A. , N. , and V. Shoshan-barmatz, Voltage-dependent anion channel 1-based peptides interact with Bcl-2 to prevent antiapoptotic activity, J Biol Chem, vol.285, pp.6053-6062, 2010.

C. , M. , E. Barksby, N. Johnson, and M. Capano, Mitochondrial intermembrane junctional complexes and their involvement in cell death, Biochimie, vol.84, pp.143-152, 2002.

D. , M. , D. Grevent, A. S. Lebre, M. Rio et al., Aspect of brain MRI in mitochondrial respiratory chain deficiency. A diagnostic algorithm of the most common mitochondrial genetic mutations], Rev Neurol, vol.170, pp.381-389, 2014.

E. , M. A. , M. Carre, and D. Braguer, Microtubules in Apoptosis Induction: Are They Necessary?, Current Cancer Drug Targets, vol.7, issue.8, pp.713-729, 2007.
DOI : 10.2174/156800907783220480

K. , J. F. , A. H. Wyllie, and A. R. Currie, Apoptosis: a basic biological phenomenon with wideranging implications in tissue kinetics, Br J Cancer, vol.26, pp.239-257, 1972.

K. , P. , H. M. Honda, and J. N. Weiss, Regulation of the mitochondrial permeability transition by matrix Ca(2+) and voltage during anoxia/reoxygenation, Am J Physiol Cell Physiol, vol.280, pp.517-526, 2001.

M. , U. E. , Z. Lin, C. Trimmer, N. Flomenberg et al., Cancer cells metabolically "fertilize" the tumor microenvironment with hydrogen peroxide, driving the Warburg effect: implications for PET imaging of human tumors, Cell Cycle, vol.10, pp.2504-2520, 2011.

M. , L. , M. Trivedi, A. Singh, M. Talekar et al., Mitochondrial biology, targets, and drug delivery, J Control Release, vol.207, pp.40-58, 2015.

R. , A. , and P. Bernardi, The mitochondrial permeability transition pore and its involvement in cell death and in disease pathogenesis, Apoptosis, vol.12, pp.815-833, 2007.

T. , A. , J. Thorburn, and A. E. Frankel, Induction of apoptosis by tumor cell-targeted toxins, Apoptosis, vol.9, pp.19-25, 2004.

W. , M. I. , and C. M. Williams, Mitochondria in the flight muscles of insects. II. Effects of the medium on the size form, and organization of isolated sarcosomes, J Gen Physiol, vol.37, pp.71-90, 1953.

J. Abramson, M. Svensson-ek, B. Byrne, and S. Iwata, Structure of cytochrome c oxidase: a comparison of the bacterial and mitochondrial enzymes, Biochimica et Biophysica Acta (BBA) - Protein Structure and Molecular Enzymology, vol.1544, issue.1-2, pp.1-9, 2001.
DOI : 10.1016/S0167-4838(00)00241-7

C. Alexander, M. Votruba, U. E. Pesch, D. L. Thiselton, and S. Mayer, OPA1, encoding a dynamin-related GTPase, is mutated in autosomal dominant optic atrophy linked to chromosome 3q28, OPA1, encoding a dynaminrelated GTPase, is mutated in autosomal dominant optic atrophy linked to chromosome 3q28, pp.211-215, 2000.
DOI : 10.1146/annurev.bi.50.070181.002025

A. A. Amchenkova, L. E. Bakeeva, Y. S. Chentsov, V. P. Skulachev, and D. B. Zorov, Coupling membranes as energy-transmitting cables. I. Filamentous mitochondria in fibroblasts and mitochondrial clusters in cardiomyocytes, The Journal of Cell Biology, vol.107, issue.2, pp.481-495, 1988.
DOI : 10.1083/jcb.107.2.481

S. Anderson, A. T. Bankier, B. G. Barrell, M. H. De-bruijn, and A. R. Coulson, Sequence and organization of the human mitochondrial genome, Sequence and organization of the human mitochondrial genome, pp.457-465, 1981.
DOI : 10.1038/290457a0

T. D. Anderson, J. P. Ross, R. K. Roby, D. A. Lee, and M. M. Holland, A Validation Study for the Extraction and Analysis of DNA from Human Nail Material and Its Application to Forensic Casework, Journal of Forensic Sciences, vol.44, issue.5, pp.1053-1056, 1999.
DOI : 10.1520/JFS12041J

R. M. Andrews, I. Kubacka, P. F. Chinnery, R. N. Lightowlers, and D. M. Turnbull, Reanalysis and revision of the Cambridge reference sequence for human mitochondrial DNA, Nat Genet, vol.23, p.147, 1999.

G. Anthony, A. Reimann, and B. Kadenbach, Tissue-specific regulation of bovine heart cytochrome-c oxidase activity by ADP via interaction with subunit VIa., Proceedings of the National Academy of Sciences, vol.90, issue.5, pp.1652-1656, 1993.
DOI : 10.1073/pnas.90.5.1652

S. Arnold and B. Kadenbach, Cell Respiration is Controlled by ATP, an Allosteric Inhibitor of Cytochrome-c Oxidase, European Journal of Biochemistry, vol.85, issue.1, pp.350-354, 1997.
DOI : 10.1086/405228

J. Arpa, A. Cruz-martinez, Y. Campos, M. Gutierrez-molina, and F. Garcia-rio, Prevalence and progression of mitochondrial diseases: A study of 50 patients, Muscle & Nerve, vol.14, issue.Suppl 3, pp.690-695, 2003.
DOI : 10.1002/mus.10507

L. E. Bakeeva, S. Yu, and V. P. Skulachev, Mitochondrial framework (reticulum mitochondriale) in rat diaphragm muscle, Biochimica et Biophysica Acta (BBA) - Bioenergetics, vol.501, issue.3, pp.349-369, 1978.
DOI : 10.1016/0005-2728(78)90104-4

H. J. Bandelt, A. Salas, R. W. Taylor, and Y. G. Yao, Exaggerated status of ???novel??? and ???pathogenic??? mtDNA sequence variants due to inadequate database searches, Human Mutation, vol.336, issue.2, pp.191-196, 2009.
DOI : 10.1002/humu.20846

S. Bannwarth, V. Procaccio, A. S. Lebre, C. Jardel, and A. Chaussenot, Prevalence of rare mitochondrial DNA mutations in mitochondrial disorders, Journal of Medical Genetics, vol.20, issue.10, pp.704-714
DOI : 10.1136/jmedgenet-2013-101604

S. Bannwarth, V. Procaccio, and V. Paquis-flucklinger, Surveyor??? Nuclease: A new strategy for a rapid identification of heteroplasmic mitochondrial DNA mutations in patients with respiratory chain defects, Human Mutation, vol.127, issue.6, pp.575-582, 2005.
DOI : 10.1002/humu.20177

