Z. M. Younossi, Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes, Hepatology, vol.64, pp.73-84, 2016.

C. Puoti, M. G. Elmo, D. Ceccarelli, and M. Ditrinco, Liver steatosis: The new epidemic of the Third Millennium. Benign liver state or silent killer?, Eur J Intern Med, vol.pii, issue.17, pp.30268-30274, 2017.

A. B. Siegel and A. X. Zhu, Metabolic syndrome and hepatocellular carcinoma: two growing epidemics with a potential link, Cancer, vol.115, pp.5651-5661, 2009.

C. R. Wong, M. H. Nguyen, and J. K. Lim, Hepatocellular carcinoma in patients with non-alcoholic fatty liver disease, World J Gastroenterol, vol.22, pp.8294-8303, 2016.

M. Noureddin and M. E. Rinella, Nonalcoholic Fatty liver disease, diabetes, obesity, and hepatocellular carcinoma, Clin Liver Dis, vol.19, pp.361-379, 2015.

S. Bellentani, The epidemiology of non-alcoholic fatty liver disease, Liver Int, vol.37, issue.1, pp.81-84, 2017.

Z. M. Younossi, The economic and clinical burden of nonalcoholic fatty liver disease in the United States and Europe, Hepatology, vol.64, pp.1577-1586, 2016.

J. J. Heindel, Metabolism disrupting chemicals and metabolic disorders, Reprod Toxicol, vol.68, pp.3-33, 2017.

R. Scientific, , vol.8, 2018.

C. E. Foulds, L. S. Treviño, B. York, and C. L. Walker, Endocrine-disrupting chemicals and fatty liver disease, Nat Rev Endocrinol, vol.13, pp.445-457, 2017.

L. Magueresse-battistoni, B. Labaronne, E. Vidal, H. Naville, and D. , Endocrine disrupting chemicals in mixture and obesity, diabetes and related metabolic disorders, World J Biol Chem, vol.8, pp.108-119, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-01848536

M. Cave, Toxicant-associated steatohepatitis in vinyl chloride workers, Hepatology, vol.51, pp.474-481, 2010.

B. Wahlang, Toxicant-associated steatohepatitis, Toxicol Pathol, vol.41, pp.343-360, 2013.

S. Joshi-barve, I. Kirpich, M. C. Cave, L. S. Marsano, and C. J. Mcclain, Alcoholic, nonalcoholic, and toxicant-associated steatohepatitis: mechanistic similarities and differences, Cell Mol Gastroenterol Hepatol, vol.1, pp.356-367, 2015.

M. A. Robin, Alcohol increases tumor necrosis factor alpha and decreases nuclear factor-kappa? to activate hepatic apoptosis in genetically obese mice, Hepatology, vol.42, pp.1280-1290, 2005.

T. Minato, Binge alcohol consumption aggravates oxidative stress and promotes pathogenesis of NASH from obesity-induced simple steatosis, Mol Med, vol.20, pp.490-502, 2014.

C. Duval, Chronic Exposure to Low Doses of Dioxin Promotes Liver Fibrosis Development in the C57BL/6J Diet-Induced Obesity Mouse Model, Environ Health Perspect, vol.125, pp.428-436, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02196317

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

E. Polycyclic, Scientific Opinion of the Panel on Contaminants in the Food Chain, The EFSA Journal, vol.724, pp.1-114, 2008.

K. Hardonnière, L. Huc, O. Sergent, J. A. Holme, and D. Lagadic-gossmann, Environmental carcinogenesis and pH homeostasis: Not only a matter of dysregulated metabolism, Semin Cancer Biol, vol.43, pp.49-65, 2017.

L. Ortiz, B. Nakamura, X. Li, B. Blumberg, U. Luderer et al., In utero exposure to benzo[a]pyrene increases adiposity and causes hepatic steatosis in female mice, and glutathione deficiency is protective, Toxicol Lett, vol.230, pp.314-321, 2014.

