E. H. Ahn, K. Hirohata, B. F. Kohrn, E. J. Fox, C. C. Chang et al., Detection of ultra-rare mitochondrial mutations in breast stem cells by duplex sequencing, PLoS ONE, vol.10, p.136216, 2015.

J. S. Amberger, C. A. Bocchini, F. Schiettecatte, A. F. Scott, and A. Hamosh, OMIM.org: Online Mendelian Inheritance in Man [OMIM(R)], an online catalog of human genes and genetic disorders, Nucleic Acids Res, vol.43, pp.789-798, 2015.

E. Ballana, N. Govea, R. De-cid, C. Garcia, C. Arribas et al., Detection of unrecognized low-level mtDNA heteroplasmy may explain the variable phenotypic expressivity of apparently homoplasmic mtDNA mutations, Hum. Mutat, vol.29, pp.248-257, 2008.

S. Bannwarth, V. Procaccio, A. S. Lebre, C. Jardel, A. Chaussenot et al., Prevalence of rare mitochondrial DNA mutations in mitochondrial disorders, J. Med. Genet, vol.50, pp.704-714, 2013.

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, Hum. Mutat, vol.25, pp.575-582, 2005.

D. R. Bentley, S. Balasubramanian, H. P. Swerdlow, G. P. Smith, J. Milton et al., Accurate whole human genome sequencing using reversible terminator chemistry, Nature, vol.456, pp.53-59, 2008.

M. Bitner-glindzicz, M. Pembrey, A. Duncan, J. Heron, S. M. Ring et al., Prevalence of mitochondrial 1555A->G mutation in European children, N. Engl. J. Med, vol.360, pp.640-642, 2009.

J. K. Blackwood, R. G. Whittaker, E. L. Blakely, C. L. Alston, D. M. Turnbull et al., The investigation and diagnosis of pathogenic mitochondrial DNA mutations in human urothelial cells, Biochem. Biophys. Res. Commun, vol.393, pp.740-745, 2010.

M. D. Brown, E. Starikovskaya, O. Derbeneva, S. Hosseini, J. C. Allen et al., The role of mtDNA background in disease expression: a new primary LHON mutation associated with Western Eurasian haplogroup, J. Hum. Genet, vol.110, pp.130-138, 2002.

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

L. Caporali, L. Iommarini, C. La-morgia, A. Olivieri, A. Achilli et al., Peculiar combinations of individually non-pathogenic missense mitochondrial DNA variants cause low penetrance Leber's hereditary optic neuropathy, PLoS Genet, vol.14, p.1007210, 2018.

L. Caporali, A. Maresca, M. Capristo, V. Del-dotto, F. Tagliavini et al., Incomplete penetrance in mitochondrial optic neuropathies, Mitochondrion, vol.36, pp.130-137, 2017.

C. Salas, P. Palma-milla, C. Lopez-montiel, J. Benito, C. Franco-freire et al., Leber hereditary optic neuropathy: usefulness of next generation sequencing to study mitochondrial mutations on apparent homoplasmy, Med. Clin, vol.146, pp.163-166, 2016.

S. Castellana, C. Fusilli, G. Mazzoccoli, T. Biagini, D. Capocefalo et al., High-confidence assessment of functional impact of human mitochondrial non-synonymous genome variations by APOGEE, PLoS Comput. Biol, vol.13, p.1005628, 2017.

S. Castellana, J. Ronai, and T. Mazza, MitImpact: an exhaustive collection of pre-computed pathogenicity predictions of human mitochondrial non-synonymous variants, Hum. Mutat, vol.36, pp.2413-2422, 2015.

R. Clima, R. Preste, C. Calabrese, M. A. Diroma, M. Santorsola et al., HmtDB 2016: data update, a better performing query system and human mitochondrial DNA haplogroup predictor, Nucleic Acids Res, vol.45, pp.698-706, 2017.

H. Cui, F. Li, D. Chen, G. Wang, C. K. Truong et al., Comprehensive next-generation sequence analyses of the entire mitochondrial genome reveal new insights into the molecular diagnosis of mitochondrial DNA disorders, Genet. Med, vol.15, pp.388-394, 2013.

