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
Date: Monday, 12/June/2023
8:00am
-
6:30pm
Slides Center
Location: Slides Center
Registration Desk
Location: Bologna Congress Center
9:00am
-
10:45am
Session 2.1: mtDNA maintenance and expression
Location: Bologna Congress Center - Sala Europa
Chair: Zofia Chrzanowska-Lightowers
Chair: Massimo Zeviani
Invited Speakers: M. Falkemberg; A. Filipovska
 
Invited

Initiation of mitochondrial DNA replication in mammalian cells.

Maria Falkenberg

Gothenburg University, Sweden



Invited

Regulation of mitochondrial gene expression in disease

Aleksandra Filipovska

University of Western Australia, Australia



Oral presentation

Mitochondrial translation termination at non-canonical stop codons

Annika Krüger1, Cristina Remes2, Dmitrii Igorevich Shiriaev1, Yong Liu1, Henrik Spåhr1, Rolf Wibom1, Ilian Atanassov3, Minh Duc Nguyen1, Barry S. Cooperman2, Joanna Rorbach1,3

1: Karolinska Institutet, Stockholm, Sweden; 2: University of Pennsylvania, Pennsylvania, USA; 3: Max-Planck-Institute for Biology of Ageing, Cologne, Germany



Oral presentation

Pathological variants in TOP3A cause distinct disorders of mitochondrial and nuclear genome stability

Direnis Erdinc1, Alejandro Rodríguez-Luis2,3, Mahmoud R. Fassad2,4, Sarah Mackenzie5, Christopher M. Watson6,7, Sebastian Valenzuela1, Xie Xie1, Katja E. Menger2,3, Kate Sergeant8, Kate Craig2,9, Sila Hopton2,9, Gavin Falkous2,9, Joanna Poulton10, Hector Garcia-Moreno11, Paola Giunti11, Carlos A. de Moura Aschoff12, Jonas A. Morales Saute12,13,14, Amelia J. Kirby15, Camilo Toro16, Lynne Wolfe16, Danica Novacic16, Lior Greenbaum17,18,19, Aviva Eliyahu17,19, Ortal Barel20, Yair Anikster19,21, Robert McFarland2,4, Gráinne S. Gorman2,4, Andrew M. Schaefer2,9, Claes M. Gustafsson1,22, Robert W. Taylor2,4,9, Maria Falkenberg1, Thomas J Nicholls1

1: Department of Medical Biochemistry and Cell Biology, University of Gothenburg, P.O. Box 440, SE-405 30 Gothenburg, Sweden; 2: Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK; 3: Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK; 4: Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK; 5: The Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK; 6: North East and Yorkshire Genomic Laboratory Hub, Central Lab, St. James's University Hospital, Leeds, UK; 7: Leeds Institute of Medical Research, University of Leeds, St. James's University Hospital, Leeds, UK; 8: Oxford Genetics Laboratories, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; 9: NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, NE1 4LP, UK; 10: Nuffield Department of Women’s & Reproductive Health, The Women's Centre, University of Oxford, Oxford, UK; 11: Ataxia Centre, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK; 12: Medical Genetics Service, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil; 13: Department of Internal Medicine, Universidade Federal do Rio Grande do Sul - Porto Alegre, Brazil; 14: Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul - Porto Alegre, Brazil; 15: Department of Pediatrics, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA; 16: Undiagnosed Diseases Program, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA; 17: The Danek Gertner Institute of Human Genetics, Sheba Medical Center, Tel Hashomer, Israel; 18: The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Israel; 19: The Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; 20: Genomics Unit, The Center for Cancer Research, Sheba Medical Center, Israel; 21: Metabolic Disease Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel; 22: Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden



Oral presentation

The role of replicative exonucleases in mitochondrial DNA replication and degradation

Christian D Gonzalez, Nadee Nissanka, Carlos T Moraes

University of Miami Miller School of Medicine, United States of America



Flash Talk

Processing of mitochondrial RNA in health and disease: the role of FASTKD5.

Hana Antonicka1, James B. Gibson2, Eric A. Shoubridge1

1: The Neuro & McGill University, Montreal, Quebec, Canada; 2: Dell School of Medicine, University of Texas at Austin, Austin, TX, USA



Flash Talk

Mechanisms of mtDNA maintenance and segregation in the female germline

Laura Kremer1, Lyuba Bozhilova2,3, Diana Rubalcava-Garcia1, Roberta Filograna1, Mamta Upadhyay1, Camilla Koolmeister1, Patrick Chinnery2,3, Nils-Göran Larsson1

1: Karolinska Institutet, Stockholm, Sweden; 2: MRC Mitochondrial Biology Unit, Cambridge, United Kingdom; 3: Department of Clinical Neurosciences, University of Cambridge, United Kingdom



Flash Talk

The human Mitochondrial mRNA Structurome reveals Mechanisms of Gene Expression in Physiology and Pathology

Antoni Barrientos1, Conor Moran1, Amir Brivanlou2, Flavia Fontanesi1, Silvi Rouskin2

1: University of Miami, United States of America; 2: Harvard Medical School, United States of America

10:45am
-
11:00am
Coffee Break
Location: Bologna Congress Center
11:00am
-
12:45pm
Session 2.2: Clinical 1: from new genes to old and novel phenotypes
Location: Bologna Congress Center - Sala Europa
Chair: Agnes Rotig
Chair: Daniele Ghezzi
Invited Speakers: R. Horvath; H. Prokisch
 
Invited

The role of mitochondria in neuromuscular diseases

Rita Horvath

Cambridge-UK, United Kingdom



Invited

Innovative approaches for the molecular diagnosis of mitochondrial disorders

Holger Prokisch

Technical University Munich Institute of Human Genetics



Oral presentation

Specialist multidisciplinary input maximises rare disease diagnoses from whole genome sequencing

William L Macken1,2, Micol Falabella1, Caroline McKittrick1, Chiara Pizzamiglio1,2, Rebecca Ellmers3, Kelly Eggleton3, Cathy E. Woodward2,3, Yogen Patel2,3, Robyn Labrum2,3, Genomics England Research Consortium9, Rahul Phadke4, Mary M. Reilly1, Catherine DeVille5, Anna Sarkozy4, Emma Footitt6, James Davison6,7, Shamima Rahman6,8, Henry Houlden1, Enrico Bugiardini1,2, Rosaline Quinlivan1,2,4, Michael G. Hanna1,2, Jana Vandrovcova1, Robert D.S. Pitceathly1,2

1: Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK; 2: NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK; 3: Neurogenetics Unit, Rare and Inherited Disease Laboratory, North Thames Genomic Laboratory Hub, London, UK; 4: Dubowitz Neuromuscular Centre, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK; 5: Department of Neurosciences, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK; 6: Metabolic Unit, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK; 7: National Institute for Health and Care Research Great Ormond Street Hospital Biomedical Research Centre, London, UK; 8: Mitochondrial Research Group, UCL Great Ormond Street Institute of Child Health, London, UK; 9: Genomics England, One Canada Square London, UK



Oral presentation

Biallelic variants in MCAT in an infant with lactic acidosis, lipoylation disorder, and early death

Melanie T. Achleitner1, Maja Hempel2,3, Konstantinos Tsiakas4, René G. Feichtinger1, Saskia B. Wortmann1,5, René Santer4, Johannes A. Mayr1

1: University Children's Hospital, Paracelsus Medical University, Salzburg, Austria; 2: Institute of Human Genetics, University Medical Center Eppendorf, Hamburg, Germany; 3: Current address: Institute of Human Genetics, University Hospital Heidelberg, Heidelberg, Germany; 4: Department of Pediatrics, University Medical Center Eppendorf, Hamburg, Germany; 5: Amalia Children’s Hospital, Radboudumc, Nijmegen, The Netherlands.