F. Neuschäfer-rube, Arylhydrocarbon receptor-dependent mIndy (Slc13a5) induction as possible contributor to benzo[a] pyrene-induced lipid accumulation in hepatocytes, Toxicology, vol.337, pp.1-9, 2015.

Q. Ba, Effects of benzo[a]pyrene exposure on human hepatocellular carcinoma cell angiogenesis, metastasis, and NF-?B signaling, Environ Health Perspect, vol.123, pp.246-254, 2015.

M. Tian, Association of environmental benzo[a]pyrene exposure and DNA methylation alterations in hepatocellular carcinoma: A Chinese case-control study, Sci Total Environ, vol.541, pp.1243-1252, 2016.

Y. Su, Interaction of benzo[a]pyrene with other risk factors in hepatocellular carcinoma: a case-control study in Xiamen, China, Ann Epidemiol, vol.24, pp.98-103, 2014.

A. Collin, Cooperative interaction of benzo[a]pyrene and ethanol on plasma membrane remodeling is responsible for enhanced oxidative stress and cell death in primary rat hepatocytes, Free Radic Biol Med, vol.72, pp.11-22, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01018147

T. B. Andersson, K. P. Kanebratt, and J. G. Kenna, The HepaRG cell line: a unique in vitro tool for understanding drug metabolism and toxicology in human, Expert Opin Drug Metab Toxicol, vol.8, pp.909-920, 2012.

C. Biagini, Cytochrome P450 expression-induction profile and chemically mediated alterations of the WIF-B9 cell line, Biol Cell, vol.98, pp.23-32, 2006.

B. L. Mcvicker, K. Rasineni, D. J. Tuma, M. A. Mcniven, and C. A. Casey, Lipid droplet accumulation and impaired fat efflux in polarized hepatic cells: consequences of ethanol metabolism, Int J Hepatol, vol.978136, 2012.

W. Goessling and K. C. Sadler, Zebrafish: an important tool for liver disease research, Gastroenterology, vol.149, pp.1361-1377, 2015.
DOI : 10.1053/j.gastro.2015.08.034

URL : http://europepmc.org/articles/pmc4762709?pdf=render

Y. Asaoka, S. Terai, I. Sakaida, and H. Nishina, The expanding role of fish models in understanding non-alcoholic fatty liver disease, Dis Model Mech, vol.6, pp.905-914, 2013.

D. H. Pham, C. Zhang, and C. Yin, Using zebrafish to model liver diseases-Where do we stand?, Curr Pathobiol Rep, vol.5, pp.207-221, 2017.
DOI : 10.1007/s40139-017-0141-y

C. Aninat, Expression of cytochromes P450, conjugating enzymes and nuclear receptors in human hepatoma HepaRG cells, Drug Metab Dispos, vol.34, pp.75-83, 2006.
DOI : 10.1124/dmd.105.006759

URL : https://hal.archives-ouvertes.fr/hal-00702085

A. Michaut, A cellular model to study drug-induced liver injury in nonalcoholic fatty liver disease: Application to acetaminophen, Toxicol Appl Pharmacol, vol.292, pp.40-55, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01255826

C. Decaens, P. Rodriguez, C. Bouchaud, and D. Cassio, Establishment of hepatic cell polarity in the rat hepatoma-human fibroblast hybrid WIF-B9. A biphasic phenomenon going from a simple epithelial polarized phenotype to an hepatic polarized one, J Cell Sci, vol.109, pp.1623-1635, 1996.

N. Podechard, Zebrafish larva as a reliable model for in vivo assessment of membrane remodeling involvement in the hepatotoxicity of chemical agents, J Appl Toxicol, vol.37, pp.732-746, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01526425

J. P. Otis and S. A. Farber, High-fat Feeding Paradigm for Larval Zebrafish: Feeding, Live Imaging, and Quantification of Food Intake, J Vis Exp, issue.116, 2016.