J. Damas, J. Carneiro, A. Amorim, and F. Pereira, MitoBreak: the mitochondrial DNA breakpoints database, Nucleic Acids Res, vol.42, pp.1261-1268, 2014.

M. M. Davidson, W. F. Walker, E. Hernandez-rosa, and C. Nesti, Evidence for nuclear modifier gene in mitochondrial cardiomyopathy, J. Mol. Cell. Cardiol, vol.46, pp.936-942, 2009.

K. Dimitriadis, M. Leonhardt, P. Yu-wai-man, M. A. Kirkman, A. Korsten et al., Leber's hereditary optic neuropathy with late disease onset: clinical and molecular characteristics of 20 patients, J. Rare Dis, vol.9, p.158, 2014.

A. El-meziane, S. K. Lehtinen, N. Hance, L. G. Nijtmans, D. Dunbar et al., A tRNA suppressor mutation in human mitochondria, Nat. Genet, vol.18, pp.350-353, 1998.

H. R. Elliott, D. C. Samuels, J. A. Eden, C. L. Relton, and P. F. Chinnery, Pathogenic mitochondrial DNA mutations are common in the general population, Am. J. Hum. Genet, vol.83, pp.254-260, 2008.

J. L. Elson, P. M. Smith, L. C. Greaves, R. N. Lightowlers, Z. M. Chrzanowska-lightowlers et al., The presence of highly disruptive 16S rRNA mutations in clinical samples indicates a wider role for mutations of the mitochondrial ribosome in human disease, Mitochondrion, vol.25, pp.17-27, 2015.

S. Emperador, M. Vidal, C. Hernández-ainsa, C. Ruiz-ruiz, D. Woods et al., The decrease in mitochondrial DNA mutation load parallels visual recovery in a leber hereditary optic neuropathy patient, Front. Neurosci, vol.12, p.61, 2018.

X. Estivill, N. Govea, E. Barcelo, C. Badenas, E. Romero et al., Familial progressive sensorineural deafness is mainly due to the mtDNA A1555G mutation and is enhanced by treatment of aminoglycosides, Am. J. Hum. Genet, vol.62, pp.27-35, 1998.

L. Fan and Y. G. Yao, MitoTool: a web server for the analysis and retrieval of human mitochondrial DNA sequence variations, Mitochondrion, vol.11, pp.351-356, 2011.

A. Fayssoil, P. Laforet, W. Bougouin, C. Jardel, A. Lombes et al., Prediction of long-term prognosis by heteroplasmy levels of the m.3243A>G mutation in patients with the mitochondrial encephalomyopathy, lactic acidosis and stroke-like episodes syndrome, Eur. J. Neurol, vol.24, pp.255-261, 2017.

S. Frey, G. Geffroy, V. Desquiret-dumas, N. Gueguen, C. Bris et al., The addition of ketone bodies alleviates mitochondrial dysfunction by restoring complex I assembly in a MELAS cellular model, Biochim. Biophys. Acta, vol.1863, pp.284-291, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01396751

A. Genasetti, M. L. Valentino, V. Carelli, D. Vigetti, M. Viola et al., Assessing heteroplasmic load in Leber's hereditary optic neuropathy mutation 3460G->A/MT-ND1 with a real-time PCR quantitative approach, J. Mol. Diagn, vol.9, pp.538-545, 2007.

A. Ghelli, A. M. Porcelli, C. Zanna, S. Vidoni, S. Mattioli et al., The background of mitochondrial DNA haplogroup J increases the sensitivity of Leber's hereditary optic neuropathy cells to 2,5-hexanedione toxicity, PLoS ONE, vol.4, p.7922, 2009.

C. Giordano, L. Iommarini, L. Giordano, A. Maresca, A. Pisano et al., Efficient mitochondrial biogenesis drives incomplete penetrance in Leber's hereditary optic neuropathy, Brain, vol.137, issue.2, pp.335-353, 2014.

