Conference Agenda

Overview and details of the sessions of this conference. Please select a date or location to show only sessions at that day or location. Please select a single session for detailed view (with abstracts and downloads if available).

 
 
Session Overview
Session
Tea Break and poster session
Time:
Monday, 12/June/2023:
3:30pm - 4:30pm

Location: Bologna Congress Center

Address: Piazza della Costituzione, 4/a, Bologna (BO), Italy

Session topics:
- Clinical 1: from new genes to old and novel phenotypes
- New technological developments and OMICS - Modelling pathogenic mechanisms: OXPHOS, metabolic rewiring and tissue specificity

Show help for 'Increase or decrease the abstract text size'
Presentations

Recessive MECR pathogenic variants cause a LHON-like optic neuropathy

Claudio Fiorini1, Andrea Degiorgi2, Maria Lucia Cascavilla3, Concetta Valentina Tropeano1, Chiara La Morgia1,4, Marco Battista3, Danara Ormanbekova1, Flavia Palombo1, Michele Carbonelli4, Francesco Bandello3, Valerio Carelli1,4, Alessandra Maresca1, Piero Barboni3, Enrico Baruffini2, Leonardo Caporali4

1IRCCS Istituto delle Scienze Neurologiche di Bologna, Italy; 2Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy; 3Department of Ophthalmology, University Vita-Salute, IRCCS Ospedale San Raffaele, Milan, Italy; 4Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Italy



Variants in ATP5F1B are associated with dominantly inherited dystonia

Alessia Nasca1, Niccolò Mencacci2, Federica Invernizzi1, Michael Zech3, Andrea Legati1, Giovanna Zorzi1, Holger Prokisch3, Steven Lubbe2, Barbara Garavaglia1, Daniele Ghezzi1,4

1Fondazione IRCCS Istituto Neurologico Besta, Milan, Italy; 2Northwestern University, Feinberg School of Medicine, Chicago, USA; 3Helmholtz Zentrum München, Technical University of Munich, Munich, Germany; 4Università di Milano, Milan, Italy



Toward clinical implementation of quantitative proteomics in the detection of mitochondrial disorders

Daniella H Hock1, Liana N Semcesen1, Nikeisha J Caruana1,2, Sumudu Amarasekera1,3, Teresa Zhao1,3,4, Ann E Frazier1,3, Shabnam Bakhshalizadeh1,3, Megan Ball1,3,4, Tegan Stait3,4, Jessie Jacobsen5,6, Emma Glamuzina5,6, Bryony Ryder5,6, Johan L K Van Hove7, Elena Tucker1,3, Andrew Sinclair1,3,4, Cas Simons8,9, Alison G Compton1,3,4, John Christodoulou1,3,4, David R Thorburn1,3,4, David A Stroud1,3,4

1University of Melbourne, Parkville, Australia; 2Victoria University, Footscray, Australia; 3Murdoch Children’s Research Institute, Melbourne, Australia; 4Victorian Clinical Genetics Services, Melbourne, Australia; 5National Metabolic Service Auckland City Hospital, Auckland, New Zealand; 6Starship Children's Hospital, Auckland, New Zealand; 7University of Colorado, Aurora, United States of America; 8Centre for Population Genomics, Melbourne, Australia; 9Garvan Institute of Medical Research, Sydney, Australia



A DNM2-related myopathy mimicking a primary mitochondrial disorder

Ignazio Giuseppe Arena1, Rosalba Carrozzo2, Mattia Porcino1, Alba Migliorato3, Carmelo Rodolico1, Olimpia Musumeci1

1Unit of Neurology and Neuromuscular Disorders, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy.; 2Department of Neurosciences, Unit of Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; 3Unit of Neurology and Neuromuscular Disorders, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy.



Assessing the association of mitochondrial DNA genes with Primary Mitochondrial Disease using the ClinGen Clinical Validity Framework

Elizabeth M. McCormick1, James T. Peterson1, Julie P. Taylor2, Krista Bluske2, Amanda R. Clause2, Anjana Chandrasekhar2, Josh Lowry2, Alison J. Coffey2, Xiaowu Gai3,4, Marni J. Falk1,5, Zarazuela Zolkipli-Cunningham1,5, Shamima Rahman6

1Mitochondrial Medicine Frontier Program, Division of Human Genetics, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA; 2Illumina Laboratory Services, Illumina Inc., San Diego, CA; 3Center for Personalized Medicine, Department of Pathology & Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA; 4Keck School of Medicine, University of Southern California, Los Angeles, CA; 5Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; 6Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, United Kingdom



Functional characterisation of the m.8424T>C MT-ATP8 variant using quantitative proteomics

Liana Semcesen1, Nikeisha Caruana1,2, Daniella Hock1, Alison Compton1,3,4, Zornitza Stark1,4,5, John Christodoulou1,3,4, David Thorburn1,3,4, David Stoud1,3,4

1University of Melbourne, Parkville, Australia; 2Victoria University, Footscray, Australia; 3Murdoch Children’s Research Institute, Melbourne, Australia; 4Victorian Clinical Genetics Services, Melbourne, Australia; 5Australian Genomics, Melbourne, Australia



COX18 variants cause isolated Complex IV deficiency associated with neonatal hypertrophic cardiomyopathy, myopathy and axonal sensory neuropathy

Dario Ronchi1, Megi Meneri1,2, Francesca Magri2, Francesca Menni2, Manuela Garbellini2, Maria Francesca Bedeschi2, Robertino Dilena2, Valeria Cecchetti2, Irene Picciolli2, Francesca Furlan2, Valentina Polimeni2, Sabrina Salani2, Laura Pezzoli2, Francesco Fortunato1, Matteo Bellini3, Sara Antognozzi2, Daniela Piga2, Michela Ripolone2, Simona Zanotti2, Laura Napoli2, Patrizia Ciscato2, Monica Sciacco2, Giovanna Mangili3, Fabio Mosca2, Stefania Corti1,2, Maria Iascone3, Giacomo Pietro Comi1,2

1Dino Ferrari Center, University of Milan, Italy; 2IRCCS Cà Granda Ospedale Maggiore Policlinico Milan, Italy; 3ASST Papa Giovanni XXIII, Bergamo, Italy



Severe mitochondrial encephalomyopathy caused by de novo variants in OPA1

Daria Diodato1, Michela Di Nottia2, Alessandra Torraco2, Teresa Rizza2, Claudia Nesti3, Enrico Baruffini3, Diego Martinelli4, Margherita Verardo1, Adele D'Amico1, Filippo Maria Santorelli5, Enrico Bertini1, Rosalba Carrozzo2

1Muscular and Neurodegenerative Disorders Unit, Children Hospital Bambino Gesù; 2Cellular biology and mitochondrial diseases diagnostics, Children Hospital Bambino Gesù; 3Department of Chemistry Life Sciences and Environmental Sustainability, University of Parma; 4Metabolism Division, Children Hospital Bambino Gesù, Rome; 5Molecular Medicine, IRCCS Stella Maris, Pisa



Bi-allelic TEFM variants are associated with a treatable mitochondrial myopathy

Alessandra Torraco1, Guido Primiano2,3, Anastasia Altobelli1, Teresa Rizza1, Michela Di Nottia1, Rosalba Carrozzo1, Serenella Servidei2,3

1Unit of Cellular Biology and Diagnosis of Mitochondrial Disease, Bambino Gesù Children’s Hospital, IRCCS, Rome Italy.; 2Dipartimento di Neuroscienze, Organi di Senso e Torace, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.; 3Dipartimento Di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy.



TOMM40L as a new causative gene for autosomal recessive mitochondrial disease.