E. Marza, Developmental expression and nutritional regulation of a zebrafish gene homologous to mammalian microsomal triglyceride transfer protein large subunit, Dev Dyn, vol.232, pp.506-518, 2005.

M. D. Burke, Ethoxy-, pentoxy-and benzyloxyphenoxazones and homologues: a series of substrates to distinguish between different induced cytochromes P-450, Biochem Pharmacol, vol.34, pp.3337-3345, 1985.
DOI : 10.1016/0006-2952(85)90355-7

T. Shimada, Selectivity of polycyclic inhibitors for human cytochrome P450s 1A1, 1A2, and 1B1, Chem Res Toxicol, vol.11, pp.1048-1056, 1998.
DOI : 10.1021/tx980090+

A. C. Capomacchia, V. Kumar, and R. N. Jennings, Internal hydrogen bonding in benzo[a]pyrene diol nd diol epoxide metabolites, J Chem Soc Perkin Trans, vol.2, pp.937-941, 1989.
DOI : 10.1039/p29890000937

W. Lee, H. Shin, J. Hong, H. Pyo, and Y. Kim, Studies on the analysis of benzo (a) pyrene and its metabolites on biological samples by using high performance liquid chromatography/fluorescence detection and gas chromatography/mass spectrometry, Bull Korean Chem So, vol.24, pp.559-565, 2003.

A. Bednàrikovà, B. Sklàrsovà, E. Kolek, M. Polovka, and P. Simko, New rapid HPLC method for separation and determination of benzo[a]pyrene hydroxyderivatives, Polycyclic Aromatic Compounds, vol.31, pp.350-369, 2011.

M. Kang, J. Kim, H. T. An, and J. Ko, Human leucine zipper protein promotes hepatic steatosis via induction of apolipoprotein A-IV, FASEB J, vol.31, pp.2548-2561, 2017.
DOI : 10.1096/fj.201601227r

N. Chalasani, Hepatic cytochrome P450 2E1 activity in nondiabetic patients with nonalcoholic steatohepatitis, Hepatology, vol.37, pp.544-550, 2003.
DOI : 10.1053/jhep.2003.50095

M. G. Emery, CYP2E1 activity before and after weight loss in morbidly obese subjects with nonalcoholic fatty liver disease, Hepatology, vol.38, pp.428-435, 2003.
DOI : 10.1053/jhep.2003.50342

URL : http://onlinelibrary.wiley.com/doi/10.1053/jhep.2003.50342/pdf

J. Aubert, K. Begriche, L. Knockaert, M. A. Robin, and B. Fromenty, Increased expression of cytochrome P450 2E1 in nonalcoholic fatty liver disease: mechanisms and pathophysiological role, Clin Res Hepatol Gastroenterol, vol.35, pp.630-637, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00739365

K. Pernelle, Automated detection of hepatotoxic compounds in human hepatocytes using HepaRG cells and image-based analysis of mitochondrial dysfunction with JC-1 dye, Toxicol Appl Pharmacol, vol.254, pp.256-266, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00740177

H. H. Gerets, Characterization of primary human hepatocytes, HepG2 cells, and HepaRG cells at the mRNA level and CYP activity in response to inducers and their predictivity for the detection of human hepatotoxins, Cell Biol Toxicol, vol.28, pp.69-87, 2012.

H. Al-attrache, Differential sensitivity of metabolically competent and non-competent HepaRG cells to apoptosis induced by diclofenac combined or not with TNF-?, Toxicol Lett, vol.258, pp.71-86, 2016.
DOI : 10.1016/j.toxlet.2016.06.008

URL : https://hal.archives-ouvertes.fr/hal-01372855

R. Scientific, , vol.8, 2018.

O. Sergent, Role for membrane fluidity in ethanol-induced oxidative stress of primary rat hepatocytes, J Pharmacol Exp Ther, vol.313, pp.104-111, 2005.