L. Giordano, S. Deceglie, P. Adamo, M. L. Valentino, C. La-morgia et al., Cigarette toxicity triggers Leber's hereditary optic neuropathy by affecting mtDNA copy number, oxidative phosphorylation and ROS detoxification pathways, Cell Death Dis, vol.6, p.2021, 2015.

A. Gomez-duran, D. Pacheu-grau, I. Martinez-romero, E. Lopez-gallardo, M. J. Lopez-perez et al., Oxidative phosphorylation differences between mitochondrial DNA haplogroups modify the risk of Leber's hereditary optic neuropathy, Biochim. Biophys. Acta, vol.1822, pp.1216-1222, 2012.

G. S. Gorman, A. M. Schaefer, Y. Ng, N. Gomez, E. L. Blakely et al., Prevalence of nuclear and mitochondrial DNA mutations related to adult mitochondrial disease, Ann. Neurol, vol.77, pp.753-759, 2015.

J. P. Grady, J. L. Murphy, E. L. Blakely, R. G. Haller, R. W. Taylor et al., Accurate measurement of mitochondrial DNA deletion level and copy number differences in human skeletal muscle, PLoS ONE, vol.9, p.114462, 2014.

J. P. Grady, S. J. Pickett, Y. S. Ng, C. L. Alston, E. L. Blakely et al., mtDNA heteroplasmy level and copy number indicate disease burden in m.3243A>G mitochondrial disease, EMBO Mol. Med, vol.10, p.8262, 2018.

R. C. Green, J. S. Berg, W. W. Grody, S. S. Kalia, B. R. Korf et al., ACMG recommendations for reporting of incidental findings in clinical exome and genome sequencing, Genet. Med, vol.15, pp.565-574, 2013.

H. R. Griffin, A. Pyle, E. L. Blakely, C. L. Alston, J. Duff et al., Accurate mitochondrial DNA sequencing using off-target reads provides a single test to identify pathogenic point mutations, Genet. Med, vol.16, pp.962-971, 2014.

Y. Guo, C. Li, Q. Sheng, J. F. Winther, Q. Cai et al., Very low-level heteroplasmy mtDNA variations are inherited in humans, J. Genet. Genomics, vol.40, pp.607-615, 2013.

L. He, P. F. Chinnery, S. E. Durham, E. L. Blakely, T. M. Wardell et al., Detection and quantification of mitochondrial DNA deletions in individual cells by real-time PCR, Nucleic Acids Res, vol.30, p.68, 2002.

A. H. Henrie, S. E. Hemphill, N. Ruiz-schultz, B. Cushman, M. T. Distefano et al., ClinVar Miner: demonstrating utility of a webbased tool for viewing and filtering ClinVar data, Hum. Mutat, vol.39, pp.1051-1060, 2018.

T. Huang, Next generation sequencing to characterize mitochondrial genomic DNA heteroplasmy, Curr. Protoc. Hum. Genet. Chapter, vol.19, 2011.

G. Hudson, M. Nalls, J. R. Evans, D. P. Breen, S. Winder-rhodes et al., Two-stage association study and meta-analysis of mitochondrial DNA variants in Parkinson disease, Neurology, vol.80, pp.2042-2048, 2013.

Y. Inagaki, Y. Mashima, N. Fuse, Y. Ohtake, T. Fujimaki et al., , 2006.

, Mitochondrial DNA mutations with Leber's hereditary optic neuropathy in Japanese patients with open-angle glaucoma, Jpn. J. Ophthalmol, vol.50, pp.128-134

M. Ingman, H. Kaessmann, S. Paabo, and U. Gyllensten, Mitochondrial genome variation and the origin of modern humans, Nature, vol.408, pp.708-713, 2000.

P. Jiang, X. Jin, Y. Peng, M. Wang, H. Liu et al., The exome sequencing identified the mutation in YARS2 encoding the mitochondrial tyrosyl-tRNA synthetase as a nuclear modifier for the phenotypic manifestation of Leber's hereditary optic neuropathy-associated mitochondrial DNA mutation, Hum. Mol. Genet, vol.25, pp.584-596, 2016.

