REFERENCES

1. Gorman GS, Chinnery PF, DiMauro S, Hirano M, Koga Y, et al. Mitochondrial diseases. Nat Rev Dis Primers 2016;2:16080.

2. DiMauro S, Schon EA, Carelli V, Hirano M. The clinical maze of mitochondrial neurology. Nat Rev Neurol 2013;9:429-44.

3. Duran J, Martinez A, Adler E. Cardiovascular manifestations of mitochondrial disease. Biology 2019;8:34.

4. Zong WX, Rabinowitz JD, White E. Mitochondria and Cancer. Mol Cell 2016;61:667-76.

5. McCormick EM, Muraresku CC, Falk MJ. Mitochondrial Genomics: a complex field now coming of age. Curr Genet Med Rep 2018;6:52-61.

6. Osellame LD, Blacker TS, Duchen MR. Cellular and molecular mechanisms of mitochondrial function. Best Pract Res Clin Endocrinol Metab 2012;26:711-23.

7. Tuppen HAL, Blakely EL, Turnbull DM, Taylor RW. Mitochondrial DNA mutations and human disease. Biochim Biophys Acta Bioenerg 2010;1797:113-28.

8. Greaves LC, Nooteboom M, Elson JL, Tuppen HAL, Taylor GA, et al. Clonal expansion of early to mid-life mitochondrial DNA point mutations drives mitochondrial dysfunction during human ageing. PLoS Genet 2014;10:e1004620.

9. Castellanos E, Lanning NJ. Phosphorylation of OXPHOS machinery subunits: functional implications in cell biology and disease. Yale J Biol Med 2019;92:523-31.

10. Natarajan V, Chawla R, Mah T, Vivekanandan R, Tan SY, et al. Mitochondrial dysfunction in age-related metabolic disorders. Proteomics 2020;1800404.

11. Nass S, Nass MM. Intramitochondrial fibers with DNA characteristics. II. enzymatic and other hydrolytic treatments. J Cell Biology 1963;19:613-29.

12. Anderson S, Bankier AT, Barrell BG, de Bruijn MHL, Coulson AR, et al. Sequence and organization of the human mitochondrial genome. Nature 1981;290:457-65.

13. Andrews RM, Kubacka I, Chinnery PF, Lightowlers RN, Turnbull DM, et al. Reanalysis and revision of the Cambridge reference sequence for human mitochondrial DNA. Nat Genet 1999;23:147.

14. D’Souza AR, Minczuk M. Mitochondrial transcription and translation: overview. Essays Biochem 2018;62:309-20.

15. Russell O, Turnbull D. Mitochondrial DNA disease-molecular insights and potential routes to a cure. Exp Cell Res 2014;325:38-43.

16. Bayir H, Kagan VE. Bench-to-bedside review: mitochondrial injury, oxidative stress and apoptosis--there is nothing more practical than a good theory. Critical Care (London, England) 2008;12:206.

17. Wallace DC, Brown MD, Lott MT. Mitochondrial DNA variation in human evolution and disease. Gene 1999;238:211-30.

18. Gorman GS, Schaefer AM, Ng Y, Gomez N, Blakely EL, et al. Prevalence of nuclear and mitochondrial DNA mutations related to adult mitochondrial disease. Ann Neurol 2015;77:753-9.

19. Skladal D, Halliday J, Thorburn DR. Minimum birth prevalence of mitochondrial respiratory chain disorders in children. Brain 2003;126:1905-12.

20. DiMauro S. Mitochondrial encephalomyopathies--fifty years on: the Robert Wartenberg Lecture. Neurology 2013;81:281-91.

21. Frazier AE, Thorburn DR, Compton AG. Mitochondrial energy generation disorders: genes, mechanisms, and clues to pathology. J Biol Chem 2019;294:5386-95.

22. Lebon S, Chol M, Benit P, Mugnier C, Chretien D, et al. Recurrent de novo mitochondrial DNA mutations in respiratory chain deficiency. J Med Genet 2003;40:896-9.

23. Chinnery PF, Craven L, Mitalipov S, Stewart JB, Herbert M, et al. The challenges of mitochondrial replacement. PLoS genet 2014;10:e1004315.

24. Schon EA, Manfredi G. Neuronal degeneration and mitochondrial dysfunction. J Clin Invest 2003;111:303-12.

25. Pek NMQ, Phua QH, Ho BX, Pang JKS, Hor JH, et al. Mitochondrial 3243A > G mutation confers pro-atherogenic and pro-inflammatory properties in MELAS iPS derived endothelial cells. Cell Death Dis 2019;10:802.

26. Kucharczyk R, Dautant A, Gombeau K, Godard F, Tribouillard-Tanvier D, et al. The pathogenic MT-ATP6 m.8851T>C mutation prevents proton movements within the n-side hydrophilic cleft of the membrane domain of ATP synthase. Biochim Biophys Acta Bioenerg 2019;1860:562-72.

27. Blanco-Grau A, Bonaventura-Ibars I, Coll-Cantí J, Melià MJ, Martinez R, et al. Identification and biochemical characterization of the novel mutation m.8839G>C in the mitochondrial ATP6 gene associated with NARP syndrome. Genes Brain Behav 2013;12:812-20.

28. Mordel P, Schaeffer S, Dupas Q, Laville MA, Gérard M, et al. A 2 bp deletion in the mitochondrial ATP 6 gene responsible for the NARP (neuropathy, ataxia, and retinitis pigmentosa) syndrome. Biochem Biophys Res Commun 2017;494:133-7.

29. Rahman S, Blok RB, Dahl HHM, Danks DM, Kirby DM, et al. Leigh syndrome: clinical features and biochemical and DNA abnormalities. Ann Neurol 1996;39:343-51.

30. Alston CL, Veling MT, Heidler J, Taylor LS, Alaimo JT, et al. Pathogenic Bi-allelic Mutations in NDUFAF8 cause leigh syndrome with an isolated complex I deficiency. Am J Hum Genet 2020;106:92-101.

31. Park SY, Kim SH, Lee YM. Molecular diagnosis of myoclonus epilepsy associated with ragged-red fibers syndrome in the absence of ragged red fibers. Front Neurol 2017;8:520.

32. Mackey DA, Oostra RJ, Rosenberg T, Nikoskelainen E, Bronte-Stewart J, et al. Primary pathogenic mtDNA mutations in multigeneration pedigrees with Leber hereditary optic neuropathy. Am J Hum Genet 1996;59:481-5.

33. Manickam AH, Michael MJ, Ramasamy S. Mitochondrial genetics and therapeutic overview of Leber’s hereditary optic neuropathy. Indian J Ophthalmol 2017;65:1087-92.

34. Emperador S, López-Gallardo E, Hernández-Ainsa C, Habbane M, Montoya J, et al. Ketogenic treatment reduces the percentage of a LHON heteroplasmic mutation and increases mtDNA amount of a LHON homoplasmic mutation. Orphanet J Rare dis 2019;14:150.

