M. Alexeyev, I. Shokolenko, G. Wilson, and S. Ledoux, The maintenance of mitochondrial DNA integrity-critical analysis and update, Cold Spring Harbor Perspect Biol, vol.5, p.12641, 2013.

I. Amelio, F. Cutruzzola, A. Antonov, M. Agostini, and G. Melino, Serine and glycine metabolism in cancer, Trends Biochem Sci, vol.39, pp.191-198, 2014.

S. Anderson, A. T. Bankier, B. G. Barrell, M. H. De-bruijn, A. R. Coulson et al., Sequence and organization of the human mitochondrial genome, Nature, vol.290, pp.457-465, 1981.

G. Antoch, N. Saoudi, H. Kuehl, G. Dahmen, S. P. Mueller et al., Accuracy of whole-body dual-modality fluorine-18-2-fluoro-2-deoxy-D-glucose positron emission tomography and computed tomography (FDG-PET/CT) for tumor staging in solid tumors: comparison with CT and PET, J Clin Oncol, vol.22, pp.4357-4368, 2004.

M. Augsten, E. Sjoberg, O. Frings, S. U. Vorrink, J. Frijhoff et al., Cancer-associated fibroblasts expressing CXCL14 rely upon NOS1-derived nitric oxide signaling for their tumor-supporting properties, Cancer Res, vol.74, pp.2999-3010, 2014.

. Gentric and . Al,

Y. Bao, K. Mukai, T. Hishiki, A. Kubo, M. Ohmura et al., Energy management by enhanced glycolysis in G1-phase in human colon cancer cells in vitro and in vivo, Mol Cancer Res, vol.11, pp.973-985, 2013.

J. I. Bardos and M. Ashcroft, Hypoxia-inducible factor-1 and oncogenic signalling, BioEssays, vol.26, pp.262-269, 2004.

L. Batista, T. Gruosso, M. , and F. , Ovarian cancer emerging subtypes: role of oxidative stress and fibrosis in tumour development and response to treatment, Int J Biochem Cell Biol, vol.45, pp.1092-1098, 2013.

T. Becker, L. Bottinger, and N. Pfanner, Mitochondrial protein import: from transport pathways to an integrated network, Trends Biochem Sci, vol.37, pp.85-91, 2012.

L. Berod, C. Friedrich, A. Nandan, J. Freitag, S. Hagemann et al., De novo fatty acid synthesis controls the fate between regulatory T and T helper 17 cells, Nat Med, vol.20, pp.1327-1333, 2014.

K. Birsoy, T. Wang, W. W. Chen, E. Freinkman, M. Abu-remaileh et al., An essential role of the mitochondrial electron transport chain in cell proliferation is to enable aspartate synthesis, Cell, vol.162, pp.540-551, 2015.

M. J. Bissell and W. C. Hines, Why don't we get more cancer? A proposed role of the microenvironment in restraining cancer progression, Nat Med, vol.17, pp.320-329, 2011.

S. K. Biswas, Metabolic reprogramming of immune cells in cancer progression, Immunity, vol.43, pp.435-449, 2015.

S. K. Biswas and A. Mantovani, Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm, Nat Immunol, vol.11, pp.889-896, 2010.

E. Blanchet, J. S. Annicotte, S. Lagarrigue, V. Aguilar, C. Clape et al., E2F transcription factor-1 regulates oxidative metabolism, Nat Cell Biol, vol.13, pp.1146-1152, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02193308

L. Bochet, C. Lehuede, S. Dauvillier, Y. Y. Wang, B. Dirat et al., Adipocyte-derived fibroblasts promote tumor progression and contribute to the desmoplastic reaction in breast cancer, Cancer Res, vol.73, pp.5657-5668, 2013.

C. Bonnans, J. Chou, and Z. Werb, Remodelling the extracellular matrix in development and disease, Nat Rev Mol Cell Biol, vol.15, pp.786-801, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01952416

S. Bonnet, S. L. Archer, J. Allalunis-turner, A. Haromy, C. Beaulieu et al., A mitochondria-K+ channel axis is suppressed in cancer and its normalization promotes apoptosis and inhibits cancer growth, Cancer Cell, vol.11, pp.37-51, 2007.

L. Bordone and L. Guarente, Calorie restriction, SIRT1 and metabolism: understanding longevity, Nat Rev Mol Cell Biol, vol.6, pp.298-305, 2005.

M. C. Brahimi-horn, G. Bellot, and P. J. , Hypoxia and energetic tumour metabolism, Curr Opin Genet Dev, vol.21, pp.67-72, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00546668

M. C. Brahimi-horn, J. Chiche, and P. J. , Hypoxia signalling controls metabolic demand, Curr Opin Cell Biol, vol.19, pp.23-32, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00319880

A. Csibi, G. Lee, S. O. Yoon, H. Tong, D. Ilter et al., The mTORC1/S6K1 pathway regulates glutamine metabolism through the eIF4B-dependent control of c-Myc translation, Curr Biol, vol.24, pp.2274-2280, 2014.

E. Currie, A. Schulze, R. Zechner, T. C. Walther, and R. V. Farese, Cellular fatty acid metabolism and cancer, Cell Metab, vol.18, pp.153-161, 2013.

L. Dang, D. W. White, S. Gross, B. D. Bennett, M. A. Bittinger et al., Cancer-associated IDH1 mutations produce 2-hydroxyglutarate, Nature, vol.462, pp.739-744, 2009.

D. Rj and T. Cheng, Q's next: the diverse functions of glutamine in metabolism, cell biology and cancer, Oncogene, vol.29, pp.313-324, 2010.

R. J. Deberardinis, J. J. Lum, G. Hatzivassiliou, and C. B. Thompson, The biology of cancer: metabolic reprogramming fuels cell growth and proliferation, Cell Metab, vol.7, pp.11-20, 2008.

G. Deblois, J. St-pierre, and V. Giguere, The PGC-1/ERR signaling axis in cancer, Oncogene, vol.32, pp.3483-3490, 2013.

G. M. Delgoffe, K. N. Pollizzi, A. T. Waickman, E. Heikamp, D. J. Meyers et al., The kinase mTOR regulates the differentiation of helper T cells through the selective activation of signaling by mTORC1 and mTORC2, Nat Immunol, vol.12, pp.295-303, 2011.

G. M. Denicola, P. H. Chen, E. Mullarky, J. A. Sudderth, Z. Hu et al., NRF2 regulates serine biosynthesis in non-small cell lung cancer, Nat Genet, vol.47, pp.1475-1481, 2015.

F. Dupuy, S. Tabaries, S. Andrzejewski, Z. Dong, J. Blagih et al., PDK1-dependent metabolic reprogramming dictates metastatic potential in breast cancer, Cell Metab, vol.22, pp.577-589, 2015.