J. T. Barron, L. Gu, and J. E. Parrillo, Malate-Aspartate Shuttle, Cytoplasmic NADH Redox Potential, and Energetics in Vascular Smooth Muscle, Journal of Molecular and Cellular Cardiology, vol.30, issue.8, pp.1571-1579, 1998.
DOI : 10.1006/jmcc.1998.0722

C. Bartenhagen and M. Dugas, Robust and exact structural variation detection with paired-end and soft-clipped alignments: SoftSV compared with eight algorithms, Briefings in Bioinformatics, vol.17, issue.1, 2015.
DOI : 10.1093/bib/bbv028

I. Beau, M. Mehrpour, and P. Codogno, Autophagosomes and human diseases, The International Journal of Biochemistry & Cell Biology, vol.43, issue.4, pp.460-464, 2011.
DOI : 10.1016/j.biocel.2011.01.006

A. Behin, C. Jardel, K. G. Claeys, J. Fagart, and M. Louha, Adult cases of mitochondrial DNA depletion due to TK2 defect: An expanding spectrum, Neurology, vol.78, issue.9, pp.644-648, 2012.
DOI : 10.1212/WNL.0b013e318248df2b

D. R. Bentley, S. Balasubramanian, H. P. Swerdlow, G. P. Smith, and . Milton, Accurate whole human genome sequencing using reversible terminator chemistry, Nature, vol.34, issue.7218, pp.53-59, 2008.
DOI : 10.1038/nature07517

E. L. Blakely, K. J. Rennie, L. Jones, M. Elstner, and Z. M. Chrzanowska-lightowlers, Sporadic Intragenic Inversion of the Mitochondrial DNA MTND1 Gene Causing Fatal Infantile Lactic Acidosis, Pediatric Research, vol.1659, issue.3, pp.440-444, 2006.
DOI : 10.1203/01.pdr.0000198771.78290.c4

E. J. Boekema and H. P. Braun, Supramolecular Structure of the Mitochondrial Oxidative Phosphorylation System, Journal of Biological Chemistry, vol.282, issue.1, pp.1-4, 2007.
DOI : 10.1074/jbc.R600031200

P. Boesch, N. Ibrahim, A. Dietrich, and R. N. Lightowlers, Membrane association of mitochondrial DNA facilitates base excision repair in mammalian mitochondria, Nucleic Acids Research, vol.38, issue.5, pp.1478-1488, 2010.
DOI : 10.1093/nar/gkp1143

P. Boesch, F. Weber-lotfi, N. Ibrahim, V. Tarasenko, and A. Cosset, DNA repair in organelles: Pathways, organization, regulation, relevance in disease and aging, Biochimica et Biophysica Acta (BBA) - Molecular Cell Research, vol.1813, issue.1, pp.186-200, 2011.
DOI : 10.1016/j.bbamcr.2010.10.002

URL : https://hal.archives-ouvertes.fr/hal-00629716

D. F. Bogenhagen, Mitochondrial DNA nucleoid structure, Mitochondrial DNA nucleoid structure, pp.914-920, 2012.
DOI : 10.1016/j.bbagrm.2011.11.005

V. B. Borisov, Defects in mitochondrial respiratory complexes III and IV, and human pathologies, Molecular Aspects of Medicine, vol.23, issue.5, pp.385-412, 2002.
DOI : 10.1016/S0098-2997(02)00013-4

H. Boumans, L. A. Grivell, and J. A. Berden, The Respiratory Chain in Yeast Behaves as a Single Functional Unit, Journal of Biological Chemistry, vol.273, issue.9, pp.4872-4877, 1998.
DOI : 10.1074/jbc.273.9.4872

L. M. Bragg, G. Stone, M. K. Butler, P. Hugenholtz, and G. W. Tyson, Shining a Light on Dark Sequencing: Characterising Errors in Ion Torrent PGM Data, PLoS Computational Biology, vol.6, issue.4, p.1003031, 2013.
DOI : 10.1371/journal.pcbi.1003031.s014

J. J. Briere, J. Favier, V. Ghouzzi, F. Djouadi, and P. Benit, Succinate dehydrogenase deficiency in human, Cellular and Molecular Life Sciences, vol.62, issue.19-20, pp.2317-2324, 2005.
DOI : 10.1007/s00018-005-5237-6

N. Brun, Y. Robitaille, A. Grignon, B. H. Robinson, and G. A. Mitchell, Pyruvate carboxylase deficiency: Prenatal onset of ischemia???like brain lesions in two sibs with the acute neonatal form, American Journal of Medical Genetics, vol.84, issue.2, 1999.
DOI : 10.1002/(SICI)1096-8628(19990521)84:2<94::AID-AJMG3>3.3.CO;2-T

H. P. Buermans and J. T. , Next generation sequencing technology: Advances and applications, Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease, vol.1842, issue.10, pp.1932-1941
DOI : 10.1016/j.bbadis.2014.06.015

C. Calabrese, D. Simone, M. A. Diroma, M. Santorsola, and C. Gutta, MToolBox: a highly automated pipeline for heteroplasmy annotation and prioritization analysis of human mitochondrial variants in high-throughput sequencing, Bioinformatics, vol.30, issue.21, pp.3115-3117, 2014.
DOI : 10.1093/bioinformatics/btu483

V. Carelli, A. Baracca, S. Barogi, F. Pallotti, and M. L. Valentino, Biochemical-Clinical Correlation in Patients With Different Loads of the Mitochondrial DNA T8993G Mutation, Archives of Neurology, vol.59, issue.2, pp.264-270, 2002.
DOI : 10.1001/archneur.59.2.264

V. Carelli, F. N. Ross-cisneros, and A. A. Sadun, Mitochondrial dysfunction as a cause of optic neuropathies, Progress in Retinal and Eye Research, vol.23, issue.1, pp.53-89, 2004.
DOI : 10.1016/j.preteyeres.2003.10.003

J. Carroll, I. M. Fearnley, J. M. Skehel, R. J. Shannon, and J. Hirst, Bovine Complex I Is a Complex of 45 Different Subunits, Journal of Biological Chemistry, vol.281, issue.43, pp.32724-32727, 2006.
DOI : 10.1074/jbc.M607135200

G. Casari, M. D. Fusco, S. Ciarmatori, M. Zeviani, and M. Mora, Spastic Paraplegia and OXPHOS Impairment Caused by Mutations in Paraplegin, a Nuclear-Encoded Mitochondrial Metalloprotease, Cell, vol.93, issue.6, pp.973-983, 1998.
DOI : 10.1016/S0092-8674(00)81203-9

S. Castellana, J. Ronai, and T. Mazza, MitImpact: an Exhaustive Collection of Pre-computed Pathogenicity Predictions of Human Mitochondrial Non-synonymous Variants, Human Mutation, vol.78, issue.Database issue, pp.2413-2422, 2015.
DOI : 10.1002/humu.22720

M. Castro-gago, V. Gonzalez-conde, M. J. Fernandez-seara, E. Rodrigo-saez, and S. Fernandez-cebrian, Early mitochondrial encephalomyopathy due to complex IV deficiency consistent with Alpers-Huttenlocher syndrome: report of two cases], Rev Neurol, vol.29, pp.912-917, 1999.