35. Remes AM, Peuhkurinen KJ, Herva R, Majamaa K, Hassinen IE. Kearns-Sayre syndrome case presenting a mitochondrial DNA deletion with unusual direct repeats and a rudimentary RNAase mitochondrial ribonucleotide processing target sequence. Genomics 1993;16:256-8.

36. Poulton J, Deadman M, Mark Gardiner R. Duplications of mitochondrial dna in mitochondrial myopathy. Lancet 1989;333:236-40.

37. Shoubridge EA, Johns T, Karpati G. Complete restoration of a wild-type mtDNA genotype in regenerating muscle fibres in a patient with a tRNA point mutation and mitochondrial encephalomyopathy. Hum Mol Genet 1997;6:2239-42.

38. Pineda M, Ormazabal A, López-Gallardo E, Nascimento A, Solano A, et al. Cerebral folate deficiency and leukoencephalopathy caused by a mitochondrial DNA deletion. Ann Neurol 2006;59:394-8.

39. Patel R, Coulter LL, Rimmer J, Parkes M, Chinnery PF, et al. Mitochondrial neurogastrointestinal encephalopathy: a clinicopathological mimic of Crohn’s disease. BMC Gastroenterol 2019;19:11.

40. Nishino I, Spinazzola A, Hirano M. Thymidine phosphorylase gene mutations in MNGIE, a human mitochondrial disorder. Science 1999;283:689-92.

41. González-Vioque E, Torres-Torronteras J, Andreu AL, Martí R. Limited dCTP availability accounts for mitochondrial DNA depletion in mitochondrial neurogastrointestinal encephalomyopathy (MNGIE). PLoS genet 2011;7:e1002035.

42. Shaibani A, Shchelochkov OA, Zhang S, Katsonis P, Lichtarge O, et al. Mitochondrial neurogastrointestinal encephalopathy due to mutations in RRM2B. Arch Neurol 2009;66:1028-32.

43. Tang S, Dimberg EL, Milone M, Wong LJC. Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE)-like phenotype: an expanded clinical spectrum of POLG1 mutations. J Neurol 2012;259:862-8.

44. Saneto RP, Cohen BH, Copeland WC, Naviaux RK. Alpers-Huttenlocher syndrome. Pediatr Neurol 2013;48:167-78.

45. El-Hattab AW, Scaglia F. Mitochondrial DNA depletion syndromes: review and updates of genetic basis, manifestations, and therapeutic options. Neurotherapeutics 2013;10:186-98.

46. Suomalainen A, Isohanni P. Mitochondrial DNA depletion syndromes--many genes, common mechanisms. Neuromuscul Disord 2010;20:429-37.

47. Rahman S, Copeland WC. POLG-related disorders and their neurological manifestations. Nat Rev Neurol 2019;15:40-52.

48. de Vries DD, van Engelen BGM, Gabreëls FJM, Ruitenbeek W, van Oost BA. A second missense mutation in the mitochondrial ATPase 6 gene in Leigh’s syndrome. Ann Neurol 1993;34:410-2.

49. Harding AE, Holt IJ, Sweeney MG, Brockington M, Davis MB. Prenatal diagnosis of mitochondrial DNA8993 T----G disease. Am J Hum Genet 1992;50:629-33.

50. Holt IJ, Harding AE, Petty RK, Morgan-Hughes JA. A new mitochondrial disease associated with mitochondrial DNA heteroplasmy. Am J Hum Genet 1990;46:428-33.

51. Carrozzo R, Murray J, Santorelli FM, Capaldi RA. The T9176G mutation of human mtDNA gives a fully assembled but inactive ATP synthase when modeled in Escherichia coli. FEBS Letters 2000;486:297-9.

52. Verny C, Guegen N, Desquiret V, Chevrollier A, Prundean A, et al. Hereditary spastic paraplegia-like disorder due to a mitochondrial ATP6 gene point mutation. Mitochondrion 2011;11:70-5.

53. Thyagarajan D, Shanske S, Vazquez-Memije M, Devivo D, Dimauro S. A novel mitochondrial ATPase 6 point mutation in familial bilateral striatal necrosis. Ann Neurol 1995;38:468-72.

54. Moslemi AR, Darin N, Tulinius M, Oldfors A, Holme E. Two new mutations in the MTATP6 gene associated with Leigh syndrome. Neuropediatrics 2005;36:314-8.

55. Tiranti V, Corona P, Greco M, Taanman JW, Carrara F, et al. A novel frameshift mutation of the mtDNA COIII gene leads to impaired assembly of cytochrome c oxidase in a patient affected by Leigh-like syndrome. Hum Mol Genet 2000;9:2733-42.

56. Crimi M, Papadimitriou A, Galbiati S, Palamidou P, Fortunato F, et al. A new mitochondrial DNA mutation in ND3 gene causing severe Leigh syndrome with early lethality. Pediatr Res 2004;55:842-6.

57. McFarland R, Kirby DM, Fowler KJ, Ohtake A, Ryan MT, et al. De novo mutations in the mitochondrial ND3 gene as a cause of infantile mitochondrial encephalopathy and complex I deficiency. Ann Neurol 2004;55:58-64.

58. Taylor RW, Singh-Kler R, Hayes CM, Smith PE, Turnbull DM. Progressive mitochondrial disease resulting from a novel missense mutation in the mitochondrial DNA ND3 gene. Ann Neurol 2001;50:104-7.

59. Sarzi E, Brown MD, Lebon S, Chretien D, Munnich A, et al. A novel recurrent mitochondrial DNA mutation in ND3 gene is associated with isolated complex I deficiency causing Leigh syndrome and dystonia. Am J Med Genet A 2007;143a:33-41.

60. Kirby DM, McFarland R, Ohtake A, Dunning C, Ryan MT, et al. Mutations of the mitochondrial ND1 gene as a cause of MELAS. J Med Genet 2004;41:784-9.

61. Deschauer M, Bamberg C, Claus D, Zierz S, Turnbull DM, et al. Late-onset encephalopathy associated with a C11777A mutation of mitochondrial DNA. Neurology 2003;60:1357-9.

62. Komaki H, Akanuma J, Iwata H, Takahashi T, Mashima Y, et al. A novel mtDNA C11777A mutation in Leigh syndrome. Mitochondrion 2003;2:293-304.

63. Taylor RW, Morris AA, Hutchinson M, Turnbull DM. Leigh disease associated with a novel mitochondrial DNA ND5 mutation. Eur J Hum Genet 2002;10:141-4.

64. Simon DK, Friedman J, Breakefield XO, Jankovic J, Brin MF, et al. A heteroplasmic mitochondrial complex I gene mutation in adult-onset dystonia. Neurogenetics 2003;4:199-205.