Francisco Javier Cotrina Vinagre1, María Elena Rodríguez García1, Lucía Del Pozo Filiú1, Elena Martín Hernández2,3, Francisco Martínez Azorín1,3

1Laboratorio de Enfermedades Mitocondriales. Instituto de Investigación Hospital 12 de Octubre (i+12), E-28041 Madrid, Spain.; 2Unidad Pediátrica de Enfermedades Raras, Enfermedades Mitocondriales y Metabólicas Hereditarias, Hospital 12 de Octubre, E-28041, Madrid, Spain.; 3Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), U723, E-28041 Madrid, Spain.



A primary cardiological phenotype caused by an inherited mtDNA single deletion: a case report from an Italian pedigree

Piervito Lopriore1, Christiane Neuhofer2,3, Vincenzo Montano1, Adriana Meli1, Annalisa Lo Gerfo4, Giulia Cecchi4, Maria Adelaide Caligo4, Riccardo Berutti2,3, Robert Kopajtich2,3, Gabriele Siciliano1, Holger Prokisch2,3, Michelangelo Mancuso1

1Department of Clinical and Experimental Medicine, Neurological Institute, University of Pisa, Pisa, Italy; 2Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany; 3Institute of Neurogenomics, Computational Health Center, Helmholtz Zentrum München, Neuherberg, Germany; 4Laboratory of Molecular Genetics, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy



Genetic characterization of a large cohort of Spanish patients with TK2 deficiency. A founder effect of two TK2 variants partially contributes to a higher prevalence of the disorder in Spain.

Cristina Domínguez-González1,2, Pablo Serrano-Lorenzo1,2, Alberto Blázquez1,2, Juan Francisco Quesada-Espinosa1, Jorge Amigo Lechuga3, Pablo Mínguez2,4, Carmen Ayuso2,4, Elena García-Arumí5, Nuria Muelas2,6, Teresa Jaijo6, Andrés Nascimento7, Beatriz Galán-Rodriguez8, Carmen Paradas8,9, Joaquín Arenas1,2, Ángel Carracedo2,3, Ramon Martí2,5, Miguel A. Martin1,2

1Hospital Universitario 12 de Octubre, imas12 Research Institute, Madrid, Spain; 2Spanish Network for Biomedical Research in Rare Diseases (CIBERER); 3Fundación Galega de Medicina Xenómica, Santiago de Compostela, Spain; 4Instituto de Investigación Sanitaria, Hospital Universitario FundaciónJiménez Díaz, Madrid, Spain; 5Research Group on Neuromuscular and Mitochondrial Diseases, Vall d’Hebron Research Institute, Autonomous University of Barcelona, Barcelona; 6Hospital Universitari I Politècnic La Fe, Neuromuscular and Ataxias Research Group, Instituto de Investigación Sanitaria La Fe, Valencia; 7Sant Joan de Déu Research Institute, Sant Joan de Déu Hospital, Barcelona, Spain.; 8Instituto de Biomedicina de Sevilla, Hospital U. Virgen del Rocío, Sevilla, Spain.; 9Center for Biomedical Network Research on Neurodegenerative Disorders (CIBERNED)



HSD17B10 interacts with CBR4 to form human mitochondrial 3-ketoacyl-acyl carrier protein reductase 2 (KAR2) in the mitochondrial fatty acid synthesis pathway

Ali Julfiker Md Masud, Guangyu Jiang, Kaija J Autio, J Kalervo Hiltunen, Alexander J Kastaniotis

Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland



Novel atypical variants causing pyruvate dehydrogenase complex deficiency

Helene Bruhn1,2, Karin Naess1,2, Sofia Ygberg2,3, Nicole Lesko2,4, Rolf Wibom1,2, Anna Wedell2,4, Anna Wredenberg1,2

1Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden; 2Centre of inherited metabolic diseases, Karolinska University Hospital, Stockholm, Sweden; 3Neuropediatric Unit, Dept of Women’s, and Children's Health, Karolinska Institutet, Stockholm, Sweden; 4Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden



Novel genetic discoveries detected using diagnostic OMICSs in patients suspected to suffer from multiple acyl-CoA dehydrogenation deficiency

Signe Mosegaard1,11, Mirjana Gusic2,3, Yasuhiko Ago4, Xiao Yue4, Vincente Yépez3,5, Julien Gagneur3,5, Robert Kopajtich3, Dmitrii Smirnov3, Sarah Stenton2, Robert Barski6, Mark Sharrard7, Stanley H. Korman8, Jolanta Sykut-Cegielska9, Anibh M. Das10, Helle H. Nygaard11, Margrethe Kjeldsen11, Yusof Rahman12, Skadi Beblo13, Mirian C. H. Janssen14, Eva Morava15, Leo A. J. Kluijtmans16, Alexander Asamoah17, Emma Buckley18, Isaque Qureshi18, Godfrey T. Gillett19, Ole H. Larsen20, Niels Gregersen11, Toshiyuki Fukao4,21, Hideo Sasai4, Simon Olpin22, Holger Prokisch2,3, Rikke K. J. Olsen11

1Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands; 2Institute of Neurogenomics, Helmholtz Zentrum München, Germany; 3Institute of Human Genetics, School of Medicine, Technical University Munich, München, Germany.; 4Department of Pediatrics, Graduate School of Medicine, Gifu University Hospital, Gifu, Japan.; 5Department of Informatics, Technical University of Munich, Garching, Germany.; 6Department of Biochemical Genetics, St James's University Hospital, Leeds, UK.; 7Department of Pediatrics, Sheffield Children's Hospital, Sheffield, UK.; 8Department of Pediatrics, Shaare Zedek Medical Center, Jerusalem, Israel.; 9Department of Inborn Errors of Metabolism and Paediatrics, The Institute of Mother and Child, Warsaw, Poland.; 10Department of Pediatrics, Hannover Medical School, Hannover, Germany.; 11Research Unit for Molecular Medicine, Aarhus University and Aarhus University Hospital, Aarhus, Denmark.; 12Center for Inherited Metabolic Disorders, Guy’s & St Thomas’ Hospital NHS Foundation Trust, London, UK.; 13University Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany.; 14Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.; 15Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota, USA; 16Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands.; 17UofL Physicians Novak Center for Children's Health, Louisville, USA.; 18Nottingham Children’s Hospital, Nottingham University Hospitals NHS Trust, Queen's Medical Centre, Nottingham, UK; 19Sheffield Teaching Hospitals NHS Trust, University of Sheffield, Sheffield, UK; 20Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark.; 21Clinical Genetics Center, Gifu University Hospital, Gifu, Japan.; 22Department of Clinical Chemistry, Sheffield Children’s Hospital, Sheffield, UK.



The Australian genomics mitochondrial flagship: a national program delivering mitochondrial diagnoses

David Thorburn1,2,3, Naomi Baker2,3, Shanti Balasubramaniam4, Stephanie Best5, Kaustuv Bhattacharya4, Kristen Boggs4, Sarah Borrie6, Drago Bratkovic6, Jeffrey Braithwaite5, Alessandra Bray4, Jo Burke7, David Coman8,9, Alison Compton1,2, Mark Cowley10, Martin Delatycki2,3, Michelle de Silva1,2,3, Carolyn Ellaway4, Michael Fahey11, Janice Fletcher12, Leah Frajman1,2, Ann Frazier1,2, Velimer Gayeskiv9, Roula Ghaoui12, Himanshu Goel13, Ilias Goranitis2, Daniella Hock2, Denise Howting14, Matilda Jackson6, Maina Kava15, Sarah King-SMith6, Nicole Lake1,16, Phillipa Lamont15,17, Joy Lee2,18, Janet Long5, Mandi MacShane15, Ellenore Martin4, Jim McGill8, Sean Murray19, Julie Panetta11, Liza Phillips20, Michael Quinn21, Rocio Rius1,2, Michael Ryan22, Nicholas Smith6, David Stroud1,2,3, Michel Tchan23, Melanie Tom21, Matthew Wallis7, Tyson Ware24, AnneMarie Welch1, Christine Wools11, Eunice Wu2, John Christodoulou1,2,3

