2;3, Nicoleta Andreescu4;5, Ioana Mozos6;7, Cristina Gug8

1“Victor Babes” University of Medicine and Pharmacy, Faculty of Medicine, Timișoara, Romania; 2Spitalul Clinic de Urgență pentru Copii, Department of Medical Genetics, Cluj-Napoca, Romania; 3“Iuliu Hațieganu” University of Medicine and Pharmacy, Department of Mother and Child, Cluj-Napoca, Romania; 4“Victor Babes” University of Medicine and Pharmacy, Department of Microscopic Morphology, Timisoara, Romania; 5“Victor Babeș” University of Medicine and Pharmacy, Genomic Medicine Centre, Timisoara; 6“Victor Babes” University of Medicine and Pharmacy, Department of Functional Sciences-Pathophysiology, Timisoara; 7“Victor Babes” University of Medicine and Pharmacy, Center for Translational Research and Systems, 300173 Timisoara; 8“Victor Babes” University of Medicine and Pharmacy, Department of Microscopic Morphology, Timisoara

Smith-Magenis syndrome (OMIM#182290) is a rare multisystemic genetic disorder with behavioral dysfunction and sleep disturbances.

We present the case of an 18-month-old child with craniofacial dysmorphia: plagiocephaly, prominent viscerocranium, high forehead, deformed, large and low-set ears, bilateral epicanthus and convergent strabismus, mouth with tent-shaped upper lip, bilateral syndactyly of the second and third toe. Neurologically, the patient presented axial hypotonia, slight hypertonia of the limbs and immature gait. The psychological evaluation revealed hyperactivity, sleep disorders, expressive language development disorder, mental instability, anxiety, developmental delay consistent with a 12-month age.

Genetic testing by NGS included sequencing analysis and deletion/duplication testing for 912 genes. Testing at Invitae Laboratories identified two pathogenic deletion variants that affected the entire coding sequence in the RAI1 and B9D1 genes. Loss-of-function variants in the retinoic acid-induced protein-1 (RAI1) gene are known to be pathogenic and associated with Smith-Magenis syndrome. The location of both genes on chromosome 17p11.2 led to the suspicion of an extensive deletion. The SNP array technique revealed a heterozygous deletion spanning approximately 3677 kb in the 17p11.2 region. Deletion involves several OMIM genes, 16 of which are associated with morbidity.

The parents have a normal karyotype, and genetic testing on the abnormal genes identified in the child did not reveal any deletions. Therefore, the posibility of a translocation is rulled out, infering that the deletion or mutation in the child occurred de novo.

Examining detailed clinical and molecular characteristics proves valuable in investigating correlations between phenotype and genotype in rare diseases.

Conflict of Interest: None declared

EP13.020 Biallelic RAB34 variants impair primary cilium assembly and cause a novel subtype of oral-facial-digital syndrome.

Ange-Line Bruel 1;2, Anil Kumar Ganga3, Lenka Nosková4, Irene Valenzuela Palafoll5, Jelena Martinovic6, Yannis Duffourd1;2, Marie Zikanova4, Filip Majer4, Stanislav Kmoch4, Marketa Mohler7, Jingbo Sun3, Lauren Sweeney3, Núria Martínez-Gil5;8, David Breslow3, Christel Thauvin-Robinet1;2;9

1Université de Bourgogne, INSERM U1231 Génétique des Anomalies du développement, DIJON, France; 2CHU Dijon Bourgogne, Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, Fédération Hospitalo-Universitaire (FHU)-TRANSLAD, DIJON, France; 3Yale University, Department of Molecular, Cellular, and Developmental Biology, New Haven, United States; 4Charles University and General University Hospital, Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Prague, Czech Republic; 5Hospital Vall d’Hebron, Àrea de Genètica Clínica i Malalties Minoritàries, Barcelona, Spain; 6Antoine Béclère Hospital, Unit of Embryo-Fetal Pathology, AP-HP, Paris, France; 7University Hospital Ostrava, Institute of Molecular and Clinical Pathology and Medical Genetics, Ostrava, Czech Republic; 8Vall d’Hebron Institut de Recerca, Grup de Medicina Genètica, Barcelona, Spain; 9Hôpital d’Enfants, CHU Dijon Bourgogne, Centre de Génétique et Centre de référence maladies rares « Anomalies du Développement et Syndromes Malformatifs », Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (FHU-TRANSLAD), Dijon, France

Oral-facial-digital syndromes (OFDS) are characterized by the association of facial dysmorphism, oral abnormalities and digital malformations. Their initial classification into 14 subtypes is based on their mode of transmission and associated clinical signs (cerebral, renal, skeletal, retinal malformations, etc.). Twenty-five causative genes are now known to affect the function and/or structure of the primary cilium at different levels: basal body involved in ciliary assembly (e.g. OFD1, C2CD3), transition zone (e.g. TMEM107) or intraflagellar transport (e.g. IFT57).

In this study, we report homozygous missense variants in the RAB34 gene in 4 male individuals (including 3 fetuses) from 3 unrelated families. Prenatal ultrasound revealed oral cleft, polydactyly, cardiac and cerebral abnormalities. Three pregnancies were terminated and one fetus born at term with respiratory distress and various complications, resulting in death at 3 months. On clinical examination, they presented with variable craniofacial abnormalities (hypertelorism, micrognathia, macrocephaly), bilateral oral clefts (4/4), lingual hamartomas (2/4), and central pre-axial or bilateral polydactyly (4/4) of the hands and feet, leading to the clinical diagnosis of OFDS. Anorectal (4/4), cardiac (3/4) and cerebral (2/4) malformations including agenesis of the corpus callosum were also noted.

RAB34 encodes a member of the Rab GTPase superfamily and has recently been described as a key mediator of ciliary membrane formation. Functional studies demonstrated a loss-of-function effect of these pathogenic variations, with defective ciliary assembly.

In conclusion, this study identifies the first small GTPase involved in a new subtype of OFDS with distinct clinical features.

Conflict of Interest: None declared

EP13.021 Clinical Features of Japanese girl with Radio-Tartaglia Syndrome due to SPEN pathogenic variant.

Eriko Nishi 1, Kumiko Yanagi2, Nobuhiko Okamoto1, Tadashi Kaname2

1Osaka Women’s and Children’s Hospital, Department of Medical Genetics, Osaka, Japan; 2Research Institute, National Center for Global Health and Medicine, Department of genome medicine, Tokyo, Japan

Background: Radio-Tartaglia syndrome (RATARS) (MIM # 619312) is a genetic disorder caused by heterozygous truncating variants in SPEN on chromosome 1p36. This syndrome is very rare with only 34 individuals reported (Radio et al.,2021). RATARS is mainly characterized by developmental delay with hypotonia, intellectual disability, and facial features such as broad forehead, bitemporal narrowing, arched elongated eyebrows, synophrys, epicanthal folds, dysplastic ears, and broad nose with bulbous/prominent nasal tip (Radio et al.,2021). In this study, we report a Japanese girl with psychomotor delay, hypotonia, and facial features resembling Down syndrome (DS), in whom we identified a de novo heterozygous pathogenic variant of SPEN and diagnosed her as RATARS.

Case report: She was born at 38 weeks and 1 day, weighing 2598 g, without respiratory or feeding difficulties. We first considered DS as a differential diagnosis based on her developmental delay with hypotonia and facial features including upslanted palpebral fissure, hypertelorism, epicanthus folds, and a low nose, but ruled it out by cytogenetic testing using peripheral blood lymphocytes and buccal mucosa cells. Microarray revealed no other aberration. We performed a trio whole exome sequencing, and identified a recurrent pathogenic variant in SPEN: NM_015001.3: c.6223_6227del: p.(Ser2075GlufsTer46).

Conclusion: Although some of the features of RATARS have been reported to be similar to those of 1p36 deletion syndrome, the facial similarity to DS was characteristic in our case. Whether this feature is unique to her or relatively common to individuals with RATARS may be discussed further as more reports of individuals with RATARS accumulate.

Grant: IRUD from AMED.

Conflict of Interest: None declared

EP13.022 A novel homozygous stop-gain variant in LRRC32 causing cleft palate, proliferative retinopathy, and developmental delay

Sameeta Kumari 1, Fizza Akbar2, Seung Woo Ryu3, Arshalooz Rahman4, Salman Kirmani4

1Aga Khan University, Karachi, Pakistan; 2Aga Khan University Hospital, Division of Woman and Child Health, Karachi, Pakistan; 33billion Inc., Seoul, Korea, Rep. of South; 4Aga Khan University Hospital, Department of Pediatrics and Child Health, Karachi, Pakistan

Background/Objectives: The leucine-rich repeat containing protein 32 (LRRC32) gene encodes for a cell-surface receptor called Glycoprotein A repetitions predominant (GARP). GARP is crucial for regulation of transforming growth factor-β (TGF-β) in various cellular processes. To date, four cases with two pathogenic variants in LRRC32 have been described. Using Trio-based clinical whole genome sequencing (WGS), we describe a male patient with a novel homozygous LRRC32 variant, expanding the phenotype of the previously reported cases.

Methods: Case Report

Results: A 13-years-old proband presented with a long-standing history of global developmental delay, congenital blindness and corneal clouding. He was also recently diagnosed with post infectious glomerulonephritis (PIGN). Notably, his parents were first cousins. His examination was significant for coarse facies, strabismus, surgically repaired cleft palate, and extremely dry, scaly skin. Moreover, he had thin extremities, an overall low muscle mass, and mild, generalized hypotonia. Relevant lab investigations reported elevated IgE levels, reduced serum albumin and complement 3 (C3) levels. Trio-based WGS was sent to 3Billion (S. Korea), which revealed a likely pathogenic homozygous variant in LRRC32, denoted as c.1261C>T (p.Arg421*).

Conclusion: Our case adds further evidence that LRRC32 variants can cause cleft palate, proliferative retinopathy, and developmental delay. The prominent skin manifestations and the PIGN in our patient suggest a possible LRRC32 phenotypic expansion. Describing more cases is crucial for understanding the potential association between PIGN and LRRC32, and whether TGF-β impacts immune system dysregulation prediposing to PIGN, or if low C3 levels are associated with this disorder?

Conflict of Interest: None declared

EP13.023 Clinical implications of copy number variations on chromosome 16

Somayyeh Heidargholizadeh 1, Birsen Karaman1, Tugba Kalayci1, Ayça Dilruba aslanger1, Gülnihal Bulut2, gözde tutku turgut1, Behiye Tuğçe yıldırım1, Bilge Ozsait Selcuk1

1Istanbul University Faculty of Medicine, Medical Genetics, İstanbul, Türkyie; 2Istanbul University Institute of Health Sciences, Medical Genetics, İstanbul, Türkyie

Background: Segmental copies in chromosome 16 render the chromosome unstable and predispose it to producing Copy Number Variations (CNVs). However incomplete penetrance and diverse phenotypes, complicate prenatal genetic counseling. We analyzed the prenatal ultrasound and postnatal phenotypic characteristics associated with chromosome 16 CNVs to improve diagnosis and monitoring of this disease in the cases.

Methods: We screened cases that underwent karyotyping and CMA from 2008 to 2023. We selected 37 cases with anomalies in chromosome 16, involving 19 prenatal and 18 postnatal cases, and characterized their phenotypes based on medical records. Additionally, the type and parental origin of these CNVs were determined.

Result: We detected 37 CNVs, including 15 microdeletions and 17 microduplications, and five showed apparently balanced rearrangement with potential clinical significance. Of these CNV’s, 20 were microdeletion/microduplication syndromes involving distinct genomic regions previously defined as 16p13.11, 16p12.2, 16p11.2 and 16p13.3 microdeletions, as well as 16 p11.2-p12.2 and 16p11.2 microduplications. The remaining 17 CNVs are reported less frequently and include 16p13.11p12.3 deletions/duplication, anomalies of the q arm of chromosome 16, marker chromosome, balanced chromosomal rearrangements, and complex chromosomal anomaly involving chromosome 16. Of these, 19 cases were verified by pedigree, with 7/19 cases de novo and the rest being familial.

Conclusion: Our findings can contribute to the creation of a chromosome 16 disease map based on regions that may be associated with disease development. The phenotypic outcome cannot be predicted owing to the variable expressivity and incomplete penetrance of these CNVs.

Grants: Supported by Research Fund/Istanbul University (project no:TYL-2021-38090)

Conflict of Interest: None declared

EP13.024 Findings in ear tomography study from patients with oculo-auriculo-vertebral syndrome.

María de la Luz Arenas-Sordo 1;2, Isabel Barradas-Hernández3, J. Karina Peñuelas-Romero1, Kyrrck A Del Real-Martínez4, JImena Solorio-Fosado5

1Instituto Nacional de Rehabilitación LGII, Genetics, Mexico City, Mexico; 2UNAM, Medicine Faculty, Mexico City, Mexico; 3Insituto Nacional de Rehabilitación LGII, Genetics, Mexico City, Mexico; 4Universidad de la Salud, Medicine, Mexico City, Mexico; 5Private consultation, Santiago de Querétaro, Mexico

Background/Objectives: Oculo-auriculo-vertebral syndrome (OAVS) is the most common craniofacial malformation after cleft lip/palate. The clinical findings are microtia, hemifacial microsomia, vertebral and renal alterations. Around 80% of cases present malformation of the auditory ossicles (middle ear) and around 20% of the inner ear. The objective is to describe the ear tomography findings of a group of patients with OAVS.

Methods: Review of tomography reports of 229 patients

Results: The records were from 107 female and 122 male. Age between 2 and 53, with mean 12.8.

166 have unilateral microtia and 63 bilateral. The great majority have conductive hearing loss.

Tomograpy findings. In external acoustic meatus: stenosis in 14 right side, 12 left and 5 both sides. Atresia in 108 cases right side, 43 left and 36 both sides. Middle ear. Ossicles dysplasia in all in 131 cases, 39 cases in malleus, 6 in incus and 4 in stapes. Agenesis: malleus 16, incus 3, stapes 14, from all of them, 8. It was also aberrant course of the facial nerve in 82 cases in right side, 29 left side and bilateral in 28 cases. Inner ear. 14 cases with hypoplasia or dysplasia of semicircular canals, 15 cases with hypoplasia or agenesis of VII and/or VIII nerves. Obliteration of the oval window, 8 right side, 6 left side and 4 bilateral.

Conclusion: The tomography study is neccesary in all the patients woth OAVS to guide the treatment and after with molecular studies try to relate the findings with the maformations.

Grants:

Conflict of Interest: None declared

EP13.025 Atypical manifestations of autoimmune polyglandular syndrome type 1

Sabina Nagieva 1, Oksana Ryzhkova1, Viktoriia Zabnenkova1, Anna Orlova1, Olga Shchagina1, Natalia Taran2, Irina Tin2, Tatyana Strokova2, Natalia Semenova1

1Research Centre for Medical Genetics, Moscow, Russian Federation; 2Federal Research Centre of Nutrition and Biotechnology, Moscow, Russian Federation

Background/Objectives: Biallelic variants affecting the AIRE gene cause Autoimmune polyglandular syndrome, type 1 (APS1; OMIM: 240300). The most common initial presentation of the syndrome is chronic mucocutaneous candidiasis in early childhood. The subsequent APS1 signs are hypoparathyroidism and Addison’s disease. However, symptoms could be variable and manifest with nonendocrine disorders even among the family members.

Methods: Analysis of clinical data, Whole Exome Sequencing (WES) and trio Sanger sequencing for variants validation were performed.

Results: А 3-year-old boy from healthy parents was examined. The patient was referred to genetic counselling because of severe diarrhea and symptoms of hepatobiliary tract involvement. Retrospective data showed absence of candidiasis and autoimmune disorders. The proband had elevation of transaminase (maximal levels of AST 2045 U/L (8-48 U/L), ALT 1609 U/L (7-45 U/L) with cholestasis: increased level of GGT 443 U/L (11-50 U/L) with levels of total and direct bilirubin 58.3 µmol/L and 48.1 µmol/L respectively. Blood tests also revealed low glucose 1.6 mmol/l (3,9-5.7 mmol/l) and cholesterol 1 mmol/l (2,9-5,2 mmol/l), elevation of lactate acid (max 4.28; normal 0.9-1.7 mmol/l), hepatic autoantibodies and celiac disease markers were negative. Ultrasound results demonstrated hepatomegaly with fibrosis. Phenotype features: megalocephaly (+3.78SDS), thick hair, enlarged size of abdomen and no skin lesions. Genetic analysis revealed two previously reported pathogenic nonsense variants in the AIRE gene: NM_000383.4:c.769C>T p.(Arg257Ter) and NM_000383.4:c.1302C>A p.(Cys434Ter) in compound-heterozygous state.

Conclusion: This case demonstrates an atypical APS1 manifestation: the most prevalent clinical features in patient were diarrhea with malabsorption and hepatitis with cholestasis.

Conflict of Interest: None declared

EP13.026 Structural variant involving SLC19A2 in a patient with thiamine-responsive megaloblastic anemia syndrome

Franziska Schnabel 1, Christina Kloetzer2, Helene Faust1, Robin-Tobias Jauss1, Denny Popp1, Anne Sophie Kubasch2, Madlen Jentzsch2, Georg-Nikolaus Franke1, Jens Uhlig3, Klaus Metzeler2, Vladan Vucinic2, Uwe Platzbecker2, Johannes Lemke1;4, Henry Oppermann1

1Institute of Human Genetics, University Leipzig Medical Center, Leipzig, Germany; 2Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University Leipzig Medical Center, Leipzig, Germany; 3Hematological Praxis Naunhof, Naunhof, Germany; 4Center for Rare Diseases, University Leipzig Medical Center, Leipzig, Germany

Background: Thiamine-responsive megaloblastic anemia (TRMA) syndrome is characterized by megaloblastic anemia, progressive sensorineural hearing loss, diabetes mellitus as well as neurological, cardiovascular, and ophthalmological abnormalities. Biallelic variants in SLC19A2 encoding a high affinity thiamine transporter had been described as disease-causing in about 200 cases worldwide.

Case Report: Here, we present a 39-years-old male patient with diabetes mellitus, optic neuropathy, and cataract since early childhood. Myelodysplastic signs in bone marrow and megaloblastic anemia was diagnosed in adulthood and resulted in transfusion-dependent symptomatic anemia twice. Coronary one-vessel-disease required a drug-eluted stent. However, no hearing impairment is known.

Results: Whole exome sequencing revealed a heterozygous pathogenic variant (NM_006996.3: c.1001G>A, p.(Gly334Asp)) in SLC19A2, while a second variant was neither detectable by whole exome sequencing nor by Nanopore sequencing of PCR products covering SLC19A2. Interestingly, whole genome sequencing uncovered a 3.4 Mb inversion in the chromosomal region 1q24.2 (NC_000001.11: g.166068365_169484680inv) with breaking points in FAM78B and SLC19A2 most probably leading to a premature truncation and a functionally null allele. Segregation analysis confirmed compound heterozygosity of both variants and hence, the diagnosis of TRMA in our patient. Thiamine supplementation was initiated and after 12 weeks hemoglobin levels had normalized and bone marrow smear showed improvement of dysplastic abnormalities.

Conclusion: TRMA should be considered as differential diagnosis in patients with megaloblastic anemia and indicative comorbidities. Hereby, genetic diagnostics necessitate whole genome sequencing in order to detect structural variants involving SLC19A2.

Grant: No

Conflict of Interest: None declared

EP13.027 EFTUD2 gene-associated mandibulofacial dysostosis, Guion-Almeida type: Case series of four Turkish patients

Serap Ketenci İşlek 1, Merve Soğukpınar1, gizem ürel-demir1;2, Pelin Simsek-Kiper1, Gulen Eda Utine1

1Hacettepe University Medicine Faculty, Department of Pediatrics, Division of Pediatric Genetics, Ankara; 2Mersin City Training and Research Hospital Pediatric Genetics, Mersin, Türkyie

Background/Objectives: Mandibulofacial dysostosis, Guion-Almeida type (MIM#610536) is an extremely rare genetic syndrome featuring microcephaly, malformations of the external ear, conductive hearing loss, and distinctive facial characteristics such as micrognathia, malar and mandibular hypoplasia, along with intellectual disability. Additional findings may comprise cardiac malformations, esophageal atresia/tracheoesophageal fistula, short stature, skeletal abnormalities, seizure, and craniofacial malformations, including cleft palate, choanal atresia, zygomatic arch cleft, and facial asymmetry. The syndrome is characterized by haploinsufficiency of EFTUD2, located on chromosome 17q21, which encodes a spliceosomal GTPase. This study included four unrelated Turkish patients diagnosed with Mandibulofacial dysostosis, Guion-Almeida type.

Methods: Whole exome sequencing was performed on all affected individuals after obtaining an informed consent.

Results: All patients presented with progressive microcephaly and dysmorphic facial features, including preauricular skin tags and ear anomalies. Three patients exhibited global developmental delay, one had bilateral choanal atresia, one presented with a cleft palate, and another experienced seizures. A pathogenic variant in the EFTUD2 gene was detected in all patients, confirming the diagnosis of Mandibulofacial dysostosis, Guion-Almeida type.

Conclusion: A defect in the EFTUD2 gene leads to abnormal differentiation of neural crest cells, resulting in first and second pharyngeal arch anomalies during embryonic development. Mandibulofacial dysostosis, Guion-Almeida type has overlapping features with better known syndromes such as Treacher Collins, Nager or CHARGE syndrome. Therefore, it should be considered among the differential diagnoses in patients presenting with progressive microcephaly, facial dysostosis, intellectual disability, and otologic issues.

Grants: None

Conflict of Interest: None declared

EP13.028 De novo splice variant in DKC1 in a female with clinical features of Hoyeraal-Hreidarsson syndrome

Dominique Braun 1, Anne Gregor1, Monika Haubitz2, Gabriela M Baerlocher2;3, Claudine Rieubland1, Cornelia Kraus4, Christiane Zweier1

1Inselspital Bern, Department of Human Genetics, Bern, Switzerland; 2University of Bern, Department for BioMedical Research (DBMR), Bern, Switzerland; 3Hirslanden Zürich, Clinic for Hematology and Oncology, Zürich, Switzerland; 4Universitätsklinikum Erlangen, Institute of Human Genetics, Erlangen, Germany

Background/Objectives: Pathogenic variants in DKC1 cause X-linked dyskeratosis congenita (DC), the most severe form of which is Hoyeraal-Hreidarsson syndrome (HHS). HHS has so far only been reported in hemizygous males and is associated with severe neurological impairment and progressive bone marrow failure. Heterozygous carrier females are usually asymptomatic. Most known pathogenic variants are missense variants with few described splice variants, while hemizygous null variants are assumed to be lethal.

Methods: After diagnostic trio exome sequencing, an exon trapping mini-gene assay, X-inactivation studies and telomere length measurements were carried out to aid variant interpretation and genotype-phenotype correlation.

Results: A de novo splice variant c.1260-3C > A in the DKC1 gene was identified in a young adult female presenting with clinical features overlapping with HHS. These included intrauterine and postnatal growth retardation, microcephaly, intellectual disability and recurrent infections, while lacking other typical aspects such as cerebellar hypoplasia or bone marrow failure. The in vitro splice assay showed skipping of exon 13 of DKC1 - predicted to lead to a frameshift - with evidence of leaky splicing. X-inactivation studies in the affected individual showed a random X-inactivation pattern, and telomere length analysis revealed very short telomere lengths (<1st centile) in five of six leukocyte subsets.

Conclusion: Our observations suggest that the de novo splice variant in DKC1, likely leading to loss of function, might be causative for the individual’s symptoms and that this and similar variants might result in a phenotype overlapping with but not being typical for HHS in females.

Conflict of Interest: None declared

EP13.029 RPL26 variants: a rare cause of Diamond-Blackfan Anemia Syndrome with multiple congenital anomalies at the forefront

Clemence Vanlerberghe 1, Frédéric Frénois1, Thomas Smol1, Anne-Sophie Jourdain1, Fabienne Escande1, Emilie Ait Yahya2, Abdulrahman Aldeeri3;4, Timothy Yu3, Valérie Cormier-Daire5, Jamal Ghoumid1, Maureen Jacob6, Ruth Newbury-Ecob7, Sylvie Manouvrier-Hanu1, Jessica Platon8, Sebastian Sailer9, Perrine Brunelle1, Lydie Da Costa8;10;11, Florence Petit1

1Univ. Lille, CHU Lille, ULR 7364 - RADEME - Maladies RAres du DÉveloppement embryonnaire et du Métabolisme, Lille, France; 2CHU Lille, Cellule bioinformatique, plateau commun de séquençage, Lille, France; 3Division of Genetics and Genomics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States; 4King Saud University Medical City, Riyadh, Saudi Arabia; 5Paris Cité University, Reference center for skeletal dysplasia, Imagine Institute, Necker Hospital, Paris, France; 6Institute of Human Genetics, Technical University of Munich, School of Medicine, Munich, Germany; 7University Hospitals Bristol, St Michael’s Hospital, NHS Foundation Trust, Department of Clinical Genetics, Bristol, United Kingdom; 8Université Picardie Jules Verne, HEMATIM EA4666, Amiens, France; 9Heidelberg University, Institute of Human Genetics, Heidelberg, Germany; 10AP-HP, Hôpital Robert Debré, Service Hématologie Biologique, Paris, France; 11Université Paris, Inserm U1134, Paris, France

Background/Objectives: Diamond-Blackfan Anemia Syndrome (DBA or DBS) is a rare congenital disorder originally characterized by bone marrow failure with or without congenital anomalies, with at least 22 genes implicated, most encoding ribosomal proteins. RPL26, encoding ribosomal protein L26, is an emerging candidate gene with only two individuals reported (DBA11, MIM#614900). We herein describe additional cases with RPL26-related DBA to better characterize the phenotype.

Methods: Leveraging exome and genome sequencing, we identified eight DBA patients from four unrelated families as carriers of heterozygous RPL26 variants. The identified variants, including two splicing variants in the non-coding exon 1 and/or intron 1, likely cause RPL26 haploinsufficiency, which was previously shown to impair ribosomal maturation in a cellular model.

Results: We show that DBA11 primarily presents as multiple congenital anomalies (particularly radial ray abnormalities), albeit with variable expression. Interestingly, in one individual with normal blood cell count, extended studies revealed subclinical impaired hematopoietic with defective proliferation and enucleation of erythrocytes and delayed differentiation. In this cohort, hematological involvement is infrequent, contributing to the mounting evidence that bone marrow failure is not universally central to all DBS genes as it was once regarded. Therefore, DBA genes should not be dismissed solely based on an absence of anemia in patients with an otherwise compatible phenotype. Additionally, we report that mandibulofacial dysostosis and neural tube defects are potential features in DBA11, expanding the growing list of DBS abnormalities.

Conclusion: We confirm RPL26 as a DBS gene and expand the phenotypic spectrum of the gene and the disease.

Conflict of Interest: None declared

EP13.030 Microphthalmia and coloboma – novel features in DHP1 neurodevelopmental syndrome

Veronica Seidel 1, Julia Suárez González2, Alejandra Aguado del Hoyo3, Andrea Palacios Bermejo4, Raquel Rodrigo Fernández5, María Molina Bañón4, Ester Sanz López6

1HGU Gregorio Marañón, Clinical Genetics/ Pediatrics Department, Madrid, Spain; 2HGU Gregorio Marañón, Genomics Unit, Madrid; 3HGU Gregorio Marañón, Pediatric Radiology; 4HGU Gregorio Marañón, Pediatrics; 5HGU Gregorio Marañón, Ophthalmology; 6HGU Gregorio Marañón, Neonatal Unit

Background: Biallelic variants in the DPH1 (diphthamide biosynthesis 1) gene cause Craniofacial dysplasia-short stature-ectodermal anomalies-intellectual disability syndrome. We present another case of this rare syndrome featuring previously undescribed bilateral microphthalmia and coloboma, and congenital deafness.

Methods: Case description: Fourth child born to consanguineous Moroccan parents with 3 healthy girls. Prenatal ultrasound detected ventriculomegaly and heart anomalies. Growth parameters at birth were within normal range. Noninvasive respiratory support was required initially. Neonatal hypotonia and dysmorphic features were noted: large fontanelles, sparse scalp hair, absent eyebrows and eyelashes, very small eyeballs with short fissures, small broad nose and mouth, cleft palate, micrognathia, crumpled ears and tapering short fingers with camptodactyly and ulnar deviation, bilateral single palmar crease, short toes, bilateral cryptorchidism and micropenis. He had auricular and ventricular septal defects and hypothyroidism. His hearing screening showed no response in both ears. At age 4 months he is on high-flow oxygen support and tube-feeding, remains hypotonic and has no adequate head control. He has developed congestive heart failure and requires palliative care.

Results: Exome sequencing detected a known pathogenic homozygous variant in the DPH1 gene; p.Leu125Pro. Brain MRI showed asymmetrical cerebellar hypoplasia and dysplasia, dysplastic corpus callosum, mild ventriculomegaly, and simplified gyral pattern. Bilateral coloboma and microphthalmia with persistent primary vitreous and cysts on the right side. The right VEP test had no response.

Conclusion: Microphthalmia and coloboma, and congenital hearing loss are additional features in this rare multisystemic recessive condition with often severe intellectual disability.

Conflict of Interest: None declared

EP13.031 Chromosomal microarray analysis in children with syndromic short stature

Nela Maksimovic 1, Tatjana Damnjanovic1, Biljana Jekic1, Milka Grk1, Marija Dusanovic Pjevic1, Ana Djuranovic1, Milica Rasic1, Parsa Barzegar2, Badr El Hayani1, Dijana Perovic1

1Institute of Human Genetics, Faculty of Medicine, University of Belgrade, Belgrade, Serbia; 2Faculty of Medicine, University of Belgrade, Belgrade, Serbia

Background/Objectives: The etiology of syndromic short stature (short stature and presence of dysmorphic features, developmental delay, and/or intellectual disability) is heterogeneous and a large fraction has a genetic cause. Copy number variations (CNVs) can be one of the important causes of growth disorders especially in children with severe growth failure, body disproportion, skeletal dysplasia and multiple pituitary hormone deficiency. The aim of our research was to identify clinically significant (pathogenic/likely pathogenic) CNVs associated with syndromic forms of short stature.

Methods: Our study included 40 children with syndromic short stature referred to the Institute of Human Genetics by clinical geneticists. The array‐CGH procedure was performed using Agilent SurePrint G3 Human CGH Microarray 8×60K.

Results:Clinically significant CNVs were detected in 12 patients (30,00%). The most frequently observed CNVs were in 9p24.3 region – two microdeletions and two microduplications all including SMARCA2 gene linked to Nikolaides-Baraitser syndrome. In two patients, 15q26.2-q26.3 microdeletions involving IGF1R gene were detected. Familial deletions including Xp22.33-p22.13 and 8q23.1-q23.3 were detected in patients with familial Turner syndrome and trichorhinophalangeal syndrome, respectively. Other detected CNVs with no clear association with short stature were: 19p13.2-13.12 microduplication, 7q35 intragenic homozygous deletion of CNTNAP2 gene, and microdeletions 2p21-p16.3 and 2q36.3-q37.3.

Conclusion: Chromosomal microarray proves to be useful diagnostic tool for evaluation of syndromic short stature. Establishing the genetic diagnosis is useful in evaluating the response to growth hormone treatment and enables recognition of other family members affected by the same genetic abnormality.

Grants:

Conflict of Interest: None declared

EP13.032 Clinical spectrum of Simpson-Golabi-Behmel syndrome type 1: insights from four cases

Nazlı Büşra Açıkgöz 1, gizem ürel-demir1;2, Pelin Simsek-Kiper1, Gulen Eda Utine1

1Hacettepe University Faculty of Medicine, Pediatric Genetics, Ankara, Türkyie; 2Mersin City Training and Research Hospital, Pediatric Genetics, Mersin, Türkyie

Background/Objectives: Simpson-Golabi-Behmel syndrome type 1 (SGBS1, OMIM #312870) is characterized by pre- and postnatal macrosomia, distinctive craniofacial features, and often mild to severe intellectual disability. The disorder is caused by mutations in the GPC3 gene located on Xq26. This study aims to present the clinical and molecular data of four patients with GPC3 mutations.

Methods: GPC3 gene sequence analysis was conducted in three patients, while GPC3 gene-targeted multiplex ligation-dependent probe amplification (MLPA) analysis was performed in one patient.

Results: The ages of the four patients at diagnosis ranged between 5 months and 7 years. All four patients had global developmental delay and characteristic craniofacial findings, including macrocephaly, coarse facial features, macrostomia, macroglossia, and palate abnormalities. Two of the patients were macrosomic. One patient underwent surgery for diaphragmatic hernia, three patients had visceral organ anomalies such as splenomegaly, double collecting system, and pelvicaliectasis, two patients had supernumerary nipples, and two patients presented with skeletal abnormalities, including scoliosis, six lumbar vertebrae, and polydactyly. Consistent with the clinical diagnosis of SGBS1, GPC3 gene sequence analysis revealed hemizygous disease-causing variants in three patients and MLPA analysis revealed a hemizygous deletion in one patient.

Conclusion: SGBS1 is an overgrowth syndrome that should be considered in the presence of macrosomia, multiple congenital anomalies and intellectual disability in males. Genetic analysis including GPC3 gene sequence analysis and targeted deletion/duplication analysis should be planned accordingly and genetic counselling should be offered, particularly to all women identified as carriers.

Grants: None

Conflict of Interest: None declared

EP13.033 Second opinion for diagnosis – more accurate by an expert in rare diseases: identification of the phenotypic overlap within the dual diagnoses

VASILICA PLAIASU 1, Diana Ozunu1, Gabriela Motei1, Rodica Cretu2, Florin Brezan2, Lucica Ghita2, Salwa Addilami2, Bogdan Pascu2, Monica Panzaru3, Roxana Popescu3, Cristina Rusu3

1INSMC Alessandrescu-Rusescu, Genetics, Bucharest, Romania; 2INSMC Alessandrescu-Rusescu, Pediatrics, Bucharest, Romania; 3Spitalul Clinic de Urgenta pentru Copii Sf Maria, Genetics, Iasi, Romania

Background: 22q11.2 deletion syndrome (DS) is the most frequent copy number variant (CNV) affecting ~1/2,000–4,000 children, resulting in recognizable but variable findings across multiple organ systems.

Duchenne muscular dystrophy (DMD) is the most common type of muscular dystrophy and usually affects males. However, females are also affected in rare instances.

SHOX is a gene (short stature homeobox-containing gene), which plays an important role in bone growth and development. SHOX deficiency as a result of SHOX gene abnormalities causes poor growth in humans, usually in the first few years of life.

More recently, complex phenotypes caused by more than one genetic defect (i.e., dual molecular diagnosis) have also been reported. A dual diagnosis compounds symptoms and impacts the management of the patients.

Material and methods: Patients with a laboratory confirmed chromosome 22q11.2 deletion via fluorescent in situ hybridization (FISH), and a dual diagnosis were retrospectively identified from our cohort.

Results: We report 2 girls with 22q11.2 deletion syndrome and coexisting genetic conditions: Duchenne muscular dystrophy, respectively SHOX deletion syndrome. They were referred to our expert center for genetic counselling after receiving the first diagnoses. Based on clinical evaluation new additional diagnosis, 22q11.2 microdeletion syndrome was suspected, and then it was confirmed for both cases. Complementary parental testing was performed, showing a de novo origin of this chromosomal abnormality.

Conclusion: Patients already diagnosed with a genetic condition and presenting with atypical features require considering the possibility of a concurrent diagnosis. Accurate diagnostics allow for more personalized and appropriate medical management.

Conflict of Interest: None declared

EP13.034 First report of the recurrent 16p11.2 duplication in homozygous state in a patient with pronounced behavioral abnormalities.

Theresia Herget 1, Christine Zühlke2, Maja Hempel1;3, Amelie van der Ven1

1Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; 2Institute for Human Genetics, University of Lübeck, Lübeck, Germany; 3Institute of Human Genetics, Heidelberg University, Heidelberg, Germany

Background/Objectives: Proximal duplication of 16p11.2 is a recurrent chromosomal aberration associated with a broad spectrum of neurodevelopmental and psychiatric symptoms. Typical clinical manifestations include attention deficit hyperactivity disorder, aggression, anxiety, autism, speech/motor delay, intellectual disability, microcephaly and seizures. The duplication is inherited in many cases and the phenotype is subject to incomplete penetrance and variable expressivity.

We here describe a ten-year-old boy, second child to healthy parents from Croatia, coming to medical attention at age four with behavioral problems, notably hyperactivity and severely limited attention span. IQ-testing revealed under-average scores. At age nine he attended special school focussing on mental development and received occupational- and speech therapy. He exhibited serious difficulties with social interaction, especially aggressions against himself/others. He further displayed slurred speech, partly impaired fine motor skills, reduced pain sensation, secondary microcephaly, rather short and slender stature, downslanting palpebral fissures, slightly backwards rotated ears, short index and long fifth fingers. The family history was remarkable for attention deficit, aggression and anxiety in the elder brother but apparently healthy parents.

Methods/Results: Array-CGH uncovered a homozygous 16p11.2 microduplication (16p11.2 (29652999_30198600)x4 (hg19, GRCh37)) in our patient. Further analyses revealed the 16p11.2 microduplication in the heterozygous state in both parents and the elder brother.

Conclusion: To our knowledge, our patient presents the first case carrying a homozygous 16p11.2 duplication. We propose that the fourfold 16p11.2-copy gain explains the aggravation of neuropsychiatric features that go far beyond the abnormalities previously reported for heterozygous 16p11.2 microduplication carriers. This case thereby supports a dosage-associated genotype-phenotype correlation of 16p11.2 CNVs.

Grants: None.

Conflict of Interest: None declared

EP13.035 Genotype-phenotype study of three patients with Snijders Blok-Campeau syndrome

Elena Sukarova-Angelovska 1, Predrag Noveski2, Dijana Plaseska-Karanfilska2, Sara Veleska3, Slavica Trajkova4, Danilo Nonkulovski5, G. Ilieva3, Liljana Tasavska -Rmus3, Eugene Lee6, Jihye Kim6

1University Clinic for Pediatric Diseases, Faculty of Medicine, Ss. Cyril and Methodius University in Skopje, 1000 Skopje, 1Department of Clinical Genetics, Skopje, Macedonia; 2Research center for genetic engineering and biotechnology, Skopje, Skopje, Macedonia; 3University Clinic for Pediatric Diseases, Faculty of Medicine, Ss. Cyril and Methodius University in Skopje, 1000 Skopje, Department of Clinical Genetics, Skopje, Macedonia; 4University of Turin, Department of Medical Sciences, Turin, Italy; 5University Clinic for Pediatric Diseases, Faculty of Medicine, Ss. Cyril and Methodius University in Skopje, 1000 Skopje, Department of Neurology, Skopje, Macedonia; 63Billion Inc, Seoul, Korea, Rep. of South

Background: Snijders Blok-Campeau syndrome (# 618205) is rare autosomal dominant developmental disorder caused by variants in CHD3 gene that impairs production of energy and disrupts chromatin remodeling of the nucleosome. The gene is mostly expressed in the developing brain, mainly affects speech development. Clinical presentation varies in described patients, both in phenotypic appearance and intellectual impairment. Variable expressivity is present in familial cases as well.

Methods: We performed exome sequencing (ES) in three patients from three unrelated non-consanguineous families. Phenotypic evaluation was performed in all.

Results: The patients aged 2, 14 and 16 years had variable presentation of dysmorphic features - macrocephaly, hypertelorism, broad forehead, midface hypoplasia, broad nose and pointed chin, one had nystagmus. One had average IQ, other two had developmental delay of variable degree, Speech impairment including minor deficiency to apraxia, dysarthria or no speech were present in all. Generalized seizures and autism-like disorder was present in some of the patients. Also other features not previously described as nystagmus were noticed. WES revealed de novo heterozygous variants of CHD3 NM_001005273.3:c.3505C>T) and (NM_001005273.3:c.2896C>T) in two patients, while in one patient where variant of unknown significance was detected (NM_001005273.3:c.1094T>C) segregation analysis was not performed.

Conclusion: Variants in CHD3 gene are predominantly in ATPase and helicase domain, however other variants along the gene are described with the similar phenotype, some with still unrevealed pathogenicity. Further delineation and broadening of the phenotypic spectrum could facilitate revealing the exact gene function in chromatin modulation.

Grants: No

Conflict of Interest: None declared

EP13.036 The Challenge of Expanding the Phenotype of a Rare Disease: HDR syndrome #38529

Shelly Lev-Hochberg 1, annick REIN ROTHSCHILD1, Ben Pode Shakhed1, Odelia Chorin1

1Sheba, Ramat Gan, Israel

Background/Objectives: Heterozygous variants in GATA3, a member of the GATA-binding family of transcription factors, are associated with Hypoparathyroidism, Sensorineural Deafness and Renal anomalies (HDR) syndrome, also termed Barakat Syndrome (OMIM, 146255).

Similarly to many rare diseases, limited information regarding clinical symptoms pose a challenge in understanding the significance of previously unreported symptoms.

We aimed to explore the clinical characteristics of HDR syndrome in a large family demonstrating clinical age-dependent variability, along with novel manifestations.

Methods: The proband, a 19-year-old female, was referred for evaluation of severe intestinal disease resistant to classical IBD therapy along with congenital bilateral hearing loss. Maternal family history significant for hearing loss and kidney disease of unknown etiology. Trio exome sequencing was pursued, with further delineation of phenotypic features by use of self-reported symptoms in digital platforms.

Results: Exome sequencing revealed a maternally-inherited GATA3 pathogenic variant (c.647_681Dup; p.Glu228ThrfsX50, NM_001002295.2) establishing the diagnosis of Barakat Syndrome in the proband and her mother. Digital patient platforms (Tik Tok, Facebook, Twitter) were utilized to collect data regarding unreported symptoms including joint hyperlaxity, autonomic dysfunction, postural tachycardia, neuropathic pain and intestinal disorders.

Conclusions: Accurate diagnosis of rare diseases is key, not only for early detection and prevention, but also for family planning. Digital platforms, which boast ~290 million views of videos with #rare diseases can be used to enhance rare disease knowledge. Data from digital platforms along with medical literature, are tools that could add to our knowledge regarding rare diseases possiblly expanding known phenotypic spectrum.

Conflict of Interest: None declared

EP13.037 Unveiling the Rarity: A Case Report on Warsaw Breakage Syndrome

Olivia Kaas Laursen 1, Janni Majgaard Jensen1

1Aalborg University Hospital, Department of Clinical Genetics, Aalborg, Denmark

Background/Objectives: Warsaw Breakage Syndrome (WABS) is an ultra-rare disorder with fewer than 30 reported cases documented in the scientific literature to date, and interestingly the vast majority of cases are children. With the aim of raising awareness, we report of a case diagnosed in Denmark. WABS is caused by pathogenic bi-allelic variants in the DDX11 gene. DDX11 is involved in DNA repair, sister chromatid cohesion and consequently involved in regulation of gene transcription.

Methods: Prenatal array CGH was performed after amniocentesis with a normal result. Trio whole-exome sequencing (WES) was conducted postnatally and revealed two heterozygous likely pathogenic variants inherited from each parent: NM_030653.4:c295G>T,p.Glu99* and NM_030653.4:c.667delG, p.Glu223Lysfs*4

Results: We report a case of a girl who was born preterm at week 28 due to intrauterine growth restriction (IUGR). Birth weight: 441 g. The patient experienced prenatal and postnatal growth retardation, severe microcephaly, and bilateral hearing loss. At 7 months old, the length was -4,9 SD, weight -5,2 SD and head circumference -7,6 SD. She exhibited dysmorphic features with hypertelorism, upslanted palpepral fissures, short nose, transversal philtrum-cleft, downturned corners of mouth, and sacral pits.

Conclusion: WABS should be considered in patients with the triad of pre- and postnatal IUGR, severe microcephaly and bilateral hearing loss due to bilateral hypoplasia of the cochlear and vestibular nerves. However, the variation in the phenotype still requires clarification and comprehensive phenotyping of patients with WABS is essential, as mild cases of the syndrome may go underdiagnosed, especially in the adult population.

Conflict of Interest: None declared

EP13.038 Expanding the clinical phenotype of patient with homozygous variant in NIN gene

Shima Zamanian Najafabadi 1, Zhila Ghaderi1, Zeinab Ghorbanoghli1, Fatemeh Fatehi1, Fatemeh Ahangari1, Hossein Najmabadi1;2, Ariana Kariminejad1

1Kariminejad-Najmabadi Pathology & Genetics Center, Tehran, Iran; 2Genetics Research Center, University of Social Welfare & Rehabilitation Sciences, Tehran, Iran

Seckel syndrome (SS) is an extremely rare autosomal recessive disorder characterized by intrauterine growth retardation, severe short stature, severe microcephaly, small ears, hypotelorism, prominent nose, developmental delay and intellectual disability. Cardiovascular, dental, and ocular manifestations have also been reported.

Seckel syndrome is associated with biallelic variants in nine genes: ATR, CENPJ, CEP63, CEP152, DNA2, NIN, NSMCE2, RBBP8, and TRAIP. To date there is one report of two sisters with severe short stature, microcephaly, and developmental delay with compound heterozygote variants in NIN gene and one paper reporting homozygote variant in NIN gene with progressive, high-frequency sensorineural hearing loss in 4 siblings. In view to the scarcity of cases with NIN variants the relationship between the phenotype and gene is provisional and our case gives further evidence regarding the phenotype related to NIN gene variants.

Here, we report a patient with a homozygous variant in exon 2 of NIN gene defined as c.3407_3409del (p.Glu1136del). Clinical findings in our patient were characteristic of Seckel syndrome including microcephaly, prominent nose, intellectual disability and severe short stature. In addition, this patient had bilateral hearing loss, which was not reported in SS cases associated with NIN gene before. WES was reanalyzed for other genes which are known for deafness and no variant was identified. Also, family history of deafness was not present in the pedigree.

This is the first report of a patient with Seckel phenotype and deafness associated with NIN gene.

Grants: There is no grants to disclose

Conflict of Interest: None declared

EP13.039 A rare case of two consecutive pregnancies with tetra-amelia syndrome due to a novel WNT3 pathogenic variant in a family of Roma origin

Kameliya Kercheva 1;2, Irena Bradinova1;2, Silvia Andonova1, Radostina Raynova1, Violeta Dimitrova2;3, Alexey Savov1;2

1UHOG “Maichin dom”, National genetic laboratory, Sofia, Bulgaria; 2Medical University Sofia; 3UHOG “Maichin dom”, High risk pregnancy clinic, Sofia, Bulgaria

Background/Objectives: Tetra-amelia is a rare congenital anomaly with an incidence of 1.5-4/100,000 births. It is characterized by complete absence of all four limbs and might include anomalies of the cranium, face, heart, lungs, urogenital system, etc. The precise etiology remains uncertain, as it can present as a sporadic anomaly or part of a broader genetic syndrome. Here we present a clinical case of a Roma couple with a reproductive history of one spontaneous abortion and one uneventful pregnancy followed by two consecutive pregnancies marked by the manifestation of tetra-amelia. A positive family history for tetra-amelia on paternal side (father’s sibling) was reported. The first affected fetus was with tetra-amelia and cleft lip and palate; the pregnancy was terminated in 19 g.w. and no genetic analysis was performed. The second affected fetus was again with tetra-amelia and facial defects and after termination of this pregnancy in 12 g.w. a genetic testing was initiated in the family.

Methods: Sanger sequencing of the WNT3 gene was performed on DNA samples from the affected fetus and the parents.

Results: A homozygous pathogenic variant WNT3:c.929G>A/p.Cys310Tyr was found in the tetra-amelia fetus. Both parents were confirmed to be carriers of the mutation. The fifth pregnancy in the family was achieved during COVID-19 pandemic and a healthy infant was born after regularly preformed ultrasound examinations.

Conclusion: The significance of genetic counseling in instances of tetra-amelia syndrome needs to be highlighted, especially in ethnic groups which adopt endogamy, even when they assert non-consanguinity.

Grants:

Conflict of Interest: None declared

EP13.040 Disease associated Chromosome 1 Copy Number Variations: A Retrospective Case Series

Gulnihal Bulut 1, Bilge Ozsait Selcuk2, Somayyeh Heidargholizadeh1, Tugba Kalayci2, Ayça Dilruba aslanger2, Selen Cerci1, Birsen Karaman3

1Istanbul University Institute of Health Sciences, Genetics Department; 2Istanbul University Faculty of Medicine, Department of Medical Genetics; 3Istanbul University Institute of Child Health, Department of Pediatric Basic Sciences

Background/Objectives: Copy number variations (CNV) are described as deletions and duplications of DNA segments ranging in size from 50 base pairs to megabases. Chromosome 1, the largest human chromosome, contains 2,061 genes associated with many rare genetic diseases. This study aims to contribute to the genotype-phenotype map by interpreting the CNVs detected on chromosome 1.

Methods: Data from a total of 3473 cases using array-based comparative genomic hybridization (a-CGH) between 2012 and 2023 were included in the study. When microscopic chromosomal anomalies were excluded, 15 (8 prenatal and 7 postnatal cases) with CNV on chromosome 1 were identified. The variants were analyzed according to the criteria of the American College of Medical Genetics and Genomics (ACMG). The clinical findings were evaluated for each CNV region. Databases and related literature were used to compare the genotype–phenotype correlations.

Results: The most frequently detected CNV was 1q21.1 in 8 cases (53.3%), followed by 1p36 in 3 cases (20%), and changes in the 1q22, 1q23.3, 1p31.3p31, and 1q43q4 regions in one case respectively. The severity of the phenotype varied, and pathological ultrasound findings such as short tubular bones, cystic hygroma, and ventriculomegaly were generally seen in prenatal cases, whereas developmental delay (DD), intellectual disability (ID), and dysmorphic features were predominant in postnatal cases.

Conclusion: The phenotypic difference in chromosome 1 alterations might be due to the change in gene expression at the breakpoints.

Grants: This project was supported by the Istanbul University Scientific Research Projects (BAP) project no: TYL-2021-38090

Conflict of Interest: None declared

EP13.041 Gross deletion in KIF11: A de novo occurrence

Hilal Şentürk 1;2, burak eraslan3, sinan akbaş1, Turkan Uygur4, Ayça Dilruba Aslanger1, Volkan Karaman1, Gozde Yesil Sayin1, Guven Toksoy1, Zehra Oya Uyguner1

1Istanbul University, Istanbul Faculty of Medical, Department of Medical Genetics, Istanbul, Turkey, Genetics, Istanbul, Türkyie; 2Istanbul University, Institute of Graduate Studies in Health Sciences, Istanbul, Turkey, Genetics, Istanbul, Türkyie; 3Adnan Menderes Univercity Faculty of Medicine, Medical Genetics; 4Altınbaş University Medical Park Bahçelievler Hospital, Pediatric Neurology, İstanbul, Türkyie

Background/Objectives: A 6-month-old female with microcephaly and retinal detachment, previously analyzed through a clinical exome sequencing at another center with no disease-associated alterations, underwent exome sequencing (ES) at our center, revealing a gross heterozygous deletion in KIF11. KIF11 (Kinesin Family Member 11), a microtubule motor protein, is involved in mitosis-related processes, encompassing microtubule sliding and cell division. Mono-allelic pathogenic variants in KIF11 are associated with autosomal dominant MCLMR syndrome (MCLMR, MIM# 152950), which is characterized by microcephaly, facial dysmorphisms, intellectual disability, chorioretinopathy.

This study presents the clinical and genetic findings of the case, as well as assessing the efficacy of the genetic diagnostic methods.

Methods: DNA and RNA were isolated from the peripheral blood of the index and family members; ES was performed on the index, and fragment analysis along with Sanger sequencing were performed on cDNA.

Results: Copy number variation (CNV) analysis of the ES data unveiled a ~5 kb deletion across exons 11-14 of the KIF11 gene, subsequently validated through fragment analysis and Sanger sequencing on cDNA. The deletion resulted loss of 219 amino acids without causing a frameshift predicted to the formation of a truncated protein. Parental analysis indicated a de novo occurrence in our index.

Conclusion: The study highlights that the initial lack of CNV analysis in ES data resulted in the absence of genetic basis detection for the clinical findings. While CNV analyses improve diagnostic yield, a second method is required for confirmation.

Grants: This study was supported by Scientific Research Projects Coordination Units of Istanbul University (Project number: TDK-2023-39856)

Conflict of Interest: None declared

EP13.043 Impact of recurrent 17q12 microdeletion across three generations: a family with complete penetrance

Inês Beleza 1, Ana Vilan2;3, Sofia Dória3, Ana Grangeia1;3

1São João University Hospital, Serviço de Genética Humana, Porto, Portugal; 2São João University Hospital, Serviço de Neonatalogia, Porto, Portugal; 3Faculdade de Medicina da Universidade do Porto - FMUP, Porto, Portugal

Background/Objectives: The 17q12 recurrent deletion syndrome (RCAD syndrome) is a contiguous gene syndrome, encompassing three major phenotypes of variable functional/structural abnormalities of the kidney and urinary track, Maturity-Onset Diabetes of the Young type 5 and neurodevelopmental/neuropsychiatric disorders.

Methods: We describe a family with three consecutive affected generations, composed of maternal grandmother, a mother and her female neonate. The clinical picture of these three relatives consisted of: prematurity, unilateral renal agenesis, chronic kidney disease and spastic diplegia (on the mother); diabetes (diagnosed at 14 years of age) and chronic kidney disease (on the maternal grandmother); unilateral multicystic dysplastic kidney (prenatal diagnosis), and preterm labour with low birth weight (on the female neonate).

Results: The genetic study of the mother, through high-resolution array comparative genomic hybridization, identified a microdeletion on the chromosomal region 17q12, consistent with her clinical findings. Subsequently, genetic testing was performed on the other two affected female relatives, thus demonstrating a multigenerational diagnosis of 17q12 recurrent deletion syndrome with variable clinical expression.

Conclusion: These three relatives demonstrate different phenotypic manifestations, associated with the 17q12 recurrent deletion syndrome, whose heritability presents in an autosomal dominant manner, with approximately 25% of cases due to parental inheritance (and the remaining 75% of cases presenting as de novo deletions). This chromosomal microdeletion has recognized phenotypic variability, but its exact penetrance is currently unknown. We believe the present case of this family supports a penetrance of virtually 100% and variable expressivity for this microdeletion syndrome, in accordance with recent studies.

Grants: Nothing to declare.

Conflict of Interest: None declared

EP13.045 Is the Homozygous Mutation Pattern Reminding Uniparental Disomy on DHCR7 Gene Responsible for Smith-Lemli-Opitz (SLO) Syndrome ?

Zeyneb Berrin KOSEM 1, İlknur Sunar2, Ayse Sena Demircioglu3, Rukiye Akarsu4, İsmail Gencay5, Ibrahim Akalin6

1Biruni University, Faculty of Engineering and Natural Sciences, Istanbul, Türkyie; 2Metagentech Center for Genetic Diseases, Istanbul, Türkyie; 3Kocaeli University, Faculty of Medicine, Istanbul, Türkyie; 4Abdullah Gul University, Faculty of Life and Natural Sciences, Kayseri, Türkyie; 5Uskudar University, Faculty of Engineering and Natural Sciences, Istanbul, Türkyie; 6Metagentech Center for Genetic Diseases, Istanbul, Türkyie

Background/Objectives: Syndactyly of the second and third toes forming a Y-shape is most pathognomonic finding of several congenital syndromes, in particular Smith-Lemli-Opitz (SLO). The latter is a rare genetic disorder caused by mutations in the 7-dehydrocholesterol reductase (DHCR7) gene encoding last step of cholesterol biosynthesis. Most common characteristic features are microcephaly, growth retardation and intellectual disability. Here we present two new cases having similar clinical and mutational pattern.

Methods: Patient A, aged 23 months and premature Patient B, aged 5 months, both presented with 2-3 toe syndactyly and developmental delay. They were examined by Whole Exome Sequencing (WES).

Results: Five similar benign homozygous mutation patterns were identified in the DHCR7 gene, where those were heterozygous for both parents of each family. They separately revealed heterozygous PPM1D and KMT2B mutations, particularly. Developmental acceleration observed for both after butter feeding.

Conclusion: Though patients’ clinical findings might conquer Jansen-de Vries Syndrome (PPM1D) and Intellectual developmental disorder-68 (KMT2B) mutations individually, the homozygous mutation pattern of DHCR7 gene might resemble micro-uniparental disomy for SLO.

Grants:

Conflict of Interest: None declared

EP13.046 First familial case of 16p13.3 microdeletion syndrome – two new cases illustrating the phenotype of this recently described syndrome

Raquel Rodrigues 1, Catarina Macedo1, Mariana Soeiro e Sá1, José Paulo Monteiro2, Ana Sousa1, Ana Berta Sousa1

1Unidade Local de Saúde de Santa Maria, Serviço de Genética Médica, Lisboa, Portugal; 2Hospital Garcia da Horta

Background/Objectives: Deletions within 16p13.3, encompassing TBC1D21 and ATP6V0C, have been associated with a novel microdeletion syndrome. This emerging clinical entity is characterized by microcephaly, developmental delay (DD)/intellectual disability (ID), and epilepsy that typically manifests within the first few years of life. We report the first familial case and contribute to the recognition of this rare syndrome as a distinct clinical condition.

Methods: We report a 2-year-old boy, referred to our clinic due to DD and behavioral problems. On examination, he presented hirsutism, microcephaly, brachycephaly, arched eyebrows, mild synophrys, hypertelorism, bilateral epicanthus, upslanted palpebral fissures, depressed nasal bridge, and 5th finger clinodactyly. EEG revealed mild diffuse slowing of brain activity, with no recorded seizures. His mother had DD/ID and refractory epilepsy since age 14, manifested as tonic-clonic seizures with right frontal focal onset and nocturnal predomination. She presented with microcephaly and a similar facial gestalt.

DNA samples were analyzed by aCGH.

Results: Index and mother aCGH analysis revealed a heterozygous deletion in 16p13.3, spanning 322.62kb (2429144_2751764; hg19).

Subsequent analysis of the unaffected maternal grandmother yielded normal results. The maternal grandfather was not available for testing.

Conclusion: This first report of familial 16p13.3 microdeletion contributes to the recognition of this syndrome as a distinct clinical condition. Mother and son display notably similar dysmorphism, however differ from patients in the literature, suggesting familial traits rather than a recognizable facial gestalt. We report the latest onset of epilepsy to date, highlighting the variable expressivity of this feature.

Conflict of Interest: None declared

EP13.047 Discrepant results between SNP array and MS-MLPA in a patient with Prader-Willi syndrome

Laura van Zutven 1, Jeroen Knijnenburg2, Gerthe Kerkhof3, Eunice Stefanou1, Marielle Alders4, Sharmila Chitoe-Ramawadhdoebe2, Diana Rongen - van Nispen1, Marga Thomassen1, Yvette Van Ierland1, Hennie Brüggenwirth1

1Erasmus MC, Department of Clinical Genetics, Rotterdam, Netherlands; 2Leiden University Medical Centre, Department of Clinical Genetics, Leiden, Netherlands; 3Erasmus MC, Department of Pediatric Endocrinology, Rotterdam, Netherlands; 4Amsterdam UMC location AMC, University of Amsterdam, Amsterdam Reproduction & Development Research Institute, Department of Human Genetics, Amsterdam, Netherlands

Background/Objectives: Prader-Willi syndrome (PWS) is an imprinting disorder characterized by low birth weight, hypotonia and feeding difficulties during infancy, followed by obesity, developmental delay and short stature later in life. PWS can be caused by a 15q11q13 deletion on the paternal allele, maternal uniparental disomy (UPD) of chromosome 15 or an imprinting defect on the paternal allele, all resulting in the loss of paternal expression of genes that are silenced on the maternal allele.

Methods: We present a 1-year old male patient with hypotonia, neonatal feeding difficulties, mild motor delay and facial features suggestive of PWS where SNP array and Methylation Sensitive(MS)-MLPA showed discrepant results.

Results: Routine trio SNP array in blood showed a maternal UPD15, consistent with PWS. However, MS-MLPA showed a pattern suggestive for mosaic hypermethylation rather than full hypermethylation. Repeated testing on DNA from a second blood sample as well as skin biopsy and buccal swap showed identical results, as well as Genome-wide methylation analysis using Illumina Infinium MethylationEPIC array.

Conclusion: We cannot explain the discrepancy between SNP array and MS-MLPA/EPIC array results in our patient. If this pattern was the result of two different trisomic rescues, resulting in one cell line with and one without UPD15mat, a mosaic homozygous stretch with SNP array would be expected. However, there was no indication for any mosaic homozygous stretch of DNA nor for a mosaic gain or loss. We speculate that our findings are caused by a rare imprinting defect, which is not detectable by the current diagnostic.

Grants:

Conflict of Interest: None declared

EP13.048 Diagnosis of challenging cases through next-generation sequencing

Darina Kachakova 1;2, Kunka Kamenarova1;2, Kalina Mihova1;2, Martin Georgiev1;2, Gergana Stancheva1;2, Valentina Peycheva1;2, Ivanka Dimova2;3, Radka Kaneva1;2

1Molecular Medicine Center, Department of Medical Chemistry and Biochemistry, Medical Faculty, Medical University – Sofia, Sofia, Bulgaria; 2Laboratory of Genomic Diagnostics, Department of Medical Chemistry and Biochemistry, Medical Faculty, Medical University – Sofia, Sofia, Bulgaria; 3Medical University of Sofia, Department of Medical Genetics, Medical Faculty, Medical University of Sofia, Sofia, Bulgaria

Background/Objectives: Diagnosis of syndromic diseases is a real defiance because of overlapping phenotypic features and variability in clinical presentation. Whole exome sequencing (WES) is a powerful tool for precise diagnosis and for shortening the diagnostic odyssey.

Methods: The following patients were referred to Molecular Medicine Center for genetic diagnosis through WES by NovaSeq6000: two patients with obscure Kabuki syndrome, one with X-linked panhypopituitarism, adrenoleukodystrophy, left ventricular hypertrophy and other symptoms and one with Noonan syndrome.

Results: In the patient with obscure X-linked panhypopituitarism we found pathogenic and variant of unknown significance (VUS) in HERC1 and VUS in CAMTA1 which explain the symptoms. The two discovered variants in HERC1 were with variant allele frequency of 0.75 and 0.796 which was also confirmed by Sanger sequencing and it is probably due to mosaicism. In the patient with Noonan syndrome point mutations explaining the symptoms were not found but CNV analysis of WES data showed two separate deletions, around 7 Mbp each in the 18p chromosome. Thereafter microarray analysis was performed which confirmed the result and showed a single 18p11.32-p11.21 - 13.93 Mbp deletion. Likely pathogenic variant in TFAP2B was found in one of the patients with Kabuki syndrome which changes the diagnosis to Char syndrome. In the other patient referred with Kabuki syndrome likely pathogenic variant in PAX3 for Waardenburg syndrome was found along with VUS in KDM6A and SETD2.

Conclusion: WES contributes for substantially to genetic diagnosis of challenging cases helps distinguishing syndromes with common symptoms.

Grants: D01-302/17.12.2021, D01-165/28.07.2022

Conflict of Interest: None declared

EP13.049 Heterozygous PBX1 gene variant associated with multiple congenital anomalies in a pediatric patient.

Karolis Slibinskas 1, Giedrė Kojelienė1;2, Roma Liutkeviciutė2, Ivan Gavriljev3, Rasa Ugenskienė1, Rasa Traberg1

1Lithuanian University of Health Sciences, Department of Genetics and Molecular medicine, Kaunas, Lithuania; 2Lithuanian University of Health Sciences, Department of Pediatrics, Kaunas, Lithuania; 3Lithuanian University of Health Sciences, Department of Children Rehabilitation, Kaunas, Lithuania

Background: Pre-B cell leukemia factor 1 (PBX1) is essential for fetal development, hematopoiesis, organogenesis. Heterozygous nonsense PBX1 variant can cause renal and urinary tract abnormalities, neurodevelopmental disorders, facial and skeletal defects, deafness. In individuals with heterozygous missense variant pulmonary hypoplasia, cardiac and sexual development defects are the most common. In the literature, the genotype - phenotype correlation remains inadequately explored, necessitating further research.

Methods: The proband was 15 months old female delivered at 38 weeks of gestation by cesarean section. Birth weight was 2884 g (3‰), birth height 49 cm (<10‰). Pregnancy was complicated by polyhydramnios, cystic hygroma. After birth due to respiratory insufficiency intubation was implemented. Phenotypically newborn was dysmorphic: orbital hyperthelorism, low-set ears, genital hypoplasia, low muscle tone. After more investigations, various pulmonary anomalies (tracheomalacia, bronchomalacia, separated sternum) were detected. Echocardiography showed atrial septal defect and patent ductus arteriosus. Brain MRI revealed slight hypoplasia of left temporal lobe. At 3 month of age seizure with muscle spasm was noticed and sleep EEG showed chronic multifocal activity. That led to the generalized epilepsy diagnosis. Diagnostic Inventory for Screening Children (DISC) was performed and motoric developmental delay confirmed. Currently, the proband is artificial ventilation dependent.

Result: Whole exome sequencing (WES) showed heterozygous, likely pathogenic variant NM_002585.4(PBX1):c.869G>A p.(Arg290Gln) (PP3 supporting, PP2, PM2, PM5 supporting, PM6) in 6th exon. Both parents were negative for the variant.

Conclusions: we report case of PBX1 missense variant with severe pulmonary developmental anomalies, patent ductus arteriosus what is compatible with phenotype reported in the literature.

Conflict of Interest: None declared

EP13.050 An atypical case of Nablus mask-like facial syndrome caused by 8q21.3q22.1 microdeletion

Anastasios Mitrakos 1;2, Kyriaki Kekou1, Faidon-Nikolaos Tilemis1;2, Maria Svingou1, Georgios Papadimas3, Christalena Sofocleous1, Jan Traeger-Synodinos1, MARIA TZETIS1

1National and Kapodistrian University of Athens, Laboratory of Medical Genetics, Athens, Greece; 2University Research Institute for the Study of Genetic and Malignant Disorders in Childhood, Athens, Greece; 3National and Kapodistrian University of Athens, 1st Department of Neurology, Athens, Greece

Background/Objectives: Nablus mask-like facial syndrome (NMLFS) is a very rare condition first described in a 4-year-old boy in the city of Nablus, caused by a deletion of the 8q22.1 region. The hallmark feature of the syndrome is a characteristic expressionless facial appearance resembling a mask, with tight glistening facial skin, blepharophimosis and upswept frontal hairline. Here we report on a 35-year-old male with 8q21.3q22.1 microdeletion who presents with hypotonia, cryptorchidism, laryngomalacia, mitral regurgitation, keratoconus, ADHD, psychomotor delay and behavioral disorder. Notably, he only has mild dysmorphic facial features and additionally presents with histopathological lesions consistent with myopathy.

Methods: Whole Exome Sequencing was performed on Illumina NextSeq-500 instrument (IDT xGen Exome Research v2). Variant calling and CNV analysis was performed with VarSome Clinical (Saphetor, Switzerland). Chromosomal microarray analysis was performed using the high-resolution 4x180K G3 CGH + SNP microarray platform. Analysis was performed with CytoGenomics v.5.2 (Agilent Technologies, USA).

Results: A heterozygous deletion of ~8.8Mb of 8q21.3-q22.2 was detected by WES analysis and confirmed by CMA (arr[GRCh37] 8q21.3 - q22.1 (89,148,404-98,017,865)x1). No additional (likely) pathogenic SNVs were revealed to explain myopathic features.

Conclusion: To this day, 26 subjects carrying 8q22.1 deletions have been reported in the literature. Of those only 13 present with typical features of NMLFS suggesting that the deletion although causative is not always sufficient for manifesting NMLFS. Since the pathogenetic mechanism remains unknown, the study additional individuals, like our patient, where a partial phenotype is observed may provide insights into the underlying processes leading to this rare syndrome.

Grants: -

Conflict of Interest: None declared

EP13.051 Expanding the phenotype associated with splice acceptor mutations in the NPHP3 gene

chumei li 1, Sarab Mohamed2

1McMaster University Medical Centre, Pediatrics, Hamilton, Canada; 2McMaster University Medical Centre, Pathology, Hamilton, Canada

Background/Objectives: Mutations in splice acceptor site in the NPHP3 gene are a very rare cause of ciliopathy. We report a case of a newborn girl, to nonconsanguinous British/German and Italian/Filipino parents, who presented with evolving lethal phenotype and homozygous mutations in the splice acceptor that predicted skipping of the second exon of the NPHP3 gene. The case expands phenotype.

Methods: Phenotype description: Antenatal multicystic enlarged kidneys, severe oligohydramnios and fetal growth restriction noted. After C-section, the placenta was also noted to be small with darkened areas ~30% of the parenchyma. Dysmorphic features noted post-delivery.

Then head ultrasound showed tiny subependymal nodules and punctate echogenic foci suggestive of mineralizing vasculopathy.

Within the first two weeks, patient developed anemia, increasing edema, hypertension and jaundice and was found to have severe left ventricular hypertrophy, fenestrasted PFO/ASD, dysplastic pulmonary valves.

Her condition further deteriorated with pericardial, peritoneal and pleural effusions, pneumothorax, portal vein thrombosis, renal failure and liver failure. She passed away before she turned 3 months.

Whole exome sequencing was performed at GeneDx.

Autopsy consent was obtained and carried out.

Results: WES revealed homozygous changes c2692-2_2694-1del, p.? known to be associated with Renal-Hepatic-Pancreatic Dysplasia. Autopsy confirmed dysmorphic findings and microscopic pathology consistent with Renal-Hepatic-Pancreatic Dysplasia. Additionally, the marrow was hypercellular, the ovaries had cystic follicles, the pancreas had diffuse fibrosis, the heart had asymmetric hypertrophy with insterstial fibrosis and dysplastic intramyocardial blood vessels and hypoplastic pulmonic valve. The brain was smaller, with Alzheimer type 2 gliosis and probable periventricular nodular hypertopia.

Grants: none

Conflict of Interest: None declared

EP13.052 Unraveling the complex phenotype of dual diagnosis - Cerebellofaciodental syndrome and RELN-related lissencephaly

Zeynep Esener 1, Nezir Özgün2, Ahmet Yaramis3

1Balikesir University, Faculty of Medicine, Medical Genetics, Balikesir, Türkyie; 2Mardin Artuklu University, Faculty of Medicine, Pediatric Neurology, Mardin, Türkyie; 3Pediatric Neurology Clinic, Pediatric Neurologist, Diyarbakir, Türkyie

Background/Objectives: Dual diagnosis refers to individuals concurrently exhibiting two or more genetic conditions, adding complexity to the clinical phenotype. Dual diagnoses complicate the diagnostic process, making it challenging to pinpoint specific conditions. Our objective is to present a distinctive case featuring the simultaneous occurrence of Cerebellofaciodental syndrome (CFDS) and RELN-related lissencephaly. This case highlights the challenges of diagnosing and managing individuals with dual genetic disorders.

Methods: Detailed family history, clinical examination and cranial imaging findings were evaluated. The diagnosis was achieved through the clinical exome sequencing (CES).

Results: A 1-year-old female, born to consanguineous parents, exhibits notable clinical features, including facial dysmorphism, low weight, short stature, tetralogy of Fallot, microcephaly, severe developmental delay, cerebellar hypoplasia and pachygria. CES analysis revealed a pathogenic homozygous variant [(NM_001519.4), c.875C>A, p.(Pro292His)] in the BRF1 gene and a pathogenic homozygous variant [(NM_005045.4), c.9800_9803dup, p.(Tyr3268*)] in the RELN gene. Facial dysmorphism findings were consistent with CFDS, while pachygria and severe neurodevelopmental delay aligned with RELN-related lissencephaly.

Conclusions: This report reveals that complicated cases present with a dual diagnosis and that comprehensive gene panels such as CES are useful in elucidating the etiology. To the best of our knowledge, this is a first presentation of a patient with co-occurrence of BRF1 and RELN related disorders in the literature. In cases where clinical findings cannot be explained by a single diagnosis, we emphasize the importance of considering the possibility of dual diagnosis and the significance of detailed clinical assessment in medically complex individuals.

Conflict of Interest: None declared.

Conflict of Interest: None declared

EP13.053 A new case of a homozygous intragenic deletion of the ATAD3A gene responsible for PHRINL syndrome

Estelle Colin 1;2, Clarisse Battault1;2, Magalie Barth1, Marine Tessarech1;2, Agnes Guichet1, Maud Blanluet1, Florence Biquard3, Françoise Boussion3, Clara Houdayer1;2, Pierre Blanc4, Alban Lermine4, Benoit Delorme5, Vincent Procaccio2;5, Céline Bris1;2

1University Hospital of Angers, Genetics, Angers Cedex 9, France; 2University of Angers, Mitovasc Unit, UMR CNRS 6015 INSERM 1083, Angers Cedex 9, France; 3University Hospital of Angers, Gynecology and Obstetrics, Angers Cedex 9, France; 4Laboratoire multisites SeqOIA, Paris, France; 5University Hospital of Angers, Radiology, Angers Cedex 9

Background/Objectives: ATAD3A (ATPase family AAA-domain containing protein 3A) is a mitochondrial membrane protein encoded by nuclear DNA, essential for interaction between mitochondria and the endoplasmic reticulum, and plays an important role in mitochondrial biosynthesis. Bi-allelic alterations in the ATAD3A gene are implicated in a neonatal lethal syndrome combining pontocerebellar hypoplasia, hypotonia, and respiratory insufficiency (PHRINL: pontocerebellar hypoplasia, hypotonia, and respiratory insufficiency syndrome, neonatal lethal - MIM # 618810). So far, only. Twelve cases have been described in the literature.

Methods: We report the case of a neonate who died at seven days of age. Antenatal ultrasound revealed a picture of hydrops fetalis, with a decrease in active fetal movements and cerebellopontine hypoplasia. In the postnatal period, neonatal respiratory distress, corneal opacities, and cardiomyopathy were identified, raising the possibility of PHRINL syndrome.

Results: Using the SNP-Array technique, ACPA identified a region of loss of heterozygosity at 1p36.33, containing the ATAD3A gene. Only one SNP in the area is deleted. Only trio genome sequencing revealed the homozygous deletion due to ATAD3B and ATAD3C paralogs positioned in tandem on chromosome 1p36.33.

Conclusion: We describe a new case of bi-allelic alteration of the ATAD3A gene and report some of the challenges of detecting and interpreting rare rearrangements from next-generation sequencing data.

Grants:

Conflict of Interest: None declared

EP13.054 Cat-eye syndrome in three members of a family with a supernumerary marker chromosome investigated by different methodologies

Michael da Silva1, Bruna Burssed1, Marco Antonio Ramos1, Eduardo Perrone1, Maria Isabel Melaragno 1

1Universidade Federal de São Paulo, Genetics Division, São Paulo

Background/Objectives: The cat-eye syndrome (CES) exhibits a broad spectrum of phenotypic variability, encompassing ocular coloboma, cleft palate, pre-auricular anomalies, congenital heart malformations as well as renal and anorectal anomalies. The syndrome arises from the presence of a small supernumerary marker chromosome (sSMC) derived from chromosome 22, resulting in tetrasomy of a chromosomal region spanning the 22p arm and the proximal region of 22q11.21. We studied a family consisting of three members (a mother, a son, and a daughter) who exhibit a highly variable CES phenotype, with the daughter displaying the most severe phenotype among them.

Methods: The family was studied clinically and through G-banding karyotype, fluorescence in situ hybridization (FISH), multiplex ligation-dependent probe amplification (MLPA), chromosome microarray (CMA), and optical genome mapping (OGM).

Results: All patients presented a karyotype with sSMCs. MLPA and FISH techniques revealed an extra dicentric and bisatellited chromosome with breakpoints in the chromosome 22 low-copy repeat A region, resulting in CES due to the chromosome 22 partial tetrasomy of 22pter → q11.2 including the cat eye chromosome region. CMA showed a ~18,6 Mb triplication, as follows: arr[GRCh37] 22q11.1q11.21(16,888,899_18,644,241)×4. OGM was not informative regarding the characterization of the rearrangement since this technique has a limitation in detecting breakpoints in highly repetitive regions such as low-copy repeats.

Conclusion: Utilizing different cytogenomic and molecular techniques enabled a more precise delineation of the chromosome rearrangement, although the cat-eye chromosome involves a complex region that is challenging to characterize accurately.

Grants: FAPESP, Brazil

Conflict of Interest: None declared

EP13.055 Diabetes mellitus in pregnancy and prevalence of congenital malformations: observations from referrals to a single clinical geneticist in Sri Lanka between 2012 to 2023

Deepthi De Silva 1, Nuwani Wijesooriya2, Romesh Gunasekera3, sriyani Basnayake4, Malka Jayathilake5

1university of Kelaniya, Physiology, Ragama, Sri Lanka; 2University of Kelaniya, Physiology, Ragama, Sri Lanka; 3Lady Ridgway hospital, Plastic surgery, Colombo; 4Lady Ridgway hospital, Orthodontics, Colombo; 5Lady Ridgway hospital, Speech and language therapy, Colombo

Background/Objectives: Poor control of diabetes mellitus (DM) in early pregnancy associated with malformations (holoprosencephaly/ caudal dysgenesis sequences and multiple (VACTER, OAV) or single developmental field defects. Highest risk of malformations in pre-pregnancy DM. Universal DM screening at first antenatal visit and 24-28/40 gestation offered in Sri Lanka.

Methods: Review of referrals to single clinical geneticist between January 2012- December 2023.

Results: 241/1983 referrals (12%) reported DM in pregnancy including 59 (25%) pregestational, 57 (24 %) diagnosed in first trimester, 111(46%) in second/ third trimester and 14 at unknown gestation. In 83 cases, malformations involved multiple developmental fields suggestive of blastogenic insult with only 40% presenting in diagnosed pregestational DM mothers.

Blastogenic insults

Total

Pregestational DM

DM diagnosis T1

DM diagnosisT2 + T3

Unknown gestation

Holoprosencephaly

09

05

04

00

-

caudal dysgenesis

19

09

09

01

-

VACTERL

06

01

01

04

-

OAV

41

13

10

17

01

Femoral-face

08

05

02

01

-

Single field malformations

Cardiac

25

03

07

13

02

Brain

44

06

05

26

07

Genito-urinary

04

-

02

01

01

Cleft lip +/- palate

52

14

11

26

01

Limb

15

02

04

09

-

Other diagnoses

18

01

02

13

02

Conclusion: Despite DM screening in pregnancy, this data suggests that potentially preventable malformations are identifiable in babies due to late diagnosis and poor glycaemic control of pre-gestational diabetic women. Prevention of this requires wider professional and public education, especially as pregnancy termination for fetal anomalies is unavailable in Sri Lanka.

Grant: Medical faculty SDG/23/4

Conflict of Interest: None declared

EP13.056 Unraveling the complexity of Phelan-McDermid syndrome: A multifaceted exploration of clinical and molecular/cytogenetic findings in four cases

Mehmet Akif Yücesoy 1, sinan akbaş1, Gülhanım Memiş1, Esma Nur Konur1, Durmuş Durmaz1, Volkan Karaman1, Leyli Şentürk1;2, Umut Altunoğlu1;3, Esma Şengenç4, Hülya Maraş Genç5, Edibe Pempegül Yıldız5, Adnan Deniz6, Bülent Kara6, Ayça Dilruba aslanger1, Bilge Ozsait Selcuk1, Gozde Yesil Sayin1, Zehra Oya Uyguner1, Birsen Karaman1;7

1Istanbul University, Faculty of Medicine, Department of Medical Genetics, İstanbul, Türkyie; 2İstanbul Bağcılar Training and Research Hospital, Department of Clinical Genetics, İstanbul, Türkyie; 3Koç University School of Medicine (KUSoM), Department of Medical Genetics, İstanbul, Türkyie; 4Biruni University Faculty of Medicine, Department of Pediatric Neurology, İstanbul, Türkyie; 5İstanbul University, Faculty of Medicine, Department of Pediatric Neurology, İstanbul, Türkyie; 6Kocaeli University, Faculty of Medicine, Department of Pediatric Neurology, Kocaeli, Türkyie; 7İstanbul University, Child Health Institute, Department of Pediatric Basic Sciences, İstanbul, Türkyie

Background/Objectives: Phelan-McDermid syndrome (PMS) is a rare neurodevelopmental disorder characterized by diverse phenotypes, including intellectual disability, delayed speech, and autistic features. PMS is caused by either pathological variants or contiguous gene deletions on chromosome 22q13, encompassing the SHANK3 gene. This study aims to present clinical, molecular/cytogenetic findings of four cases highlighting PMS’s multifaceted nature and complexity.

Methods: The diagnosis of PMS in cases was achieved through the analysis of blood samples using chromosome analysis, Fluorescence in situ Hybridization (FISH), array-comparative Genomic Hybridization (CGH), exome sequencing (ES) (covering copy number variations), and Sanger sequencing for confirmation.

Results: Three patients had multiple congenital anomalies, developmental delay, and facial dysmorphic features, while the fourth patient had only epilepsy and autism. In the first case, chromosome analysis was normal, and array-CGH analysis revealed a 5 Mb deletion involving the SHANK3 gene. In the second case, chromosomal analysis unveiled an unbalanced de novo inversion at 22q, resulting in a deletion involving the SHANK3 gene. In the third case, a 2.3 Mb deletion was detected in the 22q13.3 region in the array-CGH study due to mosaic ring chromosome 22, which was detected in chromosome analysis. In the fourth case, ES revealed a heterozygous frameshift c.3950_3962del/p.(Arg1317LeufsTer25) variant in SHANK3 gene.

Conclusion: PMS is a complex neurodevelopmental disorder characterized by diverse clinical features resulting from various molecular/cytogenetic alterations, including chromosomal abnormalities and single nucleotide variations. Understanding underlying genetic mechanisms is crucial for precise genetic counseling and improving genotype-phenotype correlations.

References:

Grants:

Conflict of Interest: None declared

EP13.057 Diagnostic utility of whole-exome sequencing in Iranian patients suspected with Joubert Syndrome

Arezou Karamzade 1, Meisam Babaei2, Neda Golchin3, Saeed Talebi4, Aysun Khalil Nejad Sani Banaei3, Mohammad Keramatipour3;5

1Tehran University of Medical Sciences, Pediatrics Center of Excellence, Children’s Medical Center, Tehran, Iran; 2North Khorasan University of Medical Sciences, Department of Pediatrics, Bojnourd, Iran; 3Watson Genetic Laboratory, North Kargar street, Tehran, Iran; 4Iran University of Medical Sciences, School of Medicine, Department of Medical Genetics and Molecular Biology, Tehran, Iran; 5Tehran University of Medical Sciences, School of medicine, Department of Medical Genetics, Tehran, Iran

Background: Joubert syndrome (JS) is a recessive neurodevelopmental ciliopathy characterized by hypotonia, ataxia, abnormal breathing, intellectual disability and a pathognomonic cerebellar-brainstem malformation recognizable as the molar tooth sign (MTS) in MRIs. Additional features including retinal, renal and/or liver diseases. Over 40 genes implicated in JS, highlighting its genetic heterogeneity. There is significant phenotypic overlap between the JS and other ciliopathies. Recognition of the MTS is crucial in distinguishing JS from other disorders particularly those with cerebellar/vermis atrophy. However, the MTS is not always properly diagnosed. Here, we investigated the utility of whole-exome sequencing (WES) as a diagnostic tool for patients suspected to have JS.

Methods: Thirteen families including 16 patients with possible diagnosis of JS evaluated and referred by pediatric neurologist for analysis. All patients revealed structural defects on brain MRI images but only 7 families showed typical MTS. WES was performed on 13 probands and segregation analysis was conducted on 37 unaffected and other 3 affected members available in pedigrees through sanger-sequencing.

Results: Thirteen strong causative variants detected homozygously in all probands. Ten had pathogenic and three had uncertain significance classifications. Only five variants were novel. One proband diagnosed as Zellweger Syndrome with PEX1 mutated gene. Detecting pathogenic variants in nine of 12 remaining probands, confirmed the JS diagnosis definitely. The most frequently mutated gene was AHI1.

Conclusion: This study demonstrated a diagnostic yield of 76.9% for WES, extended the mutations spectrum of JS in Iran and showed the WES utility in distinguishing two distinct phenotypes with overlapping features.

Conflict of Interest: None declared

EP13.058 Variant c.89G>C p.(Gly30Ala) in the DHCR7 gene as a cause of the mild phenotype in Smith-Lemli-Opitz syndrome

Júlia Martinková 1, Lukáš Ryba1, Emílie Vyhnálková1, Miroslava Balaščaková1, Martin Schwarz1;2, Romana Borská3, Lenka Fajkusova3, Anna Křepelová1

1Charles University and Motol University Hospital, Department of Biology and Medical Genetics, Prague, Czech Republic; 2PRENET - Laboratoře lékařské genetiky s.r.o., Pardubice, Czech Republic; 3University Hospital Brno, Department of Medical Genetics, Brno, Czech Republic

Background: Smith-Lemli-Opitz syndrome (SLOS) is an autosomal recessive disorder caused by pathogenic variants in the DHCR7 gene, resulting in a deficiency of the enzyme 7-dehydrocholesterol reductase. The phenotype is characterised by microcephaly, polydactyly, syndactyly of the toes, cleft palate, underdeveloped external genitalia, organ malformations, intellectual disability and behavioural disorder. SLOS type I has a milder phenotype. SLOS type II is more severe and often results in death during pregnancy or shortly after birth. The diagnosis is confirmed by the presence of an elevated level of 7-dehydrocholesterol (7-DHC) and/or by the detection of a pathogenic variant(s) in the DHCR7 gene.

Methods: Sanger sequencing was used to detect the variants.

Results: We observed 2 unrelated cases: a 4-year-old male patient with syndactyly, hexadactyly, microcephaly, short stature, and a 3-year-old female patient with syndactyly and incontinentia pigmenti. Both patients had elevated 7-DHC levels. We identified a rare heterozygous probably pathogenic variant c.89G>C p.(Gly30Ala) (rs200334114) and a pathogenic variant c.964-1G > C in trans position in the DHCR7 gene (NM_ 001360.2) in both patients.

Conclusion: The p.Gly30Ala variant has previously been reported by others in a mother and her daughter with mild microcephaly, and in a fetus from a terminated pregnancy. These cases were compound heterozygous for the p.Gly30Ala variant and p.Trp151* or p.Thr93Met. Our results suggest that the p.Gly30Ala variant causes a mild phenotype of SLOS.

References: Perez et al., 2023; Blahakova et al., 2007.

Grants: MH CZ - DRO, Motol University Hospital, Prague, Czech Republic 00064203.

Conflict of Interest: None declared

EP13.059 Developmental anomalies related to the FOX genes haploinsufficiency: a case series

Ina Ofelia Focsa1;2, Andreea Tutulan-Cunita 2, Anca Pavel2, Diana Prepelita2, VASILICA PLAIASU3, Liana Ples1;4, Anca Neacsu2, Maria Stratan2, Daniela Blanita5, Natalia Usurelu5, Danai Stambouli2

1University of Medicine and Pharmacy Carol Davila, Medical Genetics, Bucharest, Romania; 2Cytogenomic Medical Laboratory, Bucharest, Romania; 3Institute of Mother and Child Alessandrescu-Rusescu, Bucharest, Romania; 4Bucur Maternity, St. John Emergency Hospital, Bucharest, Romania; 5Institute of Mother and Child, Chisinau, Moldova

Background: The outstanding advancements of genomics have highlighted many new mechanisms at work in human pathology. However, the puzzle of genetic makeup is far from being complete. Human FOX family of transcription factors comprises 50 genes, with some of them acting ubiquitously, while the rest exhibit tissue specificity; all are under the control of multiple signalling via intricate networks involved in cell growth, proliferation, survival and migration, during development and organogenesis, being also involved in stress response, metabolism regulation, immunity, aging, carcinogenesis. Here, we present four patients, including a foetus, with anomalies in several FOX genes and various phenotypes.

Methods: The patients were referred for WES due to a broad spectrum of clinical manifestations including craniosynostosis, brain anomalies, ocular, cardiac or genital involvement.

Results: Sequencing revealed deleterious variants in three FOX genes: a novel c.477C>A in FOXC1 associated with Axenfeld-Rieger syndrome, c.460dupG and c.693C>A in FOXG1, causative for Rett syndrome, and c.1541G>A in FOXP1 linked to an intellectual developmental disorder. Interestingly, some of the clinical signs seen in our patients have not been yet described in FOX gene-related pathology, an example being lobar holoprosencephaly.

Conclusion: NGS has enabled rapid genetic diagnosis, spotlighted the broad phenotypic spectrum of many genes and expanded the clinical presentation related to a specific gene. An early diagnosis is crucial in preventing the incidence of complex and severe disorders. Our study emphasizes the value of NGS technologies in diagnosis of disorders with challenging clinical presentation, bringing new evidences for a better outline of the FOX gene-associated disorders.

Conflict of Interest: None declared

EP13.060 Two cases of WAC-Related Intellectual Disability – from point mutation to contiguous gene deletion

Sónia Custódio 1, Raquel Gouveia Silva1, Oana Moldovan1, Eva Rolo1, Ana Sousa1, Ana Berta Sousa1

1Unidade Local de Saúde Santa Maria, Departamento de Pediatria, Lisboa, Portugal

Background/Objectives: WAC-Related Intellectual Disability (ID) is a recently described recognizable neurodevelopmental disorder. WAC gene, at 10p12.1, encodes a nuclear protein involved in multiple cellular processes with a potential critical role in development.

We describe two cases of this syndrome with distinct genotypes: a microdeletion and a point mutation.

Case reports: Case 1 was a two-year-old girl referred to our Genetics Department for developmental delay (DD), hypotonia, bilateral hydronephrosis, bilateral cataracts, and dysmorphisms (hypertelorism, deep-set eyes, depressed nasal bridge, bulbous nose, bifid tongue and inverted nipples).

Case 2 was a 31-year-old male, first observed at age 8 months, presenting with mild ID, ventricular septum defect, cryptorchidism, and dysmorphism (brachycephaly, frontal bossing, hypertelorism, and cutaneous syndactyly). Previous chromosome analysis showed 46,XY/47,XYY mosaicism, and FGD1 sequencing identified a missense variant, c.614G>T, of uncertain significance.

Results: In case 1, arrayCGH revealed a 9,30 Mb interstitial deletion, at 10p12.31p11.22, spanning 37 annotated OMIM genes, including WAC. In case 2, exome sequencing detected a likely pathogenic WAC variant, c.1048C>T p.(Gln350*). The latter originates a premature stop codon resulting in a truncated protein. Variants were de novo in both patients.

Conclusion: Both patients presented DD/ID, language impairment and some overlapping facial dysmorphism. Observed clinical features were, overall, consistent with other patients in the literature. However, bifid tongue, present in case 1, wasn’t previously described; and cardiac defects, present in case 2, are, interestingly, mainly observed in microdeletion cases.

These patients, bearing distinct genotypes, further contribute to the clinical delineation of this rare syndrome.

Conflict of Interest: None declared

EP13.061 Uncovering the Molecular Etiology in Patients with Arthrogryposis

Dilek Uludağ Alkaya 1, Nilay Gunes1, Buşra Kasap1, Timur Yıldırım2, Aysegul Bursali2, Evren Akpınar2, Mehmet Müfit Orak3, Mehmet Bülent Balioğlu2, SEHIME TEMEL4, Beyhan Tüysüz1

1Istanbul University-Cerrahpaşa, Cerrahpaşa Medical Faculty, Department of Pediatric Genetics, Türkyie; 2University of Health Sciences Turkey, Baltalimani Bone Diseases Training and Research Center, Department of Orthopedics and Traumatology, Türkyie; 3Kartal Lutfi Kirdar Training and Research Center, Department of Orthopedics and Traumatology; 4Bursa Uludağ University, Department of Medical Genetics, Türkyie

Background/Objectives: Arthrogryposis, characterized by multiple joint contractures affecting two or more body parts, displays significant genetic and clinical heterogeneity, with over 400 conditions and more than 320 associated genes. It is divided into three main groups: amyoplasia, distal arthrogryposis, and syndromic group, making it crucial to distinguish sporadic amyoplasia from distal or syndromic types caused by genetic disorders. In this study, we aimed to identify molecular etiology in patients with arthrogryposis.

Methods: Whole exome sequencing analysis was performed on 36 patients with arthrogryposis. Sanger sequencing was conducted to verify the identified variants.

Results: The underlying molecular etiology was identified in 19 families (52.8%). The CHRNG was the most commonly detected gene, found in three patients, followed by the ECEL1 and VPS33B genes, which were present in two patients each. The biallelic FBN3 variants were found in a patient with Beals syndrome suggesting it is a novel candidate gene for arthrogryposis. In a patient, biallelic pathogenic variants were detected in both COL6A3 and NEB genes confirming the dual diagnosis. Additionally, a novel variant was identified in a recently discovered CEP295 gene in a patient. The other identified genes in our group were as follows; KIFBP, H1-4, SETD5, SLC6A9, FBN2, DYNC1H1, MYBPC1, CHST14, and TRPV4.

Conclusion: This study provides novel insights into the molecular mechanisms that underlie arthrogryposis.

Grants: This project is granted by the Scientific Research Projects Coordination Unit of Istanbul University-Cerrahpasa (No 36552).

Conflict of Interest: None declared

EP13.062 ACTB variant in a female with Baraitser-Winter cerebrofrontofacial syndrome and sex reversal.

Henrique Serigatto 1, Nancy Kokitsu-Nakata1, Siulan Vendramini-Pittoli1, Tania Kamiya2, Ana Krepischi3, Carla Rosenberg3, Roseli Zechi-Ceide1

1Hospital for Rehabilitation of Craniofacial Anomalies, University of São Paulo - HRAC/USP, Department of Clinical Genetics and Molecular Biology, Bauru, Brazil; 2Hospital for Rehabilitation of Craniofacial Anomalies, University of São Paulo - HRAC/USP, Department of Cytogenetics, Bauru, Brazil; 3Human Genome and Stem Cell Research Center, Institute of Biosciences, University of São Paulo - IB/USP, Department of Genetics and Evolutionary Biology, São Paulo, Brazil

Background/Objectives: Baraitser-Winter cerebrofrontofacial syndrome (BWCFF; OMIM #243310; #614583) is a very rare autosomal dominant condition characterized by distinct facial gestalt and developmental delay. The etiology of BWCFF is heterogeneous, caused by deleterious variants in two different actin genes: ACTB and ACTG1. Here, we report on a 10-year-old female, evaluated at the Hospital for Rehabilitation of Craniofacial Anomalies, University of São Paulo (HRAC/USP), with a phenotype suggestive of BWCFF but with atypical findings.