N. Erez, M. Truitt, P. Olson, S. T. Arron, and D. Hanahan, Cancerassociated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kappaBdependent manner, Cancer Cell, vol.17, pp.135-147, 2010.

M. J. Evans and R. C. Scarpulla, Interaction of nuclear factors with multiple sites in the somatic cytochrome c promoter. Characterization of upstream NRF-1, ATF, and intron Sp1 recognition sequences, J Biol Chem, vol.264, pp.14361-14368, 1989.

V. R. Fantin, J. St-pierre, and P. Leder, Attenuation of LDH-A expression uncovers a link between glycolysis, mitochondrial physiology, and tumor maintenance, Cancer Cell, vol.9, pp.425-434, 2006.

C. Feig, J. O. Jones, M. Kraman, R. J. Wells, A. Deonarine et al., Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer, Proc Natl Acad Sci U S A, vol.110, pp.20212-20217, 2013.

J. Feilchenfeldt, M. A. Brundler, C. Soravia, M. Totsch, and M. Ca, Peroxisome proliferator-activated receptors (PPARs) and associated transcription factors in colon cancer: reduced expression of PPARgamma-coactivator 1 (PGC-1), Cancer Lett, vol.203, pp.25-33, 2004.

W. Feng, A. Gentles, R. V. Nair, M. Huang, Y. Lin et al., Targeting unique metabolic properties of breast tumor initiating cells, Stem Cells, vol.32, pp.1734-1745, 2014.

T. Fiaschi, A. Marini, E. Giannoni, M. L. Taddei, P. Gandellini et al., Reciprocal metabolic reprogramming through lactate shuttle coordinately influences tumor-stroma interplay, Cancer Res, vol.72, pp.5130-5140, 2012.

D. K. Finlay, E. Rosenzweig, L. V. Sinclair, C. Feijoo-carnero, J. L. Hukelmann et al., PDK1 regulation of mTOR and hypoxiainducible factor 1 integrate metabolism and migration of CD8+ T cells, J Exp Med, vol.209, pp.2441-2453, 2012.

R. S. Finn, J. Dering, D. Conklin, O. Kalous, D. J. Cohen et al.,

, inhibitor, preferentially inhibits proliferation of luminal estrogen receptor-positive human breast cancer cell lines in vitro, Breast Cancer Res, vol.11, p.77, 2009.

A. Fiori, X. Perez-martinez, and F. Td, Overexpression of the COX2 translational activator, Pet111p, prevents translation of COX1 mRNA and cytochrome c oxidase assembly in mitochondria of Saccharomyces cerevisiae, Mol Microbiol, vol.56, pp.1689-1704, 2005.

C. Frantz, K. M. Stewart, and V. M. Weaver, The extracellular matrix at a glance, J Cell Sci, vol.123, pp.4195-4200, 2010.

K. A. Frauwirth, J. L. Riley, M. H. Harris, R. V. Parry, J. C. Rathmell et al., The CD28 signaling pathway regulates glucose metabolism, Immunity, vol.16, pp.769-777, 2002.

C. Frezza and E. Gottlieb, Mitochondria in cancer: not just innocent bystanders, Semin Cancer Biol, vol.19, pp.4-11, 2009.

T. Fujino, J. Kondo, M. Ishikawa, K. Morikawa, and Y. Tt, Acetyl-CoA synthetase 2, a mitochondrial matrix enzyme involved in the oxidation of acetate, J Biol Chem, vol.276, pp.11420-11426, 2001.

S. Galvan-pena, O. , and L. A. , Metabolic reprograming in macrophage polarization, Front Immunol, vol.5, p.420, 2014.

P. Gao, I. Tchernyshyov, T. C. Chang, Y. S. Lee, K. Kita et al., c-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism, Nature, vol.458, pp.762-765, 2009.

G. Gasparre, A. M. Porcelli, G. Lenaz, and R. G. , Relevance of mitochondrial genetics and metabolism in cancer development, Cold Spring Harbor Perspect Biol, vol.5, 2013.

R. A. Gatenby and R. J. Gillies, Why do cancers have high aerobic glycolysis?, Nat Rev Cancer, vol.4, pp.891-899, 2004.

G. Gentric, V. Maillet, V. Paradis, D. Couton, L. 'hermitte et al., Oxidative stress promotes pathologic polyploidization in nonalcoholic fatty liver disease, J Clin Invest, vol.125, pp.981-992, 2015.

D. Gerald, E. Berra, Y. M. Frapart, D. A. Chan, A. J. Giaccia et al., JunD reduces tumor angiogenesis by protecting cells from oxidative stress, Cell, vol.118, pp.781-794, 2004.
URL : https://hal.archives-ouvertes.fr/hal-00322373

E. Giannoni, F. Bianchini, L. Calorini, and P. Chiarugi, Cancer associated fibroblasts exploit reactive oxygen species through a proinflammatory signature leading to epithelial mesenchymal transition and stemness, Antioxid Redox Signal, vol.14, pp.2361-2371, 2011.

. Gentric and . Al,

E. Giannoni, M. L. Taddei, A. Morandi, G. Comito, M. Calvani et al., Targeting stromal-induced pyruvate kinase M2 nuclear translocation impairs oxphos and prostate cancer metastatic spread, Oncotarget, vol.6, pp.24061-24074, 2015.

N. Gleyzer, K. Vercauteren, and R. C. Scarpulla, Control of mitochondrial transcription specificity factors (TFB1M and TFB2M) by nuclear respiratory factors (NRF-1 and NRF-2) and PGC-1 family coactivators, Mol Cell Biol, vol.25, pp.1354-1366, 2005.

J. G. Goetz, S. Minguet, I. Navarro-lerida, J. J. Lazcano, R. Samaniego et al., Biomechanical remodeling of the microenvironment by stromal caveolin-1 favors tumor invasion and metastasis, Cell, vol.146, pp.148-163, 2011.

V. Gogvadze, S. Orrenius, and B. Zhivotovsky, Mitochondria in cancer cells: what is so special about them?, Trends Cell Biol, vol.18, pp.165-173, 2008.

G. S. Taylor and P. R. , Monocyte and macrophage heterogeneity, Nat Rev Immunol, vol.5, pp.953-964, 2005.

K. Grohmann, F. Amairic, S. Crews, and G. Attardi, Failure to detect ''cap'' structures in mitochondrial DNA-coded poly(A)-containing RNA from HeLa cells, Nucleic Acids Res, vol.5, pp.637-651, 1978.

S. Gugneja, C. M. Virbasius, and R. C. Scarpulla, Nuclear respiratory factors 1 and 2 utilize similar glutaminecontaining clusters of hydrophobic residues to activate transcription, Mol Cell Biol, vol.16, pp.5708-5716, 1996.

M. Guppy, P. Leedman, X. Zu, and R. V. , Contribution by different fuels and metabolic pathways to the total ATP turnover of proliferating MCF-7 breast cancer cells, Biochem J, vol.364, pp.309-315, 2002.

