J. M. Maglich, D. J. Parks, L. B. Moore, J. L. Collins, B. Goodwin et al., Identification of a Novel Human Constitutive Androstane Receptor (CAR) Agonist and Its Use in the Identification of CAR Target Genes, J. Biol. Chem, vol.278, pp.17277-17283, 2003.

Y. Yamamoto, T. Kawamoto, and M. Negishi, The role of the nuclear receptor CAR as a coordinate regulator of hepatic gene expression in defense against chemical toxicity, Arch. Biochem. Biophys, vol.409, pp.207-218, 2003.

S. Saini, J. Sonoda, L. Xu, D. Toma, H. Uppal et al., A Novel Constitutive Androstane Receptor-Mediated and CYP3A-Independent Pathway of Bile Acid Detoxification, Mol. Pharmacol, vol.65, pp.292-300, 2004.

W. Huang, J. Zhang, S. S. Chua, M. Qatanani, Y. Han et al., Induction of bilirubin clearance by the constitutive androstane receptor (CAR), Proc. Natl. Acad

. U. Sci, , vol.100, pp.4156-61, 2003.

B. M. Forman, I. Tzameli, H. S. Choi, J. Chen, D. Simha et al., Androstane metabolites bind to and deactivate the nuclear receptor CAR-beta, Nature, vol.395, pp.612-617, 1998.

T. Kawamoto, S. Kakizaki, K. Yoshinari, and M. Negishi, Estrogen activation of the nuclear orphan receptor CAR (constitutive active receptor) in induction of the mouse Cyp2b10 gene, Mol. Endocrinol, vol.14, pp.1897-905, 2000.

L. B. Moore, D. J. Parks, S. A. Jones, R. K. Bledsoe, T. G. Consler et al., Orphan nuclear receptors constitutive androstane receptor and pregnane X receptor share xenobiotic and steroid ligands, J. Biol. Chem, vol.275, pp.15122-15129, 2000.

J. Sugatani, H. Kojima, A. Ueda, S. Kakizaki, K. Yoshinari et al., The phenobarbital response enhancer module in the human bilirubin UDPglucuronosyltransferase UGT1A1 gene and regulation by the nuclear receptor CAR

, Hepatology, vol.33, pp.1232-1240, 2001.

K. Swales, M. Negishi, and . Car, Driving into the Future, Mol. Endocrinol, vol.18, pp.1589-1598, 2004.

J. M. Maglich, J. Watson, P. J. Mcmillen, B. Goodwin, T. M. Willson et al., The nuclear receptor CAR is a regulator of thyroid hormone metabolism during caloric restriction

, J. Biol. Chem, vol.279, pp.19832-19840, 2004.

X. Ding, K. Lichti, I. Kim, F. J. Gonzalez, and J. L. Staudinger, Regulation of constitutive androstane receptor and its target genes by fasting, cAMP, hepatocyte nuclear factor alpha, and the coactivator peroxisome proliferator-activated receptor gamma coactivator-1alpha, J. Biol. Chem, vol.281, pp.26540-51, 2006.

M. Qatanani, J. Zhang, and D. D. Moore, Role of the Constitutive Androstane Receptor in Xenobiotic-Induced Thyroid Hormone Metabolism, Endocrinology, vol.146, pp.995-1002, 2005.

J. Gao, J. He, Y. Zhai, T. Wada, and W. Xie, The constitutive androstane receptor is an antiobesity nuclear receptor that improves insulin sensitivity, J. Biol. Chem, vol.284, pp.25984-92, 2009.

B. Dong, P. K. Saha, W. Huang, W. Chen, L. A. Abu-elheiga et al., Activation of nuclear receptor CAR ameliorates diabetes and fatty liver disease, Proc. Natl. Acad

, Sci, vol.106, pp.18831-18836, 2009.

A. A. Yarushkin, E. M. Kachaylo, and P. Vo, The constitutive androstane receptor activator 4-[(4R,6R)-4,6-diphenyl-1,3-dioxan-2-yl]-N,N-dimethylaniline inhibits the gluconeogenic genes PEPCK and G6Pase through the suppression of HNF4? and FOXO1 transcriptional activity, Br. J. Pharmacol, vol.168, pp.1923-1955, 2013.

J. Gao, J. Yan, M. Xu, S. Ren, and W. Xie, CAR Suppresses Hepatic Gluconeogenesis by Facilitating the Ubiquitination and Degradation of PGC1?, Mol. Endocrinol, vol.29, pp.1558-1570, 2015.

J. Miao, S. Fang, Y. Bae, and J. K. Kemper, Functional inhibitory cross-talk between constitutive androstane receptor and hepatic nuclear factor-4 in hepatic lipid/glucose metabolism is mediated by competition for binding to the DR1 motif and to the common coactivators, GRIP-1 and PGC-1alpha, J. Biol. Chem, vol.281, pp.14537-14583, 2006.

S. Kodama, C. Koike, M. Negishi, and Y. Yamamoto, Nuclear receptors CAR and PXR cross talk with FOXO1 to regulate genes that encode drug-metabolizing and gluconeogenic enzymes, Mol. Cell. Biol, vol.24, pp.7931-7971, 2004.

A. Ueda, H. K. Hamadeh, H. K. Webb, Y. Yamamoto, T. Sueyoshi et al., Diverse roles of the nuclear orphan receptor CAR in regulating hepatic genes in response to phenobarbital, Mol. Pharmacol, vol.61, pp.1-6, 2002.

A. Kassam, C. J. Winrow, F. Fernandez-rachubinski, J. P. Capone, and R. A. Rachubinski, The peroxisome proliferator response element of the gene encoding the peroxisomal betaoxidation enzyme enoyl-CoA hydratase/3-hydroxyacyl-CoA dehydrogenase is a target for constitutive androstane receptor beta/9-cis-retinoic acid receptor-mediated transactivation, J. Biol. Chem, vol.275, pp.4345-50, 2000.

L. Yu, Z. Wang, M. Huang, Y. Li, K. Zeng et al., Evodia alkaloids suppress gluconeogenesis and lipogenesis by activating the constitutive androstane receptor

, Biochim. Biophys. Acta -Gene Regul. Mech, vol.1859, pp.1100-1111, 2016.

C. Breuker, A. Moreau, L. Lakhal, V. Tamasi, Y. Parmentier et al., Hepatic Expression of Thyroid Hormone-Responsive Spot 14 Protein Is Regulated by

, Constitutive Androstane Receptor (NR1I3), Endocrinology, vol.151, pp.1653-1661, 2010.

A. Marmugi, C. Lukowicz, F. Lasserre, A. Montagner, A. Polizzi et al., Activation of the Constitutive Androstane Receptor induces hepatic lipogenesis and regulates Pnpla3 gene expression in a LXR-independent way, Toxicol. Appl. Pharmacol, vol.303, pp.90-100, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01837194

A. P. Delitala, G. Capobianco, G. Delitala, P. L. Cherchi, and S. Dessole, Polycystic ovary syndrome, adipose tissue and metabolic syndrome, Arch. Gynecol. Obstet, vol.296, pp.405-419, 2017.

R. Pivonello, A. M. Isidori, D. Martino, M. C. Newell-price, J. Biller et al., Complications of Cushing's syndrome: state of the art, Lancet Diabetes Endocrinol, vol.4, pp.611-629, 2016.

P. Wei, J. Zhang, M. Egan-hafley, S. Liang, and D. D. Moore, The nuclear receptor CAR mediates specific xenobiotic induction of drug metabolism, Nature, vol.407, pp.920-923, 2000.

V. Tamasi, P. Juvan, M. Beer, D. Rozman, and U. A. Meyer, Transcriptional Activation of PPAR? by Phenobarbital in the Absence of CAR and PXR, Mol. Pharm, vol.6, pp.1573-1581, 2009.

A. Montagner, A. Korecka, A. Polizzi, Y. Lippi, Y. Blum et al., Hepatic circadian clock oscillators and nuclear receptors integrate microbiome-derived signals
URL : https://hal.archives-ouvertes.fr/hal-01482816

, Sci. Rep, vol.6, 2016.

M. Régnier, A. Polizzi, Y. Lippi, E. Fouché, G. Michel et al., Insights into the role of hepatocyte PPAR? activity in response to fasting, Mol. Cell. Endocrinol, vol.471, pp.75-88, 2018.

C. Lukowicz, S. Ellero-simatos, M. Régnier, A. Polizzi, F. Lasserre et al., Metabolic Effects of a Chronic Dietary Exposure to a Low-Dose Pesticide Cocktail in Mice: Sexual Dimorphism and Role of the Constitutive Androstane Receptor, Environ. Health Perspect, vol.126, p.67007, 2018.

D. E. Kleiner, E. M. Brunt, M. Van-natta, C. Behling, M. J. Contos et al., Design and validation of a histological scoring system for nonalcoholic fatty liver disease, Hepatology, vol.41, pp.1313-1321, 2005.

K. Kobayashi, M. Hashimoto, P. Honkakoski, and M. Negishi, Regulation of gene expression by CAR: an update, Arch. Toxicol, vol.89, pp.1045-1055, 2015.

J. P. Hernandez, L. C. Mota, W. Huang, D. D. Moore, and W. S. Baldwin, Sexually dimorphic regulation and induction of P450s by the constitutive androstane receptor (CAR)

, Toxicology, vol.256, pp.53-64, 2009.

C. Weigt, T. Hertrampf, U. Flenker, F. Hülsemann, P. Kurnaz et al., Effects of estradiol, estrogen receptor subtype-selective agonists and genistein on glucose metabolism in leptin resistant female Zucker diabetic fatty (ZDF) rats

, Steroid Biochem. Mol. Biol, vol.154, pp.12-22, 2015.

B. F. Palmer and D. J. Clegg, The sexual dimorphism of obesity, Mol. Cell. Endocrinol, vol.402, pp.113-119, 2015.

H. Y. Cho, J. Jung, H. Park, J. Yang, J. S. An et al., In vivo deletion of CAR resulted in high bone mass phenotypes in male mice, J. Cell. Physiol, vol.229, pp.561-71, 2014.

D. M. Kelly and T. H. Jones, Testosterone and obesity, Obes. Rev, vol.16, pp.581-606, 2015.

M. Wang, The role of glucocorticoid action in the pathophysiology of the Metabolic Syndrome, Nutr. Metab. (Lond), vol.2, p.3, 2005.

R. Pivonello, D. Martino, M. C. Iacuaniello, D. Simeoli, C. Muscogiuri et al., Metabolic Alterations and Cardiovascular Outcomes of Cortisol Excess, Frontiers of hormone research, pp.54-65, 2016.

M. A. Quinn, X. Xu, M. Ronfani, and J. A. Cidlowski, Estrogen Deficiency Promotes Hepatic Steatosis via a Glucocorticoid Receptor-Dependent Mechanism in Mice, Cell Rep, vol.22, pp.2690-2701, 2018.

D. L. Berliner, T. F. Dougherty, . Hepatic, . Extrahepatic-regulation-of, and . Corticosteroids, Pharmacol. Rev, vol.13, 1961.

M. Nixon, R. Upreti, and R. Andrew, 5?-Reduced glucocorticoids: a story of natural selection, J. Endocrinol, vol.212, pp.111-138, 2012.

D. Livingstone, P. Barat, D. Rollo, E. M. Rees, G. A. Weldin et al., 5?-Reductase Type 1 Deficiency or Inhibition Predisposes to Insulin Resistance, Hepatic Steatosis, and Liver Fibrosis in Rodents, Diabetes, vol.64, pp.447-458, 2015.

D. Livingstone, D. Rollo, E. M. Mak, T. Sooy, K. Walker et al., Metabolic dysfunction in female mice with disruption of 5?-reductase 1, J. Endocrinol, vol.232, pp.29-36, 2017.

J. M. Hazlehurst, A. I. Oprescu, N. Nikolaou, D. Guida, R. Grinbergs et al., Dual-5?-Reductase Inhibition Promotes Hepatic Lipid Accumulation in Man, J. Clin. Endocrinol. Metab, vol.101, pp.103-113, 2016.

S. Park, C. A. Wiwi, and D. J. Waxman, Signalling cross-talk between hepatocyte nuclear factor 4alpha and growth-hormone-activated STAT5b, Biochem. J, vol.397, pp.159-68, 2006.

C. A. Wiwi, M. Gupte, and D. J. Waxman, Sexually dimorphic P450 gene expression in liverspecific hepatocyte nuclear factor 4alpha-deficient mice, Mol. Endocrinol, vol.18, pp.1975-87, 2004.

M. Wortham, M. Czerwinski, L. He, A. Parkinson, and Y. Wan, Expression of constitutive androstane receptor, hepatic nuclear factor 4 alpha, and P450 oxidoreductase genes determines interindividual variability in basal expression and activity of a broad scope of xenobiotic metabolism genes in the human liver, Drug Metab. Dispos, vol.35, pp.1700-1710, 2007.

C. Kdsr, A. Go-;-,-gpx6, C. Kdsr, H. Go-;-nrp2, . Btk et al., CYP2C38 Functional categories Gene name GO:0002376~immune system process LY86, ) using DAVID Bioinformatics Resources 6.7 for the 100 and 487 genes upregulated in CAR-/-male and female mice, respectively, at age 16 weeks. Functional categories Gene name GO:0017127~cholesterol transporter activity APOA4, ABCG8, ABCG5 GO:0005783~endoplasmic reticulum HSD17B10, vol.2, p.9

P. Lysosome, . Hck, . Lgmn, . Rab39, and . H2-dma, , p.2

, Up-regulated in male CAR-/-mice Up-regulated in female CAR-/-mice androstane receptor (NR1I3)

, Mol. Pharmacol, vol.68, pp.1239-1253

S. S. Auerbach, J. G. Dekeyser, M. A. Stoner, and C. J. Omiecinski, CAR2 Displays Unique Ligand Binding and RXR Heterodimerization Characteristics, Drug Metab. Dispos, vol.35, pp.428-439, 2006.

F. M. Aynaci, F. Orhan, A. Orem, S. Yildirmis, and Y. Gedik, Effect of Antiepileptic drugs on plasma lipoprotein (a) and other lipid levels in childhood, J. Child Neurol, vol.16, pp.367-369, 2001.

F. Backhed, H. Ding, T. Wang, L. V. Hooper, G. Y. Koh et al., The gut microbiota as an environmental factor that regulates fat storage, Proc. Natl. Acad. Sci, vol.101, pp.15718-15723, 2004.

F. Bäckhed, J. K. Manchester, C. F. Semenkovich, G. , and J. I. , Mechanisms underlying the resistance to diet-induced obesity in germ-free mice, Proc. Natl. Acad. Sci, vol.104, pp.979-984, 2007.

Y. Bae, J. K. Kemper, and B. Kemper, Repression of CAR-mediated transactivation of CYP2B genes by the orphan nuclear receptor, short heterodimer partner (SHP), DNA Cell Biol, vol.23, pp.81-91, 2004.

M. Baes, T. Gulick, H. S. Choi, M. G. Martinoli, D. Simha et al., A new orphan member of the nuclear hormone receptor superfamily that interacts with a subset of retinoic acid response elements, Mol. Cell. Biol, vol.14, pp.1544-1552, 1994.

J. S. Bajaj, D. M. Heuman, P. B. Hylemon, A. J. Sanyal, M. B. White et al., Altered profile of human gut microbiome is associated with cirrhosis and its complications, J. Hepatol, vol.60, pp.940-947, 2014.

I. Baldi, P. Lebailly, S. Jean, L. Rougetet, S. Dulaurent et al., Pesticide contamination of workers in vineyards in France, J. Expo. Sci. Environ. Epidemiol, vol.16, pp.115-124, 2006.

W. S. Baldwin and J. A. Roling, A Concentration Addition Model for the Activation of the Constitutive Androstane Receptor by Xenobiotic Mixtures, Toxicol. Sci, vol.107, pp.93-105, 2009.

S. Ballestri, F. Nascimbeni, E. Baldelli, A. Marrazzo, D. Romagnoli et al., NAFLD as a Sexual Dimorphic Disease: Role of Gender and Reproductive Status in the Development and Progression of Nonalcoholic Fatty Liver Disease and Inherent Cardiovascular Risk, Adv. Ther, vol.34, pp.1291-1326, 2017.

M. L. Balmer, E. Slack, A. De-gottardi, M. A. Lawson, S. Hapfelmeier et al., The Liver May Act as a Firewall Mediating Mutualism Between the Host and Its Gut Commensal Microbiota, Sci. Transl. Med, vol.6, pp.237-66, 2014.

M. Barton, Cholesterol and atherosclerosis, Curr. Opin. Lipidol, vol.24, pp.214-220, 2013.

M. K. Basantani, M. T. Sitnick, L. Cai, D. S. Brenner, N. P. Gardner et al., Pnpla3/Adiponutrin deficiency in mice does not contribute to fatty liver disease or metabolic syndrome, J. Lipid Res, vol.52, pp.318-329, 2011.

P. Basilicata, A. Simonelli, A. Silvestre, M. Lamberti, P. Pedata et al., Evaluation by environmental monitoring of pesticide absorption in farm workers of 18 Italian tomato cultivations, Int. J. Immunopathol. Pharmacol, vol.26, pp.517-523, 2013.

J. Baudry, B. Allès, S. Péneau, M. Touvier, C. Méjean et al., Dietary intakes and diet quality according to levels of organic food consumption by French adults: crosssectional findings from the NutriNet-Santé Cohort Study, Public Health Nutr, vol.20, pp.638-648, 2017.

K. W. Beagley and C. M. Gockel, Regulation of innate and adaptive immunity by the female sex hormones oestradiol and progesterone, FEMS Immunol. Med. Microbiol, vol.38, pp.13-22, 2003.

A. P. Beigneux, A. H. Moser, J. K. Shigenaga, C. Grunfeld, and K. R. Feingold, Reduction in cytochrome P-450 enzyme expression is associated with repression of CAR (constitutive androstane receptor) and PXR (pregnane X receptor) in mouse liver during the acute phase response, Biochem. Biophys. Res. Commun, vol.293, pp.145-149, 2002.