65. Kirby DM, Kahler SG, Freckmann ML, Reddihough D, Thorburn DR. Leigh disease caused by the mitochondrial DNA G14459A mutation in unrelated families. Ann Neurol 2000;48:102-4.

66. Jun AS, Brown MD, Wallace DC. A mitochondrial DNA mutation at nucleotide pair 14459 of the NADH dehydrogenase subunit 6 gene associated with maternally inherited Leber hereditary optic neuropathy and dystonia. Proc Natl Acad Sci U S A 1994;91:6206-10.

67. Solano A, Roig M, Vives-Bauza C, Hernandez-Pena J, Garcia-Arumi E, et al. Bilateral striatal necrosis associated with a novel mutation in the mitochondrial ND6 gene. Ann Neurol 2003;54:527-30.

68. Ugalde C, Triepels RH, Coenen MJ, van den Heuvel LP, Smeets R, et al. Impaired complex I assembly in a Leigh syndrome patient with a novel missense mutation in the ND6 gene. Ann Neurol 2003;54:665-9.

69. Howell N, Bindoff LA, McCullough DA, Kubacka I, Poulton J, et al. Leber hereditary optic neuropathy: identification of the same mitochondrial ND1 mutation in six pedigrees. Am J Hum Genet 1991;49:939-50.

70. Huoponen K, Vilkki J, Aula P, Nikoskelainen EK, Savontaus ML. A new mtDNA mutation associated with Leber hereditary optic neuroretinopathy. Am J Hum Genet 1991;48:1147-53.

71. Wallace D, Singh G, Lott M, Hodge J, Schurr T, et al. Mitochondrial DNA mutation associated with Leber’s hereditary optic neuropathy. Science 1988;242:1427-30.

72. Brown MD, Voljavec AS, Lott MT, MacDonald I, Wallace DC. Leber’s hereditary optic neuropathy: a model for mitochondrial neurodegenerative diseases. Faseb J 1992;6:2791-9.

73. Johns DR, Neufeld MJ, Park RD. An ND-6 mitochondrial DNA mutation associated with Leber hereditary optic neuropathy. Biochem Biophys Res Commun 1992;187:1551-7.

74. Tawata M, Ohtaka M, Iwase E, Ikegishi Y, Aida K, et al. New mitochondrial DNA homoplasmic mutations associated with Japanese patients with type 2 diabetes. Diabetes 1998;47:276-7.

75. Blakely EL, de Silva R, King A, Schwarzer V, Harrower T, et al. LHON/MELAS overlap syndrome associated with a mitochondrial MTND1 gene mutation. Eur J Hum Genet 2005;13:623-7.

76. Sakuta R, Goto Y, Nonaka I, Horai S. An A-to-G transition at nucleotide pair 11084 in the ND4 gene may be an mtDNA polymorphism. Am J Hum Genet 1993;53:964-5.

77. Lertrit P, Noer AS, Jean-Francois MJ, Kapsa R, Dennett X, et al. A new disease-related mutation for mitochondrial encephalopathy lactic acidosis and strokelike episodes (MELAS) syndrome affects the ND4 subunit of the respiratory complex I. Am J Hum Genet 1992;51:457-68.

78. Liolitsa D, Rahman S, Benton S, Carr LJ, Hanna MG. Is the mitochondrial complex I ND5 gene a hot-spot for MELAS causing mutations? Ann Neurol 2003;53:128-32.

79. Crimi M, Galbiati S, Moroni I, Bordoni A, Perini MP, et al. A missense mutation in the mitochondrial ND5 gene associated with a Leigh-MELAS overlap syndrome. Neurology 2003;60:1857-61.

80. Santorelli FM, Tanji K, Kulikova R, Shanske S, Vilarinho L, et al. Identification of a novel mutation in the mtDNA ND5 gene associated with MELAS. Biochem Biophys Res Commun 1997;238:326-8.

81. Corona P, Antozzi C, Carrara F, D’Incerti L, Lamantea E, et al. A novel mtDNA mutation in the ND5 subunit of complex I in two MELAS patients. Ann Neurol 2001;49:106-10.

82. Ravn K, Wibrand F, Hansen FJ, Horn N, Rosenberg T, et al. An mtDNA mutation, 14453G-->A, in the NADH dehydrogenase subunit 6 associated with severe MELAS syndrome. Eur J Hum Genet 2001;9:805-9.

83. De Coo IF, Renier WO, Ruitenbeek W, Ter Laak HJ, Bakker M, et al. A 4-base pair deletion in the mitochondrial cytochrome b gene associated with parkinsonism/MELAS overlap syndrome. Ann Neurol 1999;45:130-3.

84. Goto Y, Nonaka I, Horai S. A mutation in the tRNA(Leu)(UUR) gene associated with the MELAS subgroup of mitochondrial encephalomyopathies. Nature 1990;348:651-3.

85. Varlamov DA, Kudin AP, Vielhaber S, Schroder R, Sassen R, et al. Metabolic consequences of a novel missense mutation of the mtDNA CO I gene. Hum Mol Genet 2002;11:1797-805.

86. Andreu AL, Hanna MG, Reichmann H, Bruno C, Penn AS, et al. Exercise intolerance due to mutations in the cytochrome b gene of mitochondrial DNA. N Engl J Med 1999;341:1037-44.

87. Comi GP, Bordoni A, Salani S, Franceschina L, Sciacco M, et al. Cytochrome c oxidase subunit I microdeletion in a patient with motor neuron disease. Ann Neurol 1998;43:110-6.

88. Clark KM, Taylor RW, Johnson MA, Chinnery PF, Chrzanowska-Lightowlers ZM, et al. An mtDNA mutation in the initiation codon of the cytochrome C oxidase subunit II gene results in lower levels of the protein and a mitochondrial encephalomyopathy. Am J Hum Genet 1999;64:1330-9.

89. Campos Y, Garcia-Redondo A, Fernandez-Moreno MA, Martinez-Pardo M, Goda G, et al. Early-onset multisystem mitochondrial disorder caused by a nonsense mutation in the mitochondrial DNA cytochrome C oxidase II gene. Ann Neurol 2001;50:409-13.

90. Wong LJ, Dai P, Tan D, Lipson M, Grix A, et al. Severe lactic acidosis caused by a novel frame-shift mutation in mitochondrial-encoded cytochrome c oxidase subunit II. Am J Med Genet 2001;102:95-9.

91. Hanna MG, Nelson IP, Rahman S, Lane RJ, Land J, et al. Cytochrome c oxidase deficiency associated with the first stop-codon point mutation in human mtDNA. Am J Hum Genet 1998;63:29-36.

92. Manfredi G, Schon EA, Moraes CT, Bonilla E, Berry GT, et al. A new mutation associated with MELAS is located in a mitochondrial DNA polypeptide-coding gene. Neuromuscul Disord 1995;5:391-8.