P. Nourissat, Ethanol induces oxidative stress in primary rat hepatocytes through the early involvement of lipid raft clustering, Hepatology, vol.47, pp.59-70, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00696177

T. Inagaki, Research perspectives on the regulation and physiological functions of FGF21 and its association with NAFLD. Front Endocrinol (Lausanne) 6, vol.147, 2015.

M. S. Neal, J. Zhu, and W. G. Foster, Quantification of benzo[a]pyrene and other PAHs in the serum and follicular fluid of smokers versus non-smokers, Reprod Toxicol, vol.25, pp.100-106, 2008.

W. Dai, High fat plus high cholesterol diet lead to hepatic steatosis in zebrafish larvae: a novel model for screening anti-hepatic steatosis drugs, Nutr Metab (Lond), vol.12, 2015.

M. J. Den-broeder, V. A. Kopylova, L. M. Kamminga, and J. Legler, Zebrafish as a model to study the role of peroxisome proliferatingactivated receptors in adipogenesis and obesity, PPAR Res, vol.358029, 2015.

M. J. Passeri, A. Cinaroglu, C. Gao, and K. C. Sadler, Hepatic steatosis in response to acute alcohol exposure in zebrafish requires sterol regulatory element binding protein activation, Hepatology, vol.49, pp.443-452, 2009.

A. C. Schneider, Chronic exposure to ethanol causes steatosis and inflammation in zebrafish liver, World J Hepatol, vol.9, pp.418-426, 2017.

E. J. Flynn, C. M. Trent, and J. F. Rawls, Ontogeny and nutritional control of adipogenesis in zebrafish (Danio rerio), J Lipid Res, vol.50, pp.1641-52, 2009.

J. E. Minchin and J. F. Rawls, A classification system for zebrafish adipose tissues, Dis Model Mech, vol.10, pp.797-809, 2017.

J. M. Petit, Apolipoprotein-AII concentrations are associated with liver steatosis in patients with chronic hepatitis C, Dig Dis Sci, vol.52, pp.3431-3434, 2007.

R. F. Mortensen, C-reactive protein, inflammation, and innate immunity, Immunol Res, vol.24, pp.163-176, 2001.
DOI : 10.1385/ir:24:2:163

M. ?ulc, The impact of individual cytochrome P450 enzymes on oxidative metabolism of benzo[a]pyrene in human livers, Environ Mol Mutagen, vol.57, pp.229-235, 2016.

C. Liu, Acute exposure to Tris(1,3-dichloro-2-propyl) Phosphate (TDCIPP) causes hepatic inflammation and leads to hepatotoxicity in, Zebrafish. Sci Rep, vol.6, 2016.

I. Leclercq, Y. Horsmans, J. P. Desager, N. Delzenne, and A. P. Geubel, Reduction in hepatic cytochrome P-450 is correlated to the degree of liver fat content in animal models of steatosis in the absence of inflammation, J Hepatol, vol.28, pp.410-416, 1998.

M. T. Donato, Potential impact of steatosis on cytochrome P450 enzymes of human hepatocytes isolated from fatty liver grafts, Drug Metab Dispos, vol.34, pp.1556-1562, 2006.

C. D. Fisher, Hepatic cytochrome P450 enzyme alterations in humans with progressive stages of nonalcoholic fatty liver disease, Drug Metab Dispos, vol.37, pp.2087-2094, 2009.

M. D. Merrell and N. J. Cherrington, Drug metabolism alterations in nonalcoholic fatty liver disease, Drug Metab Rev, vol.43, pp.317-334, 2011.