L. D. Beilke, L. M. Aleksunes, R. D. Holland, D. G. Besselsen, R. D. Beger et al., Constitutive androstane receptor-mediated changes in bile acid composition contributes to hepatoprotection from lithocholic acid-induced liver injury in mice, Drug Metab. Dispos, vol.37, pp.1035-1045, 2009.

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

P. Berlit, K. H. Krause, C. C. Heuck, and B. Schellenberg, Serum lipids and anticonvulsants, Acta Neurol. Scand, vol.66, pp.328-334, 1982.

R. Bhaskar and B. Mohanty, Pesticides in mixture disrupt metabolic regulation: In silico and in vivo analysis of cumulative toxicity of mancozeb and imidacloprid on body weight of mice, Gen. Comp. Endocrinol, vol.205, pp.226-234, 2014.

F. L. Bilotta, B. Arcidiacono, S. Messineo, M. Greco, E. Chiefari et al., Insulin and osteocalcin: further evidence for a mutual cross-talk, Endocrine, vol.59, pp.622-632, 2018.

Y. Bing, S. Zhu, K. Jiang, G. Dong, J. Li et al., Reduction of thyroid hormones triggers down-regulation of hepatic CYP2B through nuclear receptors CAR and TR in a rat model of acute stroke, Biochem. Pharmacol, vol.87, pp.636-649, 2014.

D. Blizard, T. Sueyoshi, M. Negishi, S. S. Dehal, and D. Kupfer, Mechanism of induction of cytochrome p450 enzymes by the proestrogenic endocrine disruptor pesticide-methoxychlor: interactions of methoxychlor metabolites with the constitutive androstane receptor system, Drug Metab. Dispos, vol.29, pp.781-785, 2001.

A. R. Boobis, J. E. Doe, B. Heinrich-hirsch, M. E. Meek, S. Munn et al., IPCS Framework for Analyzing the Relevance of a Noncancer Mode of Action for Humans, Crit. Rev. Toxicol, vol.38, pp.87-96, 2008.

S. L. Booth, A. Centi, S. R. Smith, and C. Gundberg, The role of osteocalcin in human glucose metabolism: marker or mediator?, Nat. Rev. Endocrinol, vol.9, pp.43-55, 2013.

P. J. Bosma, J. Seppen, B. Goldhoorn, C. Bakker, R. P. Oude-elferink et al., Bilirubin UDP-glucuronosyltransferase 1 is the only relevant bilirubin glucuronidating isoform in man, J. Biol. Chem, vol.269, pp.17960-17964, 1994.

W. Bourguet, M. Ruff, P. Chambon, H. Gronemeyer, and D. Moras, Crystal structure of the ligandbinding domain of the human nuclear receptor RXR-alpha, Nature, vol.375, pp.377-382, 1995.

J. Boursier, O. Mueller, M. Barret, M. Machado, L. Fizanne et al., The severity of nonalcoholic fatty liver disease is associated with gut dysbiosis and shift in the metabolic function of the gut microbiota, Hepatology, vol.63, pp.764-775, 2016.

G. Bouvier, N. Seta, A. Vigouroux-villard, O. Blanchard, and I. Momas, Insecticide Urinary Metabolites in Nonoccupationally Exposed Populations, J. Toxicol. Environ. Heal. Part B, vol.8, pp.485-512, 2005.
URL : https://hal.archives-ouvertes.fr/ineris-00962976

J. L. Boyer, Bile Formation and Secretion, Comprehensive Physiology, pp.1035-1078, 2013.

A. Bradman, L. Quirós-alcalá, R. Castorina, R. A. Schall, J. Camacho et al., Effect of Organic Diet Intervention on Pesticide Exposures in Young Children Living in Low-Income Urban and Agricultural Communities, Environ. Health Perspect, vol.123, pp.1086-1093, 2015.

F. Braet and E. Wisse, Structural and functional aspects of liver sinusoidal endothelial cell fenestrae: a review, Comp. Hepatol, vol.1, p.1, 2002.

A. Braeuning, C. Ittrich, C. Köhle, S. Hailfinger, M. Bonin et al., Differential gene expression in periportal and perivenous mouse hepatocytes, FEBS J, vol.273, pp.5051-5061, 2006.

A. Braeuning, C. Ittrich, C. Köhle, S. Hailfinger, M. Bonin et al., Differential gene expression in periportal and perivenous mouse hepatocytes, FEBS J, vol.273, pp.5051-5061, 2006.

V. Braniste, M. Leveque, C. Buisson-brenac, L. Bueno, J. Fioramonti et al., Oestradiol decreases colonic permeability through oestrogen receptor ?-mediated up-regulation of occludin and junctional adhesion molecule-A in epithelial cells, J. Physiol, vol.587, pp.3317-3328, 2009.
URL : https://hal.archives-ouvertes.fr/hal-01604720

G. A. Brent, Clinical practice. Graves' disease, N. Engl. J. Med, vol.358, pp.2594-2605, 2008.

G. A. Brent, Mechanisms of thyroid hormone action, J. Clin. Invest, vol.122, pp.3035-3043, 2012.

C. Breuker, A. Moreau, L. Lakhal, V. Tamasi, Y. Parmentier et al., Hepatic expression of thyroid hormone-responsive spot 14 protein is regulated by constitutive androstane receptor (NR1I3), Endocrinology, vol.151, pp.1653-1661, 2010.

J. Bricambert, J. Miranda, F. Benhamed, J. Girard, C. Postic et al., Salt-inducible kinase 2 links transcriptional coactivator p300 phosphorylation to the prevention of ChREBP-dependent hepatic steatosis in mice, J. Clin. Invest, vol.120, pp.4316-4331, 2010.

J. D. Browning and J. D. Horton, Molecular mediators of hepatic steatosis and liver injury, J. Clin. Invest, vol.114, pp.147-152, 2004.

J. D. Browning, L. S. Szczepaniak, R. Dobbins, P. Nuremberg, J. D. Horton et al., Prevalence of hepatic steatosis in an urban population in the United States: Impact of ethnicity, Hepatology, vol.40, pp.1387-1395, 2004.

J. D. Browning, K. S. Kumar, M. H. Saboorian, and D. L. Thiele, Ethnic differences in the prevalence of cryptogenic cirrhosis, Am. J. Gastroenterol, vol.99, pp.292-298, 2004.

R. W. Brownsey, A. N. Boone, J. E. Elliott, J. E. Kulpa, and W. M. Lee, Regulation of acetyl-CoA carboxylase, Biochem. Soc. Trans, vol.34, pp.223-227, 2006.

P. Brun, I. Castagliuolo, V. Leo, . Di, A. Buda et al., Increased intestinal permeability in obese mice: new evidence in the pathogenesis of nonalcoholic steatohepatitis, Am. J. Physiol. Liver Physiol, vol.292, pp.518-525, 2007.

G. Bryzgalova, H. Gao, B. Ahren, J. R. Zierath, D. Galuska et al., Evidence that oestrogen receptor-? plays an important role in the regulation of glucose homeostasis in mice: insulin sensitivity in the liver, Diabetologia, vol.49, pp.588-597, 2006.

S. C. Burgess, N. Hausler, M. Merritt, F. M. Jeffrey, C. Storey et al., Impaired tricarboxylic acid cycle activity in mouse livers lacking cytosolic phosphoenolpyruvate carboxykinase, J. Biol. Chem, vol.279, pp.48941-48949, 2004.

O. Burk, K. A. Arnold, A. K. Nussler, E. Schaeffeler, E. Efimova et al., Antimalarial Artemisinin Drugs Induce Cytochrome P450 and MDR1 Expression by Activation of Xenosensors Pregnane X Receptor and Constitutive Androstane Receptor, Mol. Pharmacol, vol.67, pp.1954-1965, 2005.

S. H. Caldwell, D. M. Harris, J. T. Patrie, and E. E. Hespenheide, Is NASH underdiagnosed among African Americans?, Am. J. Gastroenterol, vol.97, pp.1496-1500, 2002.

A. C. Calkin and P. Tontonoz, Transcriptional integration of metabolism by the nuclear sterol-activated receptors LXR and FXR, Nat. Rev. Mol. Cell Biol, vol.13, pp.213-224, 2012.

K. Campbell, B. Baker, A. Tovar, E. Economos, B. Williams et al., The Association Between Skin Rashes and Work Environment, Personal Protective Equipment, and Hygiene Practices Among Female Farmworkers. Workplace Health Saf, vol.65, pp.313-321, 2017.

É. Campos and C. Freire, Exposure to non-persistent pesticides and thyroid function: A systematic review of epidemiological evidence, Int. J. Hyg. Environ. Health, vol.219, pp.481-497, 2016.

C. Casals-casas and B. Desvergne, Endocrine disruptors: from endocrine to metabolic disruption, Annu. Rev. Physiol, vol.73, pp.135-162, 2011.

J. E. Casida, Curious about Pesticide Action, J. Agric. Food Chem, vol.59, pp.2762-2769, 2011.

L. Cerveny, L. Svecova, E. Anzenbacherova, R. Vrzal, F. Staud et al., Valproic Acid Induces CYP3A4 and MDR1 Gene Expression by Activation of Constitutive Androstane Receptor and Pregnane X Receptor Pathways ABSTRACT, vol.35, pp.1032-1041, 2007.

A. Chawla, J. J. Repa, R. M. Evans, and D. J. Mangelsdorf, Nuclear Receptors and Lipid Physiology: Opening the X-Files. Science, vol.294, pp.1866-1870, 2001.

W. Chen, G. Chen, D. L. Head, D. J. Mangelsdorf, and D. W. Russell, Enzymatic reduction of oxysterols impairs LXR signaling in cultured cells and the livers of mice, Cell Metab, vol.5, pp.73-79, 2007.

W. Chen, B. Chang, L. Li, C. , and L. , Patatin-like phospholipase domain-containing 3/adiponutrin deficiency in mice is not associated with fatty liver disease, Hepatology, vol.52, pp.1134-1142, 2010.

X. Chen, S. Maiti, J. Zhang, C. , and G. , Nuclear receptor interactions in methotrexate induction of human dehydroepiandrosterone sulfotransferase (hSULT2A1), J. Biochem. Mol. Toxicol, vol.20, pp.309-317, 2006.

X. Chen, J. Zhang, S. M. Baker, C. , and G. , Human constitutive androstane receptor mediated methotrexate induction of human dehydroepiandrosterone sulfotransferase (hSULT2A1), Toxicology, vol.231, pp.224-233, 2007.

Y. Chen, K. S. Mccommis, D. Ferguson, A. M. Hall, C. A. Harris et al., Inhibition of the Mitochondrial Pyruvate Carrier by Tolylfluanid, Endocrinology, vol.159, pp.609-621, 2018.

X. Cheng and C. D. Klaassen, Perfluorocarboxylic acids induce cytochrome P450 enzymes in mouse liver through activation of PPAR-alpha and CAR transcription factors, Toxicol. Sci, vol.106, pp.29-36, 2008.

Z. Cheng and M. F. White, The AKTion in non-canonical insulin signaling, Nat. Med, vol.18, pp.351-353, 2012.

S. Cheng, M. Zou, Q. Liu, J. Kuang, J. Shen et al., Activation of Constitutive Androstane Receptor Prevents Cholesterol Gallstone Formation, Am. J. Pathol, vol.187, pp.808-818, 2017.

N. J. Cherrington, D. P. Hartley, N. Li, D. R. Johnson, and C. D. Klaassen, Organ distribution of multidrug resistance proteins 1, 2, and 3 (Mrp1, 2, and 3) mRNA and hepatic induction of Mrp3 by constitutive androstane receptor activators in rats, J. Pharmacol. Exp. Ther, vol.300, pp.97-104, 2002.

H. Y. Cho, J. Jung, H. Park, J. Yang, S. Jung et al., , 2014.

, In vivo deletion of CAR resulted in high bone mass phenotypes in male mice, J. Cell. Physiol, vol.229, pp.561-571

H. Choi, M. Chung, I. Tzameli, D. Simha, Y. Lee et al., Differential Transactivation by Two Isoforms of the Orphan Nuclear Hormone Receptor CAR, J. Biol. Chem, vol.272, pp.23565-23571, 1997.

J. M. Clark, F. L. Brancati, and A. M. Diehl, The prevalence and etiology of elevated aminotransferase levels in the United States, Am. J. Gastroenterol, vol.98, pp.960-967, 2003.

S. P. Claus, H. Guillou, S. Ellero-simatos, C. E. Cerniglia, C. et al., The gut microbiota: a major player in the toxicity of environmental pollutants?, Npj Biofilms Microbiomes, vol.2, p.16003, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01602371

K. H. Clodfelter, M. G. Holloway, P. Hodor, S. Park, W. J. Ray et al., Sex-dependent liver gene expression is extensive and largely dependent upon signal transducer and activator of transcription 5b (STAT5b): STAT5b-dependent activation of male genes and repression of female genes revealed by microarray analysis, Mol. Endocrinol, vol.20, pp.1333-1351, 2006.

K. H. Clodfelter, G. D. Miles, V. Wauthier, M. G. Holloway, X. Zhang et al., Role of STAT5a in regulation of sex-specific gene expression in female but not male mouse liver revealed by microarray analysis, Physiol. Genomics, vol.31, pp.63-74, 2007.

J. C. Cohen, J. D. Horton, and H. H. Hobbs, Human fatty liver disease: old questions and new insights, Science, vol.332, pp.1519-1523, 2011.

S. P. Cole, Multidrug Resistance Protein 1 (MRP1, ABCC1), a "Multitasking" ATP-binding Cassette (ABC) Transporter, J. Biol. Chem, vol.289, pp.30880-30888, 2014.

P. Costet, C. Legendre, J. Moré, A. Edgar, P. Galtier et al., Peroxisome proliferator-activated receptor alpha-isoform deficiency leads to progressive dyslipidemia with sexually dimorphic obesity and steatosis, J. Biol. Chem, vol.273, pp.29577-29585, 1998.

D. G. Cotter, B. Ercal, X. Huang, J. M. Leid, D. A. Avignon et al., Ketogenesis prevents diet-induced fatty liver injury and hyperglycemia, J. Clin. Invest, vol.124, pp.5175-5190, 2014.

X. Coumoul, M. Diry, and R. Barouki, PXR-dependent induction of human CYP3A4 gene expression by organochlorine pesticides, Biochem. Pharmacol, vol.64, pp.1513-1519, 2002.

S. Cox, A. S. Niskar, K. M. Narayan, M. , and M. , Prevalence of Self-Reported Diabetes and Exposure to Organochlorine Pesticides among Mexican Americans: Hispanic Health and Nutrition Examination Survey, Environ. Health Perspect, vol.115, pp.1747-1752, 1982.

L. S. Csaki, R. , and K. , Lipins: multifunctional lipid metabolism proteins, Annu. Rev. Nutr, vol.30, pp.257-272, 2010.

C. L. Curl, R. A. Fenske, J. C. Kissel, J. H. Shirai, T. F. Moate et al., Evaluation of take-home organophosphorus pesticide exposure among agricultural workers and their children, Environ. Health Perspect, vol.110, pp.787-92, 2002.

C. L. Curl, R. A. Fenske, and K. Elgethun, Organophosphorus pesticide exposure of urban and suburban preschool children with organic and conventional diets, Environ. Health Perspect, vol.111, pp.377-382, 2003.

C. L. Curl, S. A. Beresford, R. A. Fenske, A. L. Fitzpatrick, C. Lu et al., Estimating Pesticide Exposure from Dietary Intake and Organic Food Choices: The Multi-Ethnic Study of Atherosclerosis (MESA), Environ. Health Perspect, vol.123, pp.475-483, 2015.

P. G. Curran and L. J. Degroot, The effect of hepatic enzyme-inducing drugs on thyroid hormones and the thyroid gland, Endocr. Rev, vol.12, pp.135-150, 1991.

R. A. Currie, R. C. Peffer, A. K. Goetz, C. J. Omiecinski, and J. I. Goodman, Phenobarbital and propiconazole toxicogenomic profiles in mice show major similarities consistent with the key role that constitutive androstane receptor (CAR) activation plays in their mode of action, Toxicology, vol.321, pp.80-88, 2014.

N. E. Daryani, N. E. Daryani, S. M. Alavian, A. Zare, S. Fereshtehnejad et al., Non-alcoholic steatohepatitis and influence of age and gender on histopathologic findings, World J. Gastroenterol, vol.16, pp.4169-4175, 2010.

M. Dashty, A quick look at biochemistry: Carbohydrate metabolism, Clin. Biochem, vol.46, pp.1339-1352, 2013.

A. Debost-legrand, C. Warembourg, C. Massart, C. Chevrier, N. Bonvallot et al., Prenatal exposure to persistent organic pollutants and organophosphate pesticides, and markers of glucose metabolism at birth, Environ. Res, vol.146, pp.207-217, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01259218

J. G. Dekeyser, M. C. Stagliano, S. S. Auerbach, K. S. Prabhu, A. D. Jones et al., Di(2-ethylhexyl) phthalate is a highly potent agonist for the human constitutive androstane receptor splice variant CAR2, Mol. Pharmacol, vol.75, pp.1005-1013, 2009.

V. Delfosse, M. Grimaldi, V. Cavaillès, P. Balaguer, and W. Bourguet, Structural and functional profiling of environmental ligands for estrogen receptors, Environ. Health Perspect, vol.122, pp.1306-1313, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-01076600

V. Delfosse, B. Dendele, T. Huet, M. Grimaldi, A. Boulahtouf et al., Synergistic activation of human pregnane X receptor by binary cocktails of pharmaceutical and environmental compounds, Nat. Commun, vol.6, p.8089, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01838034

P. Denechaud, P. Bossard, J. A. Lobaccaro, L. Millatt, B. Staels et al., ChREBP , but not LXRs , is required for the induction of glucose-regulated genes in mouse liver, J. Clin. Invest, vol.118, 2008.