93. Temperley RJ, Seneca SH, Tonska K, Bartnik E, Bindoff LA, et al. Investigation of a pathogenic mtDNA microdeletion reveals a translation-dependent deadenylation decay pathway in human mitochondria. Hum Mol Genet 2003;12:2341-8.

94. Wibrand F, Ravn K, Schwartz M, Rosenberg T, Horn N, et al. Multisystem disorder associated with a missense mutation in the mitochondrial cytochrome b gene. Ann Neurol 2001;50:540-3.

95. Schuelke M, Krude H, Finckh B, Mayatepek E, Janssen A, et al. Septo-optic dysplasia associated with a new mitochondrial cytochrome b mutation. Ann Neurol 2002;51:388-92.

96. Andreu AL, Bruno C, Dunne TC, Tanji K, Shanske S, et al. A nonsense mutation (G15059A) in the cytochrome b gene in a patient with exercise intolerance and myoglobinuria. Ann Neurol 1999;45:127-30.

97. Legros F, Chatzoglou E, Frachon P, Ogier De Baulny H, Laforet P, et al. Functional characterization of novel mutations in the human cytochrome b gene. Eur J Hum Genet 2001;9:510-8.

98. Keightley JA, Anitori R, Burton MD, Quan F, Buist NR, et al. Mitochondrial encephalomyopathy and complex III deficiency associated with a stop-codon mutation in the cytochrome b gene. Am J Hum Genet 2000;67:1400-10.

99. Tarnopolsky MA, Simon DK, Roy BD, Chorneyko K, Lowther SA, et al. Attenuation of free radical production and paracrystalline inclusions by creatine supplementation in a patient with a novel cytochrome b mutation. Muscle Nerve 2004;29:537-47.

100. Dumoulin R, Sagnol I, Ferlin T, Bozon D, Stepien G, et al. A novel gly290asp mitochondrial cytochrome b mutation linked to a complex III deficiency in progressive exercise intolerance. Mol Cell Probes 1996;10:389-91.

101. Andreu AL, Bruno C, Shanske S, Shtilbans A, Hirano M, et al. Missense mutation in the mtDNA cytochrome b gene in a patient with myopathy. Neurology 1998;51:1444-7.

102. Pulkes T, Liolitsa D, Nelson IP, Hanna MG. Classical mitochondrial phenotypes without mtDNA mutations: the possible role of nuclear genes. Neurology 2003;61:1144-7.

103. Karadimas CL, Greenstein P, Sue CM, Joseph JT, Tanji K, et al. Recurrent myoglobinuria due to a nonsense mutation in the COX I gene of mitochondrial DNA. Neurology 2000;55:644-9.

104. Kollberg G, Moslemi AR, Lindberg C, Holme E, Oldfors A. Mitochondrial myopathy and rhabdomyolysis associated with a novel nonsense mutation in the gene encoding cytochrome c oxidase subunit I. J Neuropathol Exp Neurol 2005;64:123-8.

105. Rahman S, Taanman JW, Cooper JM, Nelson I, Hargreaves I, et al. A missense mutation of cytochrome oxidase subunit II causes defective assembly and myopathy. Am J Hum Genet 1999;65:1030-9.

106. McFarland R, Taylor RW, Chinnery PF, Howell N, Turnbull DM. A novel sporadic mutation in cytochrome c oxidase subunit II as a cause of rhabdomyolysis. Neuromuscul Disord 2004;14:162-6.

107. Kennaway NG, Burton MD, Hall RE, Johnston WSW, Keightley JA, et al. Mitochondrial myopathy and cytochrome c oxidase (COX) deficiency associated with a 15 bp deletion in the gene for COX subunit III. (Abstract 827)” Neurology 1994;44:A335.

108. Musumeci O, Andreu AL, Shanske S, Bresolin N, Comi GP, et al. Intragenic inversion of mtDNA: a new type of pathogenic mutation in a patient with mitochondrial myopathy. Am J hum genet 2000;66:1900-4.

109. Valnot I, Kassis J, Chretien D, de Lonlay P, Parfait B, et al. A mitochondrial cytochrome b mutation but no mutations of nuclearly encoded subunits in ubiquinol cytochrome c reductase (complex III) deficiency. Human genetics 1999;104:460-6.

110. Andreu AL, Checcarelli N, Iwata S, Shanske S, DiMauro S. A missense mutation in the mitochondrial cytochrome b gene in a revisited case with histiocytoid cardiomyopathy. Pediatr Res 2000;48:311-4.

111. Gattermann N, Retzlaff S, Wang YL, Hofhaus G, Heinisch J, et al. Heteroplasmic point mutations of mitochondrial DNA affecting subunit I of cytochrome c oxidase in two patients with acquired idiopathic sideroblastic anemia. Blood 1997;90:4961-72.

112. Pandya A, Xia XJ, Erdenetungalag R, Amendola M, Landa B, et al. Heterogenous point mutations in the mitochondrial tRNA Ser(UCN) precursor coexisting with the A1555G mutation in deaf students from Mongolia. Am J Hum Genet 1999;65:1803-6.

113. Wang Q, Li R, Zhao H, Peters JL, Liu Q, et al. Clinical and molecular characterization of a Chinese patient with auditory neuropathy associated with mitochondrial 12S rRNA T1095C mutation. Am J Med Genet A 2005;133A:27-30.

114. Shoffner JM, Brown MD, Torroni A, Lott MT, Cabell MF, et al. Mitochondrial DNA variants observed in Alzheimer disease and Parkinson disease patients. Genomics 1993;17:171-84.

115. Kosel S, Egensperger R, Mehraein P, Graeber MB. No association of mutations at nucleotide 5460 of mitochondrial NADH dehydrogenase with Alzheimer’s disease. Biochem Biophys Res Commun 1994;203:745-9.

116. Lin FH, Lin R, Wisniewski HM, Hwang YW, Grundke-Iqbal I, et al. Detection of point mutations in codon 331 of mitochondrial NADH dehydrogenase subunit 2 in Alzheimer’s brains. Biochem Biophys Res Commun 1992;182:238-46.

117. Petruzzella V, Chen X, Schon EA. Is a point mutation in the mitochondrial ND2 gene associated with Alzheimer’s disease. Biochem Biophys Res Commun 1992;186:491-7.

118. Online Mendelian Inheritance in Man O. Online Mendelian Inheritance in Man, OMIM®. McKusick-Nathans Institute of Genetic Medicine. Baltimore, MD: Johns Hopkins University; 2019.

119. Westermann B. Mitochondrial fusion and fission in cell life and death. Nat Rev Mol Cell Biol 2010;11:872-84.

120. Geto Z, Molla MD, Challa F, Belay Y, Getahun T. Mitochondrial dynamic dysfunction as a main triggering factor for inflammation associated chronic non-communicable diseases. J Inflamm Res 2020;13:97-107.