S. S. Kalia, K. Adelman, S. J. Bale, W. K. Chung, C. Eng et al., Recommendations for reporting of secondary findings in clinical exome and genome sequencing, 2016 update (ACMG SF v2.0): a policy statement of the American College of Medical Genetics and Genomics, Genet. Med, vol.19, pp.249-255, 2017.

N. A. Khan, P. Govindaraj, V. Jyothi, A. K. Meena, and K. Thangaraj, Cooccurrence of m.1555A>G and m.11778G>A mitochondrial DNA mutations in two Indian families with strikingly different clinical penetrance of Leber hereditary optic neuropathy, Mol. Vis, vol.19, pp.1282-1289, 2013.

A. M. Kogelnik, M. T. Lott, M. D. Brown, S. B. Navathe, and D. C. Wallace, MITOMAP: a human mitochondrial genome database, Nucleic Acids Res, vol.24, pp.177-179, 1996.

M. J. Landrum, J. M. Lee, M. Benson, G. Brown, C. Chao et al., ClinVar: public archive of interpretations of clinically relevant variants, Nucleic Acids Res, vol.44, pp.862-868, 2016.

M. Lek, K. J. Karczewski, E. V. Minikel, K. E. Samocha, E. Banks et al., Analysis of protein-coding genetic variation in 60,706 humans, Nature, vol.536, pp.285-291, 2016.

D. G. Leung, J. S. Cohen, E. H. Michelle, R. Bai, A. L. Mammen et al., Mitochondrial DNA deletions with low-level heteroplasmy in adult-onset myopathy, J. Clin. Neuromuscul. Dis, vol.19, pp.117-123, 2018.

D. S. Lieber, Computational and Experimental Approaches for Evaluating the Genetic Basis of Mitochondrial Disorders, 2013.

H. Liu, Y. Ma, F. Fang, Y. Zhang, L. Zou et al., Wildtype mitochondrial DNA copy number in urinary cells as a useful marker for diagnosing severity of the mitochondrial diseases, PLoS ONE, vol.8, p.67146, 2013.

M. T. Lott, J. N. Leipzig, O. Derbeneva, H. M. Xie, D. Chalkia et al., mtDNA variation and analysis using mitomap and mitomaster, 2013.

, Curr. Protoc. Bioinformatics, vol.44, pp.1-23

L. A. Marcelino and W. G. Thilly, Mitochondrial mutagenesis in human cells and tissues, Mutat. Res, vol.434, pp.177-203, 1999.

J. Marquis, G. Lefebvre, Y. A. Kourmpetis, M. Kassam, F. Ronga et al., MitoRS, a method for high throughput, sensitive, and accurate detection of mitochondrial DNA heteroplasmy, BMC Genomics, vol.18, p.326, 2017.

A. Martin-navarro, A. Gaudioso-simon, J. Alvarez-jarreta, J. Montoya, E. Mayordomo et al., Machine learning classifier for identification of damaging missense mutations exclusive to human mitochondrial DNA-encoded polypeptides, BMC Bioinformatics, vol.18, p.158, 2017.

E. Mccormick, E. Place, and M. J. Falk, Molecular genetic testing for mitochondrial disease: from one generation to the next, Neurotherapeutics, vol.10, pp.251-261, 2013.

M. T. Mcdonnell, A. M. Schaefer, E. L. Blakely, R. Mcfarland, P. F. Chinnery et al., Noninvasive diagnosis of the 3243A > G mitochondrial DNA mutation using urinary epithelial cells, Eur. J. Hum. Genet, vol.12, pp.778-781, 2004.

J. A. Mcelhoe, M. M. Holland, K. D. Makova, M. S. Su, I. M. Paul et al., Development and assessment of an optimized nextgeneration DNA sequencing approach for the mtgenome using the Illumina MiSeq, Forensic Sci. Int. Genet, vol.13, pp.20-29, 2014.