H. Mc and D. A. Sinclair, Mammalian sirtuins: biological insights and disease relevance, Ann Rev Pathol, vol.5, pp.253-295, 2010.

W. C. Hallows, W. Yu, and J. M. Denu, Regulation of glycolytic enzyme phosphoglycerate mutase-1 by Sirt1 proteinmediated deacetylation, J Biol Chem, vol.287, pp.3850-3858, 2012.

D. Hanahan and R. A. Weinberg, Hallmarks of cancer: the next generation, Cell, vol.144, pp.646-674, 2011.

R. Haq, S. Yokoyama, E. B. Hawryluk, G. B. Jonsson, D. T. Frederick et al., BCL2A1 is a lineage-specific antiapoptotic melanoma oncogene that confers resistance to BRAF inhibition, Proc Natl Acad Sci, vol.110, pp.4321-4326, 2013.

G. C. Hard, Some biochemical aspects of the immune macrophage, Br J Exp Pathol, vol.51, pp.97-105, 1970.

S. Haxhinasto, D. Mathis, and C. Benoist, The AKT-mTOR axis regulates de novo differentiation of CD4+Foxp3+ cells, J Exp Med, vol.205, pp.565-574, 2008.

W. He, J. C. Newman, M. Z. Wang, L. Ho, and E. Verdin, Mitochondrial sirtuins: regulators of protein acylation and metabolism, Trends Endocrinol Metab, vol.23, pp.467-476, 2012.

J. M. Herrmann, M. W. Woellhaf, and N. Bonnefoy, Control of protein synthesis in yeast mitochondria: the concept of translational activators, Biochim Biophys Acta, vol.1833, pp.286-294, 2013.

P. C. Ho, J. D. Bihuniak, A. N. Macintyre, M. Staron, X. Liu et al., Phosphoenolpyruvate is a metabolic checkpoint of antitumor T cell responses, Cell, vol.162, pp.1217-1228, 2015.

B. L. Horecker, The pentose phosphate pathway, J Biol Chem, vol.277, pp.47965-47971, 2002.

A. L. Horwich, F. Kalousek, I. Mellman, and L. E. Rosenberg, A leader peptide is sufficient to direct mitochondrial import of a chimeric protein, EMBO J, vol.4, pp.1129-1135, 1985.

S. C. Huang, B. Everts, Y. Ivanova, D. O'sullivan, M. Nascimento et al., Cell-intrinsic lysosomal lipolysis is essential for alternative activation of macrophages, Nat Immunol, vol.15, pp.846-855, 2014.

F. J. Iborra, H. Kimura, and P. R. Cook, The functional organization of mitochondrial genomes in human cells, BMC Biol, vol.2, p.9, 2004.

K. Ito and T. Suda, Metabolic requirements for the maintenance of self-renewing stem cells, Nat Rev Mol Cell Biol, vol.15, pp.243-256, 2014.

S. Jager, C. Handschin, J. St-pierre, and B. M. Spiegelman, AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1alpha, Proc Natl Acad Sci, vol.104, pp.12017-12022, 2007.

M. Janiszewska, M. L. Suva, N. Riggi, R. H. Houtkooper, J. Auwerx et al., Imp2 controls oxidative phosphorylation and is crucial for preserving glioblastoma cancer stem cells, Genes Dev, vol.26, pp.1926-1944, 2012.

E. H. Jeninga, K. Schoonjans, and A. J. , Reversible acetylation of PGC-1: connecting energy sensors and effectors to guarantee metabolic flexibility, Oncogene, vol.29, pp.4617-4624, 2010.

A. Jezierska-drutel, S. A. Rosenzweig, and C. A. Neumann, Role of oxidative stress and the microenvironment in breast cancer development and progression, Adv Cancer Res, vol.119, pp.107-125, 2013.

A. K. Jha, S. C. Huang, A. Sergushichev, V. Lampropoulou, Y. Ivanova et al., Network integration of parallel metabolic and transcriptional data reveals metabolic modules that regulate macrophage polarization, Immunity, vol.42, pp.419-430, 2015.

R. G. Jones and C. B. Thompson, Revving the engine: signal transduction fuels T cell activation, Immunity, vol.27, pp.173-178, 2007.

C. Jose, N. Bellance, and R. Rossignol, Choosing between glycolysis and oxidative phosphorylation: a tumor's dilemma?, Biochim Biophys Acta, vol.1807, pp.552-561, 2011.

J. A. Joyce and D. T. Fearon, T cell exclusion, immune privilege, and the tumor microenvironment, Science, vol.348, pp.74-80, 2015.

S. C. Kalhan and R. W. Hanson, Resurgence of serine: an often neglected but indispensable amino Acid, J Biol Chem, vol.287, 2012.

M. M. Kaminski, S. W. Sauer, M. Kaminski, S. Opp, T. Ruppert et al., T cell activation is driven by an ADPdependent glucokinase linking enhanced glycolysis with mitochondrial reactive oxygen species generation, Cell Rep, vol.2, pp.1300-1315, 2012.

M. M. Kaminski, S. W. Sauer, C. D. Klemke, D. Suss, J. G. Okun et al., Mitochondrial reactive oxygen species control T cell activation by regulating IL-2 and IL-4 expression: mechanism of ciprofloxacin-mediated immunosuppression, J Immunol, vol.184, pp.4827-4841, 2010.

G. S. Karagiannis, T. Poutahidis, S. E. Erdman, R. Kirsch, R. H. Riddell et al., Cancer-associated fibroblasts drive the progression of metastasis through both paracrine and mechanical pressure on cancer tissue, Mol Cancer Res, vol.10, pp.1403-1418, 2012.

K. B. , O. , and L. A. , Metabolic reprogramming in macrophages and dendritic cells in innate immunity, Cell Res, vol.25, pp.771-784, 2015.

D. P. Kelly and R. C. Scarpulla, Transcriptional regulatory circuits controlling mitochondrial biogenesis and function, Genes Dev, vol.18, pp.357-368, 2004.

J. W. Kim, I. Tchernyshyov, G. L. Semenza, and C. V. Dang, HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia, Cell Metab, vol.3, pp.177-185, 2006.

M. P. King and G. Attardi, Human cells lacking mtDNA: repopulation with exogenous mitochondria by complementation, Science, vol.246, pp.500-503, 1989.

D. Klysz, X. Tai, P. A. Robert, M. Craveiro, G. Cretenet et al., Glutaminedependent alpha-ketoglutarate production regulates the balance between T helper 1 cell and regulatory T cell generation, Sci Signal, vol.8, p.97, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02187398

H. A. Krebs, A. Ruffo, M. Johnson, L. V. Eggleston, and H. R. Oxidative-phosphorylation, Biochem J, vol.54, pp.107-116, 1953.