Oral presentation

Biallelic PTPMT1 variants impair cardiolipin metabolism and cause mitochondrial myopathy and developmental regression

Micol Falabella1, Chiara Pizzamiglio1,2, Luis Carlos Tabara3, Ece Sonmezler4, Benjamin Munro5, William L. Macken1,2, Shanti Lu1, Lisa Tilokani3, Padraig J. Flannery6,7, Nina Patel7,8, Simon A. S. Pope7,8, Simon J. R. Heales7,8, Jana Vandrovcova1, Henry Houlden1, Robert W. Taylor9, Cathy E. Woodward6, Robyn Labrum6, Genomics England Research Consortium10, Semra Hiz11, Maha S. Zaki12, Efstathia Chronopoulou13, Germaine Pierre13, Reza Maroofian1, Michael G. Hanna1,2, Yavuz Oktay4,14,15, Rita Horvath5, Julien Prudent3, Robert D. S. Pitceathly1,2

1: Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK; 2: NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK; 3: Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge UK; 4: Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, Turkey; 5: Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK; 6: Neurogenetics Unit, Rare and Inherited Disease Laboratory, North Thames Genomic Laboratory Hub, London, UK; 7: Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, UK; 8: Neurometabolic Unit, The National Hospital for Neurology and Neurosurgery, London, UK; 9: Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK; NHS Highly Specialised Service for Rare Mitochondrial Disorders of Adults and Children, Newcastle University, Newcastle upon Tyne, UK; 10: Genomics England, London, UK; 11: Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey; 12: Clinical Genetics Department, Human Genetics and Genome Research Division, National Research Centre, Cairo, Egypt; 13: Department of Inherited Metabolic Disease, Division of Women's and Children's Services, University Hospitals Bristol NHS Foundation Trust, Bristol, UK; 14: Izmir Biomedicine and Genome Center, Izmir, Turkey; 15: Department of Medical Biology, Faculty of Medicine, Dokuz Eylül University, Izmir Turkey



Flash Talk

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

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



Flash Talk

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

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



Flash Talk

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

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



Flash Talk

Guanylate kinase 1 deficiency: a novel and potentially treatable form of mitochondrial DNA depletion/deletions syndrome

Agustin Hidalgo-Gutierrez1, Jonathan Shintaku1, Eliana Barriocanal-Casado1, Russ Saneto2, Javier Ramon4,7, Gloria Garrabou4,5, Frederic Tort3,4, Jose Cesar Milisenda6, Laura Gort3,4, Alba Pesini1, Saba Tadesse1, Mary-Claire King8, Ramon Marti4,7, Antonia Ribes3,4, Michio Hirano1

1: Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA; 2: Seattle Children’s Hospital, Seattle, WA, USA; 3: Section of Inborn Errors of Metabolism-IBC. Department of Biochemistry and Molecular Genetics. Hospital Clinic de Barcelona-IDIBAPS, Barcelona.; 4: Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Barcelona; 5: Muscle Research and Mitochondrial Function Lab, Cellex - IDIBAPS. Faculty of Medicine and Health Science - University of Barcelona (UB), Barcelona.; 6: Department of Internal Medicine, Hospital Clínic of Barcelona.; 7: Vall d’Hebron Research Institute, Autonomous University of Barcelona, Barcelona, Spain.; 8: Department of Genome Sciences, University of Washington, Seattle, WA, U.S.A.

12:45pm
-
1:45pm
Lunch
Location: Bologna Congress Center - Sala Europa
1:45pm
-
3:30pm
Session 2.3: Modelling pathogenic mechanisms: OXPHOS, metabolic rewiring and tissue specificity
Location: Bologna Congress Center - Sala Europa
Chair: Cristina Ugalde
Chair: Giovanni Manfredi
 
Invited

Metabolic adaptations of respiratory chain organization and function

Erika Fernandez-Vizarra1,2

1: Department of Biomedical Sciences, University of Padova, Italy; 2: Veneto Institute of Molecular Medicine, Padova, Italy



Invited

Pluripotent stem cells and brain organoids for drug discovery of mitochondrial diseases

Alessandro Prigione

Heinrich Heine University, Düsseldorf, Germany



Oral presentation

High-throughput single cell analysis reveals progressive mitochondrial DNA mosaicism developing throughout life

Angelos Glynos1,2, Lyuba V. Bozhilova1,2, Michele Frison1,2, Stephen P. Burr1,2, James B. Stewart3, Patrick F. Chinnery1,2

1: Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; 2: Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; 3: Biosciences Institute, Faculty of Medical Sciences, Wellcome Centre for Mitochondrial Research, Newcastle University, Newcastle upon Tyne, UK



Oral presentation

A coordinated multiorgan metabolic response contributes to human mitochondrial myopathy.

Guido Primiano3, Nneka Southwell1, Viraj Nadkarni1, Emelie Beattie1, Maria Lucia Valentino4, Valerio Carelli4, Serenella Servidei3, Giovanni Manfredi1, Qiuying Chen2, Marilena D'Aurelio1

1: Weill Cornell Medicine, Brain and Mind Research Institute, New York, NY; 2: Weill Cornell Medicine, Department of Pharmacology, New York, NY; 3: Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy; Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy; 4: IRCCS, Institute of Neurological Sciences of Bologna, Bellaria Hospital, Bologna, Italy; Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy



Oral presentation

Succinylation as a novel pathogenic mechanism in a children's mitochondrial brain disease

Pieti Elonkirjo1, Tuomas Kukkonen1, Juan Liu2, Jason W. Locasale2, Sami Jalil1, Birgit Schilling3, Eric Verdin3,4, Marco Reidelbach5, Outi Haapanen5, Vivek Sharma5,6, Elsebet Oestergaard7, Rosalba Carrozzo8, Berge Minassian9,10, Anu Suomalainen1

1: STEMM, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland; 2: Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA; 3: Buck Institute for Research on Aging, Novato, CA 94945, USA; 4: Gladstone Institutes and University of California, San Francisco, CA 94158, USA; 5: Department of Physics, University of Helsinki, Finland; 6: HiLIFE Institute of Biotechnology, University of Helsinki, Finland; 7: Department of Clinical Genetics, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark; 8: Unit of Cellular Biology and Mitochondrial Diseases, “Bambino Gesù” Children's Hospital, IRCCS, Rome, Italy; 9: Program in Genetics and Genome Biology, The Hospital for Sick Children, Institute of Medical Science University of Toronto, Toronto, Ontario, Canada; 10: Division of Neurology, Department of Pediatrics, University of Texas Southwestern, Dallas, TX, USA



Flash Talk

The levels and activation state of the pyruvate dehydrogenase complex modulate the SCAFI-dependent organization of the mitochondrial respiratory chain

Sandra Lopez-Calcerrada1, Ana Sierra-Magro1, Erika Fernández-Vizarra2, Cristina Ugalde1,3

1: Instituto de Investigación Hospital 12 de Octubre, Madrid 28041, Spain; 2: Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; 3: Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), U723, Madrid, Spain



Flash Talk

Oxphos deficiency indicates novel functions for the mitochondrial protein import subunit tim50

Jordan J Crameri1, Catherine S Palmer1, David Coman2, David A Stroud1, David R Thorburn3,4,5, Ann E Frazier3,4, Diana Stojanovski1

1: Department of Biochemistry and Pharmacology and the Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, 3010, Australia; 2: Queensland Children’s Hospital, Department of Metabolic Medicine, South Brisbane, Brisbane, Queensland, 4001, Australia; 3: Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Victoria, 3052, Australia; 4: Department of Paediatrics, University of Melbourne, Melbourne, Victoria, 3052, Australia; 5: Victorian Clinical Genetics Services, Royal Children’s Hospital, Melbourne, Victoria, 3052, Australia



Flash Talk

Microproteins in metabolic regulation

Jiemin Nah1, Baptiste Kerouanton1, David Robinson2, Kyle Dunlap3, Pooja Sridnivasan1, Sonia Chothani1, Greg Ducker3, Owen Rackham4, David Stroud2, Lena Ho1

1: Duke-NUS Medical School, Singapore; 2: University of Melbourne, Australia; 3: University of Utah, USA; 4: University of Southampton, UK

3:30pm
-
3:50pm
Industry Workshop: Abliva AB
Location: Bologna Congress Center - Sala Europa
3:30pm
-
4:30pm
Tea Break and poster session
Location: Bologna Congress Center
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
 

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

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

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

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

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

1: Mitochondrial Medicine Frontier Program, Division of Human Genetics, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA; 2: Illumina Laboratory Services, Illumina Inc., San Diego, CA; 3: Center for Personalized Medicine, Department of Pathology & Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA; 4: Keck School of Medicine, University of Southern California, Los Angeles, CA; 5: Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; 6: Genetics 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

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

1: Dino Ferrari Center, University of Milan, Italy; 2: IRCCS Cà Granda Ospedale Maggiore Policlinico Milan, Italy; 3: ASST 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

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

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

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

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

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

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

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

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

1: Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, Italy; 2: Department of Medicine, Surgery, and Health Sciences, University of Trieste, Trieste, Italy; 3: Unit 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

1: Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta; 2: Department 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

1: IRCCS, Istituto delle Scienze Neurologiche di Bologna, Italy; 2: Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy; 3: Department 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

1: IRCCS Istituto Giannina Gaslini, Genoa; 2: IRCCS Fondazione Stella Maris, Calambrone (PI); 3: IRCCS 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