R. Dentin, L. Tomas-cobos, F. Foufelle, J. Leopold, J. Girard et al., Glucose 6-phosphate, rather than xylulose 5-phosphate, is required for the activation of ChREBP in response to glucose in the liver, J. Hepatol, vol.56, pp.199-209, 2012.

B. Desvergne, L. Michalik, and W. Wahli, Transcriptional Regulation of Metabolism, Physiol. Rev, vol.86, pp.465-514, 2006.

B. Desvergne, J. N. Feige, and C. Casals-casas, PPAR-mediated activity of phthalates: A link to the obesity epidemic?, Mol. Cell. Endocrinol, vol.304, pp.43-48, 2009.

A. M. Diehl and C. Day, Nonalcoholic Steatohepatitis, N. Engl. J. Med, vol.378, pp.779-781, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01392300

X. Ding, K. Lichti, I. Kim, F. J. Gonzalez, and J. L. Staudinger, Regulation of constitutive androstane receptor and its target genes by fasting, cAMP, hepatocyte nuclear factor ?, and the coactivator peroxisome proliferator-activated receptor ? coactivator-1?, J. Biol. Chem, vol.281, pp.26540-26551, 2006.

L. J. Dixon, M. Barnes, H. Tang, M. T. Pritchard, and L. E. Nagy, Kupffer Cells in the Liver, Comprehensive Physiology, pp.785-797, 2013.

M. M. Domingos, M. F. Rodrigues, U. S. Stotzer, D. R. Bertucci, M. V. Souza et al., Resistance training restores the gene expression of molecules related to fat oxidation and lipogenesis in the liver of ovariectomized rats, Eur. J. Appl. Physiol, vol.112, pp.1437-1444, 2012.

M. T. Donato, M. J. Gómez-lechón, R. Jover, T. Nakamura, and J. Castell, Human hepatocyte growth factor down-regulates the expression of cytochrome P450 isozymes in human hepatocytes in primary culture, J. Pharmacol. Exp. Ther, vol.284, pp.760-767, 1998.

B. Dong, P. K. Saha, W. Huang, W. Chen, L. A. Abu-elheiga et al., Activation of nuclear receptor CAR ameliorates diabetes and fatty liver disease, Proc. Natl. Acad. Sci, vol.106, pp.18831-18836, 2009.

B. Dong, P. K. Saha, W. Huang, W. Chen, L. A. Abu-elheiga et al., Activation of nuclear receptor CAR ameliorates, Proc. Natl. Acad. Sci. 106, 2009.

B. Dong, P. K. Saha, W. Huang, W. Chen, L. A. Abu-elheiga et al., Activation of nuclear receptor CAR ameliorates diabetes and fatty liver disease, Proc. Natl. Acad. Sci. U. S. A, vol.106, pp.18831-18836, 2009.

K. L. Donnelly, C. I. Smith, S. J. Schwarzenberg, J. Jessurun, M. D. Boldt et al., Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease, J. Clin. Invest, vol.115, pp.1343-1351, 2005.

B. Döring and E. Petzinger, Phase 0 and phase III transport in various organs: Combined concept of phases in xenobiotic transport and metabolism, Drug Metab. Rev, vol.46, pp.261-282, 2014.

M. L. Dourson, S. P. Felter, R. , and D. , Evolution of science-based uncertainty factors in noncancer risk assessment, Regul. Toxicol. Pharmacol, vol.24, pp.108-120, 1996.

G. Du, O. Shen, H. Sun, J. Fei, C. Lu et al., Assessing hormone receptor activities of pyrethroid insecticides and their metabolites in reporter gene assays, Toxicol. Sci, vol.116, pp.58-66, 2010.

S. Ducheix, J. M. Lobaccaro, P. G. Martin, and H. Guillou, Liver X Receptor: an oxysterol sensor and a major player in the control of lipogenesis, Chem. Phys. Lipids, vol.164, pp.500-514, 2011.

M. Durazzo, P. Belci, A. Collo, V. Prandi, E. Pistone et al., Gender specific medicine in liver diseases: a point of view, World J. Gastroenterol, vol.20, pp.2127-2135, 2014.

C. Duret, M. Daujat-chavanieu, J. Pascussi, L. Pichard-garcia, P. Balaguer et al., Ketoconazole and miconazole are antagonists of the human glucocorticoid receptor: Consequences on the expression and function of the constitutive androstane receptor and the pregnane X receptor, Mol. Pharmacol, vol.70, pp.329-339, 2006.
URL : https://hal.archives-ouvertes.fr/inserm-00086861

D. Eberlé, B. Hegarty, P. Bossard, P. Ferré, and F. Foufelle, SREBP transcription factors: master regulators of lipid homeostasis, Biochimie, vol.86, pp.839-848, 2004.

P. B. Eckburg, E. M. Bik, C. N. Bernstein, E. Purdom, L. Dethlefsen et al., Diversity of the human intestinal microbial flora, Science, vol.308, pp.1635-1638, 2005.

J. Eirís, M. I. Novo-rodríguez, M. Del-río, P. Meseguer, M. C. Del-río et al., The effects on lipid and apolipoprotein serum levels of long-term carbamazepine, valproic acid and phenobarbital therapy in children with epilepsy, Epilepsy Res, vol.41, pp.1-7, 2000.

J. M. Eirís, S. Lojo, M. C. Del-río, I. Novo, M. Bravo et al., Effects of long-term treatment with antiepileptic drugs on serum lipid levels in children with epilepsy, Neurology, vol.45, pp.1155-1157, 1995.

C. R. Elcombe, B. M. Elcombe, J. R. Foster, S. Chang, D. J. Ehresman et al., Hepatocellular hypertrophy and cell proliferation in Sprague-Dawley rats from dietary exposure to potassium perfluorooctanesulfonate results from increased expression of xenosensor nuclear receptors PPAR? and CAR/PXR, Toxicology, vol.293, pp.16-29, 2012.

K. H. Elvevold, G. I. Nedredal, A. Revhaug, and B. Smedsrød, Scavenger properties of cultivated pig liver endothelial cells, Comp. Hepatol, vol.3, p.4, 2004.

E. Evangelou, G. Ntritsos, M. Chondrogiorgi, F. K. Kavvoura, A. F. Hernández et al., Exposure to pesticides and diabetes: A systematic review and meta-analysis, Environ. Int, vol.91, pp.60-68, 2016.

C. J. Everett and E. M. Matheson, Biomarkers of pesticide exposure and diabetes in the 1999-2004 national health and nutrition examination survey, Environ. Int, vol.36, pp.398-401, 2010.

L. Faessel and T. Prospective, Notes et études socio-économiques, pp.7-39, 2016.

C. N. Falany, X. Xie, J. Wang, J. Ferrer, and J. L. Falany, Molecular cloning and expression of novel sulphotransferase-like cDNAs from human and rat brain, Biochem. J. 346 Pt, vol.3, pp.857-864, 2000.

G. C. Farrell and C. Z. Larter, Nonalcoholic fatty liver disease: From steatosis to cirrhosis, Hepatology, vol.43, pp.99-112, 2006.

J. N. Feige, L. Gelman, D. Rossi, V. Zoete, R. Métivier et al., The endocrine disruptor monoethyl-hexyl-phthalate is a selective peroxisome proliferator-activated receptor gamma modulator that promotes adipogenesis, J. Biol. Chem, vol.282, pp.19152-19166, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00177041

R. A. Fenske, C. Lu, N. J. Simcox, C. Loewenherz, J. Touchstone et al., Strategies for assessing children's organophosphorus pesticide exposures in agricultural communities, J. Expo. Anal. Environ. Epidemiol, vol.10, pp.662-671

R. A. Fenske, S. J. Hamburger, and C. L. Guyton, Occupational exposure to fosetyl-Al fungicide during spraying of ornamentals in greenhouses, Arch. Environ. Contam. Toxicol, vol.16, pp.615-621, 1987.

P. Ferré and F. Foufelle, SREBP-1c Transcription Factor and Lipid Homeostasis: Clinical Perspective, Horm. Res. Paediatr, vol.68, pp.72-82, 2007.

G. Filhoulaud, S. Guilmeau, R. Dentin, J. Girard, and C. Postic, Novel insights into ChREBP regulation and function, Trends Endocrinol. Metab, vol.24, pp.257-268, 2013.

K. Flurkey, J. Currer, H. , and D. , Mouse models in aging research, Fac. Res, 2000.

M. Foretz, C. Guichard, P. Ferré, and F. Foufelle, Sterol regulatory element binding protein-1c is a major mediator of insulin action on the hepatic expression of glucokinase and lipogenesis-related genes, Proc. Natl. Acad. Sci. U. S. A, vol.96, pp.12737-12742, 1999.

B. M. Forman, I. Tzameli, H. S. Choi, J. Chen, D. Simha et al., , 1998.

, Androstane metabolites bind to and deactivate the nuclear receptor CAR-beta, Nature, vol.395, pp.612-615

B. M. Forman, I. Tzameli, H. S. Choi, J. Chen, D. Simha et al., , 1998.

, Androstane metabolites bind to and deactivate the nuclear receptor CAR-beta, Nature, vol.395, pp.612-615

F. Foufelle and P. Ferré, New perspectives in the regulation of hepatic glycolytic and lipogenic genes by insulin and glucose: a role for the transcription factor sterol regulatory element binding protein-1c, Biochem. J, vol.366, pp.377-391, 2002.

F. Foufelle, J. Girard, and P. Ferré, Regulation of lipogenic enzyme expression by glucose in liver and adipose tissue: a review of the potential cellular and molecular mechanisms, Adv. Enzyme Regul, vol.36, pp.199-226, 1996.

H. S. Fox, B. L. Bond, and T. G. Parslow, Estrogen regulates the IFN-gamma promoter, J. Immunol, vol.146, pp.4362-4367, 1991.

J. Galgani and E. Ravussin, Energy metabolism, fuel selection and body weight regulation, Int. J. Obes, vol.32, pp.109-119, 2008.

B. Gao, X. Bian, R. Mahbub, and K. Lu, Sex-Specific Effects of Organophosphate Diazinon on the Gut Microbiome and Its Metabolic Functions, Environ. Health Perspect, vol.125, pp.198-206, 2016.

J. Gao, J. He, Y. Zhai, T. Wada, and W. Xie, The constitutive androstane receptor is an anti-obesity nuclear receptor that improves insulin sensitivity, J. Biol. Chem, vol.284, pp.25984-25992, 2009.

J. Gao, J. Yan, M. Xu, S. Ren, and W. Xie, CAR Suppresses Hepatic Gluconeogenesis by Facilitating the Ubiquitination and Degradation of PGC1?, Mol. Endocrinol, vol.29, pp.1558-1570, 2015.

A. Gastaldelli, K. Cusi, M. Pettiti, J. Hardies, Y. Miyazaki et al., Relationship Between Hepatic/Visceral Fat and Hepatic Insulin Resistance in Nondiabetic and Type 2 Diabetic Subjects, Gastroenterology, vol.133, pp.496-506, 2007.

D. W. Gaylor, The use of safety factors for controlling risk, J. Toxicol. Environ. Health, vol.11, pp.329-336, 1983.

R. Gebhardt, J. Alber, H. Wegner, and D. Mecke, Different drug metabolizing capacities in cultured periportal and pericentral hepatocytes, Biochem. Pharmacol, vol.48, pp.761-766, 1994.

C. Ged, J. M. Rouillon, L. Pichard, J. Combalbert, N. Bressot et al., The increase in urinary excretion of 6 beta-hydroxycortisol as a marker of human hepatic cytochrome P450IIIA induction, Br. J. Clin. Pharmacol, vol.28, pp.373-387, 1989.

S. Gerbal-chaloin, M. Daujat, J. Pascussi, L. Pichard-garcia, M. Vilarem et al., Transcriptional Regulation of CYP2C9 Gene, J. Biol. Chem, vol.277, pp.209-217, 2002.

P. Germain, B. Staels, C. Dacquet, M. Spedding, and V. Laudet, Overview of nomenclature of nuclear receptors, Pharmacol. Rev, vol.58, pp.685-704, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00187929

Z. Ghafour-rashidi, E. Dermenaki-farahani, A. Aliahmadi, H. Esmaily, A. Mohammadirad et al., Protection by cAMP and cGMP phosphodiesterase inhibitors of diazinon-induced hyperglycemia and oxidative/nitrosative stress in rat Langerhans islets cells: Molecular evidence for involvement of non-cholinergic mechanisms, Pestic. Biochem. Physiol, vol.87, pp.261-270, 2007.

R. Ghose, O. Omoluabi, A. Gandhi, P. Shah, K. Strohacker et al., , 2011.

, Role of high-fat diet in regulation of gene expression of drug metabolizing enzymes and transporters, Life Sci, vol.89, pp.57-64

D. A. Giles, M. E. Moreno-fernandez, T. E. Stankiewicz, S. Graspeuntner, M. Cappelletti et al., Thermoneutral housing exacerbates nonalcoholic fatty liver disease in mice and allows for sex-independent disease modeling, Nat. Med, vol.23, pp.829-838, 2017.

P. Ginès, P. S. Kamath, and V. Arroyo, Chronic liver failure : mechanisms and management, 2011.

A. K. Goetz, W. Bao, H. Ren, J. E. Schmid, D. B. Tully et al., Gene expression profiling in the liver of CD-1 mice to characterize the hepatotoxicity of triazole fungicides, Toxicol. Appl. Pharmacol, vol.215, pp.274-284, 2006.

M. J. Gómez-lechón, T. Donato, R. Jover, C. Rodriguez, X. Ponsoda et al., Expression and induction of a large set of drug-metabolizing enzymes by the highly differentiated human hepatoma cell line BC2, Eur. J. Biochem, vol.268, pp.1448-1459, 2001.

B. Goodwin, E. Hodgson, and C. Liddle, The orphan human pregnane X receptor mediates the transcriptional activation of CYP3A4 by rifampicin through a distal enhancer module, Mol. Pharmacol, vol.56, pp.1329-1339, 1999.

M. Goodwin, C. Herath, Z. Jia, C. Leung, M. T. Coughlan et al., Advanced glycation end products augment experimental hepatic fibrosis, J. Gastroenterol. Hepatol, vol.28, pp.369-376, 2013.

S. Gremlich, R. Roduit, and B. Thorens, Dexamethasone induces posttranslational degradation of GLUT2 and inhibition of insulin secretion in isolated pancreatic beta cells. Comparison with the effects of fatty acids, J. Biol. Chem, vol.272, pp.3216-3222, 1997.

F. Grün and B. Blumberg, Endocrine disrupters as obesogens, Mol. Cell. Endocrinol, vol.304, pp.19-29, 2009.

F. P. Guengerich, Z. Wu, and C. J. Bartleson, Function of human cytochrome P450s: Characterization of the orphans, Biochem. Biophys. Res. Commun, vol.338, pp.465-469, 2005.

C. Guinez, G. Filhoulaud, F. Rayah-benhamed, S. Marmier, C. Dubuquoy et al., O-GlcNAcylation Increases ChREBP Protein Content and Transcriptional Activity in the Liver, Diabetes, vol.60, pp.1399-1413, 2011.

R. B. Gunier, M. H. Ward, M. Airola, E. M. Bell, J. Colt et al., Determinants of Agricultural Pesticide Concentrations in Carpet Dust, Environ. Health Perspect, vol.119, pp.970-976, 2011.

D. Guo, J. Sarkar, K. Suino-powell, Y. Xu, K. Matsumoto et al., Induction of Nuclear Translocation of Constitutive Androstane Receptor by Peroxisome Proliferatoractivated Receptor ? Synthetic Ligands in Mouse Liver, J. Biol. Chem, vol.282, pp.36766-36776, 2007.

G. L. Guo, S. Choudhuri, and C. D. Klaassen, Induction profile of rat organic anion transporting polypeptide 2 (oatp2) by prototypical drug-metabolizing enzyme inducers that activate gene expression through ligandactivated transcription factor pathways, J. Pharmacol. Exp. Ther, vol.300, pp.206-212, 2002.

G. L. Guo, G. Lambert, M. Negishi, J. M. Ward, H. B. Brewer et al., Complementary roles of farnesoid X receptor, pregnane X receptor, and constitutive androstane receptor in protection against bile acid toxicity, J. Biol. Chem, vol.278, pp.45062-45071, 2003.

K. Z. Guyton, S. Barone, R. C. Brown, S. Y. Euling, J. Jinot et al., Mode of Action Frameworks: A Critical Analysis, J. Toxicol. Environ. Heal. Part B, vol.11, pp.16-31, 2008.

G. L. Hager, C. S. Lim, C. Elbi, and C. T. Baumann, Trafficking of nuclear receptors in living cells, J. Steroid Biochem. Mol. Biol, vol.74, pp.249-254, 2000.

C. Handschin and U. A. Meyer, Induction of Drug Metabolism: The Role of Nuclear Receptors, Pharmacol. Rev, vol.55, pp.649-673, 2003.

C. Handschin, M. Podvinec, R. Amherd, R. Looser, J. Ourlin et al., Cholesterol and Bile Acids Regulate Xenosensor Signaling in Drug-mediated Induction of Cytochromes P450, J. Biol. Chem, vol.277, pp.29561-29567, 2002.

D. G. Hardie and M. L. Ashford, AMPK: regulating energy balance at the cellular and whole body levels, Physiology (Bethesda), vol.29, pp.99-107, 2014.

M. E. Harnly, A. Bradman, M. Nishioka, T. E. Mckone, D. Smith et al., Pesticides in dust from homes in an agricultural area, Environ. Sci. Technol, vol.43, pp.8767-8774, 2009.

S. A. Harrison and C. P. Day, Benefits of lifestyle modification in NAFLD, Gut, vol.56, pp.1760-1769, 2007.

E. L. Haughton, S. J. Tucker, C. J. Marek, E. Durward, V. Leel et al., Pregnane X receptor activators inhibit human hepatic stellate cell transdifferentiation in vitro, Gastroenterology, vol.131, pp.194-209, 2006.

L. Hebbard, G. , and J. , Animal models of nonalcoholic fatty liver disease, Nat. Rev. Gastroenterol. Hepatol, vol.8, pp.35-44, 2011.