1Murdoch Children's Research Institute, Melbourne, Australia; 2University of Melbourne, Melbourne, Australia; 3Victorian Clinical Genetics Services, Melbourne,; 4Sydney Children’s Hospitals Network, Westmead, Australia; 5Macquarie University, Sydney, Australia; 6Women’s and Children’s Hospital, Adelaide, Australia; 7Tasmanian Clinical Genetics Service, Hobart, Australia; 8Queensland Children’s Hospital, Brisbane, Australia; 9Wesley Hospital, Brisbane, Australia; 10Garvan Institute, Sydney, Australia; 11Royal Melbourne Hospital, Melbourne, Australia; 12Royal Adelaide Hospital, Adelaide, Australia; 13John Hunter Hospital, Newcastle, Australia; 14Harry Perkins Institute of Medical Research, Perth, Australia; 15Perth Children’s Hospital, Perth, Australia; 16Yale School of Medicine, New Haven, CT, USA; 17Royal Perth Hospital, Perth, Australia; 18Royal Children’s Hospital, Melbourne, Australia; 19Mito Foundation, Sydney, Australia; 20Mater Hospital, Brisbane, Australia; 21Genetic Health Queensland, Brisbane, Australia; 22Monash University, Melbourne, Australia; 23Westmead Hospital, Westmead, Australia; 24Royal Hobart Hospital, Hobart, Australia



A new family with a case of severe early-onset muscle fatigue and a peculiar maternally inherited painful swelling in chewing muscles associated with homoplasmic m.15992A>T mutation in mitochondrial tRNAPro

Irene Bruno1, Elena Ghirigato2, Mirko Baglivo3, Francesca Terenzi2, Nadia Zanetti3, Massimo Zeviani1, Eleonora Lamantea3

1Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, Italy; 2Department of Medicine, Surgery, and Health Sciences, University of Trieste, Trieste, Italy; 3Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.



A novel MT-ATP6 variant associated with complicated ataxia in two unrelated Italian patients: case report and functional studies. 

Daniele Sala1, Silvia Marchet1, Lorenzo Nanetti1, Andrea Legati1, Caterina Mariotti1, Eleonora Lamantea1, Daniele Ghezzi1,2, Alessia Catania1, Costanza Lamperti1

1Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta; 2Department of Pathophysiology and Transplantation (DEPT), University of Milan



Biallelic pathogenic variants of PARS2 cause Developmental and Epileptic Encephalopathy with Spike-and-Wave Activation in Sleep

Laura Licchetta1, Carlotta Stipa1, Raffaella Minardi1, Margherita Santucci1,2, Lucia Di Giorgi2,3, Martina Soldà1, Valerio Carelli1,2, Francesca Bisulli1,2

1IRCCS, Istituto delle Scienze Neurologiche di Bologna, Italy; 2Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy; 3Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Italy.



Novel KARS1 mutation causes early-onset lethal cardiomyopathy

Sara Casalini1, Silvia Buratti1, Chiara Panicucci1, Maria Elena Derchi1, Marco Scaglione1, Claudia Nesti2, Monica Traverso1, Francesca Madia1, Michela Di Nottia3, Chiara Fiorillo1, Valeria Capra1, Filippo Maria Santorelli2, Andrea Moscatelli1, Rosalba Carrozzo3, Claudio Bruno1

1IRCCS Istituto Giannina Gaslini, Genoa; 2IRCCS Fondazione Stella Maris, Calambrone (PI); 3IRCCS Ospedale Bambin Gesù, Rome



The ER-MITO (Emilia Romagna-Mitochondrial) project: prevalence and genetics of Chronic Progressive External Ophthalmoplegia (CPEO) in an Italian region

Maria Lucia Valentino1,2, Leonardo Caporali1, Chiara La Morgia1,2, Flavia Palombo1, Martina Romagnoli1, Cristina Fonti1, Alessandra Maresca1, Rocco Liguori1,2, Corrado Zenesini1, Roberto D'Alessandro1, Valerio Carelli1,2

1IRCCS Institute of Neurological Sciences of Bologna, Italy; 2Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy



UCHL1 missense and loss-of-function variants as an emerging cause of autosomal dominant optic atrophy (ADOA)

Claudio Fiorini1, Giada Capirossi1,2, Eleonora Pizzi1, Federico Sadun3, Maria Lucia Cascavilla4, Chiara La Morgia1,2, Marco Battista4, Piero Barboni4, Danara Ormanbekova1, Valentina Del Dotto2, Flavia Palombo1, Valerio Carelli1,2, Alessandra Maresca1, Leonardo Caporali2

1IRCCS Istituto delle Scienze Neurologiche di Bologna, Italy; 2Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Italy; 3Ospedale Oftalmico Roma, Rome, Italy; 4Department of Ophthalmology, University Vita-Salute, IRCCS Ospedale San Raffaele, Milan, Italy



Mitochondrial dysfunction in patients with early-onset UFM1-linked encephalopathy

Samira Ait El Mkadem - Saadi1,2, Cécile Rouzier1,2, Annabelle Chaussenot1,2, Konstantina Fragaki1,2, Sylvie Bannwarth1,2, Julien Neveu3, Aline Cano3, Brigitte Chabrol3, Véronique Paquis-Flucklinger1,2

1National Centre for Mitochondrial Diseases, Nice Teaching Hospital (CHU de Nice), Department of Medical Genetics, Nice, France; 2Université Côte d'Azur, CHU, Inserm, CNRS, IRCAN, France; 3APHM, La Timone Hospital, Department of Neuropediatrics, Marseille, France



A novel dominant variant in the ISCU gene is associated with mitochondrial myopathy

Joanna Rusecka1,2, Dominika Szczęśniak2,3, Magdalena Kacprzak2, Damian Loska2, Kamila Czerska2, Agnieszka Sobczyńska-Tomaszewska2

1Maria Sklodowska-Curie, Medical Academy in Warsaw, Poland; 2MedGen Medical Center, Warsaw, Poland; 3Institute of Psychiatry and Neurology, Warsaw, Poland



Expanding the spectrum of clinical presentations associated with COA8 pathogenic

Sara Antognozzi1, Magi Meneri1,2, Francesca Magri1, Manuela Garbellini1, Sabrina Salani1, Francesco Fortunato2, Michela Ripolone1, Simona Zanotti1, Patrizia Ciscato1, Monica Sciacco1, Stefania Corti1,2, Giacomo Pietro Comi1,2, Dario Ronchi2

1IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; 2Dino Ferrari Center, University of Milan, Milan, Italy



A novel mitochondrial DNA variant, m.14430A>C, in MT-ND6 as the likely cause of Leigh syndrome with mitochondrial complex I deficiency.

Surita Meldau1,2, Gillian T M Riordan1,3, Sally Ackermann4, Sharika Raga1,3,6, Careni Spencer1,5, George F Van der Watt1,2, Kashief Khan2, Francois H Van der Westhuizen7

1University of Cape Town, Cape Town, South Africa; 2National Health Laboratory Sevices, South Africa; 3Red Cross War Memorial Children's Hospital, Cape Town, South Africa; 4Constantiaberg Mediclinic, Cape Town, South Africa; 5Grootte Schuur Hospital, Cape Town, South Africa; 6Neuroscience Institute, University of Cape Town, Cape Town, South Africa; 7Human Metabolomics, North-West University, Potchefstroom, South Africa



Leigh syndrome and Fanconi renotubular syndrome are the main clinical phenotype due to mutations in NDUFAF6 gene.