R. Mcfarland, J. L. Elson, R. W. Taylor, N. Howell, and D. M. Turnbull, Assigning pathogenicity to mitochondrial tRNA mutations: when "definitely maybe" is not good enough, Trends Genet, vol.20, pp.591-596, 2004.

A. L. Mitchell, J. L. Elson, N. Howell, R. W. Taylor, and D. M. Turnbull, Sequence variation in mitochondrial complex I genes: mutation or polymorphism?, J. Med. Genet, vol.43, pp.175-179, 2006.

C. T. Moraes, D. P. Atencio, J. Oca-cossio, and F. Diaz, Techniques and pitfalls in the detection of pathogenic mitochondrial DNA mutations, J. Mol. Diagn, vol.5, issue.10, pp.60474-60480, 2003.

D. Navarro-gomez, J. Leipzig, L. Shen, M. Lott, A. P. Stassen et al., Phy-Mer: a novel alignment-free and reference-independent mitochondrial haplogroup classifier, Bioinformatics, vol.31, pp.1310-1312, 2015.

Y. S. Ng, N. Z. Lax, P. Maddison, C. L. Alston, E. L. Blakely et al., MT-ND5 mutation exhibits highly variable neurological manifestations at low mutant load. EBioMedicine, vol.30, pp.86-93, 2018.

A. Niroula and M. Vihinen, PON-mt-tRNA: a multifactorial probabilitybased method for classification of mitochondrial tRNA variations, Nucleic Acids Res, vol.44, pp.2020-2027, 2016.

W. Parson and A. Dur, EMPOP-a forensic mtDNA database, Forensic Sci. Int. Genet, vol.1, pp.88-92, 2007.

A. Patowary, R. Nesbitt, M. Archer, R. Bernier, and Z. Brkanac, Next generation sequencing mitochondrial DNA analysis in autism spectrum disorder, Autism Res, vol.10, pp.1338-1343, 2017.

R. Preste, O. Vitale, R. Clima, G. Gasparre, A. et al., HmtVar: a new resource for human mitochondrial variations and pathogenicity data, Nucleic Acids Res. gky1024, 2018.

V. Procaccio, N. Neckelmann, V. Paquis-flucklinger, S. Bannwarth, R. Jimenez et al., Detection of low levels of the mitochondrial tRNALeu(UUR) 3243A>G mutation in blood derived from patients with diabetes, Mol. Diagn. Ther, vol.10, pp.381-389, 2006.

J. Putz, M. Dupuis-b-fau--sissler, M. Sissler, C. Fau--florentz, and C. Florentz, Mamit-tRNA, a database of mammalian mitochondrial tRNA primary and secondary structures, RNA, vol.13, pp.1184-1190, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00200384

S. Rahman, J. Poulton, D. Marchington, and A. Suomalainen, Decrease of 3243 A->G mtDNA mutation from blood in MELAS syndrome: a longitudinal study, Am. J. Hum. Genet, vol.68, pp.238-240, 2001.

S. Richards, N. Aziz, S. Bale, D. Bick, S. Das et al., Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology, Genet. Med, vol.17, pp.405-424, 2015.

R. Rossignol, B. Faustin, C. Rocher, M. Malgat, J. P. Mazat et al., Mitochondrial threshold effects, Biochem, J, vol.370, pp.751-762, 2003.

M. Rueda and A. Torkamani, SG-ADVISER mtDNA: a web server for mitochondrial DNA annotation with data from 200 samples of a healthy aging cohort, BMC Bioinformatics, vol.18, p.373, 2017.

E. Ruiz-pesini, M. T. Lott, V. Procaccio, J. C. Poole, M. C. Brandon et al., An enhanced MITOMAP with a global mtDNA mutational phylogeny, Nucleic Acids Res, vol.35, pp.823-828, 2007.

A. M. Schaefer, R. Mcfarland, E. L. Blakely, L. He, R. G. Whittaker et al., Prevalence of mitochondrial DNA disease in adults, Ann. Neurol, vol.63, pp.35-39, 2008.

