M. W. Gray, G. Burger, and B. F. Lang, Mitochondrial evolution, Science, vol.283, pp.1476-1481, 1999.

L. Ernster and G. Schatz, Mitochondria: A historical review, J Cell Biol, vol.91, pp.227-255, 1981.

C. M. Gustafsson, M. Falkenberg, and N. G. Larsson, Maintenance and expression of mammalian mitochondrial DNA, Annu Rev Biochem, vol.85, pp.133-160, 2016.

W. C. Copeland and M. J. Longley, Mitochondrial genome maintenance in health and disease, DNA repair, vol.19, pp.190-198, 2014.

M. I. Ekstrand, M. Falkenberg, A. Rantanen, C. B. Park, M. Gaspari et al., Mitochondrial transcription factor A regulates mtDNA copy number in mammals, Hum Mol Genet, vol.13, issue.9, pp.935-944, 2004.

C. Kukat, K. M. Davies, C. A. Wurm, H. Spahr, N. A. Bonekamp et al., Cross-strand binding of TFAM to a single mtDNA molecule forms the mitochondrial nucleoid, Proc Natl Acad Sci, vol.112, issue.36, pp.11288-11293, 2015.

J. Marín-garcia, Mitochondrial DNA repair: a novel therapeutic target for heart failure, Heart Fail Rev, vol.21, issue.5, p.26940911, 2016.

S. D. Cline, Mitochondrial DNA damage and its consequences for mitochondrial gene expression, BBA-Gene Regul Mech, vol.1819, issue.9, pp.979-991, 2012.

E. Silva, Espé cies reativas de oxigênio e nitrogênio: produção e efeitos sobre a integridade estrutural e funcional dos espermatozoides. Ciência Veteriná ria, vol.13, pp.9-16, 2010.

K. Chandrasekaran, M. Anjaneyulu, J. Choi, P. Kumar, M. Salimian et al., Role of mitochondria in diabetic peripheral neuropathy: Influencing the NAD+-dependent SIRT1-PGC-1?-TFAM pathway, Int Rev Neurobiol, vol.145, pp.177-209, 2019.

A. Johri, A. Chandra, ;. Beal, and M. , PGC-1?, mitochondrial dysfunction, and Huntington's disease, Free Radic Biol Med, vol.62, pp.37-46, 2013.

J. E. Selfridge, E. L. Lu, J. Swerdlow, and R. H. , Role of mitochondrial homeostasis and dynamics in Alzheimer's disease, Neurobiol Dis, vol.51, pp.3-12, 2013.

M. Uittenbogaard and A. Chiaramello, Mitochondrial biogenesis: A therapeutic target for neurodevelopmental disorders and neurodegenerative diseases, Curr Pharm Des, vol.20, issue.35, pp.5574-5593, 2014.

I. Kang, C. T. Chu, and B. A. Kaufman, The mitochondrial transcription factor TFAM in neurodegeneration: Emerging evidence and mechanisms, FEBS Lett, vol.592, issue.5, pp.793-811, 2018.

D. C. Rubinsztein, The roles of intracellular protein-degradation pathways in neurodegeneration, Nature, vol.443, pp.780-786, 2006.

J. A. Doudna and E. Charpentier, The new frontier of genome engineering with CRISPR-Cas9, Science, vol.346, issue.6213, p.1258096, 2014.

A. Pickar-oliver and C. A. Gersbach, The next generation of CRISPR-Cas technologies and applications, Nat Rev Mol Cell Bio, vol.20, issue.8, p.31147612, 2019.

V. C. Oliveira, G. Moreira, F. F. Bressan, M. Junior, C. G. Roballo et al., Edition of TFAM gene by CRISPR/Cas9 technology in bovine model, PLoS One, vol.14, issue.3, pp.1-14, 2019.
URL : https://hal.archives-ouvertes.fr/mnhn-02866456

M. F. Raoufi, P. Tajik, M. M. Dehghan, F. Eini, and A. Barin, Isolation and differentiation of mesenchymal stem cells from bovine, Reprod Domest Anim. 2011, vol.46, issue.1, p.20345587

J. A. Nicklas, E. M. Brooks, T. C. Hunter, R. Single, and R. F. Branda, Development of a quantitative PCR (Taq-Man) assay for relative mitochondrial DNA copy number and the common mitochondrial DNA deletion in the rat, Environ Mol Mutagen, vol.44, issue.4, pp.313-320, 2004.

K. Roballo and J. Bushman, Evaluation of the host immune response and functional recovery in peripheral nerve autografts and allografts, Transpl. Immunol, vol.53, pp.61-71, 2019.