1: IRCCS Institute of Neurological Sciences of Bologna, Italy; 2: Department 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

1: IRCCS Istituto delle Scienze Neurologiche di Bologna, Italy; 2: Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Italy; 3: Ospedale Oftalmico Roma, Rome, Italy; 4: Department 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

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

1: Maria Sklodowska-Curie, Medical Academy in Warsaw, Poland; 2: MedGen Medical Center, Warsaw, Poland; 3: Institute 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

1: IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; 2: Dino 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

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

1: Bambino Gesù Children Hospital, Italy; 2: UCL Queen Square Institute of Neurology; 3: Université 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

1: Hospital Municipal Dr. José de Carvalho, Brazil; 2: Escola 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

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

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

1: University of Cape Town, Cape Town, South Africa; 2: National Health Laboratory Sevices, South Africa; 3: Red 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

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

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

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

1: Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden, Sweden; 2: Centre 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

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

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

1: Department of Genetics, Translational Metabolic Laboratory, Radboudumc, Nijmegen, The Netherlands.; 2: Department 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

1: Department of Biology, University of Padova, Italy; 2: Department 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

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

1: Secció d'Errors Congènits del Metabolisme-IBC, Servei de Bioquímica i Genètica Molecular, Hospital Clínic de Barcelona, IDIBAPS, CIBERER, Barcelona, Spain; 2: Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany; 3: CNAG-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

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

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

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

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

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

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

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

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

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

1: Hereditary 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; 2: Department of Internal Medicine, Hospital Clinic of Barcelona, Barcelona, Spain; 3: CIBERER— Spanish Biomedical Research Centre in Rare Diseases, Madrid, Spain; 4: Department 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

1: Research centre for medical genetics, Russian Federation; 2: Morozov'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

1: LUCA Science, Japan; 2: Biological 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

1: Pharmaceutical Sciences Laboratory, Åbo Akademi University, Finland; 2: Turku 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

1: Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Sweden; 2: Max-Planck Institute of Biochemistry, Martinsried, Germany; 3: Metabolomics 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

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

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

1: Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany; 2: German Center for Cardiovascular Research (DZHK), partner site Göttingen, Germany; 3: Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany; 4: Research 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

1: Heinrich Heine University Düsseldorf, Germany; 2: IUF- 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

1: Wellcome Centre for Mitochondrial Research, Newcastle University, United Kingdom; 2: Metabolism 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

1: Children's Hospital of Philadelphia, USA; 2: University of Pennsylvania, USA; 3: Boston University, USA; 4: National 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

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

1: Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH; 2: NHS 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

1: IMol Polish Academy of Sciences, Poland; 2: ReMedy 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

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

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

1: Columbia University, USA; 2: Centro 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

1: MITOVASC Université d'Angers, France; 2: Departments of Biochemistry and Molecular Biology, University Hospital Angers, Angers, France; 3: Laboratoire 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

1: National Institute of Chemical Physics and Biophysics, Estonia; 2: Clinic 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

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

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

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

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



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

Janne Purhonen1,2, Jukka Kallijarvi1,2

1: Folkhalsan Research Center, Finland; 2: Stem 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

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

4:30pm
-
6:00pm
Session 2.4: New technological developments and OMICS
Location: Bologna Congress Center - Sala Europa
Chair: Holger Prokisch
Chair: Leonid Sazanov
Invited Speaker: :S. Churchman; :H. Hillen
 
Invited

Decoding the regulatory principles of mitochondrial DNA: packaging, expression, and impact on cellular metabolism

L. Stirling Churchman

Harvard Medical School, United States of America



Invited

Mechanisms of mitochondrial RNA biogenesis in health and disease

Hauke Hillen1,2

1: Department of Cellular Biochemistry, University Medical Center Göttingen, Germany; 2: Research Group Structure and Function of Molecular Machines, Max-Planck-Institute for Multidisciplinary Sciences Göttingen, Germany



Oral presentation

Disruption of mitochondrial function induces cell lineage-specific compensatory transcriptional responses during early embryonic development

Stephen P. Burr1,2, Florian Klimm1,2,3, Angelos Glynos1,2, Malwina Prater1,2,4, Maria Falkenberg5, Michal Minczuk2, James B. Stewart6,7, Patrick F Chinnery1,2

1: Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; 2: Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; 3: Novo Nordisk Research Centre Oxford, Innovation Building, University of Oxford, Old Road Campus, Oxford, UK; 4: Functional Genomics Centre, Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK; 5: Department of Medical Biochemistry and Cell Biology, University of Gothenburg, PO Box 440, Gothenburg 405 30, Sweden; 6: Max Planck Institute for Biology of Ageing, Cologne, Germany; 7: Biosciences Institute, Faculty of Medical Sciences, Wellcome Centre for Mitochondrial Research, Newcastle University, Newcastle upon Tyne, UK



Oral presentation

Single-cell multi-omics reveals dynamics of purifying selection of pathogenic mitochondrial DNA across human immune cells

Caleb A. Lareau1,2,3,4,5, Sonia M. Dubois5, Frank A. Buquicchio1, Yu-Hsin Hsieh6,7, Kopal Garg4,5, Pauline Kautz6,7,8, Lena Nitsch6,7,9, Samantha D. Praktiknjo6,7, Patrick Maschmeyer6,7, Jeffrey M. Verboon4,5, Jacob C. Gutierrez1, Yajie Yin1, Evgenij Fiskin4, Wendy Luo4, Eleni Mimitou10,17, Christoph Muus4,11, Rhea Malhotra4, Sumit Parikh12, Mark D. Fleming13, Lena Oevermann14, Johannes Schulte14, Cornelia Eckert14, Anshul Kundaje3,15, Peter Smibert10,18, Ansuman T. Satpathy1,2, Aviv Regev4,16,19, Vijay Sankaran4,5, Suneet Agarwal5, Leif S. Ludwig4,5,6,7

1: Department of Pathology, Stanford University, Stanford, CA 94305, USA; 2: Parker Institute of Cancer Immunotherapy, San Francisco, CA 94129, USA; 3: Department of Genetics, Stanford University, Stanford, CA 94305, USA; 4: Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; 5: Division of Hematology / Oncology, Boston Children’s Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; 6: Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; 7: Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), 10115 Berlin, Germany; 8: Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany; 9: Department of Biology, Chemistry, Pharmacy, Freie Universität Berlin, Berlin, Germany; 10: Technology Innovation Lab, New York Genome Center, New York, NY 10013, USA; 11: Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02134, USA; 12: Center for Pediatric Neurosciences, Mitochondrial Medicine, Cleveland Clinic, Cleveland, OH 44195, USA; 13: Department of Pathology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; 14: Department of Pediatric Oncology, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum, 13353 Berlin, Germany; 15: Department of Computer Science, Stanford University, Stanford, CA 94305, USA; 16: Department of Biology and Koch Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; 17: Current address: Immunai, New York, NY 10114, USA; 18: Current address: 10x Genomics, San Francisco, CA 94111, USA; 19: Current address: Genentech, San Francisco, CA 94080, USA



Flash Talk

Quantifying mitochondrial proteome remodeling during macrophage polarization

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

University of Lausanne, Switzerland



Flash Talk

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

Janne Purhonen1,2, Jukka Kallijarvi1,2

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



Flash Talk

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

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

6:00pm
-
7:00pm
Poster session
Location: Bologna Congress Center
Session topics:
- Modelling pathogenic mechanisms: OXPHOS, metabolic rewiring and tissue specificity
 

Maintenance on mitochondrial complexes ensures bioenergetic function in differentiated cells

Ilka Wittig1, Julian Heidler1, Heiko Giese2, Ralf P. Brandes1

1: Institute for Cardiovascular Physiology, Goethe University Frankfurt, Germany; 2: Molecular Bioinformatics, Goethe University, Frankfurt, Germany



Investigating pathogenicity and tissue-specificity of mitochondrial aminoacyl-tRNA synthetase defects AARS2, EARS2 and RARS2 in neurons

Oliver Podmanicky, Fei Gao, Denisa Hathazi, Rita Horvath

Department of Clinical Neurosciences, University of Cambridge, United Kingdom



Mutations in Coq2 leads to severe developmental delay and early death in both zebrafish and mouse

Julia Corral-Sarasa1, Sergio López-Herrador2, Juan M. Martínez-Gálvez1,3, Pilar González-García2, Laura Jiménez-Sánchez1, Mª Elena Díaz-Casado1,2, Luis C. López1,2

1: Ibs.Granada, Granada, Spain; 2: Physiology Department, Biomedical Research Center, University of Granada, Granada, Spain; 3: Biofisika Institute (CSIC,UPV-EHU) and Department of Biochemistry and Molecular Biology, University of Basque Country, Leioa, Spain



Pathogenic variants of the mitochondrial metallochaperone SCO1 result in a severe, combined COX and copper deficiency that causes a dilated cardiomyopathy in the murine heart.