H. N. Kung, J. R. Marks, and C. Jt, Glutamine synthetase is a genetic determinant of cell type-specific glutamine independence in breast epithelia, PLoS Genet, vol.7, p.1002229, 2011.

E. D. Lagadinou, A. Sach, K. Callahan, R. M. Rossi, S. J. Neering et al., BCL-2 inhibition targets oxidative phosphorylation and selectively eradicates quiescent human leukemia stem cells, Cell Stem Cell, vol.12, pp.329-341, 2013.

L. Lapeire, A. Hendrix, K. Lambein, M. Van-bockstal, G. Braems et al., Cancer-associated adipose tissue promotes breast cancer progression by paracrine oncostatin M and Jak/STAT3 signaling, Cancer Res, vol.74, pp.6806-6819, 2014.

M. Laplante and D. M. Sabatini, mTOR signaling in growth control and disease, Cell, vol.149, pp.274-293, 2012.

G. Laurent, F. Solari, B. Mateescu, M. Karaca, J. Castel et al., Oxidative stress contributes to aging by enhancing pancreatic angiogenesis and insulin signaling, Cell Metab, vol.7, pp.113-124, 2008.

V. S. Lebleu, J. T. O'connell, G. Herrera, K. N. Wikman, H. Pantel et al., PGC-1alpha mediates mitochondrial biogenesis and oxidative phosphorylation in cancer cells to promote metastasis, Nat Cell Biol, vol.16, pp.1001-1015, 2014.

S. Lefort, C. Joffre, Y. Kieffer, A. M. Givel, B. Bourachot et al., Inhibition of autophagy as a new means of improving chemotherapy efficiency in high-LC3B triple-negative breast cancers, Autophagy, vol.10, pp.2122-2142, 2014.

J. M. Lemons, X. J. Feng, B. D. Bennett, A. Legesse-miller, J. E. Raitman et al., Quiescent fibroblasts exhibit high metabolic activity, PLoS Biol, vol.8, p.1000514, 2010.

A. J. Levine and A. M. Puzio-kuter, The control of the metabolic switch in cancers by oncogenes and tumor suppressor genes, Science, vol.330, pp.1340-1344, 2010.

F. Li, Y. Wang, K. I. Zeller, J. J. Potter, D. R. Wonsey et al., Myc stimulates nuclearly encoded mitochondrial genes and mitochondrial biogenesis, Mol Cell Biol, vol.25, pp.6225-6234, 2005.

H. X. Liao and L. L. Spremulli, Interaction of bovine mitochondrial ribosomes with messenger RNA, J Biol Chem, vol.264, pp.7518-7522, 1989.

J. H. Lim, C. Luo, F. Vazquez, and P. Puigserver, Targeting mitochondrial oxidative metabolism in melanoma causes metabolic compensation through glucose and glutamine utilization, Cancer Res, vol.74, pp.3535-3545, 2014.

J. Lin, P. Puigserver, J. Donovan, P. Tarr, and B. M. Spiegelman, Peroxisome proliferator-activated receptor gamma coactivator 1beta (PGC-1beta), a novel PGC-1-related transcription coactivator associated with host cell factor, J Biol Chem, vol.277, pp.1645-1648, 2002.

X. Lin, W. Zheng, J. Liu, Y. Zhang, H. Qin et al., Oxidative stress in malignant melanoma enhances tumor necrosis factor-alpha secretion of tumor-associated macrophages that promote cancer cell invasion, Antioxid Redox Signal, vol.19, pp.1337-1355, 2013.

W. Liu, A. Le, C. Hancock, A. N. Lane, C. V. Dang et al., Reprogramming of proline and glutamine metabolism contributes to the proliferative and metabolic responses regulated by oncogenic transcription factor c-MYC, Proc Natl Acad Sci, vol.109, pp.8983-8988, 2012.

J. W. Locasale and . Serine, glycine and one-carbon units: cancer metabolism in full circle, Nat Rev Cancer, vol.13, pp.572-583, 2013.

J. W. Locasale, A. R. Grassian, T. Melman, C. A. Lyssiotis, K. R. Mattaini et al., Phosphoglycerate dehydrogenase diverts glycolytic flux and contributes to oncogenesis, Nat Genet, vol.43, pp.869-874, 2011.

. Lopez-mejia-ic and L. Fajas, Cell cycle regulation of mitochondrial function, Curr Opin Cell Biol, vol.33, pp.19-25, 2015.

O. D. Maddocks, C. R. Berkers, S. M. Mason, L. Zheng, K. Blyth et al., Serine starvation induces stress and p53-dependent metabolic remodelling in cancer cells, Nature, vol.493, pp.542-546, 2013.

A. Mantovani, S. Sozzani, M. Locati, P. Allavena, and A. Sica, Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes, Trends Immunol, vol.23, pp.549-555, 2002.

I. Marin-valencia, C. Yang, T. Mashimo, S. Cho, H. Baek et al., Analysis of tumor metabolism reveals mitochondrial glucose oxidation in genetically diverse human glioblastomas in the mouse brain in vivo, Cell Metab, vol.15, pp.827-837, 2012.

U. E. Martinez-outschoorn, F. Sotgia, and L. Mp, Caveolae and signalling in cancer, Nat Rev Cancer, vol.15, pp.225-237, 2015.

V. Mcculloch, B. L. Seidel-rogol, and G. S. Shadel, A human mitochondrial transcription factor is related to RNA adenine methyltransferases and binds S-adenosylmethionine, Mol Cell Biol, vol.22, pp.1116-1125, 2002.

E. M. Mcinerney, D. W. Rose, S. E. Flynn, S. Westin, T. M. Mullen et al., Determinants of coactivator LXXLL motif specificity in nuclear receptor transcriptional activation, Genes Dev, vol.12, pp.3357-3368, 1998.

D. Medici and R. Kalluri, Endothelial-mesenchymal transition and its contribution to the emergence of stem cell phenotype, Semin Cancer Biol, vol.22, pp.379-384, 2012.

I. Mercier, J. Camacho, K. Titchen, D. M. Gonzales, K. Quann et al., Caveolin-1 and accelerated host aging in the breast tumor microenvironment: chemoprevention with rapamycin, an mTOR inhibitor and anti-aging drug, Am J Pathol, vol.181, pp.278-293, 2012.

R. D. Michalek, V. A. Gerriets, S. R. Jacobs, A. N. Macintyre, N. J. Maciver et al., Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets, J Immunol, vol.186, pp.3299-3303, 2011.

M. Michaud, I. Martins, A. Q. Sukkurwala, S. Adjemian, Y. Ma et al., Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice, Science, vol.334, pp.1573-1577, 2011.