T. Heinzel, R. M. Lavinsky, T. M. Mullen, M. Söderstrom, C. D. Laherty et al., A complex containing N-CoR, mSin3 and histone deacetylase mediates transcriptional repression, Nature, vol.387, pp.43-48, 1997.

J. P. Hernandez, W. Huang, L. M. Chapman, S. Chua, D. D. Moore et al., The Environmental Estrogen, Nonylphenol, Activates the Constitutive Androstane Receptor, Toxicol. Sci, vol.98, pp.416-426, 2007.

J. P. Hernandez, L. C. Mota, W. Huang, D. D. Moore, and W. S. Baldwin, Sexually dimorphic regulation and induction of P450s by the constitutive androstane receptor (CAR), Toxicology, vol.256, pp.53-64, 2009.

J. P. Hernandez, L. C. Mota, and W. S. Baldwin, Activation of CAR and PXR by Dietary, Environmental and Occupational Chemicals Alters Drug Metabolism, Intermediary Metabolism, and Cell Proliferation, Curr. Pharmacogenomics Person. Med, vol.7, pp.81-105, 2009.

J. P. Hernandez, L. C. Mota, and W. S. Baldwin, Activation of CAR and PXR by Dietary, Environmental and Occupational Chemicals Alters Drug Metabolism, Intermediary Metabolism, and Cell Proliferation, Curr. Pharmacogenomics Person. Med, vol.7, pp.81-105, 2009.

J. Herrington and C. Carter-su, Signaling pathways activated by the growth hormone receptor, Trends Endocrinol. Metab, vol.12, pp.252-257, 2001.

K. N. Hewitt, K. Pratis, M. E. Jones, and E. R. Simpson, Estrogen replacement reverses the hepatic steatosis phenotype in the male aromatase knockout mouse, Endocrinology, vol.145, pp.1842-1848, 2004.

M. G. Holloway, G. D. Miles, A. A. Dombkowski, and D. J. Waxman, Liver-specific hepatocyte nuclear factor-4alpha deficiency: greater impact on gene expression in male than in female mouse liver, Mol. Endocrinol, vol.22, pp.1274-1286, 2008.

N. Hong, K. Kim, I. Lee, P. M. Lind, L. Lind et al., The association between obesity and mortality in the elderly differs by serum concentrations of persistent organic pollutants: a possible explanation for the obesity paradox, Int. J. Obes, vol.36, pp.1170-1175, 2012.

K. Howe, F. Sanat, A. E. Thumser, T. Coleman, and N. Plant, The statin class of HMG-CoA reductase inhibitors demonstrate differential activation of the nuclear receptors PXR, CAR and FXR, as well as their downstream target genes, Xenobiotica, vol.41, pp.519-529, 2011.

P. L. Huang, A comprehensive definition for metabolic syndrome, Dis. Model. Mech, vol.2, pp.231-237, 2009.

C. Huang and H. C. Freake, Thyroid hormone regulates the acetyl-CoA carboxylase PI promoter, Biochem. Biophys. Res. Commun, vol.249, pp.704-708, 1998.

W. Huang, J. Zhang, S. S. Chua, M. Qatanani, Y. Han et al., Induction of bilirubin clearance by the constitutive androstane receptor (CAR), Proc. Natl. Acad. Sci. U. S. A, vol.100, pp.4156-4161, 2003.

W. Huang, J. Zhang, P. Wei, W. T. Schrader, and D. D. Moore, Meclizine Is an Agonist Ligand for Mouse Constitutive Androstane Receptor (CAR) and an Inverse Agonist for Human CAR, Mol. Endocrinol, vol.18, pp.2402-2408, 2004.

S. Impey, S. R. Mccorkle, H. Cha-molstad, J. M. Dwyer, G. S. Yochum et al., Defining the CREB Regulon: A Genome-Wide Analysis of Transcription Factor Regulatory Regions, Cell, vol.119, pp.1041-1054, 2004.

K. Ishimoto, H. Nakamura, K. Tachibana, D. Yamasaki, A. Ota et al., Sterol-mediated regulation of human lipin 1 gene expression in hepatoblastoma cells, J. Biol. Chem, vol.284, pp.22195-22205, 2009.

N. Jaber, Z. Dou, J. Chen, J. Catanzaro, Y. Jiang et al., Class III PI3K Vps34 plays an essential role in autophagy and in heart and liver function, Proc. Natl. Acad. Sci, vol.109, 2003.

J. P. Jackson, S. S. Ferguson, R. Moore, M. Negishi, and J. A. Goldstein, The Constitutive Active/Androstane Receptor Regulates Phenytoin Induction of Cyp2c29, Mol. Pharmacol, vol.65, pp.1397-1404, 2004.

J. P. Jackson, S. S. Ferguson, M. Negishi, and J. A. Goldstein, Phenytoin Induction of the Cyp2c37 Gene Is Mediated by the Constitutive Androstane Receptor, Drug Metab. Dispos, vol.34, 2003.

M. Jakubowski and M. Trzcinka-ochocka, Biological monitoring of exposure: trends and key developments, J. Occup. Health, vol.47, pp.22-48, 2005.

H. R. Jamshidi, M. H. Ghahremani, S. N. Ostad, M. Sharifzadeh, A. R. Dehpour et al., Effects of diazinon on the activity and gene expression of mitochondrial glutamate dehydrogenase from rat pancreatic Langerhans islets. Pestic, Biochem. Physiol, vol.93, pp.23-27, 2009.

S. M. Jandhyala, R. Talukdar, C. Subramanyam, H. Vuyyuru, M. Sasikala et al., Role of the normal gut microbiota, World J. Gastroenterol, vol.21, pp.8787-8803, 2015.

J. Jansson, S. Edén, I. , and O. , Sexual Dimorphism in the Control of Growth Hormone Secretion*, Endocr. Rev, vol.6, pp.128-150, 1985.

Y. Jin, X. Lin, W. Miao, T. Wu, H. Shen et al., Chronic exposure of mice to environmental endocrine-disrupting chemicals disturbs their energy metabolism, Toxicol. Lett, vol.225, pp.392-400, 2014.

H. Jinno, T. Tanaka-kagawa, N. Hanioka, S. Ishida, M. Saeki et al., Identification of Novel Alternative Splice Variants of Human Constitutive Androstane Receptor and Characterization of Their Expression in the Liver, Mol. Pharmacol, vol.65, pp.496-502, 2004.

S. Jitrapakdee, Transcription factors and coactivators controlling nutrient and hormonal regulation of hepatic gluconeogenesis, Int. J. Biochem. Cell Biol, vol.44, pp.33-45, 2012.

C. Joly, J. Gay-quéheillard, A. Léké, K. Chardon, S. Delanaud et al., Impact of chronic exposure to low doses of chlorpyrifos on the intestinal microbiota in the Simulator of the Human Intestinal Microbial Ecosystem (SHIME®) and in the rat, Environ. Sci. Pollut. Res, vol.20, pp.2726-2734, 2013.

M. W. Jones and J. G. Deppen, Physiology, Gallbladder, 2018.

K. Jungermann, Role of intralobular compartmentation in hepatic metabolism, Diabete Metab, vol.18, pp.81-86, 1992.

K. Jungermann and T. Keitzmann, Zonation of Parenchymal and Nonparenchymal Metabolism in Liver, Annu. Rev. Nutr, vol.16, pp.179-203, 1996.

D. Junquero and Y. Rival, Syndrome métabolique : quelle définition pour quel(s) traitement(s) ? Médecine/sciences 21, pp.1045-1053, 2005.

T. Kabashima, T. Kawaguchi, B. E. Wadzinski, and K. Uyeda, Xylulose 5-phosphate mediates glucoseinduced lipogenesis by xylulose 5-phosphate-activated protein phosphatase in rat liver, Proc. Natl. Acad. Sci, vol.100, pp.5107-5112, 2003.

S. Kakizaki, Y. Yamazaki, T. Kosone, N. Horiguchi, N. Horigichi et al., Gene expression profiles of drug-metabolizing enzymes and transporters with an overexpression of hepatocyte growth factor, Liver Int, vol.27, pp.109-119, 2007.

V. Kamath, R. , and P. , Altered glucose homeostasis and oxidative impairment in pancreas of rats subjected to dimethoate intoxication, Toxicology, vol.231, pp.137-146, 2007.

Y. Kamiyama, T. Matsubara, K. Yoshinari, K. Nagata, H. Kamimura et al., Role of human hepatocyte nuclear factor 4alpha in the expression of drug-metabolizing enzymes and transporters in human hepatocytes assessed by use of small interfering RNA, Drug Metab. Pharmacokinet, vol.22, pp.287-298, 2007.

S. Karami-mohajeri and M. Abdollahi, Toxic influence of organophosphate, carbamate, and organochlorine pesticides on cellular metabolism of lipids, proteins, and carbohydrates, Hum. Exp. Toxicol, vol.30, pp.1119-1140, 2011.

W. Karmaus, J. R. Osuch, I. Eneli, L. M. Mudd, J. Zhang et al., Maternal levels of dichlorodiphenyl-dichloroethylene (DDE) may increase weight and body mass index in adult female offspring, Occup. Environ. Med, vol.66, pp.143-149, 2009.

A. Kassam, C. J. Winrow, F. Fernandez-rachubinski, J. P. Capone, and R. A. Rachubinski, The peroxisome proliferator response element of the gene encoding the peroxisomal beta-oxidation enzyme enoyl-CoA hydratase/3-hydroxyacyl-CoA dehydrogenase is a target for constitutive androstane receptor beta/9-cis-retinoic acid receptor-mediated transac, J. Biol. Chem, vol.275, pp.4345-4350, 2000.

H. R. Kast, B. Goodwin, P. T. Tarr, S. A. Jones, A. M. Anisfeld et al., Regulation of Multidrug Resistance-associated Protein 2 (ABCC2) by the Nuclear Receptors Pregnane X Receptor, Farnesoid X-activated Receptor, and Constitutive Androstane Receptor, J. Biol. Chem, vol.277, pp.2908-2915, 2002.

T. Kawaguchi, M. Takenoshita, T. Kabashima, and K. Uyeda, Glucose and cAMP regulate the L-type pyruvate kinase gene by phosphorylation/dephosphorylation of the carbohydrate response element binding protein, Proc. Natl. Acad. Sci, vol.98, pp.13710-13715, 2001.

T. Kawamoto, T. Sueyoshi, I. Zelko, R. Moore, K. Washburn et al., Phenobarbital-responsive nuclear translocation of the receptor CAR in induction of the CYP2B gene, Mol. Cell. Biol, vol.19, pp.6318-6322, 1999.

T. Kawamoto, S. Kakizaki, K. Yoshinari, and M. Negishi, Estrogen Activation of the Nuclear Orphan Receptor CAR (Constitutive Active Receptor) in Induction of the Mouse Cyp2b10 Gene, Mol. Endocrinol, vol.14, pp.1897-1905, 2000.

T. Kawamoto, S. Kakizaki, K. Yoshinari, and M. Negishi, Estrogen activation of the nuclear orphan receptor CAR (constitutive active receptor) in induction of the mouse Cyp2b10 gene, Mol. Endocrinol, vol.14, pp.1897-1905, 2000.

N. L. Keating, A. J. O'malley, S. J. Freedland, and M. R. Smith, Diabetes and cardiovascular disease during androgen deprivation therapy: observational study of veterans with prostate cancer, J. Natl. Cancer Inst, vol.104, pp.1518-1523, 2012.

E. Kesse-guyot, J. Baudry, K. E. Assmann, P. Galan, S. Hercberg et al., Prospective association between consumption frequency of organic food and body weight change, risk of overweight or obesity: results from the NutriNet-Santé Study, Br. J. Nutr, vol.117, pp.325-334, 2017.

F. Kiernan, The Anatomy and Physiology of the Liver, Philos. Trans. R. Soc. London, vol.123, pp.711-770, 1833.

K. H. Kim and M. Lee, Autophagy-a key player in cellular and body metabolism, Nat. Rev. Endocrinol, vol.10, pp.322-337, 2014.

J. Kim, Y. Park, K. S. Yoon, J. M. Clark, and Y. Park, Imidacloprid, a neonicotinoid insecticide, induces insulin resistance, J. Toxicol. Sci, vol.38, pp.655-660, 2013.

J. Kim, Y. Park, K. S. Yoon, J. M. Clark, and Y. Park, Permethrin Alters Adipogenesis in 3T3-L1 Adipocytes and Causes Insulin Resistance in C2C12 Myotubes, J. Biochem. Mol. Toxicol, vol.28, pp.418-424, 2014.

J. K. Kim, J. J. Fillmore, Y. Chen, C. Yu, I. K. Moore et al., Tissue-specific overexpression of lipoprotein lipase causes tissue-specific insulin resistance, Proc. Natl. Acad. Sci. U. S. A, vol.98, pp.7522-7527, 2001.

C. D. Klaassen, L. Liu, and R. T. Dunn, Regulation of sulfotransferase mRNA expression in male and female rats of various ages, Chem. Biol. Interact, vol.109, pp.299-313, 1998.

S. A. Kliewer, K. Umesono, D. J. Mangelsdorf, and R. M. Evans, Retinoid X receptor interacts with nuclear receptors in retinoic acid, thyroid hormone and vitamin D3 signalling, Nature, vol.355, pp.446-449, 1992.

Y. Ko, T. Wong, Y. Hsu, K. Kuo, Y. et al., The Correlation Between Body Fat, Visceral Fat, and Nonalcoholic Fatty Liver Disease, Metab. Syndr. Relat. Disord, vol.15, pp.304-311, 2017.

K. Kobayashi, T. Sueyoshi, K. Inoue, R. Moore, and M. Negishi, Cytoplasmic accumulation of the nuclear receptor CAR by a tetratricopeptide repeat protein in HepG2 cells, Mol. Pharmacol, vol.64, pp.1069-1075, 2003.

S. Kodama, C. Koike, M. Negishi, and Y. Yamamoto, Nuclear receptors CAR and PXR cross talk with FOXO1 to regulate genes that encode drug-metabolizing and gluconeogenic enzymes, Mol. Cell. Biol, vol.24, pp.7931-7940, 2004.

S. Kodama, R. Moore, Y. Yamamoto, and M. Negishi, Human nuclear pregnane X receptor cross-talk with CREB to repress cAMP activation of the glucose-6-phosphatase gene, Biochem. J, vol.407, pp.373-381, 2007.

K. H. Koh, S. Jurkovic, K. Yang, S. Choi, J. W. Jung et al., Estradiol induces cytochrome P450 2B6 expression at high concentrations: implication in estrogen-mediated gene regulation in pregnancy, Biochem. Pharmacol, vol.84, pp.93-103, 2012.

K. Kohalmy, V. Tamási, L. Kóbori, E. Sárváry, J. Pascussi et al., Dehydroepiandrosterone induces human CYP2B6 through the constitutive androstane receptor, Drug Metab. Dispos, vol.35, pp.1495-1501, 2007.

C. Koike, R. Moore, and M. Negishi, Extracellular signal-regulated kinase is an endogenous signal retaining the nuclear constitutive active/androstane receptor (CAR) in the cytoplasm of mouse primary hepatocytes, Mol. Pharmacol, vol.71, pp.1217-1221, 2007.

H. Kojima, F. Sata, S. Takeuchi, T. Sueyoshi, and T. Nagai, Comparative study of human and mouse pregnane X receptor agonistic activity in 200 pesticides using in vitro reporter gene assays, Toxicology, vol.280, pp.77-87, 2011.

K. Kotani, K. Tokunaga, S. Fujioka, T. Kobatake, Y. Keno et al., Sexual dimorphism of age-related changes in whole-body fat distribution in the obese, Int. J. Obes. Relat. Metab. Disord, vol.18, pp.207-209, 1994.

G. G. Kuiper, J. G. Lemmen, B. Carlsson, J. C. Corton, S. H. Safe et al., Interaction of Estrogenic Chemicals and Phytoestrogens with Estrogen Receptor ?, Endocrinology, vol.139, pp.4252-4263, 1998.

J. T. Lahtela, J. Arranto, and E. Sotaniemi, Enzyme inducers improve insulin sensitivity in non-insulindependent diabetic subjects, Diabetes, vol.34, pp.911-916, 1985.

K. K. Lai, D. Kolippakkam, and L. Beretta, Comprehensive and quantitative proteome profiling of the mouse liver and plasma, Hepatology, vol.47, pp.1043-1051, 2008.

B. G. Lake, R. J. Price, and T. G. Osimitz, Mode of action analysis for pesticide-induced rodent liver tumours involving activation of the constitutive androstane receptor: relevance to human cancer risk, Pest Manag. Sci, vol.71, pp.829-834, 2015.

D. W. Lam and D. Leroith, Metabolic Syndrome, 2000.

V. Lamba, K. Yasuda, J. K. Lamba, M. Assem, J. Davila et al., PXR (NR1I2): splice variants in human tissues, including brain, and identification of neurosteroids and nicotine as PXR activators, Toxicol. Appl. Pharmacol, vol.199, pp.251-265, 2004.

J. E. Lambert, M. A. Ramos-roman, J. D. Browning, and E. J. Parks, Increased de novo lipogenesis is a distinct characteristic of individuals with nonalcoholic fatty liver disease, Gastroenterology, vol.146, pp.726-735, 2014.

M. M. Lasram, I. B. Dhouib, A. Annabi, S. El-fazaa, and N. Gharbi, A review on the molecular mechanisms involved in insulin resistance induced by organophosphorus pesticides, Toxicology, vol.322, pp.1-13, 2014.

M. M. Lasram, I. B. Dhouib, K. Bouzid, A. J. Lamine, A. Annabi et al., Association of inflammatory response and oxidative injury in the pathogenesis of liver steatosis and insulin resistance following subchronic exposure to malathion in rats, Environ. Toxicol. Pharmacol, vol.38, pp.542-553, 2014.
URL : https://hal.archives-ouvertes.fr/pasteur-01061164

T. L. Lassiter and S. Brimijoin, Rats gain excess weight after developmental exposure to the organophosphorothionate pesticide, chlorpyrifos, Neurotoxicol. Teratol, vol.30, pp.125-130, 2008.