J. B. Cavanagh and B. N. Harding, Pathogenic factors underlying the lesions in Leigh's disease, Brain, vol.117, issue.6, pp.1357-1376, 1994.
DOI : 10.1093/brain/117.6.1357

A. Chakrapani, L. Heptinstall, and J. Walter, A family with Leigh syndrome caused by the rarer T8993C mutation, Journal of Inherited Metabolic Disease, vol.21, issue.6, pp.685-686, 1998.
DOI : 10.1023/A:1005401121344

H. Chen and D. C. Chan, Critical dependence of neurons on mitochondrial dynamics, Current Opinion in Cell Biology, vol.18, issue.4, pp.453-459, 2006.
DOI : 10.1016/j.ceb.2006.06.004

X. Chen, R. Prosser, S. Simonetti, J. Sadlock, and G. Jagiello, Rearranged mitochondrial genomes are present in human oocytes, Am J Hum Genet, vol.57, pp.239-247, 1995.

P. F. Chinnery, R. M. Andrews, D. M. Turnbull, and N. N. Howell, Leber hereditary optic neuropathy: Does heteroplasmy influence the inheritance and expression of the G11778A mitochondrial DNA mutation?, American Journal of Medical Genetics, vol.42, issue.3, pp.235-243, 2001.
DOI : 10.1002/1096-8628(20010122)98:3<235::AID-AJMG1086>3.0.CO;2-O

P. F. Chinnery and G. Hudson, Mitochondrial genetics, Mitochondrial genetics, pp.135-159, 2013.
DOI : 10.1093/bmb/ldt017

P. F. Chinnery, M. A. Johnson, T. M. Wardell, R. Singh-kler, and C. Hayes, The epidemiology of pathogenic mitochondrial DNA mutations, Annals of Neurology, vol.30, issue.2, pp.188-193, 2000.
DOI : 10.1002/1531-8249(200008)48:2<188::AID-ANA8>3.0.CO;2-P

M. Chomova and P. Racay, Mitochondrial complex I in the network of known and unknown facts, General Physiology and Biophysics, vol.29, issue.1, pp.3-11, 2010.
DOI : 10.4149/gpb_2010_01_3

S. L. Chow, Z. J. Rooney, M. A. Cleary, P. T. Clayton, and J. V. Leonard, The significance of elevated CSF lactate, Archives of Disease in Childhood, vol.90, issue.11, pp.1188-1189, 2005.
DOI : 10.1136/adc.2005.075317

S. Cipolat, O. Martins-de-brito, B. Dal-zilio, and L. Scorrano, OPA1 requires mitofusin 1 to promote mitochondrial fusion, Proceedings of the National Academy of Sciences, vol.101, issue.45, pp.15927-15932, 2004.
DOI : 10.1073/pnas.0407043101

D. A. Clayton, Replication of animal mitochondrial DNA, Cell, vol.28, issue.4, pp.693-705, 1982.
DOI : 10.1016/0092-8674(82)90049-6

D. A. Clayton, Mitochondrial DNA Replication: What We Know, IUBMB Life (International Union of Biochemistry and Molecular Biology: Life), vol.55, issue.4-5, pp.213-217, 2003.
DOI : 10.1080/1521654031000134824

M. Collu-marchese, M. Shuen, M. Pauly, A. Saleem, and D. A. Hood, 2015 The regulation of mitochondrial transcription factor A (Tfam) expression during skeletal muscle cell differentiation, Biosci Rep, vol.35

W. C. Copeland, Defects in mitochondrial DNA replication and human disease, Critical Reviews in Biochemistry and Molecular Biology, vol.339, issue.2, pp.64-74
DOI : 10.1016/j.febslet.2007.06.042

M. S. Damian, P. Seibel, W. Schachenmayr, H. Reichmann, and W. Dorndorf, VACTERL with the mitochondrial NP 3243 point mutation, American Journal of Medical Genetics, vol.338, issue.4, pp.398-403, 1996.
DOI : 10.1002/(SICI)1096-8628(19960424)62:4<398::AID-AJMG13>3.0.CO;2-J

N. Darin, A. Oldfors, A. R. Moslemi, E. Holme, and M. Tulinius, The incidence of mitochondrial encephalomyopathies in childhood: Clinical features and morphological, biochemical, and DNA abnormalities, Annals of Neurology, vol.109, issue.3, pp.377-383, 2001.
DOI : 10.1002/ana.75

S. Das, N. Hajnoczky, A. N. Antony, G. Csordas, and L. D. Gaspers, Mitochondrial morphology and dynamics in hepatocytes from normal and ethanol-fed rats, Pfl??gers Archiv - European Journal of Physiology, vol.36, issue.Suppl 1, pp.101-109, 2012.
DOI : 10.1007/s00424-012-1100-4

D. Lonlay, P. , C. Mugnier, D. Sanlaville, K. Chantrel-groussard et al., human:rodent hybrids for physical mapping of novel mitochondrial respiratory chain deficiency genes, Cell complementation using Genebridge Hum Mol Genet, vol.4, issue.11, pp.3273-3281, 2002.

F. G. Debray, G. A. Mitchell, P. Allard, B. H. Robinson, and J. A. Hanley, Diagnostic Accuracy of Blood Lactate-to-Pyruvate Molar Ratio in the Differential Diagnosis of Congenital Lactic Acidosis, Clinical Chemistry, vol.53, issue.5, pp.916-921, 2007.
DOI : 10.1373/clinchem.2006.081166

M. B. Delatycki, R. Williamson, and S. M. Forrest, Friedreich ataxia: an overview, Journal of Medical Genetics, vol.37, issue.1, pp.1-8, 2000.
DOI : 10.1136/jmg.37.1.1

C. Delettre, J. M. Griffoin, J. Kaplan, H. Dollfus, and B. Lorenz, Mutation spectrum and splicing variants in the OPA1 gene, Human Genetics, vol.109, issue.6, pp.584-591, 2001.
DOI : 10.1007/s00439-001-0633-y

C. Delettre, G. Lenaers, J. M. Griffoin, N. Gigarel, and C. Lorenzo, Nuclear gene OPA1, encoding a mitochondrial dynamin-related protein, is mutated in dominant optic atrophy, Nature Genetics, vol.303, issue.2, pp.207-210, 2000.
DOI : 10.1083/JCB.143.2.351

M. Devaux-bricout, D. Grevent, A. S. Lebre, M. Rio, and I. Desguerre, Aspect en IRM c??r??brale des maladies mitochondriales. Algorithme d??cisionnel des maladies mitochondriales les plus fr??quentes, Revue Neurologique, vol.170, issue.5, pp.381-389, 2014.
DOI : 10.1016/j.neurol.2014.03.006

