V. Agier, P. Oliviero, J. Lainé, L. Hermitte-stead, C. Girard et al., Defective mitochondrial fusion, altered respiratory function, and distorted cristae structure in skin fibroblasts with heterozygous OPA1 mutations, Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease, vol.1822, issue.10, pp.1570-1580, 2012.
DOI : 10.1016/j.bbadis.2012.07.002

URL : https://doi.org/10.1016/j.bbadis.2012.07.002

S. Anthérieu, C. Chesné, R. Li, C. Guguen-guillouzo, and A. Guillouzo, Optimization of the HepaRG cell model for drug metabolism and toxicity studies, Toxicology in Vitro, vol.26, issue.8, pp.1278-1285, 2012.
DOI : 10.1016/j.tiv.2012.05.008

S. Anthérieu, A. Rogue, B. Fromenty, A. Guillouzo, and R. , Induction of vesicular steatosis by amiodarone and tetracycline is associated with up-regulation of lipogenic genes in heparg cells, Hepatology, vol.96, issue.Suppl., pp.1895-1905, 2011.
DOI : 10.1093/toxsci/kfl184

J. Aubert, K. Begriche, L. Knockaert, M. Robin, and B. Fromenty, Increased expression of cytochrome P450 2E1 in nonalcoholic fatty liver disease: Mechanisms and pathophysiological role, Clinics and Research in Hepatology and Gastroenterology, vol.35, issue.10, pp.630-637, 2011.
DOI : 10.1016/j.clinre.2011.04.015

URL : https://hal.archives-ouvertes.fr/hal-00739365

K. Begriche, A. Igoudjil, D. Pessayre, and B. Fromenty, Mitochondrial dysfunction in NASH: Causes, consequences and possible means to prevent it, Mitochondrion, vol.6, issue.1, pp.1-28, 2006.
DOI : 10.1016/j.mito.2005.10.004

K. Begriche, J. Massart, R. M. Bonnet, F. Fromenty, and B. , Mitochondrial adaptations and dysfunctions in nonalcoholic fatty liver disease, Hepatology, vol.6, issue.4, pp.1497-1507, 2013.
DOI : 10.1016/j.mito.2005.10.004

URL : https://hal.archives-ouvertes.fr/inserm-00814080

K. Begriche, J. Massart, M. Robin, A. Borgne-sanchez, and B. Fromenty, Drug-induced toxicity on mitochondria and lipid metabolism: Mechanistic diversity and deleterious consequences for the liver, Journal of Hepatology, vol.54, issue.4, pp.773-794, 2011.
DOI : 10.1016/j.jhep.2010.11.006

URL : https://hal.archives-ouvertes.fr/hal-00739371

D. Bishop, C. Granata, and N. Eynon, Can we optimise the exercise training prescription to maximise improvements in mitochondria function and content?, Biochimica et Biophysica Acta (BBA) - General Subjects, vol.1840, issue.4, pp.1266-1275, 2014.
DOI : 10.1016/j.bbagen.2013.10.012

URL : http://vuir.vu.edu.au/23768/1/Can%20we%20optimise%20ex-training%20to%20promote%20REVISED%20TEXT%20UNMARKED.pdf

S. Bucher, P. Jalili, L. Guillou, D. Begriche, K. Rondel et al., Bisphenol a induces steatosis in HepaRG cells using a model of perinatal exposure, Environmental Toxicology, vol.9, issue.471, pp.1024-1036, 2017.
DOI : 10.2217/14622416.9.11.1695

URL : https://hal.archives-ouvertes.fr/hal-01470709

B. Cohen, Pharmacologic effects on mitochondrial function, Developmental Disabilities Research Reviews, vol.2, issue.Suppl 2, pp.189-199, 2010.
DOI : 10.1212/01.WNL.0000140494.58732.83

D. Bus, L. Depuydt, P. Libbrecht, L. Vandekerckhove, L. Nollet et al., Severe Drug-induced Liver Injury Associated with Prolonged Use of Linezolid, Journal of Medical Toxicology, vol.30, issue.8, pp.322-326, 2010.
DOI : 10.1007/s13181-010-0047-0

, This article has not been copyedited and formatted. The final version may differ from this version

D. Vriese, A. Coster, R. Smet, J. Seneca, S. Lovering et al., Linezolid-Induced Inhibition of Mitochondrial Protein Synthesis, Clinical Infectious Diseases, vol.39, issue.7, pp.1111-1117, 2006.
DOI : 10.1086/423841

D. Deschamps, V. Debeco, C. Fisch, B. Fromenty, A. Guillouzo et al., Inhibition by perhexiline of oxidative phosphorylation and the ??-oxidation of fatty acids: Possible role in pseudoalcoholic liver lesions, Hepatology, vol.46, issue.4, pp.948-961, 1994.
DOI : 10.1016/0005-2760(72)90179-8