T. L. Lassiter, I. T. Ryde, E. A. Mackillop, K. K. Brown, E. D. Levin et al., Exposure of Neonatal Rats to Parathion Elicits Sex-Selective Reprogramming of Metabolism and Alters the Response to a High-Fat Diet in Adulthood, Environ. Health Perspect, vol.116, pp.1456-1462, 2008.

E. M. Laurenzana, T. Chen, M. Kannuswamy, B. E. Sell, S. C. Strom et al., The orphan nuclear receptor DAX-1 functions as a potent corepressor of the constitutive androstane receptor (NR1I3), Mol. Pharmacol, vol.82, pp.918-928, 2012.

E. M. Laurenzana, D. M. Coslo, M. V. Vigilar, A. M. Roman, and C. J. Omiecinski, Activation of the Constitutive Androstane Receptor by Monophthalates, Chem. Res. Toxicol, vol.29, pp.1651-1661, 2016.

M. J. Lebaron, R. J. Rasoulpour, B. B. Gollapudi, R. Sura, H. L. Kan et al., Characterization of Nuclear Receptor-Mediated Murine Hepatocarcinogenesis of the Herbicide Pronamide and Its Human Relevance, Toxicol. Sci, vol.142, pp.74-92, 2014.

G. M. Ledda-columbano, M. Pibiri, D. Concas, F. Molotzu, G. Simbula et al., Sex difference in the proliferative response of mouse hepatocytes to treatment with the CAR ligand, TCPOBOP. Carcinogenesis, vol.24, pp.1059-1065, 2003.

A. J. Lee, M. X. Cai, P. E. Thomas, A. H. Conney, and B. T. Zhu, Characterization of the oxidative metabolites of 17beta-estradiol and estrone formed by 15 selectively expressed human cytochrome p450 isoforms, Endocrinology, vol.144, pp.3382-3398, 2003.

D. Lee, M. W. Steffes, A. Sjödin, R. S. Jones, L. L. Needham et al., Low Dose of Some Persistent Organic Pollutants Predicts Type 2 Diabetes: A Nested Case-Control Study, Environ. Health Perspect, vol.118, pp.1235-1242, 2010.

M. Lee, J. Hwang, H. Lee, S. Jung, I. Kang et al., AMP-activated protein kinase activity is critical for hypoxia-inducible factor-1 transcriptional activity and its target gene expression under hypoxic conditions in DU145 cells, J. Biol. Chem, vol.278, pp.39653-39661, 2003.

Y. Lee, S. H. Kim, S. Kim, H. Kwon, J. Kim et al., Sex-specific metabolic interactions between liver and adipose tissue in MCD diet-induced non-alcoholic fatty liver disease, Oncotarget, vol.7, pp.46959-46971, 2016.

G. Lemaire, W. Mnif, J. Pascussi, A. Pillon, F. Rabenoelina et al., Identification of New Human Pregnane X Receptor Ligands among Pesticides Using a Stable Reporter Cell System, Toxicol. Sci, vol.91, pp.501-509, 2006.

U. Lemke, A. Krones-herzig, M. Diaz, P. Narvekar, A. Ziegler et al., The glucocorticoid receptor controls hepatic dyslipidemia through Hes1, Cell Metab, vol.8, pp.212-223, 2008.

H. Lempiäinen, F. Molnár, M. Macias-gonzalez, M. Peräkylä, and C. Carlberg, Antagonist-and inverse agonist-driven interactions of the vitamin D receptor and the constitutive androstane receptor with corepressor protein, Mol. Endocrinol, vol.19, pp.2258-2272, 2005.

S. Lenzen, A Fresh View of Glycolysis and Glucokinase Regulation: History and Current Status, J. Biol. Chem, vol.289, pp.12189-12194, 2014.

D. F. Lewis, M. N. Jacobs, and M. Dickins, Compound lipophilicity for substrate binding to human P450s in drug metabolism, Drug Discov. Today, vol.9, pp.530-537, 2004.

M. S. Lewitt and K. Brismar, Gender difference in the leptin response to feeding in peroxisomeproliferator-activated receptor-alpha knockout mice, Int. J. Obes. Relat. Metab. Disord, vol.26, pp.1296-1300, 2002.

R. E. Ley, P. J. Turnbaugh, S. Klein, G. , and J. I. , Microbial ecology: Human gut microbes associated with obesity, Nature, vol.444, pp.1022-1023, 2006.

J. Z. Li, Y. Huang, R. Karaman, P. T. Ivanova, H. A. Brown et al., Chronic overexpression of PNPLA3I148M in mouse liver causes hepatic steatosis, J. Clin. Invest, vol.122, pp.4130-4144, 2012.

L. Li, T. Chen, J. D. Stanton, T. Sueyoshi, M. Negishi et al., The peripheral benzodiazepine receptor ligand 1-(2-chlorophenyl-methylpropyl)-3-isoquinoline-carboxamide is a novel antagonist of human constitutive androstane receptor, Mol. Pharmacol, vol.74, pp.443-453, 2008.

L. Li, X. Bao, Q. Zhang, M. Negishi, and X. Ding, Role of CYP2B in Phenobarbital-Induced Hepatocyte Proliferation in Mice, Drug Metab. Dispos, vol.45, pp.977-981, 2017.

M. V. Li, W. Chen, R. N. Harmancey, A. M. Nuotio-antar, M. Imamura et al., Glucose-6-phosphate mediates activation of the carbohydrate responsive binding protein (ChREBP), 2010.

, Biochem. Biophys. Res. Commun, vol.395, pp.395-400

X. Li, H. T. Pham, A. S. Janesick, and B. Blumberg, Triflumizole is an obesogen in mice that acts through peroxisome proliferator activated receptor gamma (PPAR?), Environ. Health Perspect, vol.120, pp.1720-1726, 2012.

Y. Li, S. Xu, M. M. Mihaylova, B. Zheng, X. Hou et al., , 2011.

, AMPK Phosphorylates and Inhibits SREBP Activity to Attenuate Hepatic Steatosis and Atherosclerosis in DietInduced Insulin-Resistant Mice, Cell Metab, vol.13, pp.376-388

S. Lim, S. Y. Ahn, I. C. Song, M. H. Chung, H. C. Jang et al., Chronic exposure to the herbicide, atrazine, causes mitochondrial dysfunction and insulin resistance, PLoS One, vol.4, p.5186, 2009.

L. O. Lima, S. Almeida, M. H. Hutz, and M. Fiegenbaum, PPARA, RXRA, NR1I2 and NR1I3 gene polymorphisms and lipid and lipoprotein levels in a Southern Brazilian population, Mol. Biol. Rep, vol.40, pp.1241-1247, 2013.

H. Lin, I. Yu, R. Wang, Y. Chen, N. Liu et al., Increased hepatic steatosis and insulin resistance in mice lacking hepatic androgen receptor, Hepatology, vol.47, pp.1924-1935, 2008.

Y. Liu, R. S. Heymann, F. Moatamed, J. J. Schultz, D. Sobel et al., A mutant thyroid hormone receptor alpha antagonizes peroxisome proliferator-activated receptor alpha signaling in vivo and impairs fatty acid oxidation, Endocrinology, vol.148, pp.1206-1217, 2007.

F. Lizcano and G. Guzmán, Estrogen Deficiency and the Origin of Obesity during Menopause, Biomed Res. Int, p.757461, 2014.

C. Loewenherz, R. A. Fenske, N. J. Simcox, G. Bellamy, K. et al., Biological monitoring of organophosphorus pesticide exposure among children of agricultural workers in central Washington State, Environ. Health Perspect, vol.105, pp.1344-1353, 1997.

A. Lonardo, C. Carani, N. Carulli, L. , and P. , Endocrine NAFLD" a hormonocentric perspective of nonalcoholic fatty liver disease pathogenesis, J. Hepatol, vol.44, pp.1196-1207, 2006.

A. Lonardo, S. Bellentani, C. K. Argo, S. Ballestri, C. D. Byrne et al., Epidemiological modifiers of non-alcoholic fatty liver disease: Focus on high-risk groups, Dig. Liver Dis, vol.47, pp.997-1006, 2015.

J. A. López-velázquez, L. D. Carrillo-córdova, N. C. Chávez-tapia, M. Uribe, and N. Méndez-sánchez, Nuclear Receptors in Nonalcoholic Fatty Liver Disease, J. Lipids, vol.2012, pp.1-10, 2012.

C. Lu, K. Toepel, R. Irish, R. A. Fenske, D. B. Barr et al., Organic diets significantly lower children's dietary exposure to organophosphorus pesticides, Environ. Health Perspect, vol.114, pp.260-263, 2006.

M. Lu, J. Du, P. Zhou, H. Chen, C. Lu et al., Endocrine disrupting potential of fipronil and its metabolite in reporter gene assays, Chemosphere, vol.120, pp.246-251, 2015.

Y. Lu, T. Jin, Y. Xu, D. Zhang, Q. Wu et al., Sex Differences in the Circadian Variation of Cytochrome P450 Genes and Corresponding Nuclear Receptors in Mouse Liver, Chronobiol. Int, vol.30, pp.1135-1143, 2013.

L. Lundholm, H. Zang, A. L. Hirschberg, J. Gustafsson, P. Arner et al., Key lipogenic gene expression can be decreased by estrogen in human adipose tissue, Fertil. Steril, vol.90, pp.44-48, 2008.

A. L. Luz, C. D. Kassotis, H. M. Stapleton, and J. N. Meyer, The high-production volume fungicide pyraclostrobin induces triglyceride accumulation associated with mitochondrial dysfunction, and promotes adipocyte differentiation independent of PPAR? activation, 3T3-L1 cells, vol.393, pp.150-159, 2018.

C. Lynch, Y. Pan, L. Li, S. Heyward, T. Moeller et al., Activation of the constitutive androstane receptor inhibits gluconeogenesis without affecting lipogenesis or fatty acid synthesis in human hepatocytes, Toxicol. Appl. Pharmacol, vol.279, pp.33-42, 2014.

L. Ma, N. G. Tsatsos, and H. C. Towle, Direct role of ChREBP.Mlx in regulating hepatic glucose-responsive genes, J. Biol. Chem, vol.280, pp.12019-12027, 2005.

A. Mabuchi, I. Mullaney, P. W. Sheard, P. A. Hessian, B. L. Mallard et al., Role of hepatic stellate cell/hepatocyte interaction and activation of hepatic stellate cells in the early phase of liver regeneration in the rat, J. Hepatol, vol.40, pp.910-916, 2004.

D. L. Macintosh, C. W. Kabiru, and P. B. Ryan, Longitudinal investigation of dietary exposure to selected pesticides, Environ. Health Perspect, vol.109, pp.145-150, 2001.

J. N. Macleod, N. A. Pampori, and B. H. Shapiro, Sex differences in the ultradian pattern of plasma growth hormone concentrations in mice, J. Endocrinol, vol.131, pp.395-399, 1991.

R. M. Macsween, A. D. Burt, B. C. Portmann, K. G. Ishak, P. J. Scheurer et al., Pathology of the liver, vol.29, pp.43-43, 2003.

J. M. Maglich, J. Watson, P. J. Mcmillen, B. Goodwin, T. M. Willson et al., The nuclear receptor CAR is a regulator of thyroid hormone metabolism during caloric restriction, J. Biol. Chem, vol.279, pp.19832-19838, 2004.

J. M. Maglich, D. C. Lobe, and J. T. Moore, The nuclear receptor CAR (NR1I3) regulates serum triglyceride levels under conditions of metabolic stress, J. Lipid Res, vol.50, pp.439-445, 2009.

H. Mahdessian, A. Taxiarchis, S. Popov, A. Silveira, A. Franco-cereceda et al.,

F. Hooft, TM6SF2 is a regulator of liver fat metabolism influencing triglyceride secretion and hepatic lipid droplet content, Proc. Natl. Acad. Sci, vol.111, pp.8913-8918, 2014.

C. Malaplate-armand, L. Ferrari, C. Masson, S. Visvikis-siest, H. Lambert et al., Downregulation of astroglial CYP2C, glucocorticoid receptor and constitutive androstane receptor genes in response to cocaine in human U373 MG astrocytoma cells, Toxicol. Lett, vol.159, pp.203-211, 2005.
URL : https://hal.archives-ouvertes.fr/hal-01999436

D. E. Malarkey, K. Johnson, L. Ryan, G. Boorman, and R. R. Maronpot, New Insights into Functional Aspects of Liver Morphology, Toxicol. Pathol, vol.33, pp.27-34, 2005.

F. P. Mall, A study of the structural unit of the liver, Am. J. Anat, vol.5, pp.227-308, 1906.

D. J. Mangelsdorf, C. Thummel, M. Beato, P. Herrlich, G. Schütz et al., The nuclear receptor superfamily: the second decade, Cell, vol.83, pp.835-839, 1995.

L. H. Mangum, G. E. Howell, and J. E. Chambers, Exposure to p,p'-DDE enhances differentiation of 3T3-L1 preadipocytes in a model of sub-optimal differentiation, Toxicol. Lett, vol.238, pp.65-71, 2015.

I. J. Marie, C. Dalet, J. M. Blanchard, C. Astre, A. Szawlowski et al., Inhibition of cytochrome P-450p (P450IIIA1) gene expression during liver regeneration from two-thirds hepatectomy in the rat, Biochem. Pharmacol, vol.37, pp.3515-3521, 1988.

K. Marinou, M. Adiels, L. Hodson, K. N. Frayn, F. Karpe et al., Young women partition fatty acids towards ketone body production rather than VLDL-TAG synthesis, compared with young men, Br. J. Nutr, vol.105, pp.857-865, 2011.

S. Martin and R. G. Parton, Lipid droplets: a unified view of a dynamic organelle, Nat. Rev. Mol. Cell Biol, vol.7, pp.373-378, 2006.

C. N. Martini, M. Gabrielli, J. N. Brandani, and M. D. Vila, Glyphosate Inhibits PPAR Gamma Induction and Differentiation of Preadipocytes and is able to Induce Oxidative Stress, J. Biochem. Mol. Toxicol, vol.30, pp.404-413, 2016.

D. Masson, M. Qatanani, A. L. Sberna, R. Xiao, J. P. Pais-de-barros et al., Activation of the constitutive androstane receptor decreases HDL in wild-type and human apoA-I transgenic mice, J. Lipid Res, vol.49, pp.1682-1691, 2008.
URL : https://hal.archives-ouvertes.fr/hal-01958844

F. Mastropasqua, G. Girolimetti, and M. Shoshan, PGC1?: Friend or Foe in Cancer? Genes (Basel), vol.9, p.48, 2018.

H. Masuyama, H. Nakatsukasa, and Y. Hiramatsu, Effect of oncostatin M on uridine diphosphate-5'-glucuronosyltransferase 1A1 through cross talk with constitutive androstane receptor, Mol. Endocrinol, vol.24, pp.745-753, 2010.

H. Masuzaki, J. Paterson, H. Shinyama, N. M. Morton, J. J. Mullins et al., A Transgenic Model of Visceral Obesity and the Metabolic Syndrome, Science, vol.294, pp.2166-2170, 2001.

M. Matsumoto, A. Pocai, L. Rossetti, R. A. Depinho, A. et al., Impaired Regulation of Hepatic Glucose Production in Mice Lacking the Forkhead Transcription Factor Foxo1 in Liver, Cell Metab, vol.6, pp.208-216, 2007.

T. Matsuzaka, H. Shimano, N. Yahagi, M. Amemiya-kudo, T. Yoshikawa et al., Dual regulation of mouse Delta(5)-and Delta(6)-desaturase gene expression by SREBP-1 and PPARalpha, J. Lipid Res, vol.43, pp.107-114, 2002.

N. J. Mckenna, A. J. Cooney, F. J. Demayo, M. Downes, C. K. Glass et al., Minireview: Evolution of NURSA, the Nuclear Receptor Signaling Atlas, vol.23, pp.740-746, 2009.

S. Medjakovic, A. Zoechling, P. Gerster, M. M. Ivanova, Y. Teng et al., Effect of nonpersistent pesticides on estrogen receptor, androgen receptor, and aryl hydrocarbon receptor, Environ. Toxicol, vol.29, pp.1201-1216, 2014.

R. C. Meex and M. J. Watt, Hepatokines: linking nonalcoholic fatty liver disease and insulin resistance, Nat. Rev. Endocrinol, vol.13, pp.509-520, 2017.

M. A. Mendez, R. Garcia-esteban, M. Guxens, M. Vrijheid, M. Kogevinas et al.,

, Prenatal Organochlorine Compound Exposure, Rapid Weight Gain, and Overweight in Infancy, Environ. Health Perspect, vol.119, pp.272-278

G. Merla, C. Howald, S. E. Antonarakis, and A. Reymond, The subcellular localization of the ChoREbinding protein, encoded by the Williams-Beuren syndrome critical region gene 14, Hum. Mol. Genet, vol.13, pp.1505-1514, 2004.

J. Miao, S. Fang, Y. Bae, and J. K. Kemper, Functional inhibitory cross-talk between constitutive androstane receptor and hepatic nuclear factor-4 in hepatic lipid/glucose metabolism is mediated by competition for binding to the DR1 motif and to the common coactivators, GRIP-1 and PGC-1alpha, J. Biol. Chem, vol.281, pp.14537-14546, 2006.

G. Min, Estrogen modulates transactivations of SXR-mediated liver X receptor response element and CARmediated phenobarbital response element in HepG2 cells, Exp. Mol. Med, vol.42, pp.731-738, 2010.

G. Min, J. K. Kemper, and B. Kemper, Glucocorticoid receptor-interacting protein 1 mediates ligandindependent nuclear translocation and activation of constitutive androstane receptor in vivo, J. Biol. Chem, vol.277, pp.26356-26363, 2002.