R. A. Mccloy, S. Rogers, C. E. Caldon, T. Lorca, A. Castro et al., Partial inhibition of Cdk1 in G 2 phase overrides the SAC and decouples mitotic events, Cell Cycle, vol.13, pp.1400-1412, 2014.

C. G. Elsik, R. L. Tellam, and K. C. Worley, The genome sequence of Taurine cattle: A window to ruminant biology and evolution, Science, vol.324, pp.522-527, 2009.
URL : https://hal.archives-ouvertes.fr/hal-02667039

N. G. Larsson, J. Wang, H. Wilhelmsson, A. Oldfors, P. Rustin et al., Mitochondrial transcription factor A is necessary for mtDNA maintenance and embryogenesis in mice, Nat. Genet, vol.18, issue.3, pp.231-267, 1998.

D. Kang and N. Hamasaki, Mitochondrial Transcription Factor A in the Maintenance of Mitochondrial DNA Overview of Its Multiple Roles Annals of the, New York Academy of Sciences. Ann Ny Acad Sci, vol.1042, pp.101-108, 2005.

M. R. Chiaratti and F. V. Meirelles, Increase in mitochondrial DNA quantity and impairment of oxidative phosphorylation in bovine fibroblast cells treated with ethidium bromide for 15 passages in culture, Genet. Mol. Res, vol.5, issue.1, pp.55-62, 2006.

C. T. Moraes, What regulates mitochondrial DNA copy number in animal cells?, Trends Genet, vol.17, pp.199-205, 2001.

L. Pessoa, F. F. Bressan, M. R. Chiaratti, P. Pires, D. Perecin et al., Mitochondrial DNA dynamics during in vitro culture and pluripotency induction of a bovine Rho0 cell line, Genet. Mol. Res, vol.14, issue.4, pp.14093-14104, 2015.

B. L. Seidel-rogol and G. S. Shadel, Modulation of mitochondrial transcription in response to mtDNA depletion and repletion in HeLa cells, Nucleic Acids Res, vol.30, pp.1929-1934, 1929.

R. J. Shmookler-reis and S. Goldstein, Mitochondrial DNA in mortal and immortal human cells, J Biol Chem, vol.258, p.90789085, 1983.

A. A. Moskalev, M. V. Shaposhnikov, E. N. Plyusnina, A. Zhavoronkov, A. Budovsky et al., The role of DNA damage and repair in aging through the prism of Koch-like criteria, Ageing Res Rev, vol.12, issue.2, pp.661-684, 2013.

N. Sondheimer, C. E. Glatz, J. E. Tirone, M. A. Deardorff, A. M. Krieger et al., Neutral mitochondrial heteroplasmy and the influence of aging, Hum Mol Genet, vol.20, issue.8, pp.1653-1662, 2011.

A. R. Stiles, M. T. Simon, A. Stover, S. Eftekharian, N. Khanlou et al., Mutations in TFAM, encoding mitochondrial transcription factor A, cause neonatal liver failure associated with mtDNA depletion, Mol Genetic Metab, vol.11, pp.91-100, 2016.

A. L. Guyatt, R. R. Brennan, K. Burrows, P. Guthrie, R. Ascione et al., A genome-wide association study of mitochondrial DNA copy number in two population-based cohorts, Human genomics, vol.13, issue.1, p.30704525, 2019.

M. Ikeda, T. Ide, T. Fujino, S. Arai, K. Saku et al., Overexpression of TFAM or twinkle increases mtDNA copy number and facilitates cardioprotection associated with limited mitochondrial oxidative stress, PLos One, vol.10, issue.3, p.119687, 2015.

E. J. Tucker, R. Rius, and S. Jaillard, Genomic sequencing highlights the diverse molecular causes of Perrault syndrome: a peroxisomal disorder (PEX6), metabolic disorders (CLPP, GGPS1), and mtDNA maintenance/translation disorders (LARS2, TFAM) Hum, p.32399598

R. J. Deberardinis and C. B. Thompson, Cellular metabolism and disease: what do metabolic outliers teach us? Cell, pp.1132-1144, 2012148.

A. Jo, S. Ham, G. H. Lee, Y. I. Lee, S. Kim et al., Efficient mitochondrial genome editing by CRISPR/ Cas9, Bio Med Res international, p.10, 2015.

J. Zou, F. Yue, X. Jiang, W. Li, J. Yi et al., Mitochondrion-associated protein LRPPRC suppresses the initiation of basal levels of autophagy via enhancing Bcl-2 stability, Biochemical, vol.454, issue.3, pp.447-457, 2013.