Sampurna Ghosh1, Scot C. Leary1, Paul A. Cobine2

1: University of Saskatchewan, Canada; 2: Auburn University



Tissue-specific adaptation of stress responses upon COX10 deficiency

Lea Isermann1,2, Milica Popovic1,2, Ming Yang1,2, Christian Frezza1,2, Aleksandra Trifunovic1,2

1: CECAD Research Center, Germany; 2: Institute for Mitochondrial Diseases and Aging, Medical Faculty, University of Cologne



Using iPSC-derived neurons to unravel the pathomechanisms of Leber’s hereditary optic neuropathy

Camille Peron1, Alessandra Maresca2, Angelo Iannielli3,4, Alberto Danese5, Simone Patergnani5, Danara Ormanbekova2, Andrea Cavaliere1, Carlotta Giorgi5, Paolo Pinton5,6, Vania Broccoli3,4, Valerio Carelli2,7, Valeria Tiranti1

1: Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy; 2: IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy; 3: Division of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy; 4: National Research Council (CNR), Institute of Neuroscience, Milan, Italy; 5: Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies, University of Ferrara, 44121 Ferrara, Italy; 6: Maria Cecilia Hospital, GVM Care & Research, 48033, Cotignola, Ravenna, Italy; 7: Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy



Stem cell modelling of mitochondrial disease-linked cardiomyopathy

Ann E. Frazier1,2, Yau Chung Low1,2, Cameron L. McKnight1,2, Linden Muellner-Wong1,3, Hayley L. Pointer1, Nikeisha Caruana3, Jordan J. Crameri3, Luke E. Formosa4, Yilin Kang3, Thomas D. Jackson3, Alison G. Compton1,2,5, Michael T. Ryan4, Andrew G. Elefanty1,2,6, Enzo R. Porrello1,6,7,8, David A. Stroud1,3,5, David A. Elliott1,2,6,7, Diana Stojanovski3, David R. Thorburn1,2,5

1: Murdoch Children’s Research Institute, The Royal Children's Hospital, Melbourne, VIC, Australia; 2: Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia; 3: Department of Biochemistry and Pharmacology and The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, Australia; 4: Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia; 5: Victorian Clinical Genetics Services, The Royal Children’s Hospital, Melbourne, VIC, Australia; 6: The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, VIC, Australia; 7: Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, The Royal Children's Hospital, Melbourne, VIC, Australia; 8: Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Melbourne, VIC, Australia



Biochemical and computational approaches to dissect the effect of MT-CYB pathogenic mutations on respiratory chain activity and assembly

Gaia Tioli, Francesco Musiani, Luisa Iommarini, Anna Maria Porcelli, Anna Maria Ghelli

Department of Pharmacy and Biotechnology, University of Bologna, Italy



Exploring the assembly and maintenance of mitochondrial complex I by complexome profiling-based approaches

Alfredo Cabrera-Orefice1,2, Ilka Wittig1

1: Institute for Cardiovascular Physiology, Goethe University Frankfurt, Germany; 2: Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands



Functional involvement of actin-binding Gelsolin on mitochondrial Oxphos dysfunction

María Illescas, Ana Peñas, Cristina Ugalde

Fundación Hospital 12 de Octubre, Spain



In vivo role of respiratory complex I NDUFA10 subunit in dNTP homeostasis

Andrea Férriz Gordillo1, David Molina-Granada1,2, Javier Ramón1,2, Izaskun Izagirre-Urizar1, Marina Singla-Manau1, Maria Jesús Melià1,2, Antoni Ruiz-Vicaria1, Javier Torres-Torronteras1,2, Mònica Zamora3, Michael T. Ryan4, Cristina Ugalde2,5, Josep Antoni Villena6, Ramon Martí1,2, Yolanda Cámara1,2

1: Research Group on Neuromuscular and Mitochondrial Disorders, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain; 2: Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain; 3: BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, Barcelona 08028, Spain. and Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona 08036, Spain.; 4: Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia; 5: Instituto de Investigación, Hospital Universitario 12 de Octubre, Avda. de Córdoba s/n, 28041 Madrid, Spain.; 6: Laboratory of Metabolism and Obesity, Vall d'Hebron - Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain; CIBERDEM, CIBER on Diabetes and Associated Metabolic Diseases, Instituto de Salud Carlos III, Barcelona, Spain



Modeling POLRMT pathogenic variants in the mouse

David Alsina1,2, Roberta Filograna1,2, Rodolfo García-Villegas1,2, Camilla Koolmeister1,2, Nils-Göran Larsson1,2,3

1: Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden; 2: Max Planck Institute Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden; 3: Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden



The role of the CCR4 family member ANGEL1 in the expression of mitochondrial-targeted proteins

Kai Chang1, Paula Clemente1, Joyce Noble1, Björn Reinius1, Anna Wedell1,2, Christoph Freyer1,2, Anna Wredenberg1,2

1: Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden; 2: Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden



Tissue-specific bioenergetics in mouse models of mitochondrial disease

Valeria Balmaceda1, Timea Komoldi2, Massimo Zeviani1, Erich Gnaiger3, Anthony L. Moore4, Erika Fernandez Vizarra1, Carlo Viscomi1

1: Università di Padova; 2: Semmelweis University; 3: Universität Innsbruck; 4: University of Sussex



Yeast as a tool to investigate variants in mtARS genes associated with mitochondrial diseases

Sonia Figuccia1, Camilla Ceccatelli Berti1, Andrea Legati2, Rossella Izzo2, Alessia Nasca2, Daniele Ghezzi2,3, Paola Goffrini1

1: Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy; 2: Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy; 3: Department of Medical Physiopathology and Transplantation, University of Milan, Milan, Italy



A mutation in mouse mt-Atp6 gene induces respiration defects and opposed effects on the cell tumorigenic phenotype

Raquel Moreno-Loshuertos1, Nieves Movilla1, Joaquín Marco-Brualla2, Ruth Soler-Agesta1, Patricia Ferreira1, José Antonio Enríquez3, Patricio Fernández-Silva1

1: University of Zaragoza, Spain; 2: University of Zaragoza, Peaches Biotech Group, Spain; 3: Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Spain



A systemic Muscle-WAT crosstalk progressively depletes protein and fat stores aggravating mitochondrial myopathy.

Nneka Southwell1, Guido Primiano3, Emelie Beattie1, Nicola Rizzardi1, Serenella Servidei3, Giovanni Manfredi1, Qiuying Chen2, Marilena D'Aurelio1

1: Weill Cornell Medicine, Brain and Mind Research Institute, New York, NY; 2: Weill Cornell Medicine, Department of Pharmacology, New York, NY; 3: Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy; Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy.



A novel mitochondrial assembly factor RTN4IP1 has an essential role in the final stages of Complex I assembly

Monika Oláhová1,2, Jack J. Collier1,3, Rachel M. Guerra4, Juliana Heidler5, Kyle Thompson1, Robert N. Lightowlers1, Zofia M.A. Chrzanowska-Lightowlers1, Ilka Wittig5, David J. Pagliarini4, Robert W. Taylor1

1: Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; 2: Department of Applied Sciences, Faculty of Health & Life Sciences, Northumbria University, Newcastle upon Tyne, UK; 3: Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada; 4: Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA; 5: Functional Proteomics Group, Institute for Cardiovascular Physiology, Goethe University Frankfurt, 60590, Frankfurt am Main, Germany



ETFDH supports OXPHOS efficiency in skeletal muscle by regulating coenzyme Q homeostasis

Beñat Salegi Ansa1, Juan Cruz Herrero Martín1, José M. Cuezva1,2,3,4, Laura Formentini1,2,3,4

1: Department of Molecular Biology, Centro de Biología Molecular "Severo Ochoa" (CBMSO-UAM-CSIC), Madrid, Spain; 2: Instituto Universitario de Biología Molecular (IUBM), Autonomous University of Madrid, Madrid, Spain; 3: Centro de Investigación Biomédica en red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; 4: Instituto de Investigación Hospital 12 de octubre, i+12, Universidad Autónoma de Madrid, Madrid, Spain



Metabolic rewiring as an adaptive mechanism in COX null cells

Guillermo Puertas-Frias1,2, Kristýna Čunátová1,2, Petr Pecina1, Marek Vrbacký1, Lukáš Alán1, Tomáš Čajka3, Josef Houštěk1, Tomáš Mráček1, Alena Pecinová1