J. Dykens, J. Jamieson, L. Marroquin, S. Nadanaciva, J. Xu et al., In Vitro Assessment of Mitochondrial Dysfunction and Cytotoxicity of Nefazodone, Trazodone, and Buspirone, Toxicological Sciences, vol.1, issue.7, pp.335-345, 2008.
DOI : 10.1124/dmd.31.8.1035

J. Eakins, C. Bauch, H. Woodhouse, B. Park, S. Bevan et al., A combined in vitro approach to improve the prediction of mitochondrial toxicants, Toxicology in Vitro, vol.34, pp.161-170, 2016.
DOI : 10.1016/j.tiv.2016.03.016

B. Fromenty, Drug-induced liver injury in obesity, Journal of Hepatology, vol.58, issue.4, pp.824-826, 2013.
DOI : 10.1016/j.jhep.2012.12.018

URL : https://hal.archives-ouvertes.fr/hal-00865967

B. Fromenty, C. Fisch, A. Berson, P. Letteron, D. Larrey et al., Dual effect of amiodarone on mitochondrial respiration. Initial protonophoric uncoupling effect followed by inhibition of the respiratory chain at the levels of complex I and complex II, J Pharmacol Exp Ther, vol.255, pp.1377-1384, 1990.

I. García-ruiz, C. Rodríguez-juan, T. Díaz-sanjuán, and M. Martínez,

J. Herruzo, Effects of rosiglitazone on the liver histology and mitochondrial function in ob/ob mice, Hepatology, vol.46, pp.414-423, 2007.

G. Garrabou, À. Soriano, T. Pinós, J. Casanova-mollà, D. Pacheu-grau et al., ABSTRACT, Antimicrobial Agents and Chemotherapy, vol.61, issue.9, pp.542-559, 2017.
DOI : 10.1128/AAC.00542-17

O. Guillery, F. Malka, P. Frachon, D. Milea, M. Rojo et al., Modulation of mitochondrial morphology by bioenergetics defects in primary human fibroblasts, Neuromuscular Disorders, vol.18, issue.4, pp.319-330, 2008.
DOI : 10.1016/j.nmd.2007.12.008

S. Hobbie, S. Akshay, S. Kalapala, C. Bruell, D. Shcherbakov et al., Genetic analysis of interactions with eukaryotic rRNA identify the mitoribosome as target in aminoglycoside ototoxicity, Proceedings of the National Academy of Sciences, vol.76, issue.4, pp.20888-20893, 2008.
DOI : 10.1073/pnas.76.4.1760

T. Horie, K. Ono, K. Nagao, H. Nishi, M. Kinoshita et al., Oxidative stress induces GLUT4 translocation by activation of PI3-K/Akt and dual AMPK kinase in cardiac myocytes, Journal of Cellular Physiology, vol.97, issue.3, pp.733-742, 2008.
DOI : 10.1042/bj2810809

, This article has not been copyedited and formatted. The final version may differ from this version

H. Hosomi, T. Fukami, A. Iwamura, M. Nakajima, and T. Yokoi, Development of a Highly Sensitive Cytotoxicity Assay System for CYP3A4-Mediated Metabolic Activation, Drug Metabolism and Disposition, vol.39, issue.8, pp.1388-1395, 2011.
DOI : 10.1124/dmd.110.037077

M. Hull and J. Montaner, Ritonavir-boosted protease inhibitors in HIV therapy, Annals of Medicine, vol.25, issue.4, pp.375-388, 2011.
DOI : 10.1086/649521

W. Isley, Hepatotoxicity of thiazolidinediones, Expert Opinion on Drug Safety, vol.38, issue.6, pp.581-586, 2003.
DOI : 10.1053/jhep.2003.50229

L. Kamalian, A. Chadwick, M. Bayliss, N. French, M. Monshouwer et al., The utility of HepG2 cells to identify direct mitochondrial dysfunction in the absence of cell death, Toxicology in Vitro, vol.29, issue.4, pp.732-740, 2015.
DOI : 10.1016/j.tiv.2015.02.011

G. Kumar, A. Rodrigues, A. Buko, and J. Denissen, Cytochrome P450-mediated metabolism of the HIV-1 protease inhibitor ritonavir (ABT-538) in human liver microsomes, 1996.