S. Dimauro, Mitochondrial encephalomyopathies--Fifty years on: The Robert Wartenberg Lecture, Neurology, vol.81, issue.3, pp.281-291
DOI : 10.1212/WNL.0b013e31829bfe89

S. Dimauro and E. A. Schon, Mitochondrial DNA mutations in human disease, Mitochondrial DNA mutations in human disease, pp.18-26, 2001.
DOI : 10.1002/ajmg.1392

F. Distelmaier, W. J. Koopman, L. P. Van-den-heuvel, R. J. Rodenburg, and E. Mayatepek, Mitochondrial complex I deficiency: from organelle dysfunction to clinical disease, Brain, vol.132, issue.4, pp.833-842, 2009.
DOI : 10.1093/brain/awp058

D. K. Dowling, Evolutionary perspectives on the links between mitochondrial genotype and disease phenotype, Biochimica et Biophysica Acta (BBA) - General Subjects, vol.1840, issue.4, pp.1393-1403
DOI : 10.1016/j.bbagen.2013.11.013

N. V. Dudkina, H. Eubel, W. Keegstra, E. J. Boekema, and H. P. Braun, Structure of a mitochondrial supercomplex formed by respiratory-chain complexes I and III, Proceedings of the National Academy of Sciences, vol.102, issue.9, pp.3225-3229, 2005.
DOI : 10.1073/pnas.0408870102

T. M. Durcan and E. A. Fon, The three ???P???s of mitophagy: PARKIN, PINK1, and post-translational modifications, Genes & Development, vol.29, issue.10, pp.989-999
DOI : 10.1101/gad.262758.115

A. W. El-hattab, A. M. Adesina, J. Jones, and F. Scaglia, MELAS syndrome: Clinical manifestations, pathogenesis, and treatment options, MELAS syndrome: Clinical manifestations, pathogenesis, and treatment options, pp.4-12, 2015.
DOI : 10.1016/j.ymgme.2015.06.004

E. Meziane, A. , S. K. Lehtinen, I. J. Holt, and H. T. Jacobs, Mitochondrial tRNALeu isoforms in lung carcinoma cybrid cells containing the np 3243 mtDNA mutation, Mitochondrial tRNALeu isoforms in lung carcinoma cybrid cells containing the np 3243 mtDNA mutation, pp.2141-2147, 1998.
DOI : 10.1093/hmg/7.13.2141

M. Filippi, A. Charil, M. Rovaris, M. Absinta, and M. A. Rocca, Insights from magnetic resonance imaging, Handb Clin Neurol, vol.122, pp.115-149
DOI : 10.1016/B978-0-444-52001-2.00006-6

E. Fragouli, K. Spath, S. Alfarawati, F. Kaper, and A. Craig, Altered Levels of Mitochondrial DNA Are Associated with Female Age, Aneuploidy, and Provide an Independent Measure of Embryonic Implantation Potential, PLOS Genetics, vol.3, issue.6, p.1005241, 2015.
DOI : 10.1371/journal.pgen.1005241.t003

K. Fu, R. Hartlen, T. Johns, A. Genge, and G. Karpati, A novel heteroplasmic tRNAleu(CUN) mtDNA point mutation in a sporadic patient with mitochondrial encephalomyopathy segregates rapidly in skeletal muscle and suggests an approach to therapy, Human Molecular Genetics, vol.5, issue.11, pp.1835-1840, 1996.
DOI : 10.1093/hmg/5.11.1835

J. M. Fuste, S. Wanrooij, E. Jemt, C. E. Granycome, and T. J. Cluett, Mitochondrial RNA Polymerase Is Needed for Activation of the Origin of Light-Strand DNA Replication, Molecular Cell, vol.37, issue.1, pp.67-78, 2010.
DOI : 10.1016/j.molcel.2009.12.021

M. E. Gegg, J. M. Cooper, K. Y. Chau, M. Rojo, and A. H. Schapira, Mitofusin 1 and mitofusin 2 are ubiquitinated in a PINK1/parkin-dependent manner upon induction of mitophagy, Human Molecular Genetics, vol.19, issue.24, pp.4861-4870, 2010.
DOI : 10.1093/hmg/ddq419

M. L. Genova and G. Lenaz, Functional role of mitochondrial respiratory supercomplexes, Biochimica et Biophysica Acta (BBA) - Bioenergetics, vol.1837, issue.4, pp.427-443
DOI : 10.1016/j.bbabio.2013.11.002

D. Ghezzi, P. Goffrini, G. Uziel, R. Horvath, and T. Klopstock, SDHAF1, encoding a LYR complex-II specific assembly factor, is mutated in SDH-defective infantile leukoencephalopathy, Nature Genetics, vol.69, issue.6, pp.654-656, 2009.
DOI : 10.1016/j.tig.2008.05.008

E. E. Glater, L. J. Megeath, R. S. Stowers, and T. L. Schwarz, Axonal transport of mitochondria requires milton to recruit kinesin heavy chain and is light chain independent, The Journal of Cell Biology, vol.14, issue.4, pp.545-557, 2006.
DOI : 10.1016/S0959-4388(99)80045-2

D. Golan and P. Medvedev, Using state machines to model the Ion Torrent sequencing process and to improve read error rates, Bioinformatics, vol.29, issue.13, pp.344-351, 2013.
DOI : 10.1093/bioinformatics/btt212

Y. Goto, Clinical features of melas and mitochondrial DNA mutations, Muscle & Nerve, vol.1, issue.S14, pp.107-112, 1995.
DOI : 10.1002/mus.880181422

Y. Goto, K. Tsugane, Y. Tanabe, I. Nonaka, and S. Horai, A New Point Mutation at Nucleotide Pair-3291 of the Mitochondrial Transfer-RNALeu(Uur) Gene in a Patient with Mitochondrial Myopathy, Encephalopathy, Lactic-Acidosis, and Stroke-Like Episodes (MELAS), Biochemical and Biophysical Research Communications, vol.202, issue.3, pp.1624-1630
DOI : 10.1006/bbrc.1994.2119

L. Griparic, N. N. Van-der-wel, I. J. Orozco, P. J. Peters, and A. M. Van-der-bliek, Loss of the Intermembrane Space Protein Mgm1/OPA1 Induces Swelling and Localized Constrictions along the Lengths of Mitochondria, Journal of Biological Chemistry, vol.279, issue.18, pp.18792-18798, 2004.
DOI : 10.1074/jbc.M400920200

M. X. Guan, Mitochondrial 12S rRNA mutations associated with aminoglycoside ototoxicity, Mitochondrion, vol.11, issue.2, pp.237-245
DOI : 10.1016/j.mito.2010.10.006

R. H. Haas, Thiamin and the Brain, Annual Review of Nutrition, vol.8, issue.1, pp.483-515, 1988.
DOI : 10.1146/annurev.nu.08.070188.002411

R. H. Haas, S. Parikh, M. J. Falk, R. P. Saneto, and N. I. Wolf, The in-depth evaluation of suspected mitochondrial disease, Molecular Genetics and Metabolism, vol.94, issue.1, pp.16-37, 2008.
DOI : 10.1016/j.ymgme.2007.11.018