1: Department of Bioenergetics, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic; 2: Faculty of Science, Charles University, 12800 Prague, Czech Republic; 3: Laboratory of Translational Metabolomics, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic



Metabolic rewiring due to progressive increase in mtDNA mutation heteroplasmy reveals markers of disease severity

Karin Terburgh1, Marianne Venter1, Jeremie Z Lindeque1, Emi Ogasawara2, Mirian C H Janssen3, Jan A M Smeitink3, Kazuto Nakada4, Roan Louw1

1: North-West University, South Africa; 2: Osaka University, Japan; 3: Radboud University Medical Center, Netherlands; 4: University of Tsukuba, Japan



Novel or rare AIFM1 pathogenic variants: their impact on mitochondrial metabolism and clinical manifestation in eight patients, including 3 girls

Tereza Rakosnikova1, Jan Kulhanek1, Martin Reboun1, Hana Stufkova1, Lenka Dvorakova1, Dagmar Grecmalova2, Pavlina Plevova2, Tomas Honzik1, Hana Hansikova1, Jiri Zeman1, Marketa Tesarova1

1: Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague; 2: Institute of Molecular and Clinical Pathology and Medical Genetics, University Hospital Ostrava



Pathological molecular mechanisms underlying COA8 loss of function

Kristyna Cunatova1,2, Michele Brischigliaro1,2, Alfredo Cabrera-Orefice3, Cinzia Franchin1, Jimin Pei4, Marco Roverso5, Sara Bogialli5, Qian Cong4, Giorgio Arrigoni1, Susanne Arnold3,6, Carlo Viscomi1,2, Massimo Zeviani2,7, Erika Fernández-Vizarra1,2

1: Department of Biomedical Sciences, University of Padova, Padova, Italy; 2: Veneto Institute of Molecular Medicine, Padova, Italy; 3: Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands; 4: University of Texas Southwestern Medical Center, Dallas, TX, USA; 5: Department of Chemical Sciences, University of Padova, Padova, Italy; 6: Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; 7: Department of Neurosciences, University of Padova, Padova, Italy



Retinal pathophysiology characterisation of the novel mitochondrial heteroplasmy mouse model

Lucia Luengo-Gutierrez1, Keira Turner1, James B Stewart2, Patrick Yu-Wai-Man1, Michal Minczuk1

1: University of Cambridge, United Kingdom; 2: Newcastle University, United Kingdom



Impaired spermatogenesis driven by mitochondrial dysfunction and ferroptosis in primary spermatocytes in a mouse model of Leigh syndrome

Enrico Radaelli1, Charles-Antoine Assenmacher1, Esha Banerjee1, Florence Manero2, Salim Khiati3, Anais Girona3, Guillermo Lopez-Lluch4, Placido Navas4, Marco Spinazzi3,5

1: University of Pennsylvania,USA; 2: University of Angers, SFR ICAT, SCIAM, 49000 Angers, France; 3: MITOLAB, University of Angers, INSERM U1083, France; 4: Pablo de Olavide University, Spain; 5: Neuromuscular Reference Center CHU Angers, France



Mitophagy dysfunction in mitochondrial myopathy and therapy by mitophagy activator CAP1902

Takayuki Mito1, Amy E. Vincent2, Julie Faitg2, Kathleen Rodgers3, Kevin Gaffney3, Thomas G. McWilliams1, Orian Shirihai4, Anu Suomalainen1

1: STEMM Research Program, Biomedicum Helsinki, Faculty of Medicine, University of Helsinki, Helsinki, Finland; 2: Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; 3: Department of Pharmacology, Center for Innovations in Brain Science, University of Arizona, Tucson, AZ, USA; 4: Department of Medicine, Endocrinology, and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA



Mtfp1 controls oxidative phosphorylation and cell death in liver disease

Cecilia Patitucci1, Juan Diego Hernández-Camacho1, Elodie Vimont1, Etienne Kornobis2, Thibault Chaze3, Quentin Giai Gianetto3,4, Mariette Matondo3, Anastasia Gazi5, Ivan Nemanyy6, Daniella Hock7, Erminia Donnarumma1, Timothy Wai1

1: Institut Pasteur, Mitochondrial Biology Group, CNRS UMR 3691, Université Paris Cité, Paris, France.; 2: Institut Pasteur, Biomics Technological Platform, Université Paris Cité, Paris, France.; 3: Institut Pasteur, Bioinformatics and Biostatistics Hub, Université Paris Cité, Paris, France.; 4: Institut Pasteur, Proteomics Core Facility, MSBio UtechS, UAR CNRS 2024, Université Paris Cité, Paris, France.; 5: Institut Pasteur Ultrastructural Bio Imaging, UTechS, Université Paris Cité, Paris, France.; 6: Platform for Metabolic Analyses, SFR Necker, INSERM US24/CNRS UMS 3633, Paris, France.; 7: Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Australia.



Non-canonical function of succinate dehydrogenase assembly factor 2 (SDHAF2) during OXPHOS dysfunction

Kugapreethan Roopasingam1, Joanna Sacharz1, Tegan Stait2, Yau chung Low2,3, Ann E. Frazier2,3, David P. De souza4, David R. Thorburn2,3,5, David A. Stroud1,3,5

1: Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia; 2: Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia; 3: Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia; 4: Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia; 5: Victorian Clinical Genetics Services, Royal Children's Hospital, Parkville, VIC, Australia



NUAK1-dependent metabolic underpinnings of adult muscle stem cells

Ha-My Ly, Caroline Brun, Géraldine Meyer-Dihet, Julien Courchet, Rémi Mounier

Physiopathology and Genetics of Neurons and Muscles Laboratory, Institut NeuroMyoGène, Lyon, France



A novel approach to measure complex V ATP hydrolysis in frozen cell lysates and tissue homogenates

Lucia Fernandez del Rio1,2, Cristiane Benincá1,2, Frankie Villalobos1,2, Cynthia Shu1,2, Linsey Stiles1,2,3, Marc Liesa1,2,4,5, Ajit S. Divakaruni2,3, Rebeca Acin-Perez1,2, Orian S. Shirihai1,2,3,4

1: Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095 USA; 2: Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; 3: Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA; 4: Molecular & Cellular Integrative Physiology, University of California, Los Angeles, CA, 90095, USA.; 5: Institut de Biologia Molecular de Barcelona, IBMB-CSIC, Barcelona, Catalonia, 08028, Spain



OxPhos defects cause cell-autonomous and whole-body signs of hypermetabolism in cells and in patients with mitochondrial diseases

Gabriel Sturm1, Karan Kalpita1, Anna S Monzel1, Balaji Santhanam1, Tanja Taivassalo2, Céline Bris3, Atif Towheed1, Albert Higgins-Chen4, Meagan McManus5, Andres Cardenas6, Jue Lin7, Elissa Epel7, Shamima Rahman8, Jon Vissing9, Bruno Grassi10, Morgan Levine11, Steve Horvath11, Ronald G Haller12, Guy Lenaers3, Douglas C Wallace5, Marie-Pierre St-Onge1, Saeed Tavasoie1, Vincent Procaccio3, Brett A Kaufman13, Erin L Seifert14, Michio Hirano1, Martin Picard1

1: Columbia University Irving Medical Center, United States of America; 2: University of Florida, United States of America; 3: Angers University, UMR CNRS 6015 - INSERM U1083, MitoVasc Institute, Angers, France; 4: Yale University, United States of America; 5: University of Pennsylvania, United States of America; 6: Stanford University, United States of America; 7: University of California San Francisco, United States of America; 8: Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom; 9: University of Copenhagen, Denmark; 10: University of Udine, Italy; 11: Altos labs, United States of America; 12: University of Texas Southwestern Medical Center, United States of America; 13: University of Pittsburgh, United States of America; 14: Thomas Jefferson University, United States of America



Dysfunction of mitochondrial chaperone HSP60 triggers disruption of mitochondrial pathways activating multiple regulatory responses

Cagla Cömert1, Paula Fernandez-Guerra1, Kasper Kjær-Sørensen2, Jakob Hansen3, Jesper Just4,5, Jasper Carlsen1, Lisbeth Schmidt-Laursen2, Johan Palmfeldt1, Peter Bross1

1: Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; 2: Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark; 3: Department of Forensic Medicine, Aarhus University, Aarhus, Denmark; 4: Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark; 5: Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark



Generation of iPSCs derived neural progenitors and cardiomyocytes as cellular models to study the pathophysiology of Pearson Syndrome