E. D. Michelakis, G. Sutendra, P. Dromparis, L. Webster, A. Haromy et al., Metabolic modulation of glioblastoma with dichloroacetate, Sci Transl Med, vol.2, pp.31-34, 2010.

K. Mitra, C. Wunder, B. Roysam, G. Lin, L. et al., A hyperfused mitochondrial state achieved at G1-S regulates cyclin E buildup and entry into S phase, Proc Natl Acad Sci, vol.106, pp.11960-11965, 2009.

S. Monti, K. J. Savage, J. L. Kutok, F. Feuerhake, P. Kurtin et al., Molecular profiling of diffuse large B-cell lymphoma identifies robust subtypes including one characterized by host inflammatory response, Blood, vol.105, pp.1851-1861, 2005.

R. Moreno-sanchez, S. Rodriguez-enriquez, A. Marin-hernandez, and E. Saavedra, Energy metabolism in tumor cells, FEBS J, vol.274, pp.1393-1418, 2007.

R. Moreno-sanchez, S. Rodriguez-enriquez, E. Saavedra, A. Marin-hernandez, and J. C. Gallardo-perez, The bioenergetics of cancer: is glycolysis the main ATP supplier in all tumor cells, Biofactors, vol.35, pp.209-225, 2009.

M. Morita, S. P. Gravel, V. Chenard, K. Sikstrom, L. Zheng et al., Topisirovic I, and Sonenberg N. mTORC1 controls mitochondrial activity and biogenesis through 4E-BP-dependent translational regulation, vol.18, pp.698-711, 2013.

M. P. Murphy, A. Holmgren, N. G. Larsson, B. Halliwell, C. J. Chang et al., Unraveling the biological roles of reactive oxygen species, Cell Metab, vol.13, pp.361-366, 2011.

P. Nagley, Coordination of gene expression in the formation of mammalian mitochondria, Trends Genet, vol.7, pp.1-4, 1991.

P. Nahon, A. Sutton, D. Pessayre, P. Rufat, F. Degoul et al., Genetic dimorphism in superoxide dismutase and susceptibility to alcoholic cirrhosis, hepatocellular carcinoma, and death, Clin Gastroenterol Hepatol, vol.3, pp.292-298, 2005.

N. Dl, M. M. Cox, and . Lehninger, Principles of Biochemistry, 2013.

P. Newsholme, J. Procopio, M. M. Lima, T. C. Pithon-curi, and R. Curi, Glutamine and glutamate-their central role in cell metabolism and function, Cell Biochem Funct, vol.21, pp.1-9, 2003.

R. Noy and J. W. Pollard, Tumor-associated macrophages: from mechanisms to therapy, Immunity, vol.41, pp.49-61, 2014.

E. Obre and R. Rossignol, Emerging concepts in bioenergetics and cancer research: metabolic flexibility, coupling, symbiosis, switch, oxidative tumors, metabolic remodeling, signaling and bioenergetic therapy, Int J Biochem Cell Biol, vol.59, pp.167-181, 2015.

J. I. Odegaard and A. Chawla, Alternative macrophage activation and metabolism, Ann Rev Pathol, vol.6, pp.275-297, 2011.

D. Ohlund, E. Elyada, and D. Tuveson, Fibroblast heterogeneity in the cancer wound, J Exp Med, vol.211, pp.1503-1523, 2014.

D. Ojala, J. Montoya, and G. Attardi, tRNA punctuation model of RNA processing in human mitochondria, Nature, vol.290, pp.470-474, 1981.

I. Okoye, L. Wang, K. Pallmer, K. Richter, T. Ichimura et al., T cell metabolism. The protein LEM promotes CD8(+) T cell immunity through effects on mitochondrial respiration, Science, vol.348, pp.995-1001, 2015.

A. Ozaki, T. Tanimoto, and S. Saji, Palbociclib in Hormone-Receptor-Positive Advanced Breast Cancer, N Engl J Med, vol.373, pp.1672-1673, 2015.

S. Paget, The distribution of secondary growths in cancer of the breast, Lancet, vol.133, pp.571-573, 1889.

A. D. Panopoulos, O. Yanes, S. Ruiz, Y. S. Kida, D. Diep et al., The metabolome of induced pluripotent stem cells reveals metabolic changes occurring in somatic cell reprogramming, Cell Res, vol.22, pp.168-177, 2012.

I. Papandreou, R. A. Cairns, L. Fontana, A. L. Lim, and D. Nc, HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption, Cell Metab, vol.3, pp.187-197, 2006.

G. C. Parker, G. Acsadi, and C. A. Brenner, Mitochondria: determinants of stem cell fate?, Stem Cells Dev, vol.18, pp.803-806, 2009.

M. Parri and P. Chiarugi, Redox molecular machines involved in tumor progression, Antioxid Redox Signal, vol.19, pp.1828-1845, 2013.

D. W. Parsons, S. Jones, X. Zhang, J. C. Lin, R. J. Leary et al., An integrated genomic analysis of human glioblastoma multiforme, Science, vol.321, pp.1807-1812, 2008.

A. Pasto, C. Bellio, G. Pilotto, V. Ciminale, M. Silic-benussi et al., Cancer stem cells from epithelial ovarian cancer patients privilege oxidative phosphorylation, and resist glucose deprivation, Oncotarget, vol.5, pp.4305-4319, 2014.

S. Pavlides, D. Whitaker-menezes, R. Castello-cros, N. Flomenberg, A. K. Witkiewicz et al., The reverse Warburg effect: aerobic glycolysis in cancer associated fibroblasts and the tumor stroma, Cell Cycle, vol.8, pp.3984-4001, 2009.

L. S. Pike, A. L. Smift, N. J. Croteau, D. A. Ferrick, and M. Wu, Inhibition of fatty acid oxidation by etomoxir impairs NADPH production and increases reactive oxygen species resulting in ATP depletion and cell death in human glioblastoma cells, Biochim Biophys Acta, vol.1807, pp.726-734, 2011.

R. Possemato, K. M. Marks, Y. D. Shaul, M. E. Pacold, D. Kim et al., Functional genomics reveal that the serine synthesis pathway is essential in breast cancer, Nature, vol.476, pp.346-350, 2011.

J. Pouyssegur and F. Mechta-grigoriou, Redox regulation of the hypoxia-inducible factor, Biol Chem, vol.387, pp.1337-1346, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00321043

P. Puigserver, Z. Wu, C. W. Park, R. Graves, M. Wright et al., A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis, Cell, vol.92, pp.829-839, 1998.

D. F. Quail and J. A. Joyce, Microenvironmental regulation of tumor progression and metastasis, Nat Med, vol.19, pp.1423-1437, 2013.

D. C. Radisky, D. D. Levy, L. E. Littlepage, H. Liu, C. M. Nelson et al., Rac1b and reactive oxygen species mediate MMP-3-induced EMT and genomic instability, Nature, vol.436, pp.123-127, 2005.