Teresa Rizza1, Alessandra Torraco1, Michela Di Nottia1, Daniela Verrigni1, Anastasia Altobelli1, Diego Martinelli1, Daria Diodato1, Stephanie Efthymiou2, Carlo Dionisi-Vici1, Enrico Bertini1, Reza Maroofian2, Agnes Rotig3, Rosalba Carrozzo1

1Bambino Gesù Children Hospital, Italy; 2UCL Queen Square Institute of Neurology; 3Université Paris Descartes, Sorbonne Paris Cité



Mitochondrial encephalomyopathy associated with the m.618T>C in MT-TF

Aryane Silva Coutinho2, Tainara Zappia Tessaro1, Rodrigo Fonseca Vilanova1, Celia Harumi Tengan2

1Hospital Municipal Dr. José de Carvalho, Brazil; 2Escola Paulista de Medicina, Universidade Federal de São Paulo, Brazil



TWNK in Parkinson's disease: a Movement Disorder and Mitochondrial Disease Center perspective study

Marco Percetti1,2,3, Giulia Franco3,4, Edoardo Monfrini3,4, Leonardo Caporali5, Raffaella Minardi5, Chiara La Morgia5, Maria Lucia Valentino5,6, Rocco Liguori5,6, Ilaria Palmieri7, Donatella Ottaviani8, Maria Vizziello3, Dario Ronchi3, Federica Di Berardino9, Antoniangela Cocco10,11, Bertil Macao12, Maria Falkenberg12, Giacomo Pietro Comi3,4, Alberto Albanese11, Bruno Giometto8, Enza Maria Valente7,13, Valerio Carelli5,6, Alessio Di Fonzo3,4

1School of Medicine and Surgery and Milan Center for Neuroscience, University of Milan-Bicocca.; 2Foundation IRCCS San Gerardo dei Tintori, Monza; 3Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Milan 20122, Italy; 4Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy; 5IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy; 6Unit of Neurology, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy; 7Neurogenetics Research Center, IRCCS Mondino Foundation, Pavia, Italy; 8Neurology Unit, Rovereto Hospital, Azienda Provinciale per i Servizi Sanitari (APSS) di Trento, Trento, Italy; 9Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Audiology Unit, Milan, Italy; 10University of Milan, Milan, Italy; 11Department of Neurology, Istituto di Ricovero e Cura a Carattere Scientifico Humanitas, Research Hospital, Milan, Italy; 12Department of Medical Biochemistry and Cell Biology, University of Gothenburg, P.O. Box 440, SE 405 30 Gothenburg, Sweden; 13Department of Molecular Medicine, University of Pavia, Pavia, Italy.



Novel pathogenic MT-ND3 variant causing a particular MELAS phenotype

Lucile Riera-Navarro1, Cécile Rouzier1, Samira Saadi Ait-El-Mkadem1, Konstantina Fragaki1,2, Sylvie Bannwarth1,2, Pierre Thomas3, Luisa Villa4, Véronique Paquis Flucklinger1,2, Annabelle Chaussenot1

1CHU de Nice, France; 2Université Côte d'Azur, CNRS, INSERM, IRCAN; 3Service de Neurologie- Hôpital Pasteur 2, CHU de Nice; 4Centre de référence des Maladies neuromusculaires



Mitochondrial molecular genetic findings in the South African diagnostic setting

Surita Meldau1,2, Elizabeth Patricia Owen1,2, Kashief Khan2, Gillian Tracy Riordan3

1University of Cape Town, Cape Town, South Africa; 2National Health Laboratory Sevices, South Africa; 3Red Cross War Memorial Children's Hospital, Cape Town, South Africa



Known genes, new genes and new phenotypes in inherited mitochondrial eye diseases

Neringa Jurkute1,2,3, Gavin Arno1,2,4, Patrick Yu-Wai-Man1,2,5,6, Andrew R Webster1,2

1Moorfields Eye Hospital NHS Foundation Trust, London, UK; 2Institute of Ophthalmology, University College London, London, UK; 3The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK; 4North Thames Genomic Laboratory Hub, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK; 5John van Geest Centre for Brain Repair and MRC Mitochondrial Biology Unit, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK; 6Cambridge Eye Unit, Addenbrooke’s Hospital, Cambridge University Hospitals, Cambridge, UK



LHON spectrum disorder: new phenotypes and genotypes

Marco Battista1, Leonardo Caporali2, Enrico Borrelli1, Giorgio Lari1, Alice Galzignato3, Claudio Fiorini2, Paolo Nucci4, Francesco Bandello1, Maria Lucia Cascavilla1, Valerio Carelli2,5, Piero Barboni1

1San Raffaele Hospital, Italy; 2IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica (Bologna, Italy); 3Studio Oculistico d’Azeglio (Bologna, Italy); 4Department of Clinical Science and Community Health, University of Milan, (Milan, Italy); 5Unit of Neurology, Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna (Bologna, Italy)



Chronic asymmetric progressive external ophthalmoplegia without eyelid weakness

Jae Ho Jung

Seoul National University Hospital, Korea, Republic of (South Korea)



Bayesian inference enables discovery of functional effects of heteroplasmic mitochondrial mutations in the developing brain

Aidan Scott Marshall, Yue Nie, Nick.S Jones, Patrick.F Chinnery

Imperial College London, United Kingdom



Cell lineage-specific mitochondrial gene expression is established in the early embryo, prior to organ maturation

Stephen P. Burr1,2, Florian Klimm1,2,3, Angelos Glynos1,2, Malwina Prater1,2,4, James B. Stewart5,6, Patrick F. Chinnery1,2, Michele Frison1,2

1Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; 2Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; 3Novo Nordisk Research Centre Oxford, Innovation Building, University of Oxford, Old Road Campus, Oxford, UK; 4Functional Genomics Centre, Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK; 5Max Planck Institute for Biology of Ageing, Cologne, Germany; 6Biosciences Institute, Faculty of Medical Sciences, Wellcome Centre for Mitochondrial Research, Newcastle University, Newcastle upon Tyne, UK



Identifying mitochondrial methyltransferases using unbiased proteome-ligand profiling

Alissa Wilhalm1, David Moore1, Florian Rosenberger1, Anna Wedell2, Christoph Freyer1, Anna Wredenberg1,2

1Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden, Sweden; 2Centre of Inherited metabolic diseases, Karolinska University Hospital, Stockholm, Sweden



Engineering mitochondrial aminoacyl-tRNA synthetases as a tool to investigate mitochondrial protein synthesis

Christin A Albus, Ellyn N Willsher, Syeda Z Akthar, Robert N Lightowlers, Zofia MA Chrzanowska-Lightowlers

Newcastle University, United Kingdom



Short-read NGS for the screening of structural and copy number alterations in mtDNA as powerful diagnostic tool.

Christiane Neuhofer1,2, Rossella Izzo3, Riccardo Berutti1,2, Martin Pavlov2, Eleonora Lamantea3, Elisabeth Graf1, Costanza Lamperti3, Thomas Klopstock4, Holger Prokisch1,2, Daniele Ghezzi3,5, Andrea Legati3

1Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, (Munich, Germany); 2Institute of Neurogenomics, Helmholtz Zentrum München (Neuherberg, Germany); 3Fondazione IRCCS Istituto Neurologico Carlo Besta (Milan, Italy); 4Department of Neurology, Friedrich-Baur-Institute, LMU Hospital, Ludwig Maximilians University (Munich, Germany); 5Department of Pathophysiology and Transplantation, University of Milan (Milan, Italy)



Ethical dilemmas and diagnostic uplifts; primary mitochondrial disease the era of first line whole genome sequencing

William L. Macken1,2, Rachel L. Horton3,4, Michael G. Hanna1,2, Anneke M. Lucassen3,4, Robert D.S. Pitceathly1,2

1UCL Queen Square Institute of Neurology, United Kingdom; 2NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK.; 3Centre for Personalised Medicine, and Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK; 4Clinical Ethics, Law and Society, Faculty of Medicine, University of Southampton, Southampton, UK