J. Habersetzer, W. Ziani, I. Larrieu, C. Stines-chaumeil, and M. F. Giraud, ATP synthase oligomerization: From the enzyme models to the mitochondrial morphology, The International Journal of Biochemistry & Cell Biology, vol.45, issue.1, pp.99-105
DOI : 10.1016/j.biocel.2012.05.017

URL : https://hal.archives-ouvertes.fr/hal-00712690

C. M. Henley and J. Schacht, Pharmacokinetics of Aminoglycoside Antibiotics in Blood, Inner-Ear Fluids and Tissues and Their Relationship to Ototoxicity, International Journal of Audiology, vol.36, issue.3, pp.137-146, 1988.
DOI : 10.1172/JCI112463

D. A. Hood, I. Irrcher, V. Ljubicic, and A. M. Joseph, Coordination of metabolic plasticity in skeletal muscle, Journal of Experimental Biology, vol.209, issue.12, pp.2265-2275, 2006.
DOI : 10.1242/jeb.02182

R. Horsefield, V. Yankovskaya, G. Sexton, W. Whittingham, and K. Shiomi, Structural and Computational Analysis of the Quinone-binding Site of Complex II (Succinate-Ubiquinone Oxidoreductase), Journal of Biological Chemistry, vol.281, issue.11, pp.7309-7316, 2006.
DOI : 10.1074/jbc.M508173200

M. Huttemann, P. Pecina, M. Rainbolt, T. H. Sanderson, and V. E. Kagan, The multiple functions of cytochrome c and their regulation in life and death decisions of the mammalian cell: From respiration to apoptosis, Mitochondrion, vol.11, issue.3, pp.369-381, 2011.
DOI : 10.1016/j.mito.2011.01.010

T. Iizuka and F. Sakai, Pathogenesis of Stroke-Like Episodes in MELAS: Analysis of Neurovascular Cellular Mechanisms, Current Neurovascular Research, vol.2, issue.1, pp.29-45, 2005.
DOI : 10.2174/1567202052773544

M. Ingman and U. Gyllensten, mtDB: Human Mitochondrial Genome Database, a resource for population genetics and medical sciences, Nucleic Acids Research, vol.34, issue.90001, pp.749-751, 2006.
DOI : 10.1093/nar/gkj010

R. J. Janssen, L. G. Nijtmans, L. P. Van-den-heuvel, and J. A. Smeitink, Mitochondrial complex I: Structure, function and pathology, Journal of Inherited Metabolic Disease, vol.279, issue.4, pp.499-515, 2006.
DOI : 10.1007/s10545-006-0362-4

B. Kadenbach, R. Ramzan, L. Wen, and S. Vogt, New extension of the Mitchell Theory for oxidative phosphorylation in mitochondria of living organisms, Biochimica et Biophysica Acta (BBA) - General Subjects, vol.1800, issue.3, pp.205-212, 2010.
DOI : 10.1016/j.bbagen.2009.04.019

O. A. Kajander, T. A. Kunnas, M. Perola, S. K. Lehtinen, and P. J. Karhunen, Long-Extension PCR to Detect Deleted Mitochondrial DNA Molecules Is Compromized by Technical Artefacts, Biochemical and Biophysical Research Communications, vol.254, issue.2, pp.507-514, 1999.
DOI : 10.1006/bbrc.1998.9975

T. P. Kearns, Pigmentary Degeneration of the Retina, and Cardiomyopathy: A Newly Recognized Syndrome, External Ophthalmoplegia Trans Am Ophthalmol Soc, vol.63, pp.559-625, 1965.

D. S. Kerr, Review of Clinical Trials for Mitochondrial Disorders: 1997???2012, Neurotherapeutics, vol.131, issue.Suppl. 1, pp.307-319, 2013.
DOI : 10.1007/s13311-013-0176-7

M. Kishi, Y. Yamamura, T. Kurihara, N. Fukuhara, and K. Tsuruta, An autopsy case of mitochondrial encephalomyopathy: biochemical and electron microscopic studies of the brain, Journal of the Neurological Sciences, vol.86, issue.1, pp.31-40, 1988.
DOI : 10.1016/0022-510X(88)90005-6

A. Kloss-brandstatter, D. Pacher, S. Schonherr, H. Weissensteiner, and R. Binna, HaploGrep: a fast and reliable algorithm for automatic classification of mitochondrial DNA haplogroups, Human Mutation, vol.123, issue.Suppl 3, pp.25-32, 2011.
DOI : 10.1002/humu.21382

Y. Koga, N. Povalko, J. Nishioka, K. Katayama, and N. Kakimoto, MELAS and l-arginine therapy: pathophysiology of stroke-like episodes, Annals of the New York Academy of Sciences, vol.27, issue.230, pp.104-110, 2010.
DOI : 10.1111/j.1749-6632.2010.05624.x

A. M. Kogelnik, M. T. Lott, M. D. Brown, S. B. Navathe, and D. C. Wallace, MITOMAP: a human mitochondrial genome database--1998 update, Nucleic Acids Research, vol.26, issue.1, pp.112-115, 1998.
DOI : 10.1093/nar/26.1.112

L. Kussmaul and J. Hirst, The mechanism of superoxide production by NADH:ubiquinone oxidoreductase (complex I) from bovine heart mitochondria, Proceedings of the National Academy of Sciences, vol.103, issue.20, pp.7607-7612, 2006.
DOI : 10.1073/pnas.0510977103

M. Lagouge, C. Argmann, Z. Gerhart-hines, H. Meziane, and C. Lerin, Resveratrol Improves Mitochondrial Function and Protects against Metabolic Disease by Activating SIRT1 and PGC-1??, Cell, vol.127, issue.6, pp.1109-1122, 2006.
DOI : 10.1016/j.cell.2006.11.013

URL : https://hal.archives-ouvertes.fr/hal-00188005

B. Langmead, C. Trapnell, M. Pop, and S. L. Salzberg, Ultrafast and memory-efficient alignment of short DNA sequences to the human genome, Genome Biology, vol.10, issue.3, p.25, 2009.
DOI : 10.1186/gb-2009-10-3-r25

R. M. Lebovitz, Ownership of human genes, Nature, vol.382, issue.6586, p.17, 1996.
DOI : 10.1038/382017a0

Y. J. Lee, S. Y. Jeong, M. Karbowski, C. L. Smith, and R. J. Youle, Roles of the Mammalian Mitochondrial Fission and Fusion Mediators Fis1, Drp1, and Opa1 in Apoptosis, Molecular Biology of the Cell, vol.15, issue.11, pp.5001-5011, 2004.
DOI : 10.1091/mbc.E04-04-0294

H. Z. Li, R. Y. Li, and M. Li, A review of maternally inherited diabetes and deafness, Frontiers in Bioscience, vol.19, issue.5, pp.777-782
DOI : 10.2741/4244