Methods: Clinical examination was performed. G-banded karyotype, low-pass whole genome sequencing, and exome sequencing were performed using DNA samples extracted from peripheral blood. Variants were classified according to ACMG guidelines.

Results: Clinical evaluation showed arched eyebrows, ptosis, ocular hypertelorism, long palpebral fissures, broad nose, bifid nasal tip, bilateral cleft lip and palate, macrostomia, short neck, and developmental delay. Karyotype analysis revealed a male karyotype (46,XY). Low-pass genome sequencing confirmed the presence of SRY, and absence of clinically relevant copy number variants. Exome sequencing revealed a heterozygous missense variant in the ACTB gene [NM_001101.5:c.931G>T (p.Asp311Tyr)], classified as likely pathogenic (PM2, PM5, PP2, PP3), which was not previously reported. The Asp311 amino acid substitution have already been reported in three patients with BWCFF, in ClinVar, suggesting a causal relation with part of the patient’s phenotype. Analysis of genes associated with sex reversal showed no alterations.

Conclusion: Clinical and molecular findings are compatible with BWCFF, suggesting that the ACTB variant could partially explain the clinical signs. It remains to be investigated if the sex reversal could also be associated with this ACTB variant.

Grants: CAPES, FAPESP.

Conflict of Interest: None declared

EP13.063 LINKED syndrome- first Polish patient with pathogenic variant in OTUD5 gene

Katarzyna Wojciechowska 1, Whitley Zie2, Anna Kutkowska-Kaźmierczak3, Małgorzata Rydzanicz4, Rafał Płoski4, Monika Lejman1

1Medical University of Lublin, Independent Laboratory of Genetic Diagnostics, Lublin, Poland; 2Medical University of Lublin, 2. Studen Scientific Society, Independent Laboratory of Genetic Diagnostics, Lublin, Poland; 3Institute of Mother and Child, Medical Genetics Department, Warsaw, Poland; 4Medical University of Warsaw, Department of Medical Genetics, Warsaw, Poland

Background: LINKED (LINKage-specific deubiquitylation deficiency–induced Embryonic Defects) syndrome is a developmental disorder caused by pathogenic variants in OTUD5 gene. It is an X-linked disorder which is characterized by central nervous system, craniofacial, cardiac, skeletal, and genitourinary anomalies. Our aim is to discuss the case of a patient with a hemizygous pathogenic variant in OTUD5 gene.

Methods: The genetic analysis was performed by TRIO-based whole-exome sequencing.

Results: Our patient was born on time of healthy, nonconsanguineous parents. His birth weight was -3600g, length- 53 cm, OFC-34 cm. After birth, dysmorphic facial features, dysplastic ears, unilateral cryptorchidism, mild umbilical hernia were diagnosed. Array CGH testing showed normal balanced male karyotype. Molecular analysis showed de novo hemizygous variant c.1558C>T p.(Arg520Trp) in OTUD5 gene. At the age of six years and six months among the main health problems of our patient were: dysmorphic facial features, dysplastic ears, seizures, global developmental delay, speech delay, autistic features.

Conclusion: The case of our patient contributes to the studies of the phenotypes of patients with OTUD5-related LINKED syndrome. It’s the first patient reported in Poland so far.

Conflict of Interest: None declared

EP13.064 Clinical and molecular characteristics of patients with syndromic craniosynostosis

Hilal Onur 1, Nilay Güneş1, Dilek Uludağ Alkaya1, Ali Metin Kafadar2, Beyhan Tüysüz1

1Istanbul University- Cerrahpaşa, Cerrahpaşa Faculty of Medicine, Department of Pediatric Genetics, Istanbul, Türkyie; 2Istanbul University- Cerrahpaşa, Cerrahpaşa Faculty of Medicine, Department of Neurosurgery, Istanbul, Türkyie

Background/Objectives: Craniosynostosis (CS) is the premature fusion of one or more cranial sutures and is clinically defined by the pattern of sutural involvement. The presence of associated malformations indicates syndromic and nonsyndromic forms of CS. Our study aims to investigate clinical and molecular characteristics of syndromic CS patients.

Methods: The study comprised 38 Turkish patients including individuals with Apert (11), Crouzon (11), Saethre-Chotzen (8), Pfeiffer (2), craniosynostosis and dental anomaly (2), and Carpenter (2) syndromes and two additional patients. Eighteen patients had molecular diagnosis confirmed by sequencing analyses.

Results: Seventeen males and 21 females were included, and the median age at admission was 0.63 (0.01-27) years. The head circumference of 10 patients were below -2 standard deviations. Twelve patients underwent craniosynostosis surgery, with a median age at operation of 11 months. Nine patients had ventriculomegaly. Clinical seizures were observed in six patients. The mean IQ of 13 patients who underwent developmental testing was 76,7. Nineteen patients had digit anomalies. Hearing (8/19) and vision loss (7/20) were also present. In molecularly diagnosed patients, mutations were detected in FGFR1 (1), FGFR2 (10), FGFR3 (3), TWIST1 (2), and IL11RA (2) genes. A FGFR3 patient exhibited a novel phenotype characterized by CS and oligodactyly, while another FGFR3 patient diagnosed with hypochondroplasia had also CS. One patient had no mutations in FGFR2, FGFR3, and TWIST1.

Conclusion: CS, being a clinically heterogeneous disorder, should be evaluated through a multidisciplinary approach. To understand its pathogenesis and provide appropriate genetic counseling, both clinical and molecular diagnostic features need to be addressed together.

Conflict of Interest: None declared

EP13.065 A novel RAD21 frameshift variant in a patient with atypical Cornelia de Lange syndrome.

Amal Abd Mouleh 1;2, Syrine Hizem1;2, Yasmina Elaribi1, Houweyda Jilani1, Imen REJEB1;3, Ons Azzabi3;4, Sana Karoui1, Lamia BenJemaa1;3

1Mongi Slim hospital, Department of Congenital and Hereditary Diseases, Tunis, Tunisia; 2Faculty of Medicine of Tunis, University of Tunis ElManar, Human genetics laboratory, LR99ES10, Tunis, Tunisia; 3Mongi Slim hospital, Research laboratory « santé mère enfant » LR22SP01, Tunis, Tunisia; 4Mongi Slim hospital, Pediatrics department, Tunis, Tunisia

Background/Objectives: Cornelia de Lange syndrome (CdLS) is a genetically and clinically heterogeneous multisystem cohesinopathy caused by pathogenic variants in one of the six genes of the cohesin complex. Recently, CdLS was classified as a clinical spectrum including patients with the classical phenotype, as well as those with milder overlapping phenotypes. Variants in RAD21 are rarely observed as a cause of CdLS, and the clinical phenotype is considered as atypical.

We report a new variant in the RAD21 gene in a patient with microcephaly and mild dysmorphic features.

Methods: The clinical assessment was performed according to the Consensus Statement diagnostic algorithm of CdLS. The molecular investigation was performed by whole-exome sequencing (WES).

Results: The proband was a three-month-old female, referred for facial dysmorphism and microcephaly.

The patient’s history revealed gastroesophageal reflux and normal motor development. Prenatal and postnatal growth measurements were correct. Clinical examination showed microcephaly at -3 SD, facial dysmorphism (thin and arched eyebrows, high nasal bridge with bulbous tip, thin upper vermilion and downturned corners of the mouth), cutis marmorata and finger camptodactyly. The clinical score of CdLS was of 3, described as insufficient to indicate molecular testing for CdLS.

WES revealed a heterozygous frameshift variation in the exon 12 of the RAD21 gene; NM_006265.3:c.1599_1602del, classified as likely pathogenic (PVS1, PM2) according to the ACMG criteria.

Conclusion: This case emphasizes the role of WES in the genetic investigation of microcephaly, specially whith non specific dysmorphic features. Accurate diagnosis is essential to give the family the appropriate genetic counseling.

Grants:

Conflict of Interest: None declared

EP13.066 Case report of a homozygous missense variant in the transmembrane domain of GLDN – Further evidence for the role of GLDN in congenital malformations and arthrogryposis

Tim Bender 1, Kirsten Cremer1, Hartmut Engels1, Claudia Perne1, Isabelle Claus1, Jessica Trautmann1, Axel Schmidt1, Stefanie Heilmann-Heimbach2, Pietro Incardona3, Andreas Müller4, Hela Hundertmark1, Sophia Peters1

1University Hospital Bonn, Institute of Human Genetics, Bonn, Germany; 2University of Bonn, Medical Faculty, Next Generation Sequencing Core Facility, Bonn, Germany; 3University of Bonn, Medical Faculty, Core Unit for Bioinformatics Data Analysis, Bonn, Germany; 4University Hospital Bonn, Department of Neonatology and Pediatric Intensive Care, Bonn, Germany

Background/Objectives: Congenital arthrogryposis is a rare (prevalence of approx. 1 in 4,500 to 12,500) but complex condition that exhibits phenotypic and genotypic heterogeneity (PMID: 28726266). Biallelic pathogenic variants in GLDN (OMIM * 608603) cause Lethal Congenital Contracture Syndrome 11 (OMIM # 617194), a clinically severe form of arthrogryposis multiplex congenita. To date only a few cases have been reported.

Case Report: We report on a premature girl, born in the 36th gestational week. Prenatal ultrasound revealed polyhydramnios. She was born with pneumothorax, chylothorax, pulmonary hypoplasia, pulmonary arterial hypertension, contractures of the hands, talipes equinovarus, short forearms, hip dislocation, intraventricular hemorrhage, macrocephaly and low-set ears. She developed congestive heart failure, muscular hypotonia and dysphagia. The family history of the parents showed a stillbirth in the 30th gestational week due to hydrops fetalis and an abortion of unknown cause in the 19th gestational week.

Methods and Results: Postnatal trio whole-exome-sequencing revealed a rare homozygous missense variant in the transmembrane domain of GLDN (NM_181789.4:c.79T>C;p.(Ser27Pro)). According to the ACMG criteria, we classified the variant as of unknown significance. Both parents are healthy heterozygous carriers of the variant.

Conclusion: We report a novel homozygous variant in GLDN in a girl with multiple congenital malformations. The patient presents known symptoms of Lethal Congenital Contracture Syndrome 11 such as pulmonary hypoplasia, talipes equinovarus and shortened long bones, but also novel symptoms, which – to our knowledge – have not been reported before, such as chylothorax and macrocephaly. Hence, our case expands the clinical spectrum of the disease.

Grants: -

Conflict of Interest: None declared

EP13.067 Nijmegen breakage syndrome: complex clinical manifestation

Jelena Jovanovic 1, Gordana Stojanovic1, Olivera Miljanovic1;2;3, Sladjana Teofilov1, Luca Lovrecic4, Alenka Hodžić4, Borut Peterlin4

1Clinical Center of Montenegro, Centre for Medical Genetics and Immunology, Podgorica, Montenegro; 2; 3Faculty of Medicine University of Montenegro, Podgorica, Montenegro; 4University Medical Centre Ljubljana, Clinical Insitute of Genomic Medicine, Ljubljana, Slovenia

Background/Objectives: Nijmegen breakage syndrome (NBS) is a rare autosomal recessive DNA repair disorder, characterized by severe chromosomal instability, caused by mutations in the NBN gene. It is characterized by intrauterine and postnatal growth retardation, craniofacial dysmorphism, progressive microcephaly, immunodeficiency and predisposition for malignancy.

Less common features are congenital anomalies of central nervous, respiratory tract and urogenital system. Clinical manifestations of NBS vary greatly in severity. We report a complex patient with NBS for whom genetic diagnostic was established in early adulthood.

Methods: Three diagnostic, karyotype, aCGH and WES, were performed to establish the diagnostic of NBS.

Results: A 29-year-old women with ovarian agenesis, uterine hypoplasia, amenorrhea and severe facial granulomatous dermatitis was referred for a genetic examination. History and clinical evaluation reveald IUGR, microcephaly, frontoparietal occipital cyst, kidney ectopia, immunodeficiency, hypo end hyperpigmented skin spots. No consanguinity was reported. First diagnostic step showed translocation in karyotype: 46,XX,t(3;10)(p11.2;q23.1), followed by aCGH analysis which showed the presence of heterozygous pathogenic interstitial deletion of 3p12.1p11 region (arr[GRCh37] 3p12.1p11.1(86412145_87917869)x1), involving POU1F1 gene, which is associated with autosomal recessive pituitary hormone deficiency 1. Final diagnosis was made by WES and the homozygous pathogenic variant, c.657_661delACAAA in NBN gene was detected, which is a known cause of NBS. Focused clinical evaluation established the consistency of the findings with the patient’s clinical presentation.

Conclusion: Given that NBS is a rare disease with an unknown prevalence, with extremely variable clinical manifestations, the presentation of our patient may contribute to a more effective recognition of patients with NBS.

Conflict of Interest: None declared

EP13.069 A novel missense variant in the LZTR1 gene in a patient with an overlapping phenotype of autosomal dominant Noonan Syndrome and Schwannomatosis

Davide Calosci 1, Lisa Passaglia1, Annapaola Calò2, Chiara Rigon1;3, Dario Seif Alì4, Cinzia Bertolin2, Eva Trevisson1;2;3

1University of Padova, Department of Women’s and Children’s Health SDB, Padova, Italy; 2University Hospital of Padova, Clinical Genetics Unit, Padova, Italy; 3Istituto di Ricerca Pediatrica-IRP, Fondazione Città della Speranza, Padova, Italy; 4A.O.U. delle Marche, SOSD Genetica Medica e Coordinamento Malattie Rare, Ancona, Italy

Background/Objectives: in the germline setting, LZTR1 was first identified as a Schwannomatosis predisposing gene with AD inheritance. Subsequent evidence led to the classification of LZTR1 as one of the causative genes of Noonan Syndrome (NS), associated with either AD or AR inheritance. Schwannomatosis and AR-NS have traditionally been liked to LZTR1 loss-of-function variants, whereas AD-NS was hypothesized to be the result of a dominant negative mechanism. Consequently, heterozygous carriers may potentially exhibit either conditions, depending on the functional impact on the modulation of RAS/MAPK signaling. Although their co-occurrence has rarely been reported and clinical data are limited, the diagnosis of AD-NS may be missed in adulthood and Schwannomatosis displays a reduced penetrance.

Methods: we present the case of a 30-years-old woman with congenital pulmonary stenosis and facial dysmorphisms (hypertelorism, downslanting palpebral fissures) suggestive of NS. Next-Generation-Sequencing analysis of a panel of RASopathies-associated genes was perfomed.

Results: a novel heterozygous missense variant in LZTR1 affecting a highly conserved residue within the fourth Kelch domain was detected. Segregation analysis confirmed its maternal origin. Although the clinical history of the mother was unremarkable, an examination of her childhood pictures revealed facial features reminiscent of NS. These findings are consistent with an AD form of Noonan syndrome. Three years after genetic consultation, our proband underwent resection of a vertebral schwannoma.

Conclusion: the identification of a novel missense variant in LZTR1 associated with AD-NS and Schwannomatosis adds further evidence to the hypothesis of a dominant negative effect underlying both conditions.

Grants: PRINN2017.

Conflict of Interest: None declared

EP13.070 Two cases with Jansen-de Vries syndrome as a result of two PPM1D gene variants

Gokcen Karamik 1, sezin yakut uzuner2, Nuray Ozturk1, Öznur YILMAZ BAYER1, guldeniz oner1, banu nur1, ercan mihci1

1Akdeniz University, Department of Pediatrics, Division of Pediatric Genetics, Antalya, Türkyie; 2Akdeniz University, Department of Medical Biology and Genetics, Akdeniz University School of Medicine, Antalya, Türkyie

Background/Objectives: Jansen-de Vries syndrome(JDVS,OMIM#617450) is a rare, autosomal dominant neurodevelopmental disorder characterized by intellectual disability(ID), delayed psychomotor development, and behavioral abnormalities. Short stature, broad forehead, wide mouth/thin upper lip, small hands/feet, malnutrition, constipation, cyclic vomiting, and ophthalmological anomalies constitute the characteristics of the syndrome. All disease-causing variants in the PPM1D gene are truncated, and located in the last or penultimate exon of the gene(Exon 5,6). There are 65 patients reported in the literature. Here,two cases affected by JDVS are presented.

Methods:Patient 1(P1,9y,male) and Patient 2(P2,2y,male) were referred due to growth retardation and mental-motor retardation. Their prenatal history was normal. There was no consanguinity. P1 and P2 had retardation in weight (-2.51,-2.46), height (-3,-1.59), and head circumference (-2.9,-2) SDS scores. P1 had a prominent forehead, low-set ears, long philtrum, arched eyebrows, widow’s peak, hypertelorism, short fingers, and vision problems. His echocardiography and ultrasound were normal. P2 had a broad forehead, low-set ears, thick lips, a wide mouth, and small hands and feet. He had undescended testicles, hypospadias, feeding difficulties, constipation, and behavioral problems. Third and lateral ventricles were seen larger than normal on MRI.

Results:Clinical-exome sequencing analysis revealed that pathogenic de novo heterozygous mutation for P1(c.1262C>G,novel) and P2(c.1270dup) in the PPM1D gene(NM_003620.4, exon 6). The sequencing analysis results that the patient’s parents were normal.

Conclusion:JDVS is a newly identified ID syndrome. We wanted to increase the recognition of the disease with its typical findings and to draw attention to the molecular mechanism of truncating variants in the PPM1D gene.

Grants:None

References:https://doi.org/10.1002/ajmg.a.63226, https://doi.org/10.1016/j.ajhg.2017.02.005

Conflict of Interest: None declared

EP13.071 Somatic overgrowth and vascular malformations - high-depth massive parallel sequencing and ddPCR in a Swedish cohort of 100 individuals

Anna Hammarsjö 1;2, Karin Sollander1;2, Sofia Frisk1;2

1Karolinska Institutet, Molecular Medicine and Surgery, Stockholm, Sweden; 2Karolinska University Hospital, Clinical Genetics and Genomics, Stockholm, Sweden

Background/Objectives: Somatic variants are found to be responsible for overgrowth and vascular malformations with clinical manifestations from minor birthmarks to life-threatening systemic anomalies. The aim of this study is to describe our genetic approach and spectrum of findings in a Swedish cohort consisting of 100 individuals with somatic overgrowth and/or vascular malformations.

Methods: We used tissue-derived and/or blood-derived DNA for deep exome sequencing (400X) and analysis of the data from a defined gene panel consisted of 123 genes (associated with somatic overgrowth and/or vascular malformation conditions). As a 2nd approach, ddPCR was used as a confirmation of positive findings. With this setting, we were able to reach a limit-of-detection down to at least 0,1%.

Results: Disease-causing variants were identified in 44 individuals and variants of unknown significance in five (n = 84). The overall molecular diagnostic yield was 52%. The diagnostic yield increased significantly when sequencing tissue-derived DNA (n = 70; 61%), compared to blood (n = 14, 7%). The vast majority were in PIK3CA (n = 24), TEK (n = 8) and EPHB4 (n = 3), with the variant allele frequencies ranging from 0.8% to 50%.

Conclusion: Variant allele frequency varies depending on tissue type, and submission of affected tissue greatly increases the chance of a molecular diagnosis. Genetic diagnosis is important since clinical trials have shown efficacy with targeted therapy depending on the genotype.

Grants: Karolinska Institutet and Swedish Research Council

Conflict of Interest: None declared

EP13.072 Two novel variants in OCRL1 gene in two unrelated North-African patients with Lowe syndrome

Cherif Mortadha 1, Yasmina Elaribi1, Syrine Hizem1;2, Houweyda Jilani1, Sana Karoui1, yosra ben rejeb3, Sami Jebnoun4, Imen REJEB1;5, Lamia Ben Jamaa1;5

1Hôpital Mongi Slim, Department of Congenital and Hereditary Diseases, Marsa, Tunisia; 2Faculty of Medicine of Tunis, University of Tunis ElManar, Human genetics laboratory, Tunis, Tunisia; 3The Military Hospital of Tunis, pediatrics, Tunis, Tunisia; 4Tunis, pediatrics clinic, Tunis, Tunisia; 5Tunis, Research laboratory « santé mère enfant » LR22SP01, Tunis, Tunisia, Tunis

Background/Objectives: Lowe syndrome, is a rare X-linked genetic disorder characterised by the triad of congenital cataracts, intellectual disability, and proximal renal tubular dysfunction with slowly progressive renal failure. It is due to pathogenic variants throughout the OCRL1 gene. Next Generation Sequencing (NGS) techniques are currently the method of first resort for unravelling new variants in rare genetic diseases.

We report on two new variants in OCRL1 gene in two patients with Lowe syndrome.

Methods: We describe the clinical observations of two unrelated patients (P1 and P2) referred for infantile encephalopathy.

Genetic investigation was performed by NGS in both probands (clinical exome).

Results: P1 was a 6-year-old boy with the history of congenital cataract and renal rickets. On examination, he presented with a severe growth delay, microcephaly, facial dysmorphism (broad forehead, arched and rarefied eyebrows, marked philtrum, short neck), scoliosis, thoracic deformity and genu valgum. Neurological examination showed rotatory nystagmus, convergent strabismus and severe axial hypotonia. MRI scan showed leukodystrophy.

P2, was a 7-month-old boy with glaucoma and cataracts. Clinical examination revealed the same facial dysmorphism as P1, normal growth measurements, and severe hypotonia with no renal symptoms.

NGS revealed a new hemizygous variant in the ORCL gene in both cases: the frameshift c.2060_2061del variant in P1, and the splicing c.2470-2A > G IN P2. Both variants were classified as pathogenic according to the ACMG guidelines.

Conclusion: This study highlights the importance of the new sequencing technology in determining the causal genetic alteration, enabling appropriate genetic counselling and adapted management.

Conflict of Interest: None declared

EP13.073 A 46,XY patient with a heterozygous c.3418A>G (p.Met1140Val) mutation in MAP3K1 gene caused to Ambiguous Genitalia

Ekin Cemre Bayram Tokaç 1, Filiz Mine cizmecioglu Jones2, Esra Koçyiğit2, Aslı Ece Solmaz1

1Ege University Hospital, Medical Genetics, İzmir, Türkyie; 2Kocaeli University Hospital, Pediatric Endocrinology, Kocaeli, Türkyie

Background/Objectives: Disorders of Sexual Development involve variations in chromosomal, hormonal, and anatomical development. In 46,XY ambiguous genitalia, patients have male chromosomes (46,XY) but present external genitalia that may not clearly appear male or female at birth. While mutations in the MAP3K1 gene are typically associated with 46,XY sex reversal, cases involving ambiguous external genitalia are rare.

Methods: The patient has ambiguous genitalia, including a 1 cm phallus with deficient urethral meatus. Ultrasonographic images revealed palpable testes in the scrotum but absent ovaries and uterus. Genomic DNA was isolated from peripheral blood samples and analyzed using Next-Generation Sequencing (NGS) targeting 77 DSD genes. Sanger sequencing was performed for confirmation in first-degree relatives.

Results: A heterozygous missense mutation (c.3418A>G, p.Met1140Val) in the MAP3K1 gene was identified in the patient, classified as “variant of unknown significance” (VUS). Parental segregation with Sanger sequencing revealed the mother as heterozygous carrier of the variant, while the father was non-carrier. Literature review indicates silent carrier status in females with mutations in this gene. Similar findings at one patient with micropenis and penoscrotal hypospadias were reported in a study by Tsai MC et al. According to them, the variant was predicted to damage protein functions due to altered 3D structures with RMSDs. Genetic consultation was provided to the family.

Conclusion: The variant identified in this patient becomes significant as the second reported case in the literature. Although its significance is unclear, the mutation may affect protein function. The patient’s mother carrying the same mutation highlights the importance of genetic counseling.

Grants:

Conflict of Interest: None declared

EP13.074 The diagnosis of Schaaf-Yang syndrome in a child with suspected craniosynostosis

Anna Pecková 1, Jana Lastuvkova1, Vlasta Cejnova1, Anna Uhrova Meszarosova2

1Masaryk Hospital in Ústí nad Labem, o.z., Krajská zdravotní, a.s., Department od Medical Genetics, Ústí nad Labem, Czech Republic; 2Neurogenetic Laboratory, Department of Paediatric Neurology, 2nd Faculty of Medicine, Charles University and Motol Hospital, Prague, Praha, Czech Republic

Background/Objectives:Schaaf-Yang syndrome (OMIM #615547) is a very rare neurodevelopmental disorder that is inherited as an autosomal dominant, maternally imprinted manner. It shares multiple clinical features with the genetically related Prader-Willi syndrome. Patients commonly present with prenatal polyhydramnios, foetal akinesia, neonatal hypotonia, feeding difficulties, respiratory distress, joint contractures, autism spectrum disorder and developmental delay. It is difficult to diagnose because of the rarity and highly variable phenotypes involved.

Methods:Exome sequencing, Sanger sequencing

Results: We report the case of a girl from the first gravidity. The girl was born to young parents. Repeated ultrasound scans were performed during pregnancy, focusing on the nasal bones and there was a prenatal finding of renal pelvic dilatation in the foetus. Premature rupture of membranes and breech presentation resulted in termination of pregnancy by caesarean section at 38 weeks. Dysmorphic features were present after the birth – atypical head shape resembling craniosynostosis, frontal bossing, deep set eyes, low set ears, congenital contractures. The girl had neonatal hypotonia, feeding difficulties and respiratory distress. Tracheostomy was recommended because of respiratory distress and sleep apnoea. Exome sequencing revealed a de novo heterozygous pathogenic variant NM_019066.4 (MAGEL2):c.1912C>T (p.Gln638*) and Schaaf-Yang syndrome was confirmed. Diabetes insipidus complicates the condition and the prognosis is severe.

Conclusion:Genetic consultation was recommended for suspected craniosynostosis, but the girl was found to have a rare neurodevelopmental disorder: Schaaf-Yang syndrome. Consultation with the family can be very difficult in the case of rare disorders such as Schaaf-Yang syndrome.

Conflict of Interest: None declared

EP13.075 NELFA homozygous frameshift mutation in a case of adult syndromic hypertrophic myocardiopathy resembling RASopathies phenotype

Claudio Catalli 1, Isabel Llano Rivas1, Hiart Maortua Olabe1, Intza Garin Elkoro1, Blanca Gener Querol1

1Osakidetza Health System - Cruces Universitary Hospital, Genetics, Barakaldo, Bizkaia, Spain

Background/Objectives: A 53 y.o. man was evaluated for hypertrophic myocardiopathy. Clinical exploration and history was collected, showing a history of small for gestational age preterm delivery, psychomotor delay during first years of life, inmune, endocrinologic, cardiac and renal alterations and subtle dysmorphisms, prompting suspect of a syndromic condition, resembling RASopathies. Since parents are first cousin, autosomal recessive condition was suspected.

Methods: Conventional 60K array-CGH and whole exome trio were performed. Genes were analyzed according to HPO best resembling core phenotypes. Emedgene platform were uses for analysis, filtering and interpretation.

Results: No CNVs nor mutations in known OMIM genes were found. Amongst genes not associated to a OMIM phenotype, we identified a homozygous frameshift mutation in NELFA gene. NELFA encodes for a negative elongation factor mediating RNA polymerase II pausing. Although no consistent phenotype has been described associated to NELFA until now, there are reports indicationg that the Nelf-A protein play a role in structural myocardiopathies. Animal models support possible syndromic manifestations in case of loss of function / haploisufficiency mutations. No other candidate mutations were identified.

Conclusion: Disrupting biallelic mutations in NELFA could be associated to phenotypic manifestation of syndromic myocardiopathy and short stature, expecially in cases where RASopathies are suspected but no molecular cause has been identified.

Grants: No grants were used for this work.

Conflict of Interest: None declared

EP13.076 Pathogenesis of cognitive and neurofunctioning impairments in Noonan syndrome patients: the potential role of RAS/MAPK signaling pathway gene disturbances – the final results of the research.

Natalia Braun-Walicka 1, Agnieszka Pluta2;3, Tomasz Wolak2, Edyta Maj2;4, Agnieszka Maryniak3, Monika Gos1, Anna Abramowicz1, Aleksandra Landowska1, Ewa Obersztyn1

1Institute of Mother and Child, The Department of Medical Genetics, Warsaw, Poland; 2The Bioimaging Research Center, World Hearing Center at the Institute of Physiology and Pathology of Hearing, Nadarzyn, Poland; 3University of Warsaw, The Faculty of Psychology, Warsaw, Poland; 4Medical University of Warsaw, 2nd Department of Clinical Radiology, Warsaw, Poland

Background: Noonan syndrome (NS) is one of the most common genetic conditions inherited in autosomal dominant manner with vast heterogeneity in clinical and genetic features. Noonan syndrome is caused by mutations in particular genes of the RAS/MAPK signaling pathway. Patients with NS might have cognitive problems in language skills, memory, attention, executive functioning and decreased overall intelligence level. It was widely demonstrated that atypical functional connectivity may occur in many neurological diseases.

Methods: 34 patients with Noonan syndrome and 23 healthy controls were enrolled in a study involving gray and white matter volume evaluation using voxel-based morphometry (VBM), white matter connectivity measurements using diffusion tensor imaging (DTI), and resting-state functional magnetic resonance imaging (rs-fMRI). Fractional anisotropy (FA) and mean diffusivity (MD) probability distributions were calculated. Cognitive abilities were assessed using the Stanford Binet Intelligence Scales.

Results: Reductions in white matter connectivity were detected using DTI in NS patients. The rs-fMRI revealed hyper-connectivity in NS patients between the sensorimotor network and language network and between the sensorimotor network and salience network in comparison to healthy controls. NS patients exhibited decreased verbal and nonverbal IQ compared to healthy controls.

Conclusions: The assessment of the microstructural alterations of white matter as well as the resting-state functional connectivity (rsFC) analysis in patients with Noonan syndrome may shed light on the mechanisms responsible for cognitive and neurofunctional impairments.

Grants: Supported from NCN research projects no. 2013/09/B/NZ2/03164.

Conflict of Interest: None declared

EP13.078 Silver- Russel Syndrome- like phenotype caused by 7p12.1-7q11.23 maternal isodisomy detected by exome sequencing

Aleksandra Pietrzyk 1, Sabina Lichołai2, Patrycja Szymańska3, Damian Loska3, Agnieszka Sobczyńska-Tomaszewska3

1Institute of Medical Sciences, Collegium Medicum, University of Zielona Góra, Department of Clinical Genetics and Pathomorphology, Zielona Gora, Poland; 2University Clinical Hospital No.2 Pomeranian Medical University, Genetic Clinic, Szczecin, Poland; 3Medgen Medical Centre, Warsaw, Poland

Objectives: In recent years exome sequencing (ES) has become a routine first- tier test in various clinical settings. Regarded as one test for all, it enables detection of single nucleotide variants (SNV), copy number variations (CNV) and recently uniparental disomies (UPD) and repeat expansions. In this report we present a patient with Silver- Russel Syndrome like (SRS-like) phenotype caused by maternal isodisomy of 7p12.1-7q11.23 detected by ES.

Methods: A 12-year-old male was referred for consultation due to postnatal growth failure. Upon examination mild dysmorphic features, yellowish skin pigmentation and few café au lait spots were observed. Performed ES detected no pathogenic/ likely pathogenic SNV and CNV. Runs of homozygosity (ROH) analysis revealed approximately 13 Mbp region in 7p12.1-7q11.23 evaluated as isodisomic, what was confirmed by further MS-MLPA testing and proved to be maternal in origin.

Conclusion: ES can be successfully applied in testing for clinically relevant UPD. Evaluation for UPD in exome/ genome sequencing analyses should be considered as routine, especially in patients with abnormal growth. ES proves to be especially useful single test for postnatal growth failure, covering a wide spectrum of diseases caused by diverse mechanisms.

Grants: None

Conflict of Interest: None declared

EP13.079 New variant of ARFGEF1 gene causing developmental delay, impaired speech, and behavioral abnormalities, with or without seizures (DEDISB syndrome)

Laimutis Kucinskas 1, Rasa Traberg1, Marius Sukys1, Inga Nasvytienė1, Rasa Ugenskienė1

1LUHS, Genetics and Molecular Medicine Clinics

Background/objectives: DEDISB (Developmental delay, impaired speech, behavioral abormalities) syndrome is characterized by developmental delay, impaired speech, and behavioral abnormalities with or without seizures. This syndrome is inherited in autosomal dominant manner with incomplete penetrance and variable expressivity. The only known of DEDISB syndrome is ARFGEF1 gene.

Methods: We present a seven month age patient with DEDISB syndrome, which manifested by hypotonia and swallowing difficulties.

Results: Female patient was born from the the second pregnancy and the first labor. In the pregnancy period polyhydramanion was observed. From the first day the hypotonia and swallowing difficulty were noticed, for which gastrostoma was applied. Delayed psichomotor milestone was noted.The patient phenotype included microcephaly, flat face, low set ears. Because of hypotonia DMPK, SMN1 gene analysis and MLPA test for Prader Willi syndrome was performed and negative results was found. Whole exome sequencing revealed a heterozygous variant in ARFGEF1 gene: NM_006421.5 c.640-2A > C. Variant affect canonical splice site before exon 6, possible caused loss of function. This variant is not found in control databases, not described in literature.

Conclusion: We found likely pathogenic variant in ARFGEF1 gene, which might be causative for patients phenotype. Additional RNA studies for splice evaluating might help further to evaluate this variant.

Grant: No grant was obtained.

Conflict of Interest: None declared

EP13.080 A novel BRWD3 frameshift variant identified in a patient with suspected overgrowth syndrome

Karolis Baronas 1, Aušra Matulevičienė1, Beata Aleksiūnienė1, Kamilė Šiaurytė1, Algirdas Utkus1

1Institute of Biomedical Sciences, Faculty of Medicine, Department of Human and Medical Genetics, Vilnius, Lithuania

Introduction: The BRWD3 gene, located on chromosome Xq13.1, plays a critical role in chromatin remodeling and transcriptional regulation. Pathogenic variants in BRWD3 have been linked to a range of clinical manifestations, including intellectual disability, developmental delay, dysmorphic features, and overall overgrowth, and are known to cause MRX93 syndrome (OMIM# 300659).

Phenotype of a patient: The patient exhibited macrocephaly, alongside an elongated face and a prominent forehead with well-defined frontal bosses. Additional dysmorphic features included straight eyebrows, deep eye sockets, and a broad nose with a dominant area, coupled with a short philtrum and macrostomia featuring downturned corners of the mouth. The patient also presented with a prominent lower jaw and supraorbital area, complemented by large protruding ears. Gynecomastia, prominent striae in the waist area, and coarse skin with follicular prominence, notably concentrated around the knee and elbow regions was also prominent. Podiatric features included broad, large feet, with interdigital spacing anomalies and a potential short V-shaped metatarsal, alongside evident connective tissue accumulation in the heels. Brain MRI revealed internal hydrocephalus, while EEG showed no abnormalities. Ophthalmologic evaluation concluded hypermetropia in both eyes, alongside strabismus and angiopathy. Urological assessment indicated cryptorchidism.

Results: Whole Exome Sequencing identified a novel likely pathogenic hemizygous frameshift variant NM_153252.5(BRWD3):c.151_152insG, NP_694984.5:p.(Lys51ArgfsTer24).

Conclusions: Our findings contribute to expanding the phenotypic spectrum of MRX93 syndrome, with approximately 35 reported pathogenic variants in the BRWD3 gene associated with diverse clinical features.

Grant: The study is funded by the Research Council of Lithuania (No. S-LL-21-5).

Conflict of Interest: None declared

EP13.083 Clinical and Genomic Characterization of 4q Distal Deletion Associated with 4p Distal Duplication: Case Reaport and Literature Review

Anca Mitroi 1;2, Georgeta Cozaru1;2, Mariana Aschie1;2, Costel Brinzan1;2

1Emergency Clinical County Hospital of Constanta, Pathology, Constanta, Romania; 2Ovidius University, CEDMOG Center, Constanta

Background/Objectives: We report a very rare complex case of 6 years old boy who presents a distal 4q deletion associated with distal 4p duplication in order to clarify genotype-phenotype correlation for this complex case.

Methods: Due to clinical presentation array CGH was performed. Subtelomeric FISH was conducted to verify the copy number variants.

Results: The boy presents cranio-facial dysmorphism resembling CHARGE like syndrome, intellectual disability, autism spectrum disorder, irian and choroidal coloboma of right eye, partial optic atrophy both eyes and right cryptorchidy.

Array CGH revealed a 20.81Mb duplication on 4p16.3-15.31 associated with 3Mb deletion on 4q35.1-q35.2.

We discuss the correlation with other cases reported with distal 4p duplication and 4q deletion. We didn’t find any other case reported with association of distal 4p duplication and distal 4q deletion.

Conclusion: This very rare associated chromosomal abnormalities may be inherited from a parental chromosome 4 inversion.

Grants: -

Conflict of Interest: None declared

EP13.084 Familiar 16p terminal deletion: a case report

Marta Souto1, Márcia Martins 2, Andreia Dias3, Ana Matos1, Osvaldo Moutinho4, Rosário Leite1

1Unidade Local de Saúde de Trás-os-Montes e Alto Douro, Laboratório de Genética, Vila Real, Portugal; 2Unidade Local de Saúde de Trás-os-Montes e Alto Douro, Consulta de Genética, Vila Real, Portugal; 3Unidade Local de Saúde de Trás-os-Montes e Alto Douro, Serviço de Pediatria, Vila Real; 4Unidade Local de Saúde de Trás-os-Montes e Alto Douro, Departamento da Mulher e da Criança, Vila Real

Introdution: Terminal deletion in chromosome 16p13.3, also known by ATR-16 syndrome, is a rare genetic disorder, characterized by a heterogeneous clinical manifestations including intellectual disability, developmental abnormalities, microcephaly and alpha thalassemia.

The authors present familiar 16p terminal deletion.

Clinical report: A couple was referred to genetic consultation for a genetic study due to a 6-year-old son with global developmental delay, alpha thalassemia minor, hypertelorism and with a 255kb duplication in 11p15.5 region (comprising 13 genes) and a terminal deletion of 996 kb in 16p13.3, involving 48 genes (including HBA2, HBA1, and SOX8) detected by Array Comparative Genomic Hybridization.

Couple´s karyotype had a normal pattern and Fluorescence in situ Hybridization (FISH) technique with subtelomeric probe for chromosome 11p and 16p revealed a subtelomeric 16p deletion in the mother.

Discussion: The present case is a familiar deletion on 16p13.3 region, with different manifestations. The deletion was inherited from the mother, which showed normal phenotype, while the son presented phenotypic features consistent with ATR-16 syndrome, like alpha thalassemia minor and intellectual disability.

In ATR-16 syndrome, the deletion of alpha-globin genes (HBA1 and HBA2) causes alpha thalassemia, while SOX8 gene has been proposed as the responsible gene for intellectual disability.

In most cases of inherited syndrome, parent exhibits a normal or very mild phenotype in comparison to their offspring, these may due to parental imprinting, reduced penetrance, or variable expressivity.

All new cases detected should be reported in order to obtain a more precise correlation between genotype/phenotype to be used in genetic counseling.

Conflict of Interest: None declared

EP13.087 KCNK4 missense variant in an albanian patient with FHEIG syndrome

Anila Babameto-Laku 1;1, Aida Bushati2, Arlind Myftari3

1Faculty of Medicine, University Hospital Centre “Mother Teresa”, Service of Medical Genetics, Tirana, Albania; 2Faculty of Medicine, University Hospital Centre “Mother Teresa”, Service of Pediatric Neurology, Tirana, Albania; 3Faculty of Medicine, University Hospital Centre “Mother Teresa”, Service of Maxillo-Facial Surgery, Tirana, Albania

The KCNK4 gene, predominantly distributed in neurons, plays an essential role in controlling the resting membrane potential and regulating cellular excitability. De novo gain of function variants in the KCNK4 gene were reported to cause a recognizable syndrome characterized by facial dysmorphism, hypertrichosis, epilepsy, intellectual/developmental delay, and gingival overgrowth. FHEIG is extremely rare with only a few reported cases in the literature.

Material and method: We describe a case of 6 years old male Albanian patient with micrognathia, bushy eyebrows and long eyelashes, short deep philtrum, gingival overgrowth and everted upper lip, generalized hypertrichosis, epilepsy and intellectual delay. He had early onset bilateral tonic-clonic seizure that could be controlled with medication. Whole exome sequencing (WES) was performed on genomic DNA of the patient obtained from peripheral blood.

Results: WES identified a heterozygous like pathogenic variant (c.730G>C, p. Ala244Pro) in the KCNK4 gene. This variant is listed in ClinVar as a pathogenic and as likely pathogenic based on ACMG recommendations. It was described before in three patients and when comparing the phenotype with our patient, a phenotype–genotype correlation seems likely. It is speculated that this variant in the area with the function of conductive conformation led to the severe phenotype of FHEIG syndrome, while variants in other areas are possibly associated with mild phenotypes.

Conclusion: Our report provides evidence to support a specific phenotype of FHEIG syndrome associated with this KCNK4 missense variant. It also suggests that the phenotypic variations of KCNK4 are potentially associated with the molecular sub-regional effects.

Conflict of Interest: None declared

EP13.088 CHARGE syndrome due to novel mutation in the CHD7 gene

Daniela Avdjieva-Tzavella 1, Tsvetina Veleva2, Trayan Delchev1, Maria Sredkova-Ruskova1

1University Pediatric Hospital, Medical University, Department of Clinical Genetics, Sofia, Bulgaria; 2University Pediatric Hospital, Medical University, Department of Clinical Genetics, Sofia, Bulgaria

Background/Objectives: CHARGE syndrome (CS) is an autosomal dominant genetic condition caused by pathogenic variants in the CHD7 gene and clinically characterized by a wide range of anomalies with variable expression. The incidence is approximately1:15,000 newborns. CHARGE syndrome is an acronym for Coloboma, Heart defects, Atresia choanae, Retardation of growth and development, Genital abnormalities and Ear anomalies. Other criteria include distinctive facial appearance, limb abnormalities, gastrointestinal (GI) symptoms, feeding difficulties and immune deficiencies.

Methods: We present a 3-year-old girl born from a first pregnancy complicated with bleeding and gestational diabetes. A congenital heart defect - Tetralogy of Fallot was diagnosed prenatally. After birth, facial dysmorphism, sensorineural deafness, bilateral coloboma and developmental delay were identified. After obtaining normal results from chromosome analysis, MLPA and aCGH, a Whole exome sequencing was carried out.

Results: The patient carried one likely pathogenic variant - с.7763del p.(Asn2588Ilefs*4) in the CHD7 gene. The variant was confirmed by Sanger sequencing.

Conclusion: CHARGE syndrome is a heterogeneous condition with a wide variety of clinical manifestations. The genetic analysis is essential in the diagnosis of CS. Early supportive and corrective therapies, particularly in controlling infection, and improving breathing and feeding, are essential for the prognosis of patients.

Grants:

Conflict of Interest: None declared

EP13.089 MIRAGE syndrome due to a de novo SAMD9 c.2668A>G (p.Thr890Ala) variant.

Olga Ivanova 1, Polina Tsygankova1, Ekaterina Zakharova1

1Research Centre for Medical Genetics, laboratory of hereditary metabolic diseases, Moscow, Russian Federation

Background/Objectives: MIRAGE syndrome is a rare autosomal dominant multisystem genetic disorder characterized by Myelodysplasia, Infection, Growth restriction, Adrenal hypoplasia, Genital phenotypes, and Enteropathy. It has been established that children with this syndrome carry heterozygous gain-of-function variants in the SAMD9 gene. We present the case of a premature 46XY infant exhibiting severe intrauterine growth restriction, adrenal insufficiency, bronchopulmonary dysplasia, hypospadias, hyperpigmentation of the scrotum, neonatal pneumonia, thrombocytopenia, and polyuria.

Methods: DNA of blood cells of the proband and his parents was used. Whole-genome sequencing (WGS) trio analysis was conducted using the PE150 paired-end reading method on a T7 genetic analyzer from MGI. Sample preparation utilized the PCR-free method with enzymatic fragmentation (MGI).

Results: A de novo variant in the SAMD9 gene (chr7:93103430T > C GRCh38, NM_017654.4: c.2668A>G, p.Thr890Ala) in a heterozygous state was identified. This variant is not documented in gnomAD v.4.0.0, ClinVar, HGMD v2022.1, or LOVD databases. According to ACMG criteria, the variant is considered likely pathogenic (PS2, PM2).

Conclusion: While most SAMD9 variants arise de novo, patients with SAMD9 may inherit variants from asymptomatic parents due to germline or revertant mosaicism. Severe infections, anemia, and pulmonary issues are not exclusive to MIRAGE syndrome and are frequently observed in premature newborns with growth restriction. Moreover, in 20% of cases, MIRAGE syndrome does not manifest adrenal dysfunction. Consequently, SAMD9-dependent syndromes might be underdiagnosed. Increasing access to genetic sequencing is crucial for identifying children carrying pathogenic SAMD9 variants.

Grants:no

Conflict of Interest: None declared

EP13.091 Alazami syndrome: report of the first case in Bulgaria

Tsvetina Veleva 1, Daniela Avdjieva-Tzavella1, Trayan Delchev1, Maria Sredkova-Ruskova1

1University Children’s Hospital, Clinical genetics, Sofia

Background/Objectives: Alazami syndrome is an autosomal recessive neurodevelopmental disorder due to loss-of-function variants in LARP7 gene on chromosome 4q25. The incidence is less than 1:1 000 000 with most cases being found in consanguineous families from the Middle East, Asia and North Africa. The syndrome is characterized by severe growth restriction presented at birth, intellectual disability and distinctive facial features. Some patients have been reported with skeletal and behavioral features. We present 10-year-old Roma girl, born to consanguineous parents, with data for intrauterine hypotrophy, poor overall growth, short stature, small appearing head, intellectual disability, facial dysmorphism included malar hypoplasia, mandibular hypoplasia with retrognathia, low-set ears and widely spaced teeth, skeletal features (fifth digit clinodactyly, pes equinovarus), behavioral problems such as aggression and immune deficiency.

Methods: Laboratory investigation revealed a normal female karyotype, normal multiplex ligation-dependent probe amplification analysis, normal plasma aminoacids and acylcarnitines and urine organic acids, a bone age consistent with chronological age. MRI, which was performed on the patient, showed Chiari I malformation.

Results: At age of 10, clinical exome sequencing revealed homozygous pathogenic variant c.691_694del (NМ_016648.4; p.(Glu321ThrfsTer20); NP_057732.2) in LARP7 gene.

Conclusion: We report a patient with primordial dwarfism and severe developmental delay, by whom the diagnosis Alazami syndrome was detected due to whole exome sequencing. The clinic corresponded completely with genetic diagnosis.

Grants:

Conflict of Interest: None declared

EP13.092 Joubert syndrome: Molecular insights, clinical profiles, and ocular findings in a cohort of 13 patients

Salih Türk 1, Cengiz Yalçınkaya2, Buşra Kasap1, Filiz Geyik3, Dilek Uludağ Alkaya1, Nilay Güneş1, Esma Şengenç4, Sema Saltık5, Beyhan Tüysüz1

1Istanbul University - Cerrahpaşa Faculty of Medicine, Department of Pediatric Genetics, İstanbul, Türkyie; 2Istanbul University - Cerrahpaşa Faculty of Medicine, Department Of Internal Medicine, Department Of Neurology, Istanbul, Türkyie; 3Aziz Sancar Institute Of Experimental Medicine, Department Of Genetics, Istanbul, Türkyie; 4Biruni University Hospital, Department of Pediatrics, Pediatric Neurology, Istanbul, Türkyie; 5Istanbul University - Cerrahpaşa Faculty of Medicine, Department of Pediatric Neurology, İstanbul, Türkyie

Background/Objectives: Joubert Syndrome (JS) is a rare genetic disorder characterized by hypotonia, ataxia, intellectual disabilities, and cerebellar vermis hypoplasia. This study aimed to examine the molecular and clinical features in 13 affected individuals diagnosed with JS.

Methods: Whole-exome sequencing was carried out in 13 affected individuals from 12 families, and Sanger sequencing analysis was used for confirmation.

Results: Thirteen participants (5 males, 8 females) with an average age of 5.45(0.19-14.78) years were followed up during the mean time of 7.02(0.23-14.01) years. All 13 individuals showed cerebellar vermis hypoplasia on MRI, and the molar tooth appearance was present in 10 of them. Other neurological findings included developmental delay (11/13), intellectual disability (10/13), hypotonia (10/13), and ataxia (5/13). All patients had ocular findings: strabismus (7/13), nystagmus (5/13), oculomotor apraxia (5/13), retinal disease (4/13), severe hypermetropia (2/13), vision loss (1/13), and band keratopathy (1/13). No renal or hepatic disease was present in 12 patients on ultrasound examinations. Polydactyly (3/13) and syndactyly (2/13) were observed. Genetic analysis revealed mutations: AHI1 (4 individuals), CEP290 (4 individuals from 3 families), and MKS1, RPGRIP1L, TCTN2, TCTN3, TMEM107 (one individual, each). Four novel variants were identified. Among AHI1 patients, 3 of 4 had retinal disease, and one had vision loss. Patients with AHI1 (2/4) and CEP290 (1/4) mutations had normal intelligence, whereas intellectual disability accompanied all other mutations.

Conclusion: Severe ocular involvement in AHI1 patients indicates the need for careful follow-up. This underscores the importance of comprehensive evaluations in JS patients for accurate diagnosis and personalized treatment.

Conflict of Interest: None declared

EP13.093 Aarskog syndrome: Expanding the phenotypic and molecular spectrum through a new case series including two adult patients

Gözde Tutku Turgut 1, Umut Altunoglu1;2, Tuğba Kalaycı1, Sahin Avci1;2, Ayça Dilruba Aslanger1, Volkan Karaman1, Zehra Oya Uyguner1, Hülya Kayserili1;2

1Istanbul Faculty of Medicine, Istanbul University, Medical Genetics, Türkyie; 2Koç University School of Medicine (KUSoM), Medical Genetics, Türkyie

Background/Objectives: Aarskog-Scott syndrome (AAS, MIM#305400) is an X-linked disorder characterized by recognizable facial features, short stature, and genitourinary and skeletal malformations. AAS is attributed to pathogenic variants in the FGD1 gene, and to date, less than 60 patients with a genetic diagnosis have been reported. We hereby present a molecularly confirmed cohort of 14 male AAS patients from 13 families to further expand the clinical and molecular spectrum of AAS.

Methods: In all patients, a detailed clinical dysmorphological evaluation prompted molecular testing for the FGD1 gene through Sanger sequencing. Patients’ molecular and clinical features were compared with the literature data.

Results: Among 14 patients, 12 were referred during childhood, while two at adulthood due to infertility. Fifty-five percent (n = 6/11) of the patients with available records had antenatal manifestations. In addition to well-described AAS findings, distinctive features observed in multiple patients included variable skin findings (n = 6), renal malformations (n = 2), muscular build (n = 2), and azoo-/oligozoospermia (n = 2). Cardiac (n = 4, 29%) and ocular manifestations (n = 6, 43%) were identified at significantly higher rates than previously reported. We identified 11 different FGD1 variants, comprising five missense, five frameshift/nonsense, and one splice-site variant. Seven out of 11 variants were novel. Two variants were found to be recurrent, detected in two independent families. Genotype-phenotype correlation could not be established.

Conclusion: Our study provides additional insights into the clinical phenotype of AAS through the largest molecularly confirmed cohort including two adult patients. Seven novel FGD1 variants identified in the cohort further broaden the molecular spectrum of AAS.

Grants: None.

Conflict of Interest: None declared

EP13.094 Summary of a case: patient with de novo 36,7 mb 2p duplication syndrome

Filiz Ozen 1, Arzu Guliyeva1;2

1Goztepe Prof.Dr. Suleyman Yalchin City Hospital, Medical Genetics, Istanbul, Türkyie; 2Istanbul Medeniyet University, Faculty of Medicine, Medical Genetics Department, Istanbul, Türkyie

Background/Objectives: A rare chromosomal anomaly syndrome resulting from the partial duplication of the short arm of chromosome 2 with a highly variable phenotype. Principle characteristics are pre and post-natal growth failure, global developmental delay, facial dysmorphism and ocular anomalies. Other reported anomalies include hypotonia, pectus excavatum, long fingers and toes, syndactyly, congenital heart defects (ventricular, atrial septal) and neural tube defects, seizures, pulmonary hypoplasia, diaphragmatic hernia and urogenital anomalies.

Methods: A 4-month-old girl, born to distant relative parents, was referred to the genetic clinic with inability to hold head up and characteristic facial features. On physical examination, she had the developmental delay and frontal bossing, hemangioma on the forehead, sparse eyebrows, hypertelorism, epicanthus, long and triangular face shape, low-set ears, thin lips, faint philtrum, retrognathia. Her brain MRI results revealed thinning corpus callosum and expansion of the lateral ventricules. In her 3rd month, she had seizures. As a result of EEG, neuronal hyperexcitability and sharp slow wave activity were observed in bilateral temporoparietal regions in the right hemisphere during sleep.

Results: Primarily, karyotype and array CGH analysis planned by us. 46,XX,dup(2)(p22.2 p25.1) was found out in karyotype analysis. Microarray analysis revealed arr[GRCh37] 2p25.3 p22.2 (197427_36863392)x3, 36,7Mb heterozygous, likely pathogenic duplication. Tests done to parents found to be normal.

Conclusion: We reported a de novo heterozygous likely pathogenic duplication arr[GRCh37] 2p25.3 p22.2 (197427_36863392)x3, 36,7Mb 2p duplication syndrome. Chromosomal abnormalities should be considered in differential diagnosis of any child with developmental delay and dysmorphic facial features.

Grants: None

Conflict of Interest: None declared

EP13.095 Biallelic variants in CENPT as another etiology for Stromme syndrome

YoungBae Sohn 1, Yoong-a Suh2, Chang Ahn Seol3, Do-Wan Kim4

1Ajou University Hospital, Medical Genetics, Suwon, Korea, Rep. of South; 2Ajou University Hospital, Pediatrics, Korea, Rep. of South; 3GC Genome, Youngin, Korea, Rep. of South; 4Ajou University Graduate School of Medicine, Suwon, Korea, Rep. of South

Background: Stromme syndrome, first described in 1993, is a rare autosomal recessive genetic ciliopathy involving multiple abnormalities. The clinical triad includes apple peel intestinal atresia, anterior ocular anomalies, and microcephaly. CENPF is the only known causative gene of Stromme syndrome to date. The gene encodes the protein associated with the kinetochore and the mitotic spindle throughout the cell cycle and plays a role in ciliogenesis.

Methods: Herein, we present the case of a female neonate born at 34 weeks of gestation and weighing 2440 g at birth with multiple congenital anomalies including microcephaly, bilateral microphthalmia and jejunal atresia. The genetic diagnosis was performed by whole exome sequencing.

Results: We identified compound heterozygous variants in CENPT (c.[862 + 4A > C]; [1562G > A]), whereas no variant was identified in CENPF. The results were validated through Sanger sequencing and consequent in silico analysis using Ingenuity Pathway Analysis (IPA).

Conclusion: We described the first Korean newborn with typical clinical manifestations of Stromme syndrome carrying compound heterozygous pathogenic variants in CENPT. We report, for the first time, CENPT as the second causative gene of Stromme syndrome.

Conflict of Interest: None declared

EP13.096 Case report of rare Ichthyosis follicularis, alopecia, and photophobia (IFAP) syndrome with novel MBTPS2 gene variant

Rasa Traberg 1, Rimvydas Jonikas1, Jūratė Laurynaitienė2, Rasa Ugenskiene1

1Lithuanian University of Health Sciences, Department of Genetics and Molecular medicine, Kaunas, Lithuania; 2Lithuanian University of Health Sciences, Department of Neurology, Kaunas, Lithuania

Background/Objectives: Ichthyosis follicularis, alopecia, and photophobia (IFAP) syndrome is a rare X-linked genetic disorder characterized by ichthyosis and alopecia from birth and sometimes accompanied by short stature, intellectual disability, and seizures that develop in the first few years of life. The disorder is caused by MBTPS2 gene variant that impairs cholesterol homeostasis and the ability of cells to cope with endoplasmic reticulum stress.

Methods: We report 4 years old male patient who had dry skin, alopecia, developmental delay and epilepsy. His maternal uncle also had alopecia, photophobia, corneal damage, intellectual disability and epilepsy since infancy. Whole exome sequencing (WES) revealed NM_015884.4(MBTPS2):c.970+5G > A hemizygous variant that possibly affects splicing. The variant segregation analysis confirmed the same variant in the uncle and that both mother and grandmother were the carriers. Furthermore, we performed RNR analysis to determine the effect of c.970+5G > A variant for affected person and for carrier.

Results: For hemizygous male (compared to controls without this variant) agarose gel electrophoresis showed no normal transcript and one additional fragment with deletion. The most abundant fragment for patient has an approximate 300 base pair (bp) deletion, which is also seen in controls, and faintly seen fragment has abound 400 bp deletion. Sanger sequencing of hemizygous patient showed that 300 bp fragment seen on gel electrophoresis had 6-7th exon deletion, which do not create frameshift, and 400 bp fragment had 6-8th exon deletion that causes frameshift.

Conclusion: According to clinical and molecular data we confirm that MBTPS2 c.970+5G > A affects splicing and causes IFAP syndrome.

Grants: no

Conflict of Interest: None declared

EP13.097 Clinical and genetic spectrum of 14 Tunisian patient with Treacher-Collins syndrome.

Wiem Essalah 1, Ahlem Achour1;2, Asma Tajouri2, Madiha Trabelsi1;2, Faouzi Maazoul1, lilia kraoua1;2, Imen Chelly2;3, Rym Meddeb1;2, Neila Belghith1, Sana Gabteni1;2, ridha m’rad1;2, Ines Ouertani1;2

1Charles Nicolle Hospital, Department of Congenital and Hereditary Diseases, Tunis, Tunisia; 2Faculty of Medicine of Tunis, University of Tunis El Manar, Laboratory of Human Genetics (LR99ES10), Tunis, Tunisia; 3, Habib Bougatfa Hospital, Department of Pediatrics, Bizerte, Tunisia

Background/Objectives:Treacher Collins syndrome (TCS, OMIM #154500) arises from aberrant pharyngeal arch differentiation during fetal development, impacting 1 in 50,000 live births. It can be caused by pathogenic variants in several genes, but TCOF1(NM_001371623.1) remains the most commonly described in literature.

Methods:We carried out a retrospective descriptive study on a cohort of Tunisian patients with TCS followed up at the Department of Congenital and Hereditary Diseases of Charles Nicolle Hospital over a period of twelve years from June 2012 to January 2024. Sanger sequencing of 11/24 exons of the TCOF1 gene was performed on seven patients.

Results:In total 14 patients were included: 8 patients from 4 different families and 6 sporadic cases. The sex ratio was 1:1. All patients displayed distinctive dysmorphic features typical of TCS: The hypoplasia of the zygomatic arch, a prominent forehead, downslanting palpebral fissures, and retrognathia. Microcephaly was noted in 4/14 patients and one patient exhibited a cleft palate.The outer ear abnormalities were variable associated with hearing loss ranging from mild hypoacusis to profound deafness, related to malformation of the ear ossicles. Molecular analysis revealed two pathogenic frameshift variants in 3/7 tested patients : a novel variant the c.1926delG identified in two familial cases and c.1406-1409delACAG in one sporadic case.

Conclusion:Our study highlights the clinical and genetic heterogeneity of TCS. Gene panel analysis is still necessary to establish molecular diagnosis,define genetic spectrum and provide adequate genetic counseling.

Grants:(1)Bożena and al,2021 Sep.

Conflict of Interest: None declared

EP13.098 Upper limbs reduction defects caused by novel in-frame insertion in SALL4 gene

Adriana Bobinec 1;2;3, Ivona Sansović1;2;3, Ana-Maria Meašić1;2;3, Katarina Vulin1;2;3, Leona Morožin Pohovski1;2;3, Ljubica Odak1;2;3

1Children’s Hospital Zagreb, Department of Medical and Laboratory Genetics, Endocrinology and Diabetology with daily care unit, Zagreb, Croatia; 2, Scientific Centre of Excellence for Reproductive and Regenerative Medicine (CERRM), University of Zagreb School of Medicine, Zagreb, Croatia; 3, European Reference Network on congenital malformations and rare intellectual disability ERN-ITHACA, Zagreb, Croatia

Background/Objectives: SALL4 gene encodes a transcriptional factor known to play important role during embryogenesis, particularly in developing limbs. SALL4-related disorders are inherited in an autosomal dominant manner and include Duane-radial ray, acro-renal-ocular and SALL4-related Holt-Oram syndromes. Here we describe a 3-month-old proband with severe upper limbs reduction defects, craniofacial dysmorphia, facial asymmetry and ear and renal anomalies.

Methods: Clinical exome sequencing (CES) was performed in proband with clinical presentation of upper limbs reduction defects using Illumina’s TruSight One Sequencing kit. Sanger sequencing was performed in proband’s parents to determine variant origin.

Results: CES analysis revealed a heterozygous variant in SALL4 gene, c.689_706dup, p. (Gln230_Ile235dup). Detected in-frame insertion was 18 pb long and it affected protein’s glutamine-rich sequence that is considered important for establishing protein interactions. Sanger sequencing in proband’s parents confirmed de novo occurrence of the variant. Detected variant was not previously described in curated databases or literature and it is classified as likely pathogenic.

Conclusion: Small insertions/deletions are frequently found as form of genetic variants in humans and in-frame insertions are usually considered non-pathogenic. In-frame duplications of this size have not been previously described in SALL4-related conditions. We consider detected insertion at position c.689_706dup in SALL4 gene likely responsible for upper limbs reduction defects present in our patient.

Grants: This work was supported by Scientific Center of Excellence for Reproductive and Regenerative Medicine and by the European Union through the European Regional Development Fund, under grant agreement No. KK.01.1.1.01.0008, project „Reproductive and Regenerative Medicine - Exploring New Platforms and Potentials.

Conflict of Interest: None declared

EP13.100 Seventh patient with the Alazami-Yuan syndrome due to novel TAF6 variants: new phenotypic associations

Beyza Yavuzcan1, Manar Kaptan1, Mert Kaya 1, Cigdem Arikan2, Umut Altunoglu3

1Koc University, Graduate School of Health Sciences, Istanbul, Türkyie; 2Koc University, School of Medicine Department of Pediatric Gastroenterology, Istanbul, Türkyie; 3Koc University, School of Medicine Department of Medical Genetics, Istanbul, Türkyie

Background/Objectives: The TATA-binding protein-associated factor-6 (TAF6) homodimer stabilizes the Transcription factor IID (TFIID) complex, which is involved in transcription initiation. Loss-of-function mutations in TAF6 lead to the instability of the TFIID complex in Drosophila cells. Biallelic TAF6 variants are associated with the Alazami-Yuan syndrome (ALYUS), an ultra-rare autosomal recessive disorder characterized by global developmental delay and/or intellectual disability, dysmorphic facial features overlapping with Cornelia de Lange syndrome, hypotonia, microcephaly, and short stature.

Methods: We performed exome sequencing in a proband born to unrelated parents. Segregation analyses were performed in three unaffected family members by Sanger sequencing.

Results: The patient was a 12-year-old girl with history of growth retardation of prenatal onset, chronically elevated liver enzymes, spina bifida occulta, global developmental delay, moderate intellectual disability, bilateral sensorineural deafness, microcephaly and minor limb anomalies including clinodactyly of the fifth finger. Dysmorphic features included thick and broad eyebrows, synophrys, hyperconvex nasal ridge, low hanging columella, short philtrum, wide mouth with thick lip vermilions and a grimacing smile. She was found to have heterozygous c.2003del p.(Pro668Argfs*74) and heterozygous c.331G>C p.(Ala111Pro) in TAF6 (NM_001190415.2), shown to be in trans by parental analyses.

Conclusion: We present clinical information on the seventh ALYUS patient with a novel compound heterozygous TAF6 variant and review the literature for specific human phenotypes associated with TAF6. Chronically elevated liver enzymes in our patient may be indicative of liver disease as part of the TAF6 clinical spectrum.

Conflict of Interest: None declared

EP14 Cytogenetics, Genome Variation and Architecture

EP14.001 Comprehensive molecular cytogenetic analysis of small supernumerary marker chromosomes: insights into genomic topology and phenotypic implications.

Darya Yurchenko 1, Zhanna Markova1, Marina Minzhenkova1, Nadezda Shilova1

1Research Centre for Medical Genetics, Laboratory of Cytogenetics, Moscow, Russian Federation

Background/Objectives: Small supernumerary marker chromosomes (sSMCs) are additional chromosomes that cannot be unambiguously identified by standard cytogenetic methods alone. A cytogenomic approach involving molecular cytogenetic techniques is essential for a detailed morphological assessment of these chromosomes and for devising genetic counseling strategies for individuals carrying sSMCs. Most carriers of sSMCs (70%) have no clinical manifestations since marker chromosome contain only heterochromatin. However, there are reports of the presence of euchromatin regions on sSMCs in such carriers.

Methods: FISH with the commercial DNA probes for the centromeric/pericentromeric regions of chromosomes 15 and 22. To study the involvement of proximal euchromatin region (PE) in sSMCs, FISH with homemade DNA probes were developed.

Results: A cohort of patients without phenotypic features and with sSMC(15) (n = 9) and sSMC(22) (n = 9) in a karyotype, mainly in a mosaic state, was assembled. Four of the sSMC(15) carriers and eight of the sSMC(22) carriers contained only pericentromeric heterochromatin. In the remaining cases, PE with a size of 1.2 Mb was present in the sSMC(15) (4/18), and in one case of sSMC(22), PE with a size of 714 kb was present.

Conclusion: We hypothesize that the presence of a 1.2 Mb triplication, localized in the pericentric region of 15q (chr15(GRCh37/hg19):20,700,000–21,910,881), and a 714 kb triplication localized in the pericentric region of 22q (chr22(GRCh37/hg19):17,900,000–18,614,436), is not associated with an abnormal phenotype. These regions may not contain potentially dose-sensitive genes, or there may be a positional effect. sSMC carriers can be a model for characterizing the length of the non-dose-sensitive PE of the human genome.

Conflict of Interest: None declared

EP14.002 Unraveling Chromothripsis by HiFi long-read sequencing of a complex chromosomal rearrangement

Usha Dutta 1, Divya Bhanu1, Ashwin Dalal1

1Centre for DNA Fingerprinting and Diagnostics, Diagnostics Division, Hyderabad, India

Background/Objectives: Structural Variants (SVs) are the genomic rearrangements affecting more than 50 base pairs. These account for a significant complexity and over 50% cannot be detected due to limited methods. The advent of sequencing technologies helped to identify the disease-causing variants by short-read sequencing. However, the detection of SVs remains challenging due to the limited read length and repetitive nature but long-read sequencing is a better choice. Accurate detection of breakpoints helps understand the causative genes and molecular mechanisms. This study aims to sequence a complex rearrangement by PacBio HiFi long-read sequencing technique and to establish a quick pipeline.

Methods: Chromosomal analyses, Microarray, Genome sequencing, and Sanger validation were performed.

Results: An eight-year-old male child with Congenital Heart Disease and Gynecomastia revealed a complex translocation of 46,XY,t(4;5;16)(q33;q31.1;q22)der(5)del(5)(p15.2). Array revealed 4.1 Mb and 1.2 Mb deletions on 5q23.1 and 5q23.3-q31.1 regions respectively. Genome sequencing was performed on Sequel IIe and pbsv tool was used to detect the SVs. Filtering was done based on the chromosomal breakpoint regions; surprisingly, 75 breakpoints (11 deletions, 46 translocations, 26 intrachromosomal translocations, 1 duplication, 2 insertions, and a repeat region) were identified on chromosomes 4,5 and 16. These variants were analyzed in the IGV browser and Sanger validation was performed for one deletion and translocation.

Discussion: This is the first study from India to demonstrate and establish the HiFi long-read sequencing. All types of SVs including repeat expansion were detected. Thus, delineating the complexity of clustered breakpoints in depth and revealing the explosive mechanism of Chromothripsis.

Conflict of Interest: None declared

EP14.003 One Laboratory Perspective: Chromosomal Microarray Analysis (CMA) for Prenatal Diagnosis

Clairo Lin Hui Ng 1, Mui Li Tan1, Lee Tian Toh1, Hui Yi Yon1, Min Hwee YONG1, Angeline Hwei Meeng Lai2

1KK Women’s and Children’s Hospital, Cytogenetics, Singapore, Singapore; 2KK Women’s and Children’s Hospital, Genetics Service, Singapore

Background/Objectives: Chromosomal microarray analysis (CMA) is the first-tier recommendation for a prenatal evaluation of fetuses with structural anomalies (American College of Obstetricians and Gynaecologists Committee on Genetics, 2013; Dugoff et al., 2016). Since the launch of Prenatal CMA test in our lab back in 2018, it is progressively taking over conventional karyotyping due to increased yield and a resolution higher than conventional cytogenetics.

Methods and Results: Of 1117 samples sent for prenatal CMA test from October 2018 to December 2023, 51.4% were sent for ascertainment of abnormal ultrasound findings, 40.6% were sent due to high risk for prenatal screening (FTS, NIPT), and 8% were referred for other reasons such as family history or anxiety.

88.7% of these samples yielded normal results, 5.7% detected pathogenic or likely pathogenic deletions or duplications, and 5.6% resulted in variants of unknown significance (VOUS).

Among pathogenic findings, we found almost twice as many pathogenic copy number variants (CNVs) in fetuses sent due to abnormal ultrasound findings (7.2%) compared to prenatal screening (3.75%). This shows that prenatal CMA is more effective in ascertainment of abnormal ultrasound findings, especially cardiac abnormalities.

Our data also demonstrated that prenatal CMA has resulted in a 26.8% gain in detection rate in our lab as 1.2% of the pathogenic findings such as microdeletions and microduplications were undetectable by karyotype alone.

Conclusion: Despite its potentials, prenatal CMA has limitations of detecting mosaic SCA as well as balanced translocation. We also present how karyotyping and fluorescence-in-situ-hybridisation (FISH) still work together with CMA, for a holistic approach in prenatal diagnosis.

Conflict of Interest: None declared

EP14.004 Molecular karyotyping, MLPA and FISH of microdeletions and microduplications in the proximal and distal regions of chromosome 22q11.2

Ivana Tonkovic Djurisevic 1, Sanda Huljev Frkovic2, Morana Miklos1, Anita Pokupec Bilic1

1University Hospital Centre Zagreb, Department of Laboratory Diagnostics, Division of Cytogenetics, Zagreb, Croatia; 2University Hospital Centre Zagreb, Department of Pediatrics, Division of of Human Genetics and Metabolic Diseases, Zagreb, Croatia

Chromosome 22 contains eight low-copy repeats (LCR22A-LCR22H), also referred to as segmental duplications, that mediate meiotic non-allelic homologous recombination (NAHR) in region q11.2. These highly homologous sequences can lead to recurrent copy number variations (CNVs), either microdeletion or microduplication.

We described seven cases with different breakpoints of microdeletion / microduplication in 22q11.2 detected by molecular karyotyping, MLPA (Multiplex ligation-dependent probe amplification) or FISH (Fluorescence in situ hybridization). Array-based comparative genomic hybridization (Array-CGH) was performed on DNA samples using SurePrint G3 Human CGH Microarray 8 × 60 K chips (Agilent Technologies). Array-CGH data were confirmed with the SALSA MLPA kit P250 (MRC Holland).

The proximal region of chromosome 22q11.2 includes the most common ~ 3 Mb LCR22A-LCR22D deletion or duplication, rarely a smaller 1,5 Mb deletion or duplication involving LCR22A to LCR22B breakpoints. Phenotypes of patients with typically 22q11.2 microdeletion syndrome (A-D or A-B deletion) are similar, indicating that the penetrance is nearly complete, whereas microduplication of the same region results in variable phenotypes.

The recurrent distal 22q11.2 microdeletion and microduplication are variable in size, based on mediating LCRs. Three cases of the distal 22q11.2 region involved LCR22D-LCR22G, LCR22E-LCR22F deletion, and LCR22E-LCR22H duplication. These recurrent CNVs can occur de novo or be inherited from a parent, with incomplete penetrance and variable expressivity.

The inheritance, main candidate genes, incomplete penetrance, and variable phenotype expressivity in carriers of the recurrent CNVs have been described for better understanding of a genotype–phenotype correlation, which is essential in genetic counselling.

Conflict of Interest: None declared

EP14.005 Uncommon mosaicism within XYY syndrome manifested as a short stature

Anita Pokupec Bilic 1, Morana Miklos1, Ivana Tonkovic Djurisevic1, Ivan Bilic2, Katja Dumic Kubat3

1University Hospital Centre Zagreb, Department of Laboratory Diagnostics, Division of Cytogenetics, Zagreb, Croatia; 2School of Medicine, University of Zagreb, Department of Pathophysiology, Zagreb, Croatia; 3University Hospital Centre Zagreb, Department of Pediatrics, Division for Pediatric Endocrinology and Diabetes, Zagreb, Croatia

The XYY syndrome is characterized by an extra copy of an Y chromosome in a cell. The XYY phenotype commonly includes tall stature with possible mild delays in motor and language development. Sex chromosome mosaicism involving three different cell lines is relatively uncommon, with very variable clinical presentation, depending on a percentage of abnormal cells in a tissue

A 14-year-old boy (weight 48.9 kg (33.c.,SD -0,43), height 152.7cm (5.c., SD -1,64), BMI: 21,0 (71.c., SD 0,55)), was referred to our department, after endocrinological evaluation, due to a short stature.

The cytogenetic analysis was performed on peripheral blood. The percentage of cell lines as well as Y chromosome structure were determined by FISH method

The cytogenetic analysis showed the presence of three different cell lines, two of them containing sex chromosome aneuploidies (trisomy XYY, monosomy X) and a cell lineage of a normal male karyotype. The percentage of cell lines were 93%, 2% and 5%, respectively. FISH probes confirmed SRY and Yqh region on a chromosome Y, whereas the SHOX gene was present in all gonosomes. The pericentric inversion of chromosome 9, most common inversion of a chromosome in population, was present in all cells. Therefore, the karyotype was: mos 47,XYY,inv(9)[25]/45,X,inv(9)[1]/46,XY,inv(9)[1].

This case demonstrates an unusual phenotype with a short stature associated with a mosaic karyotype with high percentage of XYY cells found in peripheral blood. This report highlights the importance of cytogenetic workup as a part of endocrinological assessment of growth disorders.

Conflict of Interest: None declared

EP14.006 Genetic variants in exon 1 of ATXN2 gene are associated with neurodegenerative diseases.

Diksha Jethnani1, Joanna Lubieniecka 1;2, Krzysztof P Lubieniecki2, Kilian Audoire2, Maria Yusenko2, Charlotte Hippert2, Kristina Döring2, Nikolina Jovancevic2, Sabine Hoffjan2, Patrick Weydt3, Jinko Graham1, Huu Phuc Nguyen2

1Simon Fraser University, Statistics and Actuarial Science, Burnaby, Canada; 2Ruhr University Bochum, Human Genetics, Bochum, Germany; 3University of Bonn, Department of Neurodegenerative Diseases and Gerontopsychiatry, Bonn, Germany

Background/Objectives: ATXN2 encodes an RNA binding protein involved in mRNA translation and regulation. It has also been shown to be a modifier of TDP43 toxicity in yeast, flies, and mouse models. Large polyglutamine (CAG) expansions are causative of spinocerebellar ataxia type 2 (SCA2) and intermediate expansions are considered a risk factor for amyotrophic lateral sclerosis (ALS). However, most variants within this region detected by short read NGS analysis remain unreported due to difficulty with variant calling in repeat regions.

Methods: We tested genetic variants, detected in exon 1 (polyglutamine repeat region) of the ATXN2 gene during diagnostic exome sequencing for association with neurodegenerative disease (ND). In total 134 exomes from patients with various ND diagnosis, 165 patients with non-ND diseases, and 59 cases with unclear classification were included in the statistical analysis. In this sample set, 17 variants were selected based on predicted impact, clinVar classification, and frequency in control populations. The analysis was done using R programming utilizing the SKAT package for statistical testing of rare variants.

Results: Statistical analysis showed that the enrichment of ATXN2 exon 1 variants in patients with ND disease is unlikely to be random, suggesting potential importance in the pathology of neurodegenerative disease.

Conclusion: With the emergence of technologies that allow a better resolution of repeat regions (such as long read sequencing) re-investigation of ATXN2 gene may provide new insights into the gene’s role in neurodegeneration.

Conflict of Interest: None declared

EP14.007 Phenotype associated with haploinsufficiency of ACTB gene : a study of 19 new cases

marion lesieur-sebellin 1;2, Kristen Wigby3;4, Elise Schaefer5, Aurélie Gouronc6, Nicolas Chatron7;8, Anne-Lise Poulat9, Audrey Putoux7;10, Alice Goldenberg11;12, Mathilde Quibeuf11;13, Pascal Chambon11;12, Sophie Rondeau1, Giulia Barcia1;2, Jonathan Levy14, Juliette Piard15;16, Paul Kuentz17;18, Martine Doco19;20, Nathalie Bednarek19, Roseline Caumes21, Sonia Bouquillon22, Cedric Le Caignec23;24, Olivier Patat25, Philippe Khau Van Kien26, Jean Chiesa27, Geoffroy Delplancq28, Séverine Bacrot28, Sophie Brisset28, Vincent Cantagrel2, Jerica Lenberg4, Jennifer Friedman4;29, Marlene Rio1;2, sophie scheidecker6, VALÉRIE MALAN1;2

1Fédération de Génétique et Médecine Génomique, Service de Médecine Génomique des Maladies Rares, APHP.Centre, Hôpital Necker-Enfants Malades, Paris, France; 2Université Paris Cité, Génétique des Troubles du Neurodéveloppement, Institut Imagine, INSERM UMR_1163, Paris, France; 3Department of Genetics, Division of Genomic Medicine, UC Davis MIND Institute, Sacramento California, United States; 4Rady Children’s Institute for Genomic Medicine, San Diego California, United States; 5Service de Génétique Médicale, Institut de Génétique Médicale d’Alsace, Strasbourg, France; 6Laboratoire de diagnostic génétique, Hôpitaux Universitaires de Strasbourg, Strasbourg, France; 7Service de Génétique, Hospices Civils de Lyon, Lyon, France; 8Laboratoire Physiopathologie et Génétique du Neurone et du Muscle, CNRS UMR 5261, Institut NeuroMyoGène, Lyon, France; 9Service de Neuropédiatrie, Hospices Civils de Lyon, Lyon, France; 10Centre de Recherche en Neurosciences de Lyon, équipe GENDEV, INSERM U1028 CNRS UMR5292 UCBL1, Lyon, France; 11Univ Rouen Normandie, Normandie Univ, CHU de Rouen, Rouen, France; 12Inserm U1245, Département de Génétique et Centre de Référence Anomalies du Développement et syndromes Malformatifs, F 76000, Rouen, France; 1312 Inserm U1245, Département de Génétique et Centre de Référence Anomalies du Développement et syndromes Malformatifs, F 76000, Rouen, France; 14Département de Génétique, Hôpital Robert Debré, Paris, France; 15Centre de Génétique Humaine, Centre Hospitalier Universitaire, Université de Franche-Comté, Besançon, France; 16UMR 1231 GAD, Inserm, Université de Bourgogne, Dijon, France; 17Oncobiologie Génétique Bioinformatique, FHU-TRANSLAD, Besançon, France; 18Institut GIMI, CHU Besançon, Besançon, France; 19Service de génétique, CHU de Reims, CRMR anddi-rares, Reims, France; 20Service de génétique, CHU de Nantes, Nantes, France; 21Service de Génétique Clinique, Hôpital Jeanne de Flandre, Lille, France; 22Institut de Génétique Médicale, Hôpital Jeanne de Flandre, CHU Lille, Lille, France; 23Service de Génétique Médicale, CHU Toulouse Purpan, Toulouse, France; 24ToNIC, INSERM UMR1214, Toulouse, France; 25Service de Génétique Médicale, CHU Toulouse Purpan, Toulouse, France; 26Génétique Médicale, Centre de Compétences Anomalies du Développement et Syndromes Malformatifs, Hôpital Carémeau - CHU de Nîmes, Nîmes, France; 27UF de Cytogénétique et Génétique Médicale, Hôpital Carémeau - CHU de Nîmes, Nîmes, France; 28Unité de Génétique Constitutionnelle, Service de Biologie, Centre Hospitalier de Versailles, Le Chesnay, France; 29Department of Neurosciences, University of California San Diego and Rady Children’s Hospital, San Diego California, United States

Background/Objectives: Pathogenic gain of function or dominant negative effect missense variations in ACTB are associated with a neurodevelopmental disorder characterized by intellectual disability (ID), seizures, cerebral, renal and ocular abnormalities, and dysmorphic features (DF) (Baraitser-Winter syndrome, OMIM#243310). ACTB encodes beta-actin, a highly conserved protein implicated in cell motility, structure and integrity. Deletions including ACTB and loss-of-function single nucleotide variants in ACTB have recently been implicated with a distinct phenotype including developmental delay, ID, growth delay (GD), cardiac and renal abnormalities, and DF. Few patients have been reported in the literature.

Methods: Thanks to national networks (AchroPuce and AnDDI-Rares) and GeneMatcher, we gathered 15 individuals and one fetus carrying a heterozygous deletion including ACTB, and four patients with a heterozygous truncating variant. We analyzed genotypic and phenotypic data.

Results: Thirteen out of 19 individuals presented with ID, and 2/19 with learning difficulties. Language delay and behavior abnormalities were observed in 14/19 and 11/19 individuals respectively, and GD in 7/19. Most of the individuals (13/19) displayed a recognizable DF. Cardiac and renal abnormalities were occasionally observed. Eleven anomalies were de novo, while one deletion was inherited from the mother (unknown origin in 7 cases). Deletion size varied from 125 kb to 1.6 Mb (4.3 Mb in the fetus) and could arise from a FoSTeS/MMBIR mechanism.

Conclusion: This study allowed us to better characterize the phenotype associated with the haploinsufficiency of ACTB underlying the high prevalence of neurodevelopmental disorders (ID, language delay, behavior abnormalities), microcephaly and GD in this recognizable syndrome.

Conflict of Interest: None declared

EP14.008 Chromosomics Databases

Thomas Liehr 1

1Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, Jena, Germany

Background/Objectives: The term "chromosomics" was introduced to draw attention to the three-dimensional morphological changes in chromosomes that are essential elements in gene regulation. Chromosomics deals with the plasticity of chromosomes in relation to the three-dimensional positions of genes, which affect cell function in a developmental and tissue-specific manner during the cell cycle. It also deals with species-specific differences in the architecture of chromosomes, which has been overlooked in the past. Chromosomics includes research into chromatin-modification-mediated changes in the architecture of chromosomes, which may influence the functions and life-spans of cells, tissues, organs and individuals. It also addresses the occurrence and prevalence of chromosomal gaps and breaks.

Methods: Literature search was and is continuously done, acc. to topics of Chromosomics as specified in results. Also unpublished data from the lab of the author of the database is included, where appropriate.

Results: The yet 6 Chromosomics Databases available at https://cs-tl.de/DB.html provide details on chromosomal alterations like constitutional chromosomal breakpoints (CCPBs), chromosomal heteromorphisms (HMs), including UBCAs and EVS, small supernumerary marker chromosomes (sSMCs) and uniparental disomy (UPD). Furthermore, there is a small database on karyotypes of human and murine cell lines, which have been studied in the lab of the author. The sixth database was the starting point of the Chromosomics Databases, which includes a literature collection of articles dealing with multicolor-fluorescence in situ hybridization.

Conclusion: Overall, the Chromosomics Databases close a gap in the field of available, less chromosome oriented databases.

Conflict of Interest: Thomas Liehr UKJ Inst. of Human Genetics

EP14.010 Genome-wide identification of exon extension/shrinkage events induced by splice-site-creating mutations

Zhuo Qu 1, narumi sakaguchi1, Chie Kikutake1, Mikita Suyama1

1Kyushu University, Medical Institute of Bioregulation, Fukuoka, Japan

Background/Objectives: Mutations that affect phenotypes have been identified primarily as those that directly alter amino acid sequences or disrupt splice sites. However, some mutations that are not located in functionally important sites can also affect phenotypes. Splice-site-creating mutations (SCMs) are those types of mutations that create a novel splice site at a locus that is not normally present. There are some previous studies that have identified disease-associated of SCMs, but these studies are still sporadic. We aim to clarify the frequency and characteristics of abnormal splicing events induced by SCMs in normal individuals.

Methods: We used genotype data from the 1000 Genomes Project and transcriptome data from the GEUVADIS project to identify exon extension/shrinkage events and associated SCMs. We also analysed the characteristics of the identified events and their effect on the translation or protein products.

Results: We identified 383 exon extension/shrinkage events induced by SCMs, including 371 by SNVs and 12 by indels from 235 normal individuals. The average number of "exon extension" and "exon shrinkage" per individual was 4.6 and 11.5. We found that approximately 40.2% of the identified events may generate premature termination codons or disrupt protein domains.

Conclusion: Our results suggest that it is worth considering the impact of SCMs when identifying causative mutations in genetic diseases. The above results have been published in RNA Biology, 19(1), 1143-1152, 2022.

Grants: Uehara Memorial Foundation; JST SPRING; JSPS; Fukuoka Foundation for Sound Health; and JST, the establishment of university fellowships towards the creation of science technology innovation.

Conflict of Interest: Zhuo Qu JST, the establishment of university fellowships towards the creation of science technology innovation, Kyushu University, narumi sakaguchi JST SPRING, Kyushu University, Chie Kikutake Japan Society for the Promotion of Science (JSPS); Fukuoka Foundation for Sound Health, Kyushu University, Mikita Suyama Uehara Memorial Foundation, Kyushu University

EP14.011 Genetic Etiology of Children With Developmental Speech /Language Disorder

Emine Atlı 1, Engin Atli1, Yasemin Ozen1, Sinem YALÇINTEPE1, HAKAN GURKAN1

1Trakya University, Medical Genetics, Edirne, Türkyie

Genetic Etiology of Children With Developmental Speech /Language Disorder

Developmental language delay (DLD) is a frequent disability among children. The aetiology of speech and language deficits is influenced by genetic factors, as is widely established

We performed genetic analyzes based on the medical records of 33 patients who applied to our clinic with speech delay. Genetic testing was performed in those children referred to the geneticist of our center. The genetic tests performed included: Single Nucleotide Polymorphism (SNP) array (using a 180 K CGH + SNP (ISCA design, Agilent, Santa Clara, CA) array), karyotyping analysis.

Karyotype analyzes were normal in all 32 patients. A duplication of the 1p31.1p21.2 region was detected in the karyotype result of one of the patients. According to the array results, genomic changes were detected in 10 (%30,3) patients (Table 1). Deletions and duplications of different chromosomal regions were detected in our patient group. The genetic origin of approximately one-third of these children has been confirmed.

Molecular genetic testing should become part of the standard evaluation of children presenting with primary speech and language pathology. Unless there is a family history or a presence of clinical key features pointing towards a specific genetic disorder, genome-wide chromosome analysis with high resolution to detect CNVs is the test of first choice.

Patient

Genomic changes

1

6p23-22.3 deletion

2

17q21.31 deletion

3

9p24.1 duplication, 18p11.32 deletion

4

21q22.2 q22.3 deletion

5

1p31.1p21.2 duplication

6

8p11.23-p11.1 deletion

7

6q14.2-q14.5 deletion

8

13q33.3-q34 deletion

9

Yq11.223-q11.23 deletion

10

2p21 deletion

Conflict of Interest: None declared

EP14.012 Tissue-specific mitotic behavior of acrocentric ring chromosomes

Nadezda Shilova 1, Marina Minzhenkova1, Zhanna Markova1, Natalya Kuzina1, Nina Demina2, Artem Borovikov3

1Research Centre for Medical Genetics, Laboratory of Cytogenetics, Moscow, Russian Federation; 2Research Centre for Medical Genetics, Counselling Unit, Moscow, Russian Federation; 3Research Centre for Medical Genetics, Research and Counselling Department, Moscow, Russian Federation

Background/Objectives: The transition from a linear to a circular structure on ring chromosomes (r) leads to mitotic instability, resulting in somatic dynamic mosaicism (DM). This study reports on the varying degree instability of r(13) and r(21) in cultures of lymphocytes (LC) and skin fibroblasts (FC).

Methods: conventional cytogenetics, FISH with centromere, locus-specific and acro-p DNA-probes, chromosomal microarray analysis (CMA)

Results: In the case of r(13), DM was detected in both LC and FC. In LC, 75% of the cells exhibited r(13), 16% had double r(13), and 9% were monosomic for chromosome 13. The nucleolus organizer region (NOR) was observed at r(13). In FC harvested after 4 weeks cell lines with r(13) (30%), monosomy 13 (19%), and a tiny marker chromosome, mar(13), containing only the centromeric region and lacking NOR (51%) were revealed. After 12 weeks, 60% of the cells had mar(13), and 40% were monosomic for chromosome 13, with no detection of the cell line with r(13). For r(21), DM was also detected through CMA and FISH analysis. In LC, 83% of the cells exhibited double r(21), and 17% were monosomic for chromosome 21. NOR was not detected at r(21). FISH analysis of FC harvested after 4 weeks revealed cell lines with double r(21) (47%), monosomy 21 (33%), and a mar(21), containing only the centromeric region (20%).

Conclusion: This extreme degree of acrocentric (r) instability in FC is hypothesized to be the result of the initial absence or tissue-specific loss of the stalk material, which play an important role in stabilizing acrocentric chromosomes during DNA replication.

Conflict of Interest: None declared

EP14.013 Pitt-Hopkins syndrome: a comprehensive approach to diagnosis

Zhanna Markova 1, Darya Yurchenko1, Artem Borovikov2, Nadezda Shilova1

1Research Centre for Medical Genetics, Laboratory of Cytogenetics, Moscow, Russian Federation; 2Research Centre for Medical Genetics, Research and Counseling Department, Moscow, Russian Federation

Background/Objectives: Pitt–Hopkins syndrome (PTHS) is a rare inherited disease caused by a microdeletion on chromosome 18q21 or a heterozygous mutation in the TCF4 gene. The TCF4 variants responsible for typical PTHS usually arise as de novo mutations. However, germline or low-grade parental mosaicism has been reported in 2% of documented cases. Target FISH is a method that can be utilized to detect mosaic deletions

Methods: whole exome sequencing (WGS), FISH using developed home-made (hm) DNA-probes were employed.

Results: We present the case of a 1.5-year-old boy with distinct facial dysmorphisms, motor developmental delays, and congenital high myopia. WGS revealed a deletion of the long arm of chromosome 18: seq[GRCh37] del(18)(q21.2q21.31) chr18:g.52942713_54694498del. As the size of the detected deletion was 1.75 Mb and there were of commercial DNA probes available to validate the sequencing results, the task of developing hm DNA probes for the chromosomal region of interest were employed. We constructed a 40 bp probe composed of four unique PCR-derived fragments targeting the TCF4 locus on chromosome 18 and labeled in a nick-translation reaction. Subsequent FISH analysis confirmed the presence of microdeletion 18(q21.2q21.31) in the patient. Additionally, no insertion or mosaic form of deletion was detected in the patient’s parents, thereby ruling out these as possible causes of genomic imbalance formation.

Conclusion:. Based on the results of the FISH study, microdeletion 18(q21.2q21.31) was not only confirmed by an independent method, but its de novo origin was also established. The obtained data holds significant importance for genetic counseling of the family.

Conflict of Interest: None declared

EP14.014 Presentation of Sotos-like syndrome in patients with submicroscopic chromosomal abnormalities

Sara Veleska 1;1, Elena Sukarova1, Slavica Trajkova2, Learta Alili-Ademi3, Liljana Tasavska -Rmus1, Gordana Ilieva1

1Pediatrics, Department of Clinical Genetics, Skopje, Macedonia; 2University of Turin, Department of Medical Sciences, Turin, Italy; 3Pediatrics, Department of Neurology, Skopje, Macedonia

Background: Copy number variations (CNVs) are established cause for neurodevelopmental disorders. It has been proved that up to 10% of patients with intellectual disability have variations in copy number changes. Sotos syndrome (#11750) is multisystem disorder associated with CNVs in some cases. Changes in CNVs on chromosome 5 and 19 are clearly associated with the phenotype, however, several other alterations could be found in some cases.

Materials and methods: Three patients with features of Sotos syndrome were analyzed. In two of them the parents with similar features were analyzed as well. Both FISH using 5q probe (Vysis) and aCGH using Cytoscan 750K Array (Affymetrix) were performed.

Results: Phenotypic features of Sotos syndrome were found in all 3 patients (from 6 months -13 years of age) and two parents. FISH analysis (mother and child) confirmed deletion of 5q35 probe (including NSD1 gene) of in one family, later confirmed with aCGH. Deletion of 19q13 encompassing NFIX gene was found in one patient. The parents had normal intellectual presentation. In the third family CNV variations were found in two chromosomes -1q and 6q, not previously described with this phenotype.

Discussion and conclusion: Genotype-phenotype studies in Sotos syndrome patients are challenging due to the involved chromosome and gene, size of the variant and number of genes included in the region. Some of them are well described, some not referred so far. Evaluation of CNV is proved to be valuable tool for the identification of modifying factors for this phenotype.