, J Pharmacol Exp Ther, vol.277, pp.423-431

G. Labbe, D. Pessayre, and B. Fromenty, Drug-induced liver injury through mitochondrial dysfunction: mechanisms and detection during preclinical safety studies, Fundamental & Clinical Pharmacology, vol.287, issue.Suppl 2, pp.335-353, 2008.
DOI : 10.1111/j.1768-322X.1987.tb00524.x

F. Li, J. Lu, and M. X. , Metabolomic Screening and Identification of the Bioactivation Pathways of Ritonavir, Chemical Research in Toxicology, vol.24, issue.12, pp.2109-2114, 2011.
DOI : 10.1021/tx2004147

A. Macgowan, Pharmacokinetic and pharmacodynamic profile of linezolid in healthy volunteers and patients with Gram-positive infections, Journal of Antimicrobial Chemotherapy, vol.51, issue.90002, pp.17-25, 2003.
DOI : 10.1093/jac/dkg248

L. Marroquin, J. Hynes, J. Dykens, J. Jamieson, and W. Y. , Circumventing the Crabtree Effect: Replacing Media Glucose with Galactose Increases Susceptibility of HepG2 Cells to Mitochondrial Toxicants, Toxicological Sciences, vol.97, issue.2, pp.539-547, 2007.
DOI : 10.1093/toxsci/kfm052

L. Marroquin, R. Swiss, and W. Y. , Identifying Compounds that Induce Opening of the Mitochondrial Permeability Transition Pore in Isolated Rat Liver Mitochondria, Curr Protoc Toxicol, vol.39, pp.25-29, 2014.
DOI : 10.1016/j.freeradbiomed.2005.05.013

J. Massarella, L. Nazareno, S. Passe, and M. B. , The effect of probenecid on the pharmacokinetics of zalcitabine in HIV-positive patients, Pharmaceutical Research, vol.13, issue.3, pp.449-452, 1996.
DOI : 10.1023/A:1016009029536

J. Massart, K. Begriche, C. Moreau, and B. Fromenty, Role of nonalcoholic fatty liver disease as risk factor for drug-induced hepatotoxicity, J Clin Transl Res, vol.3, pp.212-232, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01580159

Y. Masubuchi, Metabolic and Non-Metabolic Factors Determining Troglitazone Hepatotoxicity: A Review, Drug Metabolism and Pharmacokinetics, vol.21, issue.5, pp.347-356, 2006.
DOI : 10.2133/dmpk.21.347

A. Michaut, L. Guillou, D. Moreau, C. Bucher, S. Mcgill et al., A cellular model to study drug-induced liver This article has not been copyedited and formatted. The final version may differ from this version, 2016.

, injury in nonalcoholic fatty liver disease: Application to acetaminophen, Toxicol Appl Pharmacol, vol.292, pp.40-55

A. Michaut, C. Moreau, R. M. Fromenty, and B. , Acetaminophen-induced liver injury in obesity and nonalcoholic fatty liver disease, Liver International, vol.36, issue.Suppl 2, pp.171-179, 2014.
DOI : 10.1016/j.hepres.2006.07.009

URL : https://hal.archives-ouvertes.fr/hal-01128195

D. Morales-alamo and J. Calbet, AMPK signaling in skeletal muscle during exercise: Role of reactive oxygen and nitrogen species, Free Radical Biology and Medicine, vol.98, pp.68-77, 2016.
DOI : 10.1016/j.freeradbiomed.2016.01.012

N. Moullan, L. Mouchiroud, X. Wang, D. Ryu, E. Williams et al., Tetracyclines Disturb Mitochondrial Function across Eukaryotic Models: A Call for Caution in Biomedical Research, Cell Reports, vol.10, issue.10, pp.1681-1691, 2015.
DOI : 10.1016/j.celrep.2015.02.034

S. Nadanaciva, J. Dykens, A. Bernal, R. Capaldi, and W. Y. , Mitochondrial impairment by PPAR agonists and statins identified via immunocaptured OXPHOS complex activities and respiration, Toxicology and Applied Pharmacology, vol.223, issue.3, pp.277-287, 2007.
DOI : 10.1016/j.taap.2007.06.003

F. Paech, J. Bouitbir, and S. Krähenbühl, Hepatocellular Toxicity Associated with Tyrosine Kinase Inhibitors: Mitochondrial Damage and Inhibition of Glycolysis, Frontiers in Pharmacology, vol.81, p.367, 2017.
DOI : 10.1016/j.bcp.2010.11.002

URL : https://www.frontiersin.org/articles/10.3389/fphar.2017.00367/pdf

L. Peyta, K. Jarnouen, M. Pinault, C. Guimaraes, P. De-barros et al., Reduced cardiolipin content decreases respiratory chain capacities and increases ATP synthesis yield in the human HepaRG cells, Biochimica et Biophysica Acta (BBA) - Bioenergetics, vol.1857, issue.4, pp.443-53, 2016.
DOI : 10.1016/j.bbabio.2016.01.002

