Page 94 - Read Online
P. 94

Page 180                                            Sulaiman et al. J Transl Genet Genom 2020;4:159-87  I  https://doi.org/10.20517/jtgg.2020.27

                   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
   89   90   91   92   93   94   95   96   97   98   99