M. W. Schmitt, S. R. Kennedy, J. J. Salk, E. J. Fox, J. B. Hiatt et al., Detection of ultra-rare mutations by next-generation sequencing, Proc. Natl. Acad. Sci. U.S.A, vol.109, pp.14508-14513, 2012.

S. Seneca, K. Vancampenhout, R. Van-coster, J. Smet, W. Lissens et al., Analysis of the whole mitochondrial genome: translation of the Ion Torrent Personal Genome Machine system to the diagnostic bench?, Eur. J. Hum. Genet, vol.23, pp.41-48, 2015.

L. Shen, M. Attimonelli, R. Bai, M. T. Lott, D. C. Wallace et al., MSeqDR mvTool: a mitochondrial DNA Web and API resource for comprehensive variant annotation, universal nomenclature collation, and reference genome conversion, Hum. Mutat, vol.39, pp.806-810, 2018.

P. M. Smith, J. L. Elson, L. C. Greaves, S. B. Wortmann, R. J. Rodenburg et al., The role of the mitochondrial ribosome in human disease: searching for mutations in 12S mitochondrial rRNA with high disruptive potential, Hum. Mol. Genet, vol.23, pp.949-967, 2014.

S. Sonney, J. Leipzig, M. T. Lott, S. Zhang, V. Procaccio et al., Predicting the pathogenicity of novel variants in mitochondrial tRNA with MitoTIP, PLoS Comput. Biol, vol.13, p.1005867, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-02149524

M. X. Sosa, I. K. Sivakumar, S. Maragh, V. Veeramachaneni, R. Hariharan et al., Next-generation sequencing of human mitochondrial reference genomes uncovers high heteroplasmy frequency, PLoS Comput. Biol, vol.8, p.1002737, 2012.

S. Tang, J. Wang, V. W. Zhang, F. Y. Li, M. Landsverk et al., Transition to next generation analysis of the whole mitochondrial genome: a summary of molecular defects, Hum. Mutat, vol.34, pp.882-893, 2013.

D. R. Thorburn, C. Sugiana, R. Salemi, D. M. Kirby, L. Worgan et al., Biochemical and molecular diagnosis of mitochondrial respiratory chain disorders, Biochim. Biophys. Acta, vol.1659, pp.121-128, 2004.

M. Urata, Y. Wada, S. H. Kim, W. Chumpia, Y. Kayamori et al., High-sensitivity detection of the A3243G mutation of mitochondrial DNA by a combination of allele-specific PCR and peptide nucleic acid-directed PCR clamping, Clin. Chem, vol.50, pp.2045-2051, 2004.

E. M. Van-der-walt, I. Smuts, R. W. Taylor, J. L. Elson, D. M. Turnbull et al., Characterization of mtDNA variation in a cohort of South African paediatric patients with mitochondrial disease, Eur. J. Hum. Genet, vol.20, pp.650-656, 2012.

J. M. Van-der-walt, Y. A. Dementieva, E. R. Martin, W. K. Scott, K. K. Nicodemus et al., Analysis of European mitochondrial haplogroups with Alzheimer disease risk, Neurosci. Lett, vol.365, pp.28-32, 2004.

M. Van-oven and M. Kayser, Updated comprehensive phylogenetic tree of global human mitochondrial DNA variation, Hum. Mutat, vol.30, pp.386-394, 2009.

K. Vancampenhout, B. Caljon, C. Spits, K. Stouffs, A. Jonckheere et al., A bumpy ride on the diagnostic bench of massive parallel sequencing, the case of the mitochondrial genome, PLoS ONE, vol.9, p.112950, 2014.

H. Vandebona, P. Mitchell, N. Manwaring, K. Griffiths, B. Gopinath et al., Prevalence of mitochondrial 1555A->G mutation in adults of European descent, N. Engl. J. Med, vol.360, pp.642-644, 2009.

S. K. Vellarikkal, H. Dhiman, K. Joshi, Y. Hasija, S. Sivasubbu et al., mit-o-matic: a comprehensive computational pipeline for clinical evaluation of mitochondrial variations from next-generation sequencing datasets, Hum. Mutat, vol.36, pp.419-424, 2015.