121. Niyazov DM, Kahler SG, Frye RE. Primary mitochondrial disease and secondary mitochondrial dysfunction: importance of distinction for diagnosis and treatment. Mol Syndromol 2016;7:122-37.

122. Morava E, van den Heuvel L, Hol F, de Vries MC, Hogeveen M, et al. Mitochondrial disease criteria: Diagnostic applications in children. Neurology 2006;67:1823-6.

123. Bris C, Goudenege D, Desquiret-Dumas V, Charif M, Colin E, et al. Bioinformatics Tools and Databases to Assess the Pathogenicity of Mitochondrial DNA Variants in the Field of Next Generation Sequencing. Front Genet 2018;9:632.

124. Xu J, Kitada M, Koya D. The impact of mitochondrial quality control by Sirtuins on the treatment of type 2 diabetes and diabetic kidney disease. Biochim Biophys Acta Mol Basis Dis 2020;1866:165756.

125. Li K, Wu L, Liu J, Lin W, Qi Q, et al. Maternally inherited diabetes mellitus associated with a Novel m.15897G>A mutation in mitochondrial tRNA(Thr) gene. J Diabetes Res 2020;2020:2057187.

126. Sangwung P, Petersen KF, Shulman GI, Knowles JW. Mitochondrial dysfunction, insulin resistance and potential genetic implications. Endocrinology 2020; doi: 10.1210/endocr/bqaa017.

127. Pereira CA, Carlos D, Ferreira NS, Silva JF, Zanotto CZ, et al. Mitochondrial DNA promotes NLRP3 inflammasome activation and contributes to endothelial dysfunction and inflammation in Type 1 diabetes. Front Physiol 2020;10:1557.

128. Docrat TF, Nagiah S, Naicker N, Baijnath S, Singh S, et al. The protective effect of metformin on mitochondrial dysfunction and endoplasmic reticulum stress in diabetic mice brain. Eur J Pharmacol 2020;875:173059.

129. Umbria M, Ramos A, Aluja MP, Santos C. The role of control region mitochondrial DNA mutations in cardiovascular disease: stroke and myocardial infarction. Sci Rep 2020;10:2766.

130. Hirata Y, Inoue A, Suzuki S, Takahashi M, Matsui R, et al. Trans-Fatty acids facilitate DNA damage-induced apoptosis through the mitochondrial JNK-Sab-ROS positive feedback loop. Sci Rep 2020;10:2743.

131. Aguilar-López BA, Moreno-Altamirano MMB, Dockrell HM, Duchen MR, Sánchez-García FJ. Mitochondria: an integrative Hub coordinating circadian rhythms, metabolism, the microbiome, and immunity. Front Cell Dev Biol 2020;8:51.

132. Yang S, Liu Y, Guo Y, Liu R, Qi F, et al. Circadian gene clock participates in mitochondrial apoptosis pathways by regulating mitochondrial membrane potential, mitochondria out membrane permeablization and apoptosis factors in AML12 hepatocytes. Mol Cell Biochem 2020; doi: 10.1007/s11010-020-03701-1.

133. Sardon Puig L, Valera-Alberni M, Cantó C, Pillon NJ. Circadian rhythms and mitochondria: connecting the dots. Front genet 2018;9:452.

134. Rahman A, Hasan AU, Nishiyama A, Kobori H. Altered circadian timing system-mediated non-dipping pattern of blood pressure and associated cardiovascular disorders in metabolic and kidney diseases. Int J Mol Sci 2018;19:400.

135. Gombert M, Carrasco-Luna J, Pin-Arboledas G, Codoñer-Franch P. The connection of circadian rhythm to inflammatory bowel disease. Trans Res 2019;206:107-18.

136. Maiese K. Moving to the rhythm with clock (Circadian) genes, autophagy, mTOR, and SIRT1 in degenerative disease and cancer. Curr Neurovasc Res 2017;14:299-304.

137. Leng Y, Musiek ES, Hu K, Cappuccio FP, Yaffe K. Association between circadian rhythms and neurodegenerative diseases. Lancet Neurol 2019;18:307-18.

138. Takaguri A, Sasano J, Akihiro O, Satoh K. The role of circadian clock gene BMAL1 in vascular proliferation. Eur J Pharmacol 2020;872:172924.

139. Škrlec I, Milić J, Steiner R. The impact of the circadian genes CLOCK and ARNTL on myocardial infarction. J Clin Med 2020;9:484.

140. Zhang ZQ, Ding JW, Wang XA, Luo CY, Yu B, et al. Abnormal circadian rhythms are associated with plaque instability in acute coronary syndrome patients. Int J Clin Exp Pathol 2019;12:3761-71.

141. Wang XB, Cui NH, Liu Xn, Liu X. Mitochondrial 8-hydroxy-2’-deoxyguanosine and coronary artery disease in patients with type 2 diabetes mellitus. Cardiovasc Diabetol 2020;19:22.

142. Ward PS, Thompson CB. Metabolic reprogramming: a cancer hallmark even warburg did not anticipate. Cancer Cell 2012;21:297-308.

143. Ganapathy-Kanniappan S. Molecular intricacies of aerobic glycolysis in cancer: current insights into the classic metabolic phenotype. Crit Rev Biochem Mol Biol 2018;53:667-82.

144. Feng Z, Hu W, de Stanchina E, Teresky AK, Jin S, et al. The regulation of AMPK β1, TSC2, and PTEN Expression by p53: stress, cell and tissue specificity, and the role of these gene products in modulating the IGF-1-AKT-mTOR pathways. Cancer Res 2007;67:3043-53.

145. Levine AJ, Puzio-Kuter AM. The control of the metabolic switch in cancers by oncogenes and tumor suppressor genes. Science 2010;330:1340-4.

146. Elstrom RL, Bauer DE, Buzzai M, Karnauskas R, Harris MH, et al. Akt stimulates aerobic glycolysis in cancer cells. Cancer Res 2004;64:3892-9.

147. Gray LR, Tompkins SC, Taylor EB. Regulation of pyruvate metabolism and human disease. Cell Mol Life Sci 2014;71:2577-604.

148. Bauer DE, Hatzivassiliou G, Zhao F, Andreadis C, Thompson CB. ATP citrate lyase is an important component of cell growth and transformation. Oncogene 2005;24:6314-22.

149. Hatzivassiliou G, Zhao F, Bauer DE, Andreadis C, Shaw AN, et al. ATP citrate lyase inhibition can suppress tumor cell growth. Cancer Cell 2005;8:311-21.

150. Land SC, Tee AR. Hypoxia-inducible factor 1α is regulated by the mammalian target of rapamycin (mTOR) via an mTOR signaling motif. J Biolog Chem 2007;282:20534-43.