A. Naik, A. Beli?, U. M. Zanger, and D. Rozman, Molecular interactions between NAFLD and xenobiotic metabolism, Front Genet, vol.4, 2013.
DOI : 10.3389/fgene.2013.00002

URL : https://www.frontiersin.org/articles/10.3389/fgene.2013.00002/pdf

E. Cobbina and F. Akhlaghi, Non-alcoholic fatty liver disease (NAFLD)-pathogenesis, classification, and effect on drug metabolizing enzymes and transporters, Drug Metab Rev, vol.17, pp.1-15, 2017.
DOI : 10.1080/03602532.2017.1293683

URL : http://europepmc.org/articles/pmc5576152?pdf=render

J. A. Cichocki, Impact of nonalcoholic fatty liver disease on toxicokinetics of tetrachloroethylene in Mice, J Pharmacol Exp Ther, vol.361, pp.17-28, 2017.

Y. Morel, I. De-waziers, and R. Barouki, A repressive cross-regulation between catalytic and promoter activities of the CYP1A1 and CYP2E1 genes: role of H(2)O(2), Mol Pharmacol, vol.57, pp.1158-1164, 2000.

Y. Morel, N. Mermod, and R. Barouki, An autoregulatory loop controlling CYP1A1 gene expression: role of H(2)O(2) and NFI, Mol Cell Biol, vol.19, pp.6825-6832, 1999.
DOI : 10.1128/mcb.19.10.6825

URL : http://mcb.asm.org/content/19/10/6825.full.pdf

R. Barouki and Y. Morel, Repression of cytochrome P450 1A1 gene expression by oxidative stress: mechanisms and biological implications, Biochem Pharmacol, vol.615, pp.511-516, 2001.
DOI : 10.1016/s0006-2952(00)00543-8

Z. Abdel-razzak, Cytokines down-regulate expression of major cytochrome P-450 enzymes in adult human hepatocytes in primary culture, Mol Pharmacol, vol.44, pp.707-715, 1993.

Y. Kusunoki, Hepatic early inflammation induces downregulation of hepatic cytochrome P450 expression and metabolic activity in the dextran sulfate sodium-induced murine colitis, Eur J Pharm Sci, vol.54, pp.17-27, 2014.

M. Zhou, S. R. Maitra, and P. Wang, The potential role of transcription factor aryl hydrocarbon receptor in downregulation of hepatic cytochrome P-450 during sepsis, Int J Mol Med, vol.21, pp.423-428, 2008.

T. M. Penning, Human aldo-keto reductases and the metabolic activation of polycyclic aromatic hydrocarbons, Chem Res Toxicol, vol.27, pp.1901-1917, 2014.
DOI : 10.1021/tx500298n

URL : https://doi.org/10.1021/tx500298n

M. Stiborová, Cytochrome b5 and epoxide hydrolase contribute to benzo[a]pyrene-DNA adduct formation catalyzed by cytochrome P450 1A1 under low NADPH:P450 oxidoreductase conditions, Toxicology, vol.318, pp.1-12, 2014.

R. N. Hardwick, C. D. Fisher, M. J. Canet, A. D. Lake, and N. J. Cherrington, Diversity in antioxidant response enzymes in progressive stages of human nonalcoholic fatty liver disease, Drug Metab Dispos, vol.38, pp.2293-2301, 2010.

M. E. Kushman, Expression of human glutathione S-transferase P1 confers resistance to benzo[a]pyrene or benzo[a]pyrene7,8-dihydrodiol mutagenesis, macromolecular alkylation and formation of stable N2-Gua-BPDE adducts in stably transfected V79MZ cells co-expressing hCYP1A1, Carcinogenesis, vol.28, pp.207-214, 2007.

Q. Shi, L. Maas, C. Veith, F. J. Van-schooten, and R. W. Godschalk, Acidic cellular microenvironment modifies carcinogen-induced DNA damage and repair, Arch Toxicol, vol.91, pp.2425-2441, 2017.
DOI : 10.1007/s00204-016-1907-4

URL : https://link.springer.com/content/pdf/10.1007%2Fs00204-016-1907-4.pdf