Analysis of mitochondrial metabolism using 13C-labeled mass isotopologue analysis and mass spectrometry as a new approach for the diagnostics of mitochondrial disorders

Liesbeth T. Wintjes1, Arno van Rooij1, Sacha Hendriks1, Coby M. Laarakkers1, Richard J.T. Rodenburg1,2

1Department of Genetics, Translational Metabolic Laboratory, Radboudumc, Nijmegen, The Netherlands.; 2Department of Pediatrics, Radboud Centre for Mitochondrial Medicine, Radboudumc, Nijmegen, The Netherlands



Subcellular metabolomics: a pipeline for compartment-specific metabolic investigations in a mouse model of Leigh syndrome

Gunter van der Walt, Jason Elferink, Zander Lindeque, Shayne Mason, Roan Louw

North-West University, South Africa



High‐content screening for modulators of mitochondria‐ER contact sites and identification of their protein targets

Tomas Knedlik1, Federica Dal Bello1, Marta Giacomello1,2

1Department of Biology, University of Padova, Italy; 2Department of Biomedical Sciences, University of Padova, Italy



A novel Approach to assess the pathogenicity of mtDNA Variants

Omar Tutakhel, Frans van den Brandt, Kai Francke, Hatice Nur Ozhan, Frans Maas, Daan Panneman, Liesbeth Wintjes, Roel Smeets, Dirk Lefeber, Bert van den Heuvel, Richard J. Rodenburg

RadboudUMC, Translational Metabolic Laboratory, Dept of Pediatrics, Nijmegen, The Netherlands



At the core of the apoptotic foci

Hector Flores-Romero, Aida Peña Blanco, Lisa Hohorst, Jonas Aufdermauer, Shashank Dadsena, Cristiana Zollo, Rodrigo Cuevas-Arenas, Ana J. Garcia-Saez.

CECAD, Germany



Clinical utility of ultra-rapid genomic testing for infants and children with a suspected mitochondrial disorder

John Christodoulou1,2,3,4, Sophie Bouffler5, Chirag Patel6, Sarah Sandaradura4,7, Meredith Wilson4,7,8, Jason Pinner8,9, Matthew Hunter10,11, Christopher Barnett12,13,14, Mathew Wallis15,16, Benjamin Kamien17, Tiong Tan1,2,3, Mary-Louise Freckmann18, Karin Kassahn13,14, Tony Roscioli19,20,21, Alison Compton1,2,3, David Thorburn1,2,3, Sebastian Lunke1,2,3,5, Zornitza Stark1,2,3,5

1Murdoch Children's Research Institute, Melbourne, Australia; 2University of Melbourne, Melbourne, Australia; 3Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, Australia; 4University of Sydney, Sydney, Australia; 5Australian Genomics, Melbourne, Australia; 6Genetic Health Queensland, Royal Brisbane and Women’s Hospital, Brisbane, Australia; 7Sydney Children’s Hospitals Network – Westmead, Sydney, Australia; 8Sydney Children’s Hospitals Network – Randwick, Sydney, Australia; 9University of New South Wales, Sydney, Australia; 10Monash Genetics, Monash Health, Melbourne, Australia; 11Department of Paediatrics, Monash University, Melbourne, Australia; 12Paediatric and Reproductive Genetics Unit, Women’s and Children’s Hospital, North Adelaide, Australia; 13Adelaide Medical School, The University of Adelaide, Adelaide, Australia; 14Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, Australia; 15Tasmanian Clinical Genetics Service, Tasmanian Health Service, Hobart, Australia; 16School of Medicine and Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia; 17Genetic Services of Western Australia, Perth, Australia; 18Department of Clinical Genetics, The Canberra Hospital, Canberra, Australia; 19Centre for Clinical Genetics, Sydney Children's Hospital, Sydney, NSW, Australia; 20Randwick Genomics Laboratory, NSW Health Pathology, Prince of Wales Hospital, Sydney, Australia; 21Neuroscience Research Australia (NeuRA) and Prince of Wales Clinical School, UNSW, Sydney, Australia



Contribution of RNA-seq to diagnosis and determination of functional impact of candidate variants in 45 patients suspected of mitochondrial disease.

Gerard Muñoz-Pujol1, Olatz Ugarteburu1, Blai Morales1, Judit García-Villoria1, Laura Gort1, Vicente A. Yépez2, Julien Gagneur2, Mirjana Gusic2, Holger Prokisch2, Marc Dabad3, Anna Esteve-Codina3, Antonia Ribes1, Frederic Tort1

1Secció d'Errors Congènits del Metabolisme-IBC, Servei de Bioquímica i Genètica Molecular, Hospital Clínic de Barcelona, IDIBAPS, CIBERER, Barcelona, Spain; 2Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany; 3CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology and Universitat Pompeu Fabra, Barcelona, Spain



Dynamics of NAD and glutathione metabolites in blood during aging, in disease and upon supplementation with NAD-booster

Liliya Euro1,2, Kimmo Haimilahti1, Sonja Jansson1,2, Jana Buzkova1,2, Anu Suomalainen-Wartiovaara1,3

1University of Helsinki, Finland; 2NADMED Ltd, Finland; 3HUS Diagnostic Centre, Finland



Enzymatic assay for UDP-GlcNAc and its application in the parallel assessment of substrate availability and protein O-GlcNAcylation

Marc Sunden1,2, Divya Upadhyay1,2, Rishi Banerjee1,2, Nina Sipari3, Vineta Fellman1,2,4, Jukka Kallijarvi1,2, Janne Purhonen1,2

1Folkhalsan Research Center, Finland; 2Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Finland; 3Viikki Metabolomics Unit, University of Helsinki, Finland; 4Children’s Hospital, Helsinki University Hospital, Finland



Genetic testing for mitochondrial disease: The United Kingdom best practice guidelines

Eleni Mavraki1,2, Robyn Labrum3, Kate Sergeant4, Charlotte L Alston1,2, Cathy Woodward3, Conrad Smith4, Charlotte V Y Knowles1,2, Yogen Patel3, Philip Hodsdon4, Jack P Baines1,2, Emma L Blakely1,2, James Polke3, Robert W Taylor1,2, Carl Fratter4

1NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK; 2Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; 3Neurogenetics Unit, National Hospital for Neurology and Neurosurgery, Queen Square, London, UK; 4Oxford Genetics Laboratories, Oxford University Hospitals NHS Foundation Trust, Oxford, UK



Identification of uncharacterized genes involved in mitochondrial OXPHOS function and integrity.

Marcos Javier Zamora Dorta, Sara Laine Menéndez, David Abia Holgado, Eduardo Balsa Martínez

Centro de Biología Molecular Severo Ochoa, Spain



Investigating the role of mito-nuclear genetic variation in determining m.3243A>G variant heteroplasmy

Alia K. Saeed1, Róisín M. Boggan1, Imogen G. Franklin1, Charlotte L. Alston1,2, Emma L. Blakely1,2, Boriana Büchner3, Enrico Bugiardini4, Kevin Colclough5, Gráinne S. Gorman1, Catherine Feeney1, Michael G. Hanna4, Andrew T. Hattersley6, Thomas Klopstock3,7,8, Cornelia Kornblum9, Michelangelo Mancuso10, Yi Shiau Ng1, Kashyap A. Patel6, Robert D. S. Pitceathly4, Chiara Pizzamiglio4, Holger Prokisch11,12, Jochen Schäfer13, Andrew M. Schaefer1, Maggie H. Shepherd6, Annemarie Thaele14, Doug M. Turnbull1, Cathy E. Woodward15, Heather J. Cordell16, Robert McFarland1, Robert W. Taylor1,2, Gavin H. Hudson1, Sarah J. Pickett1