C. R. Lincke, C. Van-den-bogert, L. G. Nijtmans, R. J. Wanders, and P. Tamminga, Cerebellar Hypoplasia in Respiratory Chain Dysfunction, Cerebellar hypoplasia in respiratory chain dysfunction, pp.216-218, 1996.
DOI : 10.1055/s-2007-973792

L. Liu, Y. Li, S. Li, N. Hu, and Y. He, 2012 Comparison of next-generation sequencing systems, J Biomed Biotechnol, vol.2012, p.251364

M. F. Lopez, B. S. Kristal, E. Chernokalskaya, A. Lazarev, and A. I. Shestopalov, High-throughput profiling of the mitochondrial proteome using affinity fractionation and automation, Electrophoresis, vol.431, issue.16, pp.3427-3440, 2000.
DOI : 10.1002/1522-2683(20001001)21:16<3427::AID-ELPS3427>3.0.CO;2-L

A. Maitra, Y. Cohen, S. E. Gillespie, E. Mambo, and N. Fukushima, The Human MitoChip: A High-Throughput Sequencing Microarray for Mitochondrial Mutation Detection, Genome Research, vol.14, issue.5, pp.812-819, 2004.
DOI : 10.1101/gr.2228504

E. Malfatti, M. Bugiani, F. Invernizzi, C. F. De-souza, and L. Farina, Novel mutations of ND genes in complex I deficiency associated with mitochondrial encephalopathy, Brain, vol.130, issue.7, pp.1894-1904, 2007.
DOI : 10.1093/brain/awm114

P. Y. Man, P. G. Griffiths, D. T. Brown, N. Howell, and D. M. Turnbull, The Epidemiology of Leber Hereditary Optic Neuropathy in the North East of England, The American Journal of Human Genetics, vol.72, issue.2, pp.333-339, 2003.
DOI : 10.1086/346066

M. Negrier, M. L. , M. Coquet, B. T. Moretto, J. Y. Lacut et al., Partial Triplication of mtDNA in Maternally Transmitted Diabetes Mellitus and Deafness, The American Journal of Human Genetics, vol.63, issue.4, pp.1227-1232, 1998.
DOI : 10.1086/302045

J. A. Mcelhoe, M. M. Holland, K. D. Makova, M. S. Su, and I. M. Paul, Development and assessment of an optimized next-generation DNA sequencing approach for the mtgenome using the Illumina MiSeq, Forensic Science International: Genetics, vol.13, pp.20-29
DOI : 10.1016/j.fsigen.2014.05.007

L. Milane, M. Trivedi, A. Singh, M. Talekar, and M. Amiji, Mitochondrial biology, targets, and drug delivery, Journal of Controlled Release, vol.207, pp.40-58, 2015.
DOI : 10.1016/j.jconrel.2015.03.036

L. Morato, E. Bertini, D. Verrigni, A. Ardissone, and M. Ruiz, Mitochondrial dysfunction in central nervous system white matter disorders, Mitochondrial dysfunction in central nervous system white matter disorders, pp.1878-1894, 2014.
DOI : 10.1002/glia.22670

A. N. Murphy, In a flurry of PINK, mitochondrial bioenergetics takes a leading role in Parkinson's disease, EMBO Molecular Medicine, vol.28, issue.2, pp.81-84, 2009.
DOI : 10.1002/emmm.200900020

O. Musumeci, A. L. Andreu, S. Shanske, N. Bresolin, and G. P. Comi, Intragenic Inversion of mtDNA: A New Type of Pathogenic Mutation in a Patient with Mitochondrial Myopathy, The American Journal of Human Genetics, vol.66, issue.6, pp.1900-1904, 2000.
DOI : 10.1086/302927

E. Ohama, S. Ohara, F. Ikuta, K. Tanaka, and M. Nishizawa, Mitochondrial angiopathy in cerebral blood vessels of mitochondrial eneephalomyopathy, Mitochondrial angiopathy in cerebral blood vessels of mitochondrial encephalomyopathy, pp.226-233, 1987.
DOI : 10.1007/BF00688185

A. Olichon, L. J. Emorine, E. Descoins, L. Pelloquin, and L. Brichese, The human dynamin-related protein OPA1 is anchored to the mitochondrial inner membrane facing the inter-membrane space, FEBS Letters, vol.2, issue.1-3, pp.171-176, 2002.
DOI : 10.1016/S0014-5793(02)02985-X

G. Paradies, G. Petrosillo, M. Pistolese, and F. M. Ruggiero, Reactive oxygen species generated by the mitochondrial respiratory chain affect the complex III activity via cardiolipin peroxidation in beef-heart submitochondrial particles, Mitochondrion, vol.1, issue.2, pp.151-159, 2001.
DOI : 10.1016/S1567-7249(01)00011-3

M. K. Park, M. C. Ashby, G. Erdemli, O. H. Petersen, and A. V. Tepikin, Perinuclear, perigranular and sub-plasmalemmal mitochondria have distinct functions in the regulation of cellular calcium transport, The EMBO Journal, vol.20, issue.8, pp.1863-1874, 2001.
DOI : 10.1093/emboj/20.8.1863

Y. Y. Park, S. Lee, M. Karbowski, A. Neutzner, and R. J. Youle, Loss of MARCH5 mitochondrial E3 ubiquitin ligase induces cellular senescence through dynamin-related protein 1 and mitofusin 1, Journal of Cell Science, vol.123, issue.4, pp.619-626, 2010.
DOI : 10.1242/jcs.061481

W. Parson, C. Strobl, G. Huber, B. Zimmermann, and S. M. Gomes, Evaluation of next generation mtGenome sequencing using the Ion Torrent Personal Genome Machine (PGM), Forensic Science International: Genetics, vol.7, issue.5, pp.543-549
DOI : 10.1016/j.fsigen.2013.06.003

S. G. Pavlakis, P. C. Phillips, S. Dimauro, D. C. De, L. P. Vivo et al., Mitochondrial myopathy, encephalopathy, lactic acidosis, and strokelike episodes: A distinctive clinical syndrome, Annals of Neurology, vol.5, issue.4, pp.481-488, 1984.
DOI : 10.1002/ana.410160409

H. A. Pearson, J. S. Lobel, S. A. Kocoshis, J. L. Naiman, and J. Windmiller, A new syndrome of refractory sideroblastic anemia with vacuolization of marrow precursors and exocrine pancreatic dysfunction, The Journal of Pediatrics, vol.95, issue.6, pp.976-984, 1979.
DOI : 10.1016/S0022-3476(79)80286-3

V. Petruzzella, C. T. Moraes, M. C. Sano, E. Bonilla, and S. Dimauro, oxidase-negative ragged-red fibers in patients harboring a point mutation at nt 3243, Human Molecular Genetics, vol.3, issue.3, pp.449-454, 1994.
DOI : 10.1093/hmg/3.3.449