151. Sakamoto T, Weng JS, Hara T, Yoshino S, Kozuka-Hata H, et al. Hypoxia-inducible factor 1 regulation through cross talk between mTOR and MT1-MMP. Mol Cell Biol 2014;34:30-42.

152. Kietzmann T, Mennerich D, Dimova EY. Hypoxia-inducible factors (HIFs) and Phosphorylation: impact on stability, localization, and transactivity. Front Cell Dev Biol 2016;4.

153. Papandreou I, Cairns RA, Fontana L, Lim AL, Denko NC. HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption. Cell Metab 2006;3:187-97.

154. Kim JW, Tchernyshyov I, Semenza GL, Dang CV. HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia. Cell Metab 2006;3:177-85.

155. Le A, Lane AN, Hamaker M, Bose S, Gouw A, et al. Glucose-independent glutamine metabolism via TCA cycling for proliferation and survival in B cells. Cell Metab 2012;15:110-21.

156. Wang JB, Erickson JW, Fuji R, Ramachandran S, Gao P, et al. Targeting mitochondrial glutaminase activity inhibits oncogenic transformation. Cancer cell 2010;18:207-19.

157. Itahana Y, Itahana K. Emerging roles of p53 family members in glucose metabolism. Int J Mol Sci 2018;19:776.

158. Schwartzenberg-Bar-Yoseph F, Armoni M, Karnieli E. The tumor suppressor p53 down-regulates glucose transporters GLUT1 and GLUT4 gene expression. Cancer Res 2004;64:2627-33.

159. Boidot R, Végran F, Meulle A, Le Breton A, Dessy C, et al. Regulation of monocarboxylate transporter MCT1 expression by p53 mediates inward and outward lactate fluxes in tumors. Cancer Res 2012;72:939-48.

160. Bensaad K, Tsuruta A, Selak MA, Vidal MNC, Nakano K, et al. TIGAR, a p53-inducible regulator of glycolysis and apoptosis. Cell 2006;126:107-20.

161. Kondoh H, Lleonart ME, Gil J, Wang J, Degan P, et al. Glycolytic enzymes can modulate cellular life Span. Cancer Res 2005;65:177-85.

162. Zhou S, Kachhap S, Singh KK. Mitochondrial impairment in p53-deficient human cancer cells. Mutagenesis 2003;18:287-92.

163. Lebedeva MA, Eaton JS, Shadel GS. Loss of p53 causes mitochondrial DNA depletion and altered mitochondrial reactive oxygen species homeostasis. Biochim Biophys Acta 2009;1787:328-34.

164. Kulawiec M, Ayyasamy V, Singh KK. p53 regulates mtDNA copy number and mitocheckpoint pathway. J carcinog 2009;8:8.

165. D’Souza AD, Parikh N, Kaech SM, Shadel GS. Convergence of multiple signaling pathways is required to coordinately up-regulate mtDNA and mitochondrial biogenesis during T cell activation. Mitochondrion 2007;7:374-85.

166. Di Donato S. Disorders related to mitochondrial membranes: pathology of the respiratory chain and neurodegeneration. J Inherit Metab Dis 2000;23:247-63.

167. Polster BM, Fiskum G. Mitochondrial mechanisms of neural cell apoptosis. J Neurochem 2004;90:1281-9.

168. Rehm M, Düßmann H, Jänicke RU, Tavaré JM, Kögel D, et al. Single-cell fluorescence resonance energy transfer analysis demonstrates that caspase activation during apoptosis is a rapid process: role of caspase-3. J Biol Chem 2002;277:24506-14.

169. Verhagen AM, Ekert PG, Pakusch M, Silke J, Connolly LM, et al. Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins. Cell 2000;102:43-53.

170. Liu X, Kim CN, Yang J, Jemmerson R, Wang X. Induction of apoptotic program in cell-free extracts: requirement for dATP and cytochrome c. Cell 1996;86:147-57.

171. Susin SA, Lorenzo HK, Zamzami N, Marzo I, Snow BE, et al. Molecular characterization of mitochondrial apoptosis-inducing factor. Nature 1999;397:441-6.

172. Du C, Fang M, Li Y, Li L, Wang X. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 2000;102:33-42.

173. Zou H, Li Y, Liu X, Wang X. An APAF-1·Cytochrome c multimeric complex is a functional apoptosome that activates procaspase-9. J Biolog Chem 1999;274:11549-56.

174. Trushina E, McMurray CT. Oxidative stress and mitochondrial dysfunction in neurodegenerative diseases. Neuroscience 2007;145:1233-48.

175. Saccon RA, Bunton-Stasyshyn RKA, Fisher EMC, Fratta P. Is SOD1 loss of function involved in amyotrophic lateral sclerosis? Brain: a journal of neurology 2013;136:2342-58.

176. Grünewald A, Kumar KR, Sue CM. New insights into the complex role of mitochondria in Parkinson’s disease. Prog Neurobiol 2019;177:73-93.

177. Antony PMA, Kondratyeva O, Mommaerts K, Ostaszewski M, Sokolowska K, et al. Fibroblast mitochondria in idiopathic Parkinson’s disease display morphological changes and enhanced resistance to depolarization. Sci Rep 2020;10:1569.

178. Sironi L, Restelli LM, Tolnay M, Neutzner A, Frank S. Dysregulated Interorganellar Crosstalk Of Mitochondria In The Pathogenesis of Parkinson’s disease. Cells 2020;9:233.

179. Suomalainen A, Battersby BJ. Mitochondrial diseases: the contribution of organelle stress responses to pathology. Nat Rev Mol Cell Biol 2018;19:77-92.

180. Rahman J, Rahman S. Mitochondrial medicine in the omics era. Lancet 2018;391:2560-74.

181. Holt IJ, Harding AE, Morgan-Hughes JA. Deletions of muscle mitochondrial DNA in patients with mitochondrial myopathies. Nature 1988;331:717-9.

182. Ingman M, Gyllensten U. mtDB: Human mitochondrial genome database, a resource for population genetics and medical sciences. Nucleic Acids Res 2006;34:D749-51.

183. Bourgeron T, Rustin P, Chretien D, Birch-Machin M, Bourgeois M, et al. Mutation of a nuclear succinate dehydrogenase gene results in mitochondrial respiratory chain deficiency. Nat Genet 1995;11:144-9.

184. Ye F, Samuels DC, Clark T, Guo Y. High-throughput sequencing in mitochondrial DNA research. Mitochondrion 2014;17:157-63.

185. Liang MH, Johnson DR, Wong LJC. Preparation and validation of PCR-generated positive controls for diagnostic dot blotting. Clin Chem 1998;44:1578-9.

186. Bai RK, Wong LJC. Detection and quantification of heteroplasmic mutant mitochondrial DNA by real-time amplification refractory mutation system quantitative PCR analysis: a single-step approach. Clin Chem 2004;50:996-1001.