R. R. Rao, Q. Li, K. Odunsi, and P. A. Shrikant, The mTOR kinase determines effector versus memory CD8+ T cell fate by regulating the expression of transcription factors T-bet and Eomesodermin, Immunity, vol.32, pp.67-78, 2010.

D. Rapaport, Finding the right organelle. Targeting signals in mitochondrial outer-membrane proteins, EMBO Rep, vol.4, pp.948-952, 2003.

Y. I. Rattigan, B. B. Patel, E. Ackerstaff, G. Sukenick, J. A. Koutcher et al., Lactate is a mediator of metabolic cooperation between stromal carcinoma associated fibroblasts and glycolytic tumor cells in the tumor microenvironment, Exp Cell Res, vol.318, pp.326-335, 2012.

L. J. Reitzer and B. M. Wice, Evidence that glutamine, not sugar, is the major energy source for cultured HeLa cells, J Biol Chem, vol.254, pp.2669-2676, 1979.

J. T. Rodgers, C. Lerin, W. Haas, S. P. Gygi, B. M. Spiegelman et al., Nutrient control of glucose homeostasis through a complex of PGC-1alpha and SIRT1, Nature, vol.434, pp.113-118, 2005.

J. C. Rodriguez-prados, P. G. Traves, J. Cuenca, D. Rico, J. Aragones et al., Substrate fate in activated macrophages: a comparison between innate, classic, and alternative activation, J Immunol, vol.185, pp.605-614, 2010.

A. Roesch, A. Vultur, I. Bogeski, H. Wang, K. M. Zimmermann et al., Overcoming intrinsic multidrug resistance in melanoma by blocking the mitochondrial respiratory chain of slow-cycling JARID1B(high) cells, Cancer Cell, vol.23, pp.811-825, 2013.

J. Rorbach, R. Richter, H. J. Wessels, M. Wydro, M. Pekalski et al., The human mitochondrial ribosome recycling factor is essential for cell viability, Nucleic Acids Res, vol.36, pp.5787-5799, 2008.

D. J. Rossi, C. H. Jamieson, and W. Il, Stems cells and the pathways to aging and cancer, Cell, vol.132, pp.681-696, 2008.

R. Rossignol, R. Gilkerson, R. Aggeler, K. Yamagata, S. J. Remington et al., Energy substrate modulates mitochondrial structure and oxidative capacity in cancer cells, Cancer Res, vol.64, pp.985-993, 2004.

A. Rynne-vidal, J. A. Jimenez-heffernan, C. Fernandez-chacon, M. Lopez-cabrera, and P. Sandoval, The mesothelial origin of carcinoma associated-fibroblasts in peritoneal metastasis, Cancers, vol.7, 1994.

T. Sakamaki, M. C. Casimiro, X. Ju, A. A. Quong, S. Katiyar et al., Cyclin D1 determines mitochondrial function in vivo, Mol Cell Biol, vol.26, pp.5449-5469, 2006.

P. Sancho, E. Burgos-ramos, A. Tavera, B. Kheir, T. Jagust et al., MYC/PGC-1alpha Balance Determines the Metabolic Phenotype and Plasticity of Pancreatic Cancer Stem Cells, Cell Metab, vol.22, pp.590-605, 2015.

V. G. Sankaran, S. H. Orkin, and C. R. Walkley, Rb intrinsically promotes erythropoiesis by coupling cell cycle exit with mitochondrial biogenesis, Genes Dev, vol.22, pp.463-475, 2008.

L. A. Sauer, J. W. Stayman, and R. T. Dauchy, Amino acid, glucose, and lactic acid utilization in vivo by rat tumors, Cancer Res, vol.42, pp.4090-4097, 1982.

R. C. Scarpulla, Metabolic control of mitochondrial biogenesis through the PGC-1 family regulatory network, Biochim Biophys Acta, vol.1813, pp.1269-1278, 2011.

R. C. Scarpulla, Nuclear control of respiratory chain expression by nuclear respiratory factors and PGC-1-related coactivator, Ann N Y Acad Sci, vol.1147, pp.321-334, 2008.

G. Schatz, Mitochondria: beyond oxidative phosphorylation, Biochim Biophys Acta, vol.1271, pp.123-126, 1995.

S. Schoors, U. Bruning, R. Missiaen, K. C. Queiroz, G. Borgers et al., Fatty acid carbon is essential for dNTP synthesis in endothelial cells, Nature, vol.520, p.482, 2015.

S. N. Schreiber, R. Emter, M. B. Hock, D. Knutti, J. Cardenas et al., The estrogen-related receptor alpha (ERRalpha) functions in PPARgamma coactivator 1alpha (PGC-1alpha)-induced mitochondrial biogenesis, Proc Natl Acad Sci, vol.101, pp.6472-6477, 2004.

Z. T. Schug, B. Peck, D. T. Jones, Q. Zhang, S. Grosskurth et al., Acetyl-CoA synthetase 2 promotes acetate utilization and maintains cancer cell growth under metabolic stress, Cancer Cell, vol.27, pp.57-71, 2015.

L. A. Sena, S. Li, A. Jairaman, M. Prakriya, T. Ezponda et al., Mitochondria are required for antigen-specific T cell activation through reactive oxygen species signaling, Immunity, vol.38, pp.225-236, 2013.

S. Shalapour and K. M. , Immunity, inflammation, and cancer: an eternal fight between good and evil, J Clin Invest, vol.125, pp.3347-3355, 2015.

X. J. Shen, H. Zhang, G. S. Tang, X. D. Wang, R. Zheng et al., Caveolin-1 is a modulator of fibroblast activation and a potential biomarker for gastric cancer, Int J Biol Sci, vol.11, pp.370-379, 2015.

J. A. Smeitink, J. L. Loeffen, R. H. Triepels, R. J. Smeets, J. M. Trijbels et al., Nuclear genes of human complex I of the mitochondrial electron transport chain: state of the art, Hum Mol Genet, vol.7, pp.1573-1579, 1998.

J. E. Smith-garvin, G. A. Koretzky, and J. Ms, T cell activation, Ann Rev Immunol, vol.27, pp.591-619, 2009.

P. Smits, J. Smeitink, and L. Van-den-heuvel, Mitochondrial translation and beyond: processes implicated in combined oxidative phosphorylation deficiencies, J Biomed Biotechnol, p.737385, 2010.

P. Smits, J. A. Smeitink, L. P. Van-den-heuvel, M. A. Huynen, and E. Tj, Reconstructing the evolution of the mitochondrial ribosomal proteome, Nucleic Acids Res, vol.35, pp.4686-4703, 2007.

H. Soh, M. Wasa, and M. Fukuzawa, Hypoxia upregulates amino acid transport in a human neuroblastoma cell line, J Pediatr Surg, vol.42, pp.608-612, 2007.