M. Phadke, M. R. Lokeshwar, S. Bhutada, C. Tampi, and R. Saxena, RETRACTED ARTICLE: Kearns Sayre Syndrome???Case Report with Review of Literature, The Indian Journal of Pediatrics, vol.106, issue.5, pp.650-654, 2012.
DOI : 10.1007/s12098-011-0618-3

J. Poulton, M. E. Deadman, and R. M. Gardiner, DUPLICATIONS OF MITOCHONDRIAL DNA IN MITOCHONDRIAL MYOPATHY, The Lancet, vol.333, issue.8632, pp.236-240, 1989.
DOI : 10.1016/S0140-6736(89)91256-7

T. R. Prezant, J. V. Agapian, M. C. Bohlman, X. Bu, and S. Oztas, Mitochondrial ribosomal RNA mutation associated with both antibiotic???induced and non???syndromic deafness, Nature Genetics, vol.51, issue.3, pp.289-294, 1993.
DOI : 10.1016/0014-5793(87)80249-1

M. Priault, B. Salin, J. Schaeffer, F. M. Vallette, and J. P. Di-rago, Impairing the bioenergetic status and the biogenesis of mitochondria triggers mitophagy in yeast, Cell Death and Differentiation, vol.16, issue.12, pp.1613-1621, 2005.
DOI : 10.1093/nar/24.13.2519

M. A. Quail, M. Smith, T. D. Coupland, S. R. Otto, and . Harris, A tale of three next generation sequencing platforms: comparison of Ion torrent, pacific biosciences and illumina MiSeq sequencers, BMC Genomics, vol.13, issue.1, p.341
DOI : 10.1186/1471-2164-13-341

C. M. Quinzii and M. Hirano, Primary and secondary CoQ10 deficiencies in humans, BioFactors, vol.5, issue.5, pp.361-365, 2011.
DOI : 10.1002/biof.155

A. Rodriguez-hernandez, M. D. Cordero, L. Salviati, R. Artuch, and M. Pineda, Coenzyme Q deficiency triggers mitochondria degradation by mitophagy, Coenzyme Q deficiency triggers mitochondria degradation by mitophagy, pp.19-32, 2009.
DOI : 10.4161/auto.5.1.7174

D. F. Rolfe and G. C. Brown, Cellular energy utilization and molecular origin of standard metabolic rate in mammals, Physiol Rev, vol.77, pp.731-758, 1997.

M. Ronaghi, S. Karamohamed, B. Pettersson, M. Uhlen, and P. Nyren, Real-Time DNA Sequencing Using Detection of Pyrophosphate Release, Analytical Biochemistry, vol.242, issue.1, pp.84-89, 1996.
DOI : 10.1006/abio.1996.0432

N. Rusk, Torrents of sequence, Nature Methods, vol.8, issue.1, pp.44-44, 2011.
DOI : 10.1038/nmeth.f.330

O. Russell and D. Turnbull, Mitochondrial DNA disease???molecular insights and potential routes to a cure, Experimental Cell Research, vol.325, issue.1, pp.38-43
DOI : 10.1016/j.yexcr.2014.03.012

P. Rustin, D. Chretien, T. Bourgeron, B. Gerard, and A. Rotig, Biochemical and molecular investigations in respiratory chain deficiencies, Clinica Chimica Acta, vol.228, issue.1, pp.35-51, 1994.
DOI : 10.1016/0009-8981(94)90055-8

J. F. Samson, P. G. Barth, J. I. De-vries, F. H. Menko, and W. Ruitenbeek, Familial mitochondrial encephalopathy with fetal ultrasonographic ventriculomegaly and intracerebral calcifications, Familial mitochondrial encephalopathy with fetal ultrasonographic ventriculomegaly and intracerebral calcifications, pp.510-516, 1994.
DOI : 10.1007/BF01957007

F. Sanger, S. Nicklen, and A. R. Coulson, DNA sequencing with chain-terminating inhibitors, Proceedings of the National Academy of Sciences, vol.74, issue.12, pp.5463-5467, 1977.
DOI : 10.1073/pnas.74.12.5463

M. Saraste, Oxidative Phosphorylation at the fin de si&egrave;cle, Science, vol.283, issue.5407, pp.1488-1493, 1999.
DOI : 10.1126/science.283.5407.1488

M. Sato and K. Sato, Maternal inheritance of mitochondrial DNA by diverse mechanisms to eliminate paternal mitochondrial DNA, Biochimica et Biophysica Acta (BBA) - Molecular Cell Research, vol.1833, issue.8, pp.1979-1984, 2013.
DOI : 10.1016/j.bbamcr.2013.03.010

L. A. Sazanov, A giant molecular proton pump: structure and mechanism of respiratory complex I, Nature Reviews Molecular Cell Biology, vol.15, issue.6, pp.375-388
DOI : 10.1038/nrm3997

H. Schagger and G. Jagow, Blue native electrophoresis for isolation of membrane protein complexes in enzymatically active form, Analytical Biochemistry, vol.199, issue.2, pp.223-231, 1991.
DOI : 10.1016/0003-2697(91)90094-A

R. Scherz-shouval and Z. Elazar, ROS, mitochondria and the regulation of autophagy, Trends in Cell Biology, vol.17, issue.9, pp.422-427, 2007.
DOI : 10.1016/j.tcb.2007.07.009

R. Scherz-shouval, E. Shvets, and Z. Elazar, Oxidation as a Post-Translational Modification that Regulates Autophagy, Autophagy, vol.3, issue.4, pp.371-373, 2007.
DOI : 10.4161/auto.4214

M. Schlame and M. Ren, Barth syndrome, a human disorder of cardiolipin metabolism, FEBS Letters, vol.288, issue.23, pp.5450-5455, 2006.
DOI : 10.1016/j.febslet.2006.07.022

S. Seneca, K. Vancampenhout, R. Van-coster, J. Smet, and W. Lissens, Analysis of the whole mitochondrial genome: translation of the Ion Torrent Personal Genome Machine system to the diagnostic bench?, European Journal of Human Genetics, vol.23, issue.1, pp.41-48, 2015.
DOI : 10.1073/pnas.0500563102

S. B. Seo, X. Zeng, J. L. King, B. L. Larue, and M. Assidi, Underlying Data for Sequencing the Mitochondrial Genome with the Massively Parallel Sequencing Platform Ion Torrent??? PGM???, Underlying Data for Sequencing the Mitochondrial Genome with the Massively Parallel Sequencing Platform Ion Torrent PGM, p.4, 2015.
DOI : 10.1101/gr.107615.110

S. Shanske, J. Coku, J. Lu, J. Ganesh, and S. Krishna, The G13513A Mutation in the ND5 Gene of Mitochondrial DNA as a Common Cause of MELAS or Leigh Syndrome, Archives of Neurology, vol.65, issue.3, pp.368-372, 2008.
DOI : 10.1001/archneurol.2007.67