Chiara Fasano1, Luca Sala2,3, Camille Peron1, Andrea Cavaliere1, Maria Nicol Colombo1, Valeria Tiranti1

1: Unit of Medical genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy; 2: Istituto Auxologico Italiano IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milan, Italy; 3: Department of Biotechnology and Biosciences, University of Milano - Bicocca, Milan, Italy



High aerobic exercise capacity predicts increased mitochondrial response to exercise training

Estelle Heyne1, Susanne Zeeb1, Luren G Koch2, Steven L Britton3, Torsten Doenst1, Michael Schwarzer1

1: Department of Cardiothoracic Surgery, University Hospital of Friedrich-Schiller-University Jena, Germany; 2: Department of Physiology and Pharmacology, University of Toledo, Toledo, OH, United States; 3: Department of Anesthesiology, University of Michigan, Ann Arbor, MI, United States



Investigating the role of LONP1 in heart and skeletal muscle metabolism

Franziska Baumann1, Dieu Hien Rozsivalova1, Katharina Senft1, Simon Geißen2, Aleksandra Trifunovic3

1: Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany; 2: Department III of Internal Medicine, Heart Center, University Hospital of Cologne, 50931 Cologne, Germany; 3: Center for Molecular Medicine Cologne (CMMC), 50931 Cologne, Germany



Mitochondrial dysfunction promotes liver fibrosis through the ACOT2-MCT6-OXCT1 axis.

Xiaoshan Zhou1, Sophie Curbo1, Wei Wang2, Xinling Li2, Jingyi Yan1, Yu Lei1, Raoul Kuiper3, Ujjwal Noegi1, Anna Karlsson1

1: Karolinska Institutet, Sweden; 2: Zhengzhou University, China; 3: Norwegian Veterinary Institute, Norway



PNC2 (SLC25A36) deficiency associated with the hyperinsulinism/hyperammonemia syndrome

Francesco Massimo Lasorsa1, Deborah Fratantonio2, Maher A. Shahroor3, Vito Porcelli1, Bassam Abu-Libdeh3, Orly Elpeleg4, Luigi Palmieri1

1: Università degli Studi di Bari Aldo Moro, Italy; 2: Libera Università Mediterranea Giuseppe Degennaro, Italy; 3: Department of Pediatrics and Genetics, Al Makassed Hospital and Al-Quds University, Palestine.; 4: Department of Genetics, Hadassah, Hebrew University Medical Center, Israel



Cultured neurons with CoQ10 deficiency reveal alterations of lipid metabolism

Alba Pesini1, Eliana Barriocanal-Casado1, Giacomo Monzio-Compagnoni2, Kleiner Giulio1, Agustin Hidalgo-Gutierrez1, Mohammed Bakkali3, Yashpal Singh Chhonker4, Saba Tadesse1, Delfina Larrea1, Daryl J Murry4, Caterina Mariotti5, Barbara Castellotti5, Luis Carlos Lopez3, Alesio Di Fonzo2, Estela Area-Gomez1, Catarina Quinzii1

1: Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, United States; 2: IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy; 3: Institute of Biotechnology, Biomedical Research Center (CIBM), Health Science Technological Park (PTS), University of Granada, Armilla, Granada, 18100, Spain; 4: Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, 986145 Nebraska Medical Center, Omaha, NE; 5: Unita` di Genetica delle Malattie Neurodegenerative e Metaboliche, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, 20126, Italy



An engineered variant of MECR reductase reveals indispensability of long-chain acyl-ACPs for mitochondrial respiration

M. Tanvir Rahman1, M. Kristian Koski2, Joanna Panecka-Hofman3,4, Werner Schmitz5, Alexander J. Kastaniotis1, Rebecca C. Wade4,6, Rik K. Wierenga1, J. Kalervo Hiltunen1, Kaija J. Autio1

1: Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland; 2: Biocenter Oulu, University of Oulu, Oulu, Finland; 3: Faculty of Physics, University of Warsaw, Warsaw, Poland; 4: Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), Heidelberg, Germany; 5: Department of Biochemistry and Molecular Biology, University of Würzburg, Würzburg, Germany; 6: Zentrum für Molekulare Biologie (ZMBH), DKFZ-ZMBH Alliance and Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Heidelberg, Germany



Antibiotics directly affect mitochondrial respiration

Judith Sailer1, Sabine Schmitt2, Hans Zischka1,3, Erich Gnaiger2

1: Technische Universität München, Germany; 2: Oroboros Instruments GmbH, Innsbruck, Austria; 3: Helmholtz Zentrum München, Germany



Can transmission of mitochondria over the species barrier promote climate change adaptation?

Kateryna Gaertner1, Craig Michell2, Riikka Tapanainen2, Steffi Goffart2, Sina Saari1, Manu Soininmäki2, Eric Dufour1, Jaakko Pohjoismäki2

1: Tampere University, Finland; 2: University of Eastern Finland



Developing an in vitro model to study the impact of the m.3243A>G mutation in iPSC-derived myofibers

Gabriel E. Valdebenito, Anitta R. Chacko, Michael R. Duchen

University College London, United Kingdom



Discordant phenotype in fibroblast cell lines generated from the same MELAS patient

Monica Moresco1, Valentina Concetta Tropeano1, Valentina Del Dotto2, Mariantonietta Capristo1, Claudio Fiorini1, Danara Ormanbekova1, Chiara La Morgia1,2, Maria Lucia Valentino1,2, Valerio Carelli1,2, Alessandra Maresca1

1: IRCCS Istituto delle Scienze Neurologiche di Bologna, Italy; 2: Department of Biomedical and Neuromuscular Sciences (DIBINEM), University of Bologna



Generation of a novel CoQ deficient mouse model to elucidate the role of COQ4

Eliana Barriocanal Casado, Agustin Hidalgo-Gutierrez, Alba Pesini, Catarina M Quinzii

Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA.



Perivascular adipose tissue remodeling impairs mitochondrial function in thermoneutral-housed rats

Amy C Keller1, Melissa M Henckel1, Leslie A Knaub1, Greg B Pott1, Georgia James2, Jane E-B Reusch1

1: University of Colorado/Rocky Mountain Regional VA Medical Center, United States of America; 2: Cornell College, United States of America



Temporal analysis of mitochondrial complexome profiling coupled to multi-omics analysis unveils implications of CIV remodelling in postnatal heart development

Milica Popovic, Aleksandra Trifunovic

University of Cologne, Germany



Mitochondrial dysfunction in immune cells leads to distinct transcriptome profile and improved immune competence in Drosophila

Yuliya Basikhina1, Laura Vesala1,2, Tea Tuomela1, Emilia Siukola1, Anssi Nurminen1, Pedro F. Vale3, Tiina S. Salminen1

1: Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; 2: Department of Molecular Biology, Umeå University, Umeå, Sweden; 3: Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, United Kingdom



Molecular mechanisms of extraocular muscle manifestation in mitochondrial myopathy

Swagat Pradhan, Takayuki Mito, Thomas McWilliams, Nahid Khan, Anu Suomalainen

STEMM, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland



Redox metabolites and transporters: Differential expression and ratios in specific Ndufs4 knockout mice organs.

Jeremie Zander Lindeque, Marthinus Theodorus Jooste, Daneël Nel, Belinda Fouché, Marianne Venter

Human Metabolomics, North-West University, South Africa



What makes folding of a mitochondrial protein dependent on the HSP60/HSP10 chaperone complex?

Peter Bross, Cagla Cömert, Paula Fernandez-Guerra, Johan Palmfeldt

Aarhus University and Aarhus University Hospital, Denmark



OXPHOS composition is altered in the FXNI151F mouse model of Friedreich Ataxia in a progressive and a tissue-specific way

Maria Pazos-Gil, Marta Medina-Carbonero, Arabela Sanz-Alcázar, Marta Portillo-Carrasquer, Fabien Delaspre, Elisa Cabiscol, Joaquim Ros, Jordi Tamarit

Dept. Ciències Mèdiques Bàsiques, Fac. Medicina, Universitat de Lleida. IRB Lleida.



Disease causing-Mfn2 mutations alter mitochondrial fusion and fission dynamics and metabolism.

Daniel Lagos1,2, Nicolas Perez2, Pamela R. de Santiago2, Diego Troncoso2, Benjamin Cartes-Saavedra2, Rita Horvath1, Veronica Eisner2

1: Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge, UK.; 2: School of Biological Sciences, Department of Cellular and Molecular Biology, Pontificia Universidad Catolica de Chile, Santiago, Chile.