References

Devriendt, Lourdes, Tattoon-Brown

Conflict of Interest: Sara Veleska full-time, Elena Sukarova: None declared, Slavica Trajkova: None declared, Learta Alili-Ademi: None declared, Liljana Tasavska -Rmus: None declared, Gordana Ilieva: None declared

EP14.015 Classification of human chromosomal heteromorphisms: a contribution to the lack of a universal scoring system

Sílvia Pires 1;2, Paula Jorge1;2, Thomas Liehr3, Natália Oliva-Teles1;2;4;5

1Centro de Genética Médica Doutor Jacinto Magalhães (CGM)/Unidade Local de Saúde de Santo António (ULSSA), Porto, Portugal; 2UMIB - Unidade Multidisciplinar de Investigação Biomédica/ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Portugal; 3Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, Jena, Germany; 4ITR - Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal; 5Faculty of Medicine, University of Porto, MEDCIDS – Departamento Medicina da Comunidade, Informação e Decisão em Saúde, Porto, Portugal

Background/Objectives: Chromosomal heteromorphisms (CHs) are morphological variations of specific constitutive heterochromatic regions, composed of satellite DNA tandemly repetitive sequences. In humans these sections are normally not transcribed and without phenotypic impact, although they are not devoid of genes (https://cs-tl.de/DB/CA/HCM/0-Start.html). However, their clinical significance is increasingly being discussed and numerous studies attempt to evaluate its influence on human diseases/susceptibilities. Several CHs classification methods were proposed, but there is no agreement on the standard sizes/scoring system of CHs; consequently, the classification of CHs is not accurate and comparing results from different scientific studies is difficult, leading to incorrect conclusions.

Methods: PubMed search for English-language publications between 1975-2023 describing CHs evaluation, its scoring systems and appropriate keywords was used for this study. 25 articles were identified, seven were excluded due to omission of a classification system and 18 were included.

Results: Several scoring systems with numerous variations each have been identified. Based on our experience and in order to clarify/simplify the scientific discussion, we have grouped scoring systems into three main types, based on the use of different methodologies/banding patterns and rating levels.

Conclusion: Prevously published approaches, CHs role, assessment and reporting in genetic diagnosis are inconsistent. Therefore, we argue that a uniform scoring system is needed for scientific reproducibility and for correct identification of human heteromorphisms. This analysis enhances their contribution for clinical significance/association with genetic diseases.

Grants: UMIB/ICBAS-UP/ITR (Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal and ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal).

Conflict of Interest: None declared

EP14.016 Utility of Exome Sequencing in the Setting of a General Pediatric Clinic for Hospitalized Children with Suspected Genetic Disorders

Georgia Christopoulou1, Alexandra Fountoulaki2, maria kassotaki2, ioanna lygerou2, maria michailou2, Ioannis Goniotakis2, rozanna poulaki2, ILIANNA MANIADAKI2, maria bitsori2, PELAGIA VORGIA3, chrissoula perdikogianni2, emmanouil paraskakis2, ioannis germanakis2, Pantelis Constantoulakis1, Eleftheria Papadopoulou 2

1GENOTYPOS Science Labs, Athens, Greece; 2University Hospital of Heraklion, Pediatrics, Heraklion, Greece; 3Hellenic Mediterranean University, Agri-Food and Life Sciences Institute, Research Centre, Heraklion, Greece

Background/Objectives: Genetic conditions contribute a significant portion of disease etiologies in children admitted to a General Pediatric Clinic (GPC). Exome Sequencing (ES) is a NGS method, identifying pathogenic or likely pathogenic single nucleotide variants (SNVs), small insertions/deletions (Indels) and copy number variants (CNVs). We aim to investigate the diagnostic impact of ES in children with suspected monogenic disorders, while receiving inpatient care

Methods: This retrospective study was performed on pediatric patients, admitted to the GPC at a tertiary medical hospital between November 2022 and November 2023, with various disorders and unrevealing workup before ES. The patients’ clinical features and molecular diagnoses based on ES results are reported in this study. ES was performed on DNA extracted from peripheral blood: library preparation was performed with Clinical Exome (Sophia Genetics) or Whole Exome (Twist Bioscience) and sequenced on NextSeq-550 (Illumina). Bioinformatics analyses were conducted by SOPHiA DDM® bioinformatics pipelines. All ES were performed in a specific laboratory.

Results: A total of 52 probands were evaluated and referred for ES. Neurological phenotype was the most common (24/52), including neurodevelopmental (11/24), neurocutaneous (7/24), neuromuscular (4/24) and neurometabolic disorders (2/24). A molecular diagnosis was reached in 23/52 patients (44.2%). 18/23 patients had pathogenic/likely pathogenic variants for monogenic disorders, 2/23 a pathogenic risk factor for multifactorial disease and 3/23 CNVs indicative of genetic syndromes. In 13/52 the results were inconclusive, considering further action.

Conclusion: ES consists a powerful tool, revealing a significant fraction of genetic etiologies among children admitted to a GPC, ending their diagnostic odyssey.

Conflict of Interest: None declared

EP14.017 New integrated approaches to solve the genomic complexity of Ring14 syndrome

Laura Formentini 1, Giulia Olivucci1;2, Anna Basile3, Mariia Zadorozhna3;4, Paola Dimartino3, Elisa Giorgio3;4, Malte Spielmann5;6;7, Tommaso Pippucci1

1IRCCS University Hospital of Bologna Sant Orsola Polyclinic, Bologna, Italy; 2University of Palermo, Department of Surgical, Oncological and Stomatological Sciences, Palermo, Italy; 3University of Pavia, Department of Molecular Medicine, Pavia, Italy; 4IRCCS Mondino Foundation, Neurogenetics Research Centre, Pavia, Italy; 5Max Planck Institute for Molecular Genetics, Development and Disease Group, Berlin, Germany; 6Institute of Human Genetics, University Hospital Schleswig-Holstein, University of Lübeck and Kiel University, Lübeck, Germany; 7DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany

Background/Objectives: Ring14 (r14) syndrome is a rare neurodevelopmental disorder characterised by a complex phenotype including drug-resistant epilepsy. It is caused by the rearrangement of chromosome 14 into a ring-shaped structure originated from two breakpoints fused together with terminal deletions. This loss of genetic material doesn’t explain some clinical features: e.g., individuals carrying comparable 14q linear deletions don’t exhibit epilepsy. The aim of our project is to define the molecular mechanisms underlying r14 phenotype through an integrated approach with state-of-the-art methods.

Methods: We obtained Lymphoblastoid Cell Lines from 10 patients and 5 healthy parents, courtesy of the Telethon Network of Genetic Biobanks. Analyses will be conducted in parallel with complementary approaches, integrating DNA sequencing with long read Oxford Nanopore Technology, genome-wide chromosome conformation capture (Hi-C) and mRNA sequencing.

Results: We present a novel approach to study r14 molecular aetiology. Hi-C sequencing data will allow characterization of the impact of the r14 on 3D genome architecture, potentially leading to Topologically Associating Domain (TAD) disruption, abnormal chromatin interaction, and subsequent misregulation of gene expression. Aligning long-reads to telomere-to-telomere assembly will enable the definition of complex rearrangements in r14 and breakpoints in previously inaccessible regions. Using RNAseq we’ll identify differentially expressed genes between controls and patients, pinpointing disease-related transcriptome changes.

Conclusion: Although results are pending, this research holds promise for contributing to a better understanding of the underlying biology and genotype-phenotype correlation in Ring14 syndrome and at eventually improving patient care and therapeutic options for this rare but extremely severe disorder.

Grants: This research is funded by Ring14 International Onlus

Conflict of Interest: None declared

EP14.018 Homozygosity for a PLOD1 pathogenic variant due to paternal segmental UPD in a patient with kyphoscoliotic Ehlers-Danlos syndrome.

Caterina Marconi 1, Lina Quteineh1, Sebastian Suchet1, Frédéric Masclaux1, Jean-Louis Blouin1;2, Joël Fluss3, Marc Abramowicz1;2, Michel Guipponi1;2

1University Hospitals of Geneva (HUG), Division of Genetic Medicine, Geneva, Switzerland; 2University of Geneva-School of Medicine, Department of Genetic Medicine and Development, Geneva, Switzerland; 3University Hospitals of Geneva (HUG), Pediatric Neurology Unit, Geneva, Switzerland

Background/Objectives:Segmental uniparental isodisomy is a rare (estimated prevalence 0.03%) mechanism that can induce homozygous variant causing autosomal recessive disease.

Methods:Here we report the case of a 7-year-old girl born premature with severe hypotonia and flaccid paralysis of the upper limbs. She has numerous dislocations associated to his hyperlaxity and severe neuromuscular kyphoscoliosis.

Results:Genome sequencing allowed us to identify a homozygous variant p.(Gln467*) in the PLOD1 gene that causes a kyphoscoliotic Ehlers-Danlos syndrome. The variant was heterozygous in the father but absent in the mother. Sample swap was excluded by microsatellites analysis. Runs of homozygosity (ROH) analysis of sequencing data excluded a high degree of consanguinity but identified a single 12Mb ROH on the distal 1p (1p36.33p36.22) including PLOD1. No deletion in this region was detected by array-CGH nor CNV analysis of sequencing data. We tested 5 SNVs distributed throughout the ROH that resulted all homozygote in the proband, heterozygote in the father and absent in the mother, confirming a segmental paternal isodisomy. Microsatellites analysis will allow us to verify a meiotic (non-disjunction) or mitotic (post-zygotic crossover) origin of this event.

Conclusion:We describe a case of segmental isodisomy of distal chromosome 1p resulting in homozygosity for a loss of function variant in the PLOD1 gene. Characterization of these molecular defects will allow us to provide appropriate recurrence risk counseling. Finally, we suggest that ROH analysis of sequencing data can be a valuable tool for causative variants identification in very rare autosomal recessive disorders even in non-consanguineous families.

Grants:QS07-38

Conflict of Interest: None declared

EP14.019 Trisomy 9p and Monosomy 2q: A rare chromosomal aberration with variable phenotypic presentation

Hiba Abu Khalil 1, Mohammed Albalwi2;3, Norah Alsaleh1;4

1King Abdullah Specialist Children’s Hospital (KASCH), King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNG-HA), Genetics & Precision Medicine, Riyadh, Saudi Arabia; 2King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNG-HA), Division of Molecular Pathology, Department of Pathology, Riyadh, Saudi Arabia; 3College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNG-HA), Riyadh, Saudi Arabia; 4King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs (MNG-HA), Medical Genomics Research Department, Riyadh, Saudi Arabia

Background/Objectives: Trisomy 9p and partial monosomy 2q represent distinct entities within the realm of chromosomal abnormalities. Trisomy 9p is an infrequent chromosomal anomaly characterized by the duplication, either complete or partial, of the short arm of chromosome 9. Conversely, partial monosomy 2q is an exceedingly rare chromosomal aberration involving chromosome 2, with only a handful cases reported globally.

Methods: The patient’s DNA underwent karyotyping, FISH and array CGH analysis that are routinely performed as standard patient care protocol.

Results: We present a 4-month-old male exhibiting various dysmorphic features, including bitemporal narrowing, microphthalmia, downslanting palpebral fissures, depressed nasal bridge, cupped ears, and retrognathia. Furthermore, he presents with ambiguous genitalia, hypotonia, and a congenital heart defect characterized by bicuspid aortic valve. Chromosomal analysis revealed an unbalanced translocation between the short arm of chromosome 9 and long arm of chromosome 2 resulting in a karyotype of 46, XY, der(2)t(2,9)(q37;p12), which was further confirmed by FISH analysis. Subsequent array CGH validated the size of the deletion on chromosome 2 (6.7 Mb) and the duplication on chromosome 9 (40.7 Mb).

Conclusion: Given the substantial impact of the deletion and duplication involving chromosome 2 and 9, respectively, which encompass numerous genes, providing precise counseling to the family regarding the prognosis and outcome of these rare chromosomal structural disorders poses a significant challenge. Furthermore, the scarcity of reported cases, compounded by the fact that many cases are identified prenatally and subsequently terminated, presents an even greater obstacle in predicting the complete phenotype and disease progression.

Grants: None

Conflict of Interest: None declared

EP14.020 A case of Immunodeficiency, Centromeric instability, Facial anomalies (ICF) syndrome diagnosed by Karyotype analysis but not Molecular testing

Omamah AlShehri 1, Abdulelah Abunadi2, Ashwaq Alghamdi2, Aziza AlKhalidi2, Bader AlMuzaini2;3, Mohammed Al Balwi2;3;4

1King Abdulaziz Medical City, Ministry of National Guard - Health Affairs, Genetics and Precision Medicine Department, King Abdullah Specialized Children’s Hospital, Riyadh, Saudi Arabia; 2King Abdulaziz Medical City, Ministry of National Guard - Health Affairs, Department of Pathology and Laboratory Medicine, Riyadh, Saudi Arabia; 3King Saud bin Abdulaziz University for Health Sciences, Abdullah International Medical Research Center, Riyadh, Saudi Arabia; 4King Saud bin Abdulaziz University for Health Sciences, College of Medicine, Riyadh, Saudi Arabia

Background/Objectives: The Immunodeficiency, Centromeric instability, Facial anomalies (ICF) syndrome is a rare autosomal recessive disease presenting with immunodeficiency, hypo- or agammaglobulinemia, developmental delay, and facial anomalies. Centromeric instability is the cytogenetic hallmark of the disorder which results from chromosomal rearrangements. Mutations in two genes have been discovered to cause ICF syndrome: DNMT3B and ZBTB24 however few cases reported with no genetic mutation.

Methods: we’re reporting a case of ICF syndrome Diagnosed by Karyotyp with negative Molcular testing.

Results: A 13 months old male with hypotonia, dysmorphisim of high forehead, upslanted palpebral fissures, microphthalmia, long philtrum, retrognathia, low seated ears and No history of infections. Karyotype showed a triradial chromosome 1 and decondensation of pericentromeric heterochromatin in 1 cell 46,XY,tr(1)(p,q,q)[1]/46,XY[49].

Conclusion: This case highlights the importance of cytogenetics in diagnosing such a rare and atypical case.

Grants: none

Conflict of Interest: None declared

EP14.021 Focus on deletions and duplications identified by Optical Genome Mapping technique in a cohort of 100 patients : comparison with MCA.

Martine Doco-Fenzy 1;2, Olivier Pichon1, Faezeh Vasheghani1, Emilie Landais3, Tony Yammine2, Sylvie Bernard4, Poirsier Céline2, nathalie Le Du5, vincent jauffret4, Noemie Celton5, Kamran Moradkhani6, nicolas gruchy7, Agnes Guichet8, Sylvie Jaillard9, Sylvie Odent9, Marie Vincent1, Mathilde Nizon1, Bertrand Isidor1, Sandra Mercier1, celine bonnet10, Marc-Antoine Belaud-Rotureau11, Clemence Jacquin3, Nathalie Bednarek12, Jonathan Levy13, Laetitia Lambert10, Mylene Dexheimer14, stéphane bezieau1

1CHU Nantes, genetics, Nantes, France; 2CHU Reims, genetics, Reims, France; 3CHU Reims, Reims, France; 4Cytogen laboratory, Nantes, France; 5CHU Tours, genetics, Tours, France; 6CHU Nantes, Nantes, France; 7CHU Caen, genetics, Caen, France; 8CHU Angers, genetics, Angers, France; 9CHU Rennes, Rennes, France; 10CHU Nancy, genetics, Nancy, France; 11CHU Rennes, genetics, Rennes, France; 12CHU Reims, Pediatrics, Reims, France; 13CHU Robert Debré, genetics, Paris, France; 14CHU Nancy, Nancy, France

Introduction: Optical Genome Mapping (OGM) Bionano ®technology is based on the fluorescent marking of long DNA molecules aligned in electrophoretic field via nano-channels. The Saphyr scanner reads the fluorescent signals and the results are interpreted using Bionano access software. The Nantes Hospital used OGM for an oncohemato-constitutional project on 150 patients.

Method: In constitutional validation process, we compared the sizes of deletions and duplications obtained by OGM and CMA (Agilent® 60k ISCA chips) through 15 duplications and 15 deletions. The CNVs studied on chromosomes 1,5,8,9,11,15,16,19,22, and X varied from 4.3kb to 39Mb.

Results : For duplications, the CNV sizes differences (DSs) OGM/CMA ranged from 2189bp to 1014459bp respectively and differences of the breakpoints (DBPs) ranged from 120bp to 824500bp (OGM/CMA).The ratio between DSs and average CNVs size (DS/MS) ranged from 0.002 (dup 35Mb) to 0.84 (dup 1.804Mb). For deletions, the DSs ranged from 1412bp to 272470bp, DBPs ranged from 2537bp to 226462bp, and DS/MS ranged from 0,26 to 1,99 (8 cases). Discrepancies are notably explained by location of breakpoints (eg. telomeric and centromeric).

Conclusion: This study participates in our laboratory in method’s validation of OGM technique confirming the concordance of CNVs seen in OGM and evaluating the differences on various genomic regions. We confirmed the reliability and evaluated the uncertainty concerning the BPs identified by OGM/CMA approaches. By mirror effect the OGM technique enables the verification of BPs identified by CMA. This work is useful for data interpretation, defining the limit and resolution of both techniques.

Conflict of Interest: None declared

EP14.023 Implementation and validation of the dicentric chromosome assay for the biological dosimetry purposes following the irradiation by various radiation fields

Jakub Vavra1, Johana Alaverdyan 1, Artur Sergunin1, Daniela Ekendahl1

1National Radiation Protection Institute, Dosimetry department, Prague 4, Czech Republic

Background/Objectives: Biological dosimetry aims to determine the dose based on the assessment of radiation exposure biomarkers. The dicentrics formation in peripheral blood lymphocytes is the most prominent of such biomarkers being highly specific to ionizing radiation. The aim of the work was to establish and validate the internal calibration curves for the dicentric chromosome assay (DCA) considering various radiation fields relevant to the clinical or emergency purposes.

Methods: The blood samples of 30 volunteers in total were irradiated in vitro either by linear accelerator (X-ray) or training reactor (mixed field of neutrons and gamma rays). The doses applied were in the range of 0.1 – 7.5 Gy. Following that, the blood samples were cultivated and the metaphases of peripheral blood lymphocytes were imaged and analysed for the dicentrics frequency by the automatic scanning microscopy system.

Results: The calibration curves for the DCA upon X-irradiation and mixed field irradiation were established and validated by the analysis of the samples irradiated by a priori blind doses. Whereas the calibration curve for X-ray exhibited the linear-quadratic character, it was rather linear in case of mixed field irradiation.

Conclusion: In this study, the applicability of DCA for the biological dosimetry purposes was proven. The key step was to establish the internal calibration curves to eliminate the possible interlaboratory differences in dose estimation. Moreover, the established calibration curve for the DCA upon the mixed field of neutrons and gamma rays irradiation is unique.

Grants: Supported by the grant VK01020052 from the Ministry of the Interior of the Czech Republic.

Conflict of Interest: None declared

EP14.024 A new case with coexistence of mosaic 48, XYYY/47, XYY and CACNA1E variant in autism spectrum disorder

Aysel Kalayci 1, Deniz Agirbasli2, Nihal Serdengeçti3, Mustafa Tarık Alay2, Mahmut Cem Tarakçıoğlu3, Mehmet Seven2

1Istanbul University-Cerrahpaşa, Medical Genetics, Istanbul, Türkyie; 2Istanbul University-Cerrahpaşa, Medical Genetics; 3Istanbul University-Cerrahpaşa, Child-adolescent psychiatry

Background/Objectives: Autism spectrum disorder (ASD), is one of the most genetically heterogeneous progressive neurobehavioral disorders. Although autistic behaviors are defined in many genetic syndromes. The etiology and the inheritance pattern are usually unknown. Here we report a case with mosaic 48,XYYY/47,XYY and CACNA1E variant in an ASD patient.

Methods: For chromosome and FISH analyses, peripheral blood was obtained from both the patient and the parents. Chromosome preparations were analyzed using conventional Giemsa staining and 100 metaphases. FISH analysis of the patient used a specific alpha satellite X chromosome and specific alpha satellite Y chromosome. For the evaluation of sequence variants associated with ASD, WES analysis was performed.

Results: The index case is a 3-year-old male with the diagnosis of ASD. The patient’s physical development was normal, the family applied to the child psychiatry outpatient clinic due to failure to speak at 30 months. Neurological examination was normal. He had mild dysmorphic features. His current Childhood Autism Rating Scale (CARS) score was 34 points (mild to moderate autism). He is diagnosed with ASD according to DSM-V criteria. Chromosomal analysis of the patient revealed mos 48,XYYY[28]/ 47,XYY[72] karyotype. In FISH analysis, nuc ish(DXZ1x1, DYZ1x3)[44]/(DXZ1x1,DYZ1x2)[156] was detected. WES results displayed a heterozygous missense VUS (c.3545G>A) in CACNA1E gene.

Conclusion: 48,XYYY/47,XYY karyotype and CACNA1E variant together may contribute to phenotypic heterogeneity. Further investigation about the functionality of the variant in CACNA1E is needed.

Grants: This research received no specific grant from any funding agency in the public, commercial, or not for profit sectors

Conflict of Interest: None declared

EP14.025 Genome sequencing uncovers a 8,8 kb deletion between L1CAM and AVPR2 in a boy with Hirschsprung disease and nephrogenic diabetes insipidus

Ingrid Bader 1, Kathrin Buder2, Stefanie Beck-Wödl1, Marc Sturm1, Petra Stöbe1, Olaf Riess1, Tobias Haack1

1Universitätsklinikum Tübingen, Institut für Medizinische Genetik und Angewandte Genomik, Tübingen, Germany; 2Universitätsklinikum Tübingen, KfHNierenzentrum für Kinder u. Jugendliche, Tübingen, Germany

Background/Objectives: Pathogenic loss-of function variants and missense-variants in AVPR2 (Arginine Vasopressin Receptor 2, OMIM * 300538) are a known cause of X-linked nephrogenic diabetes insipidus. Pathogenic loss-of function variants and missense-variants in L1CAM (L1 Cell Adhesion Molecule, OMIM * 308840) are a known cause of X-linked hydrocephalus with or without Hirschsprung disease (OMIM # 307000). Here we describe a 1,5 year old boy with Hirschsprung disease and nephrogenic diabetes insipidus.

Methods: Genome sequencing was performed using Illumina short read technology.

Results: Genome sequencing uncovered an 8,8 kb Deletion in Xq28 (NC_000023.11:g.[153883503_153892327del];[0]) located in between the two adjacent genes L1CAM and AVPR2. It affects the 5’ UTR exon 1 of L1CAM and it terminates 11 kb before the 5’-end of the nearest annotated AVPR2 -trancript (NR_027419.2). The deletion does not affect any annotated coding or non-coding transcripts of AVPR2

Conclusion: The unusual combination of the very specific and rare symptoms of the patient raises the hypothesis that this 8,8 kb deletion affects chromosomal material that regulate the expression of both genes. Further analyses need to be performed to demonstrate dysregulation of both genes. The respective deletion could be the causative variant in other families with X-linked nephrogenic diabetes insipidus were no disease causing variant in the AVPR2-gene could be found.

Conflict of Interest: None declared

EP14.026 Two cases of HDR syndrome due to a large deletion in the 10p15.3p14 region

Gülhanım Memiş 1, hatice arslan1, sinan akbaş1, Ayça Dilruba aslanger1, Gozde Yesil Sayin1, sevgi yavuz2, aslı al3, bağdagül aksu4, zeynep yıldırım4, alev yılmaz4, firdevs baş3, Bilge Ozsait Selcuk1, Birsen Karaman1;5

1Istanbul University Faculty of Medicine, Department of Medical Genetics, ISTANBUL; 2Başakşehir Çam and Sakura City Hospital, Department of Child Health and Diseases, Division of Pediatric Nephrology, ISTANBUL; 3Istanbul University Faculty of Medicine, Department of Child Health and Diseases, Division of Growth Development and Pediatric Endocrinology, ISTANBUL; 4Istanbul University Faculty of Medicine, Department of Child Health and Diseases, Division of Pediatric Nephrology, ISTANBUL; 5Istanbul University, Child Health Institute, Department of Pediatric Basic Sciences, ISTANBUL

Background/Objectives: Barakat syndrome, also known as HDR syndrome, is a rare disorder characterized by the triad of Hypoparathyroidism, sensorineural Deafness and Renal disease, caused by deletions in chromosome 10p14 or pathogenic mutations in the GATA3 gene. Our aim is to elucidate the clinical and genetic characteristics of two cases with a gross deletion in 10p15.3p14 resulting from diverse chromosomal alterations, exhibiting recognizable facial features in addition to the triad of Barakat syndrome.

Methods: Diagnosis involved conventional cytogenetic technology, Fluorescence-in-situ-hybridization/(FISH) and array-based comparative genomic hybridization/(a-CGH)

Results: Clinical examination revealed similar facial dysmorphism in both cases. Karyotype analysis of the first case indicated a terminal deletion on 10p. A-CGH analysis identified a 10.9 Mb copy number loss of the 10p15.3p14 region containing the GATA3 gene. In the second case, karyotype and FISH analyses unveiled an unbalanced de novo translocation between chromosomes 10 and 19, leading to a 10p14 deletion, as confirmed by a 9.1 Mb deletion at 10p15.3p14 through a-CGH.

Conclusion: The identification of 10p15.3p14 deletions enabled us to diagnose HDR syndrome due to the haploinsufficiency of the GATA-3 gene and also to investigate additional symptoms of the deletion. We propose that deletion of the 10p15.3p14 region represents a contiguous gene deletion syndrome with a distinct phenotype comprising variable features of HDR syndrome alongside manifestations of facial dysmorphism and intellectual disability. Furthermore, we emphasize the importance of combined use of cytogenetic and molecular cytogenetic techniques in order to obtain a more precise physical mapping of 10p terminal deletions and an accurate genotype-phenotype correlation.

Grants:

Conflict of Interest: None declared

EP14.027 A Complex Clinical Scenario: A Newborn with Multiple Congenital Anomalies with a de novo chromosomal defect of der(9)inv(9)(p24q13)dup(9)(q21.11q34.11)

Mohammed Al Balwi 1;2;3, Roaa Alahmadi4, Nedaa Aldossary4, Khalid Alshalan4, Albandri Alzaid4, Mohammed Siraj4, Mshari alhwiti4, Nabilah Alghamdi4, Razan Almudhhi1, Aziza AlKhalidi1, Malak Alghamdi5

1King Abdulaziz Medical City, Ministry of National Guard - Health Affairs, Department of Pathology and Laboratory Medicine, Riyadh, Saudi Arabia; 2King Saud bin Abdulaziz University for Health Sciences, College of Medicine, Riyadh, Saudi Arabia; 3King Saud bin Abdulaziz University for Health Sciences, Abdullah International Medical Research Center, Riyadh, Saudi Arabia; 4King Khalid University Hospital, King Saud University Medical City, Department of Medicine laboratory, Riyadh, Saudi Arabia; 5King Khalid University Hospital, King Saud University Medical City, Medical Genetic Division, Pediatrics Department, Riyadh, Saudi Arabia

Background/Objectives: Partial trisomy 9p is the most common chromosomal disorder reported in the literature. However, very few cases with duplicated 9q have been reported. Here, we reported a newborn with complex congenital anomalies and complex chromosomal rearrangement of derivative chromosome 9 that resulted from a pericentric inversion and an interstitial duplication of the long arm 9q21.11q34.11.

Methods: Clinical examination, G-banding Karyotype, FISH analysis and array-CGH were performed.

Results: A 3-week-old baby girl who was born with dysmorphic features and multiple congenital anomalies involving the central nervous system (CNS), cardiovascular system (CVS), and renal system. Diagnostic imaging revealed severe supratentorial hydrocephalus and a relatively small posterior fossa with effacement of the fourth ventricle, suggestive of Chiari II malformation. Cytogenetic banding analysis revealed a female karyotype [46,XX,der(9)inv(9)(q13p24)dup(9)(q21.11q34.11)] with de novo derivative chromosome 9 that resulted from a pericentric inversion with breakpoints in 9p24 and 9q13 and an interstitial duplication of the long arm between breakpoints 9q21.11 and 9q34.11. The pericentric inv(9) was confirmed by FISH analysis. Array CGH analysis identified ~63.14Mb a de novo heterozygous duplication on 9q21.11q34.11 (64,964,986-128,102,370) encompassing 518 genes. Parental karyotypes were normal.

Conclusion: To our best knowledge, this is the first case report with a derivative chromosome 9 that resulted from a pericentric inversion and an interstitial duplication of the long arm 9q21.11q34.11. Our clinical findings provide evidence on the possible important contribution of this precise structural chromosome abnormality to elucidate and early diagnosis of genetic disorders. Further, evaluation of partial 9q trisomy suggested specific clinical care and better patient conseling.

Grants: None

Conflict of Interest: None declared

EP14.029 Alu-element insertion as a second pathogenic hit in USH2A associated with retinitis pigmentosa

Janne Sofie Wehnert 1, Yorck Hellenbroich2, Malte Spielmann2, Inga Nagel1

1University Hospital Schleswig-Holstein, Institute of Human Genetics, Kiel, Germany; 2University Hospital Schleswig-Holstein, Institute of Human Genetics, Lübeck, Germany

Background/Objectives: Insertions of transposable elements (TEs) have rarely been found to be associated with monogenic disease. Such events have been described for conditions like Bardet-Biedl syndrome, Pompe disease, and Lynch syndrome. In this study, we identified an Alu-element insertion into the USH2A gene as the second pathogenic hit in a patient with retinitis pigmentosa.

Methods: Exome and genome sequencing were conducted on a 41-year-old female patient with retinitis pigmentosa, following standard procedures. Subsequently, Illumina’s long-read sequencing technology was used to better characterize a complex alteration in the USH2A gene.

Results: The patient was found to harbor the pathogenic variant NM_206933.4:c.10561T>C p.(Trp3521Arg) in USH2A, which is associated with autosomal recessive retinitis pigmentosa. A second hit was called as unknown feature elongation based on the exome and genome sequencing data. Interpretation of the sequence alignments suggested an insertion exceeding 150 bp. Long-read sequencing revealed a 386 bp long insertion into the USH2A gene (NC_000001.11:chr1:g.215845960_215845961ins[N[372];215,845,947_215845960]), identified as an Alu element (AluYg6).

Conclusion: Illumina’s long-read sequencing technology successfully detected a 386 bp Alu-insertion into the USH2A gene. It is tempting to speculate that, due to technical limitations, the identification of such insertions as a cause of disease is less frequent than their actual occurrence.

Grants:

Conflict of Interest: Janne Sofie Wehnert provision of free of charge sequencing reagents and software by Illumina within the framework of a collaborative research study, Yorck Hellenbroich provision of free of charge sequencing reagents and software by Illumina within the framework of a collaborative research study, Malte Spielmann provision of free of charge sequencing reagents and software by Illumina within the framework of a collaborative research study, Inga Nagel provision of free of charge sequencing reagents and software by Illumina within the framework of a collaborative research study

EP14.030 An interstitial duplication of chromosome An interstitial duplication of chromosome 15q11.2q13.1 in a Saudi baby girl with global developmental delay and aggressive behavior

Azizah AlKhaldi 1, Mohammed Adam1, Alaa Almeleihi1, Salman Alsaad1, Areej AlAtawi2, Mohammed Al Balwi1;3;4

1King Abdulaziz Medical City, Ministry of National Guard - Health Affairs, Department of Pathology and Laboratory Medicine, Riyadh, Saudi Arabia; 2King Abdulaziz Medical City, Ministry of National Guard - Health Affairs, Genetics and Precision Medicine Department, King Abdullah Specialized Children’s Hospital, Riyadh, Saudi Arabia; 3King Saud bin Abdulaziz University for Health Sciences, College of Medicine, Riyadh, Saudi Arabia; 4King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia

Consortium: None

Background/Objectives: Chromosome 15q duplication syndrome is a rare congenital developmental disorder that may presented with virable clinical presentation according to the size of duplication. Generally, Dup 15q is characterized by hypotonia, motor delays, variable intellectual disability, autism spectrum disorder, and epilepsy including infantile spasms.

Methods: Patien’s physical examination, conventional cytogenetic analysis, fluorescence in situ hybridization analysis, array-CGH analysis and multiplex ligation-dependent probe amplification were performed as standard care protocol to investigate genetic disorder.

Results: A 34-month-old baby girl with global developmental delay, central hypotonia with autism spectrum disorder, delay speech, hyperactive with aggressive behaviour and poor social skills. Her brain MRI was abnormal and suggestive of hypomyelination. Chromosomal analysis revealed a female karyotype with an interstitial heterozygous duplication within the long arm of chromosome 15 involving cytogenetic band 15q11q13. Subsequent array CGH and FISH analysis were confirmed the duplication 15q11.2-13.1 region encompasses imprinted genes within the Prader-Willi/Angelman Critical Region (PWACR). Furthermore, multiplex ligation-dependent probe amplification (MLPA) analysis revelead that this duplication was a de novo finding.

Conclusion: Clinical variability of duplicated 15q11q13 is most likely caused by more PWACR cluster imprinted genes region which are contribute to the patient phenotype. This type of chromosomal anomaly might be more common than reported, so furthermore an extensive geneic investigations with array CGH and MLPA analysis may provide a robust methodology should always be considered to improve early diagnosis and better patient conseling.

Grants: None

Conflict of Interest: None declared

EP14.031 Interstitial deletion 3p14.1p13 including the entire MITF gene

Kristina Gornik Crkvenac 1, Ana Juras1, Nevena Krnic2, Sanda Huljev Frkovic3

1University Hospital Centre Zagreb, Department of Laboratory Diagnostics, Division of Cytogenetics, Zagreb, Croatia; 2University of Zagreb School of Medicine, University Hospital Centre Zagreb, Department of Paediatrics, Division of Pediatric Endocrinology and Diabetes, Zagreb, Croatia; 3University of Zagreb School of Medicine, University Hospital Centre Zagreb, Department of Paediatrics, Division for genetics and metabolism, Zagreb, Croatia

Background/Objectives: The microphthalmia-associated transcription factor (MITF) is a basic helix-loop-helix leucine zipper transcription factor, which regulates melanocyte development and the biosynthetic melanin pathway. Heterozygous MITF mutations cause Waardenburg syndrome, type 2A (WS2A) and Tietz syndrome, with autosomal dominant inheritance.

Methods: We report a 17-year-old patient presented with bilateral perceptual anacusia, condition after right cochlear implantation, bilateral high myopia, horizontal nystagmus, dystrophic changes of the retina, intellectual disability, liver hemangioma, premature appearance of gray hairs and white coffee stains. Molecular karyotyping on proband was performed on the SurePrint G3 Human CGH 8x60 microarray platform (Agilent Technologies, Santa Clara, CA, USA).

Results: Array CGH analysis of the DNA sample showed an interstitial deletion 3p14.1p13 (66060402_70041375). The size of the found deletion is 3.98 Mb. The deleted region contains 14 OMIM genes. Pathogenic are: SLC25A26, EOGT, LMOD3 and MITF. Haploinsufficiency of the MITF gene causes Tietz syndrome. Patients with Tietz syndrome have congenital sensorineural HL, blue iris without heterochromia iridis, and skin and hair color lighter than those of their relatives. They are born with white hair and very pale skin, but their hair color often darkens over time. Haploinsufficiency of the MITF gene also causes WS2A. WS2A patients exhibit congenital deafness, blue iris, patchy depigmentation of the skin and eyes (white foelock and heterochromia iridis), but no dystopia canthorum.

Conclusion: Elucidation of the molecular genetics underlying pigmentation has revealed genes important for melanocyte development and function. It has been demonstrated that mutations of the same gene may cause different hypopigmentation syndromes that may have similar phenotypes.

Conflict of Interest: None declared

EP14.032 Unusual phenotypic manifestations in a patient with partial trisomy 10q and partial monosomy Xq due to unbalanced chromosomal translocation (X;10)

Irena Bradinova 1;2, Silvia Andonova2, Radoslava Vazharova3, Ralitsa Robeva4;5, Alexey Savov1;2

1Medical University - Sofia, Faculty of Medicine, Department of Obstetrics and Gynecology, Sofia, Bulgaria; 2UHOG “Maichin dom”, National Genetic Laboratory, Sofia, Bulgaria; 3Sofia University “St Kliment Ohridki”, Faculty of Medicine, Department of Biology, Medical Genetics, and Microbiology, Sofia, Bulgaria; 4Medical University - Sofia, Faculty of Medicine, Department of Endocrinology, Sofia, Bulgaria; 5USHATE “Acad. Iv. Penchev”, Sofia, Bulgaria

Background/Objectives: Unbalanced chromosomal translocations, either de novo or inherited, can give rise to developmental delay, intellectual disability, abnormal growth, dysmorphic features, and congenital anomalies.

We present a 24 years-old-woman referred to genetic counselling with amenorrhea, hypergonadotropic hypogonadism, mild intellectual disability, discrete dysmorphic features and abnormal ophthalmological findings of nystagmus and severe myopia. Height and weight were within normal ranges. The patient was born from first uneventful pregnancy of healthy couple with negative family history of miscarriages or congenital anomalies. At 9 months, developmental delay, growth retardation, muscular hypotonia, and nystagmus were observed, leading to a misdiagnosis of cerebral palsy.

Methods: Clinical dysmorphology assessment, endocrinological examination and hormonal tests were performed. Subsequently genetic analyses including karyotype, QF PCR and aCGH were initiated. BrDU assay was also performed for identification of the X-inactivation pattern.

Results: Normal QF PCR result and karyotype 46,XX were obtained. The aCGH revealed partial 10q trisomy (50Mb) and partial monosomy X (43Mb). Re-analysis of the karyotype identified a discreet de novo unbalanced chromosomal translocation (Xq;10q). Skewed X-inactivation with preferential inactivation of the rearranged X-chromosome was subsequently detected.

Conclusion: The unbalanced chromosomal translocation (Xq;10q) of our patient resulted to a unique clinical profile characterized by a combination of symptoms characteristic for 10q duplication and Xq deletion. Additionally, the skewed X-inactivation pattern is hypothesized to underlie the comparatively milder phenotype observed.This case highlights the diagnostic challenges associated with unbalanced chromosomal rearrangements and emphasizes the diagnostic role of combining different genetic techniques especially in individuals with dysmorphic features, intellectual disability, and disorders of sex development.

Conflict of Interest: None declared

EP14.033 Case report on Xq isochromosome in the boy

Aline Aywaz 1, Seda Hakobyan1, Susanna Midyan1, Tatevik Hovhannisyan1, Arpine Mkrtchyan1, Natella Kostandyan2

1Center of medical genetics and primary health care, Cytogenetic, Yerevan, Armenia; 2Center of medical genetics and primary health care, Genetic counseling, Yerevan, Armenia

Background: The child was from the 1st pregnancy, in the 38th week of gestation, with normal biomechanism, weight 3100 g, height 47 cml. He had insufficient weight and height gaining since birth. The motor, mental and speech development were corresponded to age. In 2019 he was diagnosed with hypothyroidism. Over the last year, mother has noticed learning difficulties, attention and concentration deficits.

His data at at 11y.o: weight 29 kg, height 126.5 cm, head circumference 52.5 cm. He has long and narrow face, downslated palpebral fissures, almond shape of eyes, mild ptosis from both side, downslated corners of mouth, big ears and brachidactyly. Proximal parts of upper limbs are short. Hypermobility of hands joints and shield shaped thorax.

Mother’s height is 150 cm, she has short arms. Mother’s paternal aunt and paternal grandmother also have short stature. They were all fertile

Methods: Karyotype and FISH studies have been performed to the child and the mother.

Results: Karyotype study of the patient revealed isochromosome X- 46,Y(iX). The patient’s mother karyotype revealed 46,Xi(X). FISH study of the boy showed the absence of SHOX gene and “p” arm on the single X chromosome. In the same study of the mother the single copy of the SHOX gene has been detected on the normal X chromosome.

Conclusion: In the literature an isochromosome of the long arm of chromosome X, is found in 15%–18% of Turner syndrome cases and it is observed exclusively in women. The case presented in the boy is casuistic and requires further investigation.

Conflict of Interest: None declared

EP14.034 Partial trisomy 3q syndrome: a case report of siblings with intellectual disability and dysmorphic features

Derya Kan Karaer1, Kadri Karaer 1

1Pamukkale University Faculty of Medicine, Medical Genetic, Denizli

Background/Objectives: Duplications of the long arm of chromosome 3 are rare and are associated with a well-defined contiguous gene syndrome known as partial trisomy 3q syndrome. While the clinical presentation varies, common features include typical facial dysmorphism, hirsutism, microcephaly, growth retardation, intellectual disability, hand and feet abnormalities, genitourinary anomalies, renal, and congenital heart defects. Most cases result from unbalanced translocations inherited from a parent carrying a balanced reciprocal translocation or inversion, but de novo occurrence of the genomic abnormality is rare. Currently, nearly 100 patients with 3q duplication have been reported.

Methods: Two siblings, a 4-year-old boy and an 8-year-old girl, were referred to our center due to intellectual disability and dysmorphic features. The parents, who were consanguineous, were healthy. The children exhibited clinical findings including severe mental retardation, microcephaly, hirschsprung, distinctive facial features (including hirsutism, synophrys, gingival hypertrophy, bifid uvula, tented mouth), and seizures. Metaphase chromosomes of our patients and their parents obtained from peripheral blood sample cell culture and HRB cell culture were examined by staining with the GTG and HRB banding method.

Results: Evaluation of both patients revealed an unbalanced chromosome structure in all metaphase cells due to a balanced t(3;6)(q26.31;q27) translocation in the father.

Conclusion: This case highlights the importance of thorough genetic evaluation, particularly in cases of intellectual disability and dysmorphic features, and underscores the role of chromosomal analysis in diagnosing rare genetic syndromes. Further studies are warranted to explore the genotype-phenotype correlations and potential therapeutic interventions for individuals affected by partial trisomy 3q syndrome.

Conflict of Interest: None declared

EP14.035 Polymorphisms in HCN4 and PLCB2 associated with taste loss during the SARS-COV-2 infection

Belmina Šarić Medić 1, Naida Lojo-Kadrić1, Jasmin Ramić1, Nikolina Tomić1, Lejla Pojskić1

1University of Sarajevo - Institute for genetic engineering and biotechnology, Laboratory for human genetics, Sarajevo

Background/Objectives: Partial or complete loss of smell (anosmia/hyposmia) and taste (hypogeusia/ageusia), with or without distorted perception of smell and taste (dysosmia/dysgeusia), has a broad differential diagnosis. Reversibility of loss of sense of smell and taste is possible in cases of inflammation, and can be sampled by various drugs, disorders, and genetic factors.

Methods: Nucleic acid isolation from the collected samples of buccal mucosa swabs was done with Nucleospin genomic DNA isolation kit (Nucleospin, Mackerey-Nagel). Quantification of isolated DNA was performed using the fluorimetric method (Qubit quantification kits, Invitrogen). Sequencing is carried out on illumina MiniSeq with custom designed kit. Processing of raw sequencing data will be done using the Variant caller application which is available through Illumina’s website, and will identify variants that are different of the reference genome.

Results: We detected gene polymorphisms in all studied genes namely 971 variants in 68 genes. Preliminary results showed that certain polymorphisms in genes responsible for sour and bitter taste HCN4 c.2393C>G and PLCB2 c.3037-55T > C are statistically significantly associated with loss of taste (p = 0,035 and p = 0,008) respectively.

Conclusion: Loss of taste in COVID19 infection may be under influence of polymorphisms in genes associated with sour and bitter taste.

Grants: This study was sponsored by Ministry of science, higher education and youth, Canton Sarajevo, Bosnia and Herzegovina, no. 27-02-11-41250-40/21.

Conflict of Interest: Belmina Šarić Medić: None declared, Naida Lojo-Kadrić Principal investigator in this study that recieved goverment grant, Jasmin Ramić: None declared, Nikolina Tomić: None declared, Lejla Pojskić: None declared

EP14.036 A case report of a family with Xq26.2q27.1 duplication, further supporting the association of this CNV with short stature in males

Nuša Trošt 1, Luca Lovrecic1, Borut Peterlin1

1University medical centre Ljubljana, Clinical Institute of Genomic Medicine, Ljubljana, Slovenia

Background/Objectives: Short stature is a common reason for referral to further evaluation in children and it is often of genetic etiology. Although several genetic causes of growth delay disorders have been identified, the specific molecular cause in many children remains unknown. Here, we report 4 members of a family with 5,9 Mb interstitial duplication at Xq26.2q27.1 region, initially detected by prenatal microarray-testing due to fetal intrauterine growth retardation.

Methods: The proband, now 7 years old, is of short stature, receives growth hormone (GH) replacement therapy and is suspected of having ADHD. His development and cognitive abilities are otherwise within normal range. The same clinical signs and symptoms are present in his younger brother, who is also treated for GH deficiency. Additionally, the younger brother was diagnosed with speech delay, but the problem resolved itself by the age of four. The boys’ mother and grandmother are phenotypically normal. Array-CGH analysis was used for genetic evaluation.

Results: Xq26.3q27.1 duplication, covering 31 OMIM genes with distal breakpoint located just before SOX3 gene, was found in all family members. So far, only a few overlapping duplications at Xq26.2q27.1 region have been described and short stature was a common finding in male individuals. Other clinical features might include learning difficulties, developmental delay, hypopituitarism and dysmorphic signs, whereas women carrying these duplications were generally unaffected.

Conclusion: Reported Xq26.2q27.1 duplication further supports the association of this rare structural variant with X-linked recessive short stature. However, additional cases are needed to further delineate the minimal critical region (responsible genes) for growth delay.

Grants: /

Conflict of Interest: None declared

EP14.037 Optical genome mapping (OGM) allows the characterization of a complex chromosome rearrangement associated with recurrent pregnancy loss

Detlef Trost 1, Aline Receveur1, Laurence Lohmann1, Mylene Valduga1, Pascale Kleinfinger1, Aïcha Boughalem1

1Laboratoire CERBA, Departmet of Human Genetics, St Ouen l’Aumône, France

We report a 57-year-old male patient referred for karyotyping because of a history of recurrent pregnancy loss. Standard karyotyping revealed a complexly rearranged chromosome 2, present in about 50% of all analysed cells.

OGM analysis enabled the detailed characterization of this chromosome 2 rearrangement, with a total of 5 breakpoints, each break being associated to small deletions within chromosome 2. The generated chromosome 2 fragments have been reassembled to new positions, creation new fusion points. A segment originally located between chromosome bands 2p13.3 and 2p24.3 has been inserted to chromosome band 2q24.2, the inserted segment itself was found to be rearranged, with segments origination from 2p23.3-p23.2 and 2p14-p14 being inserted in inversed orientation.

One of newly created fusion points affects the gene DNAJC27, do date not associated to a human pathology, the remaining new fusion points did not affect known genes.

The five deleted segments located at the break points are between 0.9 and 2.2 Mb in size, these deletions harbour autosomal dominant genes that might contribute to the patient’s pathology: MYCN (164840), DNMT3A (602769), ASXL2(612991), ASPRV1 (608507).

OGM enabled a detailed characterization of this mosaic complex chromosome rearrangement and the genes affected, this information improved pertinent genetic counselling.

Conflict of Interest: None declared

EP14.039 Double cryptic chromosomal imbalance unravelled by cytogenetics and array CGH in a carrier woman with recurrent pregnancy loss

Eunice Matoso 1;2;3, Alexandra Estevinho2, Susana Isabel Ferreira4, Sara Ribeiro5, Daniela Couto6, Joana Barbosa de Melo3;4;7, Isabel Marques Carreira3;4;7, Jorge Saraiva5;8;9

1University of Coimbra, Faculty of Medicine, Portugal; 2Hospital Pediátrico, Unidade Local de Saúde de Coimbra, Cytogenetics Laboratory, Medical Genetics Unit, Coimbra, Portugal; 3iCBR- CIMAGO/CACC – Center of Investigation on Environment Genetics and Oncobiology of iCBR/Clinical Academic Center of Coimbra, Faculty of Medicine, University of Coimbra, Portugal; 4Cytogenetics and Genomics Laboratory, Faculty of Medicine, University of Coimbra, Portugal; 5Medical Genetics Unit, Hospital Pediátrico, Unidade Local de Saúde de Coimbra, Portugal; 6Serviço de Medicina da Reprodução, Unidade Local de Saúde de Coimbra, Portugal; 7CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal, Portugal; 8University Clinic of Pediatrics, Faculty of Medicine, University of Coimbra, Portugal; 9Clinical Academic Center of Coimbra, Portugal

Recurrent pregnancy loss (RPL) affects about 2%-3% of reproductive-aged women. The etiology of approximately 50% of RPL cases remains unknown. Carriers of structural and numerical chromosomal abnormalities are detected in about 3.74%-9.88% of RPL couples. Conventional cytogenetics is still the gold standard analysis. We report a 40-year-old woman with RPL, carrier of a double cryptic imbalance.

A couple were referred for Cytogenetic High-Resolution studies because of RPL. High-resolution cytogenetic analysis, Fluorescence in situ Hybridization (FISH) and array Comparative Genomic Hybridization (Agilent 180K) were performed. Parents of the carrier-woman were also evaluated.

Cytogenetic analysis and FISH revealed the woman to be the carrier of an imbalance Karyotype (ISCN 2020): 46,XX,der(7)add(7)(p22).ish der(7)del(7)(p22.3-)(VIJ2yRM2185-) and array result was arr[GRCh37]7p22.3(45130_930601)x1,18q23(74439776_78012829)x3.

Both parents revealed normal karyotypes. Clinical relevant information was disclosed after cytogenetic diagnosis: borderline intellectual disability, diplopia, mitral valve prolapse, and a long history of muscle weakness.

The present patient is a carrier of a deletion 7(p22.3pter) and a duplication 18(q23qter); of the 17 OMIM genes involved, just the PRKAR1B (in deletion) seemed to be of challenging interpretation; the haploinsufficiency of this gene is associated with a neurodevelopmental disorder of autosomal dominant inheritance. However, the present patient has no relevant syndrome characteristics (OMIM#619680), suggesting that Marbach-Schaaf neurodevelopmental syndrome could have alternative mechanisms in its etiology. Although this double chromosomal imbalance, disclosed by cytogenetics and array CGH, does not seem to be the cause of the patient’s RPL, undoubtedly represent essential additional information, although challenging to this couple’s genetic counseling and reproductive options.

Conflict of Interest: None declared

EP14.040 A systematic comparison of genotype-to-phenotype relationships in mouse and human

Yury Barbitoff 1, Nadezhda Pavlova1, Polina Bogaichuk1

1Institute of Bioinformatics Research and Education, Belgrade, Serbia

Background/Objectives: Reconstruction of the genotype-to-phenotype relationship network is one of the major goals of modern genetics and genomics. Development of ontologies for accurate and formal phenotype description enables a systematic comparison of these relationships between species.

Methods: We used Human Phenotype Ontology (HPO) and Mouse Genome Informatics (MGI) to retrieve information on gene-phenotype associations. Only genes with a single ortholog (n = 16,985) were used for comparison, and all phenotypes were converted to upper-level Mammalian Phenotype Ontology (MPO) terms. Data on human gene constraint (according to gnomAD v2) and expression pattern (based on GTEx v8) were used to annotate orthologous gene pairs.

Results: Only 24.6% (4,184) of all gene pairs were annotated with phenotype terms in both species. While the pleiotropic effects between the two species were well correlated for these genes (Spearman’s p = 0.28), we were surprised to discover that sets of terms were identical for as little as 15 genes. Of the remaining ones, as many as 385 genes were associated with non-overlapping sets of phenotypes in humans and mice. A large fraction of these discrepancies could be explained by the different structure of HPO and MPO. In many cases, however, phenotype terms displayed species-specific annotation patterns that correlated with the degree of evolutionary constraint and expression profile of a gene.

Conclusion: Our observations highlight important discrepancies between phenotype description in humans and mice, and raise concerns regarding the use of mouse phenotype information when interpreting the impact of human genome variants.

Conflict of Interest: None declared

EP14.041 Statistical dissection of the genetic determinants of phenotypic heterogeneity in genes with multiple associated rare diseases

Tatyana Lazareva 1, Yuriy Barbitoff1, Yulia Nasykhova1, Andrey Glotov1

1D.O. Ott Research Institute of Obstetrics, Gynaecology, and Reproductology, Dpt. of Genomic Medicine, St. Petersburg, Russian Federation

Background/Objectives: Phenotypic heterogeneity is a phenomenon in which individuals with pathogenic variants in the same gene have different phenotypes. This study aimed to uncover the gene-level factors contributing to this phenomenon in genes linked to multiple rare diseases (GMDs).

Methods: We used Human Phenotype Ontology as a source of information on gene-disease associations. Information about variant-disease associations was obtained from ClinVar. These data were used to compare variant type and localization within the coding sequence between diseases linked to a single GMD. Besides, gene-level properties (constraint, expression level) were assessed.

Results: GMDs were found to be more constrained and express transcripts across many tissues, suggesting their critical roles. Differences in the localization or type of causative variants between associated diseases were observed in 49 out of 670 GMDs. Interestingly, these factors seemed more influential for autosomal dominant disease genes. Further analysis revealed an impact of variant type on phenotype determination for 30 GMDs and variant localization for 38 GMDs. Remarkably, 19 GMDs linked to two distinct disorders displayed differences in both localization and type proportions of causal variants for associated diseases. Identified differences are useful for accurate variant annotation and prioritization.

Conclusion: The study highlights the importance of gene-level factors in understanding phenotypic heterogeneity for GMDs and emphasizes the need to explore other contributing mechanisms.

Grants: This work was supported by grant 075-15-2021-1058 from the Ministry of Science and Higher Education of Russia.

Conflict of Interest: None declared

EP14.042 Trisomy 9p in the 5-year-old boy with a complex of clinical signs as a result of the dicentric chromosome (22;9).

Valentyna Kurakova 1, Vira Galagan2, Olena Baronova1, Olga Miakushko1, Diana Mykhailova1, Maryna Tsygankova2, Olga Zhurakhovska2

1Children’s hospital “OKHMATDYT”, Laboratory of medical genetic, Kyiv, Ukraine; 2Children’s hospital "OKHMATDYT", The center of medical genetics, Kyiv, Ukraine

Background/Objectives: Trisomy 9p (ORPHA:236) refers to orphan pathology and characterized by intellectual disability, craniofacial dysmorphism, fingers abnormalities and short stature. Also, less frequently patients present with cardiopathy and renal, skeletal and central nervous system malformation.

Methods: Clinical, standard karyotyping, fluorescence in situ hybridization, Chromosome microarray (CMA) on the Afymetrix platform with Software Chromosome analysis Suite (ChAS) 4.3.0.71. using Optima CytoScan Array chip.

Results: Phenotype of the proband: downslanting palpebral fissures, short philtrum, high forehead, clinodactyly of the fifth finger, palmar crease of the left hand. Developmental delay (sits from 9 months, walks from 2 years, does not speak), muscle hypotonia.

Standard chromosomal analysis showed, that the proband had an additional material on the long arm of the chromosome 22, new mutation.

CMA showed a terminal duplication of the 9p24.3q21.12, the size is 69,1 Mb and contains 345 genes. According to the available laboratory evidence, it follows that the derivative chromosome 22 was formed as a result of a translocation between chromosomes 9 and 22 and consist of the entire chromosome 22 with an inverted part of chromosome 9 (the part of the long arm from the 9q21.11 breakpoint, the centromere and the entire short arm). Morphologically, the derivative chromosome is dicentric (22;9) with an active centromere of chromosome 22 and an inactivated centromere of chromosome 9.

Result: 46,XY,psu dic(22;9)(q13.3;q21.11),dn.arr[GRCh38]9p24.3q21.12 (649,006_69,783,599)x3.

Conclusion: Using alternatives methods of research at different levels of resolution is necessary to understand the mechanism of chromosomal rearrangements and determination of the causes of the disease.

Conflict of Interest: None declared

EP14.043 G-banding chromosomal analysis: retaining its necessity in genetic diagnosis pipelines

panayiotis myrianthopoulos1, andria ketoni1, charithea ioannidou1, Liza Kokkinou1, efthymia constantinou1, elena panayiotou1, nicole salameh1, ioannis papaevripidou1, angelos alexandrou1, paola evangelidou1, carolina sismani 1

1The Cyprus Institute of Neurology and Genetics, Cytogenetics and Genomics, Nicosia, Cyprus

Background/Objectives: High-throughput genomics in the form of array-CGH and NGS have revolutionized genetic testing, significantly improving the diagnostic yield and replacing chromosomal analysis as a first-tier test for patients with neurodevelopmental disorders (NDDs) and congenital malformations (CMs). Nevertheless, conventional karyotyping may still be needed, to overcome the limitations of these technologies. Here we present six cases displaying the necessity of chromosomal analysis for accurate diagnosis and recurrence risk estimation.

Methods: The referred cases are pregnancies with increased NT and patients with NDDs and CMs. Initial testing was performed by array-CGH (Agilent inc.) followed by G-banding chromosomal analysis. Findings were further validated with FISH and qRT-PCR.

Results: Array-CGH showed normal results in three cases and identified three with single pathogenic CNVs. Chromosomal analyses of the cases with normal results, revealed two mosaic ring chromosomes and one mosaic isochromosome [45,X/46,XX,r(18)(p11.32q23)dn/46,XX,46,XX,r(20)(p13q13.33)/46,XX and 47,XX,+i(22)(p10)dn/46,XX]. Chromosomal analysis of the cases with pathogenic CNVs revealed more complex structural rearrangements: one paternally inherited unbalanced translocation [46,XY,der(14)t(14;17)(p11.2;q25.1)dpat.arr 17q25.1q25.3(73608244_81044553)x3], a complex chromosomal rearrangement [46,XX,t(2;3)(q22;q13.2)dn,inv(8)p23.1q24.1dn.arr 8q24.11(118813668_118816960)x1] and a de novo insertion [46,XY,ins(12;7)(p13.1;q31.3q22)dn.arr 1q44(244596638_245665521)x1].

Conclusion: Our findings demonstrate that, despite the progress in genomics, karyotype analysis is still vital for accurate diagnosis and recurrence risk estimation, revealing additional structural rearrangements even in cases where the pathogenic cause was identified by array-CGH. In addition, the presence of abnormalities such as ring chromosomes and isochromosomes would be missed by array-CGH. The introduction of third-generation sequencing and optical genome mapping may offer the advantages of both methodologies in a single tool.

Conflict of Interest: None declared

EP14.044 Optical Genome Mapping for comprehensive genomic rearrangement analysis

Julia Flunkert 1, Rainer Wimmer1, Markus Stumm1

1Medicover Humangenetik Berlin-Lichtenberg MVZ, Berlin

Background/Objectives: Structural variant detection is primarily reliant on conventional cytogenetic methods such as chromosome analysis, Fluorescence in situ hybridization, or microarrays. However, these approaches exhibit limitations in offering a comprehensive understanding of genomic rearrangements. Optical Genome Mapping (OGM) has emerged as a promising genome-wide technology for detecting structural variants and copy number variants, proving effective in defining and resolving complex genomic rearrangements.

Methods: In this study, Bionano OGM was employed to further characterize breakpoints and the structure of genomic rearrangements (deletions, duplications, inversions, translocations, insertions) previously identified by standard cytogenetic methods in 45 patients.

Results: OGM reached concordance in 91% (41/45 cases) compared to standard tests for various types of structural variants. Moreover, OGM yielded additional relevant information in 11 patients. This included the orientation of structural variants, especially duplications, identification of disease-causing gene disruptions, and the resolution of the rearrangement structure of complex aberrations resulting from chromothripsis. OGM was unable to confirm the original results in 9% of cases due to technical limitations. These involved aberration breakpoints located in genomic regions known to be challenging for OGM detection. Therefore, it can be concluded that the actual concordance rate is likely higher.

Conclusion: This study underscores the potential of OGM as a valuable tool for comprehensive genomic rearrangement analysis and highlights the significance of integrating OGM with established techniques for a more thorough understanding of structural variants.

Grants: None

Conflict of Interest: None declared

EP14.045 Insights Genetic Complexity: Integrated NGS and SNP Analysis in a Case of Hypogonadotropic Hypogonadism with Structural Anomalies.

Roberta Petillo1, Massimiliano Chetta2, Marina Tarsitano2, Serena Torre2, Elvira Sannino2, Marcella D’Antonio2, Maria Oro2, Maria Rivieccio2, Maria Cioce2, MariaRita Parisi2, Davide De Brasi3, Daniele De Brasi4, Manuela Priolo 2

1AORN Cardarelli Hospital, Medical genetics and genomics, Napoli, Italy; 2AORN Cardarelli Hospital, Medical genetics and genomics, napoli, Italy; 3AORN Cardarelli Hospital, Endocrinologia, napoli, Italy; 4Santobono Pausilipon, Medical genetics, napoli, Italy

Background/Objectives: This study focuses on a 16-year-old girl who has anatomical abnormalities such a filiform uterus and hypoplastic ovaries, as well as primary amenorrhea and hypergonadotropic hypogonadism. The patient had growth hormone (GH) treatment since age 8 for low height, although having no family history of genetic abnormalities and uncomplicated pre- and post-natal events. Brain MRI revealed a functionally hyperplastic pituitary gland. Upon physical examination, anomalies such as microcephaly, nasal voice, low body weight, and unique facial traits were discovered. Initial genetic testing, such as the FISH for the SRY locus, the NGS panel for genes associated to hypogonadism, the mosaic karyotype, and the Fragile-X analysis, produced negative findings.

Methods: SNP array analysis identified a 17% loss of heterozygosity (LOH) across the genome and a deletion of the GLI2 gene at 2q14.2. Subsequently, potential homozygote variants in the LOH region were found using Clinical Exome Sequencing (CES).

Results: The comprehensive study highlighted the necessity of integrating NGS and SNP array technologies and elucidated the genetic basis of hypogonadotropic hypogonadism. A likely pathogenic variant (p.Arg143Serfs*5) in the PSMC3IP gene was discovered in the LOH region.

Conclusions: The work highlights how important it is to integrate NGS sequencing with SNP array technologies. This comprehensive strategy increases the precision of diagnosis and broadens our understanding of the genetic origins of this complicated disease, offering crucial new insights for possible therapeutic strategies.

Conflict of Interest: None declared

EP14.047 Beyond absolute size: dynamic ROH thresholds tailored by total ROH percentage

Idit Maya 1, Michal Levy1, Lina Basel-Salmon1, Lena Sagi-Dain2

1Rabin Medical Center, Petah Tikva, Israel; 2Carmel Medical Centre, Haifa, Israel

Background/Objectives: The main clinical purpose of reporting Regions of Homozygosity (ROH), is to estimate the likelihood of autosomal recessive disorders and uniparental disomy (UPD). In populations with increased prevalence of consanguinity, in which ROH findings are prevalent, there is a potential for superfluous reporting, notwithstanding the negligible risk for UPD. We aimed to investigate the correlation between the size of ROH segments and the Total Sample ROH Percentage (TSRP), and to propose cutoffs for ROH disclosure.

Methods: All chromosomal SNP arrays conducted from 2017 to 2023 were collected retrospectively. We analyzed the correlation between ROH segment size and TRSP, both per chromosome (imprinted vs. non-imprinted) and segment type (terminal vs. interstitial). We calculated the 97.5th and 99th percentile cutoffs for ROH size within each TRSP category, examining these as thresholds for ROH reporting.

Results: 66,711 ROH segments from 13,035 CMA samples were analyzed. We proposed dynamic reporting thresholds based on the 99th percentile for ROH segment size within each TRSP category, effectively identifying 28 cases with previously confirmed UPD. In TRSP above 2%, the 99th percentile cutoffs exceeded 20Mb, resulting in a lower percentage of reported samples compared to previously suggested thresholds.

Conclusion: Our study pioneers a comprehensive exploration into the complex relationship between ROH segments size and TRSP. The introduction of dynamic thresholds for reporting refines the criteria, challenging previous rigid approaches based on absolute size thresholds. This research lays a solid foundation for future investigations into the intricate genomic landscape of diverse populations.

Grants: None

Conflict of Interest: None declared

EP15 Diagnostic Improvements and Quality Control

EP15.001 Molecular analysis of lactose persistence by melting curve assays

Antoine Stuitje 1, Zewdu Terefework2, Chris Hettinga1, Robert C. Akkers3, Rob Castel3

1MRC Holland, Research, Amsterdam, Netherlands; 2MRC-ET Advanced Laboratory, Research, Addis Ababa, Ethiopia; 3Result Laboratorium, Molecular diagnostics, Zevenbergen, Netherlands

Background/Objectives: Lactase non-persistence (LNP), is a universal ancestral mammalian genetic trait, that the lactase gene (LCT) is no longer expressed in the small intestine after weaning. In the current human population about 20% have lost LNP due to relatively recent single mutations, resulting in adult lactase persistence (LP). The most common LP variant (13910C > T) is found in about 60% of the Caucasian population. Eight other mutations have, however, also been reported particularly amongst African and Arab populations. Until now, identification of nine mutations in a single melting curve assay was challenging due to clustering and the limited number of fluorophores available. This issue has been solved with SALSA® MC001-X1 Lactase Persistence Mix.

Methods: This melt assay requires two separate reactions. In each of these reactions, two different melting curve profiles are generated using TEX615 and Cy5 fluorescent probes. Reaction A detects the Caucasian frequent -13910-T and rare -13914-A, -13913-C, -13909-A or -14011-T variants, as well as the East African -13907-G variant. Reaction B is used to detect the Saudi -13915-G variant and the East African -14009-G and -14010-C variants.

Results: MC001-X1 Lactase Persistence Mix was assessed using eighteen control plasmids harbouring heterozygous or homozygous LP variants. Rare variants (such as the Ethiopian, Sudan, Arab and Kenian variants) were also detected with this assay, in the Ethiopian population and the ECACC ethnic diversity panel test. Results were confirmed by sequencing.

Conclusion: MC001-X1 Lactase Persistence Mix is a simple assay to determine which of the LP mutations are present in homozygous or heterozygous configuration.

Grants: None

Conflict of Interest: Antoine Stuitje MRC Holland, Zewdu Terefework: None declared, Chris Hettinga MRC Holland, Robert C. Akkers: None declared, Rob Castel: None declared

EP15.002 A network approach to identify, diagnose, and resolve mislabels using sample genotypes and metadata

Charles Deng 1, Ryan Thompson1, Laura Sloofman1, Alexander Charney1, Noam Beckmann1

11 Gustave L. Levy Pl, New York, United States

Consortium: The Mount Sinai COVID-19 Biobank Team

Background/Objectives: Mislabeled data can result in diminished statistical power and, more critically, introduce systematic biases that affect the accuracy of results. We present a scalable network-based approach that identifies and resolves mislabels in any dataset containing 1) multiple samples per individual and 2) sufficient genetic information to genotype samples.

Methods: A label network and a genotype network, linking 1) samples with the same subject label and 2) samples with the same genotype, are constructed. An ensemble of custom search algorithms then proposes mislabel corrections by resolving discrepancies between these two networks. We evaluated algorithm performance on simulated mislabeled datasets with up to 10,000 subjects, 20 samples per subject, and 50% mislabeling rate. We validated our algorithm on the Mount Sinai COVID-19 Biobank, which spans 3756 RNAseq, WGS, and genotype array samples across 696 individuals. The algorithm’s scalability was tested on 17350 RNAseq samples across 948 individuals from GTEx.

Results: In simulated datasets with up to 40% mislabeling, the algorithm recovered correct labels with near-perfect accuracy (average 97%). On the COVID-19 Biobank, out of 190 mislabels identified by the algorithm, 186 (98%) suggested corrections matched those previously determined by manual review, 3 (1.6%) were previously unidentified, and 1 (0.5%) was determined to be incorrect. On the GTEx dataset, the algorithm reported no discrepancies between the label and genotype networks, suggesting an absence of mislabeling.

Conclusion: Our network-based approach to correcting mislabels proves to be efficient and accurate, both across simulations and on large-scale genomic datasets.

Conflict of Interest: None declared

EP15.003 DNA-microarray as an alternative diagnostic tool for targeted genetic carrier screening in population of Yakut ethnic group

Mira Savvina 1, Nadezhda Maksimova1, Aitalina Sukhomyasova1

1North-Eastern Federal unversity, Medical Institute, Yakutsk

Background/Objectives: There are populations that show less genetic variability – genetic isolates. Yakut ethnic group in Siberia demonstrate a high prevalence of rare autosomal recessive disorders due to the founder effect. We evaluated the heterozygous carrier frequencies of five monogenic disorders enriched in the population among healthy individuals using own developed low density DNA-microarray and general methods.

Methods: Oligonucleotide probes addressing point mutations 4582_4583insT in CUL7, с.5741G > A in NBAS, с.806С>Т in DIA1 c.1090G>C in FAH., c.-23 + 1G > A in GJB2 causing 3-M syndrome, SOPH-syndrome, Methaemoglobinaemia type 1, Tyrosinemia type 1, DFNB1 type 1A respectively and wild type probes were printed unto epoxy coated glass slide by non-contact manner. The assay using developed chip which include two step multiplex PCR with production of one-stranded Cy5 labeled-PCR products following its hybridization with oligonucleotide probes.

Results: We developed population specific low-density microarray and performed genotyping of 120 healthy individuals for major mutations diseases listed above. Additionally, 520 of DNA from healthy individuals were screened for same mutations real-time PCR. The results of demonstrate a high frequency of all five genetic disorders tested and were: 3-M syndrome -1:25, SOPH-syndrome- 1:50, Methaemoglobinaemia type 1-1:25, DFNB1- 1:11, Tyrosinemia type 1-1:100

Conclusion: Obtained results of high level of carrier frequencies of above disorders indicates a high need of its prevention through implementation of carrier screening programs in population. The developed oligonucleotide microarray can be considered as a low-cost tool for heterozygous carrier screening in Yakut ethnic group.

Grants: The research is conducted under the state target program: project FSRG-2024-0001 “Genomic of Arctic: diagnostic, prophylactics and therapy”

Conflict of Interest: None declared

EP15.004 Coding undiagnosed rare disease patients: RD-CODE project recommendations to make them count

Céline Angin1, Monica Mazzucato2, Stefanie Weber3, Kurt Kirch3, Houda Ali4, Sylvie Maiella 4, ANA RATH4

1Greater Paris University Hospitals (AP-HP), French National Rare Disease Registry (BNDMR), Paris, France; 2Padua University Hospital, RD Coordinating Centre, Veneto Region; 3BfArM, Germany; 4Inserm, US14-Orphanet, France

Background/Objectives: Extensive efforts to support the implementation of ORPHA nomenclature in health information systems (HIS) (electronic health records and/or registries) improved rare disease (RD) coding worldwide. However, until recently, persons suffering from a suspected RD who could not be diagnosed even after full investigation (i.e. diagnostic dead-end) could not be coded with ORPHAcodes. The RD-CODE project aimed at making those RD undiagnosed patients identifiable in HIS to produce crucial epidemiological data.

Methods: Undiagnosed patients were defined as patients for whom no clinically-known disorder could be confirmed by an expert center after all reasonable efforts to obtain a diagnosis according to the state-of-the-art and diagnostic capabilities available. The RD-CODE multi-stakeholder panel of experts produced three recommendations for the coding of undiagnosed RD patients.

Results: The recommendations are: 1/ Capture the diagnostic ascertainment for all rare disease cases; 2/ Use the newly created ORPHAcode (ORPHA:616874 “Rare disorder without a determined diagnosis after full investigation”) available in the Orphanet nomenclature, together with the guidelines to ensure its correct and homogeneous use; 3/ Use additional descriptors in registries.

Conclusion: The RD-CODE recommendations have started to be implemented in HIS and they could be a game-changer for patients, clinicians and researchers in the field. They enable assessment of the global RD population, adaptation of policy measures -including financing for care and research programs- and improved access to research programs.

Grants: European Union’s Third Health Programme

Conflict of Interest: None declared

EP15.005 Novel complementary molecular diagnostics pipeline assay development for Fabry disease

Árpád Ferenc Kovács 1, Nóra Fekete2, Dalma Ruzsics1, Luca Kamilla Li1, Sándor Dávid Kovács1, Júlia Krisztina Erhardt1, Gergely Tibor Kozma3, Éva Pállinger2, Gyorgy Fekete1

1Semmelweis University, Pediatrics Center Tűzoltó Street Department, Budapest; 2Semmelweis University, Genetics, Cell and Immunobiology, Budapest, Hungary; 3Semmelweis University, Nanomedicine Research and Education Center, Department of Translational Medicine, Budapest, Hungary

Background: Fabry disease is the most common lysosomal storage disorder, with an estimated prevalence of 1:2000-7500. In heterozygous women the diagnostic pathway is paved with difficulties, as symptomatology and organ involvement varies on a large scale. Currently used AGAL enzymatic assay often fails to uncover the dysfunction, and at gene level intronic and structural variants are missed by next-generation sequencing. The aim of the study was the development of a new assay for measuring AGAL activity in single-cells and the development of targeted nanopore based GLA sequencing.

Methods: Conventional and multispectral imaging flow cytometry was used to detect AGAL activity at single cell level. Validation was performed via CRISPR-Cas9 mediated transfection of a GLA knockout (KO) Jurkat cell line and AGAL inhibitor. GLA KO cell line was validated using exome sequencing, gene expression and substrate accumulation. Adaptive sequencing approach was used to analyse GLA gene.

Results: Using 2.83 nmol fluorescent AGAL substrate and a 6 hour incubation period AGAL activity can be detected in individual cells. 20% trypan blue solution quenching allows the specific detection of substrate accumulation, via elimination of aspecific exofacial binding of the substrate. Inputting 150 fmol of HMW DNA for long read sequencing enables the detection of pathogenic variants.

Conclusions: At protein level we have developed a flow cytometry based assay to detect AGAL activity at single cell resolution and at DNA level a GLA targeted approach to detect both nucleotide and structural variants.

Grants: Hungarian National Research, Development and Innovation Office – NKFIH, OTKA PD_21 138521.

Conflict of Interest: Árpád Ferenc Kovács speaking honoraria from Takeda Pharma, travel grants from Sanofi Genzyme and Takeda Pharma, participation of advisory board honoraria from Sanofi Genzyme, Nóra Fekete: None declared, Dalma Ruzsics: None declared, Luca Kamilla Li: None declared, Sándor Dávid Kovács: None declared, Júlia Krisztina Erhardt: None declared, Gergely Tibor Kozma: None declared, Éva Pállinger: None declared, Gyorgy Fekete speaking honoraria from Takeda Pharma, advisory board honoraria from Sanofi Genzyme

EP15.006 A systematic evaluation framework for genome coverage, offering tools for data quality control along with guidance for clinical research.

gong meihua1, yi heng 1, rao junhua1, Xu chongjun1, Deng Ziqing1, Li Jiguang1, Wang Jingjing1, Ma Kexin1, Wang yicong1, Chen Fang1

1MGI Tech, shenzhen, China

Background/Objectives:Currently, Global Alliance for Genomics and Health published the robust evaluation benchmark for germline small variant in human Whole Genome Sequencing (WGS), yet there is a deficiency in a comprehensive evaluation for genomic data coverage, particularly in complex and challenging regions. High coverage in these regions is crucial for clinical disease research. This study has established a framework for assessing the coverage performance.

Methods:The PCR-free WGS PE150 data of NA12878 were evaluated across main sequencing platforms: DNBSEQ-T7, NovaSeq 6000, NovaSeq X, and AVITI™, using 30X downsampled data. The evaluation concentrated on genome stratifications from National Institute of Standards and Technology, challenging medically relevant and other clinical relevant genes. Numerical metrics, including the length of low coverage and its genomic positioning, were utilized to produce evaluation reports.

Results:There were significant differences in the distribution and uniformity of low coverage across different sequencing platforms. The length of low coverage regions(≤5X) was much better between DNBSEQ-T7 and NovaSeq X (approximately 110k) compared to other platforms. AVITI™ with 1000bp long insert size demonstrated higher coverage in regions with GC content lower 15%, while NovaSeq 6000 achieved higher coverage in regions with GC content above 85%. All platforms exhibited low coverage (≤20X) for challenging genes such as SMN1 and HBG1.

Conclusion:This study aims to establish a systematic evaluation framework for genome coverage, detailing the performance of important regions across different sequencing platforms, visualizing the positions of low coverage region, offering tools for data quality control along with guidance for clinical research.

Conflict of Interest: None declared

EP15.007 Re-analysis of polyalanine expansions in the FOXL2 gene in whole exome sequencing data

Theresa Sujatta 1, Denny Popp1, Henry Oppermann1, Julia Hentschel1, Helene Faust1

1Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany

Background/Objectives: The detection of polyalanine expansions poses significant challenges in exome data due to high GC-content and complex motif (GCN, imperfect repeats). We asked ourselves under which conditions polyalanine expansions may be detected in exome data.

Methods: We investigated polyalanine expansions in the gene FOXL2 in 4,609 exome analyses based on TwistCore enrichment kits and 2,419 exome analyses based on the Twist2.0 enrichment kit using ExpansionHunter. Repeat expansions of pathogenic length in FOXL2 were evaluated regarding phenotype overlap, coverage, confidence interval of the ExpansionHunter calculation and quality in IGV. Clinical relevant repeat expansions were confirmed using Sanger sequencing.

Results: 261 TwistCore analyses and two Twist2.0 analyses with potential pathogenic repeat expansions in FOXL2 were identified. Of these, a repeat expansion length with assumed narrow confidence interval (+/-3 repeat counts) was determined in 29 cases and in two cases, respectively. One of the two cases of the Twist2.0 cohort had the typical 24 polyalanine repeat expansion in FOXL2, a confidence interval of +/-0 and a complete phenotype overlap. The inspection in IGV revealed high quality, the coverage was high and the repeat count was confirmed with Sanger sequencing. In contrast, we determined in all 29 cases of the TwistCore analyses and the other case of the Twist2.0 analyses a low quality of the loci in IGV, including cases with high coverage.

Conclusion: We demonstrated that given adequate sequencing quality, ExpansionHunter is capable of accurately determining polyalanine expansion length in FOXL2. Moreover, we observed that choice of enrichment kit may influence detection of polyalanine repeats.

Grants: None.

Conflict of Interest: None declared

EP15.009 A comparative study of methods for adapter dimer detection

Eva Graf 1, Whitney Pike2

1Agilent, Waldbronn, Germany; 2Agilent, Ankeny, United States

Background/Objectives: Adapter dimers are common sources of issues in NGS workflows. They occur when adapters ligate to each other without an insert between them, resulting in short fragments that interfere with library preparation and sequencing. These smaller fragments are preferentially sequenced, so even low levels of adapter dimers can lead to reduced output, lower diversity, and decreased genome coverage. Many sequencing platforms provide guidance for the maximum amount of adapter dimers. Here, methods for detection of adapter dimers are systematically compared to provide guidance for optimization of NGS libraries.

Methods: Different electrophoresis systems were used to measure the amount of adapter dimers present in both DNA and RNA NGS libraries. To create a standardized sample set, purified libraries were spiked with suitable fragments to simulate adapter dimers. In addition, libraries with existing dimer contamination were spiked with fragments of various sizes to evaluate the resolution capabilities of the analysis methods used.

Results: The methods tested provided high sensitivity for detecting adapter dimers. In most cases, the adapter dimers were well resolved from the libraries and other dimers. The results are discussed in the light of tolerance thresholds for adapter dimers as provided by library preparation protocols.

Conclusions: The selected electrophoresis systems are appropriate tools to detect even minute amounts of adapter dimers. The results of this study offer useful insights in selecting the most appropriate methods to assess library quality according to the sequencing platforms recommendations. This allows for informed decisions about the subsequent steps in the sequencing workflow.

Conflict of Interest: Eva Graf Agilent, Agilent, Whitney Pike Agilent

EP15.010 Locus-specific non-primer-mediated allelic dropout: possible mechanism and impact on DNA diagnostics yield

Anna Shestak 1, Victoria Rumyantseva1, Elena Zaklyazminskaya1

1Petrovsky National Research Center of Surgery, Medical Genetics Laboratory, Moscow, Russian Federation

Background/Objectives: All PCR-based sequencing methods have a risk of allelic dropout (ADO) phenomenon that leads to selective allele amplification during PCR process and may reduce the diagnostic yield of genetic testing. Here we demonstrate cases of non-primer-mediated allelic dropout and a possible mechanism leading to this type of ADO.

Methods: DNA diagnostics using Sanger sequencing, NGS (AmpliSeq target genes panels; WES) was performed for 250 families with hereditary cardio-vascular disorders. To reveal cases of ADO we analyzed the primer binding sites and amplicon sequences using gnomAD. All amplicons with suspected ADO cases were re-sequenced using alternative oligoprimer(s) pair(s).

Results: We have identified 8 cases of ADO in both targeted genes panels (NGS) and Sanger sequencing results. Most of revealed ADO cases (6 cases, 75%) were caused by common or rare SNVs at the annealing sites of oligoprimers. Moreover, two ADO cases (25%) in ENG and KCNQ1 genes were mediated by the presence of six- or more nucleotides indels in the amplicons studied, leading to false-positive or false-negative diagnostic results. Dependence on allele length may be the underlying mechanism of this type of ADO.

Conclusion: We have demonstrated the impact of "point" substitutions in primer-binding sites and indels in amplicons on the amplification efficiency. SNVs check at primer-binding sites per se during primer selection is not sufficient to avoid ADO. Non-primer-mediated ADO makes a significant contribution to the loss of genetic data in DNA diagnostics.

Grants: This work was supported by Research project FURG-2023-0008.

Conflict of Interest: Anna Shestak: None declared, Victoria Rumyantseva Research project FURG-2023-0008, collaborator, Elena Zaklyazminskaya Research project FURG-2023-0008, principal investigator

EP15.011 Optimizing a protein extraction protocol and deparaffinization method for fresh and fixed oral mucosa samples

Alvaro Arthur Soares1, Franklyn Emanuell Gomes dos Santos1, Josiel Santos do Nascimento1, Antonio Thomas da Silva1, Pollyanna Almeida dos Santos Abu Hana 2, Carlos Arthur Cardoso Almeida1

1UFAL - Campus A. C. Simões, Pharmacy Institute, Maceio, Brazil; 2University Of Health Sciences of Alagoas, Maceio, Brazil

Background/Objectives: Oral Mucosa (OM) Biopsies are the gold standard for diagnosing oral cancer. Samples preserved in formalin undergo changes that make difficult its analysis. In this study it was optimized a protein extraction protocol for fresh and fixed OM samples.

Methods: Fixed and fresh OM fragments and three protocols were modified. For fresh samples: P1 (TritonX-100 at 100μL/10 mg of tissue - standard for all protocols) with crushing and complete homogenization. Extract was centrifuged at 5000RPM/30min at 4°C; P2: 100mM DTT, 4%SDS; P3: 200mM DTT, 2%SDS) with centrifugation time of 20 and 10min, respectively. Buffers were added to the samples and crushed until homogenization, extracts were incubated for 20min at 100°C and centrifuged at 4ºC right after. Comassie Brilliant Blue R-250 and G-250-based dyes were compared. The best extraction and staining protocol were tested with deparaffinization methods. For fixed samples, M1: three xylene incubations/05min and four serial ethanol rehydration/1min followed by incubation in distilled water for 30min; M2: shaking and centrifugation for 3min after each incubation with xylene and serial rehydration in ethanol (three/5min). Concentration was evaluated by microplate reading.

Results: P3 combined with M2 with an extra xylene incubation for 03min and stained with Comassie R-250 showed to be more effective for SDS-PAGE analysis.

Conclusion: The combined modified P3 + M2 protocols proved to be an effective, low cost method for the extraction and analysis of proteins in OM samples through SDS-PAGE.

Grants: FAPEAL/CNPq

Project Aproved by the UFAL Ethic Committee: 65956516.1.1001.5013

Conflict of Interest: None declared

EP15.012 Utilizing Short Read Whole-Genome Sequencing for Structural Variant Identification in PKHD1

Nadav Agam 1;2, ruth schreiber2;3, Ohad Wormser1;2, ofek freund1;2, Matan M. Jean1;2, Amit Safran1;2, Tomer Poleg1;2, Ohad Shmuel Birk1;2;4

1Ben Gurion University of the Negev, National Institute of Biotechnology in the Negev, The Morris Kahn Laboratory of Human Genetics, Be’er Sheva, Israel; 2Ben Gurion University of the Negev, Faculty of Health Sciences, Be’er Sheva, Israel; 3Soroka Medical Center, Pediatric Nephrology Clinic, Be’er Sheva, Israel; 4Soroka Medical Center, Genetics Institute, Be’er Sheva, Israel

Background/Objectives: A well-established Mendelian disease, autosomal recessive polycystic kidney disease (ARPKD), is caused by biallelic aberrations in PKHD1. We present a cluster of families of Bedouin descent, in which no causal variants were found in whole-exome sequencing (WES). Whole-genome sequencing (WGS) did not identify point mutations but enabled the unraveling of a pericentric chromosomal rearrangement as the causal variant in the queried cohort.

Methods: Two affected individuals, homozygous and compound-heterozygous for the PKHD1 locus, underwent WGS. Data was analyzed using Franklin variant analysis tool and Manta structural variants identification software. Polymerase chain reaction-based tests, amplifying only intact or altered sequences, were carried out for all family members.

Results: Analysis for point mutations of WES in clinical settings and of WGS yielded no candidates. Manta analysis of WGS data identified a chromosomal breakpoint in intron 65 of the 66 PKHD1 introns, essentially eliminating the last two exons. All disrupted reads were also complementary to a region ~21M bases upstream, in which another break was found. All family members were classified as wildtype, heterozygous or homozygote and the co-segregation of the variant with the disease was verified.

Discussion: This is the first documentation (to our knowledge) of such a structural variant for ARPKD. As there are several reports of stop codon pathogenic mutations in the last two exons of PKHD1, we conclude that this variant is pathogenic. This report emphasizes the potential of implementing WGS in clinical settings, enabling elucidation of previously undetected causative structural aberrations.

Grants: the Morris Kahn Family Foundation

Conflict of Interest: None declared

EP15.013 Mapping the Swedish methylome using ONT promethION long-read genome sequencing

Jesper Eisfeldt 1;2, Kristine Bilgrav Saether1, Esmee ten Berk de Boer1;2, Marlene Ek1;2, Anna Lindstrand1;2

1karolinska institutet, Department of molecular medicine and surgery, Sweden; 2Karolinska University hospital, Department of Clinical Genetics, Sweden

BACKGROUND: Long-read genome sequencing (lrGS) enables analyses, such as structural variant detection, phasing, and methylomics in one single experiment. The methylomic analysis is of great importance and may be used to assess the impact of genetic variation directly. In contrast to previously used technologies, lrGS combines high resolution and unbiased readout alongside the phasing of CpG sites and genetic variation.

METHODS: Here we use lrGS to study the whole-blood methylome of 40 unrelated Swedish individuals. The cohort was drafted from the department of clinical genetics, Karolinska University hospital. On average, we obtain 35X median coverage, read-length N50 of 10 kbp, and error-rate of 3%. Next, we perform a diversity of epigenetic analyses, including allele-specific methylation, aberrant promoter usage, and overlay with public RNA-seq datasets.

RESULTS: Correlating the promoter methylation level with GTEX expression levels, we find that genes not expressed in blood to be heavily methylated, while genes highly expressed are unlikely to be methylated, indicating that the lrGS methylome may be used as a proxy for gene expression.

Next, we use the allele specific methylation analysis to search for novel imprinted genes, providing support for genes predicted to be imprinted by geneimprint.

Lastly, we present an algorithm for the detection of aberrant methylation patterns, which may be used for detection of aberrant pathways, as well as a quality control. In summary, we explore the methylome in our unique dataset, and show that lrGS provides valuable genetic insights and may help in the diagnosis of rare disease patients.

Grants: None

Conflict of Interest: Jesper Eisfeldt: None declared, Kristine Bilgrav Saether: None declared, Esmee ten Berk de Boer: None declared, Marlene Ek: None declared, Anna Lindstrand Honoraria from ONT

EP15.014 Improving Genetic Testing for Hereditary Cancers: A Multi-Site Assessment Using a Multiplexed Reference Material

benedicta forson1, Melissa Berenger1, Krystyna Nahlik 1, Dana Ruminski Lowe1, Yves Konigshofer1, Dianren Xia1, Edward Davis1, Catherine Huang1

1LGC Clinical Diagnostics, Gaithersburg, United States

Background/Objectives: Inherited cancers are caused by mutations in genes passed down from one generation to the next which increase the risk to developing cancers such as Lynch syndrome, Hereditary Breast and Ovarian Cancer and others. As about 5-10% of cancers are linked to an inherited pathogenic variant, comprehensive genetic testing is important to enable clinicians implement surveillance and risk-reduction strategies, detect cancer at an early, more treatable stage, and provide tailored treatment and therapies.

Inherited cancer assays are developing rapidly with some panels screening for mutations in hundreds of genes. To perform thorough validation and assess assay performance, it is usually necessary to source numerous samples spanning mutations in each gene of interest which is difficult and costly.

Methods: We designed a highly multiplexed reference material containing 61 pathogenic variants in 55 cancer-predisposing genes. Biosynthetic DNA constructs bearing the variants were blended with genomic DNA from the GM24385 cell line at a 50% allele frequency (VAF), analyzed by digital PCR and orthogonally tested by several NGS assays.

Results: Digital PCR confirmed the presence of all variants at intended VAFs. The NGS data from multiple evaluation sites helped benchmark their performance in detecting common, rare, and challenging variants.

Conclusion: Inherited cancer genetic testing is an essential component of personalized medicine which can improve cancer prevention, detection, and treatment strategies. This study shows the benefit in using Seraseq Inherited Cancer Mutation Mix v2, a highly multiplexed reference material to support assay development; assess, optimize, and routinely monitor assay performance or implement external quality assessments.

Conflict of Interest: benedicta forson LGC Clinical Diagnostics, Melissa Berenger LGC Clinical Diagnostics, Krystyna Nahlik LGC Clinical Diagnostics, Dana Ruminski Lowe LGC Clinical Diagnostics, Yves Konigshofer LGC Clinical Diagnostics, Dianren Xia LGC Clinical Diagnostics, Edward Davis LGC Clinical Diagnostics, Catherine Huang LGC Clinical Diagnostics

EP15.015 Transforming PCR protocols in pandemic response with automated non-contact liquid dispensing

Frances Pitt1, Suhayl Mulla1, Denise Grovewood 2, Russell Buckley-Taylor2, Suki Lee3, Giorgio Fedele4, Graham Hill5, Paul Harper6, Donald Fraser7, Tamsin Redgwell2

1UKHSA, London, United Kingdom; 2SPT Labtech, Melbourn, United Kingdom; 3Utrecht University; 4University of Calabria; 5Animal and Plant Health Agency; 6AstraZeneca; 7Omix

Background/Objectives: During the COVID-19 pandemic, effective Polymerase Chain Reaction (PCR) protocols were crucial for detecting SARS-CoV-2 in clinical samples, playing a key role in rapid infection control and easing the strain on healthcare services. However, with the critical need for fast and accurate pathogen surveillance, the manual execution of PCR workflows presented numerous challenges, including limited throughput, risk of human error and variability, and risk to human health.

Methods: The analytical sensitivity for the UltraDx SARS-CoV-2 N1/N2/RP assay was evaluated by the RFL development team using RNA extracted from a panel of virus inputs from a Qnostics Analytical Q Panel, Product Number: SCV2AQP. Samples were extracted using a modified RNA extraction procedure for Thermo MagMax reagents and consumables with the dragonfly® discovery automated reagent dispenser.

Results: Using the same/equivalent reagents and control materials, the inclusion of dragonfly discovery showed a limit of detection (LoD) of 70 copies/mL (.9 copies/reaction) sensitivity, in comparison to the LoD of the manual method suggested by the manufacturer (80 copies/reaction).

Conclusion: Incorporating dragonfly discovery into the UltraDx SARS-CoV-2 N1/N2/RP assay resulted in a 80 –fold decreased limit of detection for the N1 assay compared to the recommendation from the reagent manufacturer. This could be attributed to the enhanced accuracy and precision facilitated by dragonfly discovery’s non-contact positive displacement dispensing, in combination with the Auto-Feed Reservoir (AFR) pump mechanism ensuring a continuous suspension of magnetic beads.

Grants: N/A

Conflict of Interest: None declared

EP15.016 The diagnostic yield of uniparental disomy calling in a trio exome cohort of 8.567 cases

Martin Ritthaler 1, Christine Froehlich1, Florian Battke2, Günter Jäger2, Heinz Gabriel1, Björn Schulte1, Martin Schulze1, Saskia Biskup1

1Zentrum für Humangenetik Tübingen, Tübingen, Germany; 2CeGaT GmbH, Tübingen, Germany

Background/Objectives: Trio exome diagnostics is the method of choice in syndromic cases without a clear suspected diagnosis for the detection of sequence variants and copy number variants. However, common causes of imprinting disorders, such as uniparental disomies (UPDs), are typically not addressed in standard trio exome analysis. We have therefore established a standardized calling method for the detection of uniparental hetero- (hUPD) and isodisomies (iUPD) in our trio analyses.

Methods: iUPDs can be detected as ROHs in single exomes, based on clusters of homozygous variants. Detection of hUPDs requires data from both parents: For each SNV, zygosity of the index is compared to the parental data and classified as either clearly inherited from one parent, de novo, or unclear. Positions are aggregated with a method similar PLINK ROH caller. Overlapping paternal UPD and maternal UPD calls are removed. Suspected UPDs were confirmed by ME-MLPA.

Results: The UPD calling of 8.567 trios has great implications for genetic diagnostics. Diagnostic yield of trio exomes can be significantly increased using this approach.

Conclusion: Trio exome diagnostics can be used for the detection of UPDs in syndromic cases. A standardized calling method can improve the diagnostic yield but requires validation through a different technology (such as ME-MLPA).

Conflict of Interest: None declared

EP15.017 Isolated multiple meningiomas: clinical audit of patients referred to the Peninsula Schwannomatosis Service between 2017 to 2023

Susan Louw 1, Ruth Cleaver2, Helen Tomkins3

1Royal Devon University Hospital, Clinical Genetics, Exeter, United Kingdom; 2Royal Devon University Hospital, Clinical Genetics, United Kingdom; 3University Hospitals Plymouth NHS Trust, Neurology, United Kingdom

Background/Objectives: NF2-related schwannomatosis is a rare autosomal dominant genetic disorder caused by a pathogenic or likely pathogenic variant in the NF2 gene. NF2-related schwannomatosis presents with nervous system tumours and ocular abnormalities. Individuals with NF2-related schwannomatosis have an estimated lifetime risk of 50% of developing multiple meningiomas.

Individuals with two or more meningiomas are referred to the Peninsula Schwannomatosis Service, which is part of a nationally commissioned service, for further investigation. There has been a significant increase in referrals to the service, necessitating a review of management pathways. This audit investigated the diagnostic rate of NF2-related schwannomatosis following full investigation including ophthalmology, neurological review, and genetic testing.

Methods: A clinical audit was undertaken using genetics files from the Peninsula Genetics Service and referral data from the Peninsula Schwannomatosis Service, between January 2017 and December 2023.