E. A. Shoubridge, T. Johns, and G. Karpati, Complete restoration of a wild-type mtDNA genotype in regenerating muscle fibres in a patient with a tRNA point mutation and mitochondrial encephalomyopathy, Human Molecular Genetics, vol.6, issue.13, pp.2239-2242, 1997.
DOI : 10.1093/hmg/6.13.2239

G. Silvestri, M. Rana, F. Odoardi, A. Modoni, and E. Paris, Single-fiber PCR in MELAS3243 patients: Correlations between intratissue distribution and phenotypic expression of the mtDNAA3243G genotype, American Journal of Medical Genetics, vol.60, issue.3, pp.201-206, 2000.
DOI : 10.1002/1096-8628(20000918)94:3<201::AID-AJMG5>3.0.CO;2-2

D. Simone, F. M. Calabrese, M. Lang, G. Gasparre, and M. Attimonelli, The reference human nuclear mitochondrial sequences compilation validated and implemented on the UCSC genome browser, BMC Genomics, vol.453, issue.6, p.517, 2011.
DOI : 10.1038/nature06862

E. Smirnova, L. Griparic, D. L. Shurland, and A. M. Van-der-bliek, Dynamin-related Protein Drp1 Is Required for Mitochondrial Division in Mammalian Cells, Molecular Biology of the Cell, vol.12, issue.8, pp.2245-2256, 2001.
DOI : 10.1091/mbc.12.8.2245

W. Song, J. Chen, A. Petrilli, G. Liot, and E. Klinglmayr, Mutant huntingtin binds the mitochondrial fission GTPase dynamin-related protein-1 and increases its enzymatic activity, Nature Medicine, vol.404, issue.3, pp.377-382, 2011.
DOI : 10.1038/nm.2313

M. Stoneking, Hypervariable Sites in the mtDNA Control Region Are Mutational Hotspots, The American Journal of Human Genetics, vol.67, issue.4, pp.1029-1032, 2000.
DOI : 10.1086/303092

G. Szabadkai, A. M. Simoni, and R. Rizzuto, Mitochondrial Ca2+ Uptake Requires Sustained Ca2+ Release from the Endoplasmic Reticulum, Journal of Biological Chemistry, vol.278, issue.17, pp.15153-15161, 2003.
DOI : 10.1074/jbc.M300180200

J. W. Taanman, The mitochondrial genome: structure, transcription, translation and replication, Biochimica et Biophysica Acta (BBA) - Bioenergetics, vol.1410, issue.2, pp.103-123, 1999.
DOI : 10.1016/S0005-2728(98)00161-3

R. H. Triepels, L. P. Van-den-heuvel, J. L. Loeffen, C. A. Buskens, and R. J. Smeets, Leigh syndrome associated with a mutation in the NDUFS7 (PSST) nuclear encoded subunit of complex I, Annals of Neurology, vol.260, issue.6, pp.787-790, 1999.
DOI : 10.1002/1531-8249(199906)45:6<787::AID-ANA13>3.0.CO;2-6

D. M. Turnbull and P. Rustin, Genetic and biochemical intricacy shapes mitochondrial cytopathies, Neurobiology of Disease, vol.92, 2015.
DOI : 10.1016/j.nbd.2015.02.003

L. Valente, D. Piga, E. Lamantea, F. Carrara, and G. , Identification of novel mutations in five patients with mitochondrial encephalomyopathy, Biochimica et Biophysica Acta (BBA) - Bioenergetics, vol.1787, issue.5, pp.491-501, 2009.
DOI : 10.1016/j.bbabio.2008.10.001

S. A. Vithayathil, Y. Ma, and B. A. Kaipparettu, Transmitochondrial Cybrids: Tools for Functional Studies of Mutant Mitochondria, Methods Mol Biol, vol.837, pp.219-230
DOI : 10.1007/978-1-61779-504-6_15

J. C. Von-kleist-retzow, V. Cormier-daire, P. De-lonlay, B. Parfait, and D. Chretien, A High Rate (20%???30%) of Parental Consanguinity in Cytochrome-Oxidase Deficiency, The American Journal of Human Genetics, vol.63, issue.2, pp.428-435, 1998.
DOI : 10.1086/301957

D. C. Wallace, G. Singh, M. T. Lott, J. A. Hodge, and T. G. Schurr, Mitochondrial DNA mutation associated with Leber's hereditary optic neuropathy, Science, vol.242, issue.4884, pp.1427-1430, 1988.
DOI : 10.1126/science.3201231

J. Wang, E. S. Schmitt, M. L. Landsverk, V. W. Zhang, and F. Y. Li, An integrated approach for classifying mitochondrial DNA variants: one clinical diagnostic laboratory???s experience, Genetics in Medicine, vol.19, issue.6, pp.620-626
DOI : 10.1016/j.clinbiochem.2003.11.011

H. R. Waterham, J. Koster, C. W. Van-roermund, P. A. Mooyer, and R. J. Wanders, A Lethal Defect of Mitochondrial and Peroxisomal Fission, New England Journal of Medicine, vol.356, issue.17, pp.1736-1741, 2007.
DOI : 10.1056/NEJMoa064436

N. I. Wolf and J. A. Smeitink, Mitochondrial disorders: A proposal for consensus diagnostic criteria in infants and children, Neurology, vol.59, issue.9, pp.1402-1405, 2002.
DOI : 10.1212/01.WNL.0000031795.91814.D8

S. R. Yoshii, C. Kishi, N. Ishihara, and N. Mizushima, Parkin Mediates Proteasome-dependent Protein Degradation and Rupture of the Outer Mitochondrial Membrane, Journal of Biological Chemistry, vol.286, issue.22, pp.19630-19640, 2011.
DOI : 10.1074/jbc.M110.209338

M. Zhang, E. Mileykovskaya, and W. Dowhan, Cardiolipin Is Essential for Organization of Complexes III and IV into a Supercomplex in Intact Yeast Mitochondria, Journal of Biological Chemistry, vol.280, issue.33, pp.29403-29408, 2005.
DOI : 10.1074/jbc.M504955200

X. Zhu, X. Peng, M. X. Guan, and Q. Yan, Pathogenic mutations of nuclear genes associated with mitochondrial disorders, Acta Biochimica et Biophysica Sinica, vol.41, issue.3, pp.179-187, 2009.
DOI : 10.1093/abbs/gmn021

A. Zorzano and M. Claret, 2015 Implications of mitochondrial dynamics on neurodegeneration and on hypothalamic dysfunction, Front Aging Neurosci, vol.7, p.101

S. Zuchner, I. V. Mersiyanova, M. Muglia, N. Bissar-tadmouri, and J. Rochelle, Mutations in the mitochondrial GTPase mitofusin 2 cause Charcot-Marie-Tooth neuropathy type 2A, Mutations in the mitochondrial GTPase mitofusin 2 cause Charcot-Marie-Tooth neuropathy type 2A, pp.449-451, 2004.
DOI : 10.1083/jcb.200209124