T. Sollner, J. Rassow, M. Wiedmann, J. Schlossmann, P. Keil et al., Mapping of the protein import machinery in the mitochondrial outer membrane by crosslinking of translocation intermediates, Nature, vol.355, pp.84-87, 1992.

J. Son, C. A. Lyssiotis, H. Ying, X. Wang, S. Hua et al., Glutamine supports pancreatic cancer growth through a KRASregulated metabolic pathway, Nature, vol.496, pp.101-105, 2013.

V. Sosa, T. Moline, R. Somoza, R. Paciucci, H. Kondoh et al., Oxidative stress and cancer: an overview, Ageing Res Rev, vol.12, pp.376-390, 2013.

J. St-pierre, J. Lin, S. Krauss, P. T. Tarr, R. Yang et al., Bioenergetic analysis of peroxisome proliferator-activated receptor gamma coactivators 1alpha and 1beta (PGC-1alpha and PGC-1beta) in muscle cells, J Biol Chem, vol.278, pp.26597-26603, 2003.

T. Suda, K. Takubo, and G. L. Semenza, Metabolic regulation of hematopoietic stem cells in the hypoxic niche, Cell Stem Cell, vol.9, pp.298-310, 2011.

L. B. Sullivan, D. Y. Gui, A. M. Hosios, L. N. Bush, E. Freinkman et al., Supporting aspartate biosynthesis is an essential function of respiration in proliferating cells, Cell, vol.162, pp.552-563, 2015.

R. C. Sun and N. C. Denko, Hypoxic regulation of glutamine metabolism through HIF1 and SIAH2 supports lipid synthesis that is necessary for tumor growth, Cell Metab, vol.19, pp.285-292, 2014.

A. Sutton, A. Imbert, A. Igoudjil, V. Descatoire, S. Cazanave et al., The manganese superoxide dismutase Ala16Val dimorphism modulates both mitochondrial import and mRNA stability, Pharmacogenetics Genomics, vol.15, pp.311-319, 2005.

P. Swietach, R. D. Vaughan-jones, and A. L. Harris, Regulation of tumor pH and the role of carbonic anhydrase 9, Cancer Metastasis Rev, vol.26, pp.299-310, 2007.

N. Taguchi, N. Ishihara, A. Jofuku, T. Oka, and K. Mihara, Mitotic phosphorylation of dynamin-related GTPase Drp1 participates in mitochondrial fission, J Biol Chem, vol.282, pp.11521-11529, 2007.

F. Takakubo, P. Cartwright, N. Hoogenraad, D. R. Thorburn, F. Collins et al., An amino acid substitution in the pyruvate dehydrogenase E1 alpha gene, affecting mitochondrial import of the precursor protein, Am J Hum Genet, vol.57, pp.772-780, 1995.

Z. Tan, N. Xie, H. Cui, D. R. Moellering, A. E. Thannickal et al., Pyruvate dehydrogenase kinase 1 participates in macrophage polarization via regulating glucose metabolism, J Immunol, vol.194, pp.6082-6089, 2015.

G. M. Tannahill, A. M. Curtis, J. Adamik, E. M. Palsson-mcdermott, A. F. Mcgettrick et al., Succinate is an inflammatory signal that induces IL-1beta through HIF-1alpha, Nature, vol.496, pp.238-242, 2013.

A. Terunuma, N. Putluri, P. Mishra, E. A. Mathe, T. H. Dorsey et al., MYC-driven accumulation of 2-hydroxyglutarate is associated with breast cancer prognosis, J Clin Invest, vol.124, pp.398-412, 2014.

L. A. Timmerman, T. Holton, M. Yuneva, R. J. Louie, M. Padro et al., Glutamine sensitivity analysis identifies the xCT antiporter as a common triple-negative breast tumor therapeutic target, Cancer Cell, vol.24, pp.450-465, 2013.

A. Toullec, D. Gerald, G. Despouy, B. Bourachot, M. Cardon et al., Oxidative stress promotes myofibroblast differentiation and tumour spreading, EMBO Mol Med, vol.2, pp.211-230, 2010.

M. G. Vander-heiden, L. C. Cantley, and C. B. Thompson, Understanding the Warburg effect: the metabolic requirements of cell proliferation, Science, vol.324, pp.1029-1033, 2009.

D. Vats, L. Mukundan, J. I. Odegaard, L. Zhang, K. L. Smith et al., Oxidative metabolism and PGC-1beta attenuate macrophage-mediated inflammation, Cell Metab, vol.4, pp.13-24, 2006.

P. Vaupel, F. Kallinowski, and P. Okunieff, Blood flow, oxygen and nutrient supply, and metabolic microenvironment of human tumors: a review, Cancer Res, vol.49, pp.6449-6465, 1989.

F. Vazquez, J. H. Lim, H. Chim, K. Bhalla, G. Girnun et al., PGC1alpha expression defines a subset of human melanoma tumors with increased mitochondrial capacity and resistance to oxidative stress, Cancer Cell, vol.23, pp.287-301, 2013.

K. Vercauteren, N. Gleyzer, and R. C. Scarpulla, Short hairpin RNA-mediated silencing of PRC (PGC-1-related coactivator) results in a severe respiratory chain deficiency associated with the proliferation of aberrant mitochondria, J Biol Chem, vol.284, pp.2307-2319, 2009.

A. Viale, P. Pettazzoni, C. A. Lyssiotis, H. Ying, N. Sanchez et al., Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function, Nature, vol.514, pp.628-632, 2014.

J. A. Villena and A. Kralli, ERRalpha: a metabolic function for the oldest orphan, Trends Endocrinol Metab, vol.19, pp.269-276, 2008.

J. V. Virbasius and R. C. Scarpulla, Activation of the human mitochondrial transcription factor A gene by nuclear respiratory factors: a potential regulatory link between nuclear and mitochondrial gene expression in organelle biogenesis, Proc Natl Acad Sci, vol.91, pp.1309-1313, 1994.

J. V. Virbasius, C. A. Virbasius, and R. C. Scarpulla, Identity of GABP with NRF-2, a multisubunit activator of cytochrome oxidase expression, reveals a cellular role for an ETS domain activator of viral promoters, Genes Dev, vol.7, pp.380-392, 1993.

E. Vlashi, C. Lagadec, L. Vergnes, T. Matsutani, K. Masui et al., Metabolic state of glioma stem cells and nontumorigenic cells, Proc Natl Acad Sci, vol.108, pp.16062-16067, 2011.

E. Vlashi, C. Lagadec, L. Vergnes, K. Reue, P. Frohnen et al., Metabolic differences in breast cancer stem cells and differentiated progeny, Breast Cancer Res Treat, vol.146, pp.525-534, 2014.