1Wellcome Centre for Mitochondrial Research and Institute for Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, UK; 2NHS Highly Specialised Mitochondrial Diagnostic Laboratory, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK; 3Department of Neurology, Friedrich-Baur-Institute, University Hospital of the Ludwig-Maximilians-University (LMU Klinikum), Munich, Germany; 4Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK; 5Exeter Genomics Laboratory, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK; 6Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK; 7Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; 8German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; 9Department of Neurology, University Hospital Bonn, Bonn, Germany; 10Neurological Institute of Pisa, Italy; 11Institute of Human Genetics, School of Medicine, Technische Universität München, München, Germany; 12Institute of Neurogenomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; 13Department of Neurology, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; 14Department of Neurology, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; 15Neurogenetics Unit, The National Hospital for Neurology and Neurosurgery, London, UK; 16Population Health Sciences Institute, Newcastle University, UK



Mitochondrial DNA depletion and deletion analysis using smMIPs

Maaike Brink1,2, Sanne van Kraaij1,2, Sanne Sweegers1,2, Roel Smeets1,2, Richard Rodenburg1,2,3

1Translational Metabolic Laboratory, Radboudumc, Nijmegen, The Netherlands; 2Radboud Center for Mitochondrial Medicine (RCMM), Radboudumc, Nijmegen, The Netherlands; 3Department of Pediatrics, Radboudumc, Nijmegen, The Netherlands



Ultrastructure of mitochondria in 3D from volume electron microscopy

Chenhao Wang1,2, Leif Østergaard3,4, Stine Hasselholt3,4, Jon Sporring1,2

1Department of Computer Science, University of Copenhagen, Denmark; 2Center for Quantification of Imaging Data from MAX IV; 3Department of Clinical Medicine, Aarhus University, Denmark; 4Center of Functionally Integrative Neuroscience



Visualizing ATP dynamics in living mice

Masamichi Yamamoto, Jungmi Choi

National Cerebral and Cardiovascular Center, Japan



Applying sodium carbonate extraction mass spectrometry to investigate defects in the mitochondrial respiratory chain

David Robert Lindsay Robinson1, Daniella Hock1, Linden Muellner-Wong1,2, Roopingsam Kugapreethan1, Boris Reljic1,3, Elliot Surgenor3,4, Carlos Rodrigues1,5, Nikeisha Caruana1,6, David Stroud1,2

1Department of Biochemistry and Pharmacology, University of Melbourne, Melbourne, Australia; 2Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, Australia; 3Department of Biochemistry and Molecular Biology, Monash University, Melbourne,Australia; 4The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia; 5Baker Heart and Diabetes Institute, Melbourne, Australia; 6Institute for Health and Sport (IHES), Victoria University, Melbourne, Australia



Global analysis of protein methylation in the mitochondrial compartment of cancer cells: a proteomic approach

Ayusi Mondal1,2, Alessandro Vai1,2, Silvia Pedretti1,3, Marianna Maniaci1, Nico Mitro1,3, Tiziana Bonaldi1,4

1Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS Milano, Italy; 2European School of Molecular Medicine (SEMM); 3Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy; 4Department of Oncology and Hematology-Oncology, University of Milan, Milan, Italy



MITODIAG : The French network of diagnostic laboratories for mitochondrial diseases

Cécile Rouzier1, Emmanuelle Pion2, Céline Bris3, Patrizia Bonneau3, Valérie Desquiret3, Jean-Paul Bonnefont4, Pauline Gaignard5, Elise Lebigot5, Samira Ait-El-Mkadem Saadi1, Sylvie Bannwarth1, Konstantina Fragaki1, Benoit Rucheton6, Marie-Laure Martin-Negrier7, Aurélien Trimouille7, Cécile Acquaviva-Bourdain8, Cécile Pagan8, Anne-Sophie Lebre9, Gaelle Hardy10, Stéphane Allouche11, Pascal Reynier3, Mireille Cossee12, Sharam Attarian13, Véronique Paquis-Flucklinger1, Vincent Procaccio3

1Service de génétique médicale, Centre de référence des maladies mitochondriales, CHU Nice, Université Cote d’Azur, CNRS, INSERM, IRCAN, Nice; 2Filnemus, laboratoire de génétique moléculaire, CHU Montpellier; 3Service de génétique, Institut de Biologie en santé, Centre National de référence Maladies Neurodégénératives et Mitochondriales, CHU Angers; 4Fédération de génétique médicale, Service de génétique moléculaire du GH Necker-enfants malades, Hôpital Necker-Enfants Malades, Paris; 5Laboratoire de Biochimie, Pôle BPP, CHU Paris Sud, Hôpital Bicêtre-le Kremlin Bicêtre, Paris; 6Pôle de biologie et pathologie, CHU Bordeaux; 7Unité fonctionnelle d’histologie moléculaire, Service de pathologie, CHU Bordeaux-GU Pellegrin, Bordeaux; 8Service de biochimie et biologie moléculaire Grand Est, UM Maladies Héréditaires du Métabolisme, Centre de biologie et pathologie Est, CHU Lyon HCL, GH Est, Lyon; 9Laboratoire de génétique, Hématologie et Immunologie, CHU Reims; 10Laboratoire de génétique moléculaire: maladies héréditaires et oncologie, Service de biochimie, biologie moléculaire et toxicologie environnementale, CHU Grenoble et des Alpes, Institut de biologie et pathologie, Grenoble; 11Service de biochimie, Pôle Biologie, Pharmacie et Hygiène, CHU Caen, Hôpital de la Côte de Nacre, Caen; 12Laboratoire de Génétique Moléculaire, CHU Montpellier, PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier; 13Filnemus, Assistance Publique Hôpitaux Marseille, Service de Neurologie, Hôpital La Timone, Marseille



Multiomic mitochondrial and metabolic screening reveals potential biomarkers in inclusion body myositis

Judith Cantó Santos1,2,3, Laura Valls Roca1,2,3, Ester Tobías1,2,3, Clara Oliva4, Francesc Josep García-García1,2,3, Mariona Guitart Mampel1,2,3, Félix Andújar Sánchez1,2,3, Laia Farré Tarrats1,2,3, Joan Padrosa1,2,3, Raquel Aránega1,2,3, Pedro J. Moreno Lozano1,2,3, José César Milisenda1,2,3, Rafael Artuch4, Josep M. Grau Junyent1,2,3, Glòria Garrabou1,2,3

1Hereditary Metabolic Diseases and Muscular Diseases Lab, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain; 2Department of Internal Medicine, Hospital Clinic of Barcelona, Barcelona, Spain; 3CIBERER— Spanish Biomedical Research Centre in Rare Diseases, Madrid, Spain; 4Department of Clinical Biochemistry, Institut de Recerca Sant Joan de Déu; Esplugues de Llobregat, Barcelona, Spain



Network analysis of protein-protein interactions identifies intermediate filaments as a novel mitochondrial dynamics related structure

Irene M.G.M. Hemel, Carlijn Steen, Michiel Adriaens, Mike Gerards

Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, The Netherlands



Establishment of mitochondrial proline metabolic disorder patient-derived induced pluripotent stem cells as a new cellular model for aging associated disease study

Yu-Wen Huang, Tsung-Han Lee, Dar-Shong Lin

Mackay Memorial Hospital, Taiwan



Mitochondrial disorders unraveled by NGS technologies

Yulia Itkis1, Tatiana Krylova1, Natalia Pechatnikova2, Ekaterina Zakharova1

1Research centre for medical genetics, Russian Federation; 2Morozov's Moscow City Child Clinical Hospital, Moscow, Russia



Incorporation of exogenous mitochondria into cells and their effects on the cells

Hisashi Ohta1,2, Yosif El-Darawish1,2, Masae Takeda1,2, Takahiro Shibata1,2, Keiichi Sakakibara1,2, Rick Tsai1, Masashi Suganuma1

1LUCA Science, Japan; 2Biological Drug Development based DDS technology, Hokkaido Univ.