URL : https://hal.archives-ouvertes.fr/hal-01259219

C. Ploumi, I. Daskalaki, and N. Tavernarakis, Mitochondrial biogenesis and clearance: a balancing act, The FEBS Journal, vol.15, issue.147, pp.183-195, 2017.
DOI : 10.1093/hmg/ddl006

URL : http://onlinelibrary.wiley.com/doi/10.1111/febs.13820/pdf

M. Porceddu, N. Buron, C. Roussel, G. Labbe, B. Fromenty et al., Prediction of Liver Injury Induced by Chemicals in Human With a Multiparametric Assay on Isolated Mouse Liver Mitochondria, Toxicological Sciences, vol.1, issue.Suppl. 2, pp.332-345, 2012.
DOI : 10.1038/nprot.2006.351

URL : https://hal.archives-ouvertes.fr/hal-00866136

M. Porceddu, N. Buron, R. P. Fromenty, B. Sanchez, and A. , In Vitro Assessment of Mitochondrial Toxicity to Predict Drug-Induced Liver Injury, Drug-Induced Liver Toxicity, pp.283-300, 2018.
DOI : 10.1016/0006-2952(89)90580-7

URL : https://hal.archives-ouvertes.fr/hal-01777715

P. Quirós, M. Prado, N. Zamboni, D. Amico, D. Williams et al., Multi-omics analysis identifies ATF4 as a key regulator of the mitochondrial stress response in mammals, The Journal of Cell Biology, vol.216, issue.7, pp.2027-2045, 2017.
DOI : 10.1016/j.molcel.2016.02.011

R. Rossignol, B. Faustin, C. Rocher, M. Malgat, J. Mazat et al., Mitochondrial threshold effects, Biochemical Journal, vol.370, issue.3, pp.751-762, 2003.
DOI : 10.1042/bj20021594

R. Rossignol, M. Malgat, J. Mazat, and T. Letellier, Threshold Effect and Tissue Specificity, Journal of Biological Chemistry, vol.90, issue.47, pp.33426-33432, 1999.
DOI : 10.1016/0014-5793(88)81347-4

URL : http://www.jbc.org/content/274/47/33426.full.pdf

, This article has not been copyedited and formatted. The final version may differ from this version

R. Scarpulla, R. Vega, and D. Kelly, Transcriptional integration of mitochondrial biogenesis, Trends in Endocrinology & Metabolism, vol.23, issue.9, pp.459-466, 2012.
DOI : 10.1016/j.tem.2012.06.006

E. Schon and B. Fromenty, Mitochondria in Liver Disease, Mitochondria in Liver Disease pp 283?314, 2015.

U. Spengler, M. Lichterfeld, and J. Rockstroh, Antiretroviral drug toxicity ??? a challenge for the hepatologist?, Journal of Hepatology, vol.36, issue.2, pp.283-294, 2002.
DOI : 10.1016/S0168-8278(01)00311-7

N. Sunny, F. Bril, and K. Cusi, Mitochondrial Adaptation in Nonalcoholic Fatty Liver Disease: Novel Mechanisms and Treatment Strategies, Trends in Endocrinology & Metabolism, vol.28, issue.4, pp.250-260, 2017.
DOI : 10.1016/j.tem.2016.11.006

R. Vega, J. Horton, and D. Kelly, Maintaining Ancient Organelles: Mitochondrial Biogenesis and Maturation, Circulation Research, vol.116, issue.11, pp.1820-1834, 2015.
DOI : 10.1161/CIRCRESAHA.116.305420

J. Villena, New insights into PGC-1 coactivators: redefining their role in the regulation of mitochondrial function and beyond, The FEBS Journal, vol.277, issue.4, pp.647-672, 2015.
DOI : 10.1074/jbc.M210262200

U. Walker, J. Bäuerle, M. Laguno, J. Murillas, S. Mauss et al., Depletion of mitochondrial DNA in liver under antiretroviral therapy with didanosine, stavudine, or zalcitabine, Hepatology, vol.31, issue.Suppl. 4, pp.311-317, 2004.
DOI : 10.1097/00126334-200211010-00005

D. Wallace, W. Fan, and V. Procaccio, Mitochondrial Energetics and Therapeutics, Annual Review of Pathology: Mechanisms of Disease, vol.5, issue.1, pp.297-348, 2010.
DOI : 10.1146/annurev.pathol.4.110807.092314

Y. Will and J. Dykens, Mitochondrial toxicity assessment in industry ??? a decade of technology development and insight, Expert Opinion on Drug Metabolism & Toxicology, vol.138, issue.8, pp.1061-1067, 2014.
DOI : 10.3109/1354750X.2014.897757

S. Woolsey, S. Mansell, R. Kim, R. Tirona, and M. Beaton, , 2015.