Data collected from 49 patients included number of meningiomas, age of diagnosis, additional imaging findings, relevant skin findings, ophthalmology review and genetic testing. Patients with outstanding investigations were excluded.

Results: No patients presenting with multiple meningiomas had any additional features detected upon further imaging, examination, or genetic testing. None received a clinical diagnosis of NF2-related schwannomatosis.

Conclusion: Based on our small cohort, NF2-related schwannomatosis is an unlikely diagnosis in isolated multiple meningioma cases. Mosaic NF2-related schwannomatosis cannot be excluded in most cases. Collecting the same data from other Schwannomatosis services would be beneficial in shaping referral guidance for the national service and management of patients.

Grants: None

Conflict of Interest: None declared

EP15.019 ORF15 long-read: a new strategy to improve molecular diagnosis of the mutational hot spot of X-linked RP

Valérie FAUGERE 1, Christel Vache1, David BAUX1, Luke Mansard1, Charles VAN GOETHEM1, Claire-Marie Dhaenens2, Olivier Grunewald2, Isabelle Audo3, Christina Zeitz3, Isabelanne Meunier4, Beatrice Bocquet4, Mireille Cossee1, Anne BERGOUGNOUX1, Vasiliki Kalatzis5, Anne-Françoise Roux1

1Molecular Genetics Laboratory, Univ Montpellier, CHU Montpellier, Montpellier, France; 2Univ. Lille, Inserm, CHU Lille,U1172-LilNCog-Lille Neuroscience and Cognition, Lille, France; 3Sorbonne Université, Inserm, CNRS, Institut de la Vision, Paris, France; 4National Reference Center for Inherited Sensory Diseases, Univ Montpellier, CHU, Montpellier, France; 5Institute for Neurosciences of Montpellier (INM), Univ Montpellier, Inserm, Montpellier, France

Background/Objectives: X-linked retinitis pigmentosa (XLRP) is characterized by progressive vision loss leading to legal blindness in males and a broad severity spectrum in carrier females. Pathogenic alterations of the retinitis pigmentosa GTPase regulator gene (RPGR) are responsible for over 70% of XLRP cases and the large majority of them are localized among its ORF15 terminal exon.

Sequencing challenges inherent to the ORF15 region of RPGR are well known to diagnostic laboratories because of the low-complexity of its sequence. Consequently, to improve detection of pathogenic variants in ORF15, we developed a long-read sequencing approach.

Methods: A total of 32 different pathogenic variants were tested in a blind test, 20 of them in both hemizygous and heterozygous states, including substitutions, small duplications, deletions, insertion and delins. 20 of them were localized in the most difficult ORF15 portion to sequence. After amplification of a 2062 bp sequence encompassing the entire ORF15 reading frame, libraries were prepared and sequenced with a MinION (Oxford Nanopore).

Results: Combining bioinformatic analyses with visual explorations of ORF15 long-reads, we obtained a 100% detection rate compatible with a high-performance molecular diagnostic offer for both males and female carriers.

Conclusion: In line with this goal of improving mutation detection, EMQN has introduced a new EQA for Inherited Retinal Disorders focusing in particular on detecting ORF15 mutations, highlighting the importance of this region. Indeed, until now, ORF15 pathogenic variants could represent a real pitfall for accurate diagnosis. We show in this study that long-read screening of ORF15 offers a comprehensive diagnosis.

Conflict of Interest: None declared

EP15.021 X-Chromosome Truncation Artefact Observed on Analysis of FFPE Tissue with FISH

Graziella Camilleri 1;2, Rachelle Muscat2, Karen Muscat2, Jean Calleja Agius1, Edith Said1;2

1University of Malta, Department of Anatomy, Faculty of Medicine & Surgery, Msida, Malta; 2Mater Dei Hospital, Department of Pathology, Msida, Malta

Background/Objectives: A hydatidiform molar (HM) pregnancy occurs when a non-viable fertilised egg implants itself

in the uterus and develops into an abnormal embryonic mass with hydropic degeneration of

chorionic villi. HM pregnancies are divided into two broad categories, partial HM (PHM) and complete HM (CHM), which have different morphological and genetic characteristics and the correct classification is important to the patient’s management.

Methods: Five chromosomes; 13, 18, 21, X and Y, were assessed by fluorescence in-situ hybridisation (FISH) in 11 products of conception tissues with a clinical and macroscopical picture of HM that were found negative for the p57KIP2 protein by immunohistochemistry analysis.

Results: Truncation artefacts were challenging in the analysis of FISH stained FFPE tissue sections of these tissue. The higher incidence of chromosome X monosomy in these cases, a feature not as exhibited by the other chromosomes is of note.

Although, the signals for chromosome X were less distinct with a significant number of nuclei showing only 1 green signal, the percentage of monosomy X did not surpass 70% and were therefore classified as diploid XX CHMs.

Conclusion: The objective of the study was to establish a cut-off value as to correctly classify these tissue. A cut off of 70% of cells demonstrating monosomic hybridisiation of the chromosome X was used to differentiate diploidy from monosomy in p57KIP2 negative immunohistochemical product of conception tissue.

Grants: The research work disclosed in this publication is partially funded by the ENDEAVOUR Scholarships Scheme (Group B) held by Graziella Camilleri.

Conflict of Interest: None declared

EP15.022 Case validation for variants: local experience and perspectives

Aleksey Antonenko 1, Ruslan Belov1, Maria Revkova1, Ekaterina Domoratskaya1, Alena Bartsis1, Anna Panferova1, Milana Gubona1, Polina Ulanova1, Maria Makarova1, Anastasia Krinitsina1, Maxim Belenikin1

1Evogen LLC, Moscow, Russian Federation

Background/Objectives: Recent progress in NGS, bioinformatics algorithms and sequencing data processing are increasingly actualizing the question about needing for validation of the potential causative variants by alternative methods. For the last decades capillary sequencing by Sanger is “gold standard” validation tool for clinical research. A lot of published data for Sanger validation for exome data, but little information for WGS. The most recent NGS recommendations does not require alternative validation. However, sometimes validation is necessary. Current research aimed to summarize laboratory threshold quality criteria for variant validation and providing an approaches for extensive case validation.

Methods: WGS: PE150, enzymatic fragmentation, PCR-free, DNBseq-T7/G400 (MGI); Sanger sequencing: ABI3500 (ABI).

Results: For data analysis we used 20K + WGS samples (30x) from our database: ~3000 samples with unique Sanger validated orphan/rare variants and health control samples for frequency database.

Conclusion: (1) The variant quality threshold value depends on the details of the algorithm and pipeline; (2) Sanger validation was useful for ~2% of the most complex variants (e.g., indels up to 30 bp, if additional variants are located in close proximity to the validated variant, areas with repeats, GC-regions, pseudogenes, variant with coverage <10x), however, most of these variants it is possible still to call in silico with additional advanced bioinformatic processing of NGS data. (3) The most important thing is validation is required mostly for clinical cases validation, so the most effective way more informative and extensive segregation analysis would provide ~10x WGS in addition to 30x proband sequencing.

Additional: Informed consents were obtained for all cases.

Grants:

Conflict of Interest: None declared

EP15.023 Validation by capillary sequencing: is it still necessary or is already redundant?

Maria Revkova 1, Ekaterina Domoratskaya1, Dmitrii Markov1, Polina Ulanova1, Olga Chesnokova1, Natalia Sokolova1, Aleksey Antonenko1, Anna Panferova1, Natalia Doroshchuk1, Anastasia Krinitsina1, Maxim Belenikin1

1Evogen, Moskva, Russian Federation

Background: The current high throughput sequencing technologies allow to provide high-quality data. There have been many reports suggesting that NGS data are more accurate than Sanger sequencing in most cases. NGS data quality scores such as (1) depth of coverage and (2) allele representation need to be definitely specified for using in clinical settings without Sanger validation. Both were analyzed for NGS variants not confirmed by Sanger sequencing.

Methods: DNA extraction: blood (MGIEasy). WGS: PE150, enzymatic fragmentation, PCR-free, DNBseq-T7/G400 (MGI). Sanger sequencing: ABI3500 (ABI).

Results: Almost 100% of “high quality” NGS variants were confirmed by first or at least second Sanger sequencing run in some cases. 23 out of 1356 (1.7%) cases showed discrepancy between NGS and Sanger data. For 6 cases (coverage >25X) the difference was caused by allelic dropout during PCR solved by using new primers. Other 17 non-confirmed cases had coverage <25Х and mainly allele representation <15%, two of them were located in high GC region or had pseudogene(s). No alternative alleles were identified for them either.

Conclusion: The “high quality” NGS variants were mainly confirmed. The reasons of non-conformity are (1) low allele representation (<15%), (2) incorrect PCR causing by the features of the genome region where the variant is located, (3) details of bioinformatic analysis.

Conflict of Interest: None declared

EP15.024 Unreported phenotype associated to SPAST biallelic variants: cerebral palsy masquerade

Gregorio Nolasco 1, Monica Roldan2, Florencia Epifani1, Roser Urreizti3, LLuís Armengol4, Daniel Natera-de Benito5, Andrés Nascimento5, anna codina6, Maria Lluïsa Ramírez7, Cristina Hernando-Davalillo6, Carlos Ortez5, Loreto Martorell Sampol6, Mercedes Serrano8

1Sant Joan de Déu Barcelona Hospital, Institut de Recerca Sant Joan de Déu; Pediatric Neurology Department, Esplugues de Llobregat, Spain; 2Sant Joan de Déu Barcelona Hospital, Confocal Microscopy and Cellular Imaging Unit, Genetic and Molecular Medicine Department, Pediatric Institute for Rare Diseases, Esplugues de Llobregat, Spain; 3Sant Joan de Déu Barcelona Hospital, Clinical Biochemistry Department; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Instituto de Salud Carlos III, Esplugues de Llobregat, Spain; 4qGenomics, Quantitative Genomic Medicine Laboratories, qGenomics, Esplugues de Llobregat, Spain; 5Sant Joan de Déu Barcelona Hospital, Neuromuscular Unit; Pediatric Neurology Department, Esplugues de Llobregat, Spain; 6Sant Joan de Déu Barcelona Hospital, Genetic and Molecular Medicine Department, Pediatric Institute for Rare Diseases, Esplugues de Llobregat, Spain; 7Sant Joan de Déu Barcelona Hospital, Genetic and Molecular Medicine Department; Pediatric Institute for Rare Diseases, Esplugues de Llobregat, Spain; 8Sant Joan de Déu Barcelona Hospital, Institut de Recerca Sant Joan de Déu; Pediatric Neurology Department; Clinical Biochemistry Department; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Instituto de Salud Carlos III, Esplugues de Llobregat, Spain

Background/Objectives: Monoallelic variants in SPAST are the most common cause of hereditary spastic paraparesis (HSP). It is a heterogeneous group of neurodegenerative disorders. We report individuals with progressive encephalopathy with spasticity and neuronoaxonal invlvement and biallelic variants in SPAST.

Methods: Mutational analysis was performed using gene panel or trio WES. Spastin was localized and quantified in fibroblasts from patients and compared with healthy control and classic phenotype (dominant inheritance). We performed functional studies by confocal microscopy: immunofluorescence labeling of spastin, tubulin, nucleus and in vivo studies using Image J software.

Results: In silico analysis predicted high impact on protein structure. Confocal microscopy revealed differences in SPAST fluorescence intensity in individuals with biallelic variants compared to healthy individuals. We identified aberrant plasma membrane prolongations and indirect signs of mitochondrial apoptosis in patient cells.

Conclusion: This is the first report of individuals with biallelic variants in SPAST. Our findings expand the etiological possibilities under the so-called PCI mimics. Functional experimental studies are essential to unravel and understand the biological mechanisms of this recently described phenotype.

Grants: This work was supported by national grants (PI18/00101) from the National Plan on I + D + I, co-financed by ISCIII (Subdirección General de Evaluación y Fomento de la Investigación Sanitaria) and FEDER (Fondo Europeo de Desarrollo Regional). Plan on I + D + I. The funding sources had no role in the design and conduct of the study; in the collection, management, analysis, or interpretation of the data; it the preparation, review, or approval of the manuscript.

Conflict of Interest: Gregorio Nolasco This work was supported by national grants (PI18/00101) from the National Plan on I + D + I, co-financed by ISCIII (Subdirección General de Evaluación y Fomento de la Investigación Sanitaria) and FEDER (Fondo Europeo de Desarrollo Regional). Doctor Epifani was supported by a national grant (FI22/00218) from the National Plan on I + D + I. The funding sources had no role in the design and conduct of the study; in the collection, management, analysis, or interpretation of the data; it the preparation, review, or approval of the manuscript; or in the decision to submit the manuscript for publication., Monica Roldan: None declared, Florencia Epifani: None declared, Roser Urreizti This work was supported by national grants (PI18/00101) from the National Plan on I + D + I, co-financed by ISCIII (Subdirección General de Evaluación y Fomento de la Investigación Sanitaria) and FEDER (Fondo Europeo de Desarrollo Regional). Doctor Epifani was supported by a national grant (FI22/00218) from the National Plan on I + D + I. The funding sources had no role in the design and conduct of the study; in the collection, management, analysis, or interpretation of the data; it the preparation, review, or approval of the manuscript; or in the decision to submit the manuscript for publication., LLuís Armengol: None declared, Daniel Natera-de Benito: None declared, Andrés Nascimento: None declared, anna codina: None declared, Maria Lluïsa Ramírez: None declared, Cristina Hernando-Davalillo: None declared, Carlos Ortez: None declared, Loreto Martorell Sampol: None declared, Mercedes Serrano This work was supported by national grants (PI18/00101) from the National Plan on I + D + I, co-financed by ISCIII (Subdirección General de Evaluación y Fomento de la Investigación Sanitaria) and FEDER (Fondo Europeo de Desarrollo Regional). Doctor Epifani was supported by a national grant (FI22/00218) from the National Plan on I + D + I. The funding sources had no role in the design and conduct of the study; in the collection, management, analysis, or interpretation of the data; it the preparation, review, or approval of the manuscript; or in the decision to submit the manuscript for publication.

EP15.025 Uncovering XX Male Syndrome (De la Chapelle Syndrome): Insights from STR Analysis in a Paternity Testing Case

Marian Baldovic 1;2, Gabriela Bľandová1;3, Gabriela Krasnanska1, Lenka Wachsmannova1, Vladimír Eliáš1, Vladimír Ferák1, Michal Konecny1;4

1GHC GENETICS SK, Science Park Comenius University, Laboratory of Genomic Medicine, Bratislava, Slovakia; 2Faculty of Natural Sciences, Comenius Univesity, Department of Molecular Biology, Bratislava, Slovakia; 3Faculty of Medicine, Comenius University, Institute of Medical Biology, Genetics and Clinical Genetics, Bratislava, Slovakia; 4Faculty of Natural Sciences, University of Ss. Cyril and Methodius, Department of Biology, Trnava, Slovakia

Background/Objectives: XX male or De la Chapelle Syndrome is a rare genetic condition (approx. 1:20,000 newborn males) characterized by individuals with a male phenotype despite having two X chromosomes (46, XX karyotype). Majority of cases are caused by atypical crossing-over during paternal meiosis, resulting in translocations or duplications of the SRY gene into X and loss of Y chromosome (SRY positive XX male). Affected individuals manifest with varying degrees of genital ambiguity, infertility, and incomplete masculinization even modified by inactivation of X chromosome bearing SRY. Some individuals may show no or mild symptoms and can be difficult to identify.

Methods: DNA samples were initially assessed using the Investigator ESSplex SE QS Kit for STR profiling. Further characterization of suspected XX male syndrome involved additional STR analyses using the Aneufast™ QF-PCR Kit and the PowerPlex® Y23 System. Additionally, the Devyser AZF kit was utilized for detecting Y chromosomal microdeletions.

Results: Routine STR profiling revealed a lack of signal for the AMELY marker, which was further corroborated by the absence of multiple Y chromosomal markers in the Aneufast, PowerPlex, and AZF analysis. This enabled an approximate reconstruction of the extent of Y chromosomal rearrangements.

Conclusion: Anonymous paternity testing led to the identification of XX male syndrome in the alleged father, albeit with unknown clinical conditions. Nonetheless, these findings underscore the potential of utilizing forensic genetics to diagnose rare genetic conditions such as XX Male Syndrome.

Grants: The authors acknowledge the contribution of the Slovak Research and Development Agency under the project APVV-20-0070.

Conflict of Interest: None declared

EP15.026 Elucidating the emerging genotype-phenotype correlations and clinical spectrum of MARS1-related disease.

Jacob Day 1, Allison Newman1, Claire Salter1, Fathiya Al-Murshedi2, Almundher Al-Maawali2, Amna Mohammed Al-Futaisi2, Fowzan Alkuraya3, Lama Al-Abdi4, Nishanka Ubeyratna1, Joseph Leslie1, Richard Caswell5, Emma Baple1;6, Andrew Crosby1

1RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom; 2Genetic and Developmental Medicine Clinic, Department of Genetics, College of Medicine and Health Sciences, Sultan Qaboos University Hospital, Muscat, Oman; 3Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia; 4Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia; 5Exeter Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, RILD Building - Level 3, Exeter, United Kingdom; 6Peninsula Clinical Genetics Service, Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom

Background/Objectives: Mono- and biallelic methionyl-tRNA synthetase (MARS1) variants have previously been associated with a diverse range of clinical presentations spanning neurological, interstitial liver and lung, as well as skin and hair manifestations. However, the genetic evidence for some of these associations is equivocal, resulting in lack of clarity of potential genotype-phenotype correlations and disease pathomechanisms. Here we provide a comprehensive description of MARS1-related disease including descriptions of new as well as previously published affected individuals to clarify disease pathomechanisms and clinical outcomes.

Methods: Detailed clinical evaluations were undertaken alongside whole exome/genome sequencing and in-silico modelling. New affected individuals were identified through GeneMatcher, international collaboration and peer-reviewed literature.

Results: We identified new families and reviewed literature describing previously published candidate families with mono/biallelic MARS1 variants. Our findings identify multisystem presentations spanning trichothiodystrophy, hereditary spastic paraplegia with/without developmental impairment, pulmonary alveolar proteinosis and hepatic steatosis-cirrhosis as common outcomes associated with biallelic MARS1 variants. Consideration of the location of these variants identified an emerging underlying genotype-phenotype correlation and disease pathomechanisms, particularly linked with specific molecular domains. The evidence linking monoallelic MARS1 variants with disease was found to be more equivocal.

Conclusion: Our study considerably expands knowledge of the genetic basis, and identifies emerging genotype-phenotype correlations and pathomechanisms, of MARS1-related disease. This has enabled the development of guidance to support diagnostic classification of MARS1 variants.

Grants: GW4-CAT HP PhD Programme (Wellcome Trust), The Halpin Trust, Hereditary Spastic Paraplegia Support Group

Conflict of Interest: None declared

EP15.027 Next-generation sequencing for the diagnosis of hereditary angioedema

Jerneja Debeljak 1, Nina Rupar1, Julij Šelb1;2, Peter Korošec1;3, Matija Rijavec1;4

1University Clinic of Respiratory and Allergic Diseases Golnik, Laboratory for Clinical Immunology and Molecular Genetics, Golnik, Slovenia; 2Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; 3Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia; 4Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia

Background/Objectives: Hereditary angioedema with C1 inhibitor deficiency (C1-INH-HAE) is caused by a range of variants (more than 800 described to date) in the SERPING1 gene. Implementation of next-generation sequencing (NGS) for genetic testing is an efficient alternative to currently used Sanger sequencing followed by Multiplex Ligation-dependent Probe Amplification (MLPA). The aim of this study was to develop and introduce a new single-step NGS method for genetic analysis of patients suspected of C1-INH-HAE into routine clinical practice.

Methods: The QIAseq Targeted DNA Custom Panel was designed to cover the entire SERPING1 gene; sequencing was performed on Illumina MiSeq, followed by the bioinformatic analysis (standard GATK germline variant calling pipeline and Atlas-CNV or DELLY), which allowed us detection of SNVs, small indels, and CNVs, including breakpoint predictions. In total, 35 different SERPING1 variants (previously tested by Sanger sequencing and/or MLPA) and 42 controls were included in the validation process.

Results: Our targeted sequencing panel provide 100% coverage of all exonic, 5’- and 3’- untranslated regions and exon-intron boundaries, as well as most of the intronic regions, with 94% overall coverage of the entire SERPING1 gene. The sensitivity and specificity of rare variant detection was 100%. No rare variants were found in controls. Importantly, in samples with large defects, we were able to predict the precise size and location of the deleted and inserted DNA fragments.

Conclusion: Using targeted NGS panel, we could accurately identify SNVs, small indels or CNVs, thus introducing a valid single-step improvement in the genetic testing of C1-INH-HAE.

Grants: ARRS-P3-0360, ARRS-J3-2532

Conflict of Interest: None declared

EP15.028 Development of preimplantation genetic testing for monogenic reference materials using next-generation sequencing

Weihua Zhao1, Yanyan Song 2, Chuanfeng Huang3, Shan Xu2, Qi Luo1, Runsi Yao1, Nan Sun3, Bo Liang4;5, Jia Fei6, Fangfang Gao7, Jie Huang3, Shoufang Qu3

1Shenzhen Second People’s Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, China; 2MGI Tech Co., Ltd.; 3National Institutes for food and drug Control (NIFDC); 4State Key Laboratory of Microbial Metabolism; 5Basecare Medical Device Co., Ltd.; 6Peking Jabrehoo Med Tech Co., Ltd.; 7Yikon Genomics Company,Ltd

Objective: Preimplantation genetic testing for monogenic disorders (PGT-M) has been used for over 20 years to detect many serious genetic conditions. However, there is still a lack of reference materials (RMs) to validate the test performance during the development and quality control of PGT-M.

Method: Sixteen thalassemia cell lines from four thalassemia families were selected to establish the RMs. Each family consisted of parents with heterozygous mutations for α- and/or β-thalassemia and two children, at least one of whom carried a homozygous thalassemia mutation (proband). The RM panel consisted of 12 DNA samples (parents and probands in 4 families) and 4 simulated embryos (cell lines constructed from blood samples from the four nonproband children). Four accredited genetics laboratories that offer verification of thalassemia samples were invited to evaluate the performance of the RM panel. Furthermore, the stability of the RMs was determined by testing after freeze-thaw cycles and long-term storage.

Results: PGT-M RMs containing 12 genome DNA (gDNA) reference materials and 4 simulated embryo reference materials for thalassemia testing were successfully established. The genotypes and haplotypes of all 16 PGT-M RMs were concordant across the four labs, which used various testing workflows. These well-characterized PGT-M RMs retained their stability even after 3 years of storage.

Conclusion: The establishment of PGT-M RMs for thalassemia will help with the standardization and accuracy of PGT-M in clinical use.

Grants: This study received funding from the National Key R&D Program of China (2021YFC2700500) and Technology and Innovation Commission of Shenzhen Municipality (JCYJ20200109140623124).

Conflict of Interest: None declared

EP16 Bioinformatics, Machine Learning and Statistical Methods

EP16.002 Assessment of long-read assemblers for sequencing genetic regions with long and high copy number variations

Noriko Tanaka 1;2, Yuki Onishi3, Maria Yamasaki4, Masako Kifushi3

1Kyoto University, Graduate School of Biostudies, Center for Living Systems Information Science, Kyoto, Japan; 2National Center for Global Health and Medicine, Genome Medical Science Project-Toyama, Tokyo, Japan; 3Waseda University, Graduate School of Advanced Science and Engineering, Department of Life Science and Medical Bioscience, Tokyo, Japan; 4Tokyo Metropolitan Institute for Geriatrics and Gerontology, Integrated Research Initiative for Living Well with Dementia, Tokyo, Japan

Background/Objectives: Many studies suggested that structural variations, especially high copy number variations (CNVs) may have a critical role to develop rare diseases or cancers. Modern long-read technologies enable us to sequence most of genetic regions, however, it is still very challenging to genotype high CNVs. One of the very famous genetic regions with long and high CNVs is the Kringle IV 2 (KIV2) in the LPA gene, which has been measured by many research groups as bench marking of the measurement technologies.

Methods: Among 10 long-read assemblers that were benchmarking in Sun et al. (2021), four tools (Redbean (0.0 (19830203)), flye (2.9.1-b1780), flye (2.8.1-b1676), NextDenovo (v2.5), shasta (0.11.1)) were selected to assess their performance to genotype LPA-KIV2 CNV using 7 samples including NA12878, that were sequenced on the Oxford Nanopore MinION in the Whole Human Genome Sequencing Project.

Results: For all samples, total number of sequenced contigs in the flye (2.8.1-b1676) was the highest, and the repeated sequence and the primer sequence to measure CNV counts by PCR methods were included on the same contig. However, the primer sequence was not observed on the contigs in other assemblies, even in the upgrade version of flye (2.9.1). Any sample was not assembled successfully with NextDenovo.

Conclusion: The best performing tool for assembling the region including LPA-KIV2 high CNV would be the flye (2.8.1-b1676).

Grants: This study was supported in part by a Grant-in-aid for Scientific Research (C) (#17K08718) and Early Career Scientists (#18K15055) from the Ministry of Education, Culture, Sports, Science, and Technology, Japan (http://www.jsps.go.jp/english/e-grants/index.html).

Conflict of Interest: None declared

EP16.003 Automated decision support for the clinical interpretation of copy number variants

Jaroslav Budiš 1;2;3, Tomáš Sládeček1;4, Michaela Gažiová1;5, Marcel Kucharik1;4, Zuzana Pös1;2;6, Ondrej Pös1;4, Zuzana Hanzlikova1;7, Jakub Styk1;2, Monika Kubanova4, Diana Rusnakova1;4, Jozef Martis1;4, Juraj Gazdarica1;3;4, Jozef Sitarcik1;4, Werner Krampl1;2;4, Ján Radvánszky1;2;6, Tomas Szemes1;2;4

1Geneton Ltd., Bratislava IV, Slovakia; 2Comenius University Faculty of Natural Sciences, Department of Molecular Biology, Bratislava, Slovakia; 3Slovak Centre of Scientific and Technical Information, Bratislava, Slovakia; 4Comenius University Science Park, Bratislava, Slovakia; 5Comenius University, Faculty of Mathematics, Physics and Informatics, Department of Computer Science, Bratislava, Slovakia; 6Biomedical Research Center, Slovak Academy of Sciences, Institute of Clinical and Translational Research, Bratislava, Slovakia; 7Faculty of Informatics and Information Technologies STU, Bratislava, Slovakia

Background/Objectives: Clinical interpretation of copy number variants (CNVs) is a complex process that often leads to different decisions between diagnostic centers. In order to alleviate this obstacle, technical standards for the interpretation and reporting of variants have been developed, called ACMG guidelines. Several semiautomatic computational methods have been proposed to recommend appropriate choices. Alternately, the emerging machine learning-based tools showed promising ways of even fully automated predictions.

Methods: We evaluated state-of-the-art computational prediction tools on CNV records from the ClinVar database and showed the superior accuracy of their combination. We also tweaked the key parameters and underlying data sources to demonstrate their impact on the overall prediction.

Results: Here, we demonstrate that the choice of underlying data sources significantly affects the prediction of CNV pathogenicity. Automated prediction tools, especially their combination, further improve the evaluation of ACMG guidelines and thus lead to more reliable clinical decision support for clinicians.

Conclusion: Modern clinical decision support tools for the clinical interpretation of CNVs are able to provide valuable guidance to clinicians, relieving them of a great share of tedious annotation and interpretation processes.

Grants: This work was supported by the PANGAIA project H2020-MSCA-RISE-2019 (Grant agreement ID: 872539) funded under H2020-EU.1.3.3. Programme; by the ALPACA project H2020-MSCA-ITN-2020 (Grant agreement ID: 956229) funded under H2020-EU.1.3.1. Programme; by the Slovak Research and Development Agency (grants APVV-20-0472 Sepmin, and APVV-21-0296 INCAM); and by the Horizon Europe Framework Programme (Grant agreement ID: 101087124 — ADDIT-CE).

Conflict of Interest: Jaroslav Budiš The author is an employee of Geneton Ltd. and participates in the development of a commercial application for the automated annotation and interpretation of CNVs. However, the employee does not hold any financial interest in the company., Tomáš Sládeček The author is an employee of Geneton Ltd. and participates in the development of a commercial application for the automated annotation and interpretation of CNVs. However, the employee does not hold any financial interest in the company., Michaela Gažiová The author is an employee of Geneton Ltd. and participates in the development of a commercial application for the automated annotation and interpretation of CNVs. However, the employee does not hold any financial interest in the company., Marcel Kucharik The author is an employee of Geneton Ltd. and participates in the development of a commercial application for the automated annotation and interpretation of CNVs. However, the employee does not hold any financial interest in the company., Zuzana Pös The author is an employee of Geneton Ltd. and participates in the development of a commercial application for the automated annotation and interpretation of CNVs. However, the employee does not hold any financial interest in the company., Ondrej Pös The author is an employee of Geneton Ltd. and participates in the development of a commercial application for the automated annotation and interpretation of CNVs. However, the employee does not hold any financial interest in the company., Zuzana Hanzlikova The author is an employee of Geneton Ltd. and participates in the development of a commercial application for the automated annotation and interpretation of CNVs. However, the employee does not hold any financial interest in the company., Jakub Styk The author is an employee of Geneton Ltd. and participates in the development of a commercial application for the automated annotation and interpretation of CNVs. However, the employee does not hold any financial interest in the company., Monika Kubanova: None declared, Diana Rusnakova The author is an employee of Geneton Ltd. and participates in the development of a commercial application for the automated annotation and interpretation of CNVs. However, the employee does not hold any financial interest in the company., Jozef Martis The author is an employee of Geneton Ltd. and participates in the development of a commercial application for the automated annotation and interpretation of CNVs. However, the employee does not hold any financial interest in the company., Juraj Gazdarica The author is an employee of Geneton Ltd. and participates in the development of a commercial application for the automated annotation and interpretation of CNVs. However, the employee does not hold any financial interest in the company., Jozef Sitarcik The author is an employee of Geneton Ltd. and participates in the development of a commercial application for the automated annotation and interpretation of CNVs. However, the employee does not hold any financial interest in the company., Werner Krampl The author is an employee of Geneton Ltd. and participates in the development of a commercial application for the automated annotation and interpretation of CNVs. However, the employee does not hold any financial interest in the company., Ján Radvánszky The author is an employee of Geneton Ltd. and participates in the development of a commercial application for the automated annotation and interpretation of CNVs. However, the employee does not hold any financial interest in the company., Tomas Szemes The author is an employee of Geneton Ltd. and participates in the development of a commercial application for the automated annotation and interpretation of CNVs. However, the employee does not hold any financial interest in the company.

EP16.004 Transposition of great vessels associated with chromosome 1 microdeletion: A case report.

María Aguirre1, Lina Moreno-Giraldo 1, armicson felipe solano1

1universidad libre, ginecologia, Cali, Colombia

Background/Objectives: Transposition of the Great Arteries is part of 5 to 7% of major congenital heart diseases, with a survival rate up to 99% when detected early during pregnancy, and an early approach is carried out in postnatal life. Considering its incidence 2-3/10,000 live birth, it is important to recognize its presence and association with involvement of other organs and/or systems as a result of chromosomal alterations due to microdeletion and/or microduplication processes that lead to the presentation of various genetic syndromes that contribute to the overexpression of specific genes involved in these conditions.

Aim: Present the case of a patient with a microdeletion of chromosome 1q21.1, highlighting the importance of identifying gene interactions, as an essential step to understand the complexity of polygenic conditions, and understand the great phenotypic variability.

Results: Case presentation: Patient with no history, reports in anatomical detail ultrasound transposition of the great vessels, in addition to intrauterine growth restriction. During the genetic studies, she has normal karyotype and a microarray with a 1.4Mb microdeletion of chromosome 1q21.1. At birth with facial dysmorphism associated with heart disease.

Conclusion: The 1q21.1 microdeletion is a chromosomal alteration, in which multiple genes are affected, which can result in a wide variety of phenotypic alterations, thus, there is a wide range of presentation in terms of cardiac pathology, which leads to the importance of genetic studies as the method of choice for its identification and correct multidisciplinary approach, as well as to highlight the importance of 5P medicine.

Conflict of Interest: None declared

EP16.006 functional consequence of loss of heterozygosity regions: benign or pathogenic

Mert Polat 1, Zerrin Çelik1, Yunus Kasım Terzi1

1Başkent University Ankara Hospital, Department of Medical Genetic, ANKARA, Türkyie

Background/Objectives: Loss of heterozygosity (LOH) is a genetic alteration resulting from losing one allele at a heterozygous locus. Uniparental disomy and gene conversion are some of the reasons for LOH. The objective of this study is to identify the location and possible effects of LOH regions in the genome.

Methods: The study analyzed microarray data from 600 patients between 2020 and 2023 using the Affymetrix CytoScan Optima 315K (Thermo Fisher Scientific, Waltham, MA, USA). Only data from 440 patients met the analysis criteria. Genomic segments detected by at least 50 consecutive markers and equal or greater than 10 Megabase pairs’ length were classified as LOH regions.

Results: We detected a higher frequency of LOH in chromosomes 1, 2, and 5 compared to other autosomal chromosomes. Out of 440 patients, LOH was detected in 107. Among them, 71 showed no numerical abnormality, while 36 showed a numerical abnormality. Individuals with similar phenotypes and LOH regions were compared in a subgroup.

Conclusion: Loss of heterozygosity (LOH) is a significant factor in genetic diversity and disease development. LOH events are commonly found in regions with high rates of mitotic recombination, which are often susceptible to DNA damage. Genetic alterations in these regions can contribute to the progression of various genetic and somatic diseases. By establishing a strong connection between LOH and recombination events, we can gain valuable insights into disease pathogenesis, leading to better treatments for various diseases. Thus, it is a crucial aspect of medical research.

Grants: There is no grant.

Conflict of Interest: None declared

EP16.007 From Pixel to Gene: Unveiling Radiogenomics in Adult Gliomas through AI Predictions, Scoping Reviews, and the Promise of Virtual Biopsy

Xavier Le Guillou Horn 1;2, Carole Guillevin2;3, Pierre Fayolle3, Clement Giraud2;3, Mathieu Naudin2;3, François Lecellier4, Christine Fernandez4, Remy Guillevin2;3

1CHU de Poitiers, Service de Génétique Médicale, Poitiers, France; 2University of Poitiers, Labcom I3M, Chasseneuil du Poitou, France; 3CHU de POITIERS, Service de Radiologie, Poitiers, France; 4Université du CHU de Poitiers, XLIM, CNRS UMR 7252, Poitiers, France

Background/Objective: Gliomas, the primary malignant brain tumors in adults, have witnessed an escalating integration of genetics into their diagnosis and therapeutic management. With the advent of radiomics and genomics, there’s a growing interest in predicting glioma genetics from complex datasets, a concept akin to virtual biopsies. In this context, we conducted a scoping review on radiogenomics AI tools in adult gliomas.

Methods: A systematic search of EmBase and MedLine yielded 426 articles. After meticulous abstract review, articles not focusing on adult gliomas or those not constituting original research were excluded, leaving a refined dataset for analysis.

Results: The scoping review reaffirms the pivotal role of genetics in glioma classification and highlights the potential of AI models in predicting genetic features. Presently, existing AI models can predict individual genetic markers like IDH mutation, MGMT methylation, or specific caryotype abnormalities. However, these models are often specialized, providing fragmented results rather than a comprehensive genetic profile.

Conclusion: The synthesis of our scoping review underscores the current landscape of AI tools in radiogenomics for adult gliomas. While substantial progress has been made in predicting isolated genetic markers, there exists a notable gap in comprehensive models encompassing multiple genetic features. Bridging this gap could significantly enhance the clinical utility of AI in predicting glioma genetics, advancing towards a more holistic and personalized approach in glioma diagnosis and treatment planning.

Conflict of Interest: Xavier Le Guillou Horn Subject : Genetics in Cardiomyopathies and family screening for treatable forms

- TAKEDA (Fabry disease) : 2 times

- PFIZER (ATTR) : 1 time

- AMICUS (Fabry disease) : 1 time, Carole Guillevin: None declared, Pierre Fayolle: None declared, Clement Giraud: None declared, Mathieu Naudin: None declared, François Lecellier: None declared, Christine Fernandez: None declared, Remy Guillevin: None declared

EP16.008 Harmonized framework for RNA-seq-based rare disease diagnostics in a pan-continental consortium - Solve-RD

Vicente Yépez 1, Anna Esteve-Codina2, Julien Gagneur1

1Technical University of Munich; 2Centro Nacional de Análisis Genómico

Consortium: Solve-RD Consortium

RNA-seq WG of Solve-RD

Background/Objectives: RNA-seq is a complementary approach to DNA sequencing to discover disease-causing gene regulatory defects in rare disease individuals. However, as large consortia are implementing RNA-seq-based diagnostics, new challenges arise to cope with the heterogeneity of diseases, various tissues, sample sizes, and the multiplicity of interpreters. Here, we present new guidelines, algorithms, and methodologies that have been developed by the Pan-European consortium Solve-RD to address these issues. The dataset consists of >450 samples collected from four tissues from individuals tested at >20 clinics from six European Reference Networks.

Methods: Samples were analyzed using DROP, including the recent aberrant splicing caller FRASER2.0. Two RNA-seq quality control modules were added to confirm sex and tissue annotations. To identify candidate genes and variants, we considered a DNA-first approach, in which candidate variants are validated by RNA-seq and an RNA-first approach in which candidate genes are identified by aberrant expression or splicing, and the DNA data is then re-examined. For the newly-introduced RNA-first approach, we devised consortium-wide guidelines that integrate scoring of an aberrant event in combination with genotype and phenotype. To increase the consistency of the annotations, we conducted in-person workshops, Solvathons, combining training and collective analysis sessions, and offered weekly follow-ups.

Results: The approaches proved to be complementary and reported ~ten candidates per sample on average. Currently, twelve cases have been solved including intronic variants, deletions, and short tandem repeats missed by WES, some in multiple family members.

Conclusion: Altogether, our new algorithms and processes contribute to improved RNA-seq-based rare disease diagnostics methodologies.

Grants: GHGA (#1-NFDI-DFG)

Conflict of Interest: Vicente Yépez Employee at the Technical University of Munich, Anna Esteve-Codina Other grants, Employee at CNAG, Julien Gagneur Other grants, Employee at TUM

EP16.009 Benchmarking of phenotypic similarity-based metrics applied to unsolved rare diseases’ variant prioritization

Maroua Chahdil 1;1, Carolina Fabrizzi1, Caterina Lucano1, Leslie Matalonga2, Charlotte Rodwell1, Marc Hanauer1, Laurent Tichit3, David Lagorce1, ANA RATH1

1Paris, 75, Paris, France; 2Spain, Distrito de Les Corts, Barcelona, Spain; 3Marseille, 13, Marseille, France

Text:

Rare diseases (RD) have a prevalence of not more than 1/2000 in the European population, and are characterised by the difficulty of obtaining a correct and timely diagnosis. A significant proportion of patients suspected to have a genetic RD receive an inconclusive exome/genome sequencing and according to Orphanet, 72,5% of RD have a genetic origin although 35% of them do not yet have an identified causative gene. The Orphanet RD Knowledge Database stores, amongst others, a data model of known RD annotated with (i) phenotypic features using the Human Phenotype Ontology (HPO) and (ii) genetical information, both freely available on Orphadata.com. In the framework of the Solve-RD project, which aims to identify the molecular causes underlying undiagnosed RD, we published a phenotypic similarity-based variant prioritization methodology by comparing submitted patients’ cases (phenotypically annotated with HPO), as well as with known RD in Orphanet (ORPHAcodes). However, this promising methodology does not retrieve some diagnosed patient cases. To address this challenge, to enhance our method, we benchmarked existing methods for measuring phenotypic similarity distance selected in the literature (such as HPO3 or LIRICAL, amongst others), however few make their code available. Our evaluation criteria for these methods include interpretability, sensitivity, specificity, and precision highlighting the importance of genetic analysis. This benchmark was conducted on specific test sets, derived from a reference patient dataset based on the SolveRD project. Results were reviewed by a dedicated physician at Orphanet, who valuated the relevance of clinical signs with the goal of reducing human biases.

Conflict of Interest: None declared

EP16.010 Structural-Aware model in Disease-associated Variant Prioritization Reveals New Genes SUMF1 and Variants in Retinitis Pigmentosa

LiChao Huang 1, meng yang1, JiHong Wu2

1MGI, BGI-Shenzhen, Shenzhen 518083, China., X, China; 2Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan University, Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, China

Background: Retinitis Pigmentosa (RP) lacks a definitive genetic explanation in 30-50% of cases. Our goal is to use interpretable AI technology, based on WES data and clinical phenotypes, to assist clinicians in improving the diagnostic rate of difficult cases, to clarify the potential pathogenic genes and variations in these genetically unexplained RP cases, and to discover possible new pathogenic genes.

Methods: Building on clinical phenotyping, we developed the DVPP, capable of rapidly identifying pathogenic variants in patients. DVPP scores and prioritizes millions of diverse variants interpretably, considering DNA sequence, amino acid sequence, protein structure and clinical phenotype. We integrated the ESM-GVP model into DVPP, which predicts the impact of missense variants independently and provides insights into mutations at both the sequence and 3D structural levels. New RP pathogenic genes were validated through gene knockout experiments and Electroretinogram.

Results: This approach evaluates millions of candidate variants in 92 patients from multiple perspectives, including protein sequence, structure, and clinical phenotype, rapidly and accurately identifying pathogenic missense mutations in 42 uncertain cases. It recognized SUMF1 as a novel pathogenic gene for RP, validated through experimentation, effectively aiding clinicians in enhancing the diagnosis rate of uncertain cases (35.7%).

Conclusion: This advancement significantly supports clinicians in discovering causative mutations for hereditary diseases, rectifying previously unexplained cases, and expanding the potential applications of genomic data in clinical practice.

Grants: Supported by the National Key Research and Development Program of China (No.2020YFA0112703). Supported by NSFC 82271085 & 82171055. Sponsored by Program of Shanghai Municipal Commission of Science and Technology (No.21S11905900).

Conflict of Interest: LiChao Huang MGI, BGI-Shenzhen, Shenzhen 518083, China, meng yang MGI, BGI-Shenzhen, Shenzhen 518083, China, JiHong Wu Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai 200031, China.

EP16.011 Unveiling Trends in Chromosomal Abnormalities: A Comprehensive Meta-Analysis of Nearly 40,000 VeriSeqTM NIPT Studies

Beatriz Sanz-Milián 1, Albert Ferran Martin1, Luisa Aparicio2, Aroa Barba2, Rebeca Blanco2, Laura Llaó2, Lídia Parra2, Rosella Soler2, Alba Flores Bonet3, Irina Royo3

1Reference Laboratory S.A., Bioinformatics, Barcelona; 2Reference Laboratory S.A., Molecular Biology Department, Barcelona; 3Reference Laboratory S.A., Barcelona

Background/Objectives: Non-invasive prenatal testing (NIPT) is a screening technique that allows the detection of possible chromosomal abnormalities in the foetus through the analysis of foetal DNA fragments circulating in maternal blood.

As part of this study, a meta-analysis was carried out to examine how factors such as maternal age, foetal fraction, and the nature of the pregnancy (natural or IVF) may influence the likelihood of having chromosomal abnormalities during pregnancy. In addition, we sought to compare the results obtained from our Spanish cohort with data published in other regions of the world.

Methods: Clinical Data obtained from nearly 40,000 studies performed with VeriSeqTM NIPT (Illumina) were analysed bioinformatically

Results: After performing a statistical analysis of the various aspects studied, it was observed that the distribution of the different chromosomal abnormalities resembles that found in other cohorts, with trisomy 21 being the most common. In addition, the relevance of maternal age was highlighted, identifying that the 30-35 years age group is less likely to have chromosomal abnormalities during pregnancy.

It was corroborated that, statistically, IVF pregnancies, due to the pre-screening already performed, have a significantly lower positivity rate, apparently not associated with age.

Finally, it was observed that low circulating foetal fractions appear to be associated with certain chromosomal abnormalities.

Conclusion: The data obtained in this study show the effect that different factors may have on the likelihood of chromosomal abnormalities during pregnancy. These are corroborated by what has been seen in other populations.

Conflict of Interest: None declared

EP16.012 Analysis for mosaic and germline variants in pediatric cancer patients: comprehensive validation and optimization of a unified bioinformatics pipeline

Beatriz Ruz 1;2;3, Lucía de Dios1, Carmen Rodríguez Jiménez1;4, Victoria Eugenia Fernandez Montaño1, Maria Victoria Del Pozo-Gomez1, María Esther Rubio Martín1, Cristina Ortega-Patrón1, Elisa Izquierdo1;2;3, Clara Venegas1;2;3, Sonia Rodriguez Novoa1;5, adela escudero1;2;3, Antonio Pérez3;6;7, Carlos Rodríguez-Antolín1;8

1La Paz University Hospital, Department of Genetics, Madrid, Spain; 2Hospital La Paz Institute for Health Research – IdiPAZ, Pediatric Onco-Hematology, Clinical Research Unit, Madrid, Spain; 3CNIO, Pediatric Onco-Hematology, Clinical Research Unit, Madrid, Spain; 4IdiPAZ, Dyslipidemias of Genetic Origin and Metabolic Diseases Group, Madrid, Spain; 5Hospital La Paz Institute for Health Research – IdiPAZ, Dyslipidemias of Genetic Origin and Metabolic Diseases Group, Madrid, Spain; 6La Paz University Hospital, Paediatric Haemato-Oncoloy Department, Madrid, Spain; 7Hospital La Paz Institute for Health Research – IdiPAZ, Pediatric Onco-Hematology, Clinical Research Unit, Madrid, Spain; 8Hospital La Paz Institute for Health Research – IdiPAZ, Experimental therapies and biomarkers in Cancer Group, Madrid, Spain

Background/Objectives: The traditional approach in bioinformatics for high throughput sequencing studies separates germline and somatic variants in oncology patients. However, some patients may have both types of variants, needing optimization of analytical approaches.

This study aims to create, optimize, and validate a unified bioinformatics pipeline for efficiently characterizing germline and mosaic variants in pediatric cancer patients.

Methods: Common steps were merged from two independent pipelines to enhance time performance, sensitivity, and accuracy. Analyses were conducted using NIST reference samples and patient samples with validated variants.

Results: - The unified pipeline reduced storage and processing time, considering the impact of the reference genome and aligner algorithms.

- Maintaining distinct subprocesses for each variant type ensured maximum quality values, validated objectively with reference samples for each variant type.

- The reduction of two different alignments for both pipelines to a common and faster one accelerated the processing of samples from 44 hours to 19 hours.

Conclusion: -The implementation of the new pipeline resulted in enhanced recall and precision, particularly in germline analysis. This improvement was achieved by switching to a more accurate aligner with shorter analysis times. In SNPs, recall increased from 93.57% to 97.14%, and in indels, it increased from 89.06% to 93.81%. Additionally, precision improved from 99.28% to 99.46% for SNPs and from 77.41% to 95.15% for indels.

- In somatic analysis, only the processing times were improved by switching to a similar but faster aligner.

Grants: Cris Cancer Foundation, PI21/01239, CA22/00002

Conflict of Interest: None declared

EP16.013 Investigating the molecular impact of obesity on celiac disease by RNA sequence analysis of intestinal mucosa

Yeliz Ekici 1, Selcuk Candan2;3, Ikbal Billur Canbakan3, Feyza Tuncer4

1Istanbul University, Graduate School of Health Sciences, Istanbul, Türkyie; 2Sancaktepe Prof.Dr.Ilhan Varank State Hospital, Department of Gastroenterology, Istanbul, Türkyie; 3Istanbul University-Cerrahpaşa, Department of Gastroenterology, Istanbul, Türkyie; 4IU Aziz Sancar Experimental Medicine Research Institute, Department of Genetics, Istanbul, Türkyie

Background/Objectives: Diagnosis of Celiac Disease (CD) may be delayed in overweight/obese patients due to the low incidence of gastrointestinal symptoms or their confusion with other disorders. The molecular mechanisms of CD is incomplete and obesity’s impact on CD is not examined, yet. We aimed to clarify these by RNA sequence analyses in duodenal biopsies of lean and overweight celiac patients and their CD-excluded controls.

Methods: RNA was isolated from duodenal biopsies of 23 volunteers, followed by RNA-sequencing with using Illumina Novaseq 6000. Functional pathway analysis of differentially expressed genes was performed to identify statistically significant pathways that were up- or down-regulated in lean patients compared to overweight patients.

Results: The differentially expressed genes were quantified as 534 and 137 among lean celiac vs. lean control and among overweight group comparisons, respectively (p < 0.05). Lean group results were linked to immune-system pathways, while overweight groups were linked to metabolic pathways.

Conclusion: In overweight patients with digestive problems, the diagnosis of CD may be overlooked due to low bowel inflammation. Our results suggest a significant change in the active pathways when obesity co-occured with CD. This might explain the difficulty in the diagnosis of these patients. There are no RNA-seq studies in the literature that include obese celiac patients. By increasing the number of overweight/obese volunteers, mechanisms that cause differences in low intestinal inflammation can be elucidated.

Grants: This work was supported by the Scientific Research Projects Coordination Unit of Istanbul University (TDK-2022-38888).

Conflict of Interest: None declared

EP16.014 FAIR Data Cube, a FAIR data infrastructure for integrated multi-omics data analysis

Xiaofeng Liao 1, Yuliia Orlova2, Cenna Doornbos1, Anna Niehues3, Casper de Visser1, Junda Huang1, Thomas Ederveen1, Purva Kulkarni1, Joeri Van Der Velde4, Morris Swertz4, Martin Brandt2, Alain van Gool1, Peter-Bram t Hoen1

1Radboud University Medical Center, Nijmegen, Netherlands; 2SURF, Amsterdam, Netherlands; 3Leiden University Medical Center (LUMC), Leiden, Netherlands; 4University Medical Center Groningen, Groningen, Netherlands

Background/Objectives: Integrative analysis of -omics data is crucial to advance our understanding of health and disease. However, such analyses are challenging due to the lack of findability, accessibility, interoperability, and reusability (FAIR) of -omics data and metadata. The storage of human -omics data (for example the WGS and WES data) in secure silos, for privacy reasons, further complicates their reuse and asks for privacy-preserving solutions.

To address these issues, the X-omics initiative introduces the FAIR Data Cube (FDCube), a multi-omics data infrastructure that facilitates federated data analyses and data FAIRification.

Methods: The FDCube infrastructure allows dataset owners to register data on the FAIR Data Point (FDP). It incorporates the FAIR Data Station, a metadata capture platform that facilitates making data FAIR at the source. Using the Investigation-Study-Assay (ISA) metadata schema, metadata is transformed into a FAIR machine-actionable resource stored in an RDF triplestore.

Researchers can exploit the FDCube to find datasets and initiate computation requests to

dataset owners. These federated analysis requests are executed on the respective datasets through Vantage6, and the results are communicated back.

Results: The FDCube was demonstrated using the Trusted World of Corona (TWOC) project showing its FAIRification and federated data analysis capabilities. Furthermore, the FDCube is now catalogued as an SURF Research Cloud item.

Conclusion: The FDCube provides a solution to the FAIR challenges in multi-omics data. Simultaneously, it offers a federated data analysis mechanism, enhancing the usability of -omics data for securely stored health data.

Grants: The Netherlands X-omics Initiative (184.034.019).

Conflict of Interest: None declared

EP16.015 Genetic-Clinical data interconnectivity: The weight of genomic data

Albert Ferran Martin1, Anna Subirats2, Joana Fortuño2, Beatriz Rey-Fernández 2, Rebeca Blanco2, Jaume Torrents3, Albert Torrents3, Cristina Camprubí2, Manel Flores2, Irina Royo3

1Reference Laboratory S.A., Bioinformatics Department, Barcelona, Spain; 2Reference Laboratory S.A., Molecular Biology Department, Barcelona, Spain; 3Reference Laboratory S.A., Barcelona, Spain

Background/Objectives: The increased computing power and cost-effectiveness of genetic and clinical big data storage has enabled greater interconnectivity between genetic, clinical, and demographic information. Next Generation Sequencing (NGS) databases make it easier to interconnect the information from thousands of variants with thousands of patients’ data, permitting better understanding of the variants as well as narrowing-down their classification and improving clinical diagnosis.

Methods: Information from genetic databases was analysed by filtering and selecting pathogenic and probably pathogenic variants overexpressed in specific cohorts.

Results: The interconnectivity of this data has shown some interesting discoveries, such as: 1) Example of a possible founder effect: variants p.(Val905Gly) in GLDC and p.(Asp778Glu) in MYH7 were strongly established in the population of Colombia and Melilla, being the main variants associated with Glycine encephalopathy and Cardiomyopathy, respectively. 2) Improved variant reclassification: p.(Tyr400_Phe402del) in LDLR is in pathogenicity conflicts according to the scientific literature. It has been recurrently found in patients with hypercholesterolemia, so its pathogenicity would be increased. 3) Improved differential diagnosis: Variant p.(Met390Arg) in BBS1 is associated with Bardet-Bield syndrome. This variant was identified in a patient with clinical suspicion of retinitis pigmentosa. This information facilitated a conclusive diagnosis.

Conclusion: Working with curated, updated and enriched database, is a great advantage, as it allows to improve variant classification, work out the genomic configuration, and detect possible founder effects. This information can be of utmost importance in establishing the possible implication of the detected variants in the patient’s clinical condition and pointing out a diagnosis.

Conflict of Interest: None declared

EP16.016 Comprehensive evaluation of quantification methods for tissue or cell types of origin of the plasma cell-free transcriptome

Tingyu Yang 1;2, Wen-Jing Wang2, Yulong Qin1;2, shuo yan1;2, Jinghua Sun2, Qing Zhou2

1University of the Chinese Academy of Sciences, Beijing, China; 2BGI Research, Shenzhen, China

Background/Objectives: Cell-free RNA (cfRNA) in plasma originates from cells of various tissues and organs. It varies with physiological and pathological conditions, manifesting as quantitative changes in the type of tissue or cell of origin affected. Although multiple methods can be used for quantification of tissue or cell types of origin of the cfRNA, comparative evaluation is still lacking.

Methods: We used bulk RNA sequencing data and single-cell RNA sequencing data as references for tissues and cells, and applied various quantification methods to calculate the contributing tissue or cell types of origin of the cfRNA, including regression-based, deep learning-based, and gene signatures-based tools. We analyzed the differences between results of tissue and cell types of origin of the plasma cfRNA, and compared the performance of these tools on several cfRNA datasets.

Results: We observed more reliable results from the contributing cell types of origin than tissues of origin. Among different tools, Deconformer demonstrated the lowest average coefficient of variation and the highest intragroup sample correlation. Moreover, most tools were able to identify changes in the contribution of disease-affected cells. Across various datasets and diseases, the origin quantification results from CibersortX and Deconformer were relatively more effective for sample classification.

Conclusion: This study provides an important reference for analysis of tissue or cell types of origin of the plasma cell-free transcriptome, promoting further application of the cfRNA.

Grants: The Science, Technology and Innovation Commission of Shenzhen Municipality (No.JCYJ20170412152854656).

Conflict of Interest: None declared

EP16.017 Integrated CNV Detection in Clinical Exomes: A Comprehensive Analysis Using NGS data and Validation Approaches

Rimvydas Jonikas 1, Marius Šukys1;2, Zivile Zemeckiene1;2, Rasa Ugenskiene1;2

1Hospital of Lithuanian University of Health Sciences, Genetics and Molecular medicine, Kaunas, Lithuania; 2Medical Academy, Lithuanian University of Health Sciences, Genetics and Molecular medicine, Kaunas, Lithuania

Background/Objectives: Detecting copy number variants (CNVs) is crucial in clinical applications. Gold standards for CNV identification include microarray comparative genomic hybridization (aCGH) and multiplex ligation-dependent probe amplification (MLPA), known for their costliness and time consumption. While next-generation sequencing (NGS) is a standardized approach for genetic variants, precise CNV detection from clinical exomes remains challenging.

Methods: We developed a CNV calling tool using whole exome NGS data. Target regions were segmented into 100 bp fragments, and average coverages were obtained with mosdepth. Raw read coverage for each region was normalized based on GC%, region mappability, and mean total sample coverage. Normalized values were compared to batch mean for the region, and Log2 was used to convert ratios to copy numbers. Based on coefficient of variance and Z-score high variability regions were filtered out, aiding in discerning actual CNV calls.

Results: We analyzed approximately 1000 whole exome NGS data, detecting 37 clinically relevant CNVs. 15 CNVs were deletions involving single gene. Nine of those deletions were successfully validated by MLPA or real-time PCR methods. 1 MLPA result was inconsistent due to probe hybridization coinciding with an 11 bp deletion. 5 CNVs did not undergo validation testing. 24 CNVs covered multiple genes. 10 of those were selected for validation and successfully confirmed by microarray analysis.

Conclusion: This novel CNV analysis algorithm holds promise for routine diagnostic purposes, providing single nucleotide variant and CNV detection in a single analysis. This approach has the potential to reduce costs and expedite the diagnostic time for patients.

Conflict of Interest: None declared

EP16.018 Benchmarking pathogenicity prediction scores for missense variants with a focus on clinical applications

T. madhusankha Alawathurage1, Kerstin Ludwig1, Axel Schmidt 1;2

1Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany; 2Department of Pediatric Neurology, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany

Background/Objectives: In exome/genome sequencing, missense variants are frequently detected as potential causes of monogenic diseases in clinical and research applications. Classifying the pathogenicity of this type of variation is particularly challenging, as only a minority of identified missense variants is truly pathogenic. Recently, a number of novel pathogenicity prediction scores for missense variants have been developed. Still, it remains unclear how these novel tools behave in clinical diagnostics settings.

Methods: In this project, we evaluate the properties of novel pathogenicity prediction scores (among others AlphaMissense, PrimateAI-3D and CADD 1.7) in clinical settings. We first apply classical benchmarking approaches, such as correlation analyses with experimental scores from deep mutational scans (DMS) or their performance in classifying ClinVar variants. We will also simulate real-life gene panel analyses, under variable gene panel sizes and compositions as well as under varying expected diagnostic yields. We will also address the performance with relation to differences in population backgrounds. For this purpose, pathogenic variants from ClinVar will be spiked into data from the 1000 genomes and the human genome diversity project.

Results: First results of the classical approach demonstrated that AlphaMissense in particular correlated better with scores from DMS experiments than PrimateAI-3D or REVEL. Further analyses are ongoing and will be presented at the conference.

Conclusion: Our analyses will provide insights into the behavior of pathogenicity predictors in real-world applications to facilitate their use and interpretation.

Grants: BONFOR

Conflict of Interest: T. madhusankha Alawathurage: None declared, Kerstin Ludwig: None declared, Axel Schmidt Research Scholarship of the University of Bonn (BONFOR program), University and University Hospital Bonn

EP16.019 bioGWAS: a Simple and Flexible Tool for Simulating GWAS Datasets

Anton Changalidis 1, Yulia Nasykhova1, Andrey Glotov1, Yury Barbitoff1

1D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Department of genomic medicine, Saint Petersburg, Russian Federation

Background/Objectives: Genome-wide association studies (GWAS) are crucial for identifying genes, loci, and biological processes related to complex traits. However, development of tools for annotation and interpretation of GWAS findings requires datasets with predefined biological ground truth. Simulating GWAS results can address the need for such datasets for both benchmarking of tools and educational use. Still, there are no software that could simulate datasets with predefined sets of causal genetic variants and/or molecular pathways.

Methods: We introduce bioGWAS, a flexible Snakemake pipeline that utilizes a range of existing bioinformatics tools (PLINK, HAPGEN2, bedtools, and PhenotypeSimulator) to generate realistic genotypes (with predefined causal genetic variants or molecular pathways), phenotypes, and summary statistics of their association.

Results: Following the implementation of bioGWAS pipeline, we made an effort to select a set of default simulation parameters by evaluating precision and recall in detecting associations between user-defined causal genetic variants and the simulated phenotype. When the optimal parameters were used, bioGWAS yielded an average F1-score exceeding 0.95, indicating a high accuracy of simulation results. Further validation through the simulation of 120 datasets with causal variants drawn from specific biological pathways demonstrated the method’s effectiveness for benchmarking tools for GWAS results annotation.

Conclusion: bioGWAS offers a significant advancement in simulating biologically meaningful GWAS datasets, with potential applications in method development, bioinformatics software testing, and educational settings.

Grants: Supported by the Ministry of Science and Higher Education of the Russian Federation (contract 075-15-2021-1058 from 28 September 2021).

Conflict of Interest: None declared

EP16.022 Insights into the variant landscape of polycystic kidney disease in Ireland using haplotype-based approaches

Ashwini Shanmugam 1;2;3, Sophia Heneghan1;2;3, Kane Collins1;2;3, Elhussein Elhassan4;5, Omri Teltsh1;3, Sahin Sarihan1;6, Russell L. McLaughlin2;3;7, Ross Byrne7, Gianpiero Cavalleri1;2;3, Peter Conlon4;6, Katherine Benson1;3, Edmund Gilbert1;3

1Royal College of Surgeons in Ireland, School of Pharmacy and Biomolecular Sciences, Dublin, Ireland; 2University of Galway, The SFI Centre for Research Training in Genomics Data Science, School of Mathematics, Statistics and Applied Mathematics, Galway, Ireland; 3The SFI FutureNeuro Research Centre, Dublin, Ireland; 4Royal College of Surgeons in Ireland, Ireland; 5Beaumont Hospital, Department of Nephrology and Transplantation, Dublin, Ireland; 6Beaumont Hospital, Dublin, Ireland; 7Trinity College Dublin, Complex Trait Genomics Laboratory, Smurfit Institute of Genetics, School of Genetics and Microbiology, Dublin, Ireland

Background/Objectives: Polycystic Kidney Disease (PKD) is the most common Mendelian renal disorder and a leading cause of end-stage kidney disease. It is commonly assumed that pathogenic PKD variants are recent in age and that carriers of the same variant share a recent common ancestor, carrying the variant on the same ancestral haplotype. These carriers of the same variant indeed share the same haplotype, and haplotype-based prediction could be used to identify undiagnosed patients. Targeted sequencing could then be applied to confirm suspected cases, reducing costs and time-to-diagnosis.

Methods: Using genotyping and whole-exome sequencing data from a cohort of Irish PKD patients (n = 380), we utilised population genetics approaches to (1) confirm that clusters of individuals who share a common diagnostic variant also share the same haplotype across that region, (2) predict putative carriers of observed diagnostic variants though common sharing of identified “PKD haplotypes” and, (3) investigate whether these PKD haplotypes can be used to predict diagnostic variants in an external dataset such as the UK Biobank or Genomics England (GEL).

Results: Analyses are ongoing.

Conclusion: Through these approaches, we aim to confirm if the Irish and British PKD variants identified are typically founder variants and explore the utility of haplotype analysis in diagnostic prediction. Additionally, we intend to identify potential carriers of these variants and investigate if they segregate by geography within Ireland. This work highlights a potential new avenue for cost-effective clinical genetics, as well as characterising the haplotype and variant landscape of PKD in Ireland.

Grant SFI 18/CRT/214 and 16/RC/3948

Conflict of Interest: Ashwini Shanmugam Science Foundation Ireland 18/CRT/214 and 16/RC/3948, Sophia Heneghan Science Foundation Ireland 18/CRT/214, Kane Collins Science Foundation Ireland 18/CRT/214, Elhussein Elhassan: None declared, Omri Teltsh: None declared, Sahin Sarihan: None declared, Russell L. McLaughlin: None declared, Ross Byrne: None declared, Gianpiero Cavalleri: None declared, Peter Conlon: None declared, Katherine Benson: None declared, Edmund Gilbert: None declared

EP16.023 DATOS.CAT Project: Transformation of population based cohorts to the standardised format of Observational Medical Outcomes Partnership Common Data Model (OMOP-CDM) and to the European Genome-Phenome Archive (EGA)

Aikaterini Lymperidou1;2, Judith Martinez-Gonzalez2;3, Guillem Bracons Cucó2, Xavier Escribà-Montagut4, Marta Huertas2;5, Ramon Mateo-Navarro2;4, David Sarrat-González4, Santiago Frid6, Juan Ramón González4, Alberto Labarga3, Susana Iraola-Guzmán 1, Rafael de Cid1

1Genomes for Life- GCAT lab- Germans Trias i Pujol Research Institute, Badalona, Spain; 2Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain; 3Barcelona Supercomputing Center, Barcelona, Spain; 4Barcelona Institute for Global Health, Barcelona, Spain; 5Centre for Genomic Regulation, Barcelona, Spain; 6Hospital Clínic de Barcelona, Barcelona, Spain

Background/Objectives: The project DATOS-CAT aims to increase the visibility and scientific impact of the two population-based cohorts, GCAT-Genomes for life and COVICAT-CONTENT, improving the level of interoperability of their data in the context of the FAIR data principles (Findable, Accessible, Interoperable, Reusables) to facilitate their exploitation and scientific use. The Common Data Model (CDM) of Observational Medical Outcomes Partnership (OMOP) will be used for working with structured clinical and genomic data and the European Genome-Phenome Archive (EGA) infrastructure will be used for genomic data.

Methods: The GCAT and the COVICAT-CONTENT have been following 20.000 and 11.000 individuals, respectively, living in Catalonia. Extensive clinical, genomic and lifestyle data has been collected to investigate the role of the environmental and genetic factors (i.e., genomic, metabolomic, proteomic, epigenomic) in the development of chronic diseases and COVID-19. In order to assess adherence to the FAIR principles, OMOP-CDM is intended to be used by applying two different Extract-Transform-Load (ETL) processes: a traditional and a semantic one. ETL plays an important role in ensuring data consistency and integrity through the integration process, enabling a smooth transition from heterogeneous datasets to a coherent standardized structure.

Results: DATOS-CAT will examine the adaptability, efficiency and scalability of each ETL approach in achieving data consistency and integrity.

Conclusion: The efficient integration into standardized formats empowers researchers to access direct sources of information, enabling a deeper understanding of how genetic profiles relate to clinical outcomes and influence health care practices around the world, resulting in a greater benefit for society.

Conflict of Interest: None declared

EP16.025 Validating RNA-fusion detection in cancer diagnostics using nf-core/rnafusion

Eva Caceres1;2, Annick Renevey 1, Anders Jemt1;2, Yingbo Lin3, Ida Lindegaard1, Linnea La Fleur1, Susanne Månér1, Felix Haglund3;4, Henrik Stranneheim1;2;5, Anna Lyander1;6, Valtteri Wirta1;2;6

1Science for Life Laboratory, Karolinska Institutet, Department of Microbiology, Tumour and Cell Biology, Solna, Sweden; 2Karolinska University Hospital, Genomic Medicine Center Karolinska, Solna, Sweden; 3Karolinska Institutet, Department of Oncology-Pathology, Stockholm, Sweden; 4Karolinska University Hospital, Pathology and Cancer Diagnostics, Solna, Sweden; 5Karolinska University Hospital, Centre for Inherited Metabolic Diseases, Solna, Sweden; 6Science for Life Laboratory, KTH Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology and Health, Stockholm, Sweden

Background/Objectives: Gene fusions play an important role in the oncogenesis and progression of many tumors. Being able to accurately detect fusions can inform diagnostic, prognostic and therapy selection. Unlike traditional methods, whole transcriptome sequencing allows for comprehensive detection of both known and novel fusions. However, the nature of the data makes fusion detection challenging.

With healthcare moving towards large-scale sequencing and precision oncology, there is a need for robust, scalable and portable pipelines that can operate in clinical settings. The nf-core/rnafusion pipeline (https://nf-co.re/rnafusion) has been developed with this in mind and combines the results from different fusion-detection tools to increase the confidence of the calls and aid in their interpretation.

Here we describe our efforts to validate a complete workflow for RNA fusion detection.

Methods: One commercial and multiple clinical samples with previously identified gene fusions were used, covering a range of expression and heterogeneity levels, varied sample qualities (wide range of RIN values), and representing different cancer types. The robustness of the library preparation (Illumina Stranded mRNA prep) and the sensitivity as well as the performance of the pipeline were evaluated. The bioinformatic pipeline integrates the following callers: Arriba, FusionCatcher, and STAR-Fusion.

Results: In the commercial sample 19/19 fusions were detected, but one exon-skipping event was not called, demonstrating the need to address these events separately. Ongoing analyses show that integrating results from multiple fusion callers considerably reduces the number of false positives, with Arriba and STAR-Fusion being the most sensitive callers.

Conflict of Interest: None declared

EP16.026 Matched Annotation from NCBI and EMBL-EBI (MANE) in 2024

Adam Frankish1, Jane Loveland1, Shashikant Pujar2, Alex Astashyn2, Ruth Bennett1, Andrew Berry1, Eric Cox2, Claire Davidson1, Olga Ermolaeva2, Catherine Farrell2, Reham Fatima1, Tamara Goldfarb2, Jose M Gonzalez1, Diana Haddad2, Matt Hardy1, Toby Hunt 1, John Jackson2, Vinita Joardar2, Michael Kay1, Vamsi Kodali2, Kelly McGarvey2, Jonathan Mudge1, Michael Murphy2, Sanjida Rangwala2, Francoise Thibaud-Nissen2, Anjana Vatsan2, Craig Wallin2, David Webb2, Terence Murphy2

1European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom; 2National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, United States

The Matched Annotation from NCBI and EMBL-EBI (MANE) collaboration between Ensembl/GENCODE and RefSeq has defined a set of transcripts and corresponding proteins for use as a universal standard for variant reporting. RefSeq and Ensembl/GENCODE MANE transcripts are exact matches and their identifiers may be used synonymously. The MANE Select set defines a representative transcript for each human protein-coding gene with the MANE Plus Clinical set providing additional transcripts at loci where the Select alone is not sufficient to report all currently known clinical variants. MANE release 1.2 has MANE Select transcripts for 99.4% of human protein-coding genes, including genes located on GRCh38 patches.

The MANE collaboration continues with the resolution of all remaining annotatable MANE Select transcripts, the addition of new MANE Plus Clinical transcripts, the ongoing expansion of MANE Select into clinically relevant non-coding loci and the integration of MANE into the human reference pangenome. The latter raising significant challenges given the importance of anchoring MANE transcripts to GRCh38.

Please give feedback at mane-help@ebi.ac.uk or MANE-help@ncbi.nlm.nih.gov to help us ensure that the MANE collaboration provides maximum benefit to users.

This work is supported by: Wellcome Trust-WT226083/Z/22/Z; EMBL-Core-Funds; NIH-U41HG007234. This work was supported in part by the National Center for Biotechnology Information of the National Library of Medicine (NLM), National Institutes of Health (NIH).

Conflict of Interest: None declared

EP16.027 The physiological role of T-cadherin in Pulmonary Endothelial Cells

Mikhail Arbatskiy1;2, Veronika Sysoeva2, Polina Klimovich2, Kamilla Yamileva3, Ekaterina Semina3, Kseniya Rubina 3

1MSU, Russian Federation; 2Moscow State University, Faculty of Medicine, Moskva, Russian Federation; 3Moscow State University, Moskva, Russian Federation

The physiological role of T-cadherin in Pulmonary Endothelial Cells

M. Arbatskiy, V. Sysoeva, Semina E.V., Klimovich P.S., Yamileva K.I., Rubina K.A.

Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia

Background/Objectives: Previously published data and the results of our own research suggest the significant role of T-cadherin in endothelial function under normal conditions and in pathology (https://doi.org/10.1016/j.ejcb.2021.151183).

Methods: We first utilized the GSE164829 series for bioinformatic analysis. Data analysis was conducted using Seurat. Cell type annotation was performed using the automatic cell type identification tool - SingleR.

To identify cellular targets and signaling partners of T-cadherin cascades, we utilized STRING.

Total RNA was reverse-transcribed to generate cDNA, followed by real-time polymerase chain reaction (PCR). Primer design was carried out using NCBI Primer-BLAST.

Results: Bioinformatic analysis yielded 5 cell clusters expressing CDH13 (T-cadherin). Co-expression of CDH13, CDH5, and CTNNB1 genes was detected in all clusters.

Construction of protein-protein interaction maps using the STRING.ORG web platform revealed potential protein partners of T-cadherin, including CTNNB1, CDH5, CDH2, CAV1, BRCA1, DNMT1, SRFP1, and ADIPOR1.

The bioinformatic findings were validated through RT-PCR analysis. Total RNA was extracted from the human endothelial cells Eahy926 (cells overexpressing T-cadherin or control crlls). RT-PCR confirmed the expression of target genes identified by bioinformatics. Endothelial cells overexpressing T-cadherin exhibited reduced mRNA levels of β-catenin, VE-cadherin, N-cadherin, CAV1, BRCA1, SRFP1, and AdipoR2.

Grant References: The study was supported by the Russian Science Foundation, grant No. 19-75-30007, and under the State Assignment # 03р-23/110-03 of Lomonosov MSU.

Conflict of Interest: None declared

EP16.028 SpliceAI-visual – a valuable prediction tool for inconclusive PMS2 short intronic variants

Catalin-Vasile Munteanu 1;2, Adela Chirita-Emandi1;2, Maria Puiu1;2, Catalin Valer Marian2;3, Adrian Trifa2;3

1Louis Turcanu, Clinical Emergency Hospital for Children, Medical Genetics, Timisoara, Romania; 2Victor Babes University of Medicine and Pharmacy, Timisoara, Romania; 3Victor Babes Clinical Hospital of Infectious Diseases and Pneumophysiology, Timisoara, Romania

Background/Objectives: In spite of the constant improvements in DNA sequencing techniques in the last years, germline testing of PMS2 gene remains a challenge in the current clinical practice from various reasons. Clinically significant variants in PMS2 gene are reported in about 15% of individuals with Lynch syndrome (LS). However, this percentage is recently acknowledged as a significant underestimation, prompting the need to increase the diagnostic rate.

Methods: We collected all PMS2 intronic variants available in ClinVar database on 05 January 2024. Structural variants (50 base pairs or greater), point mutations and class 4-5 ACMG variants were filtered out. We annotated the filtered variants using SpliceAI-500 nt. Simultaneously, we conducted a manual analysis for each variant using SpliceAI-visual and compared the outcomes with the standard Delta score (DS) strategy.

Results: Among the 838 intronic variants documented in ClinVar, we analyzed 61 short genetic variants. 14 variants were predicted to have a potential splicing impact when we used a combined in silico approach. Remarkably, SpliceAI-visual identified 4 variants (classified as likely benign, uncertain or conflicting) with potential disruptive effects, that might be otherwise dismissed or underestimated when relying solely on DS. To date, there are no available functional studies for any of these variants.

Conclusion: Our analysis aligns with existing literature, emphasizing the utility of SpliceAI-visual predictions. The combined in silico approach holds promise for improving the diagnostic rate in PMS2-related LS. Furthermore, this strategy facilitates the variant filtering process, ensuring that only relevant variants are selected for RNA sequencing.

Grants:

Conflict of Interest: None declared

EP16.029 AI4NEF: a tool to support clinical management and genotype-phenotype dissection of patients with Neurofibromatosis type 1

Mariateresa Zanobio 1, Antonella Farina1, Claudia Santoro2, Marina Melone3, Silverio Perrotta2, Marco Benedetto4, Anna Biondi4, Stefano Tagliaferri4, Annalaura Torella1;5, Vincenzo Vincenzo Nigro1;5, Giulio Piluso1

1University of Campania L. Vanvitelli, Medicine of Precision, Naples, Italy; 2University of Campania L. Vanvitelli, Department of Women, Children and General and Specialist Surgery, Naples, Italy; 3University of Campania L. Vanvitelli, Department of Advanced Medical and Surgical Sciences, Naples, Italy; 4Kelyon s.r.l., Naples, Italy; 5Telethon Institute of Genetics and Medicine, Pozzuoli (NA), Italy

Background/Objectives: Neurofibromatosis type 1 (NF1) is a multisystemic autosomal dominant disorder caused by mutations in NF1, a tumour suppressor gene encoding neurofibromin. NF1 is characterised by predisposition to tumours, as cutaneous or plexiform neurofibromas (PNFs), benign peripheral nerve sheath tumours. Malignant evolution of pre-existing PNFs could be possible, worsening disease prognosis. NF1 has a complete penetrance with an age-dependent clinical manifestations and a wide inter-/intra-familiar phenotypic variability. In this scenario, the identification of novel genotype–phenotype correlations and genetic modifiers is an emergency. To this aim we developed AI4NEF platform, a web-based registry of NF1 patients’.

Methods and Results: For this pilot study we selected 250 patients from Campania Referral Centre for Neurofibromatosis, collected and integrated their genetic data (from WES analysis) and clinical features including lifestyle. Artificial Intelligence and Machine Learning applications was used to identify the most appropriated cohort clustering methods, which allowed us to stratify our patients in three groups, from a more severe to a less severe phenotype, in accordance with the already known literature evidence. Due to the high allelic heterogeneity, we also recognized an intermediate phenotype group that could represent the candidate to unravel novel genotype–phenotype correlations and genetic modifiers. In the next future, the established collaboration with other Italian centres should ensure the cohort expansion.

Conclusion: We believe that this integrated platform will be able to improve clinical patients’ management identifying eventual prognostic factors as well as to increase our knowledge about NF1 disease, revealing novel genotype-phenotype correlations and potential genetic modifiers.

Conflict of Interest: None declared

EP16.030 Molecular Structures of Orthologous Transcripts: to Full-Length T-Cadherin and T-Cadherin Lacking the 3rd Exon in Mice and Humans

Mikhail Arbatskiy 1, Veronika Sysoeva1, Ekaterina Semina1, Kseniya Rubina1

1Moscow State University, Moskva, Russian Federation

Molecular Structures of Orthologous Transcripts: to Full-Length T-Cadherin and T-Cadherin Lacking the 3rd Exon in Mice and Humans

M. Arbatskiy, V. Sysoeva, E. Semina, K. Rubina

Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia

Background/Objectives: We previously generated Cdh13∆Exon3 mice lacking exon 3 in the Cdh13 gene (T-cadherin) and described their phenotype. While these mice exhibited normal gross morphology, their body weight was lower than of WT mice. Upon physical training the systolic blood pressure was significantly elevated. The plasma adiponectin and the AMPK phosphorylation level in skeletal muscles and myocardium of Cdh13∆Exon3 mice were elevated. Herein, we investigated the relevance of the truncated form of murine T-cadherin in the context of human T-cadherin.

Methods: To predict the protein folding, we applied the ColabFold. The best model was refined by relaxation based on the pLDDT using AMBER.

Results: Comparative analysis of the truncated and full-length mouse T-cadherin revealed the presence of a prodomain in the full-length T-cadherin protein and its absence in the truncated form T-cadherin lacking the 3rd exon. The structure with the highest cumulative pLDDT score was selected.

Conclusion: Further investigations are needed to elucidate the underlying mechanisms of physiological effects of truncated T-cadherin, focusing on interactions between T-cadherin and its ligands.

Grant References: The study was supported by the Russian Science Foundation, grant No. 23-11-00205, under the State Assignment # 03р-23/110-03 of Lomonosov MSU.

Conflict of Interest: None declared

EP16.032 HIGESS - an evidence-based scoring tool for the identification and prioritization of novel candidate genes in Hirschsprung Disease

Alexander Schulz1, Dominik Seelow2, Denise Dawid1, Tanja Mederer1, Ralph Röth1;3, Patrick Günther3;4, Nagarajan Paramasivam3;5, Daniel Hübschmann3;5;6, Beate Niesler1;3, Gudrun Rappold1, Philipp Romero3;4, Stefanie Schmitteckert 1;3

1Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany; 2Institute of Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, Berlin, Germany; 3ICGH – Interdisciplinary Center for Gut Health, 3R Center, Heidelberg University Hospital, Heidelberg, Germany; 4Pediatric Surgery Division, Heidelberg University Hospital, Heidelberg, Germany; 5Computational Oncology Group, Molecular Precision Oncology Program, NCT Heidelberg, DKFZ Heidelberg, Heidelberg, Germany; 6Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM), Pattern Recognition and Digital Medicine, Heidelberg, Germany

Background/Objectives: A major challenge in heterogeneous and genetically complex diseases is identifying genes relevant to disease etiology. For the enteric neuropathy Hirschsprung Disease (HSCR), about 25 risk loci have been identified, which account only for a minority of patients. Despite intensive research the complete understanding of underlying pathomechanisms remains incomplete. To understand the complex genetics of HSCR, the identification of further risk loci is indispensable.

Methods: As currently no tool is available that can reliably and quickly identify disease-relevant genes, the Hirschsprung Disease Gene Scoring System HIGESS was developed.

Entered genes are rated according to their likelihood of being involved in the pathogenesis of HSCR. Therefore, HIGESS applies four categories requesting expression, disease association, function and phenotype association of each gene with regard to HSCR. Predetermined thresholds allow for scoring and rating.

Results: To prove its suitability, we applied HIGESS to the 25 known HSCR genes, among others. 18 HSCR genes received relatively high scores, while seven were rated with low or no impact. As HIGESS is evidence- and knowledge-based, the applied rating is only as good as the information available in respective databases at the time of the query. However, due to regular updates of HIGESS, the datasets used are dynamically growing.

Conclusion: Prioritization with HIGESS can facilitate the search for novel candidate genes in a reliable and time-saving way. Identified genes can be examined more closely, which will secondarily support our understanding of the underlying pathomechanisms and the development of alternative therapies in the future.

Grants: not applicable

Conflict of Interest: None declared

EP16.033 Precision genome analysis: unraveling SNVs and CNVs with a multi-variant caller WGS workflow

Marta Ferreira 1;2;3, Celina São José1, francisco almeida1;3, joaquin jurado maqueda3, ana rita monteiro4, Pedro Ferreira2;5, carla oliveira1;3;6

1i3S - Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal; 2FCUP Department of Computer Sciences, Porto, Portugal; 3IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal; 4Inovretail, SA, Maia, Portugal; 5INESC TEC - Institute for Systems and Computer Engineering, Technology and Science, Laboratory of Artificial Intelligence and Decision Support, Porto, Portugal; 6Faculdade de Medicina da Universidade do Porto - FMUP, Porto, Portugal

Background/Objectives: Genome-wide analysis emerges as a solution to understand genome variation, with Whole Genome Sequencing (WGS) being the preferred method for this purpose. We aimed at developing a WGS analysis workflow using a combination of multiple variant callers for SNV, CNV and SV calling.

Methods: We used a gold standard sample from the Genome in a Bottle project (GIAB) to access the performance of a novel pipeline encompassing alignment (BWA mem); post-processing (GATK tools); CNV (LUMPY, Delly and GRIDSS) or SNV (HaplotypeCaller-GATK (HC) and DeepVariant (DV)) calling. Additionally, we performed merging of multiple called variants through overlap analysis of SNV, CNV, and SV calls. We calculated recall, precision and F1 scores by comparing our outputs with those from GIAB for SNVs. As no gold standard is available in GIAB for CNVs, we compared our CNV outputs with a GIAB pool of high confidence to improve our pipeline.

Results: We called 4.309.554 SNVs with DV and 4.578.886 with HC, and 4.109.099 were common. The performance of DV alone (F1 score = 0.9819, 3.802.474 true positives) was better than SNV calling with HC alone (F1 score = 0.9522, 3.759.774 true positives), but worse than the combination of DV and HC outputs (F1 score = 0.9841, 3.836.476 true positives variants). Our CNV calling pipeline called a higher number of variants than the GIAB, as well as a set of inversions confirmed by visualization in samplot.

Conclusion: Our WGS-pipeline shows high performance and improves the likelihood of finding true positive germline SNVs, captures high confidence CNVs and uniquely calls inversions.

Grants: 2020.05763.BD, GenomePT(22184), LEGOH_PTDC/BTM-TEC/6706/2020,SOLVE-RD_H2020-SC1-2017, PDCC

Conflict of Interest: None declared

EP16.034 Bioinformatics analysis of pathogenic and likely pathogenic SNVs in human homeotic genes

Iurii Sadovnychenko 1, Nataliia Pastukhova2, Sofiia Rozumenko1

1Kharkiv National Medical University, Department of Medical Biology, Kharkiv, Ukraine; 2Institute of Food Biotechnology and Genomics of National Academy of Sciences of Ukraine, Department of Genomics and Molecular Biotechnology, Kyiv, Ukraine

Background: Homeotic genes play a pivotal role during human embryonic development. Mutations in homeotic genes are associated with various types of cancer, disorders of the musculoskeletal and nervous systems, sensory organs, etc. The purpose of the study was to search for pathogenic and probably pathogenic single nucleotide variants (SNVs) in the homeotic genes HOXA13, HOXB13 and HOXD13 in silico.

Methods: The list of homeotic genes’ SNVs, gene and protein sequences were retrieved from the dbSNP and Uniprot databases. Bioinformatic tools fathmm, PANTHER, PhD-SNP, PolyPhen-2, SIFT, and SNPs&GO were used to estimate pathogenic effects of SNVs on the protein structure and functions. Pathogenic SNVs were determined according to five or six different predictors. Likely pathogenic SNVs were determined according to three or four different predictors.

Results: The minimum number of homeotic genes’ SNVs were identified by SIFT tool, and the maximum one was analyzed by the PANTHER and the PhD-SNP tools. Most pathogenic and likely pathogenic SNVs were found in the HOXB13 gene. It was identified 162 pathogenic SNVs in the HOXA13, HOXB13 and HOXD13 genes, which are not listed in bioinformatics databases as deleterious, most of them belonged to the HOXD13 gene. SNV density was highest in the HOXB13 gene. The distribution of SNVs across homeotic genes was uneven and associated with biological properties of certain domains of homeodomain proteins.

Conclusion: Targeted sequencing and bioinformatic analysis of SNVs in the HOXA13, HOXB13 and HOXD13 genes would help diagnosis of homeotic gene-related disorders.

Grants: Not applicable.

Conflict of Interest: None declared

EP16.035 Investigating established and non-established susceptibility genes in generalized pustular psoriasis

Mohammad Deen Hayatu 1, Rotraut Mößner2, Nina Magnolo3, Sandra Philipp4, Jörg C. Prinz5, Knut Schaekel6, Arif B Ekici1, Steffen Uebe1, Wiebke Sondermann7, Michael Sticherling8, Sascha Gerdes9, Dagmar Wilsmann-Theis10, Ulrike Hüffmeier1

1Institute of human genetics, University hospital Erlangen, FAU, Erlangen, Germany; 2Universitätsmedizin Göttingen,Georg-August-University Göttingen, Department of Dermatology, Göttingen, Germany; 3University of Münster, Department of Dermatology, Münster, Germany; 4University of Berlin, Department of Dermatology, Berlin, Germany; 5Ludwig-Maximilian University Munich, Department of Dermatology and Allergology, München, Germany; 6University of Heidelberg, Department of Dermatology, Heidelberg; 7University of Essen, Department of Dermatology, Essen, Germany; 8Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Department of Dermatology, Deutsches Zentrum Immuntherapie, Erlangen, Germany; 9University Hospital Schleswig- Holstein – Campus Kiel, Psoriasis Center, Department of Dermatology, Venereology and Allergology, Kiel, Germany; 10University Hospital Bonn, Department and Clinic for Dermatology and Allergology, Bonn, Germany

Background/Objectives: Generalized pustular psoriasis (GPP) is a rare severe subtype of pustular psoriasis characterised by sterile, neutrophil filled pustules. Main genetic factors are bi-allelic variants in IL36RN and MPO, while an association has also been described for i.a. AP1S3 and BTN3A3, the latter more recently to heterozygous truncating variants in 282 Chinese GPP patients.

Methods: We analysed 79 exomes of European GPP patients for truncating BTN3A3 variants and assessed the cumulative effect of disease-relevant variants per gene in IL36RN, MPO and AP1S3 in ours and published patient groups (≤269 European patients). Depending on the reported state of disease-variants, we compared allele or genotype frequencies to those of controls. In case of bi-allelic variants, we considered ~4.900 individuals in whom the allele dosage could be determined.

Results: We did not identify a single truncating BTN3A3 variant in 79 patients, although the overall allele-frequency of the most common variant is 0.7% in Europeans of gnomAD. Putative disease variants in AP1S3 were not associated with GPP.

Meta-analysis revealed significant effect sizes for IL36RN and MPO variants in carriers of mono-, but especially of bi-allelic variants (IL36RN: p = 9.03E-54; 334.2 [106.5-1599.2]; MPO: p = 1.06E07), as expected for a monogenic disease.

Conclusion: Our studies confirm a significant contribution of IL36RN and MPO variants on disease susceptibility. The frequency of homozygous, healthy carriers of risk alleles in BTN3A3 and AP1S3 in gnomAD and the variants’ overall allele frequencies of up to 4.5% in certain populations render an impact on GPP’s susceptibility less likely

Grants:

Conflict of Interest: None declared

EP16.036 Assessment of genetic copy number variations yield in Whole exome sequencing

Martin Georgiev 1;2, Kunka Kamenarova1;2, Darina Kachakova-Yordanova1;2, Neviana Ivanova1;2, Olga Beltcheva1;2, Kalina Mihova1;2, Valentina Peycheva1;2, Radka Kaneva1;2

1Medical University of Sofia, Molecular Medicine Center, Medical Chemistry and Biochemistry, Sofia, Bulgaria; 2Medical University of Sofia, Laboratory of genomic diagnostics, Medical Chemistry and Biochemistry, Sofia, Bulgaria

Background/Objectives: Discovering the genetic causes for diverse human disorders is challenging for molecular geneticists and bioinformaticians on a daily basis. The Next Generation Sequencing(NGS) approach is used to assess genetic variations in patients with different level of disease complication. Identification of Single Nucleotide Polymorphisms (SNPs)/indels occasionally is not enough for confirmation of the genetic cause, imposing the need for assessment of Copy Number Variants(CNVs). This study is aiming for calculating the diagnostic yield with a respect to the CNV analysis in exome panel.

Methods: Whole exome sequencing(WES) was performed for both research and diagnostic genetic testing across 506 patients. We operated on Illumina’s NovaSeq 6000 and DRAGEN Bio-IT platform on-premise for executing the DNA and CNV pipeline analyses. Minimum mapping quality(MAPQ) for alignments is set to 30 and Copy-Ratio(CR) over 1.2 and under 0.8 are the values for variant calling. ACMG criteria was used for evaluation of the variant pathogenicity.

Results: 381(381/506 = 75.3% yield) cases had a SNP/indel mutation detected. The remaining 125 samples were in need of additional CNVs assessment, 12 of which have a CNV observed and additionally validated with an independent method(MLPA, aCGH or rt-PCR). The overall yield from the WES cases is 77.7%, resulting in higher number of solved cases(increased with ~2.4%).

Conclusion: Regardless of using the NGS approach for WES panel for CNV calling over a large exons(>100bp), a Whole genome sequencing technique might be considered for a better mutation assessment with respect to large CNVs, covering also the intronic DNA regions.

Grants:D01-302/17.12.2021, D01-165/28.07.2022

Conflict of Interest: None declared

EP16.037 A Correlation Analysis of GEO Datasets in Parkinson’s Disease and Type 2 Diabetes Research

Rahel Feleke 1, Bethany Quinton2, winston lau2, Toby Andrew1, nikolas maniatis2

1Imperial College London, London, United Kingdom; 2University College London, London, United Kingdom

Background/Objectives: The Gene Expression Omnibus (GEO) database contains thousands of genomic data, including bulk RNA sequencing (RNA-Seq) data and is major resource for genomic research. These types of data are critical for understanding cell-specific differential gene expression in complex disorders such as Parkinson’s (PD) and Type 2 diabetes (T2D). Nevertheless, study heterogeneity makes meta-analysis harder, undermines reliable inference and reduces ability to draw meaningful conclusions.

Methods: To characterise and quantify such inconsistencies, we conducted differential gene expression analyses for 14 PD bulk RNA-seq datasets and 10 T2D bulk RNA-seq datasets. The data for each condition were ascertained for similar protocols and case/control study design. Genomic correlation coefficients were analysed using heatmaps, providing a comprehensive overview.

Results: Independent T2D datasets were observed to have a maximum Pearson correlation coefficient of 0.25 between genomic studies, while a maximum of 0.58 was observed for PD datasets. The analyses also indicated that datasets did not obviously cluster by instrument used, including sequencing platforms, but inclusion of other fundamental protocol variables such as snap-freezing samples, post-mortem interval (PMI) and RNA integrity number (RIN) values measurably affected sample clustering.

Conclusion: Our work highlights the necessity for rigorous dataset selection based on experimental design and comparable protocols. While potentially wide-scale inconsistencies between RNA seq datasets are observed, genomic co-expression analyses can be used to identify samples that demonstrate homogeneity. We also recommend protocol measures that need to be included by researchers for publicly released RNA-seq data to mitigate sample heterogeneity and confounding.

Grant: MR/X011070/1

Conflict of Interest: None declared

EP16.039 Exploring the relationship between Rheumatoid Factor and Gender in Rheumatoid Arthritis on transcriptomic level: Insights from a comprehensive analytical approach

Bianka Böllering 1

1University Medical Center Mainz, Institute of Human Genetics, Mainz, Germany

Intro: Rheumatoid arthritis (RA), a complex autoimmune disease, is associated with chronic inflammation of the synovial joints leading to progressive cartilage and bone loss. The etiology of RA, which primarily affects women between the ages of 35 and 55, includes genetic predisposition, epigenetic changes and environmental factors. Although rheumatoid factor (RF) is associated with RA, its diagnostic specificity remains uncertain. This study aims to investigate possible sex-specific differences between RF-positive and RF-negative patients at the transcriptome level to improve understanding of the development and diagnosis of the disease.

Method: RNA-seq will be used to investigate the differences of transcriptomic profiles from blood samples of male and female RA patients with and without RF. With the aim of identifying potential disease subgroups that correlate with phenotypic features such as the RF, normalized gene expression data will be used to apply state-of-the-art clustering approaches.

Results: Distinct transcriptomic regulation underlying biological processes of the potential disease subgroups will be analyzed to identify key drivers of individual disease development.

Conclusion: We believe that our study will provide new insights into the complexity of RA by identifying possible associations between the presence of RF and the sex of RA patients.