Mitochondrial encapsulation technology for mitochondrial transplantation therapy

Yonghui Wang1,2, Oliver Koivisto1,2, Chang Liu1,2, Hongbo Zhang1,2

1Pharmaceutical Sciences Laboratory, Åbo Akademi University, Finland; 2Turku Bioscience Centre, University of Turku and Åbo Akademi University



Inhibition of mtDNA transcription in liver reverses diet-induced obesity and hepatosteatosis in the mouse

Shan Jiang1, Taolin Yuan1, Laura Kremer1, Florian Schober2, Fynn Hansen2, Diana Rubalcava-Gracia1, Mara Mennuni1, Roberta Filograna1, David Alsina1, Jelena Misic1, Patrick Giavalisco3, Matthias Mann2, Nils-Göran Larsson1

1Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Sweden; 2Max-Planck Institute of Biochemistry, Martinsried, Germany; 3Metabolomics Core Facility, Max Planck Institute for Biology of Ageing, Cologne, Germany



The mitochondrial phenotype of Leigh syndrome SURF1 mutant patient-derived fibroblasts and recovery using small molecules

Rachel M Hughes1, Laura M Ellis1, Naomi Hartopp1, Emily Mossman1, Gauri Bhosale2, Annachiara Gandini2, Alessandro Pristera2, Christopher Doe2, Scott P Allen1, Oliver Bandmann1, Laura Ferraiuolo1, Pamela J Shaw1, Heather J Mortiboys1

1Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom; 2Nanna Therapeutics, Cambridge, United Kingdom



Knockout of Complex III subunit Uqcrh decreases respiratory capacity and impairs cardiac contractile function independent of mitochondrial ROS production

Nadine Spielmann1,9, Christina Schenkl2,9, Tímea Komlódi3,4, Patricia da Silva-Buttkus1, Estelle Heyne2, Jana Rohde1, Oana V. Amarie1, Birgit Rathkolb1,5,6, Erich Gnaiger3, Torsten Doenst2, Helmut Fuchs1, Valérie Gailus-Durner1, Martin Hrabě de Angelis1,6,7,9, Marten Szibor2,8,9

1Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Center Munich, German Research Center for Environmental Health, Germany; 2Jena University Hospital, Friedrich-Schiller University of Jena, Germany; 3Oroboros Instruments, Innsbruck, Austria; 4Department of Biochemistry and Molecular Biology, Semmelweis University Budapest, Hungary; 5Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Germany; 6Member of German Center for Diabetes Research (DZD), Germany; 7Chair of Experimental Genetics Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, Germany; 8BioMediTech & Tampere University Hospital, Faculty of Medicine and Health Technology, Tampere University, Finland; 9Contributed equally



Molecular insights into the role of complex V deficiency in heart development, function and disease

Mario Pavez-Giani1,2, Karen an der Brügge1, Till Stephan4, Felix Lange4, Jakob Fell1,2, Alessandro Prigione3, Stefan Jakobs4, Lukas Cyganek1,2

1Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany; 2German Center for Cardiovascular Research (DZHK), partner site Göttingen, Germany; 3Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany; 4Research Group Mitochondrial Structure and Dynamics, Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany



Establishing mammalian cell models for research of NDUFS1-associated diseases

Lena Jentsch1, Laura Schröter2, Natascia Ventura1,2

1Heinrich Heine University Düsseldorf, Germany; 2IUF- Leibniz Research Institute for Environmental Medicine



A neuronal model of mtDNA disease reveals a compensatory reprogramming of the electron transfer chain during neuronal maturation

Shane Thomas Bell1, Rebeca Acín-Pérez2, Iffath Ghouri1, Orian Shirihai2, Robert Lightowlers1, Oliver Russell1

1Wellcome Centre for Mitochondrial Research, Newcastle University, United Kingdom; 2Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA, USA



Development of mutant mtDNA-targeted TALENs and their application to iPSC-based mitochondrial disease model.

Naoki Yahata, Ryuji Hata

Fujita Health University School of Medicine, Japan



Deficits in mitochondrial oxidative phosphorylation enhance SARS-CoV-2 replication

Yentli E. Soto Albrecht1,2, Ryan Morrow1, Arnold Olali1, Devin Kenney3,4, Prasanth Potluri1, Deborah Murdock1, Alessia Angelin1, Florian Douam3,4, Douglas C. Wallace1,2

1Children's Hospital of Philadelphia, USA; 2University of Pennsylvania, USA; 3Boston University, USA; 4National Emerging Infectious Diseases Laboratories, Boston, USA



Metabolic analysis of mouse sarcopenic skeletal muscle identifies new strategies to increase lifespan in C. elegans

Steffi M Jonk1, Vicki Chrysostomou2, James R Tribble1, Jonathan G Crowston2,3,4, Peter Swoboda1, Pete A Williams1

1Karolinska Institutet, Sweden; 2Centre for Vision Research Duke-NUS & Singapore National Eye Centre, Singapore; 3Save Sight Institute at the University of Sydney, Australia; 4The University of Melbourne, Australia.



Delineating mitochondrial pathology using a genome-wide CRISPR/Cas9 activation screen

Yasmin Tang1, Angela Pyle1, Krutik Patel1, Robert McFarland1,2, Robert W. Taylor1,2, Monika Oláhová1

1Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH; 2NHS Highly Specialised Rare Mitochondrial Disorders Service, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, NE2 4HH



Cancer and cellular senescence – two complementary stress models to study turnover and quality control of mitochondrial respiratory complexes.

Hanna Salmonowicz1,2, Mahdi S Mahdi1,2, Deepti Mudartha1, Szymon Gorgon1,2, Stadnik Dorota1,2, Remigiusz Serwa1,2, Karolina Szczepanowska1,2

1IMol Polish Academy of Sciences, Poland; 2ReMedy International Research Agenda



Proteotoxicity induced mitochondrial integrated stress response in CHCHD10-linked adult-onset spinal muscular atrophy

Sandra Harjuhaahto1, Bowen Hu1, Jouni Kvist1, Fuping Zhang2,3, Tomas Zárybnický2, Kimmo Haimilahti4, Eija Pirinen4, Emilia Kuuluvainen2, Ville Hietakangas2,5, Satu Kuure2,3, Manu Jokela6,7, Emil Ylikallio1,8, Henna Tyynismaa1

1Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki; 2Helsinki Institute of Life Science HiLIFE, University of Helsinki; 3Genetically Modified Rodents Unit, Laboratory Animal Center, University of Helsinki; 4Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki; 5Faculty of Biological and Environmental Sciences, University of Helsinki; 6Division of Clinical Neurosciences, Turku University Hospital and University of Turku; 7Department of Neurology, Neuromuscular Research Center, Tampere University Hospital and Tampere University; 8Clinical Neurosciences, Neurology, Helsinki University Hospital



Protective role of mitochondrial stress signaling and fragmentation in mitochondrial cardiomyopathy

Sofia Ahola, Lilli Pazurek, Fiona Mayer, Hendrik Nolte, Thomas Langer

Max Planck Institute for Biology of Ageing, Cologne, Germany



The Italian reappraisal on the most frequent genetic defects in hereditary optic neuropathies and the global top 10

Claudio Fiorini1, Danara Ormanbekova1, Flavia Palombo1, Alberto Pasti1, Michele Carbonelli2, Giulia Amore2, Martina Romagnoli2, Pietro D'Agati2, Maria Lucia Valentino1,2, Piero Barboni3, Maria Lucia Cascavilla3, Anna Maria De Negri4, Federico Sadun5, Arturo Carta6, Francesco Testa7, Vittoria Petruzzella8, Silvana Guerriero8, Stefania Bianchi Marzoli9, Valerio Carelli1,2, Chiara La Morgia1,2, Leonardo Caporali2

