J. Nakhle, A. Rodriguez, and M. Vignais, Multifaceted Roles of Mitochondrial Components and Metabolites in Metabolic Diseases and Cancer, International Journal of Molecular Sciences, vol.21, issue.12, p.4405, 2020.
URL : https://hal.archives-ouvertes.fr/inserm-02989029

P. A. Gammage and C. Frezza, Mitochondrial DNA: the overlooked oncogenome?, BMC Biology, vol.17, issue.1, p.53, 2019.

O. M. Russell, G. S. Gorman, R. N. Lightowlers, and D. M. Turnbull, Mitochondrial Diseases: Hope for the Future, Cell, vol.181, issue.1, pp.168-188, 2020.

C. A. Castellani, R. J. Longchamps, J. Sun, E. Guallar, and D. E. Arking, Thinking outside the nucleus: Mitochondrial DNA copy number in health and disease, Mitochondrion, vol.53, pp.214-223, 2020.

J. Dolcini, H. Wu, J. C. Nwanaji-enwerem, M. Kiomourtozlogu, A. Cayir et al., Mitochondria and aging in older individuals: an analysis of DNA methylation age metrics, leukocyte telomere length, and mitochondrial DNA copy number in the VA normative aging study, Aging, vol.12, issue.3, pp.2070-2083, 2020.

M. Frederick, H. D. Skinner, S. A. Kazi, A. G. Sikora, and V. C. Sandulache, High expression of oxidative phosphorylation genes predicts improved survival in squamous cell carcinomas of the head and neck and lung, Scientific Reports, vol.10, issue.1, p.6380, 2020.

P. Shukla, S. Mukherjee, and A. Patil, Identification of Variants in Mitochondrial D-Loop and OriL Region and Analysis of Mitochondrial DNA Copy Number in Women with Polycystic Ovary Syndrome, DNA and Cell Biology, vol.39, issue.8, pp.1458-1466, 2020.

. Sobenin, . Zhelankin, . Khasanova, . Sinyov, . Medvedeva et al., Heteroplasmic Variants of Mitochondrial DNA in Atherosclerotic Lesions of Human Aortic Intima, Biomolecules, vol.9, issue.9, p.455, 2019.

J. Xu, W. Chang, C. Tsai, D. Bau, J. W. Davis et al., Mitochondrial DNA copy number in peripheral blood leukocytes is associated with biochemical recurrence in prostate cancer patients in African Americans, Carcinogenesis, vol.41, issue.3, pp.267-273, 2019.

J. Ariazi, A. Benowitz, V. De-biasi, M. L. Den-boer, S. Cherqui et al., Tunneling Nanotubes and Gap Junctions?Their Role in Long-Range Intercellular Communication during Development, Health, and Disease Conditions, Frontiers in Molecular Neuroscience, vol.10, p.333, 2017.

A. Rodriguez, J. Nakhle, E. Griessinger, and M. Vignais, Intercellular mitochondria trafficking highlighting the dual role of mesenchymal stem cells as both sensors and rescuers of tissue injury, Cell Cycle, vol.17, issue.6, pp.712-721, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-01855876

Y. Hekmatshoar, J. Nakhle, M. Galloni, and M. Vignais, The role of metabolism and tunneling nanotube-mediated intercellular mitochondria exchange in cancer drug resistance, Biochemical Journal, vol.475, issue.14, pp.2305-2328, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-01855869

G. Pinto, C. Brou, and C. Zurzolo, Tunneling Nanotubes: The Fuel of Tumor Progression?, Trends in Cancer, vol.6, issue.10, pp.874-888, 2020.
URL : https://hal.archives-ouvertes.fr/pasteur-02941638

F. Merante, S. Raha, J. K. Reed, and G. Proteau, The simultaneous isolation of RNA and DNA from tissues and cultured cells, Protocols for Gene Analysis. Harwood AJ, pp.113-120, 1994.

W. Kong, Y. Wang, Q. Wang, Y. Han, and Y. Hu, Comparison of three methods for isolation of nucleic acids from membranate inner ear tissue of rats, Chinese Medical Journal, vol.119, issue.12, pp.986-990, 2006.

T. Alabi, S. B. Patel, S. Bhatia, J. A. Wolfson, and P. Singh, Isolation of DNA-free RNA from human bone marrow mononuclear cells: comparison of laboratory methods, BioTechniques, vol.68, issue.3, pp.159-162, 2020.