Conflict of Interest: None declared

EP16.040 Identification of common genes associated with development of resistance against tamoxifen and doxorubicin in MCF7 cells

Arzu Zeynep Karabay 1, Asli Koc1, Yalda Hekmatshoar2

1Ankara University Faculty of Pharmacy, Department of Biochemistry; 2Altinbas University Faculty of Medicine, Department of Medical Biology

Background/Objectives:As in other types of cancer, drug resistance to chemotherapy in breast cancer is among the main treatment failures.Many mechanisms have been proposed for chemotherapy resistance, and these mechanisms and the signaling pathways they are associated with are targets for the elimination of resistance.Tamoxifen, used in the treatment of breast cancer, is a hormone drug,while doxorubicin is an anthracycline derivative.

Methods:In this study,datasets using tamoxifen-resistant and doxorubicin-resistant MCF7 cells were selected from Gene Expression Omnibus (GEO).In each data set,differentially expressed genes with p ≤ 0.05 significance and logfc values ≥ 0.5 were selected and compared before determining PPI network and hub genes with STRING and Cytoscape.Hub genes were then analysed for their enrichments in GO terms and KEGG pathways.Hub genes were also analyzed with miRNet to determine interacting miRNAs.

Results:Comparison of two datasets revealed that 635 differently expressed genes were found to be commonly expressed in both tamoxifen and doxorubicin resistant MCF7 cells.Data analysis of these 635 genes by STRING and Cytoscape databases revealed ESR1,CCND1,H3C12,CDH1, PTEN,CREBBP,H4C6,STAT1,MAPK3 and SIRT1 as top 10 Hub genes.These hub genes were found to be enriched in GO biological processes including negative regulation of I-kappa B kinase/NF-kappa B signalling,negative regulation of intracellular signal transduction,regulation of protein phosphorylation and KEGG pathways including pathways in cancer,thyroid hormone signalling pathway and FoxO signalling pathway.Top 3 interacting miRNAs with hub genes were found to be hsa-mir-22-3p,hsa-mir-34a-5p and hsa-mir-16-5p.

Conclusion:Collectively,the 10 hub genes,their interacting signalling network and miRNAs may be targets for new therapies to reverse tamoxifen and doxorubicin resistance in breast cancer.

Conflict of Interest: None declared

EP16.041 Artificial intelligence-assisted reanalysis of targeted sequencing data by non-bioinformaticians

Radostina Valeva 1;2, Mariya Levkova1;2, Mari Hachmeriyan1;2, Milena Stoyanova1;2, Dinnar Yahya1;2, Lyudmila Angelova2

1University Hospital Sveta Marina, Laboratory of Medical genetics, Varna, Bulgaria; 2Medical University Varna, Department of Medical genetics, Varna, Bulgaria

Background/Objectives: The current application of artificial intelligence (AI) is rather limited, but it is expected to be more involved in bioinformatics analysis soon. Therefore, we aimed to analyze the performance of commercially available AI software and reanalyze previous targeted sequencing cases of patients.

Methods: We used ten VCF files - three patients were analyzed for 160 genes, and seven for 369 genes, associated with hereditary cancer syndromes. Two of the patients tested positive for pathogenic variants, while the rest were negative. The VCF files were uploaded to an AI software platform and sex, ethnicity, and phenotype were added to the patient’s description. Then we compared the reported variants with the ones received after the manual analysis of the data.

Results: The AI software successfully identified the two patients with the pathogenic variants – one in the MUTYH and another in the MSH2 gene. However, the former was classified as likely pathogenic by the AI, while the manual result was pathogenic. The software also reported at least one variant of uncertain significance (VUS) in each of the patients with negative findings. These VUSs were not included in the official reports, because they were not considered to be related to the patient’s phenotype.

Conclusion: AI software could be successfully applied to everyday clinical practice, especially by counselors who lack any bioinformatics background. This illustrates the great potential of AI to offer time- and cost-efficient data analysis.

Conflict of Interest: Radostina Valeva This study is financed by the European Union-NextGenerationEU, through the National Recovery and Resilience Plan of the Republic of Bulgaria, project № BG-RRP-2.004-0009-C02., Mariya Levkova This study is financed by the European Union-NextGenerationEU, through the National Recovery and Resilience Plan of the Republic of Bulgaria, project № BG-RRP-2.004-0009-C02., Mari Hachmeriyan This study is financed by the European Union-NextGenerationEU, through the National Recovery and Resilience Plan of the Republic of Bulgaria, project № BG-RRP-2.004-0009-C02., Milena Stoyanova: None declared, Dinnar Yahya: None declared, Lyudmila Angelova: None declared

EP16.042 Bayesian modeling in Transcriptomics: endometrium profile classification for personalised ART

Francesco Ranucci 1, Daria Marzanati1, Elisa Giacomini2, Luca Pagliardini2, Valeria Stella Vanni3, Nicoletta Barberis3, Enrico Papaleo3, Massimo Candiani3, Maria Giulia Scotti4, Dejan Lazarevic4, Giovanni Tonon4, Davide Dragani5, Laura Privitera3, Patrizia Rovere Querini6, Davide Gentilini1, Paola Viganò7

1University of Pavia, Brain and Behavioral Sciences, Pavia, Italy; 2IRCCS Ospedale San Raffaele Scientific Institute, Reproductive Sciences Laboratory, Obstetrics and Gynecology Unit, Milano, Italy; 3IRCCS Ospedale San Raffaele Scientific Institute, Department of Obstetrics and Gynecology, Milano, Italy; 4IRCCS Ospedale San Raffaele Scientific Institute, Center for Omics Sciences, Milano, Italy; 5Istituto Auxologico Italiano IRCCS, Bioinformatics and Statistical Genomics Unit, Cusano Milanino, Italy; 6IRCCS Ospedale San Raffaele Scientific Institute, Division of Immunology, Transplantation and Infectious Diseases, Milano, Italy; 7Foundation IRCCS Ca’ Granda - Ospedale Maggiore Policlinico, University of Milan, Infertility Unit, Milano, Italy

Background/Objectives: Assisted reproductive technology (ART) requires the correct identification of the implantation window (WOI) to increase the chances of embryo implantation after the embryo transfer (ET). During WOI the uterine fluid-derived extracellular vesicles (UF-EVs) may play the role of mediator in endometrium-embryo communication. This study aims to predict the endometrial receptivity leveraging UF-EVs RNA-seq in women who underwent ART treatments.

Methods: The analysis included 82 women who underwent ET: 37 women achieved pregnancy while 45 failed to achieve it. After collecting the UF-EVs transcriptome during WOI a differential gene expression (DGE) analysis has been performed. The resultant statistically significative genes were aggregated through a WGCNA analysis, whose modules were used as features of a Bayesian model to predict the probability to achieve a successful pregnancy.

Results: The DGE analysis selects 966 differentially expressed genes that are eventually aggregated in 4 modules by the WGCNA. The ORA analysis reveals crucial biological functions contained in the modules used as predictors of a Bayesian logistic regression model, along with the number of previously abortions and the vesicles’ average size. The final model achieves a leave one out F1-score of 0.86 and an AUC of 0.84.

Conclusions: Our findings underline UF-EVs transcriptome differences between women who achieved a successful pregnancy post-ET and those who did not, revealing the presence of interconnected gene clusters associated with the endometrial receptivity. This study contributes to increase the likelihood of successful pregnancy outcomes through improved timing strategies for embryo transfer.

Grant: RF-2019-12369460 from the Italian Ministry of Health.

Conflict of Interest: None declared

EP16.043 Lessons learned from our diagnostic switch to GRCh38

Alexander Seitz 1, Anica Hoppe1, Mara Burkert1, Christoph Schmidt1

1MVZ genetikum GmbH, Bioinformatics, Neu-Ulm, Germany

Background/Objectives: After 10 years, there are still many institutions that have not switched to GRCh38. Here, we present the insights from our switch, regarding our internal variant database for artefact filtering and new arising problems in genes of diagnostic importance.

Methods: We actively started to switch from GRCh37 to GRCh38 in January 2023. After software installations and updates, re-analysis of the patients, validation, and accreditation, we finalized the switch in November 2023.

Results: While it is possible to just liftover the variants with minimal losses (<1%) the importance of re-analyzing the samples stem from other aspects. In our case, the number of different variants increased by 50%. Even though most of them were sequencing artefacts, stemming from the removal of the 1000g database annotation, it is absolutely necessary to be able to identify them to not clutter the diagnostic interpretation. Additionally, it is important to take a closer look at the results: While PRSS2 has no sequence on GRCh37 and can thus only be analyzed on GRCh38, a correct interpretation is still not possible. The problem is the pseudogene PRSS3P2, which was moved to an alternative contig. Now all these reads are distributed among the genes PRSS1 and PRSS2, leading to many false positives while even true variants can’t be identified correctly. However, using all alternative contigs while allowing variant calling in homologous regions results in major problems with the CNV-calling.

Conclusion: During our switch to GRCh38 we identified and solved several problems that have not been characterized.

Grants:

Conflict of Interest: None declared

EP16.044 Assessing the added value of the protein structural analysis tool FOLDX in advanced classification of missense variants of unknown significance (VUS)

Rene Mulder 1, Jan Jongbloed1, Kristin Abbott1, B. Sikkema-Raddatz1, Joeri Van Der Velde1, Helga Westers1

1UMCG, Genetics

Background/Objectives: Next-generation sequencing has transformed genetic diagnostics, enabling simultaneous high-throughput sequencing of multiple genes. As a result, the number of (novel) disease-associated variants being identified for interpretation and classification has increased significantly. Many of these variants are of unknown significance (VUS) and advanced bioinformatics tools could play a vital role in reclassifying these as pathogenic (P) or benign (B). Current guidelines emphasize employing highly specific tools, e.g., FOLDX, for predicting functional effects. FOLDX rapidly evaluates variants’ impact on protein and nucleic acid stability, folding, and dynamics. This study assesses FOLDX’s potential for advanced classification of VUS.

Methods: Candidate genes meeting the criteria—complete experimental structures (Protein Data Bank), ≥12 VUS, ≥20 L(B), and ≥20 L(P) known—were selected from the VKGL database; a national database of classified variants. Structural analysis using FOLDX determined gene-specific cut-off values via ROC analysis; a cut-off above zero delta delta Gibbs free energy (ΔΔG) indicates protein destabilization.

Results: Twelve genes met our criteria (ABCA1, ABCC8, ABCG8, BEST1, KCNQ1, MSH2, PTCH1, SCN1A, SCN5A, SCN8A, SCN10A, SLC12A3). Gene-specific cut-offs ranged from 0.08 to 3.2 ΔΔG; seven genes (ABCA1, ABCC8, ABCG8, KCNQ1, MSH2, PTCH1, SCN8A) clearly differentiated L(B), L(P) variants, and VUS. Overall, 14% (SCN10A) to 64% (SCN1A) of VUS exceeded the cut-off for (L)P variants.

Conclusion: FOLDX protein structural analysis displayed potential in distinguishing disease-causing missense variants ("high VUS") from "regular" VUS (“low-or-middle VUS”). This facilitates preselection of VUS for re-evaluation, potential reclassification, and follow-up approaches like co-segregation or functional analysis, based on stability effects.

Grants: NA

Conflict of Interest: None declared

EP16.046 Statistical analysis of genomic in-silico pathogenicity predictors for the characterization of VUS in rare and undiagnosed disorders

Eylül Aydın 1;2, Berk Ergun3, Özlem Akgün Doğan2;4;5, Yasemin Alanay2;4;5, Ozden Hatırnaz Ng2;5;6, Özkan Özdemir2;6

1Acibadem University, Institute of Health Sciences, Genome Studies, Istanbul, Türkyie; 2Acibadem University, Rare Diseases and Orphan Drugs Research and Application Center (ACURARE), Istanbul, Türkyie; 3, Geniva Informatics and Health Services Inc., Istanbul, Türkyie; 4Acibadem University, School of Medicine, Department of Pediatrics, Division of Pediatric Genetics, Istanbul, Türkyie; 5Acibadem University, School of Medicine, Department of Medical Genetics, Istanbul, Türkyie; 6Acibadem University, School of Medicine, Department of Medical Biology, Istanbul, Türkyie

Background/Objectives: The interpretation of missense variants is a pivotal element in genomic diagnostics. Current algorithms for this task display considerable inconsistency, highlighting the urgent necessity for a standardized approach. We aimed to refine the accuracy of in-silico pathogenicity predictors (ISPPs) by evaluating their performance across distinct gene-variant clusters based on pathogenicity, using variance-based multiple and pairwise comparisons.

Methods: We sourced ClinVar variants for robust classification into three categories: Benign, Pathogenic, and Unknown based on clinical significance. The study incorporated fifty-two ISPPs from dbNSFP_v4.5a, which we divided into four groups according to their predictive methodologies. These ISPPs were subjected to rigorous statistical analysis and multidimensional scaling. A gene-ISPP pairing system was established, culminating in the development of an aggregate VAMPP (Variant Analysis using Multiple Pathogenicity Predictors)-score.

Results: Our findings revealed that ensemble predictors consistently outperformed other categories for the Mendeliome. We applied the VAMPP-score to reassess thirteen cases of rare genetic disorders with missense variants originally deemed VUS. This re-evaluation led to the reclassification of biallelic variants in genes NAGA, ST3GAL5, RINT1, and LAMB1, in addition to de-novo heterozygous variants on genes DNM1L, HECW2, and NKX2-1, as likely-pathogenic, leading to the diagnosis.

Conclusion: This study presents a novel integrated ISPP, VAMPP-score, which enhances the interpretation of missense variants through a quantitative framework aligned with ACMG/AMP guidelines by utilizing ClinVar data to ISPPs, thus augmenting the diagnostic process. This tool will be accessible for gene-specific ISPP evaluation on the open-access web platform starting February 2024 at www.vamppscore.com.

Grants: -

Conflict of Interest: Eylül Aydın Full-time employee at Geniva Informatics and Health Services Inc., Berk Ergun Full-time employee at Geniva Informatics and Health Services Inc., Özlem Akgün Doğan: None declared, Yasemin Alanay: None declared, Ozden Hatırnaz Ng: None declared, Özkan Özdemir: None declared

EP16.047 Genetic control of population diversity in human immunoglobulin G N-glycosylation

Anna Soplenkova 1, Denis Maslov1, Anna Timoshchuk1, Tim Spector2, Michel Georges3, Sodbo Sharapov1, Gordan Lauc4, Yury Aulchenko1;5

1MSU Institute for Artificial Intelligence, Moscow State University, Moscow, Russian Federation; 2King’s College London, Department of Twin Research and Genetic Epidemiology, London, United Kingdom; 3University of Liège, Unit of Animal Genomics, WELBIO, GIGA-R and Faculty of Veterinary Medicine, Liège, Belgium; 4Genos Glycoscience Research Laboratory, Zagreb, Croatia; 5Institute of Cytology and Genetics, Novosibirsk, Russian Federation

Background/Objectives: N-glycosylation is a common post-translational modification that impacts the physical and biological functions of proteins. N-glycan biosynthesis pathways are well studied, but understanding of the mechanisms of genetic regulation is limited, hindering of glycome-associated disease biomarker development.

The total blood plasma N-glycome is a superposition of N-glycomes of individual glycoproteins. Reconstructing the N-glycan concentrations of individual proteins from the concentrations of glycans of all proteins in blood plasma will allow obtaining new datasets without profiling new samples.

Methods: To predict the immunoglobulin G N-glycan concentrations from the total blood plasma N-glycome, we compared five models: a simple linear regression, the lasso and ridge regressions, an elastic net, and a canonical correlation analysis. We applied the best model to the total blood plasma N-glycosylation GWAS results and then obtained the predicted immunoglobulin G N-glycosylation GWAS results. We validated our results by comparing the set of loci significantly associated with immunoglobulin G N-glycosylation to the already published loci in Klarić et al. [1].

Results: The simple linear regression model showed the greatest accuracy. The predicted immunoglobulin G N-glycosylation GWAS results are consistent with the published loci in Klarić et al. [1].

Conclusion: The presented method opens up the possibility of conducting a meta-analysis between real and predicted GWAS results of immunoglobulin G N-glycosylation. This will strongly increase the sample size without profiling new samples.

Grants: This work is an output of a research project implemented as part of the Research Program at the MSU Institute for Artificial Intelligence.

[1] L. Klarić et al. Science advances, 2020, 6(8)

Conflict of Interest: Anna Soplenkova: None declared, Denis Maslov: None declared, Anna Timoshchuk: None declared, Tim Spector: None declared, Michel Georges: None declared, Sodbo Sharapov: None declared, Gordan Lauc a founder and owner of Genos Ltd, a biotech company that specializes in glycan analysis and has several patents in the field, Yury Aulchenko a full-time employee of GSK

EP16.048 Deep learning approaches are more accurate in predicting multifactorial diseases than polygenic risk scores

Alexey Kamelin 1;2, Vladislav Perelygin1, Layal Shaheen2, Anna Kim2, Nicolay Plotnikov2, Alexander Rakitko2, Maria Poptsova1

1Higher School of Economics, Laboratory of Bioinformatics, Faculty of Computer Science, Moskva, Russian Federation; 2Genotek, Moskva, Russian Federation

Background/Objectives: Polygenic risk score (PRS) is a common method for evaluating the risk of diseases. SNP weights are calculated through a linear regression, assuming the independent and linear influence of each SNP on the phenotype, which can be otherwise due to epistatic interactions. Here we explore the efficacy of non-linear machine learning algorithms and deep learning compared to the conventional PRS method.

Methods: We compared ensemble trees and neural networks to the LASSO linear regression using simulated data with varying types and strengths of epistasis. Then superiority of non-linear models was tested using real genetic data for multifactorial phenotypes like obesity (8 506 cases and 50 168 controls), type 1 diabetes (522 cases, 10 440 controls after propensity score matching), and psoriasis (1 543 cases, 10 801 controls after propensity score matching). All individuals were genotyped using Illumina GSA v.3 microarray.

Results: As strength of epistasis increased in simulated data, non-linear models outperformed linear models. On real data, among the non-linear models, gradient boosting was the most effective for obesity (GB AUC: 0.779 ± 0.002 vs LASSO AUC: 0.773 ± 0.002) and psoriasis (GB AUC: 0.703 ± 0.004 vs LASSO AUC: 0.699 ± 0.004), while deep learning exhibited superiority over linear approaches for type 1 diabetes (RNN AUC: 0.823 ± 0.004 vs. LASSO AUC: 0.787 ± 0.004).

Conclusion: Our study confirms the power of non-linear models and deep learning approaches to more accurately account for epistasis.

Conflict of Interest: None declared

EP16.049 Pitfalls of accurate RNA-seq in human saliva samples

Sergi Marí Alemany 1;2, Alba Hernangomez-Laderas1;2, Irati Miguel1, Ariadna Cilleros-Portet1;2, Bárbara P. González-García1;2, Iraia García-Santisteban1;2, Nora Fernandez-Jimenez1;2, Jose Ramon Bilbao1;2;3

1University of the Basque Country (UPV/EHU), Leioa, Spain; 2Biobizkaia Health Research Institute, Barakaldo, Spain; 3Center for Biomedical Research in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain

Background / objective: RNA-seq is extensively used to analyse gene expression in multiple tissues. Nevertheless, there is a notable gap in RNA-seq of saliva samples, a non-invasive and accessible biofluid that is good opportunity for understanding physiological changes. We illustrate the difficulties of RNA-seq in 380 auto-collected saliva samples.

Methods: First, we used Illumina Stranded Total RNA Library Preparation Kit (coding and multiple forms of non-coding RNAs) with Ribo-Zero plus rRNA Depletion. We then performed a second round of RNA-seq adding Microbiome rRNA Depletion Kit. Finally, we performed a third round of exome sequencing using Illumina RNA Prep with Enrichment to maximize human RNA detection.

Results: An extremely low percentage of human RNA is captured by the Total RNA kit, with a mean percentage of human reads of 1.5 ± 3.44%, and a majority of microbial RNA. Although the proportion of human reads improves with exome-capture, there is a high heterogeneity in quality and in number of human reads among samples, with a mean proportion of 52.85 ± 26.99%.

Conclusion: Standard RNA-seq in saliva samples is unfeasible for human gene expression analysis, even with high depth of sequencing, but it offers the opportunity to analyse local microbial activity. Exome-capture RNA-seq is a valid strategy to measure human gene expression in degraded samples, but auto-collection of saliva may affect the quality of the RNA sample and the result in sequencing output. We hope our results help other researchers to design RNA-seq analysis in saliva samples.

Grants: Basque Government SAN2020111043, SAN2019111085; Spanish MICINN PID2019-106382RB-I00, PI21/0149; Mexican Federal Government 2021-000007-01EXTF-00209.

Conflict of Interest: None declared

EP17 Large Scale Association Studies (GWAS)

EP17.001 Constructing a predictive polygenic model for asthma in the Tatars using GWAS data

Askar Akhmetshin 1, Alexandra Karunas1;2;3, Olga Savelieva1;2, Elza Khusnutdinova1;2

1Institute of Biochemistry and Genetics of Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa, Russian Federation; 2Ufa University of Science and Technology, Department of Genetics and Fundamental Medicine, Ufa, Russian Federation; 3Bashkir State Medical University, Department of Medical Genetics and Fundamental Medicine, Ufa, Russian Federation

Background/Objectives: Asthma, a common multifactorial disease, exhibits a significant hereditary aspect due to its polygenic pattern. This study aimed to develop an effective polygenic score (PGS) model for predicting asthma to individuals of Tatar ethnicity.

Methods: GWAS analysis and genotype data for 436 individuals with asthma (225) /control (211) of Tatars from the Volga-Ural region were used. Genotype imputation was carried out using the 1000 Genome Project data (CEU population) as a reference utilizing the Beagle 5.4. GWAS-analysis using 1,490,621 SNPs was performed by Hail.is. The polymorphisms that revealed the most statistical significance were chosen for inclusion in the PGS analysis.

Results: We identified 38 SNPs with a significant association to asthma in Tatars. The strongest association of rs12591872 located near the gene IGF1R (15q26) (β = -1.354, SE = 0.281, P-value = 1.39×10-6) with asthma in Tatars was established. For the PGS analysis were selected 11 SNPs by excluding variants with a linkage disequilibrium threshold of r2 > 0.5. The association analysis of the weighted PGS using β coefficients from logistic regression as the weights revealed a high odds ratio for asthma risk in cases compared controls (OR[95%CI] = 9.41[9.23-9.59], P-value = 4.213×10-13). An impressive area under the receiver operating characteristic (ROC) curve (AUC[95%CI] = 0.878[0.834-0.923]). The weighted PGS analysis using β-coefficients from GWAS meta-analysis of asthma (Demenais, 2018) showed low predictive ability of model (AUC[95%CI] = 0.663[0.59-0.73]).

Conclusion: The PGS model demonstrates predictive capabilities in evaluating the predisposition for asthma among individuals of Tatar ethnicity.

Grants: This work was particularly funded by the Megagrant of the Government of the Russian Federation (no. 075-15-2021-595).

Conflict of Interest: None declared

EP17.002 GWAS of plasma sodium with and without exposure to thiazide diuretics

Niklas Worm Andersson 1;2, xiaoping wu1;2, Frank Geller1;2, Jan Wohlfahrt3, Mads Melbye3;4;5, Anders Peter Hviid1;6, Jonas Ghouse7;8, Ole Pedersen9;10, Erik Sørensen2, Sisse Rye Ostrowski2;9, Henning Bundgaard9;11, Marie Lund1;9;12, Bjarke Feenstra1;2;13

1Statens Serum Institut, Department of Epidemiology Research, Denmark; 2Rigshospitalet, Copenhagen University Hospital, Department of Clinical Immunology, Denmark; 3Danish Cancer Institute; 4Stanford University School of Medicine, Department of Pediatrics; 5Norwegian University of Science and Technology, HUNT Center for Clinical and Molecular Epidemiology; 6University of Copenhagen, Pharmacovigilance Research Center, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, Denmark; 7University of Copenhagen, Laboratory for Molecular Cardiology, Department of Biomedical Sciences; 8Rigshospitalet, Copenhagen University Hospital, Laboratory for Molecular Cardiology, Department of Cardiology, Copenhagen University Hospital, Copenhagen, Denmark; 9University of Copenhagen, Department of Clinical Medicine, Faculty of Health and Medical Sciences; 10Zealand University Hospital, Department of Clinical Immunology,; 11Rigshospitalet, Copenhagen University Hospital, Department of Cardiology; 12Copenhagen University Hospital – Bispebjerg and Frederiksberg, Department of Clinical Pharmacology; 13University of Copenhagen, Department of Biology,

Background/Objectives: Abnormal plasma sodium levels represent an imbalance between total body water relative to electrolyte concentrations. Low sodium (hyponatremia) is common and associated with substantial morbidity, and ultimately mortality. Thiazide diuretics constitute a leading cause of drug-induced hyponatremia.

Methods: We conducted genome-wide association meta-analysis of plasma sodium concentrations in 188,464 individuals (142,575 from Copenhagen Hospital Biobank and the Danish Blood Donor Study together with 45,889 from previous meta-analysis), analyzing potential genetic risk factors underlying thiazide-induced hyponatremia by case-control (n = 829 cases and n = 16,368 controls), quantitative trait (n = 14,326 thiazide users) and gene-environment interaction (n = 9,237 thiazide and n = 21,692 alternative antihypertensive drug users) analyses.

Results: The genome-wide association meta-analysis of plasma sodium concentrations, yielded 33 independent association signals at P < 5×10−8, out of which 31 were novel and 2 had previously been reported, NFAT5 and SLC4A10. No genome-wide significant loci related to thiazide-induced hyponatremia were found. In a gene-environment interaction analysis, we found that 1 standard deviation lower polygenic score for plasma sodium was associated with 0.41 (95% confidence interval, CI: 0.34-0.47) mmol/L lower sodium, and that exposure to thiazide was associated with 0.84 (95% CI: 0.76-0.92) mmol/L lower sodium compared to an alternative antihypertensive drug, but we found no gene-environment interaction effect (P = 0.59).

Conclusion: We identified 33 genome-wide significant loci associated with plasma sodium concentrations in general, but found no specific genetic risk factors for thiazide-induced hyponatremia.

Conflict of Interest: None declared

EP17.004 The genetic differential diagnosis in children with Henoch–Schönlein purpura nephritis and idiopathic IgA nephropathy.

Mariia Proskura 1, Anastasiia Buianova2, Valery Cheranev2, Vera Belova2, Anna Shmitko2, Anna Pavlova2, Iuliia Vasiliadis2, Oleg Suchalko2, Denis Rebrikov2, Edita Petrosyan1, Dmitriy Korostin2

1Pirogov Russian National Research Medical University, Russian Children’s Clinical Hospital, Moscow, Russian Federation; 2Pirogov Russian National Research Medical University, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Moscow, Russian Federation

Background/Objectives: IgA nephropathy (IgAN) stands out as the predominant type of primary glomerulonephritis. The perception of IgA vasculitis (IgAV) or Henoch–Schönlein purpura nephritis as a systemic manifestation of IgAN arises due to the disease’s clinical heterogeneity and an incomplete understanding of its genetic predisposition.

Methods: This retrospective study encompassed 22 patients diagnosed with IgAV and 48 with idiopathic IgAN (25 girls and 45 boys, mean age 10.3 ± 3.8 years). The control group comprised 637 healthy children (306 girls, 331 boys). Exome-wide association analysis for patients utilized logistic regression models based on additive and recessive inheritance patterns. Additionally, HLA allele frequencies were examined across the groups.

Results: In the additive model, the most noteworthy association was observed with rs80346174 (GSDMD) and IgAV (p = 1.335 × 10-12; FDR = 0.00178, OR = 0.3189 [3.841–13.41]), in the recessive model – of rs1794868 (PKP1) (p = 7.176 × 10-6, FDR = 2.453 × 10-4, OR = 0.3366 [2.105–7.875]). A Protein-Protein Interaction network analysis revealed 15 proteins associated with kidney disease in the additive model of IgAN. The most enriched Gene Ontology (GO) category in the additive model of IgAN was ‘Differentiation of myeloid leukocytes’ (GO:0002573, p = 0.00034), and in the recessive model, it was ‘Development of the urogenital tract’ (GO:0001655, p = 3.06 × 10-5).

Conclusion: Regardless of the mode of inheritance, candidate genes for IgAN and IgAV play roles in immune system signaling pathways. However, genes associated with kidney development were exclusively identified in patients with IgAN across both inheritance patterns.

Grants: №075-15-2019-1789.

Conflict of Interest: None declared

EP17.005 Multi-trait meta-analyses based on co-morbidity categories provide novel insights into the genetic architecture of hypothyroidism

Chloé Pittman 1, Sofie Bliddal2;3, Ulla Feldt-Rasmussen2;4, Tugce Karaderi1

1Center for Health Data Science, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; 2Rigshospitalet, Department of Medical Endocrinology and Metabolism, Copenhagen, Denmark; 3Hvidovre Hospital, Department of Gynaecology and Obstetrics, Copenhagen, Denmark; 4Institute of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark

Background/Objectives: Hypothyroidism is a common endocrinological condition affecting up to 10% of the general population with extensive heterogeneity for symptoms, co-morbidities and required treatment. Its co-morbidities can be outlined in four categories: (1)Reproductive, (2)cardiometabolic, (3)psychiatric, (4)polyautoimmunity/immune-mediated. It is vital to investigate its genetic underpinnings in the context of its co-morbidities for a better understanding of the disease mechanisms.

Methods: We explored the genetic overlap between hypothyroidism and 40 selected phenotypes, with publicly available genome-wide association study (GWAS) summary statistics, based on a literature review for known genetic correlation (rg) and/or epidemiological evidence. First, we estimated rg across these phenotypes, and then, conducted four multi-trait GWAS meta-analyses including phenotypes with statistically significant rg with hypothyroidism, corrected for multiple-testing in each co-morbidity category. We investigated new genetic associations for hypothyroidism, and the shared signals across the 27 phenotypes in the meta-analyses.

Results: We identified 92 new loci (p < 5x10-8) for hypothyroidism in four meta-analyses by leveraging the common genetic architecture across the phenotypes. rs28406364, a likely regulatory intronic variant in ZNF652-AS1, was a common locus for hypothyroidism, age at menarche and preeclampsia. While hypothyroidism and sleep apnoea shared 10 of the new loci, pernicious anaemia shared seven of the associations. Of these, rs2351305, upstream the pleiotropic WDR12-CYP20A1 locus, was also associated with myasthenia gravis and type 1 diabetes.

Conclusion: We identified new genetic associations for hypothyroidism and gained further fundamental insights into the genetic architecture shared with selected phenotypes, improving the understanding of common biological processes behind hypothyroidism and its co-morbidities.

Grants: Supported by grants NNF20OC0062294(TK), NNF22OC0077221(SB) and NNF23OC0087269(SB).

Conflict of Interest: None declared

EP17.006 Progress towards the 20,000 Type 1 Diabetes (T1D) Exome Cohort In Ukraine

Taras Oleksyk 1;2, Khrystyna Shchubelka1;2, Walter Wolfsberger2, Olga Oleksyk1;3

1Uzhhorod University, Biology, Uzhhorod, Ukraine; 2Oakland Univesity, Biological Sciences, Rochester, United States; 3A. Novak Transcarpathian Regional Clinical Hospital, Endocrinology Department, Uzhhorod, Ukraine

Consortium: T1D Ukraine Consortium

Background/Objectives: Progress has been made in understanding how common genetic variants contribute to the risk of developing type 1 diabetes or T1D. We have already collected and sequenced exomes of 3,000 type T1D patients and 3,000 matching controls in Ukraine and conducted a genome-wide association study with the goal of discovering new genetic factors that affect the development and pathology.

Methods: We have established a cross-sectional study with informed consent for public data release during 2022-2023. The ~7,500 samples that have been collected to date represent all 24 oblasts (provinces), including the Autonomous Republic of Crimea. The cohort is selected among the T1D patients participating in the national T1D diabetic registry and implemented in the RedCap system.

Results: The T1D Ukraine Consortium has made a significant progress to establish a T1D Research Network and a Biobank: a comprehensive resource for future research efforts in genomics in the region. The dataset of 6,000 exomes for identification of novel functional genetic variants linked to T1D has been created. Several of these have confirmed the previous studies, and local associations were also uncovered.

Conclusion: Our T1D exome dataset is prepared for an online access for the research use though the Open-Access Genetic Database: Functionally Significant Genetic Markers that have been identify with a GWAS will offer new avenues for T1D prevention and treatment.

Grants: The Leona M. and Harry B. Helmsley Charitable Trust Phase 1 “A comprehensive study of T1DM exomes in Ukraine. Phase 1”. European Commission 2SOFT/1.2/48 “Partnership for Genomic Research in Ukraine and Romania”.

Conflict of Interest: None declared

EP17.007 Risk variants in three late-onset Alzheimer’s disease related genes in cognitively healthy Bulgarian centenarians

Mikaela Stancheva 1, Lubomir Balabanski2, Dimitar Serbezov2, Dragomira Nikolova2, Radoslava Vazharova2, Draga Toncheva2, Sena Karachanak-Yankova1;2

1Sofia University St. Kliment Ohridski, Sofia, Bulgaria; 2Medical University of Sofia, Sofia, Bulgaria

Background/Objectives: The frequency of dementia, with its most common form being Alzheimer’s disease (AD), gradually increases with age. AD’s heritability has been the subject of numerous case-control studies, which can potentially be biased by age not-matched controls. In this context, we screened cognitively healthy Bulgarian centenarians for the presence of AD-associated genetic risk variants.

Methods: We searched for AD-associated variants (from Disgenet) in data, obtained by whole genome sequencing, performed using BGISEQ-500, from 16 Bulgarian centenarians with preserved cognitive health. This study is mainly focused on three late-onset AD-related genes: ABCA7, PICALM and TREM2.

Results: In this study, we found 8 AD-associated risk variants: four in PICALM: rs17817600 (in 5 centenarian genomes); rs867611, rs639012 (in 13 genomes each) and rs527162 (in 15 genomes); 4 variants in ABCA7: rs3795065 C-T, rs12151021 A-G, rs4147929 A-G (in 15 centenarian genomes each) and rs3752241 C-G (in 6 genomes); whereas the TREM2 gene did not contain any AD-associated risk variants. The A allele of rs4147929 (found in 1 sample) leads to increased ABCA7 expression which is observed in AD patients. All variants have high MAF in gnomAD (>10%).

Conclusion: The presence of multiple risk variants in genes associated with the development of AD in the genomes of cognitively healthy centenarians suggests that either these variants are of lower risk than suggested or that there is a presence of protective variants with stronger effect, which should be further investigated.

Grants: DN 03/7 from 18.12.2016 and KP-06-N33/5 from 13.12.2019 - National Science Fund of Bulgaria

Conflict of Interest: None declared

EP17.008 Identifying determinants of hepatitis B viral immunity

Mark Seielstad 1

1University of California San Francisco, Institute for Human Genetics, San Francisco, United States

Background/Objectives: HepatitisB (HBV) infects >250 million people. Some 5%-10% of exposed adults develop a chronic infection, causing liver cirrhosis or cancer in 15%-25% of cases. Vaccination has reduced infections, but there is no cure. We identify genetic variation impacting vaccine response, and the ability to spontaneously clear HBV in a cohort of Chinese individuals from Taiwan.

Methods: The TaiwanBiobank comprises 120,552 adults tested serologically for HBsAg, anti-HBs, anti-HBc, and anti-HBe. Previously vaccinated, chronically, and formerly infected individuals were identified as shown (Table). Genotyping was performed with a custom array containing 591,048 SNPs.

Results: We performed an unadjusted GWAS analysis on an early release of 2,000 chronically and 2,000 previously infected individuals. Highly associated SNPs (p < 5 × 10-16) fall within a 36kb region, encompassing HLA-DPB1 and HLA-DPA1. There was no inflation of p-values due to population stratification. We have observed similar ClassII associations with vaccine response.

Conclusion: Shared HLA associations suggest those least protected by vaccination may be most at risk of chronic infection, a significant clinical concern. The inability to respond to vaccine and the risk of chronic infection are both characterized by absence of HBs antibodies. This in turn suggests that HLA ClassII protein sequences vary in their ability to bind and present s-antigen to B-cells, resulting in variable antibody responses. Future studies intend to examine binding affinities for s-antigen with HLA alleles identified here.

Grants: Fulbright Scholar Program and Foundation for Scholarly Exchange Taiwan. Additional support: UCSF, Academia Sinica, and Dr. Hwai-I Yang.

Conflict of Interest: None declared

EP17.009 Regional analysis of vascular retinal features

Sacha Bors 1, Sven Bergmann2

1UNIL, Department of Computational Biology, Lausanne, Switzerland; 2Lausanne, Department of Computational Biology, Lausanne, Switzerland

Background/Objectives: The assumption that retinal vasculature reflects the systemic vascular function of the body, has already motivated numbers of researches. The conventional fundus image (CFI) is a non-invasive modality that offers high-quality imaging of blood vessels in the retina. In this study, we aim to investigate the added value of a more refined phenotyping of the retina, through a comparative analysis of the heritability of several traits and their disease associations, with already known results aggregated over the whole retina.

Methods: For this purpose, we apply a complete and fully automated processing pipeline on over 130k fundus images of close to 72k UK BioBank subjects. This involves initial steps such as the segmentation and the classification of the vessels, followed by the extraction of regional vascular features, including median vessels diameter, diameter variability, vascular density, central retinal equivalents, number of bifurcations, and tortuosity. Those phenotypes are adapted to their regional versions i.e. aggregated on the top versus bottom hemispheres of the retina, as well as stratified by branching level in the vascular tree, so that both spatial and topological informations are captured. We then estimate the heritability of those parameters by performing a genome-wise association study (GWAS), and their disease trait associations. We finally compare the obtained results with the ones arising when using the standard global phenotyping.

Grants: Sinergia grant

Conflict of Interest: None declared

EP17.010 Systematic characterization of the genetic architecture of acute COVID-19 and its long-term sequelae in the German National Pandemic Cohort (NAPKON)

T. madhusankha Alawathurage 1, Ayda Abolhassani2, Axel Schmidt1, Laura Kilarski2, Thomas Illig3, Verena Kopfnagel3, Kerstin Ludwig1, Eva C. Schulte1;2

1Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn; 2Institute of Psychiatry and Psychotherapy, University of Bonn, School of Medicine & University Hospital Bonn; 3Hannover Unified Biobank (HUB), Hannover Medical School

Consortium: The NAPKON study group

Background/Objectives: Large-scale host genetic analyses are integral to the elucidation of the genetic architecture of COVID-19 and its post-acute sequelae. The National Pandemic Cohort Network (NAPKON), initiated in July 2020 as part of the German Network of University Medicine (NUM), includes >5,000 participants across the entire healthcare sector. Whole exome sequencing (WES) in this cohort will serve as a cost-efficient yet comprehensive approach to identify functionally relevant rare variants associated with COVID-19 and long-term sequelae.

Methods: We will complement existing multi-omics data for NAPKON participants (e.g. genotyping data, cytokine measurements) by generating WES data of the entire NAPKON cohort, spanning all levels of severity. Data will be generated and analyzed using standard procedures and made available through the NAPKON Use&Access procedure.

Results: We will use this novel resource to identify rare host genetic factors implicated in diverse COVID-19-related phenotypes by performing classical statistical analyses (e.g. single variant association, gene burden testing) and integrating array-based genotypes to combine variants from the entire allelic spectrum, e.g. in polygenic risk scores. Deep phenotyping information will help stratify individuals for subgroup analyses, and data from transcriptomics, proteomics, and epigenomics will be integrated to functionally interpret risk variants. The first results will be presented at the conference.

Conclusion: The present project closes the gap from rare variant identification to pathophysiologic understanding of COVID-19. It provides unique added value for the NAPKON/NUM efforts and the fields of host genetics/functional genomics by generating one of the few highly visible and reusable large-scale WES datasets in Germany.

Grants: DFG, BMBF (NUM/NAPKON)

Conflict of Interest: None declared

EP17.011 New genes for back pain-related phenotypes identified by multi-trait gene-based association analysis

Nadezhda Belonogova 1, Elizaveta Elgaeva1;2, Irina Zorkoltseva1, Anatoly Kirichenko1, Gulnara Svishcheva1;3, Maxim Freidin4, Frances Williams5, Pradeep Suri6;7;8;9, Tatiana Axenovich1, Yakov Tsepilov1

1Institute of Cytology and Genetics of SB RAS, Novosibirsk, Russian Federation; 2Novosibirsk State University, Department of Natural Sciences, Novosibirsk, Russian Federation; 3Institut Obshchey Genetiki Imeni N. I. Vavilova Ran, Moskva, Russian Federation; 4Queen Mary University of London, Department of Biology, School of Biological and Behavioral Sciences, United Kingdom; 5Department Of Twin Research & Genetic Epidemiology, Kings College London, United Kingdom; 6VA Puget Sound Health Care System, Seattle Epidemiologic Research and Information Center, Seattle, United States; 7Division of Rehabilitation Care Services, Seattle, United States; 8University of Washington, Clinical Learning, Evidence, and Research Center, Seattle, United States; 9University of Washington, Department of Rehabilitation Medicine, Seattle, United States

Background/Objectives: Back pain (BP) is a major contributor to disability worldwide. Three BP-related phenotypes: chronic BP (CBP), dorsalgia and intervertebral disc disorders (IDD), have heritability estimated at 40-60%. Less than half of the heritability is explained by common genetic variants identified by GWAS. More powerful methods of statistical analysis may offer additional insight.

Methods: Using multi-trait and imputed genotypes from the UK Biobank we performed a gene-based association analysis. A multi-trait analysis combining three BP-related phenotypes: CBP, dorsalgia, and IDD, was conducted using the SHAHER approach, which maximizes the heritability of the multi-trait phenotype.

Results: We identified and replicated 16 genes associated with BP-related traits. Seven of the detected genes, namely, MIPOL1, PTPRC, RHOA, MAML3, JADE2, MLLT10, and RERG, were previously unreported. Several new genes have been previously detected as associated with traits genetically correlated with BP or included in pathways associated with BP. Our results verify the role of these genes in BP-related traits.

Conclusion: Of 16 genes significantly associated with BP-related traits, 13 were detected on the multi-trait phenotype that is in accordance with high genetic correlation between BP-related traits.

Grants: The work was supported by the budget project of the Institute of Cytology and Genetics FWNR-2022-0020, the Russian Science Foundation (No. 22-15-20037) and Government of the Novosibirsk region grant, Versus Arthritis grant number 22467, NIH/NIAMS P30AR072572. This research has been conducted using the UK Biobank Resource under Applications #18219 and #59345.

Conflict of Interest: None declared

EP17.012 A genome-wide association study of the plasma N-glycome of 10,000 individuals significantly expands the list of associated loci

Sodbo Sharapov1, Anna Timoshchuk 1, Olga Zaytseva2, Denis Maslov1, Anna Soplenkova1, Elizaveta Elgaeva3;4, Frano Vuckovic2, Irena Trbojević-Akmačić2, Frances Williams5, Dragan Primorac6, Jan van Zundert7;8, Michel Georges9, Karsten Suhre10, Massimo Allegri11, Nish Chaturvedi12, Malcolm Dunlop13, Matthias Bernd Schulze14;15;16, Tim Spector5, Yakov Tsepilov3;17, Gordan Lauc2, Yury Aulchenko1;3

1MSU Institute for Artificial Intelligence, Lomonosov Moscow State University, Moscow, Russian Federation; 2Genos Glycoscience Research Laboratory, Zagreb, Croatia; 3Institute of Cytology and Genetics, Novosibirsk, Russian Federation; 4Novosibirsk State University, Novosibirsk, Russian Federation; 5Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King’s College London, St Thomas’ Campus, London, United Kingdom; 6St. Catherine Specialty Hospital, Zagreb, Croatia; 7Department of Anesthesiology and Multidisciplinary Paincentre, ZOL, Genk/Lanaken, Belgium; 8Department of Anesthesiology and Pain Medicine, Maastricht University Medical Centre, Maastricht, Netherlands; 9Unit of Animal Genomics, WELBIO, GIGA-R and Faculty of Veterinary Medicine, University of Liège, Liège, Belgium; 10Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar; 11Pain Therapy Department Policlinico Monza Hospital, Monza, Italy; 12MRC Unit for Lifelong Health & Ageing University College London, London, United Kingdom; 13Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, The University of Edinburgh, Edinburgh, United Kingdom; 14Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam- Rehbruecke, Nuthetal, Germany; 15German Center for Diabetes Research (DZD), Neuherberg, Germany; 16Institute of Nutrition Science, University of Potsdam, Potsdam, Germany; 17Wellcome Sanger Institute, Cambridge, United Kingdom

Background/Objectives: Glycosylation is a post-translational protein modification that impacts their physico-chemical properties as well as biological functions. While the biochemical pathways of glycosylation are well studied, its genetic regulation remains incompletely understood. Here we aimed to identify and replicate new loci associated with blood plasma N-glycosylation, propose novel genes that regulate N-glycosylation, and explore the potential role of N-glycans in the control of complex traits and multifactorial diseases.

Methods: We performed univariate and multivariate genome-wide association meta-analyses on more than 10,000 individuals, examining 117 N-glycome traits. To identify causal genes for each replicated locus, we used eight in silico-based predictors. We performed a bidirectional mendelian randomization to identify potential causal relationships between glycans and diseases.

Results: We identified 16 novel loci and prioritized 13 novel genes contributing to N-glycosylation. Among these were glycosyltransferase gene ABO, as well as other genes associated with lipid metabolism – GCKR, FADS2, TRIB1, and GRAMD1B, which, to our knowledge, is the first time lipid metabolism and its regulation has been linked to the human blood plasma protein N-glycosylation. We found causal relationships between high-mannose glycans, asthma, and lipoprotein metabolism disorders.

Conclusion: By integrating glycomics, proteomics, transcriptomics, and genomics, we provide a resource that facilitates deeper exploration of disease pathogenesis and supports drug discovery and development efforts.

Grants: This work was partially supported by the Research Program at the MSU Institute for Artificial Intelligence. The study was conducted using the UK Biobank

resource under application #59345.

Conflict of Interest: Sodbo Sharapov: None declared, Anna Timoshchuk: None declared, Olga Zaytseva: None declared, Denis Maslov: None declared, Anna Soplenkova: None declared, Elizaveta Elgaeva: None declared, Frano Vuckovic: None declared, Irena Trbojević-Akmačić: None declared, Frances Williams: None declared, Dragan Primorac: None declared, Jan van Zundert: None declared, Michel Georges: None declared, Karsten Suhre: None declared, Massimo Allegri: None declared, Nish Chaturvedi: None declared, Malcolm Dunlop: None declared, Matthias Bernd Schulze: None declared, Tim Spector: None declared, Yakov Tsepilov: None declared, Gordan Lauc G.L. is the founder and owner of Genos Glycoscience Research Laboratory,a private research organization that specializes in high-throughput glycomicanalysis and has several patents in this field., Yury Aulchenko Employee of GSK

EP18 Genetic Epidemiology and Pharmacogenomics

EP18.002 Pharmacotranscriptomic study of synthetic tuftsin analogous in hippocampus of rats in the early hours after acute restraint stress

Ivan B. Filippenkov 1, Natalia Yu. Glazova1;2, Elena A. Sebentsova1;2, Ivan V. Mozgovoy1, Vasily V. Stavchansky1, Nikolay F. Myasoedov1, Natalia G. Levitskaya1;2, Svetlana A. Limborska1, Lyudmila V. Dergunova1

1National Research Centre “Kurchatov Institute”, Moscow, Russian Federation; 2Lomonosov Moscow State University, Moscow, Russian Federation

Background/Objectives: Peptides are being actively studied as anti-stress drugs. Synthetic tuftsin analogous (TKPRPGP) has anxiolytic agent. The use of transcriptomic approaches is effective to study genesis of the processes triggered by TKPRPGP under stress. Here, a model of acute restraint stress (ARS) was implemented in rats. We used high-throughput RNA sequencing (RNA-Seq) to study the effect of TKPRPGP on the gene expression of rat hippocampus at 2h after ARS.

Methods: Wistar rats, ARS lasted 1h, RNA-Seq, real-time RT-PCR, bioinformatics.

Results: 22 differential expressed genes (DEGs) with cut-off>1.5 and Padj<0.05 were revealed 2h after ARS. The administration of TKPRPGP (300 μg/kg) to rats 30 min before ARS led to 549 DEGs related to processing and presentation of antigens and neurotransmission. In addition, the peptide predominantly downregulated those DEGs (Gpatch4, Aox1, Alox15, Hbb, Acsm5, Slc6a13, Igf2, Col3a1, Ptgds) that were upregulated under stress. Concomitantly, there were no DEGs without ARS at 3.5h after TKPRPGP administration.

Conclusion: TKPRPGP modulated hippocampal gene expression profile already 2h after ARS, without those effects when ARS was not present. However, TKPRPGP under ARS trigged more DEGs than ARS action itself. Thus, TKPRPGP not only modulated stress-induced response, but cause own more complex action. We expect that further studies of the role of non-coding RNAs may provide known more about regulation of peptide activity in the brain under normal and stress conditions.

Grants: This work was supported by the Thematic plan (5f.5.9.) of NRC “Kurchatov Institute”.

Conflict of Interest: None declared

EP18.003 Novel population specific pathogenic variants in DPYD gene revealed by exome data in a cohort from N.Macedonia; implications for a routine 5-fluorouracil therapy toxicity testing

Elizabeta Krstevska Bozhinovkj 1, Predrag Noveski2, Marija Staninova Stojovska1, Emilija Gjorgievska1, Angela Joskovska1, Nenad Mitreski3, Biljana Grozdanovska3, Aleksandar Dimovski1;2, Nadica Matevska-Geshkovska1

1Faculty of Pharmacy, University Ss. Cyril and Methodius, Center for Biomolecular Pharmaceutical Analyses, Skopje; 2Macedonian Academy of Sciences and Arts, Research Center for Genetic Engineering and Biotechnology, Skopje; 3Faculty of Medicine, University Ss. Cyril and Methodius, University Clinic for Radiotherapy and Oncology, Skopje

Background/Objectives: Fluoropyrimidines (5-FU) are the mainstream therapy for treatment of many cancers. Four variants (rs3918290 or DPYD*2A, rs55886062 or DPYD*13, rs67376798 and rs56038477) in this gene were confirmed to have impact on serious life-threatening 5-FU toxicity and are used in routine clinical practice as predictive markers. The aim of this study was to discover new potentially pathogenic variants in the DPYD gene in N. Macedonia population.

Methods: The results were extracted from exome sequencing data of 680 patients and their relatives tested for rare diseases. Alignment and variant calling were based on hg38 and star-allele haplotypes/dyplotypes were called using PharmCat v.2.5.0. The results were compared with routine clinical testing data from 4393 cancer patients.

Results: The rs55886062 was not detected both in exome and routine testing data [allele frequency in European population (AF EUR) = 0,00055]. Two new pathogenic variants were detected which had similar frequency in our population with the other 3 variants used for routine clinical testing. The first, a missense variant in exon 6 (rs72549307 c.632A>G, p.Tyr211Cys) was associated with no function of the enzyme and its AF was 0,00147 in our cohort (AF EUR = 0,00001). The second, a frameshift variant in exon 17 (rs1553251907, c.2178del, p.Gly727Valfs*15) had AF of 0,00074 in N.Macedonia (AF EUR = 0.0) and was classified as likely pathogenic and pathogenic by ClinVar and ACMG, respectively.

Conclusion: These results suggest the need for modifying the algorithm for DPYD toxicity evaluation in our country by exclusion of rs55886062 and inclusion of rs72549307 and rs1553251907 in the routine clinical practice.

Grants: No

Conflict of Interest: None declared

EP18.004 Differential effects of adrenocorticotropic hormone-like peptides after cerebral ischemia at the transcriptome level

Svetlana A. Limborska 1, Ivan B. Filippenkov1, Yana Y. Shpetko1, Alina E. Denisova2, Vasily V. Stavchansky1, Leonid V. Gubsky2;3, Nikolay F. Myasoedov1, Lyudmila V. Dergunova1

1National Research Centre “Kurchatov Institute”, Moscow, Russian Federation; 2Pirogov Russian National Research Medical University, Moscow, Russian Federation; 3Federal Center for the Brain and Neurotechnologies, Federal Biomedical Agency, Moscow, Russian Federation

Background/Objectives: Adrenocorticotropic hormone (ACTH)-like peptides are being studied as potential neuroprotective agents for stroke therapy. Previously, using RNA-Seq we revealed 315 immune-related differentially expressed genes (DEGs) under ACTH(4-7)PGP (Semax) compared to ACTH(6-9)PGP peptide administration at 4.5h after transient middle cerebral artery occlusion (tMCAO) in rat brain. These genes provided differences between structure of related ACTH-like peptides at the transcriptome level at 4.5h after tMCAO. Here, we used RNA-Seq to compare gene expression profiles induced by the same peptides at 24h after tMCAO.

Methods: Wistar rats, tMCAO, magnetic resonance imaging, RNA-Seq, real-time RT-PCR, bioinformatics.

Results: The hundreds of DEGs (cut-off>1.5 and padj<0.05) were revealed after triple (100mkg/kg) administration of each peptide compared to saline in penumbra-associated frontal cortex of rats at 24h after tMCAO. Moreover, both peptides significantly reduced expression disturbances caused by ischemia for 1171 out of 3774 DEGs (e.g. Hes5, Chrm1, Bdnf, Cd14). However, under ACTH(6-9)PGP versus Semax administration, we identified only 32 DEGs (e.g. Fosb, Egr4, Igsf9, Egr2) at 24h after tMCAO. This number was substantially lower than the number observed at 4.5h after tMCAO.

Conclusion: Both ACTH-like peptides modulated brain gene expression profile disrupted by ischemia at 24h after tMCAO, when demonstrated mostly common transcriptomic effects. Thus, early ours rather than a day after stroke should be chosen to study differences of effects of ACTH-like peptides after cerebral ischemia at the transcriptome level.

Grants: This work was supported by the Thematic plan (5f.5.9.) of NRC “Kurchatov Institute”.

Conflict of Interest: None declared

EP18.005 Genomic risk prediction for common diseases in the rural Pyrenean population

Joan Fibla 1, Marina Laplana1

1University of Lleida - IRBLleida, Basic Medical Sciences, Lleida

Background/Objectives: A singular orography and the administrative-religious limits have shaped the genetic structure of the Catalan Pyrenees population (Fibla et al 2023). It remains to be determined to what extent this structure may have generated interpopulation differences able to be reflected in differences on genetic susceptibility to diseases.

Methods: Genotype data from 397 subjects from the Catalan Pyrenean region was obtained and processed by imputation using the TopMed imputation Server. Genotypes were merged with genotype data from Iberian Peninsula samples of the 1KG3 project (107 individuals). A final set of 504 subjects with genotype data of 10.371.855 SNPs were used.

Polygenic risk score data sets were downloaded from PGS Catalog. We selected data sets based on GWAS studies performed in the European population, with available SNP identifier (rsID). A total of 337 data sets, corresponding to 100 common disorders, were considered.

PGS scores for diseases were obtained using the Plink score routine (Pourcel et al). Mean zScore values and percentile distribution were compared among populations and Pyrenean regions.

Results: zScore distribution (Iberian Peninsula and Pyrenees) was different for 16 disorders with significant adjusted pvalues for Alzheimer_disease (PGS002249) and type2_diabetes_mellitus (PGS000020). In addition, West-East gradient was observed for 11 diseases with significant adjusted pvalues for intracerebral_hemorrhage (PGS003457), Alzheimer_disease (PGS001775_PGS000811_PGS000898), prostate_carcinoma (PGS000742), osteoarthritis (PGS002728) and allergic_disease PGS003487_PGS003489).

Conclusion: Our results point that population structure is also able to shape the distribution of genomic risk to common disease in a rural population. This could be of great interest in planning and monitoring public health strategies.

Grants: Diputació de Lleida

Conflict of Interest: None declared

EP18.006 The causal effect of lipid profiles on coronary heart disease under genetic pleiotropy: a Mendelian randomization study

Ming-Wei Lin 1;2, Yu-Ting Lin1, Yung-Feng Lin3, Hui-Ying Weng2, Chien-Hsiun Chen4, Shih-Feng Tsai2;3;5

1Institute of Public Health, National Yang Ming Chiao Tung University, Taipei, Taiwan; 2Biomedical Industry PhD Program, National Yang Ming Chiao Tung University, Taipei, Taiwan; 3Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan; 4Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; 5Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan

Background: Cardiovascular disease (CVD) is the leading cause of disease burden and mortality globally. Several Mendelian randomization (MR) studies have established the causal relationship between lipid profiles and coronary heart disease (CHD), however, the results of genetic pleiotropy between lipid profiles on CHD were inconsistent. This study aimed to establish a causal relationship between lipid profiles and CHD under various genetic pleiotropic models using MR.

Methods: The study was a one-sample MR design. A total of 128,632 individuals and 9,814,944 genetic variants were obtained from the Taiwan Biobank. The outcome variable was CHD. Significant genetic variants identified from the genome-wide association study (GWAS) of lipid profiles from Taiwan Biobank were used as instrumental variables (IVs). Two-stage least squares (2SLS), Inverse-variance weighting (IVW), MR-Egger, MR-pleiotropy residual sum and outlier (MR-PRESSO) were used for balanced, directional, and horizontal pleiotropy, respectively.

Results: After adjusting for age, gender, an increased risk of CHD was observed per 10 mg/dL increase in TC (OR: 1.858, 95% CI: [1.445, 2.389]) and LDL-C (OR: 1.909, 95% CI: [1.467, 2.484]). An increased risk of CHD was also found per 10 mg/dL increase in TG (OR: 1.502, 95% CI: [1.192, 1.894]) after additionally adjusting for the first ten principle components. Moreover, a decreased risk of CHD per 10 mg/dL increase in HDL-C (OR: 0.756, 95% CI: [0.596, 0.958]) was observed by using a polygenic risk score consisting of 8 functional genetic variants with pleiotropic effects as IV.

Conclusions: Our results confirmed the causal relationships between lipid profiles and CHD.

Grant No: MOST 109-2314-B-010-045-; NSTC 112-2314-B-A49-046-

Conflict of Interest: Ming-Wei Lin NSTC 112-2314-B-A49-046-, National Yang Ming Chiao Tung University, Yu-Ting Lin: None declared, Yung-Feng Lin Taipei Medical University Hospital, Hui-Ying Weng: None declared, Chien-Hsiun Chen Academia Sinica, Shih-Feng Tsai National Health Research Institutes

EP18.007 Genetic Impacts on Early and Late Onset of Menarche: Insights from Four Twin Cohort Studies

Hyojin Pyun 1, Soo Ji Lee1;2, Joohon Sung1;2

1Seoul National University, Graduate School of Public Health, Seoul, Korea, Rep. of South; 2Institute of Health & Environment Associate Research Professor, Seoul, Korea, Rep. of South

Background/Objectives: The age at menarche (AAM) has declined over the past century. While prevailing research has explored genetic and environmental contributions of early menarche (EM) predominantly, insights from twin studies suggest that AAM’s correlations vary, underscoring the necessity for diversified approaches to decode menarche’s complex nature. This study aims to deepen the understanding of factors influencing AAM, spotlighting late menarche (LM).

Methods: We compared the heritability of LM to EM by employing twin cohort studies from Europe, America, and Asia (13,689 participants), and illustrated its fluctuation across the continuum of AAM, offering a granular view of its genetic variance. Additionally, we calculated the proportion of AAM variance explained within- and between-families.

Results: LM demonstrated significantly higher heritability (0.709, SE = 0.023, p < 0.001) than EM (0.463, SE = 0.067, p < 0.001), with a notable effect of common environment on EM. The heritability curve underscored an incremental heritability with advancing AAM, aligning with previous insights. Also, a substantial portion of the variance in AAM occurred within families.

Conclusions: The study reveals distinct heritability between LM and EM, casting light on the critical limitations of applying a uniform framework to decipher the genetic and environmental architecture of AAM. The pronounced within-family variance, especially in non-Asian populations, ignites speculation on the existence of menarche-related genes that are modulated by environmental factors. This nuanced finding beckons further investigation into AAM, advocating for a multifaceted approach to fully understand its underlying mechanisms.

Grants:

Conflict of Interest: None declared

EP18.008 Genetic variability in gene encoding vitamin D binding protein may affect bone mineral density and susceptibility for viral infections after kidney transplantation

Žiga Doljak 1, Tanja Blagus1, Katja Goricar1, Miha Arnol2, Gregor Mlinšek2, Vita Dolzan1

1University of Ljubljana, faculty of medicine, Institute of Biochemistry and Molecular Genetics, Pharmacogenetics Laboratory, Ljubljana, Slovenia; 2University Medical Centre Ljubljana, Department of Nephrology, Ljubljana, Slovenia

Background: Vitamin D has important functions beyond those of calcium and bone homeostasis which include modulation of the innate and adaptive immune responses. After kidney transplantation recipients may develop acute or chronic kidney rejection. Some, but not all studies have reported that vitamin D deficiency can predispose transplant recipients to rejection and chronic complications. It’s been observed that genetic polymorphisms in gene coding for vitamin D-binding protein (VDBP) play an important role in kidney transplant outcomes. The aim of this study was to evaluate the association of VDBP polymorphisms with bone mineral density (BMD) and development of viral infection after kidney transplantation.

Methods: Our study included 144 kidney transplant patients with good graft function. Patients were genotyped for VDBP rs7041, rs4588 and rs2282679 polymorphisms using KASP-PCR. Statistical analysis was performed using nonparametric tests and logistic regression.

Results: VDBP rs7041 polymorphism was associated with lumbar spine BMD. BMD was more frequently decreased in rs7041 AC genotype carriers when compared to AA genotype carriers (OR = 2.83, 95% CI = 1.14-7.01, P = 0.020), the association remained significant after adjustment for age (P = 0.024). VDBP rs7041 was also associated with the risk of developing Human polyomavirus 1 (BKV) and Cytomegalovirus (CMV) infections after transplantation. Carriers of at least one polymorphic rs7041 C allele were less likely to develop these viral infections compared to AA genotype carriers (OR = 0.32, 95% CI = 0.14-0.73, P = 0.007). VDBP rs4588 and rs2282679 polymorphisms didn’t show significant correlation for BMD or viral infections.

Conclusion: VDBP rs7041 polymorphism may be associated with BMD and viral infections after kidney transplantation.

Grants: ARRS grants P1-0170 and P3-0323.

Conflict of Interest: None declared

EP18.009 Getting insight into the extent and genetic basis of inverse comorbidity

Maxim Freidin 1, Nikolai Czajkowski2, Siri Solbakken2, Christopher Sivert Nielsen2

1Queen Mary University of London, London, United Kingdom; 2Norwegian Institute of Public Health, Oslo, Norway

Background: Inverse comorbidity (IC) describes mutually exclusive occurrence of diseases. The extent and genetic bases of IC are seldom investigated. Using UK Biobank (UKB; n = 501,000) and the nationwide Norwegian Patient Registry (NPR, n = 5,000,000), we carried out a systematic search for IC and underlying genetic components.

Methods: Observed-to expected (O2E) ratios were calculated in UKB and validated in the NPR for pairs of ICD-10 diagnosis codes collapsed to 3 digits in individuals aged 40 + . IC was defined as O2E < 1. Polygenic risk scores (PRS) were calculated and cross-tested in the corresponding IC pairs. Global gene expression was analyzed to reveal genes differentially expressed in the opposite direction in IC pairs.

Results: In the whole UKB sample, there were 1,243 pairs of IC (FDR < 0.05). In sex-stratified analysis 23 pairs of IC were found in males and 315 in females. There was a strong correlation between O2E ratios in UKB and the NPR (R = 0.65). An IC of alcoholic liver disease (ALD) and prostate cancer (PC) seen in males was followed up. A PRS for ALD exhibited a protective effect for PC (OR = 0.96, p = 0.002). Forty-four genes were found to be differentially expressed in opposite direction between normal and diseased tissues for PC (GSE104131) and ALD (GSE167308). Many of them are involved in cancer-related pathways and affected by anti-cancer and anti-microbial drugs.

Conclusion: The study suggests the widespread of IC in the general population and provides evidence of genetic factors involved.

Grants: Rosetrees Trust #Seedcorn2022\100003. UK Biobank project # 90865.

Conflict of Interest: Maxim Freidin Rosetrees Trust #Seedcorn2022\100003, Nikolai Czajkowski: None declared, Siri Solbakken: None declared, Christopher Sivert Nielsen: None declared

EP18.010 Causal effects of SARS-CoV-2 infection and COVID-19 hospitalization on complement system activation

Luisa Foco 1, Eva König1, Reinhard Würzner2, Florian Kronenberg3, Peter Pramstaller1, Christian Fuchsberger1, Cristian Pattaro1, Fabiola Del Greco M.1

1Institute for Biomedicine (affiliated to the University of Lübeck), Eurac Research, Bolzano, Italy; 2Institute of Hygiene & Medical Microbiology, Department of Hygiene, Microbiology and Public Health, Medical University of Innsbruck, Innsbruck, Austria; 3Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria

Background/Objectives: The complement system (CS), including classical (CP), alternative (AP) and lectin (LP) pathways, is a key component of innate immunity. CS is putatively activated by SARS-CoV-2 infection, but such causal relation was never demonstrated in general population studies. We tested the causality of SARS-CoV-2 infection and COVID-19 severity on CP, AP, and LP activation, using two-sample Mendelian Randomization (MR) analyses.

Methods: We selected instrumental variables (IV) from the COVID-19 Host Genetics Initiative European ancestry genome-wide association study (GWAS) release 7 (2021), using susceptibility to infection (ncases = 122,616; ncontrols = 2,475,240) and hospitalization (ncases = 16,512; ncontrols = 71,321) as exposures. Genetic associations with CP, LP and AP activation were retrieved from a recent GWAS of the Cooperative Health Research in South Tyrol (CHRIS) study (n = 4990). We performed inverse-variance weighted fixed effect (IVW-FE) meta-analyses of the Wald estimator statistics, using the IVW-random effect meta-analysis, weighted median, weighted mode, and Egger regression as sensitivity analyses.

Results: We identified a causal effect of susceptibility to infection on LP activation (P = 4.5×10-9), with evidence of heterogeneity and pleiotropy driven by a single IV in the ABO gene (I2 = 87.4%, Pheterogeneity = 9.8×10-25). Pleiotropy-robust methods showed direction consistent estimates. Hospitalization was causally associated with lower CP activation (P = 0.029).

Conclusion: COVID-19 severity is causally associated with lower CP activation. The evidence of pleiotropy prevents claims of causality of SARS-CoV-2 infection on LP activation, recently demonstrated by in vitro studies. This raises methodological questions on MR assumptions, which may prevent true causal effect identification.

Grants: Autonomous Province of Bolzano LG14 grant no. D52F20000770003 (PACE).

Conflict of Interest: None declared

EP18.011 Polygenic risk score of C-reactive protein is associated with cardiometabolic outcomes in patients with schizophrenia spectrum disorders

Chenxu Zhao 1, Elnaz Naderi1, Tesfa Dejenie Habtewold1, Richard Bruggeman2, Behrooz Z. Alizadeh1

1University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, Netherlands; 2University of Groningen, University Medical Center Groningen, Rob Giel Research Center, Department of Psychiatry, Groningen, Netherlands

Consortium: GROUP Investigators

Background/Objectives: Low-grade inflammation that characterized by inflammatory biomarkers like C-reactive protein (CRP) and interleukins 6 (IL-6), is highly genetically correlated with cardiometabolic disorders. We investigated the association of polygenic risk scores (PRS) for CRP and IL-6 and cardiometabolic outcomes in patients with schizophrenia spectrum disorders (SSD).

Methods: We included 671 patients with SSD from the Genetic Risk and OUtcome in Psychosis (GROUP) study with available cardiometabolic and genetic data. Clumping and threshold (C + T) and Bayesian method PRS-Continuous Shrinkage (PRS-CS) were used to calculate PRSCRP and PRSIL-6 using summary statistics obtained from the latest genome-wide association studies on CRP and IL-6. Linear regression models adjusted for age, sex, and principal components of genetic ancestry were fitted, with PRSes at P value thresholds (Pt) of <5x10−8, <5x10−6, <0.05, < 0.1, <0.2, <0.5, and <1.0 as main predictors. Cardiometabolic biomarkers included body mass index (BMI), waist circumference (WC), pulse rate, systolic and diastolic blood pressure, mean arterial pressure, glycated hemoglobin, low density lipoprotein, high density lipoprotein cholesterol, triglycerides (TG), and metabolic composite scores (MCS) were outcomes.

Results: Standardized PRSCRP was associated with increased BMI (βPt_0.5 = 0.65; 95% CI = 0.22-1.07; P = 0.003; R2 = 1.85%), WC (βPt_0.5 = 2.30; 0.99-3.60; P < 0.001; R2 = 2.52%), TG (βPt_0.2 = 0.14; 0.008-0.26; P = 0.037; R2 = 1.12%), and MCS (βPt_0.2 = 0.15; 0.03-0.27; P = 0.012; R2 = 1.79%).

Conclusion: PRSCRP but not PRSIL-6 is significantly associated with cardiometabolic outcomes. Our findings contribute to identifying individuals with high susceptibility to cardiometabolic outcomes using PRSCRP and implementation of targeted risk reduction strategies.

Grants: Geestkracht programme of the Dutch Health Research Council (Zon-Mw, grant number 10-000-1001)

Conflict of Interest: None declared

EP18.013 Assessing the association between the APOE gene and Traumatic Brain Injury during Sports: A Systematic Review and Meta-analysis

Leon Ruiter-Lopez 1

1University of Pittsburgh, Dietrich School School of Arts & Science, Pittsburgh, United States

Background/Objectives: Traumatic brain injuries (TBI) are caused by a blow, or jolt to the head which can create chemical changes and pathology. The APOE gene has been associated to an increased risk of TBI, however there have been contradictory findings.

Methods: The aim of this systematic review and meta-analysis is to assess the association between the different variants of the APOE gene and the risk/susceptibility of a TBI during sports. The following search terms were used to identify studies published up to July 2023: (APOE OR Apolipoprotein) AND (Polymorphism OR Gene OR Variance OR Allele) AND (Concussion OR "Brain Injury" OR TBI OR "Head Injury" OR Contusion). PRISMA guidelines were followed. For the statistical analyses Review Manager was used, selecting a Random Effects model. Heterogeneity was assessed using the I2 statistics.

Results: Of the total 347 identified articles, 30 were fully accessed coming to a total of 6 studies that were included in the study. In total 2,303 athletes were involved in the study. The study examined the risk of concussion associated with the APOE Polymorphisms of rs429358 and rs7412 and the promoter rs405509. None of the meta-analyses found an association between the presence of any allele and an increase in the incidence of TBI.

Conclusion: There were no statistical associations between the APOE polymorphisms rs429358 and rs7412 or the genetic variants of the APOE promoter region rs405509 and the risk TBI or concussions in athletes.

Conflict of Interest: None declared

EP18.014 Role of I/D polymorphism in ACE1 gene associated with the COVID-19 severity in young women, but not in children from Ukraine

Tetiana Harashchenko 1, Dmytro Krasnienkov2, Oleksandra Gorodna3, Anastasiia Mohylevets4, Tetiana Kaminska5, Vasiliy Podolskiy1, Tetiana Umanets1, Yuriy Antipkin1, Ludmila Livshits3

1SI “Institute of Pediatrics, Obstetrics and Gynecology named after Academician O.M. Lukyanova, National Academy of Medical Sciences of Ukraine”; 2SI “Institute of Gerontology named after Dmitry F. Chebotarev, National Academy of Medical Sciences of Ukraine”; 3Institute of Molecular Biology and Genetics National Academy of Sciences of Ukraine; 4Taras Shevchenko National University of Kyiv; 5CNE “Kyiv City Children Clinical Hospital of Infectious Diseases”

Background/Objectives: The susceptibility factors to lung injury of COVID-19 are not yet fully understood. Considering that interaction and imbalance between ACE1 and ACE2 play a crucial role in the pathogenesis of lung injury, our study aims to investigate the association of ACE1 I/D polymorphism with lung injury among patients with COVID-19.

Methods: The genotyping for the I/D polymorphism (rs4646994) ACE1 gene in patients with COVID-19 and lung injury (44 – women, 148 – children, and 100 – healthy controls from Ukraine) was performed by using analyses of melting curves of specific PCR products. Two primers were designed for real-time PCR in the presence of EvaGreen I as a fluorochrome followed by melting curve analysis.

Results: Comparative analyses of genotype distribution revealed the statistically significant prevalence of genotype II (0.34) in a group of women with lung injury compared to healthy controls (0.18) (p ≤ 0.05). At the same time, we do not fount a statistical difference between children with COVID-19 and with lung injury with II genotype (0.203) and control group (0.18) (p ≥ 0.05).

Conclusion: Based on the results obtained in this study, genotype II can be proposed as a risk marker for lung injury in adults with COVID-19 but not in children patients.

Grants: NFU “Investigate the importance of medical, biological and sociological factors in the spread of coronavirus infection among women and children in Ukraine” (no. 0120U104508) and MESU “To develop a personalized prognosis of the course of COVID-19 in children based on markers of hereditary predisposition” (no. 0123U102780).

Conflict of Interest: None declared

EP18.015 Optimising circulating tumour cell protocols for transcriptome-wide analyses

Silvia Carlés González1, Ana Fernandez-Montes2, Jose Antonio Trillo-Franco3, Paulina Piario4;5, Lorena Dieguez4;5, Roberto Piñeiro3, Ceres Fernandez-Rozadilla 1

1Santiago biomedical research institute (IDIS), Cancer Predisposition and Biomarkers Lab, Santiago de Compostela, Spain; 2Ourense Clinical University Hospital, Department of Medical Oncology, Ourense, Spain; 3Santiago biomedical research institute (IDIS), Roche Mixed Unit, Santiago de Compostela, Spain; 4International Iberian Nanotechnology Laboratory, Medical Devices, Braga, Portugal; 5RUBYnanomed, Braga, Portugal

Background/Objectives: Liquid biopsy (LB) approaches have emerged in the past 15 years as a way to assess tumour properties from bodily fluids. These offer a less invasive way into tumour biology, development and evolution than classical technologies. Amongst LB methods, the analysis of circulating tumour cells (CTCs) offers an important source of information, particularly in the context oft he metastatic process. Several studies have focused on DANN analyses of these cells, but RNA tests are difficult due to the instability of this molecule, which is further exacerbated by the complex CTC isolation technologies. In this work, weh ave trialled different technologies to isolate CTCs in order to perform downstream single-cell transcriptomic analyses.

Methods: We have used bead enrichment, micromanipulation and RUBYchips to isolate single CTCs in pancreatic cancer patients. Then, weh ave performed total RNA single-cell library preparation with the Takara SMART-Seq Stranded Kit

Results: CTC isolation from metastatic pancreatic cancer patients using EPCAM-independent technologies did not yield important CTC counts for most patients, as opposed to previous reports. Library preparation yielded sequencing-able results for about 30% of the CTCs only.

Conclusion: We envisage that our sample processing times are detrimental for CTC stability, and that thus affects viability to produce efficient RNA transcriptomic libraries.

Grants: This work was supported by project PI19-00179 of the ISCIII AES19. CFR is supported by the CRIS contra el cáncer Postdoctoral Talent Fellowship

Conflict of Interest: Silvia Carlés González: None declared, Ana Fernandez-Montes: None declared, Jose Antonio Trillo-Franco: None declared, Paulina Piario RUBYnanomed, Lorena Dieguez RUBYnanomed, RUBYnanomed, RUBYnanomed, Roberto Piñeiro Roche mixed unit, Ceres Fernandez-Rozadilla: None declared

EP18.016 Parental effects on child ADHD outcomes via DNA methylation at birth – a genetically informed prospective cohort study

Leonard Frach 1, Jean-Baptiste Pingault1

1Division of Psychology and Language Sciences, University College London, Department of Clinical, Educational and Health Psychology, London, United Kingdom

Background/Objectives: DNA methylation (DNAm) is an epigenetic process that has been associated with a variety of environmental exposures as well as with (mental) health conditions. There is mixed evidence for the mediating role of DNAm, or whether DNAm is rather a biomarker of exposures and/or outcomes. Here we investigate the role of DNAm at birth in mediating potential effects of parental factors on child attention deficit hyperactivity disorder (ADHD) outcomes using genetically informed approaches.

Methods: We calculated parental and child polygenic scores for education, smoking and ADHD to examine individual/familial genetic effects on ADHD via DNAm, indicated by methylation profile scores (MPS) for education, smoking during pregnancy and ADHD. Furthermore, we performed two-step within-family Mendelian randomisation to test causal parental effects on child ADHD via DNAm.

Results: Both parental and child polygenic scores were associated with child ADHD outcomes. Parental phenotypes (education, smoking during pregnancy) were associated with their respective MPS in cord blood. Results of the mediation analyses differed between observational regression analyses and genetically informed analyses, and we find little evidence for a causal direct effect of parental factors on ADHD.

Conclusion: Our results suggest that DNAm at birth is associated with parental factors and child ADHD outcomes, however, findings do not provide strong evidence for causal effects.

Grants: This work is supported by the ERC under the European Union’s Horizon 2020 research and innovation programme (#863981).

Conflict of Interest: None declared

EP18.017 Preemptive DPYD Testing in Bulgaria: Variant Prevalence and Implementation Challenges in Clinical Practice

Hristo Ivanov 1;2, Aleksandar Linev1;2, Nelly Miteva-Marcheva1, Dimitar Dimitrov1, Elena Bangieva1, Antonia Vardeva1, Vili Stoyanova1;2

1Medical University Plovdiv, Department of Pediatrics and Medical Genetics, Plovdiv, Bulgaria; 2University Hospital “St. George” Plovdiv, Department of Medical Genetics, Plovdiv, Bulgaria

Background/Objectives: Dihydropyrimidine dehydrogenase (DPYD) plays a crucial role in the metabolism of fluoropyrimidine-based chemotherapies, such as 5-fluorouracil (5-FU). Variants in the DPYD gene can lead to severe toxicity in patients undergoing treatment. This study aimed to investigate the prevalence of DPYD variants in Bulgarian patients and explore the challenges in implementing preemptive DPYD testing in clinical practice.

Materials and Methods: We tested 85 patients for DPYD variants and surveyed 31 oncologists to gain insights into their perspectives on DPYD testing.

Results: Five patients were heterozygous for c.1129-5923C > G, c.1236G>A (HapB3) decreased function allele, and two were heterozygous for c.1905+1G > A (*2A), a no function allele. The primary barriers identified were the financial burden on the patient and difficult access to the test.

Conclusion: The identified DPYD variants are clinically relevant. The prevalence of these variants in the Bulgarian population is similar to the incidence reported in the gnomAD database, suggesting the need for preemptive testing in this population. The barriers to testing underscore the necessity for strategies to improve access and affordability of DPYD testing. These strategies could include the development of pharmacogenetic guidelines, genotyping infrastructure standardization, education activity for all potential stakeholders and national health politics of test reimbursement.

Our findings underscore the importance of preemptive DPYD testing in enhancing patient effective and safe treatment. Further efforts are needed to integrate this test into clinical practice.

Grants: Project № BG-RRP-2.004-0007-С01 Strategic Research and Innovation Program for the Development of MU - PLOVDIV–(SRIPD-MUP)

Conflict of Interest: None declared

EP18.018 Association of the C3953T (rs1143634) variant of the interleukin 1 beta gene with features of the complicated course of COVID-19-associated pneumonia

Natalia Gorovenko 1, Liliia Fishchuk2, Valeriy Pokylko3, Yuliia Cherniavska3, Liudmyla Zhuk3, Viktoriia Vershyhora4, Olena Popova4, Svitlana Podolska1, Olena Ievseienkova1, Oleksandr Buriak2, Volodymyr Olkhovych2, Zoia Rossokha4

1Shupyk National Healthcare University of Ukraine, Kyiv, Ukraine; 2SI National Scientific Center named after M.D. Strazhesko NAMS of Ukraine, Kyiv, Ukraine; 3Poltava State Medical University, Poltava, Ukraine; 4SI “Reference-centre for molecular diagnostic of Public Health Ministry of Ukraine”, Kyiv, Ukraine

Background: The pro-inflammatory cytokine IL-1 has an important role in severe of the COVID-19. A change in IL-1 production may be associated with a mutation in the IL1Β gene. The aim of our study was to analyze the impact of IL1Β gene variants (rs1143634) on disease progression in patients with severe COVID-19 pneumonia, taking into account treatment strategies.

Methods: The study enrolls 117 patients with severe COVID-19 pneumonia. Determination of IL1Β gene variants was performed by PCR-RFLP method.

Results: In the group of patients, the following genotype frequencies were found based on the investigated variant rs1143634 of the IL1Β gene: CC – 65.8%, CT – 28.2%, and TT – 6.0%. Our results show that the group of patients with the CC genotype (compared to CT + TT) of the IL1Β gene had lower leukocyte counts (10.9 ± 8.9*109/L vs. 12.5 ± 4.7*109/L, p = 0.040) and less pronounced lymphopenia (12.3 ± 11.4% vs. 6.6 ± 5.4%, p = 0.007). It was determined that patients carrying the CC genotype stayed on ventilators significantly less (8.4 ± 8.8 days vs. 11.0 ± 8.0 days, p = 0.049) and required shorter treatment with corticosteroids (11.0 ± 5.4 days vs. 13.9 ± 6.2 days, p = 0.045).

Conclusion: The determination of variants of the IL1Β gene can be used as a predictor of the adverse course of COVID-19 pneumonia and for the selection of personalized treatment tactics after conducting studies in the direction of a larger sample of patients.

Grants: No

Conflict of Interest: None declared

EP18.019 Genetic determinants of Helicobacter pylori resistance to clarithromycin and levofloxacin

Larisa Tsapkova 1, Natalia Bodunova1, Vera Polyakova1, Tatiana Yanova1, Anzhelika Chegodar1, Konstantin Rumyantsev1, Dmitriy Bordin1;2;3, Natalya Dekhnich4, Natali Ivanchik4

1SBHI Moscow Clinical Scientific and Practical Center named after A.S. Loginov of DOH, Russian Federation; 2A.I. Evdokimova Moscow State University of Medicine and Dentistry, Russian Federation; 3Tver State Medical University, Russian Federation; 4FSBEIHE Smolensk State Medical University, Russian Federation

Background/Objectives: The Maastricht VI/ Florence consensus recommends, as one of the measures to enhance the efficacy of Helicobacter pylori (H.pylori) infection eradication, a personalized treatment approach involving the selection of an antimicrobial agent based on the pre-determined resistance of H.pylori. To meet the need to develop test systems for personalized drug selection, we conducted a study of H.pylori molecular resistance in Moscow using a self-developed Sanger sequencing test platform.

Methods: We validated our test system on 25 pure culture samples of H.pylori with known re-sistance. Then we tested our platform on 112 H.pylori-positive patients who had not previously received proton pump inhibitors (PPI) or antibacterial drugs. The research method is Sanger sequencing.

Results: The sensitivity and specificity for detecting resistance to clarithromycin was 100%, to levofloxacin - 93% and 92%, respectively. The most common mutations leading to resistance to clarithromycin were 2143G and 2142G, and to levofloxacin - 261A and 271A in the gyrA gene, which accounted for 69% of all identified genetic determinants in levofloxacin-resistant bacteria.

Conclusion: A high level of detection of H.pylori resistance to clarithromycin and levofloxacin in Moscow was demonstrated, comparable to recent studies conducted in Europe. Additionally, specific features of the spectrum and nature of genetic determinants of molecular re-sistance of H. pylori to these antibiotics were identified.

Grants:

Conflict of Interest: None declared

EP18.020 Development of germline NGS Panel for autoimmune diseases: Rheumatoid Arthritis, Systemic Lupus Erythematosus, and Systemic Sclerosis

Maxim Solomadin 1, Abay Baigenzhin2;2, Lina Zaripova2, Larissa Kozina2, Alyona Boltanova2, Talgat Iglikov2

1Karaganda Medical University, Karaganda, Kazakhstan; 2, Astana, Kazakhstan

Background/Objectives: Autoimmune diseases such as Rheumatoid Arthritis (RA), Systemic Lupus Erythematosus (SLE), and Systemic Sclerosis (SSc) present complex pathogeneses with significant genetic predispositions. Current genetic screenings are insufficient in capturing the broad spectrum of genetic variations associated with these conditions.This study aims to develop a comprehensive 120-gene Next-Generation Sequencing panel to identify germline mutations implicated in RA, SLE, and SSc, leveraging extensive PubMed and Google Scholar data and robust in-silico analyses.

Methods: Genes were meticulously selected based on their association with the diseases, as evidenced by literature and database reviews. Each gene’s inclusion was scored for relevance, with a high-throughput NGS platform established for the sequencing process. The panel’s performance was gauged by coverage and uniformity metrics to ensure analytical validity.

Results: The finalized gene panel exhibited high diversity, covering key research areas in skin and connective tissue diseases, musculoskeletal disorders, and immune system pathologies. Preliminary in-silico assessments demonstrate a coverage of >99% and gene uniformity of >90% in wet-lab conditions, indicating a high potential to detect known and novel pathogenic variants.

Conclusion: This gene panel offers a targeted approach to uncover the genetic basis of RA, SLE, and SSc, facilitating the early diagnosis, personalized treatment and a deeper understanding of the molecular mechanisms underlying these autoimmune diseases. Ongoing work will validate this panel in a clinical setting and assess its utility in patient stratification, prognostic evaluations, and as a tool for unveiling the genetic landscape of autoimmune diseases.

Grants: Funding: Ministry of Science and Higher Education of the Republic of Kazakhstan (grant number АR19576783).

Conflict of Interest: None declared

EP18.021 Relationship of rs4680 COMT gene polymorphism, cervical dilation and pain perception during labour

Isabel Erenas1, Nuria García2, Sandra Estepa1, Veronica Velasco3;4, Celia Chicharro1;5, María Sáinz4, Ana Fernández Araque3;4, Fernando Bandres5;6, ZORAIDA VERDE 1;4;5

1Universidad de Valladolid, Biochemistry, Molecular Biology and Physiology,; 2Hospital Comarcal Medina del Campo, Valladolid; 3Universidad de Valladolid, Nursery; 4Universidad de Valladolid, GIR Pharmacogenetics, Cancer Genetics, Genetic Polymorphisms and Pharmacoepidemiology; 5Fundación Ortega-Marañón, Research Group Centro de Estudios Gregorio Marañón, Madrid; 6Universidad Complutense de Madrid, Department of Legal Medicine, Psychiatry and Pathology, Biopathology-Toxicology, Madrid

Background/Objectives: Even though the involvement of COMT rs4680 polymorphism in labour’s anxiety and analgesia has been studied, the role in pain or length of the labour has not been accurately defined. The authors hypothesized that polymorphism in COMT may influence labour progress in a cohort of parturients.

Methods: a prospective observational study was conducted in several obstetric Units of Valladolid Health area. The study was approved by the Valladolid Este Ethics Committee (PI-GR-21-2418), following Helsinki Declaration and data protection law. After blood sample collection, DNA was extracted following standard methods. TaqMan Drug Metabolism Genotyping Assay (Applied Biosystems, Foster City, Ca, USA) was used to genotype all subjects’ COMTVal1158Met (rs4680). The pain perception of parturients was self-evaluated two times according to the VAS (Visual Analogue Scale).

Results: 432 healthy pregnant women in the third trimester were enrolled. The mean age was 33.4 ± 5.8. 21.2% of the deliveries needed cesarean section at term. The mean of pain scores provided for each patient previous and after epidural were 8.2 ± 1.6 and 23. ±2.4, respectively. The frequency of genotypes for COMT rs4680 were (301 AA, 137 GA and 17 GG). Homozygotes for the AA genotype showed less cervical dilation after epidural measurement than those with the GA or GG genotype (p = 0,008). Cervical dilation after epidural was also related to cesarean section (p < 0,001).

Conclusion: There are demographic, clinical and genetic factors that influence individual’s rate of labour progress. This information could be used to improve the prediction of the length of labour.

Conflict of Interest: None declared

EP18.022 Assessing the influence of CYP2C19 gene variants on atorvastatin’s lipid-lowering efficacy in acute coronary syndrome patients

Darius Čereškevičius 1, Kristina Zubielienė1, Ali Aldujeli1, Vacis Tatarūnas1

1Cardiology Institute, Laboratory of Molecular Cardiology, Kaunas, Lithuania

Background/Objectives: Hypercholesterolemia, characterized by elevated levels of cholesterol in the blood, is a major risk factor for cardiovascular diseases, contributing significantly to the global burden of morbidity and mortality (Pirillo & Norata, 2023). Lowering cholesterol levels through the use of 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGCR) inhibitors such as atorvastatin is a key tool in the prevention of atherosclerotic cardiovascular disease (Razavi et al., 2022).

Methods: The study included (n = 93) patients who underwent percutaneous coronary intervention at the Hospital of the Lithuanian University of Health Sciences Kaunas Clinics. All of these patients received atorvastatin treatment for 6 months. Next-generation sequencing, using a custom panel that covered the coding region of CYP2C19, was performed for all patients.

Results: After sequencing, patients were grouped on the basis of their CYP2C19 metabolism into two groups: poor metabolizers and normal metabolizers. Carriers of the CYP2C19*2, CYP2C19*4, and CYP2C19*8 alleles were considered poor metabolizers, whereas all other patients were considered normal metabolizers. The CYP2C19 poor metabolizer phenotype was detected in 29 % of patients. The CYP2C19 poor metabolizer phenotype increased the odss of undertreatment in patients who received standard atorvastatin cholesterol-lowering therapy (OR = 3.603, 95% CI 1.410 – 9.209, p = 0.007).

Conclusion: This study has shown that CYP2C19 variants may have a significant effect on lipid-lowering therapy using atorvastatin. More detailed functional studies are required to show the exact role of CYP2C19 in determining the metabolism and effect of atorvastatin.

Conflict of Interest: Darius Čereškevičius Sequencing equipment supplied by the Department of Genetics and Molecular Medcine of LSMU hospital “Kauno Klinikos”., Funding provided by LSMU science fund., I am employed full time at the Department of Genetics and Molecular Medcine of LSMU hospital “Kauno Klinikos”., Kristina Zubielienė: None declared, Ali Aldujeli: None declared, Vacis Tatarūnas: None declared

EP18.024 Statin pharmacogenomics in patients with familial hypercholesterolemia

Maria Apellaniz-Ruiz 1, Edurne Urrutia Lafuente1;2, Alberto Maillo3, Oscar Teijido Hermida1, Maria Miranda Perez1, David Cobos Hermosa1, David Gomez-Cabrero2;3, Juan Jose Beloqui Lizaso4, Ander Ernaga Lorea5

1Navarra Medical Research Institute - Navarrabiomed - IdiSNA - HUN - UPNA, Genomics Medicine Unit, Pamplona, Spain; 2Navarra Medical Research Institute - Navarrabiomed - IdiSNA - HUN - UPNA, Translational Bioinformatics Unit, Pamplona, Spain; 3King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering Division, Thuwal, Saudi Arabia; 4University Hospital of Navarra (HUN), Pharmacy Department, Pamplona, Spain; 5García Orcoyen Hospital, Endocrinology Department, Estella, Spain

Background/Objectives: Statins are widely used cholesterol-lowering drugs and consequently, reduce cardiovascular disease (CVD) risk. However, considerable interindividual variability exists in response to statins. This is of paramount importance for familial hypercholesterolemia (FH) patients, considered at high-risk for premature CVD. Although some genetic markers associated with statin efficacy/toxicity have been described, most therapeutic decisions remain based on clinical factors. We aimed to perform a comprehensive study analyzing pharmacogenetic alleles associated with statin efficacy/toxicity to personalize FH patients’ treatment.

Methods: WGS data from 501 Spanish clinically-diagnosed FH patients was used to detect variants in genes involved in statin pharmacokinetics and pharmacodynamics (e.g. SLCO1B1, ABCG2, CYP2C9, HMGCR, LPA). Stargazer and Cyrius callers were used. Diplotypes-Phenotypes were annotated following CPIC guidelines. Clinical and treatment data were collected.

Results: In this series, 26.5% of the patients were carriers of LPA-rs3798220 or LPA-rs10455872, associated with lower statin efficacy and increased CVD risk. Concerning phenotypes linked to statin dosing recommendations: 28.1% have decreased or poor SLCO1B1 function, 43.7% were intermediate or poor CYP2C9 metabolizers, and 9.2% have decreased or poor ABCG2 function. Interestingly, 15.8% have ≥two phenotypes associated with increased statin toxicity. Regarding treatment, 89% received lipid-lowering drugs, and of them, 18% experienced statin-associated muscle toxicity, mainly myalgia.

Conclusion: More than 60% of these patients had at least one phenotype associated with statin therapeutic recommendations. Therefore, pharmacogenetic testing, especially of SLCO1B1, ABCG2 and CYP2C9, would help personalize FH treatment. With the goal of reducing cholesterol levels, minimizing adverse reactions, in order to decrease CVD risk.

Grants: La_Caixa_Postdoctoral_Junior_Leader_fellowship code LCF/BQ/PI21/11830009.

Conflict of Interest: Maria Apellaniz-Ruiz Postdoctoral Junior Leader - INCOMING Fellowship from “la Caixa” Foundation (ID: 100010434) and from the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska Curie grant agreement No: 847648. Fellowship code: LCF/BQ/PI21/11830009., Regional strategic project from GEMA call 2019-2021granted by the Government of Navarre (project code: 0011-1411-2019-000049)., Edurne Urrutia Lafuente: None declared, Alberto Maillo: None declared, Oscar Teijido Hermida: None declared, Maria Miranda Perez: None declared, David Cobos Hermosa: None declared, David Gomez-Cabrero: None declared, Juan Jose Beloqui Lizaso: None declared, Ander Ernaga Lorea Regional strategic project from GEMA call 2019-2021granted by the Government of Navarre (project code: 0011-1411-2019-000049).

EP18.025 Study of genetic predisposition to injuries in elite athletes of Kazakhstan

Dauren Yerezhepov 1, Aidana Gabdulkayum1, Saya Amangeldikyzy2, Ainur Akilzhanova1

1Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan; 2Center for Sports Medicine and Rehabilitation, Astana, Kazakhstan

Objective/Background: Athletes carry an increased risk of acute and chronic musculoskeletal injuries. It is known that the composition of muscle fibers can define their strength, elasticity, and exercise tolerance. There have been many genetic studies of predisposition to injuries. Since injuries affect both physically and mentally, leading to a negative impact on performance, this study aimed to identify the genetic predisposition to injuries in Kazakhstani athletes.