Dissecting the mitochondrial disease-associated ATAD3 gene cluster and its pathogenic variants

Linden Muellner-Wong1,2, Ann E. Frazier2,3, Eric Hanssen4, Tegan Stait2,3,5, Alice J. Sharpe6, Danielle L. Rudler7,8,9, Shuai Nie10, Luke E. Formosa6, Michael T. Ryan6, Aleksandra Filipovska7,8,9, David R. Thorburn2,3,5, David A. Stroud1,2,5

1: Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria,Australia; 2: Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia; 3: Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia; 4: Ian Holmes Imaging Centre, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria, Australia; 5: Victorian Clinical Genetics Services, Royal Children's Hospital, Melbourne, Victoria, Australia; 6: Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia; 7: Harry Perkins Institute of Medical Research and The University of Western Australia Centre for Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia; 8: ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre and University of Western Australia, Nedlands, Western Australia, Australia; 9: Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, 15 Hospital Avenue, Nedlands, Western Australia, Australia; 10: Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Victoria, Australia



Dynamics of adenine nucleotides in colorectal cancer clinical material

Sten Miller1,2, Leenu Reinsalu1,2, Marju Puurand1, Natalja Timohhina1, Kersti Tepp1, Igor Sevchuk1, Indrek Reile1, Heiki Vija1, Vahur Valvere3, Jelena Bogovskaja3, Tuuli Käämbre1

1: National Institute of Chemical Physics and Biophysics, Estonia; 2: Tallinn University of Technology, Estonia; 3: North Estonia Medical Centre



The role of SURF1 protein in cytochrome c oxidase biogenesis

Maria Jose Saucedo Rodriguez1, Petr Pecina1, Kristýna Čunátová1, Marek Vrbacký1, Alena Pecinová1, Roman Sobotka2, Carlo Viscomi3, Massimo Zeviani4, Tomáš Mráček1, Josef Houštěk1

1: Institute of Physiology of the Czech Academy of Sciences, Czech Republic; 2: Institute of Microbiology, Czech Academy of Sciences, Trebon, Czech Republic; 3: Departement of Biomedical Sciences, University of Padova, Padova, Italy; 4: Departement of Neurosciences, University of Padova, Padova, Italy



Depicting inclusion body myositis using a patient-derived fibroblast model

Judith Cantó Santos1,2,3, Laura Valls Roca1,2,3, Ester Tobías1,2,3, Francesc Josep García García1,2,3, Mariona Guitart Mampel1,2,3, Félix Andújar Sánchez1,2,3, Anna Esteve Codina4,5, Beatriz Martín Mur4, Mercedes Casado3,6, Rafael Artuch3,6, Estel Solsona Vilarrasa7,8, José Carlos Fernández Checa7,8, Carmen García Ruiz7,8, Carles Rentero9, Carlos Enrich9, Pedro Juan Moreno Lozano1,2,3, José César Milisenda1,2,3, Francesc Cardellach1,2,3, Josep Maria Grau Junyent1,2,3, Glòria Garrabou1,2,3

1: Laboratory of Inherited Metabolic Disorders and Muscle Disease, Centre de Recerca Biomèdica CELLEX - Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain; 2: Department of Internal Medicine, Hospital Clinic of Barcelona, Barcelona, Spain; 3: CIBERER— Spanish Biomedical Research Centre in Rare Diseases, Madrid, Spain; 4: CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain; 5: Universitat Pompeu Fabra (UPF), Barcelona, Spain; 6: Department of Clinical Biochemistry, Institut de Recerca Sant Joan de Déu; Esplugues de Llobregat, Barcelona, Spain; 7: Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB-CSIC), Liver Unit-HCB-IDIBAPS, Barcelona, Spain; 8: CIBEREHD-Spanish Biomedical Research Centre in Hepatic and Digestive Diseases, Madrid, Spain; 9: Department of Biomedicine, Cell Biology Unit, CELLEX-IDIBAPS, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain



Effect of physiological cell culture media on cell viability and NRF2 activation

Anton Terasmaa, Rutt Taba, Marie Põlluaed, Tuuli Käämbre

National Institute of Chemical and Biological Physics, Estonia



Genetic and functional characterization of a new patient with COX4I1 deficiency

Frederic Tort1, Olatz Ugarteburu1, Gerard Muñoz-Pujol1, María Unceta2, Ainhoa García2, Arantza Arza2, Javier de las Heras2, Antonia Ribes1, Laura Gort1

1: Hospital Clinic, IDIBAPS, CIBERER, Barcelona, Spain; 2: Hospital Universitario de Cruces, Spain



Application of the Escherichia coli Model System to Study the Human Polyribonucleotide Phosphorylase

Roberto Pizzoccheri, Federica Anna Falchi, Andrea Alloni, Francesca Forti, Sarah Sertic, Giulio Pavesi, Federica Briani

Università degli Studi di Milano, Italy



Phase two biotransformation is highly affected by mitochondrial disease: considerations for pharmacological therapies.

Marianne Venter, Belinda Fouché, Louis Mostert, Zander Lindeque, Rencia van der Sluis

Human Metabolomics, North-West University, South Africa



Mitochondrial phenotyping of fibroblasts from Kearns Sayre’s patients to model the disease

Laura Valls-Roca1,2,3, Judith Cantó-Santos1,2,3, Ester Tobías1,2,3, Francesc Josep García-García1,2,3, Félix Andújar-Sánchez1,2, Laia Farré-Tarrats1,2, Cristina Núñez de Arenas3,6, Rocío Garrido-Moraga5, Joan Padrosa1,2, Raquel Aránega1,2, Pedro J. Moreno-Lozano1,2,3, José César Milisenda1,2, Mar O’Callaghan3,4, Teresa García-Silva3,5, Montserrat Morales-Conejo3,5, Rafael Artuch3,4, Miguel Ángel Martín3,5, José M. Cuezva3,6, Josep M. Grau-Junyent1,2,3, Mariona Guitart-Mampel1,2,3, Glòria Garrabou1,2,3

1: Laboratory of Inherited Metabolic Disorders and Muscle Disease, Centre de Recerca Biomèdica CELLEX - Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Faculty of Medicine and Health Sciences - Universitat de Barcelona (UB); Barcelona, Spain.; 2: Internal Medicine Department - Hospital Clínic de Barcelona; Barcelona, Spain.; 3: CIBERER—Spanish Biomedical Research Centre in Rare Diseases; Madrid, Spain.; 4: Hospital Sant Joan de Déu (HSJdD) de Barcelona, Barcelona, Spain.; 5: Grupo de Enfermedades Mitocondriales, Instituto de Investigación Hospital 12 de Octubre (imas12). Madrid. Spain.; 6: Centro de Biología Molecular S.O., Universidad Autónoma de Madrid (UAM); Madrid, Spain.



Effect of various mutations in the GTPase and middle domain of Drp1 on the mitochondrial network, nucleoids, and peroxisomes

Nikol Volfová1, Aleš Hnízda1, Lukáš Alán2, Robert Dobrovolný1, Jakub Sikora1, Jana Křížová1, Lucie Zdražilová1, Hana Hansíková1, Jiří Zeman1, Markéta Tesařová1

1: Department of Paediatrics and Inherited Metabolic Disorders, Charles University and General University Hospital in Prague, Prague, Czech Republic; 2: Institute of Physiology, The Czech Academy of Sciences, Prague, Czech Republic



Importance of human ClpXP protease for mitochondrial function

Daniela Burska, Jana Tesarova, Jana Krizova, Nikol Volfova, Hana Hansikova, Jiri Zeman, Lukas Stiburek

First Faculty of Medicine, Charles University; and General University Hospital in Prague



Ketogenic diet mitigates the pathogenic phenotype in TMEM70 deficient animal models

Aleksandra Marković1, Petr Pecina1, Alena Pecinová1, Marek Vrbacký1, Jana Mikešová1, Hana Nuskova1, Kateřina Tauchmannová1, Otto Kučera3, Zuzana Cervinkova3, Radislav Sedláček2, Josef Houštěk1, Tomáš Mráček1

1: Institute of Physiology of the Czech Acad. Sci., Prague, Czech Republic; 2: Institute of Molecular Genetics of the Czech Acad. Sci., Prague, Czech Republic; 3: Faculty of Medicine, Charles University, Hradec Kralove, Czech Republic



Mutation in Coq5 leads to CoQ10 deficiency, developmental delay and early death in zebrafish

Sergio López-Herrador1, Julia Corral-Sarasa2, Macarena Gil1, Yaco Morillas1, Luis C. López1,2, Mª. Elena Díaz-Casado1,2