G. Min, H. Kim, Y. Bae, L. Petz, and J. K. Kemper, Inhibitory cross-talk between estrogen receptor (ER) and constitutively activated androstane receptor (CAR). CAR inhibits ER-mediated signaling pathway by squelching p160 coactivators, J. Biol. Chem, vol.277, pp.34626-34633, 2002.

J. O. Miners, J. Attwood, and D. J. Birkett, Influence of sex and oral contraceptive steroids on paracetamol metabolism, Br. J. Clin. Pharmacol, vol.16, pp.503-509, 1983.

M. A. Mitsche, H. H. Hobbs, and J. C. Cohen, Patatin-like phospholipase domain-containing protein 3 promotes transfers of essential fatty acids from triglycerides to phospholipids in hepatic lipid droplets, J. Biol. Chem, vol.293, pp.6958-6968, 2018.

Z. Mokhtari, D. L. Gibson, and A. Hekmatdoost, Nonalcoholic Fatty Liver Disease, the Gut Microbiome, and Diet, Adv. Nutr. An Int. Rev. J, vol.8, pp.240-252, 2017.

A. Montagner, A. Polizzi, E. Fouché, S. Ducheix, Y. Lippi et al., Liver PPAR? is crucial for whole-body fatty acid homeostasis and is protective against NAFLD, Gut, vol.65, pp.1202-1214, 2016.

A. Montagner, A. Korecka, A. Polizzi, Y. Lippi, Y. Blum et al., Hepatic circadian clock oscillators and nuclear receptors integrate microbiome-derived signals, Sci. Rep, vol.6, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01482816

M. P. Montgomery, F. Kamel, T. M. Saldana, M. C. Alavanja, and D. P. Sandler, Incident diabetes and pesticide exposure among licensed pesticide applicators: Agricultural Health Study, Am. J. Epidemiol, vol.167, pp.1235-1246, 1993.

G. A. Montoya, V. Strauss, E. Fabian, H. Kamp, W. Mellert et al., Mechanistic analysis of metabolomics patterns in rat plasma during administration of direct thyroid hormone synthesis inhibitors or compounds increasing thyroid hormone clearance, Toxicol. Lett, vol.225, pp.240-251, 2014.

Y. Moon, G. Liang, X. Xie, M. Frank-kamenetsky, K. Fitzgerald et al., The Scap/SREBP Pathway Is Essential for Developing Diabetic Fatty Liver and Carbohydrate-Induced Hypertriglyceridemia in Animals, Cell Metab, vol.15, pp.240-246, 2012.

D. D. Moore, P. Wei, J. Zhang, M. Egan-hafley, and S. Liang, The nuclear receptor CAR mediates specific xenobiotic induction of drug metabolism, Nature, vol.407, pp.920-923, 2000.

L. B. Moore, D. J. Parks, S. A. Jones, R. K. Bledsoe, T. G. Consler et al., Orphan nuclear receptors constitutive androstane receptor and pregnane X receptor share xenobiotic and steroid ligands, J. Biol. Chem, vol.275, pp.15122-15127, 2000.

M. J. Moreno-aliaga and F. Matsumura, Endrin inhibits adipocyte differentiation by selectively altering expression pattern of CCAAT/enhancer binding protein-alpha in 3T3-L1 cells, Mol. Pharmacol, vol.56, pp.91-101, 1999.

P. Morigny, M. Houssier, E. Mouisel, and D. Langin, Adipocyte lipolysis and insulin resistance, Biochimie, vol.125, pp.259-266, 2016.

S. Mostafalou, Persistent Organic Pollutants and Concern Over the Link with Insulin Resistance Related Metabolic Diseases, Reviews of Environmental Contamination and Toxicology, pp.69-89, 2016.

M. Mouzaki and J. P. Allard, The Role of Nutrients in the Development, Progression, and Treatment of Nonalcoholic Fatty Liver Disease, J. Clin. Gastroenterol, vol.46, pp.457-467, 2012.

M. Mouzaki, E. M. Comelli, B. M. Arendt, J. Bonengel, S. K. Fung et al., Intestinal microbiota in patients with nonalcoholic fatty liver disease, Hepatology, vol.58, pp.120-127, 2013.

M. Mueckler and B. Thorens, The SLC2 (GLUT) family of membrane transporters, Mol. Aspects Med, vol.34, pp.121-138, 2013.

N. T. Mueller, M. A. Pereira, E. W. Demerath, J. G. Dreyfus, R. F. Maclehose et al., Earlier menarche is associated with fatty liver and abdominal ectopic fat in midlife, independent of young adult BMI: The CARDIA study, Obesity, vol.23, pp.468-474, 2015.

M. Murray, E. Fiala-beer, and D. Sutton, Upregulation of cytochromes P 450 2B in rat liver by orphenadrine, Br. J. Pharmacol, vol.139, pp.787-796, 2003.

E. Mutel, A. Abdul-wahed, N. Ramamonjisoa, A. Stefanutti, I. Houberdon et al., Targeted deletion of liver glucose-6 phosphatase mimics glycogen storage disease type 1a including development of multiple adenomas, J. Hepatol, vol.54, pp.529-537, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00575314

S. Mutoh, M. Osabe, K. Inoue, R. Moore, L. Pedersen et al., Dephosphorylation of threonine 38 is required for nuclear translocation and activation of human xenobiotic receptor CAR (NR1I3), J. Biol. Chem, vol.284, pp.34785-34792, 2009.

S. Mutoh, M. Sobhany, R. Moore, L. Perera, L. Pedersen et al., Phenobarbital indirectly activates the constitutive active androstane receptor (CAR) by inhibition of epidermal growth factor receptor signaling, Sci. Signal, vol.6, p.31, 2013.

O. J. Naderer, R. E. Dupuis, E. L. Heinzen, K. Wiwattanawongsa, M. W. Johnson et al., The influence of norfloxacin and metronidazole on the disposition of mycophenolate mofetil, J. Clin. Pharmacol, vol.45, pp.219-226, 2005.

R. Nagaraju, A. K. Joshi, and P. S. Rajini, Organophosphorus insecticide, monocrotophos, possesses the propensity to induce insulin resistance in rats on chronic exposure, J. Diabetes, vol.7, pp.47-59, 2015.

K. Nakamura, R. Moore, M. Negishi, and T. Sueyoshi, Nuclear Pregnane X Receptor Cross-talk with FoxA2 to Mediate Drug-induced Regulation of Lipid Metabolism in Fasting Mouse Liver, J. Biol. Chem, vol.282, pp.9768-9776, 2007.

K. Nakayama, Y. Sudo, Y. Sasaki, H. Iwata, M. Takahashi et al., Studies on Transcriptional Regulation of Cyp3a16 Gene in Mouse Livers by Application of Direct DNA Injection Method, Biochem. Biophys. Res. Commun, vol.287, pp.820-824, 2001.

D. W. Nebert, A. L. Roe, M. Z. Dieter, W. A. Solis, Y. Yang et al., Role of the aromatic hydrocarbon receptor and [Ah] gene battery in the oxidative stress response, cell cycle control, and apoptosis, Biochem. Pharmacol, vol.59, pp.65-85, 2000.

B. A. Neel and R. M. Sargis, The paradox of progress: environmental disruption of metabolism and the diabetes epidemic, Diabetes, vol.60, pp.1838-1848, 2011.

D. R. Nelson, L. Koymans, T. Kamataki, J. J. Stegeman, R. Feyereisen et al., P450 superfamily: update on new sequences, gene mapping, accession numbers and nomenclature, Pharmacogenetics, vol.6, pp.1-42, 1996.

L. K. Nieman, C. Turner, and M. L. , Addison's disease, Clin. Dermatol, vol.24, pp.276-280, 2006.

L. Oates, M. Cohen, L. Braun, A. Schembri, and R. Taskova, Reduction in urinary organophosphate pesticide metabolites in adults after a week-long organic diet, Environ. Res, vol.132, pp.105-111, 2014.

H. Ooe, J. Kon, H. Oshima, and T. Mitaka, Thyroid hormone is necessary for expression of constitutive androstane receptor in rat hepatocytes, Drug Metab. Dispos, vol.37, pp.1963-1969, 2009.

C. Opherk, F. Tronche, C. Kellendonk, D. Kohlmüller, A. Schulze et al., Inactivation of the Glucocorticoid Receptor in Hepatocytes Leads to Fasting Hypoglycemia and Ameliorates Hyperglycemia in Streptozotocin-Induced Diabetes Mellitus, Mol. Endocrinol, vol.18, pp.1346-1353, 2004.

U. C. Oppermann and E. Maser, Molecular and structural aspects of xenobiotic carbonyl metabolizing enzymes. Role of reductases and dehydrogenases in xenobiotic phase I reactions, Toxicology, vol.144, pp.71-81, 2000.

M. Osabe, J. Sugatani, A. Takemura, M. Kurosawa, Y. Yamazaki et al., Up-regulation of CAR expression through Elk-1 in HepG2 and SW480 cells by serum starvation stress, FEBS Lett, vol.583, pp.885-889, 2009.

K. Oshida, N. Vasani, C. Jones, T. Moore, S. Hester et al., Identification of chemical modulators of the constitutive activated receptor (CAR) in a gene expression compendium, Nucl. Recept. Signal, vol.13, p.2, 2015.

H. Ozen, Glycogen storage diseases: new perspectives, World J. Gastroenterol, vol.13, pp.2541-2553, 2007.

M. Y. Pakharukova, M. A. Smetanina, V. I. Kaledin, V. F. Kobzev, I. V. Romanova et al., Activation of constitutive androstane receptor under the effect of hepatocarcinogenic aminoazo dyes in mouse and rat liver, Bull. Exp. Biol. Med, vol.144, pp.338-341, 2007.

P. Panahi, S. Vosough-ghanbari, S. Pournourmohammadi, S. N. Ostad, S. Nikfar et al., Stimulatory effects of malathion on the key enzymes activities of insulin secretion in langerhans islets, glutamate dehydrogenase and glucokinase, Toxicol. Mech. Methods, vol.16, pp.161-167, 2006.

P. Panahi, S. Vosough-ghanbari, S. Pournourmohammadi, S. N. Ostad, S. Nikfar et al., Stimulatory effects of malathion on the key enzymes activities of insulin secretion in langerhans islets, glutamate dehydrogenase and glucokinase, Toxicol. Mech. Methods, vol.16, pp.161-167, 2006.

C. Pantoja, J. T. Huff, and K. R. Yamamoto, Glucocorticoid signaling defines a novel commitment state during adipogenesis in vitro, Mol. Biol. Cell, vol.19, pp.4032-4041, 2008.

E. A. Park, S. Song, C. Vinson, and W. J. Roesler, Role of CCAAT enhancer-binding protein beta in the thyroid hormone and cAMP induction of phosphoenolpyruvate carboxykinase gene transcription, J. Biol. Chem, vol.274, pp.211-217, 1999.

S. Park, C. A. Wiwi, and D. J. Waxman, Signalling cross-talk between hepatocyte nuclear factor 4? and growth-hormone-activated STAT5b, Biochem. J, vol.397, pp.159-168, 2006.

Y. Park, Y. Kim, J. Kim, K. S. Yoon, J. Clark et al., Imidacloprid, a Neonicotinoid Insecticide, Potentiates Adipogenesis in 3T3-L1 Adipocytes, J. Agric. Food Chem, vol.61, pp.255-259, 2013.

Y. J. Park, E. K. Lee, Y. K. Lee, D. J. Park, H. C. Jang et al., Opposing regulation of cytochrome P450 expression by CAR and PXR in hypothyroid mice, Toxicol. Appl. Pharmacol, vol.263, pp.131-137, 2012.

M. Pascual, M. J. Gomez-lechon, J. V. Castell, and R. Jover, ATF5 Is a Highly Abundant Liver-Enriched Transcription Factor that Cooperates with Constitutive Androstane Receptor in the Transactivation of CYP2B6: Implications in Hepatic Stress Responses, Drug Metab. Dispos, vol.36, pp.1063-1072, 2008.

J. M. Pascussi, S. Gerbal-chaloin, J. M. Fabre, P. Maurel, and M. J. Vilarem, Dexamethasone enhances constitutive androstane receptor expression in human hepatocytes: consequences on cytochrome P450 gene regulation, Mol. Pharmacol, vol.58, pp.1441-1450, 2000.

J. M. Pascussi, . Busson-le, M. Coniat, P. Maurel, and M. Vilarem, Transcriptional analysis of the orphan nuclear receptor constitutive androstane receptor (NR1I3) gene promoter: identification of a distal glucocorticoid response element, Mol. Endocrinol, vol.17, pp.42-55, 2003.

J. M. Pascussi, . Busson-le, M. Coniat, P. Maurel, and M. Vilarem, Transcriptional Analysis of the Orphan Nuclear Receptor Constitutive Androstane Receptor (NR1I3) Gene Promoter: Identification of a Distal Glucocorticoid Response Element, Mol. Endocrinol, vol.17, pp.42-55, 2003.

C. J. Patel, J. Bhattacharya, and A. J. Butte, An Environment-Wide Association Study (EWAS) on type 2 diabetes mellitus, PLoS One, vol.5, 2010.

R. Patel, M. Patel, R. Tsai, V. Lin, A. L. Bookout et al.,

, LXR? is required for glucocorticoid-induced hyperglycemia and hepatosteatosis in mice, J. Clin. Invest, vol.121, pp.431-441

R. D. Patel, B. D. Hollingshead, C. J. Omiecinski, and G. H. Perdew, Aryl-hydrocarbon receptor activation regulates constitutive androstane receptor levels in murine and human liver, Hepatology, vol.46, pp.209-218, 2007.

K. C. Patra and N. Hay, The pentose phosphate pathway and cancer, Trends Biochem. Sci, vol.39, pp.347-354, 2014.

D. J. Peet, B. A. Janowski, and D. J. Mangelsdorf, The LXRs: a new class of oxysterol receptors, Curr. Opin. Genet. Dev, vol.8, pp.571-575, 1998.

R. C. Peffer, J. G. Moggs, T. Pastoor, R. A. Currie, J. Wright et al., Mouse Liver Effects of Cyproconazole, a Triazole Fungicide: Role of the Constitutive Androstane Receptor, Toxicol. Sci, vol.99, pp.315-325, 2007.

C. Pelletier, J. Després, and A. Tremblay, Plasma organochlorine concentrations in endurance athletes and obese individuals, Med. Sci. Sports Exerc, vol.34, pp.1971-1975, 2002.

C. Pelletier, P. Imbeault, and A. Tremblay, Energy balance and pollution by organochlorines and polychlorinated biphenyls, Obes. Rev, vol.4, pp.17-24, 2003.

F. Peris-sampedro, M. Cabré, P. Basaure, I. Reverte, J. L. Domingo et al., Adulthood dietary exposure to a common pesticide leads to an obese-like phenotype and a diabetic profile in apoE3 mice, Environ. Res, vol.142, pp.169-176, 2015.

M. C. Petersen, D. F. Vatner, and G. I. Shulman, Regulation of hepatic glucose metabolism in health and disease, Nat. Rev. Endocrinol, vol.13, pp.572-587, 2017.

J. S. Petrick and C. D. Klaassen, Importance of hepatic induction of constitutive androstane receptor and other transcription factors that regulate xenobiotic metabolism and transport, Drug Metab. Dispos, vol.35, pp.1806-1815, 2007.

E. Petzinger and J. Geyer, Drug transporters in pharmacokinetics, Naunyn. Schmiedebergs. Arch. Pharmacol, vol.372, pp.465-475, 2006.

A. Pighon, J. Gutkowska, M. Jankowski, R. Rabasa-lhoret, and J. Lavoie, , 2011.

R. Pivonello, A. M. Isidori, M. C. De-martino, J. Newell-price, B. M. Biller et al., Complications of Cushing's syndrome: state of the art, Lancet Diabetes Endocrinol, vol.4, pp.611-629, 2016.

T. D. Porter and M. J. Coon, Cytochrome P-450. Multiplicity of isoforms, substrates, and catalytic and regulatory mechanisms, J. Biol. Chem, vol.266, pp.13469-13472, 1991.

W. P. Porter, S. M. Green, N. L. Debbink, and I. Carlson, Groundwater pesticides: interactive effects of low concentrations of carbamates aldicarb and methomyl and the triazine metribuzin on thyroxine and somatotropin levels in white rats, J. Toxicol. Environ. Health, vol.40, pp.15-34, 1993.

C. Postic and J. Girard, Contribution of de novo fatty acid synthesis to hepatic steatosis and insulin resistance: lessons from genetically engineered mice, J. Clin. Invest, vol.118, pp.829-838, 2008.

C. Postic, R. Dentin, P. Denechaud, and J. Girard, ChREBP, a Transcriptional Regulator of Glucose and Lipid Metabolism, Annu. Rev. Nutr, vol.27, pp.179-192, 2007.

S. D. Prokopec, J. D. Watson, J. Lee, R. Pohjanvirta, and P. C. Boutros, Sex-related differences in murine hepatic transcriptional and proteomic responses to TCDD, Toxicol. Appl. Pharmacol, vol.284, pp.188-196, 2015.

M. Qatanani, P. Wei, and D. D. Moore, Alterations in the distribution and orexigenic effects of dexamethasone in CAR-null mice, Pharmacol. Biochem. Behav, vol.78, pp.285-291, 2004.

M. Qatanani, J. Zhang, and D. D. Moore, Role of the constitutive androstane receptor in xenobioticinduced thyroid hormone metabolism, Endocrinology, vol.146, pp.995-1002, 2005.

J. Qin, Y. Li, Z. Cai, S. Li, J. Zhu et al., A metagenomewide association study of gut microbiota in type 2 diabetes, Nature, vol.490, pp.55-60, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01204262

Y. Qin, K. T. Dalen, J. Gustafsson, and H. I. Nebb, Regulation of hepatic fatty acid elongase 5 by LXR?-SREBP-1c, Biochim. Biophys. Acta -Mol. Cell Biol. Lipids, vol.1791, pp.140-147, 2009.

E. M. Quinet, D. Savio, A. R. Halpern, L. Chen, C. P. Miller et al., Gene-selective modulation by a synthetic oxysterol ligand of the liver X receptor, J. Lipid Res, vol.45, pp.1929-1942, 2004.