Methods: We recruited 84 elite athletes from Kazakhstan’s National Olympic Team of various sport types. The control group consisted of 84 matched non-athlete individuals. All recruited individuals were Kazakhs. Data for injuries was obtained from medical records. Genotyping SNPs of ACTN3 (rs1815739), COL1A1 (rs1800012), COL3A1 (rs1800255), and COL5A1 (rs12722) genes was performed using TaqMan allelic discrimination assay.

Results: ACTN3, COL1A1, and COL3A1 did not show a significant association with injuries in the studied cohort. COL5A1 (rs12722) showed a significant association with injuries in the dominant (CC vs CT + TT, OR 1.91, CI 1.01-3.62, p = 0.046) and overdominant (CC + TT vs. C/T, OR 2.04, CI 1.10-3.79, p = 0.023) models in our studied group.

Conclusion: The SNPs of COL3A1 (rs1800255) and COL5A1 (rs12722) can be used as biomarkers of genetic predisposition to musculoskeletal injuries in Kazakh athletes. Our findings can help develop personalized training injury-preventing programs for athletes. However, more genetic research on larger cohorts is needed.

Grants: This research is funded by the Science Committee of the Ministry of Science and Higher Education of the Republic of Kazakhstan (Grant No. AP19680003).

Conflict of Interest: None declared

EP18.026 Genetic Variants Associated with Iloperidone Response in Acute and Mixed Mania Associated with Bipolar I Disorder

Haimeng Bai 1, Sandra Smiezek1, Rosarelis Torres1, Changfu Xiao1, Christos Polymeropoulos1, Gunther Birznieks1, Mihael Polymeropoulos1

1Vanda Pharmaceuticals Inc., Washington, DC, United States

Background/Objectives: Bipolar I disorder is characterized by episodes of manic and hypomanic activity. Iloperidone is a second-generation antipsychotic that has anti-manic effects. Here we report genetic associations on iloperidone efficacy for treatment of bipolar manic episodes from a phase 3, randomized, placebo-controlled study.

Methods: Whole genome sequencing was conducted using whole blood samples from the study subjects. The efficacy of iloperidone was evaluated using the Young Mania Rating Scale (YMRS).

Results: In order to investigate the genetic factors that impacts the iloperidone efficacy, we conducted GWAS in the Iloperidone group (n = 167) using linear regression on YMRS change with adjustment of AGE, SEX, BMI and the first 2 PCs. Multiple variants were found to be associated with YMRS change at 1e-5 level. Some top hits are RELN (rs55837573, b = -4.61 ± 0.94, p = 2.48e-6), NAV2 (rs1118464, b = -4.15 ± 0.89, p = 6.98e-6), FAT3 (rs76876307, b = 15.44 ± 3.28, p = 5.27e-6), and SHROOM3 (rs62300864, b = 12.08 ± 2.43, p = 1.64e-6). RELN encodes a secretory glycoprotein critical for brain development and synaptic plasticity. Lower Reelin protein levels were reported in bipolar disorder patient brains. These variants are not associated with YMRS at baseline, which suggests potential drug mechanisms. We then defined the drug responders as subjects that having EOS YMRS decreased >=50% compared to baseline. GWAS with logistic regression were conducted, and the association of RELN (rs55837573) was robustly detected in this analysis.

Conclusion: In summary, we report genetic factors associated with the efficacy of iloperidone in treating bipolar manic episodes. This result suggests that the treatment potentially leads to improvement through acting on the identified genes, setting a foundation for further investigation.

Grants:

Conflict of Interest: None declared

EP18.027 High-accuracy prediction of blood biomarker levels using polygenic risk scores in Eastern European patients

Dmitrii Kharitonov 1, Elena Kovalenko1, Alexey Kamelin1, Anna Kim1, Nicolay Plotnikov1, Alexander Rakitko1

1Genotek, Moskva, Russian Federation

Background/Objectives: Polygenic risk scores (PRS), which aggregate the effects of thousands of genetic variants, present a promising method for predicting disease risks. Previous studies have highlighted that machine learning models incorporating PRS can accurately predict the levels of human biomarkers in blood and urine. In this study, we sought to evaluate the efficacy of PRS in predicting biomarker levels within an Eastern European cohort.

Methods: Our cohort comprised 6,690 patients from Eastern Europe.. Each individual was genotyped using the Illumina Global Screening Array version 3 microarray. To extract blood test results, we employed Optical Character Recognition (OCR) and Artificial Intelligence (AI) techniques, processing reports from over 50 distinct laboratories. The resulting dataset included more than 122,000 biomarker observations. We calculated 42 pre-trained polygenic risk scores using Plink 2.0. Additionally, to ensure accurate PRS interpretation for individuals with mixed ancestry, we applied ancestry-specific PRS normalization.

Results: All 42 polygenic risk scores demonstrated significant associations with biomarker levels in the cohort under study. The highest R-squared values were observed for testosterone (0.85), hemoglobin concentration (0.47), and total bilirubin (0.43). Additionally, 49.3% of males in the highest 5% PRS stratum exhibited very high levels of LDL cholesterol (over 4 mmol/L), compared to just 9.7% in the lowest 5% PRS stratum.

Conclusion: Our results indicate a substantial correlation between polygenic risk scores (PRS) and biomarker values, underscoring their potential utility in predicting disease risk and facilitating early diagnosis.

Grants:

Conflict of Interest: None declared

EP18.028 Unravelling the role of FCGR2/3 Locus in disease susceptibility and severity in patients with Kawasaki Disease

VIBHU JOSHI 1, kanika arora1, rakesh kumar pilania1, amit rawat1, Saniya Sharma1, Manpreet Dhaliwal1, surjit singh1, Deepti Suri1

1Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, Pediatrics, Chandigarh, India

Background/Objectives: Kawasaki disease (KD) is an acute febrile systemic vasculitis affecting coronary arteries. Intravenous immunoglobulin (IVIg) is the standard of care for patients with KD. The constant region of IgG mediates its effector functions through Fc gamma receptors (FcγRs). FCGR gene has been implicated as one of the candidate genes for susceptibility to KD. Non Allelic Homologous Recombination in the FCGR2/3 locus leads to genetic variations like Copy Number Region Variations (CNRs), polymorphisms and chimeric genes. Altered FcγR expression can have significant implications on response to IVIg.

The present work was designed to study the role of functionally relevant variations in the FCGR2/3 locus in Indian patients with KD.

Methods: Patients with KD (n = 70), diagnosed based on American Heart Association (AHA) criteria 2017, and healthy controls (n = 24) were enrolled for screening FCGR2/3 locus using MLPA assay, MRC Holland.

Results: CNRs were found in 22/70 (31.4%) patients. None of the controls tested revealed these variations. Odds Ratio (OR) showed that there is causal association of CNRs with KD. Odds of development of Coronary Artery Aneurysms (CAAs) (1.27 (0.47,3.27)) and IVIg resistance (1.65 (0.55,4.67)) were greater in patients with CNRs than without CNRs. Most common polymorphism (rs1801274) affecting the binding affinity of IgG had odds ratio of 1.65 (0.58, 4.55) with respect to development of CAAs.

Conclusion: Patients with KD have CNRs in FCGR2/3 locus. Effect of CNRs correlates with development of CAAs and treatment resistance in these patients. These variants can provide better insights and help understand pathogenesis of KD and response to IVIg.

Grants: N/A

Conflict of Interest: None declared

EP18.029 Estimation of carrier frequencies utilizing the gnomAD database for ACMG recommended carrier screening and Finnish disease heritage conditions in non-Finnish European, Finnish and Ashkenazi Jewish populations

Miska Kandolin 1;2, Minna Pöyhönen1;2, Eveliina Salminen1;2

1University of Helsinki, Dept of Medical and Clinical Genetics, Helsinki, Finland; 2HUSLAB, Diagnostic Center, HUH, Dept of Clinical Genetics, Helsinki, Finland

Background/Objectives: ACMG recommends offering Tier 3 carrier screening to pregnant patients and those planning a pregnancy for conditions with a carrier frequency of ≥1/200 (96 genes for autosomal recessive (AR) conditions). Certain AR conditions referred to as Finnish disease heritage (FINDIS) have a higher prevalence in Finland than elsewhere.

Methods: Data from gnomAD v2.1 was extracted to assess carrier frequencies for ACMG-recommended AR and FINDIS AR and X-linked genes in Finnish, non-Finnish European, and Ashkenazi Jewish populations. The following variants were considered: ClinVar pathogenic or likely pathogenic, loss-of-function, and Finnish founder variants. Gene carrier (GCR), cumulative carrier (CCR), and at-risk couple rates (ACR) were estimated.

Results: In the Finnish population, 47 genes had a GCR of ≥0.5%. CCRs were 52.5% (Finnish), 48.9% (non-Finnish European), and 58.3% (Ashkenazi Jewish), while ACRs were 1.4%, 0.93%, and 2.3% respectively. Approximately 141 affected children with analyzed AR conditions are estimated to be born in Finland annually.

Conclusion: 18 genes causing FINDIS conditions had a GCR of ≥0.5% in the Finnish population but were absent in the ACMG Tier 3 gene list. Two genes (RECQL4, RMRP) had GCR of ≥0.5% either in non-Finnish Europeans or Ashkenazi Jewish populations. Results highlight the need for careful curation of carrier screening panels.

Grants: This study was supported by Helsinki University Hospital Competitive Fund. Biomedicum Helsinki Foundation granted a Start-Up Grant to Miska Kandolin for executing a research project while studying medicine.

Conflict of Interest: None declared

EP18.030 Evaluation of secondary findings detected in exome sequencing data: The experience of our center with 213 cases.

Hasan Baş 1;2, Aysel Ünal1;3, Zeynep Esener1;4

1Diyarbakir Gazi Yasargil Training and Research Hospital, Medical Genetics, Diyarbakir, Türkyie; 2Intergen Genetics and Rare Diseases Diagnosis Center, Medical Genetics, Ankara, Türkyie; 3Antalya Training and Research Hospital, Medical Genetics, Antalya, Türkyie; 4Balikesir University, Faculty of Medicine, Medical Genetics, Balikesir, Türkyie

Background/Objectives: Exome sequencing can diagnose suspicious disease groups, but assessing the extensive dataset may reveal secondary findings of medical significance not reflected in the patient’s phenotype. Our study aims to assess secondary findings identified in cases planned for whole-exome sequencing (WES) or clinical exome sequencing (CES) at our center in relevant categories.

Methods: The data of 129 WES and 84 CES patients were evaluated for secondary findings. Secondary findings were examined in three separate groups: ACMG secondary findings v3.1, carrier status for recessive diseases, and others. Low-penetrance variants or variants associated with non-Mendelian diseases, as well as variants with poor read quality, were excluded from the study. Only pathogenic and likely pathogenic variants were collected.

Results: ACMG secondary findings v3.1 variants were detected in 13 cases (6.1%), with the RYR1 gene variants being the most frequent, observed in 5 cases. Carrier status for at least one disease was identified in 88.3% of patients. While 29.1% of cases had carrier status for a single disease, others were carrier for multiple diseases. Genetic variants were identified in 31% of cases for conditions with adult onset or diseases unknown at the time of diagnosis. The most common were found in G6PD and MEFV genes.

Conclusion: The rate and variety of identified variants were found to be significantly different from other populations. Preliminary data regarding the necessity of community genetic screenings for situations that can impact public health and for which preventive measures can be taken were highlighted.

Grants: None.

Conflict of Interest: None declared

EP18.032 Comprehensive analysis of DPYD variants in a large cohort of 1956 cancer patients undergoing capecitabine-cased therapy: Implications for personalized treatment.

Alessandra Franzoni 1, Jessica Zucco1, Dora Fabbro1, Elena Betto1, Giuseppe Damante1;2

1Institute of Human Genetics, Laboratory medicine Department, Udine; 2University of Udine, Department of Medicine, Udine

Background: Capecitabine, an oral prodrug converted to 5-fluorouracil (5-FU), is a cornerstone in cancer treatment. Dihydropyrimidine dehydrogenase (DPD) encoded by DPYD plays a pivotal role in capecitabine metabolism. Genetic variants in DPYD have been associated with variable drug responses and adverse reactions.

Aim: DPYD variants analysis is important for personalized medicine, as individuals with certain DPYD gene variants may require lower doses of medications or alternative treatment options.

Materials and methods: The study analyzed 1956 oncological patients treated with 5-fluoruracil between 2009 and 2023. DPYD variants (DPYD*2A: c.1905+1G > A, rs3918290; DPYD*13: c.1679T>G, rs55886062; D949V: c.2846A>T, rs67376798; HapB3: c.1236G>A, rs56038477; DPYD*6: c.2194G>A, rs1801160) were evaluated using allelic discrimination. The frequencies of DPYD variants were determined, and associations with clinical parameters and adverse events were explored.

Results: The 1956 analyzed patients are 52% women and 48% men. The genotypes identified were: 1678 DPYD*1/*1 (85.8%), 181 DPYD*1/*6 (9.3%), 59 heterozygous for HapB3 (3%), 11 DPYD *1/*2A (0.6%), 11 heterozygous for D949V (0.6%), 5 DPYD*6/*6 (0.3%), 1 DPYD*2A/*6 (0.1%), 3 compound heterozygous for *2A/HapB3 (0.2%), 3 compound heterozygous for HapB3/*6 (0.3%). No statistical differences between sex (P = 0.058) and the median age is 69.4 years.

Conclusion: The identified variant frequencies in this large-scale cohort and their potential associations with clinical outcomes underscore the need for personalized genotyping to guide treatment decisions. This data-driven approach can contribute to the implementation of precision medicine in oncology, facilitating optimized capecitabine dosing and minimizing the risk of adverse reactions.

Conflict of Interest: None declared

EP18.034 Interaction between genetic variability in rosuvastatin metabolism and infection may explain rhabdomyolysis and consequent acute renal failure

Gregor Mlinšek1;2, Tanja Blagus3, Marija Malgaj Vrečko1, Andreja Aleš Rigler1, Vita Dolzan 3

1University Medical Centre Ljubljana, Department of Nephrology, Ljubljana, Slovenia; 2University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia; 3University of Ljubljana, Faculty of Medicine, Institute of Biochemistry and Molecular Genetics, Pharmacogenetics Laboratory, Ljubljana, Slovenia

Background/Objectives: Rosuvastatin is one of the most frequently prescribed statins and also one of the most frequent causes of rhabdomyolysis with consequent acute renal failure (ARF).We present a case of a 79-years old caucasian woman with an acute myocardial infarction in October 2023, treated with percutaneous coronary intervention, antiaggregation therapy and hypolipidemic therapy (rosuvastatin 40 mg/ezetimibe 10 mg). Her kidney function was good at the time of rosuvastatin prescription with creatinine 55 mM (eGFR > 60 ml/min). A couple of months later she was admitted septic with myoglobin > 50.000 mg/L indicating rhabdomyolysis and creatinine approximately 1600 mM, indicating ARF. Infection was treated with piperacillin/tazobactam. Myoglobin was removed with Theranova dialyzer. The patient was dismissed home two weeks later with creatinine 230 mM. We investigated if interaction between genetic variability in rosuvastatin metabolism and transport and sepsis played a role in the development of rosuvastatin side effects.

Methods: Genotyping for CYP2C9 (rs1799853, rs1057910, rs28371686, rs28371685), SLCO1B1 (rs4149056, rs2306283) and ABCG2 (rs2231142) was performed using competitive allele specific polymerase chain reaction (KASPar).

Results: The patient’s genotypes were CYP2C9 *2/*3 (poor metabolizer), SLCO1B1*37/*37 (normal funcion), and ABCG2 c.421CA (decreased function).

Conclusion: Patient’s risk factors for rhabdomyolysis were high dose of rosuvastatin, slow rosuvastatin metabolism because of polymorphisms in two of the three crucial genes, and patient’s higher age. Sepsis may have been an addidional contributory factor, as infection may have a major effect on drug metabolism and transport through downregulation of CYP enzymes as well as drug transporters.

Grants: ARIS P1-0170

Conflict of Interest: Gregor Mlinšek: None declared, Tanja Blagus: None declared, Marija Malgaj Vrečko: None declared, Andreja Aleš Rigler: None declared, Vita Dolzan Slovenian research Agency grant P1-0170

EP18.035 Calretinin as a predictive biomarker of response to cisplatin-based chemotherapy in malignant mesothelioma

Cita Zupanc1;2, Alenka Franko2;3, Danijela Strbac2;4, Viljem Kovac2;4, Vita Dolzan5, Katja Goricar 5

1Military Medical Unit-Slovenian Army, Ljubljana, Slovenia; 2University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia; 3University Medical Centre Ljubljana, Clinical Institute of Occupational Medicine, Ljubljana, Slovenia; 4Institute of Oncology Ljubljana, Ljubljana, Slovenia; 5University of Ljubljana, Faculty of Medicine, Institute of Biochemistry and Molecular Genetics, Ljubljana, Slovenia

Background: Cisplatin-based chemotherapy is commonly used for treatment of malignant mesothelioma (MM), an aggressive cancer with poor prognosis. Calretinin is an immunohistochemical biomarker used for confirmation of MM diagnosis. Even though calretinin downregulation was associated with cisplatin resistance in cell lines, less is known about its potential as a circulating predictive biomarker of cisplatin response. Our aim was to evaluate the association of serum calretinin concentration and genetic factors associated with calretinin expression with the outcome of cisplatin-based chemotherapy in MM.

Methods: Our study included 265 MM patients. Serum calretinin concentration was determined using ELISA. Patients were genotyped for seven polymorphisms in CALB2, E2F2, MIR335, NRF1 and SEPTIN7 genes using competitive allele-specific PCR. Nonparametric tests, logistic regression and survival analysis were used for statistical analysis.

Results: Calretinin concentration was higher in MM patients who progressed after treatment with cisplatin-based chemotherapy (1.68 versus 0.45 ng/ml, P = 0.001). At a cutoff value of 0.89 ng/ml, sensitivity for predicting disease progression was 0.750 and specificity was 0.714. Calretinin concentration above 0.89 ng/ml was associated with shorter progression-free survival (HR = 1.88, 95% CI = 1.28-2.77, P = 0.001) and overall survival from the start of chemotherapy (HR = 1.91, 95% CI = 1.22-2.97, P = 0.004), even after adjustment for clinical factors (P = 0.006 and 0.049, respectively). MIR335 rs3807348 was associated with better response to cisplatin-based chemotherapy (OR = 2.69, 95% CI = 1.17-6.18, P = 0.020).

Conclusion: We showed that serum calretinin is associated with chemotherapy outcome and survival of MM patients and could serve as a predictive biomarker of cisplatin-based chemotherapy in MM.

Grants: ARIS-P1-0170, L3-8203, L3-2622.

Conflict of Interest: None declared

EP18.036 Hearing difficulty as stroke and atrial fibrillation risk : A Mendelian Randomization study

Jong Hun Kim 1

1Ilsan Hospital, Neurology, Koyang

Consortium: Not applicable

Background/Objectives: Hearing difficulty (HD) is common in the general population, especially among the elderly, and is associated with conditions such as depression and dementia. This study employed Mendelian Randomization (MR) to identify diseases that may be induced by HD and sought to validate these findings using data from Korean National Health Insurance Service (K-NHISS) Health Examination database and the UK Biobank (UKB).

Methods: Two-sample MR analysis was utilized, ensuring that individual populations did not overlap by using GWAS data derived from the UKB for the exposure, and GWAS data from FinnGen for the outcomes. The discovered causal relationships were tested using the K-NHISS health examination database (n = 514,866) and UKB (n = 502,414) through replication studies. In the replication study, we used time-dependent Cox regression to correct for immortal time bias in cross-sectional studies.

Results: A phenome-wide MR study identified stroke (b = -1.71, p = 2.38E-06) and atrial fibrillation (b = -2.60, p = 8.11E-05) as diseases that could be caused by HD. In the K-NHISS Health Examination database, HD showed a higher risk for stroke (HR 1.83, 95%CI 1.69-1.98, p = <0.001) and atrial fibrillation (HR 2.00, 95%CI 1.48-2.69, p < 0.001). In the UKB, HD was associated with an increased risk of stroke (HR 1.60, 95%CI 1.37-1.88, p < 0.001) and atrial fibrillation (HR 1.46, 95%CI 1.33-1.61, p < 0.001).

Conclusion: This study, utilizing Mendelian Randomization (MR) and large-scale population data, provides compelling evidence that HD may be a causal risk factor for both stroke and atrial fibrillation. These findings highlight the potential importance of addressing HD as a preventative measure for broader cardiovascular health.

Grants: Intramural fund

Conflict of Interest: Jong Hun Kim Intramural fund, Full

EP18.037 Exploring causal mechanisms of sex dimorphism in lipid metabolism using the UK Biobank Pharma proteomics data

Daniela Zanetti 1, Mine Koprulu2, Fabio Busonero1, Andrea Maschio1, Francesca Crobu1, Claudia Langenberg2;3;4, Serena Sanna1;5

1Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), Cagliari, Italy; 2University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, MRC Epidemiology Unit, Cambridge, United Kingdom; 3Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Computational Medicine, Berlin, Germany; 4Queen Mary University of London, Precision Healthcare University Research Institute, London, United Kingdom; 5University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, Netherlands

Background/Objectives: Lipid traits are known to be sex-dimorphic, but the sex-specific molecular players are largely unknown. Since protein levels are downstream products of gene expression and can be more directly related to biological processes, proteomics can provide further information for understanding sex-differences in lipids metabolism.

Methods: We used sex-specific pQTL summary statistics for circulating proteins measured in the UK Biobank Pharma Proteomics Project (UKB-PPP), and sex-specific summary statistics of high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C) and triglycerides (TG) from the Global Lipids Genetics Consortium. We selected independently (r2 < 0.001) associated variants (p < 5E10-8) in the UKB-PPP and performed two-sample Mendelian Randomization (MR) analyses.

Results: Out of the 1,463 proteins studied, a total of 1,366 and 1,389 -in men and women respectively- had genome-wide significant hits and were thus selected for MR analyses. Using a Bonferroni-adjusted threshold of pMR < 3.60E10-5, we identified significant causal associations with HDL-C for 5.4% and 6.2% of the proteins analyzed, in men and women, respectively. A slightly higher proportion of significant causal links was detected for LDL-C (6.8% in men and 6.6% in women) while the proportion was lower for TG (4.1% in men and 5.4% in women). Intriguingly, a large fraction of these significant causal links was sex-specific (only causal-significant in men or women) for HDL-C and TG (59.3% and 61.8%, respectively) compared to LDL-C (39.8%).

Conclusions: These results highlight the need to perform sex-stratified QTL mapping and integrative analysis as a route to understanding sex dimorphism in lipid metabolism.

Grants: MSCA Postdoctoral Fellowships 2021 (HORIZON-MSCA2021-PF-01-01).

Conflict of Interest: None declared

EP18.038 Gene-based collapsing analysis in muscle repair genes associated with radiation-induced toxicities across breast, lung and prostate international REQUITE cohorts

Carlos López 1;2, Miguel E. Aguado-Barrera1;2, Olivia Fuentes-Ríos1;2, Javier Manuel Galego Carro1;2, Carla Coedo-Costa1;2, Ana Crujeiras1;2, Patricia Calvo1;3, Ana Carballo1;3, Paula Peleteiro1;3, Begoña Taboada1;3, Antonio Gomez-Caamaño1;3, David Azria4, Manuel Altabas5, Jenny Chang-Claude6, Ananya Choudhury7, Dirk R. de Ruysscher8, Alison M Dunning9, Alexandra Giraldo10, Sara Gutiérrez-Enríquez5, Sarah Kerns11, Maarten Lambrecht12, Monica Ramos10, Tiziana Rancati13, Tim Rattay14, Petra Seibold15, Barry Rosenstein16, Victoria Reyes10, Elena Sperk17, Christopher Talbot14, Liv Veldeman18, Adam Webb14, Catharine West7, Ana Vega1;2;19

1Instituto de Investigación Sanitaria (IDIS), c041-Cancer Genetics and Rare Diseases, Santiago de Compostela, Spain; 2Fundación Pública Galega de Medicina Xenómica, Santiago de Compostela, Spain; 3Hospital Clínico Universitario de Santiago de Compostela CHUS, Santiago de Compostela, Spain; 4ICM (Cancer Institute of Montpellier), Fédération Universitaire d’Oncologie Radiothérapie, Montpellier, France; 5VHIO Vall d’Hebron Institut d’Oncologia, Hereditary Cancer Genetics Group, Barcelona, Spain; 6German Cancer Research Center, Division of cancer epidemiology, Heidelberg, Germany; 7The University of Manchester, Division of Cancer Sciences, Manchester, United Kingdom; 8GROW School for Oncology and Reproduction, Department of Radiation Oncology (Maastro clinic), Maastricht, Netherlands; 9University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Oncology, Cambridge, United Kingdom; 10Vall d’Hebron University Hospital, Department of Radiation Oncology, Barcelona, Spain; 11Medical College of Wisconsin, Department of Radiation Oncology, Milwaukee, United States; 12KU Leuven, Radiation Oncology, Leuven, Belgium; 13Istituto Nazionale dei Tumori, Unit of Data Science, Milano, Italy; 14University of Leicester, Department of Genetics and Genome Biology, Leicester, United Kingdom; 15German Cancer Research Center, Division of Cancer Epidemiology, Heidelberg, Germany; 16Icahn School of Medicine at Mount Sinai, Department of Radiation Oncology, New York, United States; 17Medical Faculty Mannheim, Heidelberg University, Department of Radiation Oncology, Mannheim, Germany; 18University of Ghent, Department of Human Structure and Repair, Gent, Belgium; 19CIBER - Center for Biomedical Research Network, Rare Diseases, Madrid, Spain

Consortium: REQUITE consortium

Background/Objective:_As in pharmacogenomics, radiogenomics studies genetic variation associated with response to radiotherapy (RT). Although RT targets the tumor, adjacent tissues are unavoidably irradiated, often leading to quality-of-life-limiting adverse effects (AE). GWAS on common variants in radiogenomics pinpointed a few candidate genes, including one participating in muscle repair (MR), TANC1[PMID:24974847]. Considering that most organs at risk of developing RT-AE have muscular layers, our objective is to assess the role of MR pathways in the progression of RT-AE through the association study between variants and acute/late RT-toxicities among prostate, breast, and lung cancer patients.

Methodology:_4347_cancer patients treated with radiotherapy (prostate = 1760; breast = 2057; lung = 530) were included (www.requite.eu). Acute/late toxicity was estimated as the residuals of regressing Standardized Total Average Toxicities against clinical covariates and dichotomized by the mean. Genotypes (OncoArray) were imputed, quality-controlled and filtered (0.01 ≤ MAF ≤ 0.1). Association between phenotype and variants within myogenesis (15,361 SNPs) or myoblast fusion (9,908 SNPs) pathways was tested through the Optimal Sequence-Kernel-Association-Test adjusting for population stratification.

Results:_In the myogenesis pathway, CDH4 and CTNNA2 were associated (P-value ≤ 0.05) with late toxicity in breast cancer; and MEF2B with acute toxicity in lung and, late toxicities in prostate and when combining the three cancers. In the myoblast fusion pathway, ADGRB3, FER1LS, KCNH1, MYH9, MYMK and PLEKHD1 were associated (P-value ≤ 0.05) with acute toxicity and, PITX2 and TANC1 with late toxicities when considering all cancers_together.

Conclusions:_Low-frequency variants are potential biomarkers in RT patients. Moreover, we found that both pathways involved in MR are associated with toxicities post-RT and deserve to be further investigated.

Conflict of Interest: None declared

EP19 Population Genetics and Evolutionary Genetics

EP19.001 Ancient DNA analysis of mitochondrial haplogroup variation in a late Avar population (8th century) from the Eastern Croatian site of Šarengrad - Klopare

Dubravka Havaš Auguštin 1, Brina Zagorc2;3, Jelena Šarac1, Mario Novak1, Mario Carić1, Andrea Rimpf4, Daniel Fernandes2;3;5, Inigo Olalde6;7;8, David Reich8;9, Ron Pinhasi2;3

1Institute for Anthropological Research, Centre for Applied Bioanthropology, Zagreb, Croatia; 2University of Vienna, Department of Evolutionary Anthropology, Wien, Austria; 3University of Vienna, Human Evolution and Archaeological Sciences (HEAS), Wien; 4Ilok Town museum, Ilok, Croatia; 5University of Coimbra, Research Centre for Anthropology and Health (CIAS), Department of Life Sciences, Coimbra, Portugal; 6University of the Basque Country UPV/EHU, Department of Zoology and Animal Cell Biology, Vitoria-Gasteiz, Spain; 7Ikerbasque—Basque Foundation of Science, Bilbao, Spain; 8Harvard University, Department of Human Evolutionary Biology, Cambridge, United States; 9Harvard Medical School, Department of Genetics, Boston, United States

Background/Objectives: Avars arrived in the 6th century from the Eurasian steppe to the Pannonian Basin and controlled the region for over 200 years. Here we present maternal genetic ancestry patterns in 43 individuals from the late Avar (8th century) site Šarengrad–Klopare, Croatia to assess their ancestry and level of intermixing with local communities.

Methods: Extraction of aDNA and library preparation were performed in dedicated clean aDNA facilities. Sequencing was performed on Illumina NextSeq500 platform. Haplogrep2 was used to assign mtDNA haplogroups.

Results: 43 mitogenomes were assigned to several predominately European haplogroups (over 95%). Similar to contemporary European populations, haplogroup H is the most prevalent (41.8%). It is followed by T (16.3%), U (14%), J, HV and X (7%, respectively). Two male individuals (4.6%) assigned to haplogroup C, show direct Asian ancestry - one belonging to C4a1a4 (buried with a horse), and his relative to C5c1.

Conclusion: Our findings complement studies of other late Avar sites (8th-9th century) in showing predominantly local haplogroup composition, with minor, but traceable Asian elements in mitochondrial genomes. In our sample Asian ancestry is present only in men of higher social status according to the type of inhumation and kinship analysis.

Grants: Reconstruction of genetic diversity and migratory patterns of late Avar population using ancient DNA analysis“, Austrian Science Foundation, to D.H.A; „The Migration that shaped Europe: Health, Diet and Population Dynamics during Migration Period“, Croatian Ministry of Science and Education to M.N.; “Ancient DNA Atlas of Humanity”, John Templeton Foundation to D.R. and R.P.

Conflict of Interest: None declared

EP19.002 Sports-related polygenic profile in Lithuanian population

Gabija Anikeviciute 1, Valentina Ginevičienė1;2

1Vilnius University, Vilnius, Lithuania; 2Vytautas Magnus University, Kaunas, Lithuania

Total genotype score (TGS) represent polygenic profile or additive effect of genotypes on predicting a complex trait as physical performance. The aim of study was to investigate an athletic polygenic profile of six sports-related SNPs (BDNF rs6265 G/A; VEGF rs2010963 C/G; IL6 rs1800795 G/C; HIF1A rs11549465 C/T; ACE rs4341 I/D; MB rs7293 G/A) in Lithuanian population.

Methods: A total of 191 elite athletes (endurance, sprint/power, mixed) and 205 nonathletic controls (healthy unrelated Lithuanian citizens) were genotyped for 6 SNPs by PCR and restriction fragment length polymorphism method (for ACErs4341, MBrs7293, HIF1Ars11549465) and Real-Time PCR (for BDNFrs6265, VEGFrs2010963, IL6rs1800795). We determined TGS for 6 SNPs in a group of 59 Lithuanian elite athletes and 67 nonathletic controls.

Results: The ACE DD, BDNF CT and HIF1A CT were found as promising markers for sprint/power, MB GG, ACE DD, IL6 CC for mixed athletes, and MB AA for endurance. The mean TGS was significantly higher in sprint/power athletes (60.2 ± 11.5) compared to endurance athletes (54.2 ± 9.2; P = 0.04) and controls (53.4 ± 12.7; P = 0.02). Only one of the best Lithuanian sprint/power athletes (who are also amongst the best in the world) had the best polygenic profile (TGS 75) for up to six SNPs and none of all athletes or controls had the optimal profile (TGS 100).

Conclusion: The TGS was higher in elite sprint/power athletes than in the endurance group or controls. We have identified a polygenic profile that allows us, at least partly, to distinguish elite sprint/power athletes from both endurance athletes and nonathletic population.

Conflict of Interest: None declared

EP19.003 Distribution of polymorphic disease-associated loci of the ATM gene in Northern Eurasian populations

Marina Olkova 1, Igor Gorin1, Andrey Alimov1

1Research Centre for Medical Genetics, Moscow, Russian Federation

Background/Objectives: The ATM gene encodes a serine-threonine protein kinase that is activated by DNA strand breaks. Mutations in this gene are associated with the development of various forms of cancer and are the cause of ataxia-telangiectasia syndrome. The aim of the study was to assess the distribution of polymorphic loci of the ATM gene, presumably associated with the disease, in populations inhabiting Northern Eurasia.

Methods: Seventy-eight SNPs were selected for the study, presumably associated with the development of ataxia-telangiectasia, hereditary tumor syndromes and complications after radiation therapy. Twenty-seven populations were analyzed. The sample size per population ranged from 35 to 123 people. A total of 2,073 DNA samples were kindly provided by the Biobank of Northern Eurasia and genotyped using the Infinium Omni5Exome BeadChip Kit. Statistical data processing was performed in the RStudio environment (snpStats package).

Results: The frequency of alleles and genotypes was determined for each locus. The extended analysis revealed Hardy Weinberg equilibrium disturbances for 13 polymorphic loci, in particular, in “Chukchi/Koryaks/Itelmen” population for rs189037, rs609261, rs664143, rs227092 and rs4585, in “Eastern Transcaucasia” for rs609261, rs664143 and rs4585, and in “Russians of the North of the Arkhangelsk Region” at rs148590073, rs3218674 and rs1801516.

Conclusion: The collected data are of interest for research to assess the risk of developing diseases associated with АТМ gene in populations of Northern Eurasia.

Grants: The research was carried out within the state assignment of MSHE of RF for RCMG.

Conflict of Interest: None declared

EP19.004 Possibly protective role of DNA methylation regulation against ionizing radiation effects in the cohort of the Lithuanian clean-up workers of the Chornobyl nuclear disaster

Katažyna Samaitė 1, Ingrida Domarkienė1, Gabrielė Žukauskaitė1, Vaidutis Kučinskas1, Laima Ambrozaitytė1

1Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania

Background/Objectives: Ionizing radiation (IR) induces genotoxic damage which triggers genomic stability, gene expression and leads to carcinogenesis. We find that some Lithuanian clean-up workers of the Chornobyl catastrophe (LCWC) survived and have aged relatively healthy decades after exposure to high-dose IR. Therefore, we hypothesized that unique genomic variation in LCWC determines their reaction to IR and aimed to perform the analysis in genes that might be involved in genome adaptiveness from the perspective of DNA methylation regulation.

Methods: We compiled a list of 32 genes representing molecular mechanisms of DNA methylation potentially related to the genome integrity after exposure to IR. The study group included 38 male LCWC without a history of cancer. The control group consisted of 27 unrelated males of Lithuanian descent. Genome variants were retrieved from whole-genome short-read sequencing data. T(max) permutation procedure was performed for multiple comparisons using PLINK v1.9 software.

Results: Statistically significant (p ≤ 0.05) differences in the MeCP2 gene variation at the Xq28 locus between LCWC and the control group were identified. Determined only intronic variants that are benign or likely benign.

Conclusion: This pilot study revealed that the genomic variability at the Xq28 locus might be involved in adaptive and protective processes. Possibly through DNA hypermethylation and more effective gene expression repression to avoid mutations occurring after exposure to IR. Though, for more conclusive results further studies are needed.

Grants: This study is a part of the ADAPT/ANELGEMIA (agreement No. S-MIP-20-35/S-MIP-20-34) projects, funded by the Research Council of Lithuania (LMTLT).

Conflict of Interest: None declared

EP19.005 Genome-wide association study of mental disorders in a group of the Lithuanian clean-up workers of the Chornobyl nuclear disaster

Gabrielė Žukauskaitė 1, Ingrida Domarkienė1, Aušra Matulevičienė1, Vaidutis Kučinskas1, Laima Ambrozaitytė1

1Vilnius University, Faculty of Medicine, Institute of Biomedical Science, Department of Human and Medical Genetics, Vilnius, Lithuania

Background. Among the surviving Lithuanian clean-up workers of the Chornobyl nuclear disaster (LCWC), a group of individuals is suffering from anxiety, post-traumatic stress disorder [Gailienė, 2015], and 5.9 times elevated depression risk, even 38 years after the disaster [Kazlauskas et al., 2023]. However, some of LCWC are aging relatively healthy. Therefore, this study aims to determine the genetic factors associated with mental disorders (MD) and protection in LCWC.

Methods. Microarray genotyping (Illumina Infinium OmniExpress–24v1.3) for 93 male LCWC was performed. Mental disorders were assessed during the clinical examination and using the HAD scale. Individuals were categorised into the case (37 LCWC with MD) and control groups (56 LCWC or 182 individuals from the general Lithuanian population without MD). Association analysis was performed using PLINK’s χ2 test. The Humanbase in silico tool was used to analyse gene expression and interaction networks in the central nervous system to evaluate functional significance.

Results. Data analysis indicated that 40% of LCWC suffer from MD, primarily anxiety, depression, emotional instability, and sleep disorders. Conjoint association analysis of MD showed statistical significance (p < 0.05) in 43 SNPs in 15 genes, with ZMIZ1 and TSC22D1 emerging as protective candidates.

Conclusions. The identified loci may influence LCWC mental health and adaptive qualities to extreme conditions, offering insights into the etiopathogenesis of MD. These candidate loci provide a foundation for further research to identify genes (variants) crucial for protection from MD.

Grants. ADAPT (LMTLT- S-MIP-20-35), LITGEN (VP1-3.1-ŠMM-07-K01-013) projects funded by the Research Council of Lithuania.

Conflict of Interest: None declared

EP19.006 Tracing the Slavic origins of Croatians - ancient Y haplogroup distribution at the Jagodnjak site

Jelena Šarac 1, Dubravka Havaš Auguštin1, Brina Zagorc2;3, Mario Novak1, Mario Carić1, Dinko Tresić Pavičić4, Daniel Fernandes2;3;5, Ron Pinhasi2;3

1Institute for Anthropological Research, Centre for Applied Bioanthropology, Zagreb, Croatia; 2University of Vienna, Department of Evolutionary Anthropology, Vienna, Austria; 3Human Evolution and Archaeological Sciences (HEAS), University of Vienna, Vienna, Austria; 4Kaducej d.o.o., Split, Croatia; 5University of Coimbra, Coimbra, Portugal, Research Centre for Anthropology and Health (CIAS), Department of Life Sciences, Coimbra, Portugal

Background/Objectives: Dispersals of Slavs to Croatia and the extent to which this cultural transformation influenced its genetic landscape in Middle Ages is still under debate. The study of Y chromosome haplogroup (hg) diversity is informative for the reconstruction of historic migrations and population admixture. Here, we assess Y chromosome hgs to identify traces associated with Slavic movements in the archaeological site Jagodnjak (9th century CE).

Methods: Laboratory work was performed at the Department of Evolutionary Anthropology, University of Vienna. Shotgun sequencing of 9 male individuals was performed on Illumina NextSeq500 platform and Yleaf program was used to infer Y hgs.

Results: Paternal genetic diversity was high. Half of males were assigned to typical local E1b1 hg. Second most prevalent hg was R1a1a (33%), previously associated with Slavic migrations in Southeastern Europe. An intriguing discovery was an individual carrying R1b1 hg, typical of northern/western Europe. Additionally, a Neolithic signal was embodied in a G2a2 individual.

Conclusion: High Y hgs genetic diversity at Jagodnjak suggests gene flow from diverse regions of Eurasia in Early Middle Ages. The distribution and frequency of main Y hgs aligns with the broader European paternal landscape in medieval times. The high prevalence of R1a hg in the sampled population mirrors modern-day Croatian landscape, suggesting widespread presence of Slavic input in Early Middle Ages.

Grants: Tracing Slavic Origins of Croatians using Ancient Genomes“(JESH, Austrian Science Foundation to J.Š.); „The Migration that shaped Europe: Health, Diet and Population Dynamics during the Migration Period“ (Croatian Ministry of Science and Education).

Conflict of Interest: None declared

EP19.007 Understanding genetic changes between generations in human populations

Alina Urnikyte 1, Laura Pranckeniene1, Vaidutis Kučinskas1

1Vilnius University, Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius, Lithuania

Differences in the relative fitness of genomic variants are foundational, without these, neither natural selection nor adaption can exist. Here, we aim to answer the main question—how commonly are mutations under selection in modern humans, and how do allele frequencies change from generation to generation?

The detection of mutations and the signatures of positive selection and relative fitness across different age groups, including newborns (generation I), their parents (generation II), and the root ancestor (considered as a generation of the 1000 genome project (generation III)), was performed using whole genome sequencing data. Signs of positive natural selection were inferred using Tajima’s D, FST and XP-EHH statistical methods. Selected SNPs were annotated using ANNOVAR. Visual Studio 2017 and C# language were used to write the calculation software for relative fitness, and a graphical presentation and analysis of the results was performed using the Rcmdr and ggplot packages.

This study demonstrated that when exploring the landscapes of relative fitness on each chromosome, the overall pattern of relative fitness background remains consistent across both generations. However, a general tendency of relative fitness decrease was observed. Consequently, in the subsequent generation, the force of natural selection acting upon them is stronger, resulting in a decrease in their cumulative relative fitness. Furthermore, the strong natural selection pressure on the genetic regions that encode the NEGR1 and PTPN1/PTNP21 genes were also identified.

The research has received funding from European Social Fund (project No 09.3.3-LMT-K-712-23-0104) under grant agreement with the Research Council of Lithuania (LMTLT).

Conflict of Interest: None declared

EP19.008 Explaining close relatedness and the founding of the Faroe Islands from a bottleneck effect observed in ROHs

Hannes Gislason 1

1University of the Faroe Islands, Faculty of Science and Technology, Tórshavn, Faroe Islands

Background/Objectives: The population of the Faroe Islands, about 50,000 people, is an isolated community with a high incidence of rare diseases, e.g., Primary Carnitine Deficiency (1:300). For diagnostic purposes, some cases not affected by known mutations were whole-genome sequenced at high coverage (x37). Here, the genomes from consenting individuals are utilized to explore the genomic variation and the ancestral history of the population.

Methods: We determine the number of SNPs and heterozygosity for eight individual and merged samples. We study the pairwise relatedness through kinship, inbreeding from runs of homozygosity (ROHs), and analyse the minor allele frequency distribution. Additionally, the population ancestry and the timing of the founding event are estimated.

Results: Close relatedness was found between all the supposedly unrelated individuals. From ROHs, the high relatedness is interpreted as an ancient property of the isolated population. A bottleneck effect is estimated, starting between years 50 and 300 AD, with a maximum effect in the year 600 AD. A distinct clustering near the central European and British populations of the 1000 Genome Project is likely the result of the population isolation and genetic drift. The minor allele frequency distribution suggests many rare variants.

Conclusion: The ancestry is European, while inbreeding is higher compared to contemporary European populations and population isolates. The Faroese population exhibits inbreeding like ancient Europeans. We discovered a bottlenecked and consanguineous population event starting in the 1st-4th century, potentially 2-3 centuries earlier than previously believed based on archaeological findings.

Grants:

References:

[1] https://doi.org/10.1186/s12864-023-09763-x

[2] https://blogs.biomedcentral.com/bmcseriesblog/2023/12/15/whole-genomes-from-the-population-of-the-faroe-islands/

[3] https://blogs.biomedcentral.com/bmcseriesblog/2023/12/22/highlights-of-the-bmc-series-november-2023/

Conflict of Interest: None declared

EP19.009 A genetic study of the Albanian population using 16 autosomal STR Loci

Ela Zaimi 1, Merita Xhetani2, Alban Haxhia1, Zlatko Jakovski3

1Institute of Forensic Police, Department of Biology and DNA expertise, Tirana, Albania; 2University of Tirana, Faculty of Natural Sciences, Department of Biology, Tirane, Albania; 3University of ST Cyril and Methodius, Institute for forensic medicine, criminology and medical deontology, Skopje, Macedonia

Consortium: NanoAlb

A genetic study of the Albanian population using 15 autosomal STR Loci

Background/Objectives: Allele frequencies change over time due to factors like natural selection, genetic drift, migration and mutation. Larger population samples provide more accurate estimates of allele frequencies compared to small, potentially non-representative samples. Our objective is to study the genetic polymorphism and population genetic parameters of 16 STR loci in a large sample of population.

Methods: DNA was extracted from saliva swabs relating to reference samples from 2000 individuals. Genotyping was performed using AmpFLSTRTM NGM SElectTM kit to obtain allele frequencies for 16 polymorphic STR including D3S1358, vWA, D16S539, D2S1338, D8S1179, D21S11, D18S51, D19S433, THO1, FGA, D10S1248, D22S1045, D2S441, D1S1656, D12S391 and SE33. Allele frequencies and population genetic parameters were analyzed statistically.

Results: Statistical tests on STR genotype data showed no significant deviance from Hardy-Weinberg equilibrium. Similar values for power of discrimination were obtained, but the locus with the highest value PD of 0.9928 is SE33. Polymorphic information content (PIC) greater than 0.70 was observed in all loci, the highest PIC value 0.9368 resulted for SE33 locus. This locus has also the highest values in expected and observed heterozigozity and power of exclusion PE. Lowest values of PIC and expected heterozigozity were detected at D22S1045, whereas the lowest PE value and observed heterozigozity were detected at D10S1248.

Conclusion: These statistical parameters confirm that this combination of loci can provide a powerful information for the study of individual identification, paternity identification and population genetics.

Conflict of Interest: None declared

EP19.010 Distribution of the number of CGG repeats of the FMR1 gene in the Ukrainian cohort

Ivanna Shymanska 1, Volodymyr Kravets1;2, Maria Dushar1, Marta Tyrkus1, Nataliya Matiytsiv1;2, Dmytro Mykytenko3, Halyna Makukh1

1Scientific medical genetic center LeoGENE, Lviv, Ukraine; 2Lviv National Franko University, Lviv, Ukraine; 3Mykytenko Mother and Child Genetic Center, Kyiv, Ukraine

Background/Objectives: Expanse in FMR1 gene are characterized by intellectual disability among males and females, and premature ovarian depletion syndrome. The aim of this study was to establish frequency of the repeats in the FMR1 gene in Ukrainian cohort.

Methods: Fragment analysis was performed by SeqStudio (ThermoFisher) with CarrierMax™ FMR1 Reagent and QD1200 Size Standard. Study included 302 females and 60 males.

Results: The most frequent numbers of CGG repeats in the FMR1 gene among females from Ukrainian cohort are 30, they were detected in 23,1% cases. Premutation (50 – 200 repeats) were detected in compound heterozygous stage with 33 repeats (16,6%), 32 repeats (14,4%), 31 repeats (5,5%). The frequency of the premutation among females from Ukrainian cohort is 3%, that is higher, then in literature – 0,5-0,8%. There wasn’t detected full mutation among females. Among men, the most frequent allele was also 30 repeats (21,4%). There were 3 men with premutation (60, 78, 179 repeats), which is 5%. There were 6 men with a complete mutation (10%), with the number of repeats >200 and >330. We compared results which were got after exome and genome sequencing (short reads) individuals with pre- and full mutations and neither premutation nor mutation didn’t detect. So, the most specificity and sensitivity method for FMR1 gene analysis is fragment analysis

Conclusion: The most frequent numbers of CGG repeats in the FMR1 gene among Ukrainian cohort are 30. Fragment analysis is the most specificity and sensitivity method for FMR1 gene analysis.

Grants:

Conflict of Interest: None declared

EP19.011 Molecular investigation of congenital methemoglobinemia in Tunisian patients

Emna BOUATROUS 1;2, Dorra Chaouachi1, imen boudrigua1, Samia Menif1, Houyem Ouragini1

1Pasteur Institute of Tunis, Laboratory of Molecular and Cellular Hematology, Tunis; 2Faculty of Sciences of Tunis (FST), Tunis, Tunisia

Background/Objectives: Congenital methemoglobinemia is a rare clinical disorder characterized by lifelong cyanosis caused by autosomal dominant mutations in the genes (HBB/HBA) that code for globin proteins collectively known as hemoglobin M. It can also result from autosomal recessive defects in the enzyme NADH cytochrome b5 reductase (NADH-CYB5R), leading to two types of recessive congenital methemoglobinemia (RCM). Type I manifests with cyanosis as the major symptom, while type II is associated with severe mental retardation. These two types are caused by mutations in the CYB5R3 gene, encoding for NADH-CYB5R. The objective of this study is to investigate congenital methemoglobinemia in Tunisian patients.

Methods: Patients having a history of mild to severe cyanosis were enrolled. Methemoglobin levels and NADH-CYB5R activity were measured, hemoglobin profiles were determined, and molecular analysis was conducted.

Results: Preliminary results identified three mutations in four patients. The first mutation, c.190C > T (H64Y) in the HBB gene, led to hemoglobin M-Saskatoon formation. The second mutation, c.535G>A (A179T) in the CYB5R3 gene, was found in two unrelated patients with RCM types I and II. This mutation has been previously associated with RCM type I but not with RCM type II. The third mutation, c.757G>A (V253M), also in CYB5R3, was identified in an RCM type I patient. This mutation is the most commonly reported worldwide and is associated with both RCM type I and type II.

Conclusion: This study reports the first identification of these mutations in the Tunisian population, contributing to precise diagnosis, treatment, genetic counseling, and prenatal diagnosis for severe RCM type II cases.

Conflict of Interest: None declared

EP19.012 Molecular-genetic study of Glucose-6-Phosphate dehydrogenase enzyme deficiency in the Azerbaijani population

Elvira Tahmazli 1, Gunay Akbarova - Ben Tzvi2

1Baku State University, Baku, Azerbaijan; 2, Baku, Azerbaijan

Background/Objectives: Deficiency of the enzyme glucose-6-phosphate dehydrogenase is the most common enzymopathy of the hexose-monophosphate cycle, inherited through an X-linked codominant gene (Xq28) and leading to hemolytic anemia. G6PD deficiency is the most common inherited enzymopathy, affecting 400 million people worldwide, with the highest incidence in Africa, Asia, the Mediterranean and the Middle-East. Material was collected during screening from pupils of 7-11 grades from Masalli discrit of Azerbaijan. As a result of screening of 276 pupils, we found 23 boys with inherited hemizygous type of G6PD enzyme.

Methods: Material collected for biochemical studies was venous blood samples in EDTA- tubes anticoagulant. G6PD enzyme activity was identified with modified fluorescence method. To make the analysis accurate, to identify the inheritance type, we got their parents and family members involved into the study. Totally, there were 302 blood samples processed. Purification of enzyme preparations, their classification were done according to the WHO standardized methods.

Results: As a result of 23 pupils’ screening on G6PD enzyme, different proportion of enzyme deficiency (activity 0-60%) was identified.

Conclusion: So, G6PD enzyme deficiency for Masalli area has shown the following values: 8.33% for phenotypic frequency and 0.0833 (d.f.) for gene frequency. According to the WHO requirements, according to the biochemical characteristics of the identified enzyme deficiency, findings were related to three classes: 13 pupils to the 2nd class, 6 pupils to the 3rd class, and 4 pupils to the 4th. Molecular analysis of G6PD gene identified substitution of c.1178G>A. As a result of the mutation c.1178G-A there was substitution in protein- Arg393His.

Conflict of Interest: None declared

EP19.013 Further analysis of the Transylvanian Seklers based on high-resolution autosomal markers

Zsolt Bánfai 1, Valerián Ádám1, Katalin Sümegi1;2, András Szabó1, Gergely Büki1, Krisztián Erős2, Kinga Hadzsiev1, Ferenc Gallyas2, Attila Miseta3, Miklós Kásler4, Bela Melegh1

1University of Pécs, Clinical Centre, Department of Medical Genetics, Pécs, Hungary; 2University of Pécs, Medical School, Department of Biochemistry and Medical Chemistry, Pécs, Hungary; 3University of Pécs, Medical School, Department of Laboratory Medicine, Pécs, Hungary; 4Institute of Hungarian Research, Budapest, Hungary

Background/Objectives: The Seklers (Szeklers, Székelys) are a Hungarian-speaking, Hungarian-derived catholic ethnic group mainly from Transylvania, Romania. Studies about their within-population structure are scarce and do not discuss their subpopulations. Here we intended to describe the characteristics of Seklers according to our new Transylvanian dataset, which has been expanded since last year.

Methods: We analyzed 554 samples of Sekler individuals collected from 18 distinct regions of Transylvania. Based on their pedigree, their ancestors are from the same ethnicity for at least 3 succeeding generations and lived within a maximum 10 km radius.

Using Illumina 720K genotype data, we conducted investigations analyzing allele frequency and haplotype data, like principal component analysis, ancestry analysis based on maximum likelihood estimation, formal tests of admixture, and various DNA segment analysis methods e.g. identity-by-descent and autozygous segment analysis.

Results: According to the results, most of the Seklers show high homogeneity. Besides the previously identified two settlements Szentegyháza and Szováta, the commune of Tordaszentlászló shows an even more significantly different ancestry, which in contrast to the two previous towns are detectable with all our applied methods.

Conclusion: Based on our results, Seklers in Transylvania are more diverse and structured than we previously thought. One of the newly analyzed regions show well-detectable ancestry difference compared to the other samples with a very high extent, which ancestry component is likely derived from the Asian continent. These samples can help our investigations focusing on the genetic makeup and origin of the Transylvanian Seklers.

Grants: Supported by the NKFIH 138669.

Conflict of Interest: None declared

EP19.014 Matching suicide alleles in 1000G populations and suicide rates in corresponding countries

Erinija Pranckevičienė 1;2, Judita Kasperiuniene2, Adomas Hendrixon3, Laimonas Januska1

1Vilnius University, Department of Human and Medical Genetics, Institute of Biomedical Sciences, Vilnius, Lithuania; 2Vytautas Magnus University, Systems Analysis Department, Faculty of Informatics, Kaunas, Lithuania; 3Vilnius University, Systems Biology Program, Faculty of Medicine, Vilnius, Lithuania

Background/Objectives: Analysis of male and female suicide rates in 159 countries from 2000 till 2019 revealed 8 clusters of countries arranged on high-to-low suicide rate axis. The allele frequencies of variants associated with the suicide phenotype are available for 1000 Genomes Project populations in Ensembl database. We investigated whether a higher/lower prevalence of the suicide-associated alleles in 1000G populations on average correspond to the higher/lower comparative suicide rates in the countries associated with those populations.

Methods: Male and females suicide rate (per 100,000 population) time series were retrieved from the World Bank World Development Indicators database for 159 counties during 2000-2019 (September, 2023). Hierarchical clustering of suicide rates using Euclidean distance and Ward linkage revealed 8 clusters. Ratios of allele frequencies (effect over other) of 13 selected SNPs with no missing values rs336284, rs7049850, rs1402514, rs500042, rs3741042, rs8041188, rs61994674, rs320461, rs9479131, rs62314109, rs6913400, rs1395973, rs9672835 were analyzed by PCA. Deviations of each SNP ratio from its median value across populations was used to score a total prevalence of suicide alleles in each population.

Results: Populations formed distinct clusters in first PCs. Low prevalence of suicide alleles in AFR and AMR populations corresponded to low suicide rates in Nigeria, Gambia Sierra Leone, Kenya, Columbia and Peru. Higher prevalence of suicide-alleles in EUR and SAS populations were consistent with medium and high suicide rates in their corresponding countries.

Conclusion: Majority of the 1000G populations regarding suicide-alleles prevalence on average are in agreement with the suicide rate gradient in their corresponding countries.

Grants:

Conflict of Interest: None declared

EP19.015 Haplogroup structure and genetic variation analyses of complete mtDNA genomes in Iranian ethnolinguistic population

Masoumeh Ghasemi 1, Marzieh Mohseni1, Masoud Edizadeh2, Yasser Riazalhosseini3;4, Kimia Kahrizi1, Hossein Najmabadi1

1Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran; 2GENOKS Genetic Disease Diagnostic Center, Ankara 06560, Turkey; 3Victor Philip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada; 4Department of Human Genetics, McGill University, Montreal, QC, Canada

Background: Iran is an important place in the Middle East to study human variation, ancestry, and history due to numerous human migrations that have left a high level of ethnolinguistic diversity in the region. Mitochondrial DNA (mtDNA) is a useful tool to study human movement and evolutionary history because it has unique features such as no recombination and inheritance.

Methods: We extend a previous comprehensive study of autosomal variation and provide new insights on the maternal genetic history of Iranian ethnolinguistic groups by analyzing the whole mtDNA genomes of four of the most prevalence ethnic groups in Iran population including Persian, Azeri, Lur, and Kurdish. We examined genetic variations in 193 unrelated individuals of Iranian ethnic groups and assigned their haplogroups.

Results: The most frequent haplogroups in the Iranian population are U, followed by H, J1, HV, and T. Every ethnic group was found to have haplogroups from Europe and Eurasia, with Azeris and Persian ethnic groupings having Asian haplogroups as well. These four groupings are Central Iranian Cluster (CIC) people from populations. Additionally African/African American in the Persian group, and Latino/admixed American population in Kurdish were found.

Conclusion: These findings provide support to the racial diversity among the various Iranian ethnic groups.

Grants: USWR/GRC/2023/2878

Institutional ethical approval number: IR.USWR.REC.1402.072

Conflict of Interest: None declared

EP19.017 Association between CCDC3 rs7909670 and smoking on pulmonary function by integrated study of genetics and epigenetics

Hyun Jeong Kim1, Han Byul Jang1, Young-Youl Kim1, Hye-Ja Lee 1

1National Institute of Health Korea, Respiratory and Allergy Diseases, Cheongju, Korea, Rep. of South

Background/Objectives: Smoking is a major factor of the decline in adult pulmonary function. However, comprehensive longitudinal studies related to discover the causality of smoking effects on the decline of lung function. We aim to find out genetic variants associated with the reduction of pulmonary function causing pre-COPD in COPD registry and cohort study.

Methods: We performed a genome-wide association study in KOCOSS patient registry (n = 1,114) using K-chip and confirmed the association in KoGES cohort study (n = 4,335). Also, we integrated DNA methylation and RNA transcription data of CCDC3 gene region.

Results: GWAS identified CCDC3 rs11258050 as a top signal in LD(D’ = 0.98, r2 = 0.82) with rs7909670, which was used for the further study of two data set. CCDC3 rs7909670 was associated with forced expiratory volume at 1 second (FEV1) % and subjects carrying T allele of rs7909670 showed rate of decline in FEV1. In smokers, there was no difference between the genotypes of CC + CT vs. TT. In patients from normal to pre-COPD after 10-year follow-up, smokers carrying TT genotype showed more decreased FEV1 when compared to non-smokers or smokers carrying CC + CT genotypes. In addition, cg23526518 CpG site of TT genotype carriers was hypo-methylated in both KOCOSS and KoGES subjects. Inversely, common genotype was associated with higher rate of pulmonary function and hyper-methylated CpG with decreased RNA expression.

Conclusion: We demonstrated that the risk allele of CCDC3 variant in combination with smoking habits could be a potential risk factor of pulmonary function. It could be helpful understand the decline in lung function by genetic background in relation to smoking.

Conflict of Interest: None declared

EP19.019 Variation of TLR4 gene among Bulgarians revealed by nanopore sequencing – a pilot study

Atanas Syarov 1;2, Adelina Yosifova3, Theodor Todorov2;3, Angelina Trifonova3, Velislava Terzieva3, Radoslava Vazharova2;3

1Lozenetz University Hospital, Laboratory of Medical genetics and molecular biology, Sofia, Bulgaria; 2Sofia University St. Kliment Ohridski, Sofia, Bulgaria; 3Lozenetz University Hospital, Sofia, Bulgaria

Background: Toll-like receptor 4 is a protein that serves as a pattern recognition receptor to induce innate immune responses via downstream signaling pathways. Data about variation of TLR4 and their possible association with infectious or autoimmune diseases in our population are scarce. As part of a project on innate immunity we performed end-to-end nanopore sequencing of the gene.

Materials and methods: The investigated group includes 116 unrelated adult volunteers of Bulgarian descend (46 males/70 females). DNA samples were collected between 05.2020 and 11.2022. 54 individuals not infected by SARS-CoV-2 at that time were considered the control group. 62 volunteers who at least once tested positive for SARS-CoV-2 were referred cases. Targeted end to end sequencing of TLR4 gene was performed using a strategy for LR-PCR amplification of the target region followed by nanopore sequencing on MinION Mk1C system. Software packages MinKNOW, Guppy v.6.2.1, minimap2 v.2.22, VarAFT, GenomeBrowse v.3.0.0 and Haploview 4.1 were used for data analysis.

Results: A total of 74 variants in TLR4 were detected, 59 with MAF > 0.01: 7 in exons, 29 in untranslated regions, 38 intronic, two variants were not reported in public databases. Three variants rs12344353(T), rs10983756(C) and rs11734502(G) revealed a possible association to COVID-19 susceptibility. After applying the Bonferonni correction SNPs remained significant with P values of 0.0113, 0.0138 and 0.0157 respectively (corrected P = .017).

Conclusion: This is the first study using long-read sequencing to characterize TLR4 variation among Bulgarians. Given the small sample size, these encouraging results should be viewed with caution. Funding: BNSF, KP-06-N43/8

Conflict of Interest: None declared

EP19.020 Analysis of the distribution of Factor V (G1691A; H1299R), Prothrombin (G20210A), MTHFR (C677T; A1298C) and PAI-1/SERPINE1 (4G/5G) genetic variants in Georgian population patients with Thrombosis, Pregnancy complications and infertility

Ketevan Kartvelishvili1;2, Nino Pirtskhelani 1;2;3, Nino Kochiashvili1, Tea Mukhuradze4, Tinatin Lezhava1

1Levan Samkharauli National Forensics Bureau, Biology Department, Tbilisi, Georgia; 2Tbilisi State Medical University, Molecular and Medical Genetics, Tbilisi, Georgia; 3Ken Walker International University, Tbilisi, Georgia; 4Bokhua Memorial Cardiovascular Center, Tbilisi, Georgia

Background/Objectives: Thrombophilia, a predisposition to thrombosis, is a significant source of morbidity and mortality due to genetic or/and acquired changes in the clotting mechanism. Inherited thrombophilia may increase the risk of thrombotic events and pregnancy complications. Our previous studies showed high prevalence of thrombophilia in Georgian population and importance of identification of additional genes and variants (FactorVR2, MTHFR(A1298C) and PAI-1/SERPINE1(4G/5G) associated with thromboembolism and pregnancy complications, for successful management of thrombotic disorders. We aimed to analyze prevalence of genetic variants in two groups of patients: with thromboembolism and pregnancy complications/infertility.

Methods: 650 patients were genotyped by multiplex PCR for detection and distribution analyses of six genetic variants (Factor V(G1691A;H1299R), Prothrombin(G20210A), MTHFR(C677T;A1298C) and PAI-1/SERPINE1(4G/5G) in Georgian patients with thromboembolism and pregnancy complications/infertility.

Results: Results of our study, represented in the table, showed statistically significant prevalence of FVL, Factor II, MTHFR(C677T) in patients with thrombosis and MTHFR(A1298C) and PAI-1/SERPINE1(4G/4G) variants in patients with pregnancy complications/infertility. Distribution of Factor VR2 and compound heterozygotes MTHFR(C677T/A1298C) wasn’t statistically different in studied groups.

Conclusion: Based on our study of Georgian patients, implication of thrombophilia testing is important due to different prevalence of genetic variants in studied groups and for the proper personalized management of thrombosis and pregnancy complications/infertility.

Table. Distribution of genetic variants.

Genetic variants

Pregnancy complications/infertility

523(100%)

Thrombosis

127(100%)

Hetero

Homo

Hetero

Homo

FVL(G1691A)

26(5%)

1(0.2%)

15(11.8%)

4(3.1%)

chi^=52.0411,p < .00001

Prot(G20210A)

14(2.7%)

8(6.3%)

chi^=15.0785,p = .000103

MTHFR(C677T)

199(38%)

42(8%)

42(33.1%)

15(11.8%)

chi^=8.0943, p = .004441

MTHFR(A1298C)

261(50%)

82(15.7%)

74(58.3%)

14(11%)

chi^=9.5481,p = .002002

PAI-1/SERPINE1(4G/5G)

245(46.8%)

146(27.9%)

63(49.6%)

28(22%)

chi^=9.2951,p = .002298

FV(H1299R)

54(10.3%)

1(0.2%)

14(11%)

Chi^=0.1303,p = .718137

MTHFR(C677T/A1298C)COMPAUND

109(20.8%)

30(23.6%)

Chi^=2.2696,p = .131932

Conflict of Interest: None declared

EP19.021 Hereditary transthyretin amyloidosis: 5 years study in Spanish population

Marta Domínguez Martínez 1, Alfonso Caro Llopis1, Carla Martín-Grau1, Mónica Roselló Piera1, Juan Silvestre Oltra Soler1, Laia Pedrola1, Sandra Monfort Membrado1, Alba Gabaldon Albero1, Francisco Martinez1, Carmen Orellana1

1Instituto de Investigación Sanitaria La Fe, Valencia, Spain

Background/Objectives: Hereditary transthyretin amyloidosis (hATTR) is an autosomal dominantly inherited disease. It’s characterized by deposits of amyloid fibrils of transthyretin (TTR). It is caused by TTR gene variants, the destabilized TTR protein is prone to dissociate, then to misfold and finally to aggregate into amyloid fibrils in different organs causing a multisystem disease.

More than 100 TTR gene variants have been reported.

Usually appears in the third or fourth decade, affecting both sexes.

Main objectives are: to contribute to early etiological diagnosis of hATTR; to estimate the relative prevalence of the mutations in this gene; to define the mutational spectrum in the Spanish population and its relationship with the clinical presentation

Methods: Blood samples on FTA paper; DNA extraction and analysis of four exons of the TTR gene PCR amplified and Sanger Sequencing.

Results: Over 5 years, a cohort of 2466 patients has been studied with hATTR symptoms: 2118 probands and 348 relatives. We have detected mutations in 8.5% of the cases; 3.1% for probands and 41.7% for relative cases.

The main mutations detected are Val50Met and Val142Ile, in exons 2 and 4 respectively. Both mutations are described like the most prevalent of the disease. Furthermore, we have found another 7 pathological mutations less prevalent and 6 uncertain variants.

Conclusion: Variant spectrum observed is not quite different to the spectrum described for other population; rare variants are less common.

Val142Ile TTR variant is associated with cardiac disease while Val50Met is associated with neurological and/or cardiac disease.

Grants:

Conflict of Interest: None declared

EP19.022 Retrospective Investigation of the Frequency of Pathogenic and Likely Pathogenic Variants in 81 Genes Associated with ACMG Secondary Findings from 1600 Clinical Exome Sequencing

SEHIME TEMEL 1, Mustafa Pir2, Feride Sahin3, Sebnem Ozemri Sag1, Yunus Kasım Terzi3

1Bursa Uludag University, Department of Medical Genetics, BURSA, Türkyie; 2Acibadem University, Department of Biostatistics and Medical Informatics, İstanbul, Türkyie; 3Baskent University, Faculty of Medicine, Department of Medical Genetics, Ankara, Türkyie

Clinical exome sequencing has the potential to detect secondary findings recommended for reporting by the American College of Genetics and Genomics Obtaining unbiased data on the prevalence of these secondary findings is essential to provide a deeper understanding of the potential risks and benefits. Clinical exome sequencing is becoming part of the routine clinical approach Despite the implications of the results, there are challenges in identifying, interpreting, and communicating these findings to patients. The American College of Genetics and Genomics has published guidelines on reporting genomic data in 56 genes associated with clinically important and clinically severe phenotypes to address this issue. As a result of new studies, the guidelines were expanded to 59, 78, and 81 genes, with updates in 2017, 2022, and 2023, respectively. Therefore, within the scope of this study, 1600 clinical exome data from our center were evaluated for the genes belonging to ACMG 81 secondary findings. According to the results of our study, the most common variants reported as pathogenic/likely pathogenic in ClinVar as secondary findings in ACMG 81 gene were MUTYH (%20,69), ATP7B (%17,24), BTD (%8,05), GAA (%8,05) respectively. In the variants that were not presented in ClinVar but considered to be pathogenic/likely pathogenic by InterVar and ANNOVAR prediction algorithms, TTN (61.62%), RYR2 (4.32%), SCN5A (3.24%), respectively. Obtaining these genomic data will enable the management of diseases and determine the population-specific frequency of these genes.

Conflict of Interest: None declared

EP19.024 Haplotype analysis of the VPS33A gene

Saina Novgorodova 1, Aitalina Sukhomyasova1;2, Nadezhda Maksimova1

1North Eastern Federal University, Res. Lab. “Molecular Med. and Human Genetics”, Yakutsk, Russian Federation; 2Republican Hosp. No. 1, Medical Genetical Center, Yakutsk, Russian Federation

Background/Objectives: The objective of this study was to conduct haplotype analysis of the VPS33A gene in individuals affected by mucopolysaccharidosis-plus syndrome (MPSPS).

Methods: DNA samples from 15 patients, 30 healthy relatives, and 50 unrelated healthy individuals were used for screening. Haplotype reconstruction was performed using 11 microsatellite markers located near the VPS33A gene within the 140 cM region. Marker selection was based on databases available on the UCSC Genome Browser. PCR amplification was carried out using a S1000 Touch thermocycler in a 15 μl reaction mixture. Fragment analysis using STR markers and visualization on an Applied Biosystems 3500 DNA sequencer were performed for genetic analysis. GeneMapper Generic software was used for result analysis To identify alleles associated with the VPS33A gene mutation the family analysis of polymorphic markers was conducted. The statistical significance of allele frequency differences was assessed using Pearson’s χ2 criterion with likelihood correction.

Resuls: The identified haplotype D12S385(2)-D12S321(5)-D12S2073(3)-D12S1349(7)-D12S195(4)-D12S1603(2)-D12S378(2)-D12S1612(2)-D12S1614 (3)-D12S342(7) is likely a remain of the founder’s haplotype.

Conclusion: Our findings suggest the presence of a single "founder haplotype" at the MPS-PS locus, indicating that the mutation accumulated through the founder effect. The mutation is estimated to have spread in Yakutia approximately 3997 ± 174.5 years ago. It is estimated that, on average, 159.8 generations have passed since the initial appearance of the c.1492C>T mutation in the Yakut population.

Grants: This study was supported by the grant FSRG-2020-0014 and the "Heads of RS(Y) to young scientists" grant in 2023.

Conflict of Interest: None declared

EP19.026 High frequency of the rare CFTR variant, c.1545_1546del, in Georgian cystic fibrosis patients

Eka Kvaratskhelia1;2, Mariam Ghughunishvili3, Sandro Surmava 1, Elene Abzianidze1;4, Arndt Rolfs5;6;7, Volha Skrahina6, Tinatin Tkemaladze1;3

1Tbilisi State Medical University, Department of Molecular and Medical Genetics, Tbilisi, Georgia; 2Tbilisi State Medical University, V. Bakhutashvili Institute of Medical Boitechnology, Tbilisi, Georgia; 3Tbilisi State Medical University, G.Zhvania Pediatric Academic Clinic, Tbilisi, Georgia; 4Ivane Beritashvili Center Of Experimental Biomedicine, Tbilisi, Georgia; 5Centogene GmbH, Rostock, Germany; 6Agyany Pharmaceutical, Jerusalem, Israel; 7University of Rostock, Rostock, Germany

Background/Objectives: Cystic fibrosis (CF) is one of the most common autosomal recessive diseases in the Caucasian population (MIM#219700) caused by mutations in the CF transmembrane conductance regulator gene (CFTR). The most common CFTR pathogenic variant is c.1521_1523del (p.Phe508del), accounting for ~70% of CFTR mutations worldwide, depending on ethnic group. In this study we analyzed the frequency of the rare c.1545_1546delTA (p.Tyr515X; 1677delTA) variant in Georgian patients with cystic fibrosis. This variant is classified as pathogenic in the context of cystic fibrosis and is associated with the classic form of disease.

Methods: Whole blood from CF patients (n = 45; age 3.7 ± 2.294) was collected via venipuncture at the Givi Zhvania Academic Clinic of Pediatrics (Tbilisi, Georgia). The study was approved by the Ethics Committee of the Tbilisi State Medical University. Written informed consent forms were signed by all parents or legal guardians. Genetic testing of the patients was performed at Centogene GmbH, where DNA extraction, amplification, and CFTR sequencing were carried out.

Results: 21 patients (46.7%) were found to be compound heterozygous with one c.1545_1546del allele and 9 (20%) were homozygous for this allele. Frequency of c.1545_1546del allele was 0.43, while genotype frequency c.1545_1546del/c.1545_1546del was 0.2.

Conclusion: Our data confirm previous studies that have shown the high frequency of the c.1545_1546del variant. Better knowledge of the CFTR variation spectrum may contribute to development of molecular diagnostic tests to optimize medical care of CF patients in Georgia.

Grants: Shota Rustaveli National Science Foundation of Georgia (FR-22-2601). Centogene GmbH BioCyFi (NCT02710383).

Conflict of Interest: None declared

EP20 Functional Genomics

EP20.001 Combining full-length gene assay and SpliceAI to interpret the splicing impact of all possible SPINK1 coding variants

Hao Wu1;2, Jin-Huan Lin1;2, Xin-Ying Tang2;3, Gaëlle Marenne4, Wen-Bin Zou1;2, sacha schutz4;5, Emmanuelle Masson4;5, Emmanuelle Génin4, Yann Fichou4, Gerald Le Gac4;5, Claude Férec4, Zhuan Liao1;2, Jian-Min Chen 4

1Changhai Hospital, Naval Medical University, Department of Gastroenterology, Shanghai, China; 2Shanghai Institute of Pancreatic Diseases, Shanghai, China; 3Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China; 4Univ Brest, Inserm, EFS, UMR 1078, GGB, Brest, France; 5CHRU Brest, Brest, France

Background/Objectives: Single-nucleotide variants (SNVs) in gene coding sequences significantly impact pre-mRNA splicing, crucial for pathogenic mechanisms and precision medicine. This study aims to harness the well-established full-length gene splicing assay (FLGSA), in conjunction with SpliceAI, to prospectively interpret the splicing effects of all potential coding SNVs in the SPINK1 gene, associated with chronic pancreatitis.

Methods: The study involves retrospective and prospective analyses of 27 and 35 SPINK1 SNVs, respectively, followed by data extrapolation and a further experimental validation step (using 5 new SNVs).

Results: Of the 67 FLGSA-analyzed SPINK1 coding SNVs (9.3% of all possible coding SNVs), 12 influenced splicing. Cross-correlation with SpliceAI data suggests that the remaining 653 unanalyzed coding SNVs in SPINK1 are unlikely to affect splicing significantly. Among the 12 splice-altering SNVs, nine produced both normal and aberrant transcripts, and three resulted exclusively in aberrant transcripts. Eleven were missense (2.17% of potential missense variants) and one synonymous (0.61% of synonymous variants). Notably, adjusting SpliceAI cut-off to 0.30 enhanced specificity without reducing sensitivity.

Conclusion: Integrating FLGSA with SpliceAI, we demonstrate that less than 2% (1.67%) of SPINK1 coding SNVs influence splicing outcomes. Our findings underscore the importance of performing splicing analysis in the broader genomic sequence context of the study gene, highlight the inherent uncertainties associated with intermediate SpliceAI scores (ranging from 0.20 to 0.80), and suggest broader implications for transitioning from retrospective to prospective variant analysis in the era of exome and genome sequencing.

Grants: National Natural Science Foundation of China; Shanghai Sailing Program; INSERM; Association des Pancréatites Chroniques Héréditaires; Association Gaétan Saleün.

Conflict of Interest: None declared

EP20.002 Genetic contribution to placental miRNA levels

Antonio Gómez-Martín1;2, Marta Cosín-Tomàs3;4;5, Sofía Aguilar Lacasaña3;4;5, Olga Sánchez6;7, Zoraida García-Ruiz6;7, Hana Vespalcová3;4;5, Payam Dadvand3;4;5, Elisa Llurba6;7;8, Lola Gómez-Roig7;9, Jordi Sunyer3;4;5, Mariona Bustamante 3;4;5

1Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain; 2Escuela Andaluza de Salud Pública (EASP), Granada, Spain; 3ISGlobal, Institute for Global Health, Barcelona, Spain; 4Universitat Pompeu Fabra (UPF), Barcelona, Spain; 5CIBER Epidemiología y Salud Pública, Madrid, Spain; 6Women and Perinatal Health Research Group, Institut de Recerca (IR SANT PAU), Barcelona, Spain; 7Instituto de Salud Carlos III, Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin Network (RICORS-SAMID) (RD21/0012), Barcelona, Spain; 8Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Department of Obstetrics and Gynecology, Barcelona, Spain; 9BCNatal, Hospital Clínic of Barcelona and Hospital Sant Joan de Déu, Barcelona, Spain

Background/Objectives: The placenta, originating from the foetus and situated at the interface between mother and foetus, plays a central role in foetal growth and development. Placental microRNAs (miRNAs), small non-coding RNAs with regulatory functions, are important for various placental activities and have been implicated in obstetric complications. This study aims to elucidate the foetal and maternal genetic contributions to placental miRNA levels by identifying miRNA expression quantitative trait loci (miQTLs).

Methods: The study leverages data from 271 mother-child pairs participating in the population-based Barcelona Life Study Cohort (BiSC). Genotyping of 0.5 million single nucleotide polymorphisms (SNPs) was performed using the GSA array, and the levels of miRNAs were measured through next-generation sequencing (388 after quality control). The association between maternal SNPs and miRNA was tested using linear regression models adjusted for confounders with the TensorQTL tool.

Results: After multiple-testing correction, 11 maternal miQTLs representing 10 loci and 12 miRNAs, were identified. Two of these miQTLs may be attributed to cis effects of the foetal genome, as the SNP-miRNA pairs were closely located in the genome. Other maternal miQTLs pointed to genes related to inflammation and hormone regulation, crucial processes for pregnancy progression. Moreover, the associated miRNAs had previously been linked to pregnancy complications such as preeclampsia or gestational diabetes.

Conclusion: Eleven potential maternal miQTLs linked to inflammation and hormone regulation were identified in the placenta. The next steps will involve identifying foetal miQTLs and distinguishing between maternal and foetal effects.

Grants: ERC (785994), HEI (4959-RFA17-1/18-1), ISCIII-ERDF a way to make Europe (PI20/01116)

Conflict of Interest: None declared

EP20.003 Examining ferroptosis indicators in the neuroblastoma cell line

PINAR KOSEOGLU-BUYUKKAYA 1, Neslihan Coban1

1Istanbul University, Aziz Sancar Institute of Experimental Medicine-Genetics, İstanbul, Türkyie

Background/Objectives: Ferroptosis is a recently discovered form of lipid peroxide-induced iron-dependent cell death. Iron accumulation within the cell and lipid peroxidation are the primary markers of ferroptosis. This study aims to induce ferroptosis in the SH-SY5Y cell line and examine the markers of ferroptosis.

Methods: 500 nM Erastin and 1 µM Ferrostatin-1 (Fer-1) were used in the SH-SY5Y cells to induce and suppress ferroptosis, respectively. In order to verify the proper doses of Erastin and Ferrostatin-1, confocal microscopy and flow cytometry were utilized, and lipid peroxidation levels in cells were determined using BODIPY 581/591 C11 fluorescent dye. The viability of the cells was examined using Cell Counting Kit-8.

Results: It was discovered that cells treated with 500 nM Erastin showed 64% cell viability than control, while it was 103% in the condition of 500 nM Erastin+1 μM Ferrostatin-1. After treatment with Erastin and Fer-1 for 24 hours, lipid peroxidation increased by 75% in Erastin-treated cells compared to control, and lipid peroxidation decreased by 60% in cells treated with Erastin and Ferrostatin-1, according to the analysis performed using confocal microscopy images. In flow cytometry, 30% of Erastin-treated cells were lipid peroxidation positive after an incubation of 6 hours.

Conclusion: Consequently, Erastin and Ferrostatin-1 successfully induced ferroptosis in cell culture. It was shown that, compared to control, Erastin-treated cells had higher levels of lipid peroxidation and lower cell viability. Ferroptosis-related changes in gene expression will be examined.

Grants: This study was supported by Istanbul University Scientific Research Projects Unit. Project number: TDK-2023-39714.

Conflict of Interest: None declared

EP20.004 The canonical splice site variant LZTR1:c.2325+1G > A promotes alternative splicing

Ann-Cathrine Berking 1, Brigitte Pabst1, Uwe R. Kordes2, Lucas Müntnich3, Christian Kratz3, Jens Bohne4, Bernd Auber1, Tim Ripperger1

1Hannover Medical School, Department of Human Genetics, Hannover, Germany; 2University Medical Center Hamburg-Eppendorf, Department of Pediatric Hematology and Oncology, Hamburg, Germany; 3Hannover Medical School, Pediatric Hematology and Oncology, Hannover, Germany; 4Hannover Medical School, Institute of Virology, Hannover, Germany

Background/Objectives: Pathogenic germline variants in LZTR1 are associated with autosomal dominant schwannomatosis and autosomal recessive Noonan syndrome. The germline variant NM_006767.4(LZTR1):c.2325+1G > A was identified in a female index patient with presumed schwannomatosis (i.e., ependymoma and unilateral vestibular schwannoma at age 2 and 13, respectively). The variant has been previously reported in trans with a disease-causing variant in Noonan syndrome. There were two entries in ClinVar (2574541) and two heterozygous carriers in gnomAD v3.1.2 (non-cancer). To elucidate the consequence of this variant on splicing, functional studies were performed.

Methods: Index-derived lymphoblastoid cell lines and various controls were investigated by targeted cDNA-based PCR (exon 17-21), TOPO-TA-cloning and subsequent sequencing.

Results: Besides the wild-type amplicon, we observed an additional larger PCR product in the index and in all controls. While the amplicons were almost equally present in the index patient, controls predominantly showed the smaller PCR product. By sequencing, we showed that the larger amplicon observed in the index and in all controls was caused by intron 19 retention.

Conclusion: Intron 19 retention is an alternative previously unknown splice product of LZTR1 probably due to the small size of intron 19. Alternative splicing can be promoted by the donor splice site variant NM_006767.4(LZTR1):c.2325+1G > A. The alternative splice product leads to a neo-exon with a premature stop codon that is assumed to cause nonsense-mediated decay (NMD) and thus loss-of-function. Further investigations are planned to confirm the expected NMD, to prove alternative splicing in other tissues and to understand the role of this alternative transcript in health and disease.

Grants:

Conflict of Interest: None declared

EP20.005 Differential expression of circular RNAs in the frontal cortex and striatum of the damaged hemisphere of the rat brain after cerebral ischemia

Ivan B. Filippenkov1, Ivan V. Mozgovoy 1, Yana Y. Shpetko1, Alina E. Denisova2, Vasily V. Stavchansky1, Leonid V. Gubsky2;3, Svetlana A. Limborska1, Lyudmila V. Dergunova1

1National Research Centre “Kurchatov Institute”, Laboratory of Human Molecular Genetics, Moscow, Russian Federation; 2Pirogov Russian National Research Medical University, Moscow, Russian Federation; 3Federal Center for the Brain and Neurotechnologies, Federal Biomedical Agency, Moscow, Russian Federation

Background/Objectives: Cerebral ischemia is one of the most severe brain diseases. Previously, we used RNA-Seq to study differential expressed genes (DEGs) with cut-off>1.5 and padj<0.05 in rat brain 24h after transient middle cerebral artery occlusion (tMCAO) model under magnetic resonance imaging (MRI) and histological examination (HE) control. We revealed 2609 DEGs that were overlapped between striatum (S) and dorsolateral region of the frontal cortex (DRFC), which both contained the penumbra area. However, hundreds DEGs were unique for both S and DRFC. Circular RNAs (circRNAs) are covalently closed RNAs that have increased metabolic stability and tissue-specific gene expression regulation properties. Here, we used RNA-Seq to study differentially expressed circRNAs (DECs) in DRFC and S at 24h after tMCAO.

Methods: Wistar rats, tMCAO, MRI, HE, RNA-Seq, real-time RT-PCR, bioinformatics.

Results: We identified 64 and 597 DECs (cut-off>1.5; padj<0.05) associated with DRFC and S, respectively, 24h after tMCAO. Of these, only 7 DECs, encoded by the Cdkn1a, Fgfr1, Elmo1, Ppm1h, Mast3, Elmo1, and Adamts9 genes, overlapped in both tissues. Concurrently, 590 DECs were unique to S and primarily associated with the neurosignaling system. Furthermore, 99 of these DECs were derived from non-DEGs in S.

Conclusion: We revealed the circRNA expression profiles in DRFC and S at 24h after tMCAO. It’s worth noting that circRNAs can have expression profiles unrelated to mRNAs. The study of such circRNAs could be crucial in developing promising systems for diagnosing and treating strokes.

Grants: This work was supported by the Thematic plan (5f.5.9.) of NRC “Kurchatov Institute”.

Conflict of Interest: None declared

EP20.006 De novo missense variants in KDM1A cause a neurodevelopmental disorder

Christian Stockhaus 1, Steffen Syrbe2, Alexandra Afenjar3, Francisco Martinez4, Denisa Weis5, Nicolas Chatron6, Arjan Bouman7, Konrad Platzer1

1Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany; 2Center for Pediatrics and Adolescent Medicine, Division of Pediatric Epileptology, Heidelberg, Germany; 3University Hospitals Pitié Salpêtrière - Charles Foix, Département de Génétique, Paris, France; 4La Fe University and Polytechnic Hospital, Unidad de Genética, València, Spain; 5Johannes Kepler University Linz - JKU, Department of Medical Genetics, Linz, Austria; 6University Hospital of Lyon, Department of Medical Genetics, Lyon, France; 7Erasmus University Medical Center, Department of Clinical Genetics, Rotterdam, Netherlands

Background/Objectives: KDM1A encodes the lysine demethylase 1A and is part of the epigenetic machinery, of which multiple components have been implicated in neurodevelopmental disorders. In 2016 de novo missense variants in KDM1A were first described as causative for developmental delay and distinctive facial morphology in three individuals. A detailed description of a larger cohort of individuals with this rare disease is still lacking. In this study, we describe four additional individuals with de novo variants in KDM1A.

Methods: We collected detailed phenotypic and genetic data with a standardized questionnaire.

Results: The most prevalent features observed in individuals with de novo variants in KDM1A include global developmental delay, behavioral disorders and facial dysmorphism including epicanthus and widely spaced teeth. Causative variants comprise missense variants only. In addition, we were made aware of further individuals with predicted loss-of-function (pLoF) variants in KDM1A. However, with the recent gnomAD v4 release and a LOEUF of 0.64 (pLI = 0) compared to a LOEUF of 0.28 (pLI = 1) in the previous v2.1.1 release, the disease causality of pLoF variants in KDM1A has been challenged.

Conclusion: Our study sheds light on the phenotype of a KDM1A-related neurodevelopmental disorder caused by de novo missense variants. Data on an even larger cohort of affected individuals in addition to, e.g. methylome analyses, are needed to better understand this rare disorder.

Grants: None.

Conflict of Interest: None declared

EP20.007 Exploration of the complex functional consequences of pathogenic variation within the USP7 gene associated with Hao-Fountain syndrome.

Liselot van der Laan 1, Rob Zwart1, Martin A. Haagmans1, Adri N. Mul1, David Dyment2, Pilar Caro3, Sebastian Sailer3, Christian Schaaf3, Marcel Mannens1, Bekim Sadikovic4, Mieke van Haelst1, Peter Henneman1

1Amsterdam UMC, Human Genetics, Amsterdam, Netherlands; 2Children’s Hospital of Eastern Ontario (CHEO), Ottawa, Canada; 3Institute of Human Genetics, Heidelberg University, Heidelberg, Germany; 4London Health Science Centre, Verspeeten Clinical Genome Centre, London, Canada

Background/Objectives: Hao-Fountain syndrome (HAFOUS) is a neurodevelopmental disorder caused by pathogenic variants in USP7, characterized by developmental delay, intellectual disability, speech delay, behavioral abnormalities, autism spectrum disorder, seizures, hypogonadism, and mild dysmorphic features. USP7 encodes a deubiquitinating proteolytic enzyme involved in chromatin remodeling and aberrant DNA methylation (DNAm). A robust episignature for HAFOUS has been reported, and this study aims to elucidate the complex functional consequences of USP7 gene variations.

Methods: For a total set of five HAFOUS patients, of which one was previously confirmed inconclusive with an episignature, and four with pathogenic variants in USP7, fibroblasts (P4-7) were obtained. Five control fibroblast cell lines (P4-11) from healthy controls were achieved. All fibroblasts were cultured and maintained using standard protocols. Genetic variation was confirmed using Nanopore and Sanger sequencing. Moreover, we utilized (1) total mRNA sequencing to analyse the transcriptomic profile of these fibroblasts, and (2) DNA methylation profiles using the EPIC array of Illumina.

Results: Sufficient and good-quality DNA, RNA, and protein fractions were successfully extracted. Differential analyses are pending to date (January 2024) and are expected to be relevant regarding the aim of the study.

Conclusion: While the definitive results are pending, our analysis will quantify the potential relationship between the USP7 DNAm signature in blood vs. fibroblasts. Moreover, we expect to gain insights into the complex functional effect by expression quantitative trait methylation (eQTM) analyses. These forthcoming results from translation analyses are anticipated to enhance our understanding of these molecular mechanisms.

Conflict of Interest: None declared

EP20.008 Stabilized saliva as a non-invasive proxy to blood for whole genome bisulfite sequencing.

Christina Dillane 1, Juan Carmona2, Mike Tayeb1

1DNA Genotek Inc., Ottawa, Canada; 2OraSure Technologies, Inc., Bethlehem, United States

Background/Objectives: Whole genome bisulfite sequencing (WGBS) is a valuable tool to explore the epigenome, offering better resolution of regional methylation than arrays. Due to similarities in cell content between blood and saliva, we investigated the concordance of CpG methylation amongst the two sample types. Additionally, two collection methods of stabilized saliva were assessed: whole saliva and swab-based collection.

Methods: Ten healthy participants collected finger prick blood into EDTA tubes, stabilized whole saliva, and stabilized saliva swabs. Libraries (Accel-NGS Methyl-Seq, Swift Biosciences) were sequenced (Illumina) to 30M reads and aligned to the human genome with Bismark/Bowtie2. R packages utilized included Methylkit, MethylSig, bsseq, and DMRcate.

Results: Percent CpG methylation was not significantly different between blood and whole saliva and Pearson correlation average was high (0.90). Conversely, saliva swabs show a significantly lower percent of CpG methylation than blood or whole saliva (p < 0.002) and lower Pearson correlation to blood (0.78).

To investigate confidence in CpG loci calls, increasing coverage was evaluated. 10X coverage resulted in even, equivalent coverage of saliva to blood along with substantial coverage of the genome including within shores, shelves, and islands.

Comparing sample types, ‘saliva swabs vs. blood’ resulted in the identification of 66,845 Differentially Methylated Regions (DMRs). In contrast, ‘whole saliva vs. blood’ only found 1,451 DMRs.

Conclusion: Stabilized saliva samples are suitable for WGBS. Whole saliva has a higher concordance with blood and fewer DMRs than swab-based collections and could be a strategic, non-invasive proxy to blood-based WGBS in certain studies.

Grants: This study was funded by DNA Genotek.

Conflict of Interest: None declared

EP20.009 GJB2 cis-regulatory elements characterisation in DFNB1 deafness

Valentine Hoyau 1, clara blotas1, Claude Férec1, Stéphanie Moisan2

1Université de bretagne occidentale, UMR1078, Brest; 2CHU Brest, UMR 1078, Brest, France

Background/Objectives: DNA is compacted at different levels of organisation. The smallest consists of chromatin wrapped around nucleosomes. At a larger scale, we see the formation of loops between distant regions. These loops allow the relationship between cis-regulatory elements (CRE) and the promoter to be specifically regulated in time, space and cell type. This control of gene expression is essential for the establishment of mechanisms such as development, differentiation or cell metabolism. Consequently, deregulation of gene expression can lead to disease.

In this case, we are interested in the non-syndromic recessive deafness DFNB1, which is responsible for 1/3 of congenital deafness. These deafness are associated with GJB2 gene variations and large deletions in the DFNB1 locus. Cases of DFNB1 deafness remaining unexplained, these large deletions could lead to the loss of cis-regulatory elements of GJB2. To verify this, some research has focused on the identification of GJB2 cis-regulatory elements.

Methods: The circular chromosome conformation capture technique (4C) is used to reveal interactions between regions. What’s more, activity tests are performed in order to determine the effect of these regions on GJB2 expression.

Results: Previous studies lead to propose a first three-dimensional model of GJB2 regulation and highlight a strong enhancer of GJB2 in endogenous conditions. Some other regions probably involved in GJB2’s regulation remain to be studied.

Conclusion: Elements having an impact on the GJB2 transcription level have been identified in airway epithelial cells, interactions need to be confirmed in mouse cochlea and primary human ear cells obtained through a non-interventional research protocol.

Grants:

Conflict of Interest: None declared

EP20.010 C16ORF70, a novel gene that regulates aging in C. elegans

Valeria Morbidoni 1, Anais Franco Romero2, luca pannone3, Paolo Grumati4, Simone Martinelli3, Marco Sandri2, Eva Trevisson1

1Istituto di Ricerca Pediatrica - Città della Speranza, Dipartimento di Salute della Donna e del Bambino, Università di Padova, Padova, Italy; 2Veneto Institute of Molecular Medicine, VIMM, Dipartimento di Scienze Biomediche, Università di Padova, Padova, Italy; 3Istituto Superiore di Sanità, Dipartimento di Oncologia e Medicina Molecolare, Roma, Italy; 4Telethon Institute of Genetics and Medicine - TIGEM, Pozzuoli (NA), Italy

Background/Objectives: Our group employs the roundworm Caenorhabditis elegans to study uncharacterized genes potentially involved in rare inherited conditions. Considering that some human protein-coding genes could be involved in proteostasis and aging, we started to investigate the function of C16ORF70, a recently identified FoxO-dependent gene, that we renamed MYTHO in H. sapiens and myt-1 in C. elegans.