1: Physiology Department, Biomedical Research Center, University of Granada, Granada, Spain; 2: Ibs.Granada, Granada, Spain



Omega-3 supplementation effects on mitochondrial and metabolic profile in a rabbit model of intrauterine growth restriction

Félix Andújar-Sánchez1,2,3, Mariona Guitart-Mampel1,2,3, Míriam Illa3,4, Ester Tobías1,2,3, Laura Valls-Roca1,2,3, Judith Cantó-Santos1,2,3, Laia Farré-Tarrats1,2,3, Clara Oliva3,5, Francesc Cardellach1,2,3, Rafael Artuch3,5, Fàtima Crispi3,4, Glòria Garrabou1,2,3, Francesc J García-García1,2,3

1: Inherited metabolic diseases and muscular disorders Lab, Cellex - Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Faculty of Medicine and Health Science - University of Barcelona (UB), 08036 Barcelona, Spain; 2: Internal Medicine Unit, Medicine Department, Hospital Clínic of Barcelona, 08036 Barcelona, Spain; 3: Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain; 4: BCNatal—Barcelona Centre for Maternal-Foetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Déu), IDIBAPS, University of Barcelona, 08036 Barcelona, Spain; 5: Department of Clinical Biochemistry, Institut de Recerca de Sant Joan de Deu, Esplugues de Llobregat, 08036 Barcelona, Spain



Redundant and divergent roles of COQ8A and COQ8B in cell metabolism.

Agata Valentino1, Elisa Baschiera1, Iolanda Spera2, Luna Laera2, Valentina Giorgio3, Alessandra Castegna2, Leonardo Salviati1, Maria Andrea Desbats1

1: Clinical Genetics Unit, Department of Women and Children’s Health, University of Padova and “Fondazione Istituto di Ricerca Pediatrica Città Della Speranza”, 35127 Padova, Italy.; 2: Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, 70121 Bari, Italy; 3: Department of Biomedical and Neuromotor Sciences, University of Bologna, I-40126 Bologna, Italy.



Loss of CHCHD8 (COA4) caused mitochondrial respiratory Complex IV deficiency

Taku Amo, Yuga Hikage

National Defense Academy, Japan



Delving into the phenotypic heterogeneity of Coenzyme Q biosynthesis defects

Ariadna Crespo-González1, María del Mar Blanquer-Rosselló1,2, Laura García-Corzo1,2, Carmine Staiano1,2, María Chacón1, Ana Belén Cortés-Rodríguez3, Estefanía Sanabria-Reinoso1, María Almuedo-Castillo1, Miguel Ángel Moreno-Mateos1,2, Gloria Brea-Calvo1,2

1: Centro Andaluz de Biología del Desarrollo/Universidad Pablo de Olavide-CSIC-JA, Seville, Spain; 2: CIBERER, Instituto de Salud Carlos III, Madrid, Spain; 3: Laboratorio de Fisiopatología Celular y Bioenergética, Seville, Spain.



Investigating the impact of mtDNA point mutations on mitochondrial function and bioenergetics using patient fibroblasts and hiPSC derived neuronal models

Anitta Rose Chacko, Gabriel Esteban Valdebenito, Michael R Duchen

University College London, United Kingdom



Human COQ10A and COQ10B genes are essential for Coenzyme Q function in mitochondrial respiration

Elisa Baschiera1, Carlo Viscomi1, Maria Andrea Desbats2, Placido Navas3, Leonardo Salviati1,2

1: University of Padova, Italy; 2: Isituto di Ricerca Pediatrica - Cittá della Speranza, Italy; 3: Pablo de Olavide University, Sevilla, Spain



The use of β-RA in leptin-deficient mice reveals novel mechanisms of this compound for the treatment of obesity

Sara Torres-Rusillo1, Sergio López-Herrador1, Pilar González-García1, Mª. Elena Díaz-Casado1,2, Laura Jiménez-Sánchez2, Julia Corral-Sarasa2, Julio Ruiz-Travé1, Luis C. López1,2

1: Physiology Department, Biomedical Research Center, University of Granada, Granada, Spain; 2: Ibs.Granada, Granada, Spain



Oocyte-specific mitofusin 2 knockout enhances the metabolic disfunction of offspring born to obese mothers

Jaiane Santana da Paz, Angélica Camargo dos Santos, Lindomar Oliveira Alves, Julio Cesar Valerio Roncato, Renan Omete Ferreira, Victória Hass Gonçalves, Mirela Souza Cáceres, Marcos Roberto Chiaratti

Federal University of Sao Carlos, Brazil



Off-target effects of etomoxir: inhibition of mitochondrial Complex I and fatty acid oxidation

Timea Komlódi1,2, Filomena SG Silva3, Ana I Duarte3,4,5, Débora Mena3,5,6, Luiz F Garcia-Souza1, Marina Makrecka-Kuka7, Guida Bento3, Luís F Grilo3,5,6, Paulo J Oliveira3, Erich Gnaiger1

1: Oroboros Instruments, Innsbruck, Austria; 2: Dept Biochem, Semmelweis Univ, Budapest, Hungary; 3: CNC-Center Neurosci and Cell Biol, Univ Coimbra, Portugal; 4: IIUC-Inst Interdisciplinary Research, Univ Coimbra, Portugal; 5: CIBB-Center for Innovative Biomed Biotechnol, Univ Coimbra, Portugal; 6: PDBEB-PhD Programme in Exp Biol Biomed, IIUC, Univ Coimbra, Portugal; 7: Lab Pharmaceut Pharmacol, Latvian Inst Organic Synthesis, Riga, Latvia



Mitochondrial alterations in sirtuin1 heterozygous mice fed high fat diet and melatonin

Alessandra Stacchiotti1,2, Francesca Arnaboldi1, Gaia Favero3, Aleksandra Korac4, Maria Monsalve5, Rita Rezzani3

1: Dept Biomedical Sciences for Health, University of Milan, Milan, Italy; 2: Laboratorio Morfologia Umana Applicata, IRCCS Policlinico San Donato, Milan, Italy; 3: Dept Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; 4: Center for Electron Microscopy, University of Belgrade, Belgrade, Serbia; 5: Instituto de Investigaciones Biomedicas “Alberto Sols” (CSIC-UAM), Madrid, Spain



Microproteins in metabolic regulation

Jiemin Nah1, Baptiste Kerouanton1, David Robinson2, Kyle Dunlap3, Pooja Sridnivasan1, Sonia Chothani1, Greg Ducker3, Owen Rackham4, David Stroud2, Lena Ho1

1: Duke-NUS Medical School, Singapore; 2: University of Melbourne, Australia; 3: University of Utah, USA; 4: University of Southampton, UK



Oxphos deficiency indicates novel functions for the mitochondrial protein import subunit tim50

Jordan J Crameri1, Catherine S Palmer1, David Coman2, David A Stroud1, David R Thorburn3,4,5, Ann E Frazier3,4, Diana Stojanovski1

1: Department of Biochemistry and Pharmacology and the Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, 3010, Australia; 2: Queensland Children’s Hospital, Department of Metabolic Medicine, South Brisbane, Brisbane, Queensland, 4001, Australia; 3: Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Victoria, 3052, Australia; 4: Department of Paediatrics, University of Melbourne, Melbourne, Victoria, 3052, Australia; 5: Victorian Clinical Genetics Services, Royal Children’s Hospital, Melbourne, Victoria, 3052, Australia



The levels and activation state of the pyruvate dehydrogenase complex modulate the SCAFI-dependent organization of the mitochondrial respiratory chain

Sandra Lopez-Calcerrada1, Ana Sierra-Magro1, Erika Fernández-Vizarra2, Cristina Ugalde1,3

1: Instituto de Investigación Hospital 12 de Octubre, Madrid 28041, Spain; 2: Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; 3: Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), U723, Madrid, Spain



Development of a yeast model to characterize OPA1 mutations associated with different neuromuscular disorders

Cristina Calderan1, Marco Marchi1, Mara Doimo1, Maria Andrea Desbats1, Geppo Sartori2, Leonardo Salviati1

1: Clinical Genetics Unit, Department of Women’s and Children’s Health, University of Padua, and Istituto di Ricerca Pediatrica (IRP) Città della Speranza, Padua, Italy; 2: Department of Biomedical Sciences, University of Padua, Padua, Italy



An ultra-special family with an ultra-rare condition: three children with mithochondrial complex III deficiency due to homozygous mutations in Lyrm7

Francesca Manzoni, Titia Anita Wischmeijer, Elisa Boni, Lucio Parmeggiani, Andrea Bordugo, Francesca Pellegrini

Bolzano Hospital, Italy