187. White HE, Durston VJ, Seller A, Fratter C, Harvey JF, et al. Accurate detection and quantitation of heteroplasmic mitochondrial point mutations by pyrosequencing. Genet Test 2005;9:190-9.

188. Pronicka E, Piekutowska-Abramczuk D, Ciara E, Trubicka J, Rokicki D, et al. New perspective in diagnostics of mitochondrial disorders: two years’ experience with whole-exome sequencing at a national paediatric centre. J Transl Med 2016;14:174.

189. Kohda M, Tokuzawa Y, Kishita Y, Nyuzuki H, Moriyama Y, et al. A comprehensive genomic analysis reveals the genetic landscape of mitochondrial respiratory chain complex deficiencies. PLoS Genet 2016;12:e1005679.

190. Calvo SE, Compton AG, Hershman SG, Lim SC, Lieber DS, et al. Molecular diagnosis of infantile mitochondrial disease with targeted next-generation sequencing. Sci Transl Med 2012;4:118ra10.

191. Gould MP, Bosworth CM, McMahon S, Grandhi S, Grimberg BT, et al. PCR-free enrichment of mitochondrial DNA from human blood and cell lines for high quality next-generation DNA sequencing. PLoS One 2015;10:e0139253.

192. Vasta V, Ng SB, Turner EH, Shendure J, Hahn SH. Next generation sequence analysis for mitochondrial disorders. Genome Med 2009;1:100.

193. Sosa MX, Sivakumar IKA, Maragh S, Veeramachaneni V, Hariharan R, et al. Next-generation sequencing of human mitochondrial reference genomes uncovers high heteroplasmy frequency. PLoS Comput Biol 2012;8:e1002737.

194. Dayama G, Emery SB, Kidd JM, Mills RE. The genomic landscape of polymorphic human nuclear mitochondrial insertions. Nucleic Acids Res 2014;42:12640-9.

195. Picardi E, Pesole G. Mitochondrial genomes gleaned from human whole-exome sequencing. Nat Methods 2012;9:523-4.

196. Guo Y, Li J, Li CI, Shyr Y, Samuels DC. MitoSeek: extracting mitochondria information and performing high-throughput mitochondria sequencing analysis. Bioinformatics 2013;29:1210-1.

197. Falk MJ, Shen L, Gonzalez M, Leipzig J, Lott MT, et al. Mitochondrial disease sequence data resource (MSeqDR): a global grass-roots consortium to facilitate deposition, curation, annotation, and integrated analysis of genomic data for the mitochondrial disease clinical and research communities. Mol Genet Metabol 2015;114:388-96.

198. Kogelnik AM, Lott MT, Brown MD, Navathe SB, Wallace DC. MITOMAP: a human mitochondrial genome database. Nucleic acids Res 1996;24:177-9.

199. Preste R, Vitale O, Clima R, Gasparre G, Attimonelli M. HmtVar: a new resource for human mitochondrial variations and pathogenicity data. Nucleic Acids Res 2019;47:D1202-10.

200. Clima R, Preste R, Calabrese C, Diroma MA, Santorsola M, et al. HmtDB 2016: data update, a better performing query system and human mitochondrial DNA haplogroup predictor. Nucleic Acids Res 2017;45:D698-706.

201. Rahman J, Noronha A, Thiele I, Rahman S. Leigh map: a novel computational diagnostic resource for mitochondrial disease. Ann Neurol 2017;81:9-16.

202. Abicht A, Scharf F, Kleinle S, Schön U, Holinski-Feder E, et al. Mitochondrial and nuclear disease panel (Mito-aND-Panel): combined sequencing of mitochondrial and nuclear DNA by a cost-effective and sensitive NGS-based method. Mol Genet Genomic Med 2018;6:1188-98.

203. Dames S, Chou LS, Xiao Y, Wayman T, Stocks J, et al. The development of next-generation sequencing assays for the mitochondrial genome and 108 nuclear genes associated with mitochondrial disorders. J Mol Diagnos 2013;15:526-34.

204. Pereira V, Longobardi A, Borsting C. Sequencing of mitochondrial genomes using the precision ID mtDNA whole genome panel. Electrophoresis 2018;39:2766-75.

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

206. Kremer LS, Bader DM, Mertes C, Kopajtich R, Pichler G, et al. Genetic diagnosis of Mendelian disorders via RNA sequencing. Nat Commun 2017;8:15824.

207. Cummings BB, Marshall JL, Tukiainen T, Lek M, Donkervoort S, et al. Improving genetic diagnosis in Mendelian disease with transcriptome sequencing. Sci Transl Med 2017;9:eaal5209.

208. Gómez-Serrano M, Camafeita E, Loureiro M, Peral B. Mitoproteomics: tackling mitochondrial dysfunction in human disease. Oxid Med Cell Longev 2018;2018:1435934.

209. Calvo SE, Clauser KR, Mootha VK. MitoCarta2.0: an updated inventory of mammalian mitochondrial proteins. Nucleic Acids Res 2016;44:D1251-7.

210. Scharfe C, Zaccaria P, Hoertnagel K, Jaksch M, Klopstock T, et al. MITOP: database for mitochondria-related proteins, genes and diseases. Nucleic Acids Res 1999;27:153-5.

211. Cotter D, Guda P, Fahy E, Subramaniam S. MitoProteome: mitochondrial protein sequence database and annotation system. Nucleic Acids Res 2004;32:D463-7.

212. Elstner M, Andreoli C, Ahting U, Tetko I, Klopstock T, et al. MitoP2: an integrative tool for the analysis of the mitochondrial proteome. Mol Biotechnol 2008;40:306-15.

213. Smith AC, Robinson AJ. MitoMiner, an integrated database for the storage and analysis of mitochondrial proteomics data. Mol Cell Proteomics 2009;8:1324-37.

214. Floyd BJ, Wilkerson EM, Veling MT, Minogue CE, Xia C, et al. Mitochondrial protein interaction mapping identifies regulators of respiratory chain function. Mol Cell 2016;63:621-32.

215. Lake NJ, Webb BD, Stroud DA, Richman TR, Ruzzenente B, et al. Biallelic mutations in MRPS34 lead to instability of the small mitoribosomal subunit and leigh syndrome. Am J Hum Genet 2017;101:239-54.

216. Debray FG, Mitchell GA, Allard P, Robinson BH, Hanley JA, et al. Diagnostic accuracy of blood lactate-to-pyruvate molar ratio in the differential diagnosis of congenital lactic acidosis. Clin Chem 2007;53:916-21.

217. Finsterer J, Eichberger H, Jarius C, Boltzmann L. Lactate-stress testing in 54 patients with mitochondriopathy. Eur Arch Psychiatry Clin Neurosci 2000;250:36-9.