M. A. Quinn, X. Xu, M. Ronfani, and J. A. Cidlowski, Estrogen Deficiency Promotes Hepatic Steatosis via a Glucocorticoid Receptor-Dependent Mechanism in Mice, Cell Rep, vol.22, pp.2690-2701, 2018.

N. Raafat, M. A. Abass, and H. M. Salem, Malathion exposure and insulin resistance among a group of farmers in Al-Sharkia governorate, Clin. Biochem, vol.45, pp.1591-1595, 2012.

V. Racanelli and B. Rehermann, The liver as an immunological organ, Hepatology, vol.43, pp.54-62, 2006.

A. M. Rappaport, Z. J. Borowy, W. M. Lougheed, L. , and W. N. , Subdivision of hexagonal liver lobules into a structural and functional unit; role in hepatic physiology and pathology, Anat. Rec, vol.119, pp.11-33, 1954.

V. Ratziu, P. Giral, F. Charlotte, E. Bruckert, V. Thibault et al., Liver fibrosis in overweight patients, Gastroenterology, vol.118, pp.1117-1123, 2000.

V. Regitz-zagrosek, E. Lehmkuhl, and M. O. Weickert, Gender differences in the metabolic syndrome and their role for cardiovascular disease, Clin. Res. Cardiol, vol.95, pp.136-147, 2006.

J. J. Repa, G. Liang, J. Ou, Y. Bashmakov, J. A. Lobaccaro et al., Regulation of mouse sterol regulatory by oxysterol receptors, pp.2819-2830, 2000.

J. J. Repa, G. Liang, J. Ou, Y. Bashmakov, J. M. Lobaccaro et al., Regulation of mouse sterol regulatory element-binding protein-1c gene (SREBP1c) by oxysterol receptors, LXRalpha and LXRbeta, Genes Dev, vol.14, pp.2819-2830, 2000.

T. Rezen, V. Tamasi, A. Lövgren-sandblom, I. Björkhem, U. Meyer et al., Effect of CAR activation on selected metabolic pathways in normal and hyperlipidemic mouse livers, BMC Genomics, vol.10, p.384, 2009.

T. Rezen, D. Rozman, J. Pascussi, and K. Monostory, Interplay between cholesterol and drug metabolism, Biochim. Biophys. Acta -Proteins Proteomics, vol.1814, pp.146-160, 2011.

T. Re?en, M. Hafner, S. Kortagere, S. Ekins, V. Hodnik et al., Rosuvastatin and Atorvastatin Are Ligands of the Human Constitutive Androstane Receptor/Retinoid X Receptor ? Complex, Drug Metab. Dispos, vol.45, pp.974-976, 2017.

E. Riant, A. Waget, H. Cogo, J. Arnal, R. Burcelin et al., Estrogens Protect against High-Fat Diet-Induced Insulin Resistance and Glucose Intolerance in Mice, Endocrinology, vol.150, pp.2109-2117, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-00410271

E. Riant, A. Waget, H. Cogo, J. Arnal, R. Burcelin et al., Estrogens Protect against High-Fat Diet-Induced Insulin Resistance and Glucose Intolerance in Mice, Endocrinology, vol.150, pp.2109-2117, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-00410271

J. M. Ridlon, D. Kang, and P. B. Hylemon, Bile salt biotransformations by human intestinal bacteria, J. Lipid Res, vol.47, pp.241-259, 2006.

M. H. Rodrigues, A. S. Bruno, J. Nahas-neto, V. C. Sandrim, L. G. Muniz et al., Evaluation of Clinical and Inflammatory Markers of Nonalcoholic Fatty Liver Disease in Postmenopausal Women with Metabolic Syndrome, Metab. Syndr. Relat. Disord, vol.12, pp.330-338, 2014.

S. Romeo, J. Kozlitina, C. Xing, A. Pertsemlidis, D. Cox et al., Genetic variation in PNPLA3 confers susceptibility to nonalcoholic fatty liver disease, Nat. Genet, vol.40, pp.1461-1465, 2008.

G. Romero-navarro, T. Lopez-aceves, A. Rojas-ochoa, F. Mejia, and C. , Effect of dichlorvos on hepatic and pancreatic glucokinase activity and gene expression, and on insulin mRNA levels, Life Sci, vol.78, pp.1015-1020, 2006.

M. G. Rosenfeld and C. K. Glass, Coregulator codes of transcriptional regulation by nuclear receptors, J. Biol. Chem, vol.276, pp.36865-36868, 2001.

A. Roth, R. Looser, M. Kaufmann, and M. , Sterol regulatory element binding protein 1 interacts with pregnane X receptor and constitutive androstane receptor and represses their target genes, Pharmacogenet. Genomics, vol.18, pp.325-337, 2008.

D. L. Rothman, I. Magnusson, L. D. Katz, R. G. Shulman, and G. I. Shulman, Quantitation of hepatic glycogenolysis and gluconeogenesis in fasting humans with 13C NMR, Science, vol.254, pp.573-576, 1991.

L. R. Roust and M. D. Jensen, Postprandial free fatty acid kinetics are abnormal in upper body obesity, Diabetes, vol.42, pp.1567-1573, 1993.

L. Roy, T. Llopis, M. Lepage, P. Bruneau, A. Rabot et al., Intestinal microbiota determines development of non-alcoholic fatty liver disease in mice, Gut, vol.62, pp.1787-1794, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01193804

K. Ruckpaul, H. Rein, and J. Blanck, , 1985.

, Biomed. Biochim. Acta, vol.44, pp.351-379

C. Rufo, M. Teran-garcia, M. T. Nakamura, S. Koo, H. C. Towle et al., Involvement of a Unique Carbohydrate-responsive Factor in the Glucose Regulation of Rat Liver Fatty-acid Synthase Gene Transcription, J. Biol. Chem, vol.276, pp.21969-21975, 2001.

L. Rui, Energy Metabolism in the Liver, Comprehensive Physiology, pp.177-197, 2014.

K. Saito, K. Kobayashi, Y. Mizuno, Y. Fukuchi, T. Furihata et al., Peroxisome proliferatoractivated receptor alpha (PPARalpha) agonists induce constitutive androstane receptor (CAR) and cytochrome P450 2B in rat primary hepatocytes, Drug Metab. Pharmacokinet, vol.25, pp.108-111, 2010.

K. Saito, R. Moore, and M. Negishi, p38 Mitogen-activated protein kinase regulates nuclear receptor CAR that activates the CYP2B6 gene, Drug Metab. Dispos, vol.41, pp.1170-1173, 2013.

Y. Sakakura, H. Shimano, H. Sone, A. Takahashi, N. Inoue et al., Sterol regulatory element-binding proteins induce an entire pathway of cholesterol synthesis, Biochem. Biophys. Res. Commun, vol.286, pp.176-183, 2001.

T. M. Saldana, O. Basso, J. A. Hoppin, D. D. Baird, C. Knott et al., , 2007.

, Pesticide Exposure and Self-Reported Gestational Diabetes Mellitus in the Agricultural Health Study, Diabetes Care, vol.30, pp.529-534

Y. Sasaki, Y. Takahashi, K. Nakayama, and T. Kamataki, Cooperative regulation of CYP2C12 gene expression by STAT5 and liver-specific factors in female rats, J. Biol. Chem, vol.274, pp.37117-37124, 1999.

D. Sasse, Dynamics of liver glycogen: the topochemistry of glycogen synthesis, glycogen content and glycogenolysis under the experimental conditions of glycogen accumulation and depletion, Histochemistry, vol.45, pp.237-254, 1975.

T. Saussele, O. Burk, J. K. Blievernicht, K. Klein, A. Nussler et al., Selective induction of human hepatic cytochromes P450 2B6 and 3A4 by metamizole, Clin. Pharmacol. Ther, vol.82, pp.265-274, 2007.

C. C. Savary, R. Josse, A. Bruyere, F. Guillet, M. Robin et al., Interactions of Endosulfan and Methoxychlor Involving CYP3A4 and CYP2B6 in Human HepaRG Cells, Drug Metab. Dispos, vol.42, pp.1235-1240, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01128192

R. S. Savkur, Y. Wu, K. S. Bramlett, M. Wang, S. Yao et al., Alternative splicing within the ligand binding domain of the human constitutive androstane receptor, Mol. Genet. Metab, vol.80, pp.216-226

A. L. Sberna, M. Assem, T. Gautier, J. Grober, B. Guiu et al., Constitutive androstane receptor activation stimulates faecal bile acid excretion and reverse cholesterol transport in mice, J. Hepatol, vol.55, pp.154-161, 2011.

B. Schnabl and D. A. Brenner, Interactions Between the Intestinal Microbiome and Liver Diseases, Gastroenterology, vol.146, pp.1513-1524, 2014.

D. M. Schreinemachers, Mortality from ischemic heart disease and diabetes mellitus (type 2) in four U.S. wheat-producing states: a hypothesis-generating study, Environ. Health Perspect, vol.114, pp.186-193, 2006.

T. T. Schug, A. Janesick, B. Blumberg, and J. J. Heindel, Endocrine disrupting chemicals and disease susceptibility, J. Steroid Biochem. Mol. Biol, vol.127, pp.204-215, 2011.

B. Seeger, F. Klawonn, B. Nguema-bekale, and P. Steinberg, Mixture Effects of Estrogenic Pesticides at the Human Estrogen Receptor ? and ?, PLoS One, vol.11, 2016.

K. Semchuk, H. Mcduffie, A. Senthilselvan, A. Cessna, and D. Irvine, Body mass index and bromoxynil exposure in a sample of rural residents during spring herbicide application, J. Toxicol. Environ. Health. A, vol.67, pp.1321-1352, 2004.

S. Seok, T. Fu, S. Choi, Y. Li, R. Zhu et al., Transcriptional regulation of autophagy by an FXR-CREB axis, Nature, vol.516, pp.108-111, 2014.

A. Seppälä-lindroos, S. Vehkavaara, A. Häkkinen, T. Goto, J. Westerbacka et al., Fat Accumulation in the Liver Is Associated with Defects in Insulin Suppression of Glucose Production and Serum Free Fatty Acids Independent of Obesity in Normal Men, J. Clin. Endocrinol. Metab, vol.87, pp.3023-3028, 2002.

A. A. Shehata, W. Schrödl, A. A. Aldin, H. M. Hafez, and M. Krüger, The Effect of Glyphosate on Potential Pathogens and Beneficial Members of Poultry Microbiota In Vitro, Curr. Microbiol, vol.66, pp.350-358, 2013.

N. Shen, S. Jiang, J. Lu, X. Yu, S. Lai et al., , 2015.

, The Constitutive Activation of Egr-1/C/EBPa Mediates the Development of Type 2 Diabetes Mellitus by Enhancing Hepatic Gluconeogenesis, Am. J. Pathol, vol.185, pp.513-523

P. Shen, T. Hsieh, Y. Yue, Q. Sun, J. M. Clark et al., Deltamethrin increases the fat accumulation in 3T3-L1 adipocytes and Caenorhabditis elegans, Food Chem. Toxicol, vol.101, pp.149-156, 2017.

W. Shen, M. Punyanitya, A. M. Silva, J. Chen, D. Gallagher et al., Sexual dimorphism of adipose tissue distribution across the lifespan: a cross-sectional whole-body magnetic resonance imaging study, Nutr. Metab, issue.6, p.17, 2009.

X. Shi, Q. Cheng, L. Xu, J. Yan, M. Jiang et al., Cholesterol sulfate and cholesterol sulfotransferase inhibit gluconeogenesis by targeting hepatocyte nuclear factor 4?, Mol. Cell. Biol, vol.34, pp.485-497, 2014.

Y. Shibayama, K. Ushinohama, R. Ikeda, Y. Yoshikawa, T. Motoya et al., Effect of methotrexate treatment on expression levels of multidrug resistance protein 2, breast cancer resistance protein and organic anion transporters Oat1, Oat2 and Oat3 in rats, Cancer Sci, vol.97, pp.1260-1266, 2006.

A. Shibli-rahhal, M. Van-beek, and J. A. Schlechte, Cushing's syndrome, Clin. Dermatol, vol.24, pp.260-265, 2006.

I. Shimomura, H. Shimano, B. S. Korn, Y. Bashmakov, and J. D. Horton, Nuclear sterol regulatory elementbinding proteins activate genes responsible for the entire program of unsaturated fatty acid biosynthesis in transgenic mouse liver, J. Biol. Chem, vol.273, pp.35299-35306, 1998.

S. Shindo, S. Numazawa, Y. , and T. , A physiological role of AMP-activated protein kinase in phenobarbital-mediated constitutive androstane receptor activation and CYP2B induction, Biochem. J, vol.401, pp.735-741, 2007.

T. Shiraki, N. Sakai, E. Kanaya, J. , and H. , Activation of orphan nuclear constitutive androstane receptor requires subnuclear targeting by peroxisome proliferator-activated receptor ? coactivator-1?: A possible link between xenobiotic response and nutritional state, J. Biol. Chem, vol.278, pp.11344-11350, 2003.

R. Shizu, S. Shindo, T. Yoshida, and S. Numazawa, Cross-talk between constitutive androstane receptor and hypoxia-inducible factor in the regulation of gene expression, Toxicol. Lett, vol.219, pp.143-150, 2013.

C. J. Sinal, M. Tohkin, M. Miyata, J. M. Ward, G. Lambert et al., Targeted disruption of the nuclear receptor FXR/BAR impairs bile acid and lipid homeostasis, Cell, vol.102, pp.731-744, 2000.

A. G. Singal, H. Manjunath, A. C. Yopp, M. S. Beg, J. A. Marrero et al., The Effect of PNPLA3 on Fibrosis Progression and Development of Hepatocellular Carcinoma: A Meta-analysis, Am. J. Gastroenterol, vol.109, pp.325-334, 2014.

R. Singh, S. Kaushik, Y. Wang, Y. Xiang, I. Novak et al., Autophagy regulates lipid metabolism, Nature, vol.458, pp.1131-1135, 2009.

R. Singh, S. Kaushik, Y. Wang, Y. Xiang, I. Novak et al., Autophagy regulates lipid metabolism, Nature, vol.458, pp.1131-1135, 2009.

T. A. Slotkin, K. K. Brown, and F. J. Seidler, Developmental exposure of rats to chlorpyrifos elicits sexselective hyperlipidemia and hyperinsulinemia in adulthood, Environ. Health Perspect, vol.113, pp.1291-1294, 2005.

A. Smink, N. Ribas-fito, R. Garcia, M. Torrent, M. A. Mendez et al., Exposure to hexachlorobenzene during pregnancy increases the risk of overweight in children aged 6 years, Acta Paediatr, vol.97, pp.1465-1469, 2008.

S. M. Snedeker and A. G. Hay, Do interactions between gut ecology and environmental chemicals contribute to obesity and diabetes?, Environ. Health Perspect, vol.120, pp.332-339, 2012.

J. Soares, P. Pimentel-nunes, R. Roncon-albuquerque, A. Leite-moreira, and A. Leite-moreira, The role of lipopolysaccharide/toll-like receptor 4 signaling in chronic liver diseases, Hepatol. Int, vol.4, pp.659-672, 2010.

W. C. Song, Biochemistry and reproductive endocrinology of estrogen sulfotransferase, Ann. N. Y. Acad. Sci, vol.948, pp.43-50, 2001.

E. A. Sotaniemi, A. J. Arranto, S. Sutinen, and J. H. Stengård, Treatment of noninsulin-dependent diabetes mellitus with enzyme inducers, Clin. Pharmacol. Ther, vol.33, pp.826-835, 1983.

E. E. Spangenburg, L. M. Wohlers, and A. P. Valencia, Metabolic Dysfunction Under Reduced Estrogen Levels, Exerc. Sport Sci. Rev, vol.40, pp.195-203, 2012.

A. P. Starling, D. M. Umbach, F. Kamel, S. Long, D. P. Sandler et al., Pesticide use and incident diabetes among wives of farmers in the Agricultural Health Study, Occup. Environ. Med, vol.71, pp.629-635, 2014.

J. L. Staudinger, A. Madan, K. M. Carol, and A. Parkinson, Regulation of drug transporter gene expression by nuclear receptors, Drug Metab. Dispos, vol.31, pp.523-527, 2003.

C. A. Stedman, C. Liddle, S. A. Coulter, J. Sonoda, J. G. Alvarez et al., Nuclear receptors constitutive androstane receptor and pregnane X receptor ameliorate cholestatic liver injury, Proc. Natl. Acad. Sci. U. S. A, vol.102, pp.2063-2068, 2005.

N. Stefan, H. Staiger, and H. Häring, Dissociation between fatty liver and insulin resistance: the role of adipose triacylglycerol lipase, Diabetologia, vol.54, pp.7-9, 2011.

C. A. Strott, Sulfonation and Molecular Action, Endocr. Rev, vol.23, pp.703-732, 2002.

T. Sueyoshi, T. Kawamoto, I. Zelko, P. Honkakoski, and M. Negishi, The repressed nuclear receptor CAR responds to phenobarbital in activating the human CYP2B6 gene, J. Biol. Chem, vol.274, pp.6043-6046, 1999.

T. Sueyoshi, R. Moore, J. Sugatani, Y. Matsumura, and M. Negishi, PPP1R16A, the membrane subunit of protein phosphatase 1beta, signals nuclear translocation of the nuclear receptor constitutive active/androstane receptor, Mol. Pharmacol, vol.73, pp.1113-1121, 2008.

T. Sueyoshi, W. D. Green, K. Vinal, T. S. Woodrum, R. Moore et al., Garlic Extract Diallyl Sulfide (DAS) Activates Nuclear Receptor CAR to Induce the Sult1e1 Gene in Mouse Liver, PLoS One, vol.6, p.21229, 2011.

J. Sugatani, H. Kojima, A. Ueda, S. Kakizaki, K. Yoshinari et al., The phenobarbital response enhancer module in the human bilirubin UDP-glucuronosyltransferase UGT1A1 gene and regulation by the nuclear receptor CAR, Hepatology, vol.33, pp.1232-1238, 2001.