D. C. Wallace, W. Fan, and V. Procaccio, Mitochondrial energetics and therapeutics, Annu. Rev. Pathol, vol.5, pp.297-348, 2010.

D. C. Wallace, M. T. Lott, and V. Procaccio, Mitochondrial medicine: the mitochondrial biology and genetics of metabolic and degenerative diseases, cancer, and aging, " in Emery and Rimoin's Principle and Practice of Medical Genetics, pp.1-153, 2013.

J. Wang, R. Al-ouran, Y. Hu, S. Y. Kim, Y. W. Wan et al., MARRVEL: integration of human and model organism genetic resources to facilitate functional annotation of the human genome, Am. J. Hum. Genet, vol.100, pp.843-853, 2017.

J. Wang, E. S. Schmitt, M. L. Landsverk, V. W. Zhang, F. Y. Li et al., An integrated approach for classifying mitochondrial DNA variants: one clinical diagnostic laboratory's experience, Genet. Med, vol.14, pp.620-626, 2012.

W. Wei, A. Gomez-duran, G. Hudson, and P. F. Chinnery, Background sequence characteristics influence the occurrence and severity of disease-causing mtDNA mutations, PLoS Genet, vol.13, p.1007126, 2017.

H. Weissensteiner, L. Forer, C. Fuchsberger, B. Schopf, A. Kloss-brandstatter et al., mtDNA-Server: next-generation sequencing data analysis of human mitochondrial DNA in the cloud, Nucleic Acids Res, vol.44, pp.64-69, 2016.

H. Weissensteiner, D. Pacher, A. Kloss-brandstatter, L. Forer, G. Specht et al., HaploGrep 2: mitochondrial haplogroup classification in the era of high-throughput sequencing, Nucleic Acids Res, vol.44, pp.58-63, 2016.

C. Wolf, E. Gramer, B. Muller-myhsok, F. Pasutto, B. Wissinger et al., Mitochondrial haplogroup U is associated with a reduced risk to develop exfoliation glaucoma in the German population, BMC Genet, vol.11, issue.8, 2010.

L. J. Wong, Pathogenic mitochondrial DNA mutations in protein-coding genes, Muscle Nerve, vol.36, pp.279-293, 2007.

L. J. Wong, Molecular genetics of mitochondrial disorders, Dev. Disabil. Res. Rev, vol.16, pp.154-162, 2010.

L. J. Wong, Challenges of bringing next generation sequencing technologies to clinical molecular diagnostic laboratories, Neurotherapeutics, vol.10, pp.262-272, 2013.

L. J. Wong and R. G. Boles, Mitochondrial DNA analysis in clinical laboratory diagnostics, Clin. Chim. Acta, vol.354, pp.1-20, 2005.

H. Yang, R. Liu, and C. C. Wang, Searching the co-occurrence of pathogenic mutations for Leber's hereditary optic neuropathy and hearing loss in more than 26,000 whole mitochondrial genomes. Mitochondrial DNA A DNA Mapp, Seq. Anal, vol.27, pp.3399-3402, 2016.

F. Ye, D. C. Samuels, T. Clark, and Y. Guo, High-throughput sequencing in mitochondrial DNA research, Mitochondrion, vol.17, pp.157-163, 2014.

P. Yu-wai-man, G. Hudson, T. Klopstock, and P. F. Chinnery, Reply: parsing the differences in affected with LHON: genetic versus environmental triggers of disease conversion, Brain, vol.139, p.18, 2016.

M. V. Zaragoza, J. Fass, M. Diegoli, D. Lin, A. et al., , 2010.

, Mitochondrial DNA variant discovery and evaluation in human Cardiomyopathies through next-generation sequencing, PLoS ONE, vol.5, p.12295

W. Zhang, H. Cui, and L. J. Wong, Comprehensive one-step molecular analyses of mitochondrial genome by massively parallel sequencing, Clin. Chem, vol.58, pp.1322-1331, 2012.