1IRCCS Istituto di Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy; 2Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy; 3Department of Ophthalmology, University Vita-Salute, IRCCS Ospedale San Raffaele, Milan, Italy; 4Azienda Ospedaliera San Camillo-Forlanini, Rome, Italy; 5Ospedale Oftalmico Roma, Rome, Italy; 6Ophthalmology Unit, University Hospital of Parma, Parma, Italy; 7Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania Luigi Vanvitelli, Naples, Italy; 8Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari Aldo Moro, Bari, Italy; 9Neuroophthalmology Service and Ocular Electrophysiology laboratory, Department of Ophthalmology, IRCCS Istituto Auxologico Italiano, Milan, Italy



Aberrant ER-mitochondria communication in human mitochondrial disease

Eric A. Schon1, Khushbu Kabra1, Patricia Morcillo1, Delfina Larrea1, Estela Area-Gomez1,2, Orhan Akman1

1Columbia University, USA; 2Centro de Investigaciones Biológicas “Margarita Salas”, Madrid, Spain



Mitochondrial F0F1-ATP synthase conditions the responsiveness of mitochondria to fission

Charlène Lhuissier1, Julien Cassereau3, Valérie Desquiret-Dumas2, Guy Lenaers1, Naïg Gueguen2, Arnaud Chevrollier1

1MITOVASC Université d'Angers, France; 2Departments of Biochemistry and Molecular Biology, University Hospital Angers, Angers, France; 3Laboratoire de Neurobiologie et Neuropathologie, Centre Hospitalier Universitaire d'Angers, Angers, France



A screening method for mitochondrial disorders by high-resolution respirometry

Kersti Tepp1, Kairit Joost2, Natalja Timohhina1, Marju Puurand1, Tuuli Kaambre1

1National Institute of Chemical Physics and Biophysics, Estonia; 2Clinic of Internal Medicine, East-Tallinn Central Hospital, Estonia



AK3, adenylate kinase isozyme 3, is a new gene associated with PEO and multiple mtDNA deletions

Alessia Nasca1, Andrea Legati1, Teresa Ciavattini1, Nadia Zanetti1, Eleonora Lamantea1, Javier Ramón2, Ramon Martí2, Maria Antonietta Maioli3, Costanza Lamperti1, Holger Prokisch4,5, Daniele Ghezzi1,6

1Fondazione IRCCS Istituto Neurologico Besta, Italy; 2Vall d'Hebron Research Institute, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Autonomous University of Barcelona, Barcelona, Spain; 3Centro Sclerosi Multipla, P.O. Binaghi, ASL Cagliari, Italy; 4Technical University of Munich, School of Medicine, Institute of Human Genetics, 81675 Munich, Germany; 5Institute of Neurogenomics, Helmholtz Zentrum München, 85764 Munich, Germany; 6Department of Pathophysiology and Transplantation (DEPT), University of Milan, Italy



Heterozygous missense variants in NUTF2 (nuclear transport factor 2) gene, mapping at the OPA8 locus, cause Dominant Optic Atrophy

Agnese Macaluso1, Alessandra Maresca1, Concetta Valentina Tropeano1, Maria Antonietta Capristo1, Flavia Palombo1, Leonardo Caporali1, Claudio Fiorini1, Danara Ormanbekova1, Chiara La Morgia1, Piero Barboni2,3, Cristina Villaverde4,5, Carmen Ayuso4,5, Maria Esther Gallardo6,5, Majida Charif7, Sylvie Gerber8, Patrizia Amati-Bonneau7, Guy Lanaers7,9, Jean-Michel Rozet7, Bernd Wissinger10, Valerio Carelli1,11, Valentina Del Dotto11

1IRCCS - Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica - Bologna (Italy); 2Studio Oculistico d'Azeglio - Bologna (Italy); 3Department of Ophthalmology, University Vita-Salute, IRCCS Ospedale San Raffaele - Milano (Italy); 4Department of Genetics & Genomics, Instituto de Investigación Sanitaria - Fundación Jiménez Díaz University Hospital - Universidad Autónoma de Madrid (IIS-FJD-UAM) - Madrid (Spain); 5Center for Biomedical Network Research on Rare Diseases (CIBERER), ISCIII - Madrid (Spain); 6Grupo de investigación traslacional con células iPS, Instituto de Investigación Sanitaria Hospital 12 de Octubre (i+12), Madrid, Spain; Centro de Investigación Biomédica en Red (CIBERER) - Madrid (Spain); 7Université d’Angers, MitoLab team, UMR CNRS 6015 - INSERM U1083, Unité MitoVasc - Angers (France); 8Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetic Diseases, Imagine and Paris Descartes University - Paris (France); 9Departments of Biochemistry and Genetics, University Hospital Angers - Angers (France); 10Molecular Genetics Laboratory, Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany; 11Depart. of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna - Bologna (Italy)



Southern African paediatric patients with King Denborough syndrome are exclusively associated with an autosomal recessive STAC3 variant: is this a highly prevalent secondary mitochondrial disease in this African population?

Francois Hendrikus van der Westhuizen1, Maryke Schoonen1, Michelle Bisschoff1, Ronel Human2, Elsa Lubbe2, Malebo Nonyane2, Armand Vorster1, Karin Terburgh1, Robert McFarland3, Robert Taylor3, Mahmoud Fassad3, Krutik Patel3, Wilson Lindsay4, Michael Hanna4, Jana Vandrovcova4, The ICGNMD Consortium5, Izelle Smuts2

1Human Metabolomics, North-West University, Potchefstroom, South Africa; 2Department of Paediatrics, Steve Biko Academic Hospital, University of Pretoria, Pretoria, South Africa; 3Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom; 4Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; 5https://www.ucl.ac.uk/genomic-medicine-neuromuscular-diseases/global-contributor-list



Long-read NGS for detection of mitochondrial DNA large-scale deletions and complex rearrangements

Chiara Frascarelli1, Nadia Zanetti1, Alessia Nasca1, Rossella Izzo1, Costanza Lamperti1, Eleonora Lamantea1, Daniele Ghezzi1,2, Andrea Legati1

1Fondazione IRCCS Istituto Neurologico Carlo Besta (Milan, Italy); 2University of Milan (Milan, Italy)



Quantification of all 12 canonical ribonucleotides by real-time fluorogenic in vitro transcription

Janne Purhonen1,2, Jukka Kallijarvi1,2

1Folkhalsan Research Center, Finland; 2Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki



Quantifying mitochondrial proteome remodeling during macrophage polarization

Joan Blanco-Fernandez, Manfredo Quadroni, Alexis A. Jourdain

University of Lausanne, Switzerland



Mitochondrial injury in warm ischemia studied by high-resolution respirometry

Alba Timón-Gómez, Luiza HD Cardoso, Eleonora Baglivo, Carolina Doerrier, Erich Gnaiger

Oroboros Instruments GmBH, Austria



MitoCluster: integrated phenotyping and mouse model generation platform for mitochondrial disease and dysfunction

Micol Falabella1, Patrick F. Chinnery2, Laura C. Greaves3, Michael G. Hanna1,4, Thomas M. Keane5, Robert McFarland3, Michal Minczuk2, Owen J. Sansom6,7, James B. Stewart3, Michelle Stewart8, Lydia Teboul8, Keira Turner2, Jelle van den Ameele2, Carlo Viscomi9, Sara Wells8, Alexander J. Whitworth2, Robert D. S. Pitceathly1,4

1Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK; 2Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge UK; 3Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, UK; 4NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK; 5European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, UK; 6Cancer Research UK Beatson Institute, Glasgow, UK; 7Institute of Cancer Sciences, University of Glasgow, Glasgow, UK; 8Mary Lyon Centre MRC Harwell, UK; 9University of Padua, Italy