W. Quispe-tintaya, R. R. White, V. N. Popov, J. Vijg, and A. Y. Maslov, Fast mitochondrial DNA isolation from mammalian cells for next-generation sequencing, BioTechniques, vol.55, issue.3, pp.133-136, 2013.

M. Devall, J. Burrage, and R. Caswell, A comparison of mitochondrial DNA isolation methods in frozen post-mortem human brain tissue -applications for studies of mitochondrial genetics in brain disorders, BioTechniques, vol.59, issue.4, pp.241-246, 2015.

D. J. Clark, C. M. Moore, and M. Flanagan, An efficient and novel technology for the extraction of parasite genomic DNA from whole blood or culture, BioTechniques, vol.68, issue.2, pp.79-84, 2019.

P. Chomczynski, A reagent for the single-step simultaneous isolation of RNA, DNA and proteins from cell and tissue samples, BioTechniques, vol.15, issue.3, pp.532-534, 1993.

P. E. Vandeventer, J. S. Lin, T. J. Zwang, A. Nadim, M. S. Johal et al., Multiphasic DNA adsorption to silica surfaces under varying buffer, pH, and ionic strength conditions, J. Phys. Chem. B, vol.116, pp.5661-5670, 2012.

P. Guichet, I. Bieche, and M. Teigell, Cell death and neuronal differentiation of glioblastoma stem-like cells induced by neurogenic transcription factors, Glia, vol.61, issue.2, pp.225-239, 2013.

A. Caicedo, V. Fritz, and J. Brondello, MitoCeption as a new tool to assess the effects of mesenchymal stem/stromal cell mitochondria on cancer cell metabolism and function, Sci. Rep, vol.5, p.9073, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01834600

N. Mombo, B. Gerbal-chaloin, S. Bokus, and A. , MitoCeption: transferring isolated human MSC mitochondria to glioblastoma stem cells, J. Vis. Exp, vol.120, p.55245, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01777706

E. A. Lyons, M. K. Scheible, K. Sturk-andreaggi, J. A. Irwin, and R. S. Just, A high-throughput Sanger strategy for human mitochondrial genome sequencing, BMC Genomics, vol.14, issue.1, pp.1-16, 2013.

A. L. Andreu, R. Martinez, R. Marti, and E. García-arumí, Quantification of mitochondrial DNA copy number: pre-analytical factors, Mitochondrion, vol.9, issue.4, pp.242-246, 2009.

W. Guo, L. Jiang, S. Bhasin, S. M. Khan, and R. H. Swerdlow, DNA extraction procedures meaningfully influence qPCR-based mtDNA copy number determination, Mitochondrion, vol.9, issue.4, pp.261-265, 2009.

E. Nacheva, K. Mokretar, and A. Soenmez, DNA isolation protocol effects on nuclear DNA analysis by microarrays, droplet digital PCR, and whole genome sequencing, and on mitochondrial DNA copy number estimation, PLoS ONE, vol.12, issue.7, p.180467, 2017.

R. O'hara, E. Tedone, and A. Ludlow, Quantitative mitochondrial DNA copy number determination using droplet digital PCR with single-cell resolution, Genome Res, vol.29, issue.11, pp.1878-1888, 2019.

, C 2020 Marie-Luce Vignais www, BioTechniques.com, vol.69, issue.6

R. Maeda, D. Kami, H. Maeda, A. Shikuma, and S. Gojo, High throughput single cell analysis of mitochondrial heteroplasmy in mitochondrial diseases, Sci. Rep, vol.10, issue.1, p.10821, 2020.

R. J. Longchamps, C. A. Castellani, and Y. Sy, Evaluation of mitochondrial DNA copy number estimation techniques, PLoS ONE, vol.15, issue.1, p.228166, 2020.

C. B. Jackson, D. M. Turnbull, M. Minczuk, and P. A. Gammage, Therapeutic manipulation of mtDNA heteroplasmy: a shifting perspective, Trends Mol. Med, vol.26, issue.7, pp.698-709, 2020.

Y. Jang and K. Lim, Recent advances in mitochondria-targeted gene delivery, Molecules, vol.23, issue.9, p.2316, 2018.

B. Y. Mok, M. H. De-moraes, and J. Zeng, A bacterial cytidine deaminase toxin enables CRISPR-free mitochondrial base editing, Nature, vol.583, issue.7817, pp.631-637, 2020.

, C 2020 Marie-Luce Vignais www, BioTechniques.com, vol.69, issue.6