Materials and Methods: To functionally characterize MYTHO in this multicellular organism, we generated several worm models: 1) a transgenic worm line expressing GFP under the control of the endogenous myt-1 promoter to investigate its expression and 2) worm myt-1 KO strains by CRISPR/Cas9 technology.

Results: myt-1 is mostly expressed in skeletal muscles and neurons and its expression increases in old worms, suggesting an age-related regulation. Worms devoid of myt-1 show a precocious aging phenotype, which results in movement impairment, decreased animal survival upon exposure to oxidative stress and eventually reduced lifespan in KO animals compared to controls. Moreover, myt-1 is involved in some well-known worm longevity pathways, suggesting its importance in longevity promotion. Mechanistically, we found that MYTHO acts as scaffold to recruit and assemble the conjugation system at the phagophore assembly site.

Conclusions: All these data suggest that MYTHO is important, possibly through its crucial role in autophagy, in promoting stress resistance to ensure healthy aging and set the basis for further studies of its mechanisms of action. We are currently analyzing the effect on worm lifespan and health span of ubiquitous and tissue-specific overexpression of myt-1.

Grants: AFM-Telethon (22982); CARIPARO (20/19 FCR).

Conflict of Interest: None declared

EP20.011 Functional characterization of an enhancer regulatory variant rs74477937 via regulating YAE1 expression and potential role in substance use disorder

Hiba Alblooshi 1, Anjali Bharathan2, Anne Jhone1

1The United Arab Emirates University, Genetics and Genomics, United Arab Emirates; 2The United Arab Emirates University, Genomics and Genomics, United Arab Emirates

Background/Objectives: We have identified three novel regulatory single nucleotides polymorphisms (SNPs) rs74477937 (p-value = 8.56 × 10-8), rs118129027 (p-value = 6.24 × 10-8), and rs78707086 (p-value = 8.55 × 10-8) on Ch7p14.1 that were highly suggestive of an association with substance use disorder from GWAS in the United Arab Emirates population. Further delineation is required to unlock the enhancer role of rs74477937 and evaluate the allele-specific enhancer activity and its subsequent effect on the YAE1.

Methods: Enhancer region was cloned into a PGL4.26, then alternative SNP allele “A” was introduced by site-directed mutagenesis using QuikChange II Site-directed mutagenesis kit. Dual-luciferase reporter assay system was used to test the enhancer activity of rs74477937 in SH-SY5Y cells. To construct expression clones for the CRISPR-Cas9 knock out, target-specific CRISPR guide RNAs were designed and cloned into pSpCas9(BB)-2A-Puro(PX459)V2.O plasmid, then the construct was transfected to SH-SY5Y cells. TaqMan gene expression assay for YAE1 was performed on a QuantStudio Flex7.

Results: An enhancer dual-luciferase assay indicates that the rs74477937 risk allele “A” showed significantly higher activity than that the non-risk allele “G” in SH-SY5Y cell lines which supports allele-specific enhancer activity (P = 0.01). Deletion of rs74477937 showed upregulation of the target gene YAE1 (P = 0.0002326) on SH-SY5Y cells consistent with the luciferase assay.

Conclusion: We studied the effect of the rs74477937 enhancer variants on YAE1 gene expression in neural cell lines via CRISPR-Cas9 which supports the enhancer activity of this region to regulate YAE1 expression.

Grants: United Arab Emirates University Start Up Grant (31M502)

Conflict of Interest: Hiba Alblooshi UAE University Start up grant, Full Time, Anjali Bharathan Full time, Anne Jhone: None declared

EP20.012 Exploring the impact of a novel EGLN1 variant on the development of erythrocytosis

Aleša Kristan 1, Aleš Maver2, Tadej Pajič2;3, Saša Anžej Doma4;5, Martina Fink4, Špela Žula4, Rok Količ6, Andrej Vuga6, Petra Hudler1, Tadeja Rezen1, Jurij Stojan1, Tanja Kunej7, Irena Preložnik Zupan4;5, Nataša Debeljak1

1University of Ljubljana, Faculty of Medicine, Institute of Biochemistry and Molecular Genetics, Ljubljana, Slovenia; 2University Medical Centre Ljubljana, Clinical Institute of Genomic Medicine, Ljubljana, Slovenia; 3University of Maribor, Faculty of Medicine, Clinical Biochemistry, Maribor, Slovenia; 4University Medical Centre Ljubljana, Clinical Department of Haematology, Ljubljana, Slovenia; 5University of Ljubljana, Faculty of Medicine, Department of Internal Medicine, Ljubljana, Slovenia; 6Kemomed, Kemomed Research and Development, Ljubljana, Slovenia; 7University of Ljubljana, Biotechnical Faculty, Department of Animal Science, Domžale, Slovenia

Background/Objectives: Hereditary erythrocytosis is a rare disorder with increased erythrocyte count. In long term erythrocytosis, complications like thromboembolic events could emerge, therefore accurate diagnosis is essential for treatment and patients management. The study objective was to identify rare variants with potential clinical impact in a national cohort of hereditary erythrocytosis patients.

Methods: A genetic testing of 28 patients was performed using targeted NGS that covered 39 genes associated with erythropoiesis. Selected variant was further localized on protein structure, and protein expression and activity were assessed with quantitative western blotting and luciferase reporter assay.

Results: One pathogenic variant in the EPAS1 and four variants of unknown significance (VUS) in the EGLN1, JAK2, EPAS1, and SH2B3 were identified. A novel EGLN1 VUS, c.1072C>T (p.(Pro358Ser)), was observed in co-segregation with a disease in one family. We showed that variant is positioned in an active site of EGLN1, suggesting the effect on reduced activity of the protein. Protein quantification results showed statistically significant reduced protein expression of EGLN1 with variant p.(Pro358Ser) in comparison to the wild-type protein. However, luciferase reporter assay did not confirm the effect of variant p.(Pro358Ser) on the EGLN1 activity.

Conclusion: We confirmed the cause for erythrocytosis in one patient and identified novel rare variants with yet unknown significance. We showed that EGLN1 VUS had an effect on EGLN1 expression, however further tests are necessary to resolve the contribution of the variant to erythrocytosis development.

Grants: Slovenian Research Agency grant no. L3-4511, L3-9279, P1-0390, and Young Researcher founding; University Medical Centre Ljubljana grant no. 20170073, 20200231.

Conflict of Interest: Aleša Kristan: None declared, Aleš Maver: None declared, Tadej Pajič: None declared, Saša Anžej Doma: None declared, Martina Fink: None declared, Špela Žula: None declared, Rok Količ: None declared, Andrej Vuga: None declared, Petra Hudler: None declared, Tadeja Rezen: None declared, Jurij Stojan: None declared, Tanja Kunej: None declared, Irena Preložnik Zupan: None declared, Nataša Debeljak Principal investigator of the research grants no. L3-4511 and L3-9279, that are co-financed by Kemomed

EP20.013 Two case reports of Danon Disease: genomics importance in persistent HyperCKemia

Sara Franco Freire 1, Maria Isabel Cabrera Gonzalez1, Marta García de Burgos1, Eva Pitters1, María del Carmen Benito López1, Francisco Javier Mérida de la Torre1, Elena Colastra Ugena1, Yolanda de Diego Otero1, Francisco Miguel rodriguez peña1, RAQUEL YAHYAOUI MACIAS1

1Maternal and Child Hospital, Málaga, Spain

Background: Danon Disease (DD) is a X-Linked dominant skeletal and cardiac muscle disorder caused by the lysosome-associated membrane protein 2 (LAMP2) deficiency. It is a rare disease characterized by severe cardiomyopathy, muscle weakness and intellectual difficulties. Genetic study is crucial in patients with hyperCKemia and myopathic pattern to identify its cause.

Case reports: In our hospital, two cases of Danon disease with variants in LAMP2 gene have been described.

The case 1 was a 7-year-old male with overweight, moderate signs of myopathic involvement and learning difficulties. He presented hypertransaminasemia and hyperCKemia. His mother died because of peritoneal carcinomatosis, his father presented hypertransaminemia and his brother presented low muscle strength, hyperCKemia and learning difficulties. The NGS panel of genes related with muscle diseases was carried out and a hemizygous variant was found, c.183+4_183+5insA (NM_002294.2); g.119590503_119590504insT (NC_000023.10) classified as probably pathogenic. His brother was hemizygous carrier.

The case 2 was a 4-year-old male who presented fatigue in sport, language delay and moderate signs of nonspecific myopathic involvement. He presented hypertrophic cardiomyopathy (HCM), hipertransaminasemia and hiperCKemia. His maternal uncles suffered sudden cardiac death. His mother presented arrhythmogenic cardiomyopathy VI and underwent a heart transplantation. NGS exome showed an unknown stop variant in LAMP2 gene in hemizygosity. His mother was heterozygous carrier.

Conclusion: Myopathic involvement, intellectual disabilities and hyperCKemia in children can be explained by genetic studies, whichidentify cardiomyopathies as Danon Disease.

Conflict of Interest: None declared

EP20.014 Glycemic control in type 1 diabetes and DNA methylation profile

Barbara Cugalj Kern 1;2, Jernej Kovač1;2, Robert Sket1;2, Barbara Jenko Bizjan1;2, Tine Tesovnik1;2, Klemen Dovč1;2, Tadej Battelino1;2, Nataša Bratina1;2

1University Medical Center Ljubljana, Ljubljana, Slovenia; 2Faculty of Medicine, Ljubljana, Slovenia

Background/Objectives: Regulating glycemic control is crucial in preventing diabetes-related complications. Increasing evidence indicates that elevated blood sugar levels affect the development of vascular complications through DNA methylation. Our aim was to identify alterations in the DNA methylation pattern between individuals with type 1 diabetes (T1D) without clinical manifestation of complication with suboptimal and optimal glycemic control.

Methods: The study involved 20 participants with T1D for at least 5 years and aged between 13 and 21 years. Participants’ DNA was isolated from blood samples and pooled together according to their mean values of glycated hemoglobin (HbA1c): HbA1c < 7% (10 participants) vs. HbA1c > 8% (10 participants). Genome-wide DNA methylation was detected on a native DNA using PromethION platform (Oxford Nanopore Technologies). Differential analysis was done with DSS.

Results: Between the two groups 8385 differentially methylated sites were detected in 1802 genes. Among them 4575 CpG sites were hypomethylated and 3810 CpG sites were hypermethylated according to group HbA1c > 8%. Genes annotated to hypomethylated CpG sites were enriched for 48 KEGG signaling pathways.

Conclusion: Our results show changed DNA methylation pattern between the two groups with different regulation of glycemic control in T1D. Furthermore, several of statistically significant signaling pathways were already shown to play a role in vascular complications. These findings contribute to an early recognition of the risk for diabetes-related complications.

Grants: Young Research Fellowship SRA#38769; UMC-tertiary grants: J7-1820, TP20210119 and TP20210130

Conflict of Interest: None declared

EP20.015 Enabling systems biology resources for rare genetic diseases

Friederike Ehrhart 1, Aishwarya Iyer1, Chris Evelo1, Cenna Doornbos2, Peter-Bram t Hoen2

1Maastricht University, Department of Bioinformatics - BiGCaT, Maastricht, Netherlands; 2Radboud University Nijmegen, Department of Medical BioSciences, Nijmegen, Netherlands

Background/Objectives: The identification of genetic variants from whole exome or genome sequencing analysis that contribute to rare genetic disorders depends strongly on what is known about the function of the associated genes. Systems biology approaches allow linking of information from different levels: genetic, gene expression, signalling pathways, and phenotypes. The core challenge here is the availability of well-curated data in a machine-readable way.

Methods: Within the European Joint Programme on Rare Diseases (EJP RD) we have created and curated databases for application in (multi-omics) data analysis and the creation of rare disease networks.

Results: WikiPathways, an open, community-created, expert-curated database for molecular pathways. The rare disease collection within WikiPathways currently contains over 120 rare disease pathways that have been created by clinical and biomedical experts supported by pathway modelling experts. Furthermore, analysis methods and tools on systems biology using molecular pathways, or other resources, were published as an ELIXIR service bundle. The consequent use of identifiers and identifier mapping systems allow integration and extension with other databases - e.g. chemical compounds for drug, nutrition, and toxicology. All of these resources can be found through the EJP RD virtual platform, which links molecular knowledge bases with patient cohorts and clinical expert networks to allow the combined usage in workflows.

Conclusion: Enabling these linked resources contributes to: 1) the understanding of molecular pathways of rare diseases, 2) provides potential targets for treatment, and helps with the understanding of environmental influences when combining networks from toxicology and nutrition.

Grants: EU H2020 N°825575

Conflict of Interest: None declared

EP20.016 Association between core clock genes’ expression and covid-19 severity

Dilek Colak 1, Ibrahim Kaya2, Mustafa Kaya3, Hala Khalil4, Fatimah AlHamlan5, Ayodele Alaiya6, Olfat Alharazi1

1King Faisal Specialist Hospital and Research Centre (KFSHRC), Department of Molecular Oncology, Riyadh, Saudi Arabia; 2Alfaisal University, College of medicine, Riyadh, Saudi Arabia; 3Hacettepe University Medicine Faculty, Türkyie; 4King Faisal Specialist Hospital and Research Centre (KFSHRC), BESC, Riyadh, Saudi Arabia, Saudi Arabia; 5King Faisal Specialist Hospital and Research Centre (KFSHRC), Infection and Immunity Department, Riyadh, Saudi Arabia; 6King Faisal Specialist Hospital and Research Centre (KFSHRC), Department of Cell Therapy and Immunobiology, Riyadh, Saudi Arabia

Introduction: Circadian rhythms, controlled by core clock genes such as CLOCK, BMAL1, PER1-3, and CRY1-2, are fundamental to the regulation of physiological processes including immune responses, and their disruption may have profound effects on the body’s ability to respond to pathogens. Hence, core clock gene expression alterations may contribute to COVID-19 symptom severity among patients. In this study, we aimed to investigate the association between the dysregulation of core clock genes and increased severity of COVID-19.

Methods: We gathered transcriptomic datasets from patients with COVID-19 with detailed clinical information from NCBI-GEO. We performed RNAseq data analysis and studied the expressions of core clock genes and their associations with COVID-19. We then defined circadian-expression score and investigated the COVID-19 severity and immunity-associated genes-score correlation using transcriptomic datasets.

Results: The core clock genes displayed significant expression differences in COVID-19 vs Healthy as well as at different levels of disease severity. Unsupervised hierarchical clustering using the clock genes differentiated patients from health controls. The investigation of the circadian-expression score with the COVID-status indicated that high score was significantly associated with severe disease outcome. Moreover, high gene-set score is significantly correlated with the immunity-associated genes-score (r = 0.86, p < 0.0001).

Conclusions: The severity of COVID-19 appears to be closely linked with changes in the expression of core clock genes that highlights the importance of considering circadian biology in the context of infectious diseases and offers a new perspective for understanding and managing infectious diseases.

Acknowledgment: We would like to thank KFSHRC for supporting this study (RAC#2200022 to DC).

Conflict of Interest: None declared

EP20.018 Functional studies of an enigmatic protein, PLPHP

Jolita Ciapaite1;2, Fried Zwartkruis3, Niklas Darin4, Ralf Husain5, Shimriet Zeidler6, Clara van Karnebeek2;7, Philippa Mills8, Judith Jans1;2, Nanda Verhoeven-Duif 1;2

1UMC Utrecht, Genetics, Utrecht, Netherlands; 2United for Metabolic Diseases; 3UMC Utrecht, Center for Molecular Medicin, Utrecht, Netherlands; 4The Queen Silvia Children’s Hospital at Sahlgrenska University Hospital, Child Neurology, Gothenburg, Sweden; 5Jena University Hospital, Neuropediatrics, Jena, Germany; 6Erasmus Medical Center, Genetics, Rotterdam; 7Amsterdam UMC, Pediatrics, Amsterdam, Netherlands; 8UCL Great Ormond Street Institute of Child Health, London, United Kingdom

Background/Objectives: Biallelic variants in PLPBP, the gene encoding pyridoxal 5′-phosphate homeostasis protein (PLPHP) cause vitamin B6-dependent epilepsy. The molecular function of PLPHP is unknown. The diagnosis can be challenging, as no biomarkers or functional studies are available.

We showed that pyridoxal 5′-phosphate was decreased in patient’s fibroblasts and PLPHP-deficient HEK293 cells. Culturing with pyridoxine or pyridoxamine led to accumulation of pyridoxine 5′-phosphate and pyridoxamine 5′-phosphate. Experiments with 13C4-pyridoxine revealed increased fractional turnover of pyridoxal and pyridoxal phosphate. Although these findings did not lead to the clear determination of the protein’s function, we hypothesized that flux experiments could be used for the study of variants of unknown significance.

Methods: We examined formation of pyridoxine phosphate and pyridoxal phosphate in fibroblasts from patients with proven PLPHP-deficiency and from patients with new variants in this gene.

Results: The findings in the cells with new variants suggested normal function of PLPHP in one patient, but were inconclusive in another, as results were only partly abnormal (pyridoxal phosphate decreased, but pyridoxine phosphate was not increased).

Conclusion: A definite conclusion in the variant could not be made. These results illustrate the difficulties of classifying variants in genes of unknown function.

Grants: -

Conflict of Interest: None declared

EP20.019 uORF creation as a new mechanism of CDKN1C loss-of-function in Beckwith-Wiedemann Syndrome

Luisa Vergori 1;2, Frederic Brioude3;4, Omar Soukarieh5;6, Caroline Meguerditchian7, Yves Delneste1, Alain Morel1;2, David-Alexandre Tregouët5, Isabelle Tournier1;2

1Univ Angers, UMR Inserm 1307 - CNRS 6075, CRCI2NA, Innate Immunity and Cancer, Angers, France; 2Integrative Center for Oncology, ICO, Functional Genomics Unit, Angers, France; 3Sorbonne University, UMR Inserm 938, Saint-Antoine Research Centre, Institute of Cardiometabolism and Nutrition, Paris, France; 4Assistance Publique-Hôpitaux de Paris, Armand Trousseau University Hospital, Department of Molecular Biology and Genetics, Paris, France; 5Univ. Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR 1219, Bordeaux, France; 6Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, Pessac, France; 7Univ. Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR 1219, Bordeaux, France

Background / Objectives: Loss-of-function mutations in Cyclin-dependent kinase inhibitor 1C (CDKN1C) are associated with Beckwith–Wiedemann Syndrome (BWS), an overgrowth disorder. We have identified novel variants in CDKN1C 5’-untranslated region (5’UTR) in two cases of BWS. These variants were predicted by the MORFEE bioinformatics tool to create upstream open reading frames (uORFs) that can potentially interfere with the translation of the main ORF. Interestingly, CDKN1C has an alternative 5’UTR resulting from a splicing event. Depending on the isoform considered, the created uORFs can be in-frame or out-of-frame and may have a different effect. Here, we have characterized these functional impacts.

Methods: To test their impact on protein levels, the variants were introduced in constructs containing the two different 5’UTR of CDKN1C upstream of a luciferase reporter. After transfection, the Luciferase levels were compared between the wild-type and mutant constructs. All other possible nucleotide changes at the same positions were also tested to check if the effect was specific of the created ATGs.

Results: Both variants reduced luciferase activity in contrast with the other substitutions at the same positions. This effect was different depending on the 5’UTR region tested. When the created ORF is in frame or out-of-frame, the protein levels were down by \( \sim \)30% and by \( \sim \)60% respectively. Thus, these variations of CDKN1C 5’UTR can differentially reduce the protein levels from the two isoforms, depending on the type of the created uORFs.

Conclusion: uORF-creating variants of the 5’UTR represent a new class of loss-of-function variants of CDKN1C implicated in BWS.

Conflict of Interest: None declared

EP20.020 The methylation episignature in patients with CHD4 variants reveals new disease sub-types

Karin Weiss 1;2, Karim Karimi3, Yael Lichtenstein1, Jack Reilly3, Haley McConkey3, Raissa Relator3, Jennifer Kerkhof3, Michael Levy3, Arjan Bouman4, Peter Turnpenny5, Joseph Symonds6, Philippe M. Campeau7, Matthew Tedder8, Tugce Balci9, Bekim Sadikovic3;10

1Rambam Health Care Campus, Medical Genetics, Haifa, Israel; 2Technion-Israel Institute of Technology, Ruth and Bruce Rappaport Faculty of Medicine, Haifa, Israel; 3London Health Sciences Centre, Molecular Genetics Laboratory, Molecular Diagnostics Division, London, ON, Canada; 4Erasmus MC University Medical Center, Department of Clinical Genetics, Rotterdam, Netherlands; 5Royal Devon University Healthcare NHS Foundation Trust, Department of Clinical Genetics, Exeter, United Kingdom; 6Royal Hospital for Children, Member of the ERN EpiCAR, The Paediatric Neurosciences Research Group, Glasgow, United Kingdom; 7University of Montreal, Department of Pediatrics, Montreal, QC, Canada; 8Greenwood Genetic Center, Greenwood, SC, United States; 9Schulich School of Medicine and Dentistry, Western University, Department of Paediatrics, London, ON, Canada; 10Western University, Department of Pathology and Laboratory Medicine, London ON, Canada

Background: Pathogenic variants in CHD4 cause Sifrim-Hitz-Weiss syndrome, a neurodevelopmental disorder associated with brain anomalies, heart defects, macrocephaly, hearing impairment, hypogonadism and additional features with variable expressivity. Most patients have non-recurrent missense variants complicating variant interpretation. Only few cases were reported with truncating variants and their role in disease is unclear. We aimed to study the effect of different CHD4 variants on the DNA methylation pattern.

Methods: We collected blood DNA samples from 27 individuals with CHD4 variants including missense and truncating variants, and performed methylation array testing. Clinical data was recorded as well.

Results: Using the EpiSign™ discovery software we identified a sensitive and specific DNA methylation episignature in cases with pathogenic missense variants within the ATPase/ helicase domain, in relation to controls and other episignature conditions within EpiSign Knowledge Database. This episignature was also seen a case with dominant inheritance and variable expressivity, a case with a de novo near PHD domain variant, and variants of uncertain significance within the ATPase/ helicase domain. The episignature had the highest overlap with Helsmoortel-Van der Aa syndrome caused by ADNP variants, and interestingly, ADNP is known to form the ChAHP complex with CHD4. A suspected different episignature was identified for the previously described p.(Met954Val) variant and for truncating variants. Together with the clinical features associated with these cases, we propose they cause distinct disease sub-types. Further cases are needed to confirm this finding.

Conclusion: Taken together our study supports DNA methylation is a useful tool for understanding variant pathogenicity and potentially disease mechanism in the CHD4-related syndrome.

Conflict of Interest: Karin Weiss: None declared, Karim Karimi: None declared, Yael Lichtenstein: None declared, Jack Reilly: None declared, Haley McConkey: None declared, Raissa Relator: None declared, Jennifer Kerkhof: None declared, Michael Levy: None declared, Arjan Bouman: None declared, Peter Turnpenny: None declared, Joseph Symonds: None declared, Philippe M. Campeau: None declared, Matthew Tedder: None declared, Tugce Balci: None declared, Bekim Sadikovic Own shares Iin EpiSign Inc.

EP20.021 Single-cell functional genomics of stress granules and their disease association

Carlo Maj1, Gabriel Schweizer1, Johannes Schumacher1, Pouria Dasmeh 1;2

1Universitätsklinikum Marburg (UKGM), Marburg, Germany; 2Harvard University, Chemistry and Chemical Biology, Cambridge, United States

Background: Dysregulation of stress granules – dynamic membrane-less organelles that form through liquid-liquid phase separation – has been implicated in various human diseases, including tumors, cardiovascular diseases, viral infections, and neurodegeneration.

Methods: To gain insights into the cell-type specific expression of stress granule genes and their diseases relevance, we investigated the single-cell preferential expression of an experimentally validated set of 38 genes in approximately 4 million cells from the mouse whole brain and 500,000 cells from 24 human organs.

Results: We found a preferential expression of stress granule genes within oligodendrocyte precursor cells in the brain as well as subpopulations of immune cells (natural killer cells and T-cells), fibroblasts and endothelial cells in various human tissues (adjusted p ~ 10-4, permutation test). We identified several co-expressed genes with stress granule genes, including the tyrosine kinase FYN that are implicated in synaptic plasticity (R ~ 0.4, p ~ 10-16; Spearman’s rank sum test). Transcription factor binding analysis revealed an overrepresentation of the ZF5 family, known for stabilizing mRNA upon stress stimulation (adjusted p ~ 10-9, hypergeometric test). We observed an enrichment of stress granule genes and their cell-type specific co-expressed counterparts within gene sets implicated in diverse conditions, including liver carcinoma, hereditary vascular anomalies, and X-linked intellectual diseases (adjusted p ~ 10-4, hypergeometric test).

Conclusions: Our single-cell comprehensive analysis extends the disease relevance of stress granules beyond established associations with motor neuronopathies and contributes to the exploration of a cellular mechanism with potential broader implications in health and pathological conditions.

Count: 246 words

Conflict of Interest: None declared

EP21 New Technologies

EP21.001 A New Generation Chronic Myeloid Leukemia Treatment Strategy Through The Utilization Of CRISPR/Cas9 Mediated Gene Therapy

Makbule SOMUNCU 1, Mahmut Selman Yıldırım1, Cihan Aydın2, Ayşe Gül Zamani1, Esra Albayrak3, Tuğçe Duran4, İbrahim Halil Kavaklı5

1Necmettin Erbakan University, Medicine Faculty Medical genetics, Konya; 2Istanbul Medeniyet University, Molecular Biology And Genetics, Molecular Microbiology, Konya; 3Ondokuzmayıs University, Faculty of Health Sciences, Department of medical services and techniques, Samsun, Turkey; 4Karatay University, Medicine Faculty, Konya; 5Koç University, İstanbul

Consortium: Scientific research organization of Necmettin Erbakan University, Project Number:181418003

Background:Chronic myeloid leukemia (CML) is based on BCR/ABL1p210 fusion encoding the BCR:ABL1p210 oncoprotein with excessive and irregular tyrosine kinase activity and eventually causes the CML phenotype.

Methods:This study has been aimed to silence BCR:ABL1oncogene and suppress chimeric protein production via CRISPR/Cas9 (Clustered Regularly Interspaced Short Palindromic Repeats and CRISPR-associated Cas9).This research has consisted of multi-step methods and analysis.Long-term culture optimization of the K562 cell line has been provided and its cytogenetic and molecular characterization has been performed. Fusion breakpoints of the BCR:ABLp2101 chimeric oncogene were detected. Genome editing in K562 cells was done by transfection of lipofectamine and electroporation.The efficiency of CRISPR/Cas9 on the BCR:ABL1p210 was analyzed by RT-qPCR and the sanger sequence. Intronic sequences were detected in the translocation of the BCR:ABL1fusion gene.

Results:We recorded that BCR:ABL1p210manipulation showed by 100-fold decrease in expression as (+1 log) before (-1 log) after CRISPR/Cas9 manipulation.

Conclusion:Therefore, could the decrease in the fusion gene expression we detected after the modification have resulted from the effect of the CRISPR on the recombination or the splicing mechanism?As a result, the effect of the CRISPR/Cas9 genome editing was revealed via the knockdown of the BCR:ABL1in this study.So, CRISPR/Cas9 can target the BCR:ABL1 fusion gene due to the interference effect. We believe that the findings of our study will lead to clinical studies in personal medicine with the CRISPR genome editing system.

Grants:This project was financed by the budget given for doctoral theses by the scientific research organization of Necmettin Erbakan University, Medicine Faculty, Project number; 181418003).

Key Words: BCR:ABL1p210,CML,CRISPR/Cas9

Conflict of Interest: None declared

EP21.005 The NanoporE Enhances Diagnosis in rarE Disease (NEEDED) study

Hannah Titheradge 1;2, Lorraine Hartles-Spencer3, Jessica Woodley3, Emma Douglas1, Madhura Chandrashekara1, Derek Lim1, Esther Kinning1, Ataf Sabir1, Helen Brittain1, Deborah Osio1, Kai Ren Ong1, Mary O’Driscoll1, Helen Cox1, Joanna Jarvis1, Denise Williams1, Rebecca Igbokwe1, Joanne Stockton2, Luke Ames2, Andrew Beggs2

1Birmingham Women’s and Children’s NHS Foundation Trust, Clinical Genetics, Birmingham, United Kingdom; 2University of Birmingham, Institute of Cancer and Genomic Sciences, Birmingham, United Kingdom; 3Birmingham Women’s and Children’s NHS Foundation Trust, Regional Genetics Laboratory, Birmingham, United Kingdom

Background/Objectives: Currently patients in England with monogenic developmental disorders will undergo NHS genetic testing including microarray and Whole Genome Sequencing (WGS) using short read sequencing (SRS) technology, with additional relevant genetic tests, such as methylation studies. Within this group a genetic diagnosis is made in about 30-40% of individuals. This means that a significant proportion of patients remain undiagnosed. We aim to evaluate the utility of Nanopore long read sequencing (LRS) as a tool to increase the diagnostic rate of suspected monogenic rare developmental disorders.

Methods: We identified 25 individuals highly likely to have a monogenic disorder, who have no molecular genetic diagnosis, and have reached the ceiling of investigation within the NHS genomic medicine service. They were recruited as trios and quads (affected patient(s), and their parents) to Understanding the Genomic Basis for Human Disease (Ethics reference: 15/WM/0076, IRAS reference: 159499). Trio (or quad) WGS is performed using the Oxford Nanopore Promethion P24 instrument. We are using a range of existing software packages to study participants’ genomes for potentially disease-causing variants including, sequence, copy number, methylation variants, and tandem repeats.

Results: We have analysed the first 4 trio samples, identifying a diagnosis of Coffin Siris syndrome in one of these patients. We will discuss the results of all 25 families. We also will discuss the feasibility of implementation of Nanopore sequencing into clinical diagnostic testing pathway.

Conclusion: Nanopore LRS has increased the diagnostic rate for patients with likely monogenic developmental disorders.

Grants: Lifearc Pathfinder award and NIHR Research Scholarship.

Conflict of Interest: Hannah Titheradge: None declared, Lorraine Hartles-Spencer: None declared, Jessica Woodley: None declared, Emma Douglas: None declared, Madhura Chandrashekara: None declared, Derek Lim: None declared, Esther Kinning: None declared, Ataf Sabir: None declared, Helen Brittain: None declared, Deborah Osio: None declared, Kai Ren Ong: None declared, Mary O’Driscoll: None declared, Helen Cox: None declared, Joanna Jarvis: None declared, Denise Williams: None declared, Rebecca Igbokwe: None declared, Joanne Stockton: None declared, Luke Ames: None declared, Andrew Beggs Scientific consultant to Oxford Nanopore Plc

EP21.006 Integrated spatial transcriptomic and proteomic analysis of fresh frozen tissue with Stereo-CITE-seq

Yang Heng 1;2;3, Sha Liao1;2;3, Weiqing Liu1, Yong Ma1, Uma Sangumathi Kamaraj4, Dongmei Jia1, Xiuwen Feng1, Diyan Liang1, Jinqiong Xiang1, Caili Huang1, Jiajun Zhang1;2;3, Min Jian1, Kui Su1, Mei Li1, Yuin-Han Loh4, Ao Chen2;3, Xun Xu1

1BGI-Shenzhen, Shenzhen, China; 2BGI Research-Southwest, Chongqing, China; 3JFL-BGI STOmics Center, Jinfeng Laboratory, Chongqing, China; 4Cell Fate Engineering and Therapeutics Lab, Cell Biology and Therapies Division, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore

Consortium: SpatioTemporal Omics Consortium (STOC)

Background/Objectives: Resolving the spatial distribution of mRNAs and proteins at the single-cell resolution is important for a comprehensive understanding of biological processes in health and disease. However, current spatial multi-omics technologies that simultaneously detect mRNAs and proteins on the same tissue section are either limited by low spatial resolution (e.g. SPOTS, SM-Omics and DBiT-seq), or lack the detection multiplicity (e.g. CosMx, Xenium). Therefore, it is urgent to develop a multi-omics method with high spatial resolution and detection multiplicity.

Methods: We combined Cellular Indexing of Transcriptomes and Epitopes by Sequencing (CITE-seq) and Stereo-seq to develop the Stereo-CITE-seq workflow. Using poly(dT) on Stereo-seq chips to capture antibody-derived tags (ADTs) and mRNAs, Stereo-CITE-seq simultaneously detects whole transcriptome and high-plex protein on the same tissue section.

Results: Here we demonstrated that Stereo-CITE-seq co-mapped whole transcriptome and proteins in mouse thymus with good reproducibility and accuracy. In addition, scaling up the protein multiplexity does not influence both mRNAs and proteins detection. Further, we co-detected 128 proteins and whole transcriptome in mouse thymus, liver and spleen with Stereo-CITE-seq. We observed much higher UMI counts of proteins compared with respective mRNAs. Integration of transcriptomic and proteomic data in liver enabled more accurate identification of cell populations such as B cells, macrophages and endothelial cells at single cell resolution.

Conclusion: Collectively, Stereo-CITE-seq co-detects whole transcriptome and high-plex protein at single-cell resolution with good reproducibility and accuracy. Spatial transcriptomics and proteomics provide complementary information for accurately identifying cell types.

Grants: Not applicable.

Conflict of Interest: Yang Heng A provisional patent related to this work has been filed by the BGI, and employees of BGI have stock holdings in BGI., full time employees of BGI, Sha Liao A provisional patent related to this work has been filed by the BGI, and employees of BGI have stock holdings in BGI., fulltime employee of BGI, Weiqing Liu A provisional patent related to this work has been filed by the BGI, and employees of BGI have stock holdings in BGI., full employee of BGI, Yong Ma A provisional patent related to this work has been filed by the BGI, and employees of BGI have stock holdings in BGI., fulltime employee of BGI, Uma Sangumathi Kamaraj fulltime employee of ASTAR, Dongmei Jia A provisional patent related to this work has been filed by the BGI, and employees of BGI have stock holdings in BGI., fulltime employee of BGI, Xiuwen Feng A provisional patent related to this work has been filed by the BGI, and employees of BGI have stock holdings in BGI., fulltime employee of BGI, Diyan Liang A provisional patent related to this work has been filed by the BGI, and employees of BGI have stock holdings in BGI., fulltime employee of BGI, Jinqiong Xiang A provisional patent related to this work has been filed by the BGI, and employees of BGI have stock holdings in BGI., full time employee of BGI, Caili Huang fulltime employee of BGI, Jiajun Zhang A provisional patent related to this work has been filed by the BGI, and employees of BGI have stock holdings in BGI., fulltime employee of BGI, Min Jian A provisional patent related to this work has been filed by the BGI, and employees of BGI have stock holdings in BGI., fulltime employee of BGI, Kui Su A provisional patent related to this work has been filed by the BGI, and employees of BGI have stock holdings in BGI., fulltime employee of BGI, Mei Li A provisional patent related to this work has been filed by the BGI, and employees of BGI have stock holdings in BGI., fulltime employee of BGI, Yuin-Han Loh fulltime employee of ASTAR, Ao Chen A provisional patent related to this work has been filed by the BGI, and employees of BGI have stock holdings in BGI., fulltime employee of BGI, Xun Xu A provisional patent related to this work has been filed by the BGI, and employees of BGI have stock holdings in BGI., fulltime employee of BGI

EP21.007 Buccal swab: a noninvasive approach to obtain human DNA for genetic testing

Anna Franzoni 1, Cristiano Sabelli1

1Copan Italia SpA, Scientific Affairs, Brescia

Background: While blood is commonly the sample of choice for genetic testing, this study aimed to investigate the success of use of a cutting-edge technology of buccal swabs – Copan FLOQSwabs® - in clinical and research genetic investigations.

Methods: A literature search was conducted using the keywords: “copan buccal swabs”, “swab hdna free copan” or “4N6FLOQSwabs”. Selected studies were limited to those published in English and featured in international, peer-reviewed scientific journals.

Results: Eipel et al (2016) demonstrated that buccal FLOQSwabs® are a suitable specimen for epigenetic age predictions while Barrett et al (2022) and Herzog et al (2023) for epigenetic investigations related to cancer risk assessment. Schöfl et al (2017), Wagner et al (2018), Schöne et al (2018) and Klussmeier et al (2020) used buccal FLOQSwabs® for genotyping genes for haematopoietic stem-cell transplantation. Zaorska et al (2019) predicted skin phenotypes from buccal FLOQSwabs® DNA. Prenatal and paternity testing benefited from this non-invasive sample, as reported by Koumbaris et al (2019), Tam et al (2019) and Zednikova et al (2020). Blum et al (2020) used buccal FLOQSwabs® for genotyping genetic variants involved in substance use disorder, while Minelli et al (2021) for investigating a pharmacogenomic approach for major depressive disorder. Shan et al (2020) and De Paolis et al (2022) supported buccal FLOQSwabs® use for evaluating SNPs and genes related to pancreatic and breast cancer, respectively.

Conclusions: Buccal FLOQSwabs® showed to be a valid alternative to blood for several genetic investigations. Tests are in progress to conduct a quantitative and qualitative comparison between the two sample types.

Conflict of Interest: None declared

EP21.008 in vitro qualitative analysis of the BCR/ABL1 gene fusion with CRISPR/Cas9 system

Deniz Aksu1, Selin Akad Dincer1, Zerrin Çelik1, Feride Sahin1, Sema Karakus2, Yunus Kasım Terzi 1

1Baskent University Faculty of Medicine, Department of Medical Genetics, Ankara, Türkyie; 2Baskent University Faculty of Medicine, Department of Hematology, Ankara, Türkyie

in vitro Qualitative Analysis of the BCR/ABL1 Gene Fusion with CRISPR/Cas9 System

Background: Chronic myeloid leukemia (CML) accounts for 15% of adult leukemia. The product of the BCR-ABL1 fusion gene is a protein with tyrosine kinase activity that plays an important role in the pathogenesis of CML. Real-time PCR-based methods are used to detect BCR-ABL1 fusion genes. This study aims to detect the BCR-ABL1 fusion gene with the CRISPR/Cas9 system.

Methods: Two patients’ cDNAs with the p210 BCR-ABL1 fusion gene were used as target molecules. Sanger sequencing was performed to detect the fusion region and its’ sequence. A guide RNA was designed according to the BCR-ABL1 fusion gene of a patient, and the CRISPR/Cas9 method was used to detect the fusion gene.

Results: In this study, we tested the presence of the p210 transcript. We showed that the CRISPR/Cas9 method can detect BCR-ABL1 fusion transcript if the sequence of the fusion region is known. The calculated sensitivity of the CRISPR/Cas9 mediated analysis was 2%.

Conclusion: The CRISPR/Cas9 technique is a useful tool for editing genomes and identifying gene rearrangements of the translocation type. It is believed that the test’s sensitivity and the number of fusion regions it can detect simultaneously may be enhanced in upcoming research.

Grant: The Baskent University local ethics committee approved the study and received support from the institutional review board of the Baskent University Research Fund (Project No: KA18/418)

Conflict of Interest: None declared

EP21.009 AI cloud-based end-to-end technology for accurate, fast & affordable diagnosis of genetic disorders in Egypt

Chris Kyriakidis 1, Ahmed Saad Elmorshedy2, Walaa Elamir Mohamed3, Carmen Garrido Navas1;4;5, Sara Chulián Prado4, Anne Kristine Schack1;6, Karmele Alapont1, Zoran Velkoski1, Marija Chaushevska1;7, Gjorgji Madjarov1;7

1gMendel, Copenhagen, Denmark; 2Chatby Pediatric University Hospital, Alexandria, Egypt; 3Cairo University, Department of Pediatrics, Cairo, Egypt; 4Asesoramiento Genetico SL, Granada, Spain; 5University of Granada, Granada, Spain; 6University of Copenhagen, Copenhagen, Denmark; 7Ss. Cyril and Methodius University, Skopje, Macedonia

Background/Objectives: Different technologies are used to diagnose genetic disorders depending on the genetic alteration. For chromosomal abnormalities such as Klinefelter syndrome (KS) or Turner syndrome (TS), karyotyping/FISH/microarrays are used although the resolution is limited for small chromosomal rearrangements and mosaicism. For point mutations such as cystic fibrosis (CF), standard NGS is performed after a clinical suggestion by sweat chloride test (SCT). Finally, diagnosis of spinal muscular atrophy (SMA) caused by bi-allelic mutations in the SMN1 is diagnosed by RT-PCR for mutations in SMN1 and for copy number of SMN2. The need to use diverse methodologies in a disease-dependent case is suboptimal in newborn screening approaches.

Methods: After demonstrating that the Phivea® platform can detect chromosomal abnormalities such as KS and TS with high sensitivity/specificity (>99%) and good diagnostic accuracy and point mutations for CF and copy number alterations for SMA, gMendel® has launched a pilot study in Egypt including 60 samples from patients with KS, TS, CF or SMA. Samples are multiplexed into one single ONTR10.4 flowcell and sequencing libraries are analyzed for the four diseases following internal SOP. Sequencing libraries are run into a GridION device.

Results: As expected from preliminary data using DNA from biobanks, Phivea® platform was able to correctly identify the different genetic disorders, by reducing the costs, accelerating the diagnostic process, and demonstrating its utility for newborn screening.

Conclusion: Phivea® platform can detect chromosomal aneuploidies, point mutations and copy number alterations, being suitable as a screening tool for KS, TS, CF and SMA among other potential genetic disorders.

Grants: N/A

Conflict of Interest: Chris Kyriakidis co-founder of gMendel, gMendel, Ahmed Saad Elmorshedy: None declared, Walaa Elamir Mohamed: None declared, Carmen Garrido Navas Founder of Asesoramiento Genetico SL, Asesoramiento Genetico SL, CSO at gMendel, Sara Chulián Prado Asesoramiento Genetico SL, Anne Kristine Schack gMendel, Karmele Alapont gMendel, Zoran Velkoski co-founder of gMendel, gMendel, Marija Chaushevska gMendel, Gjorgji Madjarov IPR with gMendel and warrants of Mendel, Chief AI officer at gMendel

EP21.011 An improved and updated assay for the detection of cystic fibrosis-causing variants in CFTR gene by Next Generation Sequencing

Carmen Hernandez 1, Virginia Perez1, alicia hernandez1, mario garcía mayo1, andreea lupsor1, candela montaner1, Inés Valledor1

1Certest Biotec, San Mateo de Gállego, Spain

Background/Objectives: Cystic fibrosis (CF, OMIM 219700) is caused by homozygous or compound heterozygous mutations in the cystic fibrosis conductance regulator gene (CFTR). More than 2000 genetic variants in CFTR gene have been described and more than 700 are annotated as CF-causing. Therefore, optimized NGS-based assays are essential for accurate patient diagnosis.

We present the analytical evaluation of an optimized and easy-to-use NGS assay for specific detection of CF-causing variants in CFTR gene, called VIASURE Cystic Fibrosis NGS Amplicon Library Preparation Kit.

Methods: The test is based on a target enrichment PCR, an indexing PCR and two intermediate clean-up steps. It has been optimized for simultaneous detection of 697 variants classified as CF-causing in the “CFTR2_7April2023” update of the CFTR2 database, as well as three benign variants I506V, I507V y F508C.

Processing of fastq files was performed by a self-developed software which integrates an improved bioinformatic pipeline, a display for results visualization and a tool for reports generation.

For analytical evaluation assays, a total of 57 previously characterized clinical and synthetic samples were used in two DNA concentration inputs, allowing validation of 170 target variants.

Results: VIASURE Cystic Fibrosis NGS Amplicon Library Preparation kit showed an Overall Percent Agreement (OPA) of 100%, with 100% of the samples being attributed with correct genotype in sensitivity and specificity assays as well as repeatability and reproducibility studies.

Conclusion: These results demonstrate the analytical excellence of this assay, making it a reliable method for the detection of cystic fibrosis-related genetic variants by NGS.

Conflict of Interest: None declared

EP21.012 Investigation of different library preparation and tissue of origin deconvolution methods for urine and plasma cfDNA methylome analysis

Nicholas Kueng1, Daniel Sidler2, Banz Vanessa3, Carlo Largiadèr1, Charlotte Ng4, Ursula Amstutz 1

1Inselspital Bern University Hospital, University of Bern, Department of Clinical Chemistry, Bern, Switzerland; 2Inselspital Bern University Hospital, University of Bern, Department of Nephrology and Hypertension, Bern, Switzerland; 3Inselspital Bern University Hospital, University of Bern, Department of Visceral Surgery and Medicine, Bern, Switzerland; 4University of Bern, Department for BioMedical Research (DBMR), Bern, Switzerland

Background/Objectives: Donor-derived cell-free DNA (dd-cfDNA) has potential value in allograft surveillance to reduce the need for graft biopsies, but has limitations including multi-organ transplantation from the same donor, blood transfusions and the early transplantation phase. Analysis of graft tissue-specific methylation signatures in cfDNA could serve as an alternative approach, potentially with higher specificity to the relevant transplanted tissue.

Methods: We evaluated laboratory methods for the tissue-of-origin deconvolution of urinary (n = 7) and plasma (n = 9) cfDNA from organ transplant recipients (n = 13) and healthy volunteers (n = 3) using a novel enzymatic-based cytosine conversion method together with single- (ssLP) and double-stranded (dsLP) library preparation methods followed by low-coverage whole-genome bisulfite sequencing.

Results: The dsLP method showed significantly reduced global methylation in urinary cfDNA (56%) compared to plasma-derived cfDNA (75.8%; p < 0.0001). Conversely, no differences in global methylation were observed between plasma and urinary cfDNA with the ssLP method (plasma: 80.2%; urine: 79.4%, p > 0.05). There was a significantly higher proportion of sequences with undetermined tissue-of-origin with the dsLP method, which was more pronounced in urine (1.9% vs 32.6%, p < 0.001) than in plasma (0.2% vs 7.4%, p < 0.001), whereas no significant differences were detectable between library preparation methods for the tissue-of-origin proportions.

Conclusion: This study outlines potential biases and pitfalls with methylome-based cfDNA tissue-of-origin deconvolution and detection of cfDNA from tissues of interest. Based on these results, enzymatic conversion coupled with a single-stranded library preparation appears to be a robust and bias-free choice, especially for urinary-derived cfDNA, where DNase activity causes a reduction in intact double-stranded fragments.

Grants: Swiss National Science Foundation grant no. 310030_188762

Conflict of Interest: None declared

EP21.013 Whole Genome Sequencing in daily clinical practice: Are we there yet?

Nikolaos Marinakis 1;2, Anastasios Mitrakos1;2, Danai Veltra1, Maria Svingou1, Faidon-Nikolaos Tilemis1, Kyriaki Kekou1, Konstantina Kosma1, Aphrodite Kampouraki1, Hane Lee3, Yongjun Song3, Katherine Anagnostopoulou4, Yannis Loukas4, Periklis Makrythanasis1;5;6, Christalena Sofocleous1, MARIA TZETIS1, Jan Traeger-Synodinos1

1National and Kapodistrian University of Athens, Laboratory of Medical Genetics, Athens, Greece; 2National and Kapodistrian University of Athens, Research University Institute for the Study and Prevention of Genetic and Malignant Disease of Childhood, Athens, Greece; 33billion Inc, Seoul, Korea, Rep. of South; 4RARE-ID Research Programme, Neoscreen Diagnostic Laboratory, Athens, Greece; 5University of Geneva, Department of Genetic Medicine and Development, Geneva, Switzerland; 6Biomedical Research Foundation of the Academy of Athens, Athens, Greece

NM and AM were equally contributing

Background/Objectives: Genetic diagnosis has been revolutionized by high-throughput sequencing technologies and data analysis. In the last decade Whole Exome Sequencing (WES) has drastically impacted molecular diagnosis of rare disorders. Nowadays, Whole Genome Sequencing (WGS) holds promise as a first-tier test, however interpretation and reporting represent considerable challenges for laboratories seeking to maximize routine diagnostic applications.

Materials/Methods: We report findings from 30 cases analysed by short-read WGS (16 trios, 14 proband-only) due to neurodevelopmental, neuromuscular, retinal, or metabolic disorders, for which previous genetic tests, including array-CGH, exome sequencing and classic karyotype, were inconclusive. Sequencing was implemented with TruSeq DNA PCR-Free kit and Illumina NovaSeq 6000. Bioinformatics analysis used VarSome Clinical, Franklin (Genoox) and eVai (enGenome).

Results: Three (likely) pathogenic variants (KMT2D:c.12694C>T, H1-4:c.436_458del, and CREBBP:c.5614A>G) were detected allowing a confirmatory diagnosis in three cases with a diagnostic yield of 10%. In two more cases genotypes identified remain under further investigation towards final classification and correlation to the phenotype.

Discussion: WGS potentially supports marked improvements in the diagnosis of inherited disorders. Although, all variants detected in the present cohort are in exonic regions, (indicating that WES reanalysis may have been equally effective), this limited experience of WGS implementation highlights its potential. Once more/all non-coding regions have been annotated relative to their impact on genomic function towards resolution of underlying diseases mechanisms, WGS has potential to support more comprehensive genomic evaluation in patients/individuals becoming a true gamechanger for studying and diagnosing rare diseases.

Conflict of Interest: None declared

EP21.014 Unraveling the mysteries of balanced rearrangements: optical genome mapping spotlights BCL11B misregulation in a familial translocation

Adrián Alcalá San Martín 1, Neus Fornés Garcia1, Didac Casas-Alba1, Patricia Fernández1, Patricia Peral García1, Sara Manresa García1, Gisela Márquez1, Enric Balius Fort1, Cristina Hernando-Davalillo1

1Sant Joan de Déu Barcelona Hospital, Department of Genetic Medicine – IPER, Esplugues de Llobregat, Spain

Background/Objectives: Reciprocal translocations are common chromosomal structural rearrangements. While some carriers remain asymptomatic, others may exhibit manifestations due to genomic imbalance, gene disruption and/or position effect. Despite advances in genetics knowledge, establishing a direct causal relation between a translocation and a phenotype may still be challenging. Furthermore, traditional genetic studies often focus solely on variations directly affecting gene sequence, potentially overlooking variants without immediate gene-related effects. Optical genome mapping (OGM) has emerged as a valuable tool in elucidating such cases.

Methods: OGM was performed on a patient with an apparently balanced reciprocal translocation, previously detected by conventional karyotyping and also present in her mother. Both of them were affected with a similar neurodevelopmental phenotype, characterized by intellectual disability, behavioral disorders and dysmorphic traits.

Results: A balanced translocation involving the chromosomes 8 and 14 was confirmed at higher resolution. Detailed analysis revealed a potentially deleterious position effect at the breakpoint on chromosome 14, disrupting the regulation of the BCL11B gene. BCL11B haploinsufficiency has been associated with a rare neurodevelopmental syndrome compatible with the phenotype of our patient.

Conclusion: Although a rearrangement does not directly disrupt a gene sequence, its potential relationship with a phenotype cannot be ruled out lightly. Breakage located within genomic regulatory regions can impact proper gene regulation, requiring thorough examination. OGM has proved to be a new promising next-generation cytogenomics tool in clinical genetics, shedding light on cases as the present, where detailed delineation of the breakpoints has pointed to BCL11B misregulation as the plausible cause of the phenotype.

Grants:

Conflict of Interest: None declared

EP21.015 Improving molecular diagnosis of sexual development disorders with a gene panel strategy

Iris Pereira Caetano 1, Joana Mendonça1, Alice Mirante2, Rita Cardoso2, Marcia Rodrigues3, João Paulo Oliveira4, Ana Grangeia4, David Barbosa5, Luís Vieira1;6, João Gonçalves1;6

1Instituto Nacional de Saúde Dr Ricardo Jorge, Human Genetics Department, Lisboa, Portugal; 2Hospital Pediátrico de Coimbra, Department of Paediatric Endocrinology, Diabetes and Growth, Coimbra, Portugal; 3Hospital de Santa Maria, Centro Hospitalar Universitário de Lisboa Norte, Medical Genetics Service, Pediatric Department, Lisboa; 4Centro Hospitalar Universitário de São João, Medical Genetics Service, Porto, Portugal; 5Hospital Garcia de Orta, Endocrinology and Diabetes Service, Almada, Portugal; 6Nova Medical School, Center for Toxicogenomics and Human Health, Lisboa, Portugal

Background/Objectives: Next-generation sequencing (NGS) has emerged as a valuable tool for molecular diagnosis, including rare diseases such as disorders of sexual development (DSD). A multigene NGS approach allows the simultaneous analysis of a broad spectrum of genes, ensuring fast and cost-effective results and the likelihood of identifying the genetic cause and novel variants.

Methods: DNA samples from 38 DSD patients were sequenced using Ampliseq technology (Illumina) and analysed according clinical indication with a customized gene panel (40 genes) grouped into specific subpanels: primary sexual determination/differentiation, hypogonadotropic hypogonadism/infertility, steroidogenesis and premature ovarian insufficiency. ACMG-AMP guidelines, bioinformatics tools and clinical/population databases were used for variant classification. Pathogenic, likely pathogenic and VUS variants were validated by Sanger sequencing.

Results: We studied 38 DSD patients, 16 of which firstly screened for one key DSD-associated genes (SRY, AR, ANOS1, GNRHR, CYP21A2) presented negative results. Using the multigene approach, we identified 10 causative variants, with diverse inheritance patterns, in 8 patients (21.1%), affecting AR, HSD17B3, LHCGR, FGFR1, NR5A1, including one patient with a complex genotype, being homozygous for LHCGR and hemizygous for AR.

Conclusion: Given the complexity and phenotypic overlap of DSD, specific clinical diagnosing of these patients is difficult and requires molecular confirmation. The multigene studies by NGS, in comparison with classical gene step approach (Sanger sequencing), proved to increase the rate of variant detection, playing an important role in confirming clinical diagnoses, guiding treatment decisions, reproductive management and genetic counselling of these patients and families.

Grants: FCT/MCTES, Projects: ToxOmics and Human Health(UIDB/00009/2020) GenomePT(POCI-01-0145-FEDER-022184).

Conflict of Interest: None declared

EP21.016 Automated Library Preparation Using Watchmaker DNA Library Prep Kit with Fragmentation on the firefly®

Krystal Sheridan1, Eleanor Matrullo2, Melissa Netwal3, Paul Lomax 3

1The Translational Genomics Research Institute (TGen), Phoenix, AZ, United States; 2Watchmaker Genomics, Boulder, CO, United States; 3SPT Labtech, Melbourn, United Kingdom

Background/Objectives: The HIVE Laboratory at TGen North plays a pivotal role in Arizona’s pathogen surveillance initiatives, aiding public health efforts and supporting underserved communities. By employing whole genome sequencing, the lab tracks emerging outbreaks, fostering well-informed public health responses. The demand for high-throughput sample processing and precise pathogen surveillance raised challenges in their manual NGS library preparation workflows.

This work describes TGen North’s process of evaluating several library prep solutions on SPT Labtech’s firefly® liquid handling platform, with the goal of transitioning from manual library prep to an automated workflow.

Methods: 7 DNA library prep kits were compared across a series of evaluation criteria using 8 independent DNA isolates from GM12978 cells extracted with the Chemagic Cell Pellet protocol from Revvity, eliminating process variability. Fragmenting and A-tailing, adapter ligation, post-ligation purification, library amplification and post-amplification cleanup were performed on firefly using a combination of its 96-pipetting head and its non-contact reagent dispensing head.

Results: Out of the 7 kits tested, the Watchmaker DNA Library Prep Kit with Fragmentation offered the highest conversion rates, with even size and yield across all samples and no additional size selection required. This kit offers a streamlined automation-friendly workflow, compatible with PCR-free options.

Conclusion: The adoption of automated library preparation using Watchmaker DNA Library Prep Kit with Fragmentation on firefly offers greater flexibility, minimizes reagent wastage, and enables more effective handling of valuable samples. Consequently, this has bolstered TGen North’s capacity for timely pathogen surveillance, while also freeing resources for more complex laboratory tasks.

Grants:

Conflict of Interest: None declared

EP21.017 A streamlined EM-seq method for whole genome methylation detection

Chaithanya Ponnaluri1, Vaishnavi Panchapakesa1, Daniel Evanich1, Matthew Campbell1, Bradley Langhorst1, Louise Williams 1

1Ipswich, New England Biolabs, Ipswich, United States

DNA methylation is an important epigenetic regulator of gene expression. In the human genome cytosines are methylated in the CpG context and are often clustered in CpG rich regions associated gene regulation. Traditionally, sodium-bisulfite conversion was used to distinguish 5-methylcytosines (5mC) and 5-hydroxymethylcytosines (5hmC) from cytosines. However, this method damages DNA and introduces significant sequencing bias. NEBNext® Enzymatic Methyl-seq (EM-seq™) is an enzymatic approach that minimizes DNA damage therefore enabling longer insert sizes, lower duplication rates and a more accurate quantification of methylation.

EM-seq has been optimized further. Here we show EM-seq v2 data for DNA inputs ranging from 0.1 ng pg to 200 ng. EM-seq v2 is a streamlined protocol that reduces library preparation time via a reduction in end preparation and adaptor ligation time as well as by eliminating a cleanup step. Additionally, removal of the cleanup also results in savings through reduced plastics usage. Libraries typically had increased yield that will reduce sequencing duplications. Furthermore, all libraries displayed an even GC bias representation as well as consistent global methylation between the 0.1 ng to 200 ng DNA inputs. Coverage of genomic features such as CpG islands and enhancers was maintained to 1 ng but dropped as expected with the 0.1 ng inputs. EM-seq v2 is an enhanced, streamlined protocol that provides consistent methylation metrics for inputs ranging from 0.1 ng to 200 ng.

Conflict of Interest: Chaithanya Ponnaluri Employee of New England Biolabs, Vaishnavi Panchapakesa Employee of New England Biolabs, Daniel Evanich Employee of New England Biolabs, Matthew Campbell Employee of New England Biolabs, Bradley Langhorst Employee of New England Biolabs, Louise Williams Employee of New England Biolabs

EP21.018 Establishment of MassARRAY technology for screening of spinal muscular atrophy in pregnant women

Wantanai Satheanwongsa1, Chayada Tangshewinsirikul2, Donniphat Dejsuphong3, Preyaporn Onsod1, Budsaba Rerkamnuaychoke1, Teerapong Siriboonpiputtana1, Takol Chareonsirisuthigul 1

1Faculty of Medicine Ramathibodi Hospital, Mahidol University, Department of Pathology, Bangkok, Thailand; 2Faculty of Medicine Ramathibodi Hospital, Mahidol University, Department of Obstetrics and Gynecology, Bangkok, Thailand; 3Faculty of Medicine Ramathibodi Hospital, Mahidol University, Division of Translational Medicine, Bangkok, Thailand

Background/Objectives: Alpha motor neuron failure and degradation are hallmarks of spinal muscular atrophy (SMA), a severe autosomal recessive neurodegenerative disorder that causes progressive muscular weakening. MassARRAY technology is a new, powerful, and valuable tool for identifying common genetic changes that have recently come to light. This study aimed to validate the MassARRAY assay for SMA molecular testing in pregnant women.

Methods: Using the well-established MassARRAY and MLPA technique, the genomic DNA (n = 14) isolated from the blood specimens of Thai pregnant women was examined for mutations and copy number variation of the survival motor neuron 1 (SMN1) and survival motor neuron 2 (SMN2) genes exons 7 and 8.

Results: The findings showed that the accuracy of the two approaches in identifying SMA mutations was 100% in sensitivity and specificity. Nevertheless, in four samples (28.6%), the copy number determination results were inconsistent; MassARRAY determined that one case was an SMA carrier, although MLPA was normal.

Conclusion: More internal control markers and clinical samples were needed to explore its effectiveness further. The outcome of this study will aid with carrier screening and diagnosis in the clinical management of SMA.

Grants: Faculty of Medicine Ramathibodi Hospital, Mahidol University

Conflict of Interest: None declared

EP21.019 Optical genome mapping: lighting the way in complex clinical cases

Iván Monge Lobo 1, Barbara Fernández Garoz1, Diana Monclús Arroyo1, Elena M Ruiz Ballesteros2, Claudia Toledo Pacheco2, Ana Isabel Quinteiro García1, Nelmar Valentina Ortiz Cabrera1

1Hospital Infantil Universitario Niño Jesús, Madrid, Spain; 2Hospital General Universitario de Toledo, Toledo, Spain

Background/Objectives: Optical genome mapping (OGM) is a novel technology that could offer the information of various conventional cytogenetic techniques into a single test. We validated and implemented this technique as first tier as cytogenetic techniques in our institution in 2022. Here we present some curious cases that shows the virtues and pitfalls of this technique.

Methods: We have analysed 700 samples so far. Here we present four different cases analysed with De Novo pipeline.

Results: Case 1. Incontinencia pigmenti diagnosis. Partial deletion of IKBKG gene. Confirmatory MLPA analysis detected exons 4-10 loss, but it couldn´t specify if the deletion was within IKBKG or its pseudogene. OGM clearly located the deletion in IKBKG gene.

Case 2. 8p inverted duplication/deletion syndrome diagnosis. Deletion on 8p23.3p23.1 of 7.48 Mb in heterozygosis and a duplication on 8p23.1p11.21 of 30.43 Mb in heterozygosis. Inversion was not detected.

Case 3. Pregnant woman with history of previous miscarriages due to a deletion in chromosome 5. Transposition of 4.81 Mb from 5q35.1q35.2 into 6q23.3, and deletion on 6q23.3 of 582 kb in heterozygosis. This complex chromosomal rearrangement results in two different derivatives.

Case 4. Suspicious complex rearrangement of chromosome 4. Fusion between 4p16.1 and 4q34.1, with 4q34.1q35.2 deletion of 16.77 Mb in heterozygosis and 4p16.3p16.1 duplication of 10.09 Mb in heterozygosis.

Conclusion: OGM offers valuable insights into different chromosomal rearrangements to improve diagnosis and guide towards better genetic counselling. Even though, this technique has its limitations that needs to be foreseen to avoid diagnostic errors.

Conflict of Interest: None declared

EP21.020 Automated solid phase extraction of ultra-high molecular weight (uHMW) DNA using cross-linking reagent for long-read sequencing.

Elian Rakhmanaliev 1, Hwei Ling Khor1, Swee Yin Lim1, Shi Hui Ang1, Mohammed Irfan1, Siti Rafei1;2, Karen Wang1;2, Mi Kyoung Park1

1One BioMed Pte Ltd, R&D, Singapore, Singapore; 2Institute of Microelectronics, R&D, Singapore, Singapore

Background/Objectives: The extraction of high-quality uHMW DNA is essential for human genome research. However, it remains predominantly manual and tedious process. To simplify uHMW DNA extraction procedure, we used a unique feature of dimethyl adipimidate dihydrochloride (DMA), a cross-linking reagent, that covalently link free amino groups, for developing an automated nucleic acid (NA) extraction system. The amidine bonds formed between DMA and NA are reversable which allows capture and release NA by changing buffering conditions.

Methods: gDNA was extracted from rat tissues, HeLa cell line and bacteria using the Xceler8™ system. DNA integrity and size were analysed by Pulse-Field Gel Electrophoresis (PFGE) and Femto Pulse System. Long-read sequencing was conducted on Oxford Nanopore’s PromethION, GridION and MinION instruments using the Ultra-Long DNA V14 and Ligation Sequencing kits and PacBio’s Sequel IIe system using the SMRTbell® prep kit.

Results: PFGE shows the size of HMW DNA around 300-400Kbp and uHMW DNA > 2.2Mbp. Results for representative ONT sequencing are summarised in the table.

System

Kit

Quality

Read length N50

Longest read, nt

PromethION

Ultra-Long DNA

>Q15 72.5%

84,935

668,658

GridION

Ligation

>Q12 69.1%

17,270

619,890

MinION

Ligation

>Q12 60.1%

22,634

293,001

For PacBio distribution of read lengths was between 2,000bp and 25,000bp. This is typical for PacBio’s HiFi chemistry. Even distribution of mapped reads across the reference sequences demonstrates non-selective and uniform extraction of gDNA.

Conclusion: The results demonstrate that our sold-surface DNA-DMA binding technology enables extraction of high-quality uHMW DNA and compatible with both widely used long-read sequencing technologies.

Grants: None.

Conflict of Interest: None declared

EP21.021 Towards robust clinical genome interpretation: developing a consistent terminology to characterise Mendelian disease-gene relationships - allelic requirement, inheritance modes and disease mechanisms

Angharad Roberts 1;2;3, Marina DiStefano4, Erin Rooney Riggs5, Katherine Josephs1, Fowzan Alkuraya6, Joanna Amberger7, Mutaz Amin8, Jonathan Berg9, Alison Coffey10, Fiona Cunningham11, Karen Eilbeck12, Helen Firth13, Julia Foreman14, Ada Hamosh7, Eleanor Hay2, Sarah Leigh15, Christa Martin5, Ellie McDonagh14, Daniel Perrett14, Erin Ramos16, Peter N. Robinson17, ANA RATH8, Zornitza Stark18, David Sant12, Nicola Whiffin19, Heidi Rehm4;20, James Ware3;4;21

1Imperial College London, United Kingdom; 2Great Ormond Street Hospital for Children, United Kingdom; 3Royal Brompton & Harefield Hospitals, United Kingdom; 4Broad Institute of MIT and Harvard, Medical and Population Genetics, Cambridge, United States; 5Geisinger Medical Center, Danville, United States; 6KFSHRC, Department of Translational Genomics, Riyadh, Saudi Arabia; 7Johns Hopkins University School Of Medicine-Geriatric Medicine & Gerontology, Online Mendelian Inheritance in Man (OMIM), Baltimore, United States; 8Inserm, US14-Orphanet, Paris, France; 9University of North Carolina at Chapel Hill, Department of Genetics, Chapel Hill, United States; 10Illumina, United States; 11EMBL’s European Bioinformatics Institute (EMBL-EBI), Hinxton, United Kingdom; 12The University of Utah, Department of Biomedical Informatics, Salt Lake City, United States; 13Cambridge University Hospitals, Dept of Medical Genetics, United Kingdom; 14EMBL’s European Bioinformatics Institute (EMBL-EBI), United Kingdom; 15Genomics England, United Kingdom; 16NHGRI National Human Genome Research Institute, United States; 17The Jackson Laboratory For Genomic Medicine, United States; 18Australian Genomics, PanelApp Australia, Australia; 19Big Data Institute and Centre for Human Genetics, University of Oxford, United Kingdom; 20Massachusetts General Hospital, Center for Genomic Medicine, United States; 21Imperial College London, 1. National Heart & Lung Institute & MRC London Institute of Medical Sciences, United Kingdom

Consortium: The Gene Curation Coalition (GenCC)

Background/Objectives: The current lack of standardisation in the terminology for gene-disease curation and variant annotation poses challenges. We aim to address this by establishing uniform terminology specifically to describe inheritance, allelic requirement, and both sequence and functional consequences to characterise disease-gene relationships. This will promote global harmonisation in curation and aid variant classification within the ACMG/AMP framework.

Methods: Terminology for inheritance, allelic requirement, and both structural and functional consequences of a variant used by Gene Curation Coalition (GenCC) members and partner organizations was collated and reviewed. Harmonised terminology with definitions and use examples was created, reviewed, and validated.

Results: We present a standardised terminology to describe gene-disease relationships and support variant annotation. Application of this terminology ws piloted by classifying variation within the ACMG SF 2.0 genes recommended for reporting secondary findings. Consensus terms are formalised in sequence ontology (SO) and human phenotype ontology (HPO) ontologies. GenCC member groups commit to utilising or aligning with these terms in their resources.

Conclusion: Our standardised terminology enhances global harmonisation, enabling seamless integration of curation datasets across international efforts. This fosters improved consistency in variant classification and enhances the interpretation of genetic tests, addressing the critical need for clarity in genomic research.

Adapted from PMID: 37982373 https://doi.org/10.1016/j.gim.2023.101029

Grants: Sir Jules Thorn Trust [21JTA], Wellcome Trust [107469/Z/15/Z; 200990/A/16/Z; 222883/Z/21/Z, WT223718/Z/21/Z, 220540/Z/20/A, 220134/Z/20/Z, WT200990/Z/16/Z, Sanger Institute Quinquennial Review 2021-2026], BHF [RE/18/4/34215; FS/CRLF/21/23011], MRC (UK), NHLI Foundation Royston Centre for Cardiomyopathy Research, NIHR Imperial College Biomedical Research Centre, NHGRI [U24HG006834, U24HG009650, U24HG011449], EMBL, NIH: [T15LM007124], NHMRC [GNT1113531, GNT2000001], Rosetrees Trust [PGL19-2/10025]

Conflict of Interest: Angharad Roberts: None declared, Marina DiStefano: None declared, Erin Rooney Riggs: None declared, Katherine Josephs: None declared, Fowzan Alkuraya: None declared, Joanna Amberger: None declared, Mutaz Amin: None declared, Jonathan Berg: None declared, Alison Coffey: None declared, Fiona Cunningham: None declared, Karen Eilbeck: None declared, Helen Firth: None declared, Julia Foreman: None declared, Ada Hamosh: None declared, Eleanor Hay: None declared, Sarah Leigh: None declared, Christa Martin: None declared, Ellie McDonagh: None declared, Daniel Perrett: None declared, Erin Ramos: None declared, Peter N. Robinson: None declared, ANA RATH: None declared, Zornitza Stark: None declared, David Sant: None declared, Nicola Whiffin: None declared, Heidi Rehm: None declared, James Ware J.S.W. has received research support or consultancy fees from Myokardia, Bristol-Myers Squibb, Pfizer, and Foresite Labs

EP21.022 Optimization of a method for directed differentiation of Induced Pluripotent Stem Cells into Liver Progenitor Cells

Irina Panchuk 1, Svetlana Smirnikhina1

1Research Centre for Medical Genetics, Laboratory of genome editing, Moscow

Background/Objectives: The development of appropriate liver models is crucial for investigating liver diseases and drug development. Liver progenitor cells (LPCs) can be generated from the differentiation of human induced pluripotent stem cells (hiPSCs), used to create 2D and 3D liver models, and establish transgene delivery protocols.

Methods: The hiPSC differentiation protocol involved sequential differentiation into definitive endoderm (DE), LPCs and liver stellate cells. To induce the growth and differentiation of 3D liver organoids, we utilized the HepatiCult Organoid kit. The differentiation stages were confirmed through analysis of characteristic markers using immunocytochemistry and flow cytometry. Electroporation was used to deliver plasmid pAAV-GFP using Neon equipment. Adeno-associated virus (AAV) serotypes 2, 5, 6, and 9 with GFP cassette were used for transduction at MOI of 10,000 and 100,000. Transfection efficiency was evaluated by flow cytometry at 48 and 72 hours.

Results: The presence of DE in the cells was confirmed by the markers SOX17 (96.9 ± 0.6%) and FOXA2 (95.3 ± 1.9%), while the presence of LPCs was confirmed by AFP + /HNF4α + (74.9 ± 21.1%). During 3D differentiation, markers for not only hepatocytes (CK18 + , ALB + , AFP + ) but also cholangiocytes (CK7 + ) were defined. The presence of hepatic stellate cells was confirmed by Oil-red-O staining of fat droplets and the presence of specific morphology. A protocol for electroporation of the LPCs with a plasmid delivery efficiency of 54.3 ± 10.6% was established. The maximum transduction efficiency was marked for AAV serotype 2 at MOI of 100,000 and 93.5 ± 6.9%.

Conclusion: In conclusion, this study has optimized the method for differentiating hiPSCs into LPCs and transgene delivery protocols.

Grants:

Conflict of Interest: None declared

EP21.023 Cas9-targeted-based long-read sequencing for genetic screening of RPE65 locus

Cristina Rodilla 1;2, Gonzalo Nuñez Moreno1;2;3, Raquel Romero1;2;3, Pablo Mínguez1;2;3, Marta Corton1;2, Carmen Ayuso1;2

1Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Department of Genetics & Genomics, Madrid, Spain; 2Instituto de Salud Carlos III, Center for Biomedical Network Research on Rare Diseases (CIBERER), Madrid, Spain; 3Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Bioinformatics Unit, Madrid, Spain

Background/Objectives: Long-read sequencing (LRS) is a cutting-edge technology that enables structural and non-coding variants detection. Employing target enrichment can reduce the cost and duration of sequencing when a molecular diagnosis is suspected. This study presents our experience using CRISPR-Cas9 targeted nanopore sequencing to detect RPE65 variants in a group of inherited retinal dystrophies (IRD) patients.

Methods: Long-read sequencing was conducted in 5 IRD patients harboring a monoallelic variants in RPE65 and remained uncharacterized after clinical exome sequencing. Libraries were prepared using high-weight molecular DNA. Guide RNA probes were designed to target a 31kb targeted region including the entire RPE65 locus following Oxford Nanopore Technologies guidelines. DNA was then sequenced on a MinION platform for 24 hours. For 3 patients, short-read 30x whole genome sequencing (WGS) was performed to validate nanopore results.

Results: The sequencing process yielded a median of 60 reads within the targeted region and a median read size of 10kb. All previously identified variants have been detected using this approach, no additional RPE65 gene variants were found. CRISPR-Cas9 enrichment ensured a homogenous coverage of the region of interest and an average of 623 reads aligned to the RPE65 locus. Nanopore variant detection demonstrated performance akin to WGS at similar coverage levels, although exhibiting increased false positive calls at lower coverage.

Conclusion: In this study, we explore the advantages of using a targeted approach together with long-read sequencing for identifying variants associated with IRD. The results underscore the utility of this approach for the characterization of patients affected by rare diseases.

Conflict of Interest: None declared

EP22 Treatments for Genetic Disorders

EP22.001 Correction of p.F508del using CRISPR/Cas9 in iPSCs and airway basal stem cells derived from cystic fibrosis patients

Ekaterina Kondrateva 1;1, Anna Demchenko1, Olga Volodina1, Alexander Lavrov1, Svetlana Smirnikhina1

1Research Centre for Medical Genetics

Background/Objectives: Cystic fibrosis is the most common fatal and still incurable genetic disease. The most frequent reason is the mutation p.F508del in the CFTR gene which theoretically can be corrected using gene editing methods. We created several CRISPR/Cas9 based systems for editing this mutation and tested them in CFTE29o- line, induced pluripotent stem cells (iPSCs) and induced airway basal stem cells (iBCs).

Methods: iPSC lines were generated by reprogramming of skin fibroblasts from three patients with homozygous p.F508del mutation. iBCs lines were generated by directed differentiation of mentioned iPSCs. We created nine editing systems consisting of three variants of Cas9 (eSpCas9(1.1), SpCas9(HF4) and SaCas9), three variants of sgRNAs and three variants of ssODNs and used them for transfection of CFTE29- cells (by lipofection), iPSCs and iBCs (by electroporation). Then NHEJ, indels and HDR levels were analyzed by NGS.

Results: CFTE29o- line is hardly transfected and edited; the best HDR efficiency was 1,5% alleles in transfected cells. Three iPSCs lines showed a large spread of values, however, the best HDR efficiency for all the lines were observed with SpCas9(1.1) and sgRNA#1 — it’s ranged from 0,6% to 3,3% alleles in transfected cells. Three iBCs lines were edited with high HDR efficiency from 4,3% to 27,5% alleles in transfected cells.

Conclusion: CFTE29o- cells and iPSCs are difficult targets for gene editing but allow the selection of the best editing systems. iBCs derived from iPSCs are a promising platform for the development of gene therapy and can be edited in the p.F508del locus with high efficiency.

Conflict of Interest: None declared

EP22.002 Frequency of MC4R pathway variants in a European cohort of individuals with early-onset severe obesity

Anthony Goldstone 1, JESUS DOMINGUEZ RISCART2, Giusi Umano3, Orit Hamiel4, Melek Yildiz5, Melania Manco6, Patrick Sleiman7, Charles Savoie7, Dinesh Giri8, Jesús Argente9

1PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom; 2Instituto de Investigación e Innovación Biomédica de Cádiz, Hospital Universitario Puerta del Mar, Universidad de Cádiz, Cádiz, Spain; 3Department of Woman, Child and of General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Naples, Italy; 4Pediatric Endocrinology and Diabetes Unit, Sheba Medical Center, Sackler School of Medicine Tel Aviv University, Tel Aviv, Israel; 5Department of Pediatric Endocrinology, Istanbul University Faculty of Medicine, Istanbul, Türkyie; 6IRCCS Bambino Gesù Children’s Hospital, Rome, Italy; 7Rhythm Pharmaceuticals, Inc., Boston, United States; 8Paediatric Endocrinology, Bristol Royal Hospital of Children, University of Bristol, Bristol, United Kingdom; 9Department of Pediatrics & Pediatric Endocrinology, Universidad Autónoma de Madrid, University Hospital Niño Jesús, CIBER “Fisiopatología de la obesidad y nutrición” (CIBEROBN), Instituto de Salud Carlos III, IMDEA Institute, Madrid, Spain

Background/Objectives: The melanocortin-4 receptor (MC4R) pathway is critical for hunger regulation, energy balance, and weight regulation. Individuals with variants in MC4R pathway genes may present with early-onset severe obesity and hyperphagia. The Rare Obesity Advanced Diagnosis™ genetic testing program aims to enhance access to genetic testing for individuals with suspected rare genetic causes of obesity.

Methods: MC4R pathway genes (POMC, PCSK1, LEPR, NCOA1, SH2B1, MC4R, MC3R, CPE, LEP, KSR2, SIM1), associated with genetic obesity, from individuals with early-onset, severe obesity were sequenced. Variants were classified as pathogenic/likely pathogenic/variants of unknown significance (P/LP/VUS) according to ACMG criteria. VUS were further divided into suspected pathogenic (SP), uncertain, or suspected benign based on available evidence.

Results: Among 2,253 individuals, 6.0%, 20.7%, 26.9%, and 46.4% were aged <6, 6- < 12, 12-18, and >18 years, respectively, with mean age of obesity onset 6.9 years. Overall, 20.4% of individuals carried ≥1 variant in the 11 studied genes; 5.0% had a P/LP/VUS-SP variant. The variant frequency for genes with demonstrated responsiveness to the MC4R agonist setmelanotide (POMC, PCSK1, LEPR, NCOA1, SH2B1) was 15.7%. Stratified by age, 30.4% (41/135), 20.1% (94/467), 19.5% (118/606), and 19.7% (206/1045) of individuals aged <6, 6- < 12, 12-18, and >18 years, respectively, carried ≥1 of the 11 studied variants.

Conclusion: In our large European-based cohort of individuals with early-onset, severe obesity, 20.4% carried a variant in ≥1 of the 11 studied MC4R pathway-related genes. Genetic testing of individuals with severe obesity may be an important part of clinical care at any age.

Grants:

Conflict of Interest: Anthony Goldstone received speaker fees from Rhythm Pharmaceuticals, received speaker fees from Rhythm Pharmaceuticals, JESUS DOMINGUEZ RISCART: None declared, Giusi Umano: None declared, Orit Hamiel: None declared, Melek Yildiz: None declared, Melania Manco has received a speaker fee from Rhythm Pharmaceuticals, Patrick Sleiman holds company-awarded stocks/options from Rhythm Pharmaceuticals, employee of Rhythm Pharmaceuticals, Charles Savoie holds company-awarded stocks/options from Rhythm Pharmaceuticals, employee of Rhythm Pharmaceuticals, Dinesh Giri: None declared, Jesús Argente has received institutional study funding from Rhythm Pharmaceuticals as a clinical investigator, has received compensation for speaking engagements from Rhythm Pharmaceuticals, has received compensation for consulting; participated in an advisory board for Rhythm Pharmaceuticals

EP22.003 The multigenic analysis revolution: a retrospective review of probands with unclear genetic conditions

Massimiliano Chetta 1, Marina Tarsitano2, Maria Oro3, Maria Rivieccio3, Liberato Marzullo4, Alessandra Rosati5, Nenad Bukvic6

1Cardarelli Hospital, Medical genetics and genomics, napoli, Italy; 2AORN Cardarelli Hospital, Medical genetics and genomics, napoli; 3AORN Cardarelli Hospital, Medical genetics and genomics, Naples; 4Unisa, Department of Medicine, Salerno, Italy; 5Salerno, Department of Medicine, Salerno, Italy; 6Policlinico di Bari, Medical genetics and genomics, Bari

Background/Objectives: Next Generation Sequencing (NGS) has revolutionized the genetic study of the human genome, although its diagnostic accuracy is approximately 25–50% accurate. Despite evidence showing several genes interact to produce complicated disorders, classical genetics still only finds one damaging mutation in a single connected gene. The idea of a multi-genic enriched approach to analysis shifts the focus from patient- phenotype to individual specific genes.

Methods: Clinical exome sequencing (Sophia-CES-V2) was performed retrospectively on 32 probands with various pathologic traits The investigation evaluated the possible gene correlation for all variants of unknown significance (VUS), pathogenic, and likely pathogenic variants by applying the text mining tools.

Results: Enriched analysis was used to uncover novel relationships and to identify modifying genes that improved diagnosis. Compared to conventional methods, text mining provided significantly more information regarding the links between genes and diseases. Individual patient-specific gene variations provide insight into the underlying causes of disease, enabling more precise diagnosis and treatment planning.

Conclusion: NGS evolution presents a challenge in genetically variable illnesses because to its variable diagnostic rates. Current instruments improve accuracy by employing OMIM and HPO filters; nonetheless, their capacity to explore outside preset limits is restricted. In complicated situations when genetic relationships are unclear, multigenic analysis provides a more comprehensive picture and is essential. The study suggests that a multigenic strategy might be applied to improve diagnostic accuracy, provide individualized treatment for uncommon genetic illnesses, and strengthen the basis of precision medicine.

Conflict of Interest: None declared

EP22.004 Evaluation of Hsp90 inhibitors as optimal drug modulators.

Emanuela Pesce 1, Elisa Giorgio2;3, Tiziano Bandiera4, Stefano Ratti5, Ilaria Cani5, Pietro Cortelli5;6, Nicoletta Pedemonte1

1IRCCS Istituto Giannina Gaslini, UOC Genetica Medica, Genova, Italy; 2IRCCS Mondino Foundation, Neurogenetics Research Centre, Pavia; 3University of Pavia, Department of Molecular Medicine, Pavia; 4Istituto Italiano di Tecnologia, D3 PharmaChemistry Line, Genova; 5Università di Bologna, Dipartimento di Scienze Biomediche e Neuromotorie, Bologna; 6IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna

Background/Objectives: In our previous study (Giorgio et al, 2020) we used a pharmacological screening approach to identify compounds able to reduce lamin B1 levels, whose overexpression is responsible for the autosomal dominant leukodystrophy disorder. We screened a library of 717 biologically active compounds and approved drugs and identified alvespimycin, an Hsp90 inhibitor, as a positive hit. We will test additional Hsp90 inhibitors to gain evidence of the role of specific Hsp90 isoform(s) involved in lamin B1 biogenesis and intracellular processing.

Methods: We tested a second set of 10 Hsp90 inhibitors. Experiments were performed on dual-reporter CHO cells by using high‐content imaging methodologies. For each compound we measured the expression level of lamin B1 fused to an AcGFP, of aDsRed-RFP (to normalize the signal), and their ratio. Finally, the number of cells informed us about possible cytotoxic effects of the compound tested.

Results: Among those Hsp90 inhibitors, only two compounds reduced cell AcGFP fluorescence intensity in a dose-dependent manner. All other Hsp90 inhibitors have no major effect on lamin B1. However, it was not possible to highlight the role of a specific Hsp90 isoform as a lamin B1 regulator. This result is in agreement with the promiscuous role of Hsp90 towards misfolded proteins, such as DYRK1A.

Conclusion: Further studies will be necessary to understand whether Hsp90 plays a direct role in lamin B1 folding or degradation, or whether this effect occurs indirectly, through modulation of other proteins that regulate lamin B1 protein levels.

Grants: “Ricerca Finalizzata GR-2021-12373348” from the Italian Ministry of Health to EP

Conflict of Interest: None declared

EP22.005 SINEUP in Lafora Disease: an innovative approach to treat Lafora bodies accumulation

Lorenzo Allegri1, Federica Baldan 1, Catia Mio1, Maimouna Shaar1, Giuseppe Damante1

1University of Udine, Department of medicine, Udine, Italy

Background/Objectives: Lafora Disease (LD) is a rare autosomal recessive progressive myoclonus epilepsy with no curative therapies. It affects healthy children/adolescents, causing epilepsy, myoclonus, and psychomotor deterioration, leading to loss of autonomy and death after a median of 6 and 11 years, respectively [Pondrelli2021]. LD is caused by loss of function mutations in either EPM2A, encoding the glycogen phosphatase laforin, or EPM2B, encoding the E3-ubiquitin ligase malin. These mutations lead to the accumulation in the brain of insoluble polyglucosan aggregates, known as Lafora bodies (LBs) [Nitschke2018], responsible for neurodegeneration and clinical manifestations. While gene replacement therapy appears to be an obvious treatment option for LD, it is still in early stages of development and does not act on already formed LBs. Considering that the elimination of LBs has been shown to rescue the neurological phenotype in LD mouse models [Duran2014], our alternative therapeutic approach involves the degradation of accumulated LBs.

Methods: Since the pancreatic alpha-amylase has been shown to degrade LBs and restore neurological functions, we have explored an innovative approach for the treatment of LD patients by using SINEUP to increase alpha-amylase expression in vitro to levels capable of degrading LBs.

Results: The data obtained so far have allowed us to demonstrate that SINEUPs are capable of increasing the protein levels of alpha-amylase in various cellular models.

Conclusion: Further experiments will be needed to prove that this increase is sufficient to induce an enhancement of amylase activity and the degradation of LBs.

Grants: PNRR Investimento 2.1 M6C2. PNRR-MAD-2022-12376430

Conflict of Interest: None declared

EP22.006 Developing a framework for precision therapy for rare and genetic diseases

Siobhan Kerr 1, Kinga Groß2, Johannes Zschocke2, jonathan berg1

1School of Medicine - University of Dundee, United Kingdom; 2University of Innsbruck, Innsbruck, Austria

Background/Objectives: The identification of the majority of genes that cause monogenic disorders in humans over the past 30 years has led to the possibility of developing highly specific therapies that truly modify disease progression. Multiple different approaches have been developed that are effective in different genetic disorders, and an exponential increase in highly specific therapies (that address a specific gene activity or even a specific pathogenic variant) is expected. A conceptual framework for classifying precision therapies is required to improve understanding amongst all healthcare professionals, and to facilitate design of therapies for new rare indications.

Methods: We identified newly emerging therapies for rare and genetic diseases through a journalistic review and from online clinical trials databases. Common mechanisms of action were identified and mapped to the central dogma of protein production.

Results: We identified 10 specific mechanisms of action.

I. Gene Replacement,

II. Gene Editing

III. Epigenetic Modification

IV. RNA Knockdown

V. RNA Editing

VI. Translation (stop codon readthrough)

VII. Post Translational Modification

VIII. Protein Interaction

IX. Protein Replacement

X. Environmental Modification

Conclusion: This framework supports a common understanding of the place of action of different therapeutic interventions. It also provides a structure for teaching about emerging new classes of therapeutic agent. Such therapies are currently largely restricted to the precise treatment of rare disease, but application of the same technologies to the treatments of a wide range of common medical disorders is already emerging.

Grants:

Conflict of Interest: None declared

EP22.007 Investigation of the effect of vosoritide treatment on linear growth in pediatric patients with achondroplasia: A preliminary study

Nilay Gunes 1, Dilek Uludag Alkaya1, Beyhan Tüysüz1

1Istanbul University-Cerrahpasa, Cerrahpaşa Medical Faculty, Department of Pediatric Genetics, Istanbul, Türkyie

Background/Objectives: Achondroplasia is an autosomal dominant disorder caused by heterozygous pathogenic variants in the FGFR3 gene. Gain of function variants of FGFR3 inhibit chondrocyte proliferation and differentiation, which negatively regulates bone growth. Vosoritide is a C-type natriuretic peptide analog designed to increase linear growth that has been FDA-approved for treatment in achondroplasia patients with open epiphyses aged 5 years and older. This study has aimed to investigate the follow-up findings of a small achondroplasia cohort receiving vosoritide treatment.

Methods: Clinical findings of five achondroplasia patients receiving vosoritide treatment and a control group according to age including 10 achondroplasia patients who are not receiving the treatment were evaluated.

Results: In the vosoritide-treatment group, the median annualized growth velocity was 5.63 cm/year during the median treatment period of 0.44 (0.24-1.11) years. No significant treatment-related adverse effects were detected. The mean growth velocity was 4.44 cm/year in the control group. The growth velocity value was higher in the treatment group with a difference of 1.19 cm/year. In the molecular studies of study and patient groups, 14 patients including four study and 10 control patients had the FGFR3:c.1138G>A variant, whereas one study patient had the FGFR3:c.1138G>C variant.

Conclusion: We have found a higher growth velocity value in the vosoritide-treatment group with a difference of 1.19 cm/year despite the relatively short treatment period. We believe that it is important to share early experiences to increase understanding of vosoritide management in children with achondroplasia.

Conflict of Interest: None declared

EP22.008 Frequency of obesity-related gene variants in a European population with early-onset, severe obesity

Carel Le Roux1, JESUS DOMINGUEZ RISCART2, Maria Rosaria Licenziati3, Leandro Soriano-Guillén4, Belma Haliloglu5, Anjali Zalin6, Simona Filomena Madeo7, Patrick Sleiman8, Charles Savoie8, Liya Kerem9, Jesús Argente 10

1Diabetes Complications Research Centre, University College Dublin, Dublin, Ireland; 2Instituto de Investigación e Innovación Biomédica de Cádiz, Hospital Universitario Puerta del Mar, Universidad de Cádiz, Cádiz, Spain; 3Neuro-Endocrine Diseases and Obesity Unit, Department of Neurosciences, Santobono-Pausilipon Children’s Hospital, Naples, Italy; 4Hospital Universitario Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain; 5Department of Pediatrics and Pediatric Endocrinology and Diabetes, Marmara University Medical School, Istanbul, Türkyie; 6Barts Health NHS Trust, London, UK and Bedfordshire Hospitals NHS Foundation Trust, Bedford, United Kingdom; 7Pediatric Unit, Department of Medical and Surgical Sciences of the Mother, Children and Adults, University of Modena, Modena, Italy; 8Rhythm Pharmaceuticals, Inc., Boston, United States; 9Pediatric Endocrinology, Hadassah University Medical Center, Jerusalem, Israel; 10Department of Pediatrics & Pediatric Endocrinology, Universidad Autónoma de Madrid, University Hospital Niño Jesús, CIBER “Fisiopatología de la obesidad y nutrición” (CIBEROBN), Instituto de Salud Carlos III, IMDEA Institute, Madrid, Spain

Background/Objectives: Patients with genetic variants in the melanocortin-4 receptor (MC4R) pathway may present with early-onset, severe obesity and hyperphagia. The Rare Obesity Advanced Diagnosis™ (ROAD) genetic testing program aims to enhance access to genetic testing for patients with suspected rare genetic causes of obesity.

Methods: The sequencing panel included 79 genes and 1 chromosomal region with well-established associations with obesity and the MC4R pathway. Eligible individuals had early-onset, severe obesity, were an immediate family member of selected previously tested patients, or showed clinical symptoms that suggested Bardet-Biedl syndrome.

Results: Overall, 2,253 individuals were sequenced in Spain (n = 1,020), Italy (n = 508), Ireland (n = 216), Turkey (n = 178), Israel (n = 189), the United Kingdom (n = 138), and Germany (n = 4), of whom 1,208 (54%) were aged <18 years. Mean BMI Z score of individuals aged ≤18 years was 3.40, mean BMI of individuals aged >18 years 44.2 kg/m2, and mean age of obesity onset for all individuals 6.9 years. Genetic variants that have been indicated for treatment in Europe with the MC4R agonist setmelanotide or that are being investigated in setmelanotide clinical trials were identified in 46 (2.0%) and 661 (29.3%) individuals, respectively. Additionally, 481 (21.3%) individuals had variants that might support a diagnosis of genetic obesity but are not being indicated or investigated for setmelanotide. Genetic variants were not identified in 1065 (47.3%) individuals.

Conclusion: In this cohort, ~35% of individuals with early-onset, severe obesity carried potentially actionable variants. Genetic testing of these patients may be an important component of understanding the etiology of the disease.

Grants:

Conflict of Interest: Carel Le Roux is a member of the Irish Society for Nutrition and Metabolism outside the area of work commented on here; was the chief medical officer and director of the Medical Device Division of Keyron in 2011, reports grants from the Irish Research Council, Science Foundation Ireland, Anabio, and the Health Research Board, serves on advisory boards of Novo Nordisk, Herbalife, GI Dynamics, Eli Lilly, Johnson & Johnson, Glia, and Boehringer Ingelheim, JESUS DOMINGUEZ RISCART: None declared, Maria Rosaria Licenziati: None declared, Leandro Soriano-Guillén: None declared, Belma Haliloglu: None declared, Anjali Zalin: None declared, Simona Filomena Madeo: None declared, Patrick Sleiman holds company-awarded stocks/options from Rhythm Pharmaceuticals, is an employee of Rhythm Pharmaceuticals, Charles Savoie holds company-awarded stocks/options from Rhythm Pharmaceuticals, is an employee of Rhythm Pharmaceuticals, Liya Kerem: None declared, Jesús Argente has received institutional study funding from Rhythm Pharmaceuticals as a clinical investigator, has received compensation for speaking engagements from Rhythm Pharmaceuticals, has received compensation for consulting from Rhythm Pharmaceuticals; participated in an advisory board for Rhythm Pharmaceuticals

EP22.010 Genetic correction of CD 36/37 (-T) β-thalassemia by CRISPR/Cas9 system in human hematopoietic stem cells as a common mutation of southwest of IRAN

nasim mayeli fereydani 1, mansoureh ajami2, Elham Hoveizi1, Arash Alghasi3, Hamid Galehdari1

1Shahid Chamran University, Department of biology, Ahvaz, Iran; 2Tehran Medical Sciences, Islamic Azad, Department of Hematology, Tehran, Iran; 3JondiShapour University of Medical Science, Thalassemia & Hemoglobinopathy Research center, Ahvaz, Iran

Background/Objectives: Beta-thalassemia is one of the most common monogenic inherited disorders across Mediterranean countries, the Middle East, and Southeast Asia. Genome-editing based on clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 system (CRISPR/Cas9) has raised the hope for life-long gene therapy of beta-thalassemia. This study aimed to correct the frame-shift mutation CD 36/37 (-T), the most common mutation in southwest IRAN, using a CRISPR/Cas9 gene editing strategy based on HDR in Hematopoietic stem cells.