C. Wallace and D. Keast, Glutamine and macrophage function, Metabolism, vol.41, pp.1016-1020, 1992.

D. C. Wallace, Mitochondrial diseases in man and mouse, Science, vol.283, pp.1482-1488, 1999.

C. Wang, Z. Li, Y. Lu, R. Du, S. Katiyar et al., Cyclin D1 repression of nuclear respiratory factor 1 integrates nuclear DNA synthesis and mitochondrial function, Proc Natl Acad Sci, vol.103, pp.11567-11572, 2006.

J. Wang, G. Ying, J. Wang, Y. Jung, J. Lu et al., Characterization of phosphoglycerate kinase-1 expression of stromal cells derived from tumor microenvironment in prostate cancer progression, Cancer Res, vol.70, pp.471-480, 2010.

Z. Wang, M. Fan, D. Candas, T. Q. Zhang, L. Qin et al., Cyclin B1/Cdk1 coordinates mitochondrial respiration for cell-cycle G2/M progression, Dev Cell, vol.29, pp.217-232, 2014.

O. Warburg, On respiratory impairment in cancer cells, Science, vol.124, pp.269-270, 1956.

O. Warburg, F. Wind, and E. Negelein, The metabolism of tumors in the body, J Gener Physiol, vol.8, pp.519-530, 1927.

P. S. Ward, J. Patel, D. R. Wise, A. , O. Bennett et al., The common feature of leukemiaassociated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate, Cancer Cell, vol.17, pp.225-234, 2010.

P. S. Ward and C. B. Thompson, Metabolic reprogramming: a cancer hallmark even warburg did not anticipate, Cancer Cell, vol.21, pp.297-308, 2012.

G. Watkins, D. , A. Mansel, R. E. , and J. Wg, The localisation and reduction of nuclear staining of PPARgamma and PGC-1 in human breast cancer, Oncol Rep, vol.12, pp.483-488, 2004.

B. T. Weinert, S. A. Wagner, H. Horn, P. Henriksen, W. R. Liu et al., Proteome-wide mapping of the Drosophila acetylome demonstrates a high degree of conservation of lysine acetylation, Sci Signal, vol.4, p.48, 2011.

S. Weinhouse, On respiratory impairment in cancer cells, Science, vol.124, pp.267-269, 1956.

S. Weinhouse, Oxidative metabolism of neoplastic tissues, Adv Cancer Res, vol.3, pp.269-325, 1955.

S. Weinhouse, The Warburg hypothesis fifty years later, Z Krebsforsch klin Onkol Cancer Res Clin Oncol, vol.87, pp.115-126, 1976.

K. E. Wellen, G. Hatzivassiliou, U. M. Sachdeva, T. V. Bui, J. R. Cross et al., ATP-citrate lyase links cellular metabolism to histone acetylation, Science, vol.324, pp.1076-1080, 2009.

K. E. Wellen and C. B. Thompson, A two-way street: reciprocal regulation of metabolism and signalling, Nat Rev Mol Cell Biol, vol.13, pp.270-276, 2012.

C. E. Wenner, M. A. Spirtes, and S. Weinhouse, Metabolism of neoplastic tissue. II. A survey of enzymes of the citric acid cycle in transplanted tumors, Cancer Res, vol.12, pp.44-49, 1952.

W. Weraarpachai, H. Antonicka, F. Sasarman, J. Seeger, B. Schrank et al., Mutation in TACO1, encoding a translational activator of COX I, results in cytochrome c oxidase deficiency and late-onset Leigh syndrome, Nat Genet, vol.41, pp.833-837, 2009.

D. R. Wise, R. J. Deberardinis, A. Mancuso, N. Sayed, X. Y. Zhang et al., Myc regulates a transcriptional program that stimulates mitochondrial glutaminolysis and leads to glutamine addiction, Proc Natl Acad Sci, vol.105, pp.18782-18787, 2008.

G. Wright, K. Terada, M. Yano, I. Sergeev, M. Mori et al., Oxidative stress inhibits the mitochondrial import of 484

, preproteins and leads to their degradation, Exp Cell Res, vol.263, pp.107-117, 2001.

L. Yang, T. Moss, L. S. Mangala, J. Marini, H. Zhao et al., Metabolic shifts toward glutamine regulate tumor growth, invasion and bioenergetics in ovarian cancer, Mol Syst Biol, vol.10, p.728, 2014.

P. Yao and P. L. Fox, Aminoacyl-tRNA synthetases in medicine and disease, EMBO Mol Med, vol.5, pp.332-343, 2013.

J. Ye, A. Mancuso, X. Tong, P. S. Ward, J. Fan et al., Pyruvate kinase M2 promotes de novo serine synthesis to sustain mTORC1 activity and cell proliferation, Proc Natl Acad Sci, vol.109, pp.6904-6909, 2012.

X. Q. Ye, Q. Li, G. H. Wang, F. F. Sun, G. J. Huang et al., Mitochondrial and energy metabolism-related properties as novel indicators of lung cancer stem cells, Int J Cancer, vol.129, pp.820-831, 2011.

Q. Yu, Y. Geng, and P. Sicinski, Specific protection against breast cancers by cyclin D1 ablation, Nature, vol.411, pp.1017-1021, 2001.

Y. Yuan, Z. X. Gu, X. F. Tao, and L. Sy, Computer tomography, magnetic resonance imaging, and positron emission tomography or positron emission tomography/ computer tomography for detection of metastatic lymph nodes in patients with ovarian cancer: a meta-analysis

, Eur J Radiol, vol.81, pp.1002-1006, 2012.

M. Yuneva, N. Zamboni, P. Oefner, R. Sachidanandam, and Y. Lazebnik, Deficiency in glutamine but not glucose induces MYC-dependent apoptosis in human cells, J Cell Biol, vol.178, pp.93-105, 2007.

D. Zhang, Y. Wang, Z. Shi, J. Liu, P. Sun et al., Metabolic reprogramming of cancer-associated fibroblasts by IDH3alpha downregulation, Cell Rep, vol.10, pp.1335-1348, 2015.

Y. Zhang, Y. Ba, C. Liu, G. Sun, L. Ding et al., PGC-1alpha induces apoptosis in human epithelial ovarian cancer cells through a PPARgamma-dependent pathway, Cell Res, vol.17, pp.363-373, 2007.

Y. Zhang, S. Choksi, K. Chen, Y. Pobezinskaya, I. Linnoila et al., ROS play a critical role in the differentiation of alternatively activated macrophages and the occurrence of tumor-associated macrophages, Cell Res, vol.23, pp.898-914, 2013.

X. L. Zu and M. Guppy, Address correspondence to: Dr. Fatima Mechta-Grigoriou Stress and Cancer Laboratory É quipe Labelisée LNCC Institut Curie, Biochem Biophys Res Commun, vol.313, pp.459-465, 2004.

, France E-mail: fatima.mechta-grigoriou@curie.fr Date of first submission to ARS Central, 2016.