218. Finsterer J, Milvay E. Stress lactate in mitochondrial myopathy under constant, unadjusted workload. Eur J Neurol 2004;11:811-6.

219. Thompson Legault J, Strittmatter L, Tardif J, Sharma R, Tremblay-Vaillancourt V, et al. A metabolic signature of mitochondrial dysfunction revealed through a monogenic form of leigh syndrome. Cell Rep 2015;13:981-9.

220. Rodenburg RJ. The functional genomics laboratory: functional validation of genetic variants. J Inherit Metab Dis 2018;41:297-307.

221. Gasperskaja E, Kučinskas V. The most common technologies and tools for functional genome analysis. Acta Med Litu 2017;24:1-11.

222. Thompson K, Collier JJ, Glasgow RIC, Robertson FM, Pyle A, et al. Recent advances in understanding the molecular genetic basis of mitochondrial disease. J Inherit Metab Dis 2020;43:36-50.

223. Consortium GT. Human genomics. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science 2015;348:648-60.

224. Gropman AL. Neuroimaging in mitochondrial disorders. Neurotherapeutics 2013;10:273-85.

225. Lunsing RJ, Strating K, de Koning TJ, Sijens PE. Diagnostic value of MRS-quantified brain tissue lactate level in identifying children with mitochondrial disorders. Eur Radiol 2017;27:976-84.

226. Chi CS, Lee HF, Tsai CR, Chen WS, Tung JN, et al. Lactate peak on brain MRS in children with syndromic mitochondrial diseases. J Chin Med Assoc 2011;74:305-9.

227. Dinopoulos A, Cecil KM, Schapiro MB, Papadimitriou A, Hadjigeorgiou GM, et al. Brain MRI and proton MRS findings in infants and children with respiratory chain defects. Neuropediatrics 2005;36:290-301.

228. Matthews PM, Allaire C, Shoubridge EA, Karpati G, Carpenter S, et al. In vivo muscle magnetic resonance spectroscopy in the clinical investigation of mitochondria1 disease. Neurology 1991;41:114.

229. Saito S, Takahashi Y, Ohki A, Shintani Y, Higuchi T. Early detection of elevated lactate levels in a mitochondrial disease model using chemical exchange saturation transfer (CEST) and magnetic resonance spectroscopy (MRS) at 7T-MRI. Radiol Phys Technol 2019;12:46-54.

230. Farina L, Chiapparini L, Uziel G, Bugiani M, Zeviani M, et al. MR findings in Leigh syndrome with COX deficiency and SURF-1 mutations. Am J neuroradiol 2002;23:1095-100.

231. Bluml S, Seymour KJ, Ross BD. Developmental changes in choline- and ethanolamine-containing compounds measured with proton-decoupled (31)P MRS in in vivo human brain. Magn Reson Med 1999;42:643-54.

232. Pouwels PJ, Brockmann K, Kruse B, Wilken B, Wick M, et al. Regional age dependence of human brain metabolites from infancy to adulthood as detected by quantitative localized proton MRS. Pediatric Res 1999;46:474-85.

233. Thompson K, Majd H, Dallabona C, Reinson K, King MS, et al. Recurrent de novo dominant mutations in SLC25A4 cause severe early-onset mitochondrial disease and loss of mitochondrial DNA copy number. Am J Hum Genet 2016;99:860-76.

234. Deciphering Developmental Disorders Study. Large-scale discovery of novel genetic causes of developmental disorders. Nature 2015;519:223-8.

235. Griffin BH, Chitty LS, Bitner-Glindzicz M. The 100 000 genomes project: what it means for paediatrics. Arch Dis Child Educ Pract Ed 2017;102:105-7.

236. Stewart JB, Chinnery PF. The dynamics of mitochondrial DNA heteroplasmy: implications for human health and disease. Nat Rev Genet 2015;16:530-42.

237. Perales-Clemente E, Cook AN, Evans JM, Roellinger S, Secreto F, et al. Natural underlying mtDNA heteroplasmy as a potential source of intra-person hiPSC variability. EMBO J 2016;35:1979-90.

238. Hall AM, Vilasi A, Garcia-Perez I, Lapsley M, Alston CL, et al. The urinary proteome and metabonome differ from normal in adults with mitochondrial disease. Kidney Int 2015;87:610-22.

239. Wettmarshausen J, Perocchi F. Isolation of functional mitochondria from cultured cells and mouse tissues. Mitochondria: Practical Protocols. New York: Springer New York; 2017. pp. 15-32.

240. Frezza C, Cipolat S, Scorrano L. Organelle isolation: functional mitochondria from mouse liver, muscle and cultured filroblasts. Nat Protoc 2007;2:287-95.

241. Tang B, Zhao L, Liang R, Zhang Y, Wang L. Magnetic nanoparticles: an improved method for mitochondrial isolation. Mol Med Rep 2012;5:1271-6.

242. Pagliarini DJ, Calvo SE, Chang B, Sheth SA, Vafai SB, et al. A mitochondrial protein compendium elucidates complex I disease biology. Cell 2008;134:112-23.

243. Mootha VK, Bunkenborg J, Olsen JV, Hjerrild M, Wisniewski JR, et al. Integrated analysis of protein composition, tissue diversity, and gene regulation in mouse mitochondria. Cell 2003;115:629-40.

244. Foster LJ, de Hoog CL, Zhang Y, Zhang Y, Xie X, et al. A mammalian organelle map by protein correlation profiling. Cell 2006;125:187-99.

245. Costain G, Jobling R, Walker S, Reuter MS, Snell M, et al. Periodic reanalysis of whole-genome sequencing data enhances the diagnostic advantage over standard clinical genetic testing. Eur J Hum Genet 2018;26:740-4.

246. Yang Y, Muzny DM, Reid JG, Bainbridge MN, Willis A, et al. Clinical whole-exome sequencing for the diagnosis of mendelian disorders. N Engl J Med 2013;369:1502-11.

247. Sun Y, Xiang J, Liu Y, Chen S, Yu J, et al. Increased diagnostic yield by reanalysis of data from a hearing loss gene panel. BMC Med Genom 2019;12:76.

248. Salfati EL, Spencer EG, Topol SE, Muse ED, Rueda M, et al. Re-analysis of whole-exome sequencing data uncovers novel diagnostic variants and improves molecular diagnostic yields for sudden death and idiopathic diseases. Genome Med 2019;11:83.

249. Rehm HL, Berg JS, Brooks LD, Bustamante CD, Evans JP, et al. ClinGen--the clinical genome resource. N Engl J Med 2015;372:2235-42.

250. Landrum MJ, Chitipiralla S, Brown GR, Chen C, Gu B, et al. ClinVar: improvements to accessing data. Nucleic Acids Res 2019;48:D835-44.

Journal of Translational Genetics and Genomics
ISSN 2578-5281 (Online)
Follow Us

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/