Methods: The selected sgRNAs were designed and cloned into a PX458 plasmid. DNA template sequence containing phosphorothioate modification and two silent mutations at the Cas9 enzyme recognition site (PAM) was designed and synthesized. The efficiency of on-target mutagenesis by the SpCas9/sgRNAs was assessed by T7EI assays and non-denaturing PAGE in HEK293 cells. HDR-positive cells were isolated via qPCR techniques using specific-designed probe sequences for generated donors’ SNP and PCR-RFLP based on the new cleavage site created for the Hinf I enzyme on the donor DNA sequence.

Results: Our results showed that among four different gRNAs designed with different distances relative to the CD 36/37 (-T) target locus, gRNA1 had the highest cutting efficiency of 66% in HEK293 cells. In the following, qPCR data showed ~ 30% HDR efficiency in HEK293 cells and 23.91% in HSCs. Additionally, the results of enzymatic digestion revealed that the average occurrence of HDR in HSCs-positive clones was 48.52%.

Conclusion: Our HDR-based gene-editing strategy could efficiently target this specific HBB locus and induce HDR-positive cells.

Grants: Scu-1402

Conflict of Interest: None declared

EP22.011 The Impact of nutritional ketosis on autophagy in the brain of Tay-Sachs mouse model

Orhan Kerim Inci 1, Volkan Seyrantepe1

1Izmir Institute of Technology, Molecular Biology and Genetics, Izmir, Türkyie

Background/Objectives: Tay-Sachs disease is a rare genetic disorder primarily affecting the central nervous system which arises from an aberrant accumulation of GM2 ganglioside due to the deficiency of β-hexosaminidase-A. Tay-Sachs mouse model (Hexa-/-Neu3-/-) displayed severe neuropathology mimicking Tay-Sachs patients. Recently, impaired autophagic flux was reported by the accumulation of autophagosomes and autophagolysosomes in Hexa-/-Neu3-/- mice brains. The ketogenic diet is a high-fat/low-carbohydrate diet that has been used based on its neuroprotective properties. However, the mechanisms responsible for these effects are partially elucidated. Autophagy induction may be one of the mediators of the neuroprotection provided by the ketogenic diet. In this study, we aimed to investigate the effect of ketogenic diet treatment on autophagic impairment in the Tay-Sachs mouse model.

Methods: 140-day-old WT, Hexa-/- and Hexa-/-Neu3-/- mice were treated under the following groups: i) chow diet, ii) ketogenic diet (10-days). Autophagy markers were analyzed by RT-PCR, western blotting, and immunofluorescence in the cortex and cerebellum.

Results: The ketogenic diet treatment in Hexa-/-Neu3-/- mice paved the way for a significant reduction of Beclin-1, p62, and Lamp-2 expression levels indicating modulation of impaired autophagy. Consistently, western blotting analysis showed decreasing levels of p62 and LC3-II. Immunofluorescence staining of the p62 and LC3 also indicated attenuation of the accumulated autophagic cargoes in the cortex and cerebellum

Conclusion: Altogether, our results suggest that a ketogenic diet could be a potential treatment to correct impaired autophagy in the Tay-Sachs disease mouse model.

Grants: The research was partially funded by TUBITAK-218S824 through COST Action (CA17103) and TUBITAK-123Z087.

Conflict of Interest: None declared

EP23 Genetic Counselling / Services / Education

EP23.001 Evaluating co-designed interventions supporting pediatricians to order funded genomic tests: a website, ‘ask-a-genetics-expert’ contact service and awareness raising activities

Belinda McClaren 1;2;3, Melissa Martyn1;2;3, Jessica Ince1;2, Emma Weisz1;2;3;4, Katie Arkell1, Natasha J Brown3;5, Michael C. Fahey4, Clara Gaff1;2;3

1Murdoch Children’s Research Institute, Genomics in Society, Melbourne, Australia; 2Melbourne Genomics Health Alliance, Walter and Eliza Hall Institute, Melbourne, Australia; 3University of Melbourne, Melbourne, Australia; 4Monash Children’s Hospital, Melbourne, Australia; 5Victorian Clinical Genetics Service, Melbourne, Australia

Background/Objectives: Funded genomic tests for childhood syndromes can be ordered by paediatricians in Australia; however, use has been low. We previously interviewed paediatricians and identified barriers including: low confidence, low knowledge, limited access to clinical geneticists. Paediatricians suggested interventions to: raise awareness, provide resources in one place, facilitate access to genetic experts, strengthen professional networks. Such interventions may reduce the burden on genetic services by supporting paediatricians to order genomic testing. We aimed to co-design interventions to support paediatricians to order funded genomic tests.

Methods: Two cycles of co-design were completed with: paediatricians, parents/support groups, genetic health professionals. Each group participated in two 2-hour facilitated workshops: 1) validating and prioritising identified barriers and enablers; 2) designing interventions. Evaluation is informed by the Re-AIM framework.

Results: There were 32 workshop attendees. A website intervention, to provide a ‘one-place-to-go’ for paediatricians, was conceptualised and designed with three key components: 1) a step-by-step guide; 2) a curated collection of resources; 3) a phone/email contact service staffed by a genetics expert (a genetic counsellor). The URL chosen was: www.paediatricgenomics.org.au.

An awareness raising and educational campaign continues, led by the project clinician, EW, a paediatrician. Activities include: presentations at conferences and department meetings; social media; newsletters. To date, n = 155 paediatricians have provided their email address to access the full website and the genetic expert service is staffed and in use.

Conclusion: Co-design has generated interventions for paediatricians, by paediatricians. Evaluation will explore behaviour change and inform future sustainable supports for paediatricians.

Grants: none

Conflict of Interest: None declared

EP23.002 Collaboration between ART clinic and hospital regarding Genetic Counselling for PGT-A

Akane Kondo 1;2, Mikio Morine1, Takehiko Matsuyama3, Kazuhisa Maeda1

1Shikoku Medical Center for Children and Adults, Medical Genetics Center, Zentsuji, Japan; 2Aiiku Clinic, Clinical Genetics, Fetal Medicine, Minato City, Japan; 3Koujin Hospital, Marugame, Japan

Background: In Japan, Preimplantation genetic testing for aneuploidy or chromosomal structural rearrangement (PGT- A/SR) has been introduced as advanced medical care in 2023. Obviously, it is recommended to provide genetic counselling before and after those tests. However, there is only few geneticist or genetic counsellors at reproduciton clinic for now. We reviewd genetic counselling of all the PGT-A cases under collaborative medical care between medical genetic center at tertiary hospital and regional reproduction clinic.

Methods: We conducted retrospective observational study focusing on genetic counselling and patients atitude from hospital chart and PGT-A report of 47 embryos in 2023.

Results: The average age of patients was 39.2 y/o. Out of 47 embryos, only 6 embryos were euploid withoug any mosaicism (12.8%). 8 embryos shows low frequency mosaicism (17.0%). 13 embryos showed high frequency mosaicism (27.7%) and 27 embryos were aneuploidy (57.4%). High-risk mosaicism including chromosome 8,9,13,16,18,21 were shown in 15 embryos and 19 embryos were High-risk for UPD. Most of patients assume PGT-A is for having “Normal Baby” and did not consider further prenatal testing.

Conclusion: Since we have rather advanced aged patients, only 12.8% was diagnosed as euploid. Majority of PGT-A result showed mosaicism and patients needed careful genetic counselling. This collaborative medical service could be feasible to provide good genetic counselling and prenatal follow up including further diagnostic test.

Conflict of Interest: None declared

EP23.003 Genetic counselling for hereditary tumor risk syndromes – experience from a single center

Mari Hachmeriyan 1;2, Mariya Levkova1;2, Trifon Chervenkov2;3, Eleonora Dimitrova4;5, Milena Stoyanova1;2, Dinnar Yahya2;4, Lyudmila Angelova2

1University Hospital Sveta Marina, Laboratory of Medical genetics, Varna, Bulgaria; 2Medical University, Department of Medical genetics, Varna, Bulgaria; 3University Hospital Sveta Marina, Laboratory of clinical immunology, Varna, Bulgaria; 4University Hospital Sveta Marina, Medical Oncology Clinic, Varna, Bulgaria; 5Medical University, Department of medical oncology, Varna, Bulgaria

Background/Objectives: Genetic counselling (GC) is crucial in identifying and providing complex care for individuals at risk for hereditary tumor syndromes. This study aims to evaluate the group of patients with probable tumor risk syndromes undergoing GC and further suggests possibilities for improvement.

Methods: We assessed our experience in GC provision based on available guidelines for the last four years (2020-2023). Risk calculation, pre- and post-test genetic counseling were available to all 123 patients considering and/or undergoing genetic testing.

Results: Oncogenetic patients represented 6.5% of all counseled in our center. Their number increased twice over the period predominantly affecting patient-paid service. The majority were women (65.9%) and aged between 18-50y (65.9%). Children remained to be the smallest group (6.5%). As for referrals, the majority were by a clinician (77.2%) instead of self-referred. Also, the indication of cancer diagnosis (54%) dominated slightly over being healthy with a positive family history or other predisposing factors (42%). Only 4% underwent oncogenetic counselling as a proactive prophylactic attitude.

Conclusion: There is a tendency for escalation in GC for hereditary tumor risk syndromes, mostly concerning outpatient younger women referred by clinicians, despite being patient-paid. Regardless of the initial indications and the restrictions of cancer genetic tests, we notice an increasingly active attitude toward GC. Improved public education about hereditary cancer is needed but clinicians’ role remains essential.

Grants: This study is financed by the European Union-NextGenerationEU, through the National Recovery and Resilience Plan of the Republic of Bulgaria, project № BG-RRP-2.004-0009-C02.

Conflict of Interest: None declared

EP23.004 Leveraging a Genetic Counselling Community of Practice to support the use of genomics in medical specialties

Trang Do 1;2, Melissa Martyn1;2;3, Belinda McClaren1;2;3, Clara Gaff1;2;3, Alison McEwen4

1Murdoch Children’s Research Institute, Parkville, Australia; 2Melbourne Genomics Health Alliance, Australia; 3The University of Melbourne, Parkville, Australia; 4University of Technology Sydney, Ultimo, Australia

Background/Objectives: A Community of Practice (CoP), a group of people with shared interests (Wenger, 2011), can be a powerful approach to support healthcare professionals and advance systems change. Genetic counsellors are at the centre of change in genomic healthcare, working in new ways beyond clinical genetics departments. We established a CoP for genetic counsellors in metropolitan Melbourne (Australia) with meetings held fortnightly with an experienced facilitator and evaluated its impacts on participants.

Methods: Guided by socio-cultural learning and knowledge translation theories and the Promoting Action on Research Implementation in Health Services (PARIHS) framework, we analysed data using 25 qualitative interviews, discussion notes of 28 sessions, meeting minutes, and self-reflection over a 12-month period.

Results: This CoP was characterised by hybrid organisation, evolving structure, diverse seniority, regular reflection, and a dynamic focus. CoP members acknowledged the emotional and professional value of participating in the CoP, as they benefited from regular interactions, a safe knowledge-sharing space, and professional development opportunities. Participants reported applying the skills and knowledge from the CoP to advance interprofessional communication about genomics and address real-world situations encountered while working outside of clinical genetics services. They identified their own diverse perspectives (from current employment and past mainstream experience) as contributing to the CoP. Additionally, the CoP served as an effective channel for identifying operational issues that required management action.

Conclusion: Our evaluation demonstrates that a CoP to support practice and professional development is beneficial for genetic counsellors transitioning to work beyond clinical genetics in response to growing demands for genomic healthcare.

Conflict of Interest: Trang Do Fulltime, Melissa Martyn: None declared, Belinda McClaren: None declared, Clara Gaff: None declared, Alison McEwen: None declared

EP23.006 Analytical system QazGenIs with artificial intelligence for monitoring genetic pathology

Gulshara Abildinova1, Meruert Zhumazhanova2, Laura Ainabekova2, Dana Makhsutova2, Maral Muzar2, Manshuk Zhulkarova2, Tattikul Tazhenova3, Zhulduz Zhumadilova3, Nurbibi Suindikova4, Gulnara Abdresheva 5

1Medical Center Hospital of the President’s Affairs Administration of the Republic of Kazakhstan, Laboratory genetics, Astana; 2City multidisciplinary hospital №2, Medical Genetics Department, Astana, Kazakhstan; 3City multidisciplinary hospital №2, Astana; 4Сity perinatal center, Medical Genetics Department, Shymkent, Kazakhstan; 5City Center for Human Reproduction, Medical Genetics Department, Almaty

Background/Objectives: Congenital and inherited diseases are essential problem of medicine and society. During the last decade congenital anomalies remain unchanged on leading positions in the structure of causes of perinatal, neonatal, infant morbidity, disability and mortality.

Aiming studying frequency, structure of congenital and inherited pathologies to estimate genetical load in population of Kazakhstan information and analytical system QazGenIs was created.

Methods: system has all essential information about pregnant women from risk group that appeared to have congenital malformations diagnosed during ultrasound; fetuses with chromosomal abnormalities from 10 weeks to 22 weeks; patients with congenital malformations, chromosomal and monogenic pathologies.

The system contains four sections: congenital malformations, prenatal diagnosis, chromosomal pathology, monogenic pathology and is equipped with artificial intelligence (AI).

Results: Preliminary results of the structure of genetic pathology: 48% - congenital malformation, 38% - chromosomal and 14% - monogenic pathology. Among congenital malformations, the largest share is occupied by defects of the circulatory system - 23.7%, in second place are unilateral malformations - 16.9% and congenital malformations of the urinary system were found in 15.3% of patients. Congenital malformations of the digestive organs were registered in 14.3% of patients, the nervous system in 13.1%, the musculoskeletal system - 6.4%, and the facial structures in 5.3% of cases.

Conclusion: Thus, the formation of a database of congenital and hereditary pathologies with full coverage of all newborns, children, and fetuses will make it possible to assess the genetic load of the population and propose measures for practical healthcare.

Conflict of Interest: None declared

EP23.007 Childhood Dementia Insight: A mixed-methods study investigating the psychosocial, quality of life, and psychological impacts of childhood dementia on families

Suzanne Nevin1;2, lauren kelada 1, Claire Wakefield1, gail hilton3, meg maack3, jason djafar1, kristina elvidge3, michelle farrar1

1UNSW Sydney, school of clinical medicine, Sydney, Australia; 2Sydney Children’s Hospital, Randwick, Randwick, Australia; 3Childhood Dementia Initiative, Sydney, Australia

Background/Aims: Childhood dementia is a devastating group of life-threatening and life-limiting disorders, defined by progressive decline of neurocognitive function and enduring loss of previously acquired skills. With an incidence of 1:2900 births and >300 causal genes identified, genetic testing has become standard care. Children and their families experience substantial biopsychosocial impacts associated with the complex and idiosyncratic nature of individually rare genetic childhood dementias1. Less than five percent of childhood dementias have disease-modifying therapies and family-centered psychosocial resources following diagnosis are lacking. Taking a collective approach, this research involves an in-depth mixed-methods, multi-perspective study to investigate and address the shared psychosocial impacts of childhood dementia on families.

Methods: Parents of children with clinically confirmed or suspected genetic childhood dementia will complete a series of validated and purpose-designed measures to quantify the psychosocial, psychological and economic impacts of their child’s diagnosis. Additionally, parents will be invited to complete an individual in-depth, semi-structured interview to elucidate their unique perspectives, experiences undergoing genetic testing and to identify parents’ unmet needs. A convergent, mixed-methods approach, combining the results from the quantitative and qualitative measures will be adopted to enhance interpretation of parents’ experiences.

Results: This research will quantify and qualify important themes reflecting the collective impacts of childhood dementia on parents and bereaved parents.

Conclusion: Co-designed and evidence-based resources are crucially needed to provide integrated, multidisciplinary support for parents and bereaved parents of children with dementia. Our findings will inform future best-practice genetic healthcare services and psychosocial supports for children and affected families.

Conflict of Interest: None declared

EP23.008 The importance of revising previously established diagnoses and genetic test results in clinical practice - a case report

Monika Pokorna 1, Štefan Lednický1, Monika Koudová1, David Stejskal1

1Gennet, Prague, Czech Republic

Background/Objectives: A 30-year-old neurologically healthy woman in her first pregnancy was consulted in our genetic centre due to clinically present motoneuron disease in her father (proband). According to the available neurological and genetic history, the father was diagnosed with adult-onset SMA in 2006, despite negative genetic testing for SMA and spinobulbar muscular atrophy.

Methods: Panel of 143 genes associated with the development of neurological diseases was analyzed with NGS in the proband with negative result. Bioinformatics processing of the NGS data suggested the expansion of CAG repeats in exon 1 of the AR gene. Confirmation was performed using the PCR sequencing.

Results: Molecular genetic testing confirmed expansion of CAG repeats of AR gene, which leads to the development of X-linked recessive spinobulbar muscular atrophy. Our patient and her sister were confirmed as carriers of Kennedy’s disease by molecular genetic examination.

Conclusion: The possibilities of genetic diagnostics in the past were very limited and did not allow accurate classification of the disease. In the case we present, diagnosis of adult-onset SMA was very improbable because of negative SMN1 gene testing. The confirmation of the Kennedy’s disease, despite the previously negative results of genetical examination in 2006 was surprising. Establishing an accurate molecular and clinical diagnosis allowed for the investigation in relatives while allowing the opportunity to offer healthy carriers the option of invasive prenatal testing and preimplantation genetic testing as part of genetic prevention.

Conflict of Interest: None declared

EP23.009 Incorporating the patient voice into the development of a clinical guideline to support a direct contact approach to information sharing in families.

Angela Bedard1;2;3, Samantha Pollard4;5, Melanie O’Loughlin3;6, Anna Tsoy2, James Bedard2, Christina Rajsic7, Fabio Feldman6, Farah Jamal8, Alice Virani3;9, Sophie Sun6, Kasmintan Schrader3;6;10, Jennifer Nuk 3;11

1BC Cancer, Hereditary Cancer Program, Abbotsford, Canada; 2University of the Fraser Valley, Abbotsford, Canada; 3University of British Columbia, Medical Genetics, Vancouver, Canada; 4Fraser Health, Department of Evaluation and Research Services, Surrey, Canada; 5BC Cancer Research Institute, Vancouver, Canada; 6BC Cancer, Hereditary Cancer Program, Vancouver, Canada; 7Provincial Health Services Authority, Risk Management, Vancouver, Canada; 8BC Cancer, Policy Office, Vancouver, Canada; 9Provincial Health Services Authority, Ethics Service, Vancouver, Canada; 10BC Cancer, Molecular Oncology, Vancouver, Canada; 11BC Cancer, Hereditary Cancer Program, Victoria, Canada

Background/Objectives:To improve cascade carrier testing uptake, some hereditary cancer clinics provide the option of genetic testing directly to relatives rather than relying on a family-mediated approach. To support the development of a direct contact clinical guideline for our provincial program, we explored the attitudes of hereditary cancer patients to this approach to better understand their preferences, concerns and expectations.

Methods: Individuals with a pathogenic variant in a hereditary cancer gene were identified. Purposive sampling guided participant selection to support diversity. Semi-structured interviews were conducted via Zoom or telephone. The interview guide was co-developed alongside patient partners. Interviews were recorded, professionally transcribed, de-identified, and reviewed for accuracy prior to transcript coding using Nvivo qualitative coding software. Two qualitative analysts applied thematic analysis.

Results: Eleven females and 4 males ages 35-70 years were interviewed. In general, participants supported enhanced assistance from the clinic in sharing genetic testing information with their family. There was variation in preferences around timing and level of clinic involvement along with detail and format of information provided to relatives. Perceived benefits included reducing burden for cancer patients and navigating complex family relationships. Concerns included violations of privacy and impact of learning about cancer in the family in this way.

Conclusion: Our findings suggest strong support for a collaborative and tailored approach to results communication. Results were integrated into a patient values-informed supported-direct contact clinical practice guideline to facilitating cascade carrier testing in British Columbia, endorsed by our institution’s policy office, ethics service, risk management and operational leadership.

Conflict of Interest: None declared

EP23.010 Family screening for germline mutations in the BRCA1 and BRCA2 genes

Mariana Mazza Santos1, Pricila Bernardi2, Anelize Baranzeli1, Thais Assis1, Bruna Kellet Coelho1, Bruno Hech Dominski1, Sara Silva Lima1, Maristela Ocampos 1

1Mercolab Diagnósticos, Molecular Biology, Palhoça, Brazil; 2Polydoro Ernani de São Thiago University Hospital, Florianópolis, Brazil

Background: Hereditary cancer syndromes increase the chance that individuals in the same family will develop certain types of tumors throughout their lives. They are caused by pathogenic or possibly pathogenic germline variants that are transmitted between generations. BRCA mutation identification is important for both genetic counseling and treatment options in patients with metastatic disease. The aim of this study was to screen for BRCA1 and BRCA 2 genes in families diagnosed with breast cancer in patients from the public health system in Santa Catarina.

Methods: Whole blood was collected from three families, with at least one case of breast cancer diagnosis, totaling 18 patients, including symptomatic and asymptomatic. The preparation of the amplicon library used the Oncomine™ BRCA Research Assay kit on the Ion Torrent platform. The variant call was performed in the Ion Reporter software and the variants were classified according to the ACMG criteria.

Results: In the families investigated, two pathogenic variants were identified, p.Ala938ProfsTer21 (BRCA2) and p.Gin1756ProfsTer74 (BRCA1). The pathogenic variant p.Ala938ProfsTer21 is one of the most common variants in the Brazilian population and is present in two of the families analyzed. The presence of this mutation is associated with early and familial cancer of the breast and/or ovarian, prostate, and male breast.

Conclusion: Of the total number of patients analyzed, only one person was a carrier of a pathogenic and asymptomatic variant, reinforcing the need for molecular screening and genetic counseling for the treatment of families with cancer cases.

Grants: Unimed Mercosul, Laboratório Mercolab Diagnósticos e HU -SC

Conflict of Interest: None declared

EP23.011 GenNatal: a genetic newborn screening pilot project in Spain

Diana Salinas-Chaparro 1, Laura Ortega2, mar borregán prats1, Gema Escribano-Serrano1, alba pascual rodríguez3;4, Natalia Castejón-Fernández2, Guerau Fernandez1;3;4, Judith Armstrong1;3;4, Delia Yubero1;3;4, Cristina Hernando-Davalillo1, Adrián Alcalá San Martín1, Maria Victoria Dominguez5, Sandra González Castejón5, Laura Mallen5, Federico Mayor Zaragoza6, Magdalena Ugarte2, Belén Pérez2;4, Francesc Palau1;3;4;7

1Hospital Sant Joan de Déu, Servicio de Genética Clínica y Molecular, Barcelona, Spain; 2CIBERER. IdiPAZ. Universidad Autónoma de Madrid, Centro de Diagnóstico de Enfermedades Moleculares, Madrid, Spain; 3Institut de Recerca Sant Joan de Déu, Barcelona, Spain; 4CIBER de Enfermedades Raras (CIBERER). ISCIII., Madrid, Spain; 5BCNatal, Hospital Sant Joan de Déu, Departamentos de Obstetricia y Ginecología, Barcelona, Spain; 6Fundación Cultura de Paz, Madrid, Spain; 7Facultad de Medicina y Ciencias de la Salud, Universitat de Barcelona, Barcelona, Spain

Background/Objectives: Newborn screening constitutes an important public health initiative for secondary prevention of diseases in the neonatal period. Recent research initiatives worldwide have explored the integration of genomic sequencing in newborns. Genetic Newborn Screening (GNBS) proposes Next Generation Sequencing (NGS) technology to detect a broader range of actionable disorders expanding the scope of the current neonatal screening. GenNatal is a pilot project that addresses medical, genetic, ethical, and societal challenges of incorporating NGS into neonatal screening in Spain.

Methods: The study involved a first cohort of pregnant women and their partners in the third trimester of pregnancy who received comprehensive genetic counselling. Their perceptions regarding GNBS-related aspects were collected. Exome sequencing and analysis of the genes initially selected by the project team were performed on DNA from dried blood samples of newborns. A standardized bioinformatic tool for annotation and filtering for pathogenic/likely pathogenic variants was applied. Couples received appropriate post-test genetic counselling.

Results: To date, samples from 56 neonates have been obtained. None of the identified variants met the pre-established criteria in the project for reporting. The perceptions of participants have been analyzed, shedding light on aspects such as GNBS acceptance and the nature of findings warranting disclosure.

Conclusion: GenNatal has provided insights into the impact and implications of GNBS. Experience with the initial patient cohort enabled the evaluation and redesign of the inclusion criteria for screened conditions based on actionability. The acquired data will contribute to defining the importance of genetic counselling and the challenges of incorporating genetic screening into routine neonatal care

Conflict of Interest: None declared

EP23.012 Genetic counseling for "low-risk" pregnant women in a single institute, Shinshu University Hospital

Emiko Kise 1;2, Tomomi Kojima1, Akiko Sakyu1, Tomoki Kosho1;3;4

1Shinshu University Hospital, Center for Medical Genetics, Matsumoto, Japan; 2Shinshu University Hospital, Department of Nursing, Matsumoto, Japan; 3Shinshu University School of Medicine, Department of Medical Genetics, Matsumoto, Japan; 4Shinshu University School of Medicine, Division of Clinical Sequencing, Matsumoto, Japan

Background/Objectives: Shinshu University Hospital offers careful prenatal genetic counseling (GC) for the decision whether or not to have the prenatal genetic testing for any reasons. Most of the cases are ‘high-risk’ pregnant women who are advanced maternal age(AMA), etc. But there are also many cases of ‘low-risk’ pregnant women who do not have such risks. The purpose of this study is to demonstrate the prevalence of having the prenatal genetic testing after GC and factors influencing the decision whether or not to have the prenatal genetic testing.

Methods: Pregnant women who were ‘low-risk’ pregnant receiving GC for having the prenaltal genetic testing our institute from April 2013 to March 2019 were recruited. ‘low-risk’ pregnant women were not AMA, had no fetal ultrasound abnormalities, or had no any genetic disorder and carrier. Detailed information was collected through their medical records and thoroughly reviewed.

Results: There were 35 women. 8 women had prenatal genetic testing (average age 31.3 years), and 27 women did not have any testing (average age 29.8 years). Reasons for having the test were "I just want to find out what I know“, etc. Reasons for not having the test were "I can’t choose termination of this pregnancy“, etc.

Conclusion: The couples reconsidered what they wanted to be and realized that the test was not necessary for them, while they obtained correct information about the test and the typical chromosomal abnormalities. GC may need to provide a place where couples can imagine to be parents and to childcare even for ‘low-risk’ pregnant women.

Grants: None

Conflict of Interest: Emiko Kise full, Tomomi Kojima: None declared, Akiko Sakyu: None declared, Tomoki Kosho: None declared

EP23.013 Clinical genetics’ role in the diagnosis of hereditary cancer syndromes in children

Milena Stoyanova 1;2, Mari Hachmeriyan1;2, Mariya Levkova1;2, Dinnar Yahya1;2, Hristina Hristozova3;4, Lyudmila Angelova1

1Medical University Varna, Medical Genetics, Varna, Bulgaria; 2University Hospital St Marina, Laboratory of Medical Genetics, Varna, Bulgaria; 3Medical University Varna, Clinical Medical Scinces, Varna, Bulgaria; 4University Hospital St Marina, Pediatric Hematology and Oncology Clinic, Varna, Bulgaria

Background/Objectives: Clinical genetics is a field that takes an essential part in the multidisciplinary approach to pediatric patient care. This study aims to evaluate our experience in identifying children with probable cancer-predisposing syndromes (CPS) with primary manifestations other than cancer.

Methods: The study includes pediatric patients referred to the Genetics Unit of the University Hospital Saint Marina, Varna, for a period of four years (2020-2023). All children were assessed for the presence of dysmorphic features and further genetic counseling for different testing options was provided.

Results: During the study period, genetic counseling was provided to 535 children suspected of having genetic disorders. 260 children (48.6%) underwent molecular genetic testing, and 76 (29.2%) of them were diagnosed with rare single-gene disorders. A CPS was confirmed in 13 of those (17%), including 6 girls and 7 boys, aged 5 days to 7 years. The majority of them - 9 (69%), had a characteristic phenotype and CPS syndrome was suspected before genetic testing. The disorders confirmed were Neurofibromatosis type 1, Tuberous sclerosis, Fanconi anemia, Ataxia telangiectasia Cowden, Sotos, and Beckwith-Wiedemann syndromes. In the other four cases, the genetic testing revealed an unexpectedly ultrarare CPS: ACVR1-related, Shwachman-Diamond, SMARCAL1-related and Hermansky-Pudlak syndromes.

Conclusion: The pediatric field encompasses a diverse spectrum of disorders. CPS are a small subgroup but the genetic diagnosis has a great impact on the further management of both the patient and relatives at risk. Even though genetic testing is becoming more affordable and accessible, precise phenotype as a first-line approach is crucial to increase the diagnostic yield.

Conflict of Interest: None declared

EP23.014 The importance of pre- and post-genetic counselling for genetic testing in hereditary cancer

Konstantinos Agiannitopoulos 1, Natasa Katseli1, Kevisa Potska1, Christina Dogka1, Nikolaos Tsoulos1, Athanasios Papathanasiou1, Dimitrios Grigoriadis1, Georgios Tsaousis1, Konstantinos Papazisis2, Ioannis Natsiopoulos3, Ioannis Xanthakis4, Nikolas Bredakis5, Sofia Karageorgopoulou5, Dimitrios Mavroudis6, Christos Markopoulos7, Ioannis Boukovinas8, Christos Papadimitriou9, Vasileios Venizelos10, Athina Christopoulou11, Anna Koumarianou12, Eirini Papadopoulou1, George Nasioulas1

1Genekor Medical S.A., Athens, Greece; 2Euromedica, Thessaloniki, Greece; 3Interbalkan Medical Center of Thessaloniki, Thessaloniki, Greece; 4Interbalkan Medical Center of Thessaloniki, Thessaloniki, Greece; 5IASO Maternity - Gynecology Hospital, Marousi, Greece; 6University General Hospital of Heraklion, Iraklio, Greece; 7Athens Medical Center, Marousi, Greece; 8Bioclinic, Thessaloniki, Greece; 9Aretaieion University Hospital, Athina, Greece; 10Metropolitan Hospital, Pireas, Greece; 11Andrew Hospital, Patra, Greece; 12Attikon, Chaidari, Greece

Background/Objectives: Pre- and post-genetic counselling are important for the performance of genetic testing in hereditary cancer. The purpose of this study is to highlight the importance of pre- and post-genetic counselling based on the experience of Genekor laboratory.

Methods: 650 patients with cancer referred to the laboratory for multi-gene testing.

Results: Out of 650 patients with cancer referred to the laboratory for multi-gene testing 630 (96.9%) underwent pre-genetic counselling, whereas 3.1% declined to provide information. In the majority of patients (98%), the recommendation for genetic testing was made by the referring physician, while 2% were informed by another source. A large majority of patients (99%) received information about potential outcomes from their physician. It is noteworthy that 86.6% of those examined were also motivated to undergo genetic testing for their children. The average duration for pre-genetic counselling and family history collection was 15 minutes. Among the 650 patients, post-genetic counselling was conducted to 90 (13.8%) by experts from our laboratory. All patients seemed to fully understand the significance of the result and intended to share it with their relatives. In 80% of those examined, relief was expressed upon receiving their results, while 2% expressed surprise at the final outcome.

Conclusion: In conclusion, pre- and post-genetic counselling are essential stages for implementing genetic testing in hereditary cancer. Counselling should be conducted by specialized personnel (physicians, molecular biologists - geneticists) as the outcome received by the individuals examined may change their medical management.

Grants: None

Conflict of Interest: None declared

EP23.015 Deliberating genetic risks: protocol of a prospective, multi-method study on patient decision-making and disclosure from genetic counselling to the family

Milena Paneque 1;2, Maria Barbosa1;3, Angus Clarke4, Sofia Dias5, Filipa Júlio6, Alison Metcalfe7, Jorge Sequeiros1, Liliana Sousa5, Álvaro Mendes1

1CGPP-IBMC at i3S; 2Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Porto, Portugal; 3Faculty of Psychology and Educational Sciences, University of Porto, Portugal; 4Division of Cancer & Genetics, Institute of Medical Genetics, Cardiff University School of Medicine, UK; 5Department of Education and Psychology, CINTESIS@RISE, University of Aveiro, Portugal; 6European Huntington Association and Portuguese Huntington Association; 7LOHA Health Ltd, UK

With the increase in genetic and genomic testing, the need to discuss genetic risks with family members grows accordingly. Family disclosure is commonly encouraged in genetic counselling (GC), but this communication is often challenging. Research exploring the decision-making process about family disclosure is limited and mostly retrospective.

We describe a protocol for the DECIDE project, aimed at examining the impact of GC on how patients deliberate their family disclosure, and how the family and other lifeworld factors are implicated in that process. This project draws on a prospective, sequential, multi-methods study involving Portuguese medical genetics services. First, we will observe GC clinics to investigate the counselling base in which patients draw upon. Secondly, patients will be invited to keep diaries, through which we will examine their process of reflection and how it translates into communication with the family. Finally, in-depth interviews with patients will be conducted to delve into their overarching perspectives around family disclosure. We will focus on how patients’ thoughts about disclosure unfold over time, connecting GC with the negotiation of meaning regarding self and family life. This will provide an empirical foundation upon which a multi-stakeholder panel will engage (involving patients, family members, representatives of patient organizations, genetics healthcare professionals and researchers) to co-produce a consensus-based counselling framework with specific values, strategies, attitudes and practical points, validated through a Delphi study.

We plan to discuss the projects’ methodology and expected contribution with genetics healthcare professionals to fill gaps in GC research and support professional development.

Conflict of Interest: Milena Paneque This project has received funding from the FCT - Fundação para a Ciência e Tecnologia (2022. 04025.PTDC)., Maria Barbosa This project has received funding from the FCT - Fundação para a Ciência e Tecnologia (2022. 04025.PTDC)., Angus Clarke This project has received funding from the FCT - Fundação para a Ciência e Tecnologia (2022. 04025.PTDC)., Sofia Dias This project has received funding from the FCT - Fundação para a Ciência e Tecnologia (2022. 04025.PTDC)., Filipa Júlio This project has received funding from the FCT - Fundação para a Ciência e Tecnologia (2022. 04025.PTDC)., Alison Metcalfe This project has received funding from the FCT - Fundação para a Ciência e Tecnologia (2022. 04025.PTDC)., Jorge Sequeiros This project has received funding from the FCT - Fundação para a Ciência e Tecnologia (2022. 04025.PTDC)., Liliana Sousa This project has received funding from the FCT - Fundação para a Ciência e Tecnologia (2022. 04025.PTDC)., Álvaro Mendes This project has received funding from the FCT - Fundação para a Ciência e Tecnologia (2022. 04025.PTDC).

EP23.016 Addressing ethical challenges in genetic counselling: Navigating non-disclosure within families

Eulàlia Rovira-Moreno 1, Anna Abulí1, Patricia Muñoz-Cabello1, Marta Codina1, Clara Serra-Juhé2, Eva Bailles3, Eduardo Tizzano1

1Hospital Vall d’Hebron, Department of Clinical and Molecular Genetics, Barcelona, Spain; 2Hospital de la Santa Creu i Sant Pau, Genetics Department, Barcelona, Spain; 3Hospital Vall d’Hebron, Department of Psychiatry, Barcelona, Spain

The process of genetic counselling often arises ethical issues such as sharing information to the relatives at risk of developing a disease. Frontotemporal dementia is a neurodegenerative disorder with both clinical and genetic heterogeneity. After the disclosure of a positive genetic test, presymptomatic testing could be an option for other family members. This work aims to explore ethical issues surrounding familial communication in a particular case.

A 53-year-old man was referred for a genetic counselling appointment after being diagnosed with frontotemporal dementia due to a de novo pathogenic mutation in the MAPT gene. The patient had three descendants aged 19, 17 and 15 years old respectively, who were unaware of the molecular diagnosis. During the appointment, the patient’s wife expressed her unwillingness to disclose the results with her offspring requesting a delay for the communication.

This situation highlights a common dilemma in genetic counselling and the need to balance confidentiality against the potential consequences of nondisclosure. During several appointments, we adeptly addressed this conflict, considering the ethical principles. In this context, the implication of multiple health care professionals was crucial to help the patient’s wife identifying the right moment for disclosing the diagnosis, upholding the offspring’s autonomy for future life planning and support.

Facilitating the consultants the disclosure of information about genetic health risks to their relatives is key to maximize the benefits of counselling. Counsellors should balance ethical values and ensure an adequate support to their patients on planning how and when to best disclose information to the family.

Conflict of Interest: None declared

EP23.017 Women with hereditary breast cancer are willing to share information with their relatives: the effect of genetic and psychological counselling?

Elena Baranova 1, Vera Polyakova2, Tatyana Yanova2, Marina Tripolskaya2, Ekaterina Efremova1, Vera Izhevskaya3, Natalya Bodunova2

1FSBEI FPE RMACPE MOH Russia, Medical genetics, Moscow; 2Moscow Clinical Research Center named after A. S. Loginov, center for personalized medicine, Moscow, Russian Federation; 3Federal State Budgetary Scientific Institution Research Centre for Medical Genetics, Moscow, Russian Federation

Prerequisites/Purposes. With the development of genetic technologies, it has become possible to identify high-risk individuals for early detection of cancer. Our investigation aims to find out whether women who have undergone genetic testing report the test results to their relatives.

Methods. Questionnaire survey, analyses of studies, statistical methods.

We conducted a questionnaire survey among 2 groups of women with breast cancer who underwent genetic testing and mutations in breast cancer predisposition genes were identified. The first group (200) were women who underwent pre-test genetic counselling and psychologist’s consultation; the second group were women who underwent genetic testing without pre-test genetic counselling and psychologist’s consultation.

Results. When analyzing the responses of the first group of participants, we found that they disclose the results to family members (200; 100%). Many participants noted that family history (135; 67,5%), as well as the recommendation of a specialist (65; 32.5%), contributed significantly to undergoing genetic testing.

Analysis of the answers of the participants of the second group is to be continued.

Conclusions. One of the important tasks before genetic testing is the psychological preparation of the patient. Thus, the patient should be prepared to inform family members about the positive genetic risk result when it is detected. Currently, the importance of pre-test genetic counselling has increased significantly.

Grants. «The study was carried out at the expense of a grant from the Russian Science Foundation (project #19-18-00422)»

Conflict of Interest: Elena Baranova The study was carried out at the expense of a grant from the Russian Science Foundation (project #19-18-00422), collaborator, Vera Polyakova: None declared, Tatyana Yanova: None declared, Marina Tripolskaya: None declared, Ekaterina Efremova: None declared, Vera Izhevskaya The study was carried out at the expense of a grant from the Russian Science Foundation (project #19-18-00422), collaborator, Natalya Bodunova: None declared

EP23.018 neurofibromatosis type-1 independent variants in a neurofibromatosis type-1 family

rafaella metaxa 1, Andri Miltiadous2, Petroula Gerasimou2, natasa petridou1, Emily Athanasiou3, Paul Costeas4, Violetta Anastasiadou3

1Karaiskakio Foundation, Clinical Genetics Clinic, Strovolos, Cyprus; 2Karaiskakio Foundation, Molecular Diagnostics Laboratory, Strovolos, Cyprus; 3Makario Children’s Hospital, Clinical Genetics Clinic, Strovolos, Cyprus; 4Karaiskakio Foundation, Molecular Hematology - Oncology Department, Strovolos, Cyprus

Background/Objectives: Neurofibromatosis type 1 is one of the most common inherited conditions with autosomal dominant inheritance and prevalence of at least 1:3000 worldwide. The clinical picture varies significantly among affected individuals and is characterised by café au lait spots, axillary or inguinal freckling, iris Lisch nodules and multiple cutaneous neurofibromas. NF1 gene is one of the largest genes in the human genome and has among the highest mutation rates with almost half of affected individuals harbouring de novo mutations.

Methods: A 13-year old boy was referred with cafe au lait spots, freckles and macrocephaly and the suspected clinical diagnosis of Neurofibromatosis type 1. His father and brother were also affected, and mother was healthy. NGS sequencing was performed with Agilent’s Exome V8 and data analysed using Franklin software. Confirmatory testing was performed with Sanger sequencing.

Results: A known NF1 c.4537C>T p.Arg1513Ter pathogenic variant was detected in the peripheral blood of the proband. Interestingly, the affected father and brother of the proband carried a different pathogenic NF1 variant c.2326-2A > G, which was absent from the proband.

Conclusion: We report a rare case of a family presenting with Neurofibromatosis in which two different NF1 variants presented in three affected family members, The presence of independent variants in the same family has been reported before and is explained by the high mutation rate of this large gene. This case further emphasizes the role of comprehensive gene investigation in families with multiple affected family members.

Grants: No grants were received for this study

Conflict of Interest: None declared

EP23.019 Are secondary findings actionable? Availability of guidelines for 38 diseases associated with 81 genes recommended for reporting of secondary findings in clinical exome and genome sequencing.

Alisa Förster 1, Clara Köppen1, Gunnar Schmidt1, Bernd Auber1

1, Department of Human Genetics, Hannover Medical School, Hannover, Germany

Background/Objectives: Define the number of specific guidelines addressing clinical actionability of secondary findings in clinical exome/genome sequencing regarding the recommendations of the ACMG Secondary Findings list (v3.2), including 81 medically actionable genes associated with 38 diseases. The identification of secondary findings variants in exome or genome sequencing can be medically actionable, if for example additional surveillance can be offered to carriers.

Methods: Literature review including guidelines and articles that provide recommendations for diseases associated with ACMG-defined actionable genes. Applicable guidelines were identified by using the search function of the Arbeitsgemeinschaft der Wissenschaftlichen Medizinischen Fachgesellschaften (AWMF) database, the National Comprehensive Cancer Network webpage, the medical literature database PubMed, the webpage of the European Society of Cardiology (ESC), and the Orphanet portal for rare diseases.

Results: Internationally, guidelines for 37 of the 38 diseases were identified. In Germany, we identified currently valid guidelines for 17 out of 38 diseases. For 6 out of 16 cancer diseases, guidelines published by the AWMF were available. For diseases of inborn errors of metabolism and miscellaneous phenotypes, we detected 11 German guidelines. International guidelines issue specific treatment recommendations for healthy persons at risk for 7 diseases (breast, ovarian, and colorectal cancer, TTR-related amyloidosis, hypercholesterolemia, MEN 1, MEN 2, German guidelines only for the three cancer diseases.

Conclusion: While recommendations are published for almost all diseases addressed by ACMG’s secondary findings gene list, the specific situation of asymptomatic carriers is rarely addressed in guidelines.

Grants:-

Conflict of Interest: None declared

EP23.020 Medical students opinion on the importance of omics study in research and medicine

Rodica Dragotoiu 1

1University of Medicine and Pharmacy Carol Davila, Pediatrics - Clinical Genetics, Bucuresti, Romania

Objective: The aim of this study was to view the opinion of first year medical students on the importance of different types of omics in the medical field and research.

Methods: Between October 2023 and February 2024 during their one term training in clinical genetics first year students of the Carol Davila Medical and Pharmacy University attended weekly workshops and lectures. At the end of their training in medical genetics 99 first year students answered an anonymous questionnaire. They were asked to answer 7 questions with either one or multiple answers specifying which type of omics appeared to them as most interesting, or most important for medicine, or more useful for investigating and diagnosing a disease, or more useful in research. The answers they could choose from were firstly: genomics, transcriptomics, proteomics, lipidomics, glycomics, metabolomics and multiomics; and secondly: pharmacogenomics, oncogenomics, nutrigenomics, metagenomics, cardiogenomics, neurogenomics, epigenomics, prenatal genomics, reproductive genomics, or neither one.

Results:, 44,4% of students chose genomics as the most interesting type of omics and also 36.4% of them chose it as most important for medicine. Transcriptomics came as secondly interesting (16.2% of students), while multiomics (22.2% of students) as secondly important for medicine. 55 students chose neurogenomics as the most interesting and 46 of them chose oncogenomics as well as neurogenomics as the most important for medicine.

Conclusion: Possibly biased by the fact that the questionnaire was given at the end of their training in medical genetics, a follow-up could show if students maintain the same opinions over the years.

Conflict of Interest: None declared

EP23.021 ERN-ITHACA: European Reference Network on congenital malformations and rare neurodevelopmental disabilities

Marianne Le Dref 1, Alain Verloes1, Anne Hugon1, Nicholas Szeto1, Klea Vyshka1

1Robert Debré Hospital, Clinical Genetics

Consortium: ERN-ITHACA Consortium

Background/Objectives: ERN-ITHACA is a coordinated network of more than 70 clinical genetics departments across EU academic hospitals. ITHACA stands for Intellectual disability, TeleHealth, Autism and Congenital Anomalies, and echoes the diagnostic odyssey experienced by so many patients with developmental anomalies

Methods: Our network brings together experts in rare multiple congenital anomalies and rare neurodevelopmental disorders - the latter field mainly covering intellectual disability and autism spectrum disorder. ITHACA’s field of expertise covers the clinical and genetic diagnosis ofdevelopmental anomalies, their prenatal diagnosis and fetal pathology, and the coordination of their multidisciplinary treatment.

Results: Through our different dedicated task forces that gather researchers, clinicians and patient representatives, the ERN-ITHACA produces European guidelines (Phelan McDermid syndrome, Cornelia de Lange syndrome, Beckwith–Wiedemann syndrome, among others), facilitates collaborative research by disseminating calls for collaborative clinical studies to a network of 600 researchers or clinicians specialised into developmental disorders, supports the develoment a European patient registry (ILIAD), organises webinars, congresses, workshops, and more.

Conclusion: Overall, ERN-ITHACA seeks to improve the diagnosis, care and cure of patients with rare developmental anomalies at the EU-level.

Grants: EU4 Health Programme (EU4H), Grant Agreement nr. 101085231

Conflict of Interest: None declared

EP23.022 Cytogenetics: The Lost Art Reclaimed in Genomic Diagnosis

Vijayalakshmi Salem Ramakumaran 1, Neeta Lakhani1, emily craft1, Pradeep Vasudevan1

1University Hospitals of Leicester, LNR Genomics Medicine Service, Leicester, United Kingdom

Title: Cytogenetics: The Lost Art Reclaimed in Genomic Diagnosis

Author: Vijayalakshmi Ramakumaran, Neeta Lakhani, Emily Craft, Pradeep Vasudevan

Cytogenetics, once overshadowed by the advent of genome sequencing, is experiencing a renaissance in the realm of genetic diagnosis. We highlight the indispensable role of cytogenetics in resolving diagnoses where sequencing alone falls short. Despite the remarkable advancements in sequencing technologies, we present a case series emphasizing the role of cytogenetics in the genomics era for diagnosis of monogenic conditions from a large teaching hospital in the UK.

We describe a case series of monogenic conditions with copy number variants diagnosed with conventional cytogenetic methods and microarray techniques. These include both copy number gains and losses as well as chromosomal rearrangements. The monogenic conditions include the Dystrophinopathies, Neurofibromatosis, Ectrodactyly, Congenital aural atresia, concurrent Tuberous sclerosis, and Polycystic kidney disease. We also compare the phenotype in such cases compared to the phenotype with sequence variants and specific counselling aspects. Furthermore, we delve into the complexities surrounding contiguous gene deletions, CNV/sequence change phenotypic similarities, and translocations.

These cases underscore the critical nature of considering cytogenetics as a complementary tool in the diagnostic armamentarium. These instances serve as poignant reminders of the intricate interplay between genomic alterations and their phenotypic manifestations. By recognizing the nuances and synergies between cytogenetics and sequencing technologies, clinicians can navigate diagnostic challenges more effectively and provide patients with accurate and timely diagnoses. In conclusion, while genome sequencing has revolutionized genetic diagnostics, the art of cytogenetics remains indispensable in unravelling the complexities of the human genome.

Conflict of Interest: None declared

EP23.023 Genetic counsellors need to know - Fetal sex chromosome aneuploidy detection by NIPT

LAI-PING LISA SIU 1, Min, Linda Ou1, Wing-Yiu, Yoyo Chu1, Stephanie Ho1, Wing-Yan, Sharon To1, Sze-Wing, Shirley Cheng1, Ho-Ming Luk1, Ivan Fai-Man Lo1

1Hong Kong Children’s Hospital, Clinical Genetics Service Unit, Hong Kong

Background/Objectives: NIPT has been adopted as a prenatal screening test for fetal chromosomal aneuploidy. The positive predictive value (PPV) for trisomy 21 is 91% while PPV for fetal sex chromosome aneuploidy range from 26% for monosomy X and 86% for XXY. Parents who receive an abnormal NIPT report are faced with difficult decisions on choices of continuation or termination of pregnancy.

Methods: We retrieved and systematically reviewed the records of prenatal cases referred to a genetic counselling clinic over a 19-month period since May-2022 because of abnormal NIPT findings suggestive of fetal sex chromosome aneuploidy.

Results: A total of 100 cases have been reviewed, monosomy X, XXX, XXY and XYY have been found in 3, 19, 25 and 20 cases respectively. There were 13 cases with mosaic monosomy X, XXY or XYY; 8 cases with segmental gain / loss of chromosome X in XX female; 6 cases with mosaic X/XY or XX with Y; 6 cases with indeterminate X/Y.

Conclusion: Most cases of fetal sex chromosome aneuploidy are phenotypically normal at birth and some may go unnoticed till they experience reproductive issues. Only a few of these cases require immediate clinical intervention. Genetic counsellors have a role to provide informed choices and respect reproductive autonomy. Moreover, comprehensive pre-test counselling can better prepare the consultand for unexpected findings and reduce the anxiety while making decision on termination of pregnancy. Through appropriate genetic counselling, we hope that couples can be better prepared for the best option in their family.

Grants: All authors have no grant to disclose.

Conflict of Interest: LAI-PING LISA SIU FULL time, Min, Linda Ou FULL time, Wing-Yiu, Yoyo Chu FULL time, Stephanie Ho FULL time, Wing-Yan, Sharon To FULL time, Sze-Wing, Shirley Cheng FULL time, Ho-Ming Luk FULL time, Ivan Fai-Man Lo FULL time

EP23.024 Dysmorphology handbook: from features to syndromes and vice versa

Nicholas Szeto 1, Marion Gerard2, Yves Gillerot3, jonathan berg4, Maurizio Genuardi5, Martin Krahn6, Ute Moog7, Edward S. Tobias8, Peter Turnpenny9, Johannes Zschocke10, Dorica Dan11, Sofia Douzgou HOUGE12, Marianne Le Dref1, Laurence Faivre13, Raoul Hennekam14, Anne Hugon1, Jean-Marie Jouannic15, Tjitske Kleefstra16, David Koolen16, Giovanni Mosiello17, Alessandra Renieri18, Marco Tartaglia17, Zeynep TÜMER19, Birute Tumiene20, Agnies van Eeghen14, Lisenka Vissers16, Klea Vyshka1, Dagmar Wieczorek21, Giuseppe Zampino5, Christiane Zweier22, Alain Verloes1

1Hospital Robert Debré Ap-Hp, Department of Clinical Genetics, Paris, France; 2University Hospital Center of Caen, Department of Clinical Genetics, Caen, France; 3Institute Pathology And De Génétique, Department of Clinical Genetics, Charleroi, Belgium; 4University of Dundee, Department of Clinical Genetics, Dundee, United Kingdom; 5Università Cattolica del Sacro Cuore, Department of Clinical Genetics, Roma, Italy; 6Aix-Marseille University, Department of Clinical Genetics, Marseille, France; 7Heidelberg University, Department of Clinical Genetics, Heidelberg, Germany; 8University of Glasgow, Department of Clinical Genetics, Glasgow, United Kingdom; 9Royal Devon and Exeter Hospital, Department of Clinical Genetics, Exeter, United Kingdom; 10University of Innsbruck, Department of Clinical Genetics, Innsbruck, Austria; 11EURORDIS - Rare Diseases Europe, Bruxelles, Belgium; 12Helse Bergen, Department of Clinical Genetics, Bergen, Norway; 13Chu Dijon, Department of Clinical Genetics, Dijon, France; 14Amsterdam UMC, locatie AMC, Department of Clinical Genetics, Amsterdam, Netherlands; 15Hospital Armand Trousseau Ap-Hp, Department of Fetal medecine, Paris, France; 16Radboud University Medical Center, Department of Clinical Genetics, Nijmegen, Netherlands; 17Bambino Gesù Pediatric Hospital, Department of Clinical Genetics, Roma, Italy; 18Università di Siena - Polo Scientifico di San Miniato, Department of Clinical Genetics, Siena, Italy; 19Copenhagen University, Department of Clinical Genetics, København, Denmark; 20Vilnius university hospital Santaros klinikos, Department of Clinical Genetics, Vilnius, Lithuania; 21University Hospital Düsseldorf, Department of Clinical Genetics, Düsseldorf, Germany; 22University of Bern, Department of Clinical Genetics, Bern, Switzerland

Consortium: ERN-ITHACA Consortium

Background/Objectives: As a complementary project to the free APOGeE medical genetics textbook, the Dysmorphology handbook presents short summaries of 148 dysmorphic features conditions with illustrations divided into 9 categories: skull, eye, nose, mouth, ear, jaw, thorax, skin, extremities. In the second section, 61 syndromes are presented with their dysmorphic features. It aims to provide cross-referencing between features and syndromes, all in one book.

Methods: The text is a translation of the French handbook by Bouhanna and Gérard (2019), whilst the photos were provided by Gillerot.

Results: The illustrated text will be uploaded to the APOGeE moodle as a separate course. The Dysmorphology handbook is freely accessible without sign-up/in.

Conclusion: The Dysmorphology handbook will be available from June 2024 onwards, alongside the pre-release version of the main APOGeE textbook. The handbook may be used as a standalone guide to dysmorphisms or a companion to the main textbook.

Grants: EU4Health Grant Agreement nr. 101085231

Conflict of Interest: None declared

EP23.025 Improving patient care in rare and genetic syndromes with epilepsy: The role of the Patient Journey Framework in the Epi-ID Partnership", a collaborative approach on common syndromes

Anne Hugon 1;2, Dorica Dan3, Isabella Brambilla4, Vedrana Bibic5

1France, Assistance Publique-Hôpitaux de Paris - Université de Paris, Dept. of Genetics, ERN ITHACA, Paris, France; 2ERN ITHACA Consortium - Patient Advisory Board ERN ITHACA; 3ePAG Patient Advisory Board ERN ITHACA Chair; 4ePAG Patient Group ERN EpiCARE Coordinator; 5ePAG Patient Group ERN EpiCARE

Consortium: ERN ITHACA Consortium - WG Patient Advisory Board

Background/objectives: This study explores the transformative Patient Journey Framework within the Epi-ID Partnership projects of ePags from ERN EpiCARE and ITHACA. It focuses on improving the patient experience across the healthcare continuum. This approach goes beyond clinical pathways to include the physical, emotional, and psychological aspects of the patient experience, thereby improving healthcare delivery. The Patient Journey framework remains a key element, encompassing the entire healthcare continuum from symptom onset to post-treatment care from a family vision. For Rett syndrome, we have already developed patient journey documents and are in the process of developing fact sheets on Rett syndrome and Fragile X.

Methods: ERN ITHACA integrates patient representatives’ insights into over 5000 rare genetic and complex syndromes through syndrome-specific ‘patient pathways’, promoting a patient-centred approach. This integration facilitates the development of a standard of care through clinical pathways that address the holistic needs of patient communities.

Results: The project fosters collaborative efforts with clinicians and the European Federation, to create valuable resources for patient communication and understanding. This include the development of Patient Leaflets and Journeys for common needs syndromes, providing comprehensive information in a unique perspective

Conclusion: The importance of the Patient Journey Framework in the Epi-ID partnership project, highlighting its role in improving patient engagement, treatment outcomes, and overall satisfaction. The ongoing work on Rett Syndrome and Fragile X showcases the framework’s effectiveness in developing informative resources for better patient care and communication be developed in the future.

Grants: ERN-ITHACA, EU4Health Grant Agreement nr. 101085231

Conflict of Interest: None declared

EP23.026 Independent research projects in human and medical genetics with a final report and oral presentation at the Tbilisi State Medical University

Eka Kvaratskhelia 1;2, Sandro Surmava1, Tinatin Tkemaladze2, Nona Janikashvili3, Eka Ekaladze4, Elene Abzianidze1;5

1Tbilisi State Medical University, Department of Molecular and Medical Genetics, Tbilisi, Georgia; 2Tbilisi State Medical University, V. Bakhutashvili Institute of Medical Boitechnology, Tbilisi, Georgia; 3Tbilisi State Medical University, Department of Immunology, Tbilisi, Georgia; 4Tbilisi State Medical University, Department of Biochemistry, Tbilisi, Georgia; 5Ivane Beritashvili Center Of Experimental Biomedicine, Tbilisi, Georgia

Background/Objectives: Independent research projects in human and medical genetics were conducted at the Tbilisi State Medical University (TSMU) Department of Molecular and Medical Genetics by the 11th semester American MD program (USMD) students. The projects were carried out within a frame of the discovery phase course which has been initiated in 2022 by the faculty members of the TSMU American MD Program.

Methods: Within the course, students submitted hypothesis-driven research projects based on their interests and the expertise of the mentor. During the discovery phase, medical students worked on their projects for 5 months. Upon completion of the projects, all students presented their results at a Poster Discovery Conference.

Results: In 2022, in the research laboratory of the Human Genome and Epigenome students were engaged in a project related to single nucleotide polymorphisms in folate metabolism genes and its association with breast cancer risk and progression. The authors presented the poster and oral presentation at the Poster Discovery Conference 2022 and received the first-place award certificate. A scientific manuscript was accepted in a peer-reviewed scientific journal “Clinical Medicine Insights: Oncology” (SAGE Publishing, IF2.2). In 2023, students carried out the project “MTHFD1 & MTRR Single Nucleotide Polymorphisms and their Association with Migraine & Subclinical Hypothyroidism in the Georgian Population”. Results were presented at the Poster Discovery Conference 2023.

Conclusion: Implementation of research projects within the medical curriculum provides a mechanism for students to gain undergraduate science experience. Molecular genetics studies, in addition, acquires them with experimental laboratory skills.

Conflict of Interest: None declared

EP23.027 When misleading signs lead to the correct diagnosis: familial macrocephaly and a de novo PTEN novel variant in a boy with neurodevelopmental delay.

Ludovico Graziani 1, Francesci Piceci Sparascio2, Mario Bengala3, Allessandra Terracciano2, Cinzia Galasso4, Antonio Novelli2, Giuseppe Novelli1;3

1Tor Vergata, Department of Biomedicine and Prevention, Roma, Italy; 2Bambino Gesù Pediatric Hospital, Translational Cytogenomics Research Unit, Roma, Italy; 3Hospital Tor Vergata Roma, Medical Genetics Unit, Roma, Italy; 4Hospital Tor Vergata Roma, Child Neurology and Psychiatry Unit, Roma, Italy

Background/Objectives: Family history evaluation is the backbone of genetic counselling and a major driver of proper clinical management and molecular diagnosis. However, the emergence of new phenotypic expressions, the coexistence of unrelated genetic and non-hereditary diseases, or simple clinical misclassification may lead to erroneous conclusions in risk assessment.

Methods: A 6-year-old boy presented with autistic spectrum disorder (ASD), mild facial traits, and macrocephaly. His phenotype, in addition to the strong familiarity with neurodevelopmental disorders and macrocephaly, which included the father and the uncle, led to a next-generation sequencing analysis of the genes within the ASD/macrocephaly spectrum. Thus molecular analysis detected a de novo PTEN (NM_000314.8: c.226T>A;p.Tyr76Asn) novel heterozygous variant.

Results: PTEN (MIM: 601728) is a ubiquitously expressed tumor suppressor gene that antagonizes the phosphatidylinositol 3-phosphate kinase/AKT signalling and negatively regulates the MAPK pathway. Although germline variants of PTEN are strongly associated with cancer syndromes, they have also been described in a subgroup of patients with ASD and macrocephaly.

Conclusion: The finding confirmed the molecular diagnosis in the boy but further intricated the search for possible genetic modifiers common to the shared phenotypic traits in the family. Further analysis will be needed to understand the role of the identified variant in the development of ASD, and possible oncological correlations, which are absent in our patient to date. This report highlights the complex need to identify relevant clinical signs to conduct a correct genetic diagnosis, disentangling possible familial traits (as macrocephaly could be), phenocopies, and distinct clinical entities.

Conflict of Interest: None declared

EP23.028 Evaluating the Economic Impact on Caregivers of Pediatric Patients with Congenital Heart Disorders in Lithuania

Evelina Vaitėnienė 1, Algirdas Utkus1

1Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Department of Human and Medical Genetics, Vilnius, Lithuania

Background: Congenital heart disorders (CHD), the most prevalent birth anomalies globally, not only affect patients but impose substantial economic burdens on caregivers. These burdens impact employment, well-being, and life quality. Despite CHD’s high prevalence, data quantifying these impacts are lacking.

Methods: This study evaluated the economic burden on caregivers of pediatric CHD patients in Lithuania. Using questionnaires adapted from Landfeldt E., et al. (2018), the research focused on employment changes and informal care burden. Lithuania’s 2023 average income of 1,684.90 EUR served as the economic evaluation reference.

Results: A total of 76 primary caregivers of children aged 5-18 years were included. Data were analyzed based on comorbidity: group 1 comprised solely of patients with CHD (N = 53), group 2 included patients with CHD and additional congenital and chromosomal disorders (N = 23). 11.3% of caregivers in group 1 reported a reduction in work by 18 hours weekly, leading to an average annual income loss of 9,098.46 EUR. In group 2, 43.5% of caregivers reduced work by 29.4 hours weekly, with an average annual income loss of 14,961.9 EUR. Additionally, the study employed the proxy good method to estimate the value of unpaid informal care, which was calculated at 1,928.37 EUR for group 1 and 8,352.9 EUR for group 2.

Conclusion: This study highlights the significant economic burden on caregivers of children with CHD, especially when additional anomalies are present. The findings, consistent with global research, underscore the need for targeted support and further investigation into the socioeconomic impacts of this burden.

Grants: -

Conflict of Interest: None declared

EP23.029 GeneEQUAL Schools: Health and genetic health literacy education for students with intellectual disabilities: Teachers’ and students’ experiences and perspectives

Iva Strnadova1;2, Karen-Maia Jackaman1, Jennifer Hansen1, Julie Loblinzk1, Manjekah Dunn1, Skie Sarfaraz1;2, Sam Hurd1, Joanne Danker1, Michelle Tso1, Sierra Angelina Willow1, Chloe Molnar1, Jackie Boyle3, Jackie Leach Scully1, Elizabeth Palmer1, Bronwyn Terrill 4;5

1UNSW Sydney, Randwick, Australia; 2Self-advocacy sydney; 3Genetics of Learning Disability Service, Waratah, Australia; 4Australian Genomics, Melbourne, Australia; 5The Garvan Institute of Medical Research, Sydney, Australia

Background/Objectives: People with intellectual disability have asked to know more about genetic conditions but need to be empowered with the knowledge and skills to make informed genetic healthcare choices. Despite integration of genetics into the school curriculum, little is known about whether teachers are equipped to engage students with intellectual disability with this content and what students think about their educational experience.

Methods: We conducted and analysed semi-structured interviews and focus groups with 15 teachers in mainstream and special schools who teach genetic content to students with intellectual disability, and 15 students and young people with intellectual disability.

Results: Teachers lack confidence and competence in delivering content to improve students’ health and genetic health literacy, despite this being part of the curriculum. When genetic content is taught, it is not done in an accessible way, and evidenced-based practices are used minimally. Teachers lack the skills, expertise, and resources to educate young people with intellectual disability about genetics, and report concerns about their capability, considering the potential sensitivity of the topic. Students and young people with intellectual disability reported that they were not taught about genetic and health literacy, how to make health choices, and/or how to make life decisions. They felt this will disadvantage them in future.

Conclusion: A key recommendation is that professional learning and accessible, multimodal, online resources are required to build foundational genetic health literacy in schools. Upskilling teachers is necessary to teach students with intellectual disability in a respectful, supportive and trauma-informed way.

Grants: ADA-UNSW 2022-2023.

Conflict of Interest: None declared

EP23.030 Whole exome sequencing for diagnostics in muscular dystrophy: a case report

Zeynep Derin 1, ayşe büşra akalın2, Elif EROĞLU ŞİMŞEK3, İlknur Sunar3, Ibrahim Akalin3

1Istanbul Bilgi University, Genetics and Bioengineering, İstanbul, Türkyie; 2Bahçeşehir Üniversitesi Tıp Fakültesi, Medicine, İstanbul, Türkyie; 3Metagentech Center for Genetic Diseases, İstanbul, Türkyie

Background/Objectives: Muscular dystrophies are a rare and genetically heterogeneous group of inherited diseases that lead to skeletal muscle weakness. Accompanying other organs or clinically relevant symptoms such as contractures is important for differential diagnosis and definite genotype-phenotype correlations.

Methods: Here we report a 20-year-old male patient born to third-degree consanguinity. He was suffering from progressive myopathy. His initial symptoms started at 10 years old with gait disturbances and postural misalignment. Physical examination revealed an inability to independently walk, sit, and stand up, contracture of the arms, thick lower limp, spaced and sparse teeth, short philtrum, winged scapula, kyphosis, and proximal thumb with normal intelligence. The cytogenetic, FISH, CTG repeat expansion, and whole exome sequencing were done.

Results: Chromosomal abnormalities were excluded by cytogenetic analysis and Rubinstein-Taybi was excluded using a CREBBP-specific probe. Whole exome sequencing revealed heterozygous COL6A3:NM_004369.4:c.107C>T;p.A36V variant and heterozygous SYNE2:NM_182914.3:c.12001_12002inv:TG > CA(p.Trp4001Gln) variant.

Conclusion: COL6A3 variation has been linked to autosomal dominant Bethlem myopathy and autosomal dominant Ullrich-congenital muscular dystrophy-1. SYNE2 variant was linked to autosomal dominant Emery-Dreifuss muscular dystrophy-5. According to predominant upper extremity contractures and lower extremity myopathy and progression of the disease. Hence, Bethlem myopathy is the more likely diagnosis of the patient.

Grants:.

Conflict of Interest: None declared

EP24 Ethical, Legal and Psychosocial Aspects in Genetics

EP24.001 Daily life in patients with hereditary transthyretin amyloidosis: a qualitative study

Aina Isabel Gayá Barroso 1;1, Juan Gonzalez Moreno2, Adrián Rodríguez1, Milena Paneque3, Inés Losada López1, Eugenia Cisneros-Barroso1

1Health Research Institute of the Balearic Islands (IdISBa), Son Llàtzer University Hospital, Palma de Mallorca, Spain; 2Health Research Institute of the Balearic Islands (IdISBa), Palma de Mallorca, Spain; 3Center for Predictive and Preventive Genetics, Institute for Molecular and Cell Biology (CGPP-IBMC), Porto, Portugal

Background/Objectives: Variant transthyretin amyloidosis (ATTRv) is a rare genetic disease that negatively influences patients’ wellbeing by affecting multiple organs and tissues. Despite significant investigation into medical and psychosocial interventions, the influence of occupational therapy (OT) on ATTRv patients remains unclear. This study emphasizes the evaluation of daily activities and occupations as a vital measure in the management of patients.

Methods: Following a semi-structured interview, the study enrolled fourteen patients with ATTRv. The occupational therapist collaborated with patients to devise short- and medium-term occupational goals and a work strategy rooted in the Model of Human Occupation (MOHO). Sessions were arranged every week or ten days over the six-month intervention period to track progress and modify guidelines as needed. Activities of daily living and psychological well-being scores were used to evaluate outcomes.

Results: The research suggests that occupational therapy can offer a positive contribution to medical treatments and other psychosocial interventions. Based on the results, twelve patients maintained the same level of performance in their daily activities, two patients displayed a decline, and eight patients demonstrated an improvement in their psychological outcomes. Moreover, it observes that patients who received occupational therapy intervention reported inadequacy of six months and highlights the pivotal function of therapists in stimulating motivation, promoting meaningful activities and enhancing everyday life.

Conclusion: This study underscores the necessity for additional research in this field and the significance of occupational therapy (OT) in managing patients with ATTRv.

Grants: This project has been funded through the Pfizer Independent Research Grants Program ID#64965375.

Conflict of Interest: None declared

EP24.002 Strategy for handling unsolicited findings in syndrome diagnostics with whole genome sequencing – a retrospective study over detected and reported unsolicited findings

Kristina Karrman 1

1Office for Medical Services, Department of Clinical Genetics, Pathology and Molecular Diagnostics, Lund, Sweden

Background: Whole genome sequencing (WGS) enables identification of pathological sequences important for diagnostic purposes. Pathogenic findings that do not answer the primary diagnostic question, unsolicited findings, may be encountered and adds a medical-ethical problem to the diagnostics.

Method: Data of unsolicited findings, identified through cases of trio-WGS performed during 2020-2022, were collected from the department of Clinical Genetics in Region Skåne, Lund, Sweden. These data provided a basis for a retrospective medical-ethical comparison with international guidelines, in particular American College of Medical Genetics and Genomics (ACMG), regarding disclosure of unsolicited findings.

Results: Out of 981 trio-WGS 60 trios were identified with one or more unsolicited findings in the proband and/or the parents. This implies a 6 % frequency of encountering unsolicited findings. In total, 66 unsolicited findings were identified and of which 45 were reported to referring clinician and patient. The proportion of disclosure was 68 %. Of the disclosed variants a majority was on the ACMG list, mainly in cancer predisposition genes.

Conclusion: The international guidelines are important when reporting unsolicited findings locally. The guidelines should be a recommendation and most importantly the context must be considered. This includes the medical, ethical, social, economic, physical and psychological aspects of the patient. However, all agrees on the importance of consensus and thus the discussion and development must continue.

Grants: no grants.

Conflict of Interest: None declared

EP24.005 Perceptions of genomic newborn screening among UK medical students

Lydia Seed 1;2, Anna Scott2;3, Michelle Peter4, Shereen Tadros5, Amanda Pichini6, Cristine Sortica da Costa2;7, Melissa Hill4

1University of Cambridge, School of Clinical Medicine; 2Great Ormond Street Hospital for Children NHS Foundation Trust, Postgraduate Medical Education (PGME) Department, London; 3University of Southampton, School of Medicine; 4Great Ormond Street Hospital for Children NHS Foundation Trust, NHS North Thames Genomic Laboratory Hub, London; 5Great Ormond Street Hospital, North East Thames Regional Genetics Service, London; 6Genomics England, London; 7Great Ormond Street Hospital for Children NHS Foundation Trust, Neonatal Intensive Care Unit, London

Background/Objectives: With the potential to identify a vast number of rare diseases soon after birth, genomic newborn screening (gNBS) could facilitate earlier interventions and improve health outcomes. Designing an effective gNBS programme will involve balancing stakeholders’ opinions and addressing concerns.

Methods: We conducted a nationwide survey of UK medical students in the final two years of study. Participants’ perceptions of gNBS were explored through multiple choice and free-text questions.

Results: In total, 116 medical students across 16 UK medical schools participated. Overall, 45% supported gNBS, with a mean support score of 3.24 out of 5.0, and 55% felt it relevant to their future practice. Almost all agreed that infant-onset (94%) and childhood-onset (86%) diseases and conditions with effective treatments (95%) should be included. Most felt that earlier interventions and personalised care would be the most important benefit of gNBS (70%). Other benefits included earlier diagnoses, diagnosing more patients, and enabling research for new treatments. However, several challenges were highlighted including the risk of discrimination based on genomic information, risk of incidental or uncertain findings, maintaining privacy and security of data, and breaching the child’s future autonomy. Many were also concerned about the parental psychological impact and lack of support available.

Conclusion: As future clinicians, medical students’ opinions are important to inform the design of a novel public health programme. Although some support for gNBS was demonstrated, important ethical, legal and social challenges were raised, emphasising a need for ongoing discussions about the implications of introducing gNBS.

Grants:

Conflict of Interest: None declared

EP24.006 Finding the right time to initiate genetic diagnostics

Sophie Stoesslein 1, Dagmar Wahl1, Konstanze Hörtnagel1

1Medicover Genetics, MVZ Martinsried GmbH, Martinsried, Germany

Background/Objectives: We present the diagnostic journey of our patient initially referred at 4.5 months due to marginal microcephaly. At this time, his development appeared age-appropriate, so we decided not to initiate genetic diagnostics, but arranged a follow-up appointment at the age of 12 months. At the age of 8.5 months, the patient was presented again ahead of schedule with increasing muscular hypotonia, explicit microcephaly (P < 1) and a significant developmental regression. Now, we immediately initiated comprehensive genetic diagnostics.

Methods: This is a retrospective case report from our genetic counselling.

Results: Whole exome sequencing identified a pathogenic de novo variant in the ATRX gene, aligning with X-linked inherited ATRX syndrome, characterized by a variable developmental delay, intellectual disability, muscular hypotonia, microcephaly and characteristic facies. Seizures and a mild form of alpha-thalassemia may also occur, so far not in our patient. Given the lack of curative options in this case, recommendations encompassed vigilant surveillance and symptomatic support.

Conclusion: Finding the right time to initiate comprehensive genetic diagnostics can be challenging especially in infants, as the clinical symptoms may manifest subsequently. This case highlights the need for a multidisciplinary approach in managing genetic disorders and the ongoing challenge of navigating diagnostic uncertainties. Initiating diagnostics too early can lead to uncertainty and disruption of the parent-child-interaction. Thus, it is important to stay in regular contact with other clinical disciplines involved in the case in order not to miss potential therapeutically relevant time windows.

Grant: The legal guardians have granted consent for this report.

Conflict of Interest: None declared

EP24.007 Genome-based approach to improve precision medicine (GENOMED): First results of a patient survey to evaluate their view on genome sequencing

Larissa Mattern 1, Cordula Knopp1, Julia Suh1, Katja Eggermann1, Annette Lischka1, Jeremias Krause1, Robert Meyer1, Nergis Güzel1, Daniela Dey1, Eva Lausberg1, Thomas Eggermann1, Florian Kraft1, Matthias Begemann1, Ingo Kurth1, Miriam Elbracht1

1Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany

Background/Objectives: With the introduction of whole genome sequencing (WGS) in the care of patients with rare diseases, the medical benefits of early diagnosis and the opportunity for more targeted support become obvious. However, what are the patients’ attitudes towards comprehensive genetic diagnostics, consent management, and discussion of (incidental) findings? First results of a questionnaire within the GENOMED study are presented.

Methods: WGS is integrated into a regular outpatient setting. A questionnaire is collected anonymously before and after WGS. Aspects such as patients’ feelings and opinions about WGS, study information, time passed since the onset of the first symptoms, previous medical support, and stress caused by an unclear diagnosis are addressed. Subsequently, significance of the result for future medical treatment and life planning, and overall study satisfaction are evaluated.

Results: The patients’ feedback indicates a high acceptance of WGS and a clear interest in comprehensive genetic diagnostics embedded in counseling. Overall, about half of the patients have been living without a diagnosis for more than five years, feeling (heavily) burdened. Almost all patients expect an improved prognosis and treatment from a molecular diagnosis, but even without a diagnosis, a benefit can be derived after extensive diagnostics.

Conclusion: WGS has high acceptance among patients and their relatives, and they attributed great satisfaction if WGS is performed with personal counseling before and after the analysis. The vast majority perceive the process of informed consent, genetic analysis, and reporting of findings as less complicated.

Grants: The study was supported by the company ILMN.

Conflict of Interest: None declared

EP24.008 Mapping interdisciplinary approaches to genomic medicine through the human and social sciences: a view from the annual meetings held in the context of the France Genomic Medicine 2025 Plan

Anne Cambon-Thomsen 1, Marc Billaud2, Catherine Bourgain3, Sarah Carvallo4, Nicolas Chatauret5, Isabelle Durand-Zaleski6, Christine Lemaitre7, Lucie Morillon7, Laurent Pasquier8, Clément Pimouguet9, Marie Préau10, Emmanuelle Rial-Sebbag1, Dominique Stoppa-Lyonnet11, Frédérique Nowak7

1CERPOP, Inserm and University of Toulouse, Team BIOETHICS, Toulouse; 2Centre Léon Bérard, Lyon, France; 3CERMES 3 Inserm, Paris, France; 4University Lyon 1, Lyon, France; 5Agence de la biomedecine, Paris, France; 6AP-HP, URCEco, Paris, France; 7Inserm, Paris, France; 8CHU Rennes, Medical Genetics, Rennes, France; 9Alliance des maladies rares, Paris; 10Université Lumière Lyon 2, Lyon, France; 11Institut Curie, Paris, France

Background/Objectives: The “Ethics, regulation and society” group of the France Genomic Medicine Plan 2025 (PFMG 2025), initiated the “Human and Social Sciences (HSS)/Genomic Medicine” meetings in 2022 (1.5 days annually). They aim to federate a community of actors about the HSS issues of genomic medicine in France, encourage exchanges between research and actors of genomic medicine, foster collective reflections, encourage relevant HSS research projects.

Methods: The meetings’ programme is prepared by an interdisciplinary committee based both on thematic proposals and on abstracts submitted.

Results: The two first editions (Paris 2022; Dijon 2023), each gathered ~ hundred people from various backgrounds. The 3rd hybrid edition will occur in Toulouse on 2-3 July 2024. An overview of projects involving HSS and relevant for genomic medicine was carried out via posters/oral communications. History of genomic medicine preceded news on the PFMG 2025 and thematic sessions addressed the following through interdisciplinary panels: health policies, legal, economic, professional regulations; massive data in genomic medicine; care/research continuum; patients as actors; challenges of a reference population for genomic medicine; genomic data cycle and their reuse. In 2024 the planned topics are: beyond PFMG 2025, incidental findings (including international comparisons), challenges of genomic medicine in clinical practice, economic issues and equity. Difficulties include creating a synergy between clinical/organisational dimensions and research, agreeing on the scope of genomic medicine, promoting the PFMG 2025 while guaranteeing HSS critical views, foster new HSS research projects.

Conclusion: These meetings allow for rich exchanges thanks to their interdisciplinary dynamics. Increased geneticists’ participation and more international input are desired.

Grants:

Conflict of Interest: Anne Cambon-Thomsen European Commission, For European project GDI, Marc Billaud: None declared, Catherine Bourgain: None declared, Sarah Carvallo: None declared, Nicolas Chatauret: None declared, Isabelle Durand-Zaleski: None declared, Christine Lemaitre: None declared, Lucie Morillon: None declared, Laurent Pasquier: None declared, Clément Pimouguet: None declared, Marie Préau: None declared, Emmanuelle Rial-Sebbag: None declared, Dominique Stoppa-Lyonnet: None declared, Frédérique Nowak: None declared

EP24.009 Debating opt-in/opt-out on genetic/genomic data sharing for scientific research in the EHDS Regulation: an invitation to look behind the tree that hides the forest

Lisa Feriol 1, Gauthier Chassang2, Emmanuelle Rial-Sebbag2

1Inserm and University of Toulouse Paul Sabatier, CERPOP UMR1295, Research Team BIOETHICS ; Ekitia, Not-for-profit data sharing organisation, Toulouse, France; 2Inserm and University of Toulouse Paul Sabatier, CERPOP UMR1295, Research Team BIOETHICS, Genotoul Societal Platform “Ethics and biosciences”, Toulouse (GIS Genotoul), Toulouse, France

Background/Objectives: The European Health Data Space Regulation (EHDSR) under discussion within the European institutions is the occasion to debate once again on the major issues surrounding the options for individuals to (dis)agree to the secondary use of genetic/genomic data. As genetic/genomic data are being particularly sensitive personal data, opt-in consent/opt-out is questioned as the best way to ensure control by individuals over their secondary use for research within the European Research Area. Prolonging the efforts made for equilibrating research needs with data subject’s control along GDPR negotiations which established opt-out as a standard and opt-in as a discretionary choice for Member States, the EHDSR offers an opportunity for the research community and policy-makers to decide about harmonised rules for facilitating genetic/genomic data sharing for secondary uses in research.

Methods: This communication will analyse the main positions of actors such as EU institutions, Scientific Societies, European Patients’ Organisations to compare their principal arguments to favour either opt-in or opt-out positions.

Results: We will highlight the rationale and advantages of opt-out approach to support secondary use of health data for research that has been defended by many genetic research stakeholders for several years and backed by national laws and discuss the challenges posed by opt-out.

Conclusion: Ultimately, we will formulate suggestions for optimal conditions allowing opt-out to be used as a mechanism ensuring efficient involvement of individuals in a trustworthy multi-level data sharing governance system serving research.

Grants: KidneySign, ERA-PerMed JTC2022 programme

Conflict of Interest: Lisa Feriol KidneySign, ERA-PerMed JTC2022 programme, Gauthier Chassang KidneySign, ERA-PerMed JTC2022 programme, Emmanuelle Rial-Sebbag KidneySign, ERA-PerMed JTC2022 programme

EP24.010 Psychological factors in the adaptation of patients undergoing the genetic counseling process for hereditary breast and ovarian cancer

Ines Martin Villalba 1

1Hospital Clínic de Barcelona, Barcelona, Spain

Background: Between 5-10% of all cancers are caused by inherited mutation.People go to genetic counseling units for hereditary cancer seeking advice for their situation. However, the uncertainty and risk of the genetic counseling process can give rise to emotional discomfort that is necessary to know in depth in order to manage it in the most effective way possible.

Objectives: The objective of this work is to analyze the process of genetic counseling for patients carriers of the BRCA1/2 gene responsible for breast cancer and hereditary ovarian cancer (HBOC) syndrom, if the perception of control is a key variable that modulates concern about cancer, psychological well-being and experiencing positive consequences.

Methods: 48 patients make up the sample, of which 24 are affected carriers of HBOC and 25 are healthy carriers and aged between 20 and 71 years at the time of the study.

Results: the results indicate that patients consider that feelings of control over the detection and early treatment of cancer were a positive consequence of the genetic counseling process. Emotional distress is determined in part by Perception of Control, but also by other variables such as perceived risk or level of worry about cancer.

Conclusions: The uncertainty and risk of the genetic counseling process can give rise to emotional discomfort that must be assessed and psychological care offered if necessary. Training in strategies that increase the perception of control are essential.

Conflict of Interest: None declared

EP24.011 Center for treatment and support of people with Huntington disease – an experience from Bulgaria

Iva Ivanova 1;2, Galina Chaneva1, Dragomira Nikolova1;3

1Medical University - Sofia, Faculty of Public Health, Sofia, Bulgaria; 2Bulgarian Huntington Association, Sofia, Bulgaria; 3Medical University - Sofia, Medical Faculty, Department of medical genetics, Sofia, Bulgaria

Background/Objectives: Huntington disease (HD) is a rare genetic disorder which affects 3-5 people per 100 000 worldwide. It is a neurodegenerative disease of the central nervous system characterized by choreic movements, behavioral and psychiatric disturbances. The Bulgarian Huntington Association has worked in the last 9 years aiming to ensure clinical, psychological support and counseling to people with HD and other rare diseases. They are more than 150 000 only in the capital and more than half a million in the whole country.

Methods: Our study focuses mainly on the social needs of HD. We carried out two questionnaires addressed to patients about their requirements and most urgent needs.

Results: The results show that the isolation, the deteriorated psychiatric health, the decrease in the quality of life, the loss of work and the lack of access to inclusive education are the main problems accompanying the diagnose “rare disease”. Only 40% of the patients know their rights as socially secured citizens regarding their recovery and rehabilitation. Other problems are connected with the treatment options. The second approved VMAT2 inhibitor - deutetrabenazine and amatadine, is not registered in Bulgaria and it is too expensive and inaccessible to patients, despite its benefits.

Conclusion: At the moment, Bulgarian patients do not participate in clinical trials of drugs for HD. Due to the fact that we have not adopted the unified European framework for rare diseases, many patients do not have access to orphan drugs, treatment abroad, transplants, which is the bigger problem in our country.

Conflict of Interest: None declared

EP24.012 Care of the patient requires care of the provider

annick REIN ROTHSCHILD 1;2, michael tsur3, Ben Pode-Shakked1;2

1Sheba Medical Center, The Institute for Rare Diseases, Edmond and Lily Safra Children’s Hospital, Ramat Gan, Israel; 2Tel-Aviv University, Faculty of Medicine, Tel-Aviv, Israel; 3Shakla and Tariya multidisciplinary negotiation program, Herzelya, Israel

Care of the Patient Requires Care of the Provider

Background: During the last few years, results of emerging research show that physicians’ stress, fatigue, burnout, depression, or general psychological distress negatively affects patient care. Clinical genetics providers are faced with increased demands not only related the emotionally charged interactions with patients and families, but also to more frequent challenging situations such as the interpretation of uncertain genomic results.

Methods: In a structured survey on the amount of time demands in the practice of medical genetics (published in 2016), participating medical geneticists and genetic counsellors expressed frustration over the workload, the complexity of the profession, lack of time and inadequate compensation. Some of the respondents’ narrative comments reflected the complexity and challenges currently faced by the medical geneticists. How geneticists and patients communicate about and act within these situations has significant implications for clinical care.

Results: Here, we wish to present a review of the literature on the geneticists’ provider wellness, present tools from the negotiation world that could be used to manage and mitigate the emotional burden and uncertainty within the context of a clinician-patient relationship and discuss how these tools could be implemented in the clinic.

Conclusions: There is clearly a need for creative or innovative practice models, such as negotiation practices that might be key for the care of the provider, which is needed for providing better care of the patients and their families.

Conflict of Interest: annick REIN ROTHSCHILD: None declared, michael tsur Adv. Michael Tsur is employed by the Shakla and Tariya Multidisciplinary Negotiation Program, Ben Pode-Shakked: None declared

EP24.014 Should recipient parents have access to gamete donors’ raw genomic data? Ethical and clinical considerations

Shiri Shkedi Rafid 1, roy gilbar2, aviad raz3

1Hadassah Ein Kerem Hospital, Genetics, Jerusalem, Israel; 2Netanya Academic College, Law, Natanya, Israel; 3Ben Gurion University of the Negev, Be’er Sheva, Israel

Introduction: Genomic sequencing yields enormous amounts of data. Access to raw genomic data by patients and research participants raises ethical and practical dilemmas, which have been theoretically and empirically explored, yet concrete guidelines are missing. Consequently, various labs and sequencing projects come up with their own practice. A specific challenging (yet underdiscussed) question is whether gamete donors’ raw-data should be provided to recipient parents, what the advantages and disadvantages are, and what alternatives exist?

Methods: Using a clinical case, this paper aims to explore key ethical and clinical implications of access to gamete donors’ raw genomic data by recipient parents.

Results: Ethical implications include e.g. feasibility of meaningful informed consent of donors for complex genetic testing, sometimes years after donation; privacy; potential conflict between the donor’s and the child’s best interest; access to donors’ raw genomic data as a potential cause for raised costs and commercial interests. Clinical implications involve implementation of alternative types of consent (e.g., dynamic or meta consent) and systems of re-contacting past donors.

Conclusions: With the ever-growing prevalence of diverse families that are created using gamete donation, and a constant increase in age-related infertility, the number of third-party pregnancies utilizing gamete donation are likely to increase. Advanced-genomic-tests are becoming an integral part of pre-conception and prenatal diagnosis. It is therefore important to discuss ethical, legal, and clinical implications of access to raw genomic data also in the context of gamete donation. This may inform guidelines, which will assist clinicians involved in genetic testing and fertility.

Conflict of Interest: None declared

EP24.015 Translational justice for prenatal gene editing: understanding views and impact on patient communities

Kirsten Riggan1, Yvonne Bombard2, Natasha Bonhomme3, Roel Feys4, Heidi Carmen Howard5, McKenna Horstmann6, Karen Meagher1, Marsha Michie6, Kiran Musunuru7, Kelly Ormond 8, Sabina Rubeck6, Jane Yap9, Rosario Isasi4, Megan Allyse9

1Mayo Clinic, Rochester, United States; 2University of Toronto, Toronto, Canada; 3Genetic Alliance, Damascus, United States; 4University of Miami, Miami, United States; 5Lund University, Lund, Sweden; 6Case Western Reserve University, Cleveland, United States; 7University of Pennsylvania, Philadelphia, United States; 8ETH Zürich, Zürich, Switzerland; 9Mayo Clinic, Jacksonville, United States

Background/Objectives: Somatic gene editing during pregnancy holds therapeutic promise to mitigate genetic conditions in a fetus. However, limited attention has been given to the broader translational issues of prenatal gene editing, including its potential impact on patient communities. This study elicits and synthesizes values and priorities across stakeholder groups (patients, clinicians/scientists, policymakers) into a proposed governance pathway for prenatal human gene editing.

Methods: Qualitative thematic analysis of in-depth interviews with ten patient communities across the Family System Genetic Illness framework was performed along with a parallel ethical analysis of the translation of prenatal gene editing for genetic disease.

Results: Preliminary analysis of patient interviews supports the importance of understanding how patient communities conceptualize their condition, whether they view prenatal gene editing as an acceptable therapeutic approach, and concerns about societal impacts. Patients also stressed the importance of ensuring treatments are accessible and appropriately regulated. Ethical analysis suggests that many factors influence successful clinical translation beyond traditional benchmarks of safety and efficacy. Translational challenges include lack of market incentives and research investment into rare sub-variants and conditions, the high economic costs of genetic therapies and associated care, limited partnership with patient communities for alignment with their therapeutic priorities, and longitudinal impacts to the cohesion of patient communities.

Conclusion: Perspectives and values of patient communities, clinicians, and scientists must be integrated into governance structures to address the translational justice challenges posed by prenatal gene editing.

Grants: National Human Genome Research Institute grant R01 HG011461.

Conflict of Interest: None declared

EP24.016 Expanding clinical utility and ending the diagnostic odyssey: the impact of exome/genome sequencing in rare disease

Salma Shickh 1, Viji Venkataramanan1, Katharine Fooks1, Karen MacDonald2, Trevor Seeger2, Taila Hartley3, Francois Bernier2, Kym Boycott3, Deborah Marshall2, Robin Hayeems1;4

1The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, Canada; 2University of Calgary, Calgary, Canada; 3Children’s Hospital of Eastern Ontario (CHEO), Ottawa, Canada; 4Institute of Health Policy, Management and Evaluation, Toronto, Canada

Background/Objectives: Genomic sequencing technologies (GS;exome and genome sequencing) have significantly improved diagnosis of patients with rare diseases. A diagnosis often leads to tailored medical management and ends a diagnostic odyssey, marked by invasive medical investigations and emotional and financial burden for families. However, most evaluations of GS only report diagnostic rate and new recommendations made, potentially underestimating clinical utility.

We provide a comprehensive description of clinical utility for a cohort of patients undergoing GS including all diagnostic investigations and medical management recommendations prompted or averted by GS results.

Methods: We conducted a prospective, observational study of a cohort of patients undergoing GS from seven genetics clinics in two Canadian jurisdictions, Alberta and Ontario.

Clinical utility data were collected from medical records and clinician-reported checklists, and reported using descriptive statistics.

Results: Overall, 718 participants underwent GS, 68.1% (n = 526) presented with intellectual disability, and 55% were male. Mean age at enrollment was 11.4 years (SD:11.96).

Approximately 34% of participants (n = 246) received a molecular diagnosis from GS. GS results triggered 250 diagnostic investigations and prevented 1641 investigations.

Moreover, GS results triggered 1538 management recommendations for 23.8% (n = 171) of participants, including 106 for surveillance, 615 referrals to sub-specialists, and 46 medication or procedure-related recommendations. Three surveillance activities and 61 referrals were discontinued or avoided.

Conclusion: Our findings provide evidence that GS can prevent unnecessary diagnostic investigations and medical interventions. In addition to highlighting cost-savings and reducing burden on patients and families, our results suggest that current evaluations may underestimate the clinical utility of GS, potentially limiting funding and access.

Grants:OGI-147

Conflict of Interest: None declared

EP24.017 Improving access to genetic information of the deceased: new insights from French law

Pauline Rateau 1, Emmanuelle Rial-Sebbag1

1Inserm and University of Toulouse Paul Sabatier, CERPOP UMR1295, Research Team BIOETHICS, Genotoul Societal Platform “Ethics and biosciences”, Toulouse (GIS Genotoul), Toulouse, France

Background/Objectives: The current French legal framework for genetic testing in a medical context is governed by the patient’s written informed consent, as provided for in the Civil Code. The last bioethics law and the decree of 11 September 2023 clarify the legal provisions for carrying out a genetic test on a deceased person on the basis of an opt-out, justified by medical purposes in the interest of family members. This consent waiver favours the circulation of genetic information and challenges the autonomy of the deceased and of their family members. We analyse this new medical framework and the criteria set by the law, which aims to balance the interests of the individual and of the family.

Methods: A review of French legislation and of French literature was carried out.

Results: While the law facilitates access to genetic information by family members through the opt-out procedure, the decree establishes a framework for the study of genetic characteristics of deceased persons, which requires several conditions to be met before the analysis can be performed. The decree provides criteria for assessing the seriousness of the genetic disease, the prevention and care measures that could be offered and conditions relating to the role of family members, thus legitimising this post-mortem framework.

Conclusion: These clarifications still raise legal and ethical questions relating to respect for the wishes of the deceased, the integrity of the human body and genetic data, access to genetic testing for the benefit of family members and professional practice.

Grants: UNESCO Chair on Ethics, Science and Society

Conflict of Interest: None declared

Abstracts from the 57th European Society of Human Genetics (ESHG) Conference: e-Posters (2025)

References

Top Articles
Latest Posts
Recommended Articles
Article information

Author: Gov. Deandrea McKenzie

Last Updated:

Views: 5397

Rating: 4.6 / 5 (46 voted)

Reviews: 85% of readers found this page helpful

Author information

Name: Gov. Deandrea McKenzie

Birthday: 2001-01-17

Address: Suite 769 2454 Marsha Coves, Debbieton, MS 95002

Phone: +813077629322

Job: Real-Estate Executive

Hobby: Archery, Metal detecting, Kitesurfing, Genealogy, Kitesurfing, Calligraphy, Roller skating

Introduction: My name is Gov. Deandrea McKenzie, I am a spotless, clean, glamorous, sparkling, adventurous, nice, brainy person who loves writing and wants to share my knowledge and understanding with you.