K. Suino, L. Peng, R. Reynolds, Y. Li, J. Cha et al., The nuclear xenobiotic receptor CAR: structural determinants of constitutive activation and heterodimerization, Mol. Cell, vol.16, pp.893-905, 2004.

H. Sun, O. Shen, X. Xu, L. Song, and X. Wang, Carbaryl, 1-naphthol and 2-naphthol inhibit the beta-1 thyroid hormone receptor-mediated transcription in vitro, Toxicology, vol.249, pp.238-242, 2008.

Q. Sun, W. Qi, J. J. Yang, K. S. Yoon, J. M. Clark et al., Fipronil promotes adipogenesis via AMPK?-mediated pathway in 3T3-L1 adipocytes, Food Chem. Toxicol, vol.92, pp.217-223, 2016.

K. Sung, S. H. Wild, and C. D. Byrne, Resolution of Fatty Liver and Risk of Incident Diabetes, J. Clin. Endocrinol. Metab, vol.98, pp.3637-3643, 2013.

K. Swales and M. Negishi, CAR, driving into the future, Mol. Endocrinol, vol.18, pp.1589-1598, 2004.

J. H. Tabibian, A. I. Masyuk, T. V. Masyuk, S. P. O'hara, and N. F. Larusso, Physiology of Cholangiocytes, Comprehensive Physiology, pp.541-565, 2013.

S. Takeuchi, T. Matsuda, S. Kobayashi, T. Takahashi, and H. Kojima, In vitro screening of 200 pesticides for agonistic activity via mouse peroxisome proliferator-activated receptor (PPAR)? and PPAR? and quantitative analysis of in vivo induction pathway, Toxicol. Appl. Pharmacol, vol.217, pp.235-244, 2006.

G. S. Tannenbaum, M. , and J. B. , Evidence for an Endogenous Ultradian Rhythm Governing Growth Hormone Secretion in the Rat, Endocrinology, vol.98, pp.562-570, 1976.

G. Tarantino, S. Savastano, and A. Colao, Hepatic steatosis, low-grade chronic inflammation and hormone/growth factor/adipokine imbalance, World J. Gastroenterol, vol.16, pp.4773-4783, 2010.

H. F. Teutsch, A. Goellner, and W. Mueller-klieser, Glucose levels and succinate and lactate dehydrogenase activity in EMT6/Ro tumor spheroids, Eur. J. Cell Biol, vol.66, pp.302-307, 1995.

P. D. Thompson, P. W. Jurutka, C. A. Haussler, G. K. Whitfield, and M. R. Haussler, Heterodimeric DNA binding by the vitamin D receptor and retinoid X receptors is enhanced by 1,25-dihydroxyvitamin D3 and inhibited by 9-cis-retinoic acid. Evidence for allosteric receptor interactions, J. Biol. Chem, vol.273, pp.8483-8491, 1998.

I. Tikhanovich, J. Cox, and S. A. Weinman, Forkhead box class O transcription factors in liver function and disease, J. Gastroenterol. Hepatol, vol.28, pp.125-131, 2013.

H. Tilg, A. R. Moschen, and M. Roden, NAFLD and diabetes mellitus, Nat. Rev. Gastroenterol. Hepatol, vol.14, pp.32-42, 2017.

Y. E. Timsit and M. Negishi, CAR and PXR: The xenobiotic-sensing receptors, Steroids, vol.72, pp.231-246, 2007.

P. M. Titchenell, Q. Chu, B. R. Monks, and M. J. Birnbaum, Hepatic insulin signalling is dispensable for suppression of glucose output by insulin in vivo, Nat. Commun, vol.6, p.7078, 2015.

P. M. Titchenell, W. J. Quinn, M. Lu, J. Chen, J. D. Rabinowitz et al., Direct Hepatocyte Insulin Signaling Is Required for Lipogenesis but Is Dispensable for the Suppression of Glucose Production, Cell Metab, vol.23, pp.1154-1166, 2016.

A. H. Tolson, W. , and H. , Regulation of drug-metabolizing enzymes by xenobiotic receptors: PXR and CAR, Adv. Drug Deliv. Rev, vol.62, pp.1238-1249, 2010.

C. Torre, C. Perret, and S. Colnot, Transcription dynamics in a physiological process: ?-Catenin signaling directs liver metabolic zonation, Int. J. Biochem. Cell Biol, vol.43, pp.271-278, 2011.

E. Turola, S. Petta, E. Vanni, F. Milosa, L. Valenti et al., Ovarian senescence increases liver fibrosis in humans and zebrafish with steatosis, Dis. Model. Mech, vol.8, pp.1037-1046, 2015.

I. Tzameli, P. Pissios, E. G. Schuetz, and D. D. Moore, The xenobiotic compound 1,4-bis[2-(3,5-dichloropyridyloxy)]benzene is an agonist ligand for the nuclear receptor CAR, Mol. Cell. Biol, vol.20, pp.2951-2958, 2000.

A. Ueda, H. K. Hamadeh, H. K. Webb, Y. Yamamoto, T. Sueyoshi et al., Diverse roles of the nuclear orphan receptor CAR in regulating hepatic genes in response to phenobarbital, Mol. Pharmacol, vol.61, pp.1-6, 2002.

B. Ugele, H. J. Kempen, J. M. Kempen, R. Gebhardt, P. Meijer et al., , 1991.

, Heterogeneity of rat liver parenchyma in cholesterol 7 alpha-hydroxylase and bile acid synthesis, Biochem. J, vol.276, pp.73-77

J. Ukropec, Z. Radikova, M. Huckova, J. Koska, A. Kocan et al., High prevalence of prediabetes and diabetes in a population exposed to high levels of an organochlorine cocktail, Diabetologia, vol.53, pp.899-906, 2010.

K. A. Usmani, R. L. Rose, and E. Hodgson, Inhibition and activation of the human liver microsomal and human cytochrome P450 3A4 metabolism of testosterone by deployment-related chemicals, Drug Metab. Dispos, vol.31, pp.384-391, 2003.

K. Uyeda, H. Yamashita, and T. Kawaguchi, Carbohydrate responsive element-binding protein (ChREBP): a key regulator of glucose metabolism and fat storage, Biochem. Pharmacol, vol.63, pp.2075-2080, 2002.

C. Vecchia, . La, and E. Negri, A review of epidemiological data on epilepsy, phenobarbital, and risk of liver cancer, Eur. J. Cancer Prev, vol.23, pp.1-7, 2014.

A. Vegiopoulos and S. Herzig, Glucocorticoids, metabolism and metabolic diseases, Mol. Cell. Endocrinol, vol.275, pp.43-61, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00531932

J. D. Veldhuis and C. Y. Bowers, Human GH pulsatility: an ensemble property regulated by age and gender, J. Endocrinol. Invest, vol.26, pp.799-813, 2003.

G. Velmurugan, T. Ramprasath, K. Swaminathan, G. Mithieux, J. Rajendhran et al., Gut microbial degradation of organophosphate insecticides-induces glucose intolerance via gluconeogenesis, Genome Biol, vol.18, 2017.

S. L. Verhulst, V. Nelen, E. Hond, . Den, G. Koppen et al., , 2009.

, Intrauterine exposure to environmental pollutants and body mass index during the first 3 years of life, Environ. Health Perspect, vol.117, pp.122-126

T. Vermeire, H. Stevenson, M. N. Peiters, M. Rennen, W. Slob et al., Assessment factors for human health risk assessment: a discussion paper, Crit. Rev. Toxicol, vol.29, pp.439-490, 1999.

M. Vernay, B. Salanave, C. De-peretti, C. Druet, A. Malon et al., Metabolic syndrome and socioeconomic status in France: The French Nutrition and Health Survey, Int. J. Public Health, vol.58, pp.855-864, 2006.

A. Verrotti, S. Agostinelli, P. Parisi, F. Chiarelli, and G. Coppola, Nonalcoholic fatty liver disease in adolescents receiving valproic acid, Epilepsy Behav, vol.20, pp.382-385, 2011.

J. H. Vincent, M. , and D. , Comparison of Criteria for Defining Inspirable Aerosol and the Development of Appropriate Samplers, Am. Ind. Hyg. Assoc. J, vol.48, pp.454-457, 1987.

T. J. Visser, E. Kaptein, H. Glatt, I. Bartsch, M. Hagen et al., Characterization of thyroid hormone sulfotransferases, Chem. Biol. Interact, vol.109, pp.279-291, 1998.

V. Volynets, M. A. Küper, S. Strahl, I. B. Maier, A. Spruss et al., Nutrition, Intestinal Permeability, and Blood Ethanol Levels Are Altered in Patients with Nonalcoholic Fatty Liver Disease (NAFLD), Dig. Dis. Sci, vol.57, pp.1932-1941, 2012.

H. Volzke, S. Schwarz, S. E. Baumeister, H. Wallaschofski, C. Schwahn et al., Menopausal status and hepatic steatosis in a general female population, Gut, vol.56, pp.594-595, 2007.

S. Vosough-ghanbari, P. Sayyar, S. Pournourmohammadi, A. Aliahmadi, S. N. Ostad et al., Stimulation of insulin and glucagon synthesis in rat Langerhans islets by malathion in vitro: Evidence for mitochondrial interaction and involvement of subcellular non-cholinergic mechanisms, Pestic. Biochem. Physiol, vol.89, pp.130-136, 2007.

T. Wada, J. Gao, and W. Xie, PXR and CAR in energy metabolism, Trends Endocrinol. Metab, vol.20, pp.273-279, 2009.

B. Wahlang, K. C. Falkner, B. Gregory, D. Ansert, D. Young et al., Polychlorinated biphenyl 153 is a diet-dependent obesogen that worsens nonalcoholic fatty liver disease in male C57BL6/J mice, J. Nutr. Biochem, vol.24, pp.1587-1595, 2013.

H. Wang, S. Faucette, R. Moore, T. Sueyoshi, M. Negishi et al., Human Constitutive Androstane Receptor Mediates Induction of CYP2B6 Gene Expression by Phenytoin, J. Biol. Chem, vol.279, pp.29295-29301, 2004.

J. Wang, M. Olin, B. Rozell, I. Björkhem, C. Einarsson et al., Differential hepatocellular zonation pattern of cholesterol 7?-hydroxylase (Cyp7a1) and sterol 12?-hydroxylase (Cyp8b1) in the mouse, Histochem. Cell Biol, vol.127, pp.253-261, 2007.

J. Wang, Y. Zhu, X. Cai, J. Yu, X. Yang et al., Abnormal glucose regulation in pyrethroid pesticide factory workers, Chemosphere, vol.82, pp.1080-1082, 2011.

P. Wang, H. Wang, M. Xu, Y. Liang, Y. Sun et al., Combined subchronic toxicity of dichlorvos with malathion or pirimicarb in mice liver and serum: A metabonomic study, Food Chem. Toxicol, vol.70, pp.222-230, 2014.

S. Wang, N. Shi, Z. Ji, and G. Pinna, , 2002.

, Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi, vol.20, pp.173-176

T. J. Wang, D. Ngo, N. Psychogios, A. Dejam, M. G. Larson et al., 2-Aminoadipic acid is a biomarker for diabetes risk, J. Clin. Invest, vol.123, pp.4309-4317, 2013.

Y. Wang, D. Botolin, J. Xu, B. Christian, E. Mitchell et al., Regulation of hepatic fatty acid elongase and desaturase expression in diabetes and obesity, J. Lipid Res, vol.47, pp.2028-2041, 2006.

Y. Wang, J. Viscarra, S. Kim, and H. S. Sul, Transcriptional regulation of hepatic lipogenesis, Nat. Rev. Mol. Cell Biol, vol.16, pp.678-689, 2015.

C. M. Weghorst and J. E. Klaunig, Phenobarbital promotion in diethylnitrosamine-initiated infant B6C3F1 mice: influence of gender, Carcinogenesis, vol.10, pp.609-612, 1989.

P. Wei, J. Zhang, D. H. Dowhan, Y. Han, and D. D. Moore, Specific and overlapping functions of the nuclear hormone receptors CAR and PXR in xenobiotic response, Pharmacogenomics J, vol.2, pp.117-126, 2002.

P. Wei, J. Zhang, D. H. Dowhan, Y. Han, and D. D. Moore, Specific and overlapping functions of the nuclear hormone receptors CAR and PXR in xenobiotic response, Pharmacogenomics J, vol.2, pp.117-126, 2002.

N. Wieneke, K. I. Hirsch-ernst, M. Kuna, S. Kersten, and G. P. Püschel, PPAR?-dependent induction of the energy homeostasis-regulating nuclear receptor NR1i3 (CAR) in rat hepatocytes: Potential role in starvation adaptation, FEBS Lett, vol.581, pp.5617-5626, 2007.

C. D. Williams, J. Stengel, M. I. Asike, D. M. Torres, J. Shaw et al., Prevalence of Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis Among a Largely Middle-Aged Population Utilizing Ultrasound and Liver Biopsy: A Prospective Study, Gastroenterology, vol.140, pp.124-131, 2011.

T. M. Willson and S. A. Kliewer, PXR, CAR AND DRUG METABOLISM, Nat. Rev. Drug Discov, vol.1, pp.259-266, 2002.

J. D. Wilson, Presentation of the Southern Society for Clinical Investigation Founder's Medal to Dr. Daniel W. Foster, Am. J. Med. Sci, vol.304, pp.1-2, 1992.

V. S. Wilson, J. B. Mclachlan, J. G. Falls, and G. A. Leblanc, Alteration in sexually dimorphic testosterone biotransformation profiles as a biomarker of chemically induced androgen disruption in mice, Environ. Health Perspect, vol.107, pp.377-384, 1999.

C. A. Wiwi, M. Gupte, and D. J. Waxman, Sexually dimorphic P450 gene expression in liver-specific hepatocyte nuclear factor 4alpha-deficient mice, Mol. Endocrinol, vol.18, pp.1975-1987, 2004.

C. Wohlfahrt-veje, K. M. Main, I. M. Schmidt, M. Boas, T. K. Jensen et al., Lower birth weight and increased body fat at school age in children prenatally exposed to modern pesticides: a prospective study, Environ. Heal, vol.10, p.79, 2011.

M. Wortham, M. Czerwinski, L. He, A. Parkinson, and Y. Y. Wan, Expression of constitutive androstane receptor, hepatic nuclear factor 4 alpha, and P450 oxidoreductase genes determines interindividual variability in basal expression and activity of a broad scope of xenobiotic metabolism genes in the human liver, Drug Metab. Dispos, vol.35, pp.1700-1710, 2007.

M. C. Wright, The impact of pregnane X receptor activation on liver fibrosis, Biochem. Soc. Trans, vol.34, pp.1119-1123, 2006.

E. Wright, J. Vincent, and E. J. Fernandez, Thermodynamic characterization of the interaction between CAR-RXR and SRC-1 peptide by isothermal titration calorimetry, Biochemistry, vol.46, pp.862-870, 2007.

M. E. Wyde, E. Bartolucci, A. Ueda, H. Zhang, B. Yan et al., The environmental pollutant 1,1-dichloro-2,2-bis (p-chlorophenyl)ethylene induces rat hepatic cytochrome P450 2B and 3A expression through the constitutive androstane receptor and pregnane X receptor, Mol. Pharmacol, vol.64, pp.474-481, 2003.

M. E. Wyde, S. E. Kirwan, F. Zhang, A. Laughter, H. B. Hoffman et al., Di-n-butyl phthalate activates constitutive androstane receptor and pregnane X receptor and enhances the expression of steroid-metabolizing enzymes in the liver of rat fetuses, Toxicol. Sci, vol.86, pp.281-290, 2005.

L. Xiao, X. Xie, and Y. Zhai, Functional crosstalk of CAR-LXR and ROR-LXR in drug metabolism and lipid metabolism, Adv. Drug Deliv. Rev, vol.62, pp.1316-1321, 2010.

X. Xiao, Y. Kim, D. Kim, K. S. Yoon, J. M. Clark et al., Permethrin alters glucose metabolism in conjunction with high fat diet by potentiating insulin resistance and decreases voluntary activities in female C57BL/6J mice, Food Chem. Toxicol, vol.108, pp.161-170, 2017.

Y. Xie, B. Nedumaran, and H. Choi, Molecular characterization of SMILE as a novel corepressor of nuclear receptors, Nucleic Acids Res, vol.37, pp.4100-4115, 2009.

C. Xu, C. Y. Li, .. Kong, and A. T. , Induction of phase I, II and III drug metabolism/transport by xenobiotics, Arch. Pharm. Res, vol.28, pp.249-268, 2005.

Y. Yamamoto, R. Moore, H. A. Hess, G. L. Guo, F. J. Gonzalez et al., Estrogen receptor alpha mediates 17alpha-ethynylestradiol causing hepatotoxicity, J. Biol. Chem, vol.281, pp.16625-16631, 2006.

H. Yamashita, M. Takenoshita, M. Sakurai, R. K. Bruick, W. J. Henzel et al., A glucose-responsive transcription factor that regulates carbohydrate metabolism in the liver, Proc. Natl. Acad. Sci, vol.98, pp.9116-9121, 2001.

H. Yamazaki and T. Shimada, Progesterone and Testosterone Hydroxylation by Cytochromes P450 2C19, 2C9, and 3A4 in Human Liver Microsomes, Arch. Biochem. Biophys, vol.346, pp.161-169, 1997.

H. Yang, B. Garzel, S. Heyward, T. Moeller, P. Shapiro et al., Metformin represses drug-induced expression of CYP2B6 by modulating the constitutive androstane receptor signaling, Mol. Pharmacol, vol.85, pp.249-260, 2014.

X. Yang, M. Downes, R. T. Yu, A. L. Bookout, W. He et al., Nuclear Receptor Expression Links the Circadian Clock to Metabolism, Cell, vol.126, pp.801-810, 2006.

J. L. Yecies, H. H. Zhang, S. Menon, S. Liu, D. Yecies et al., Akt Stimulates Hepatic SREBP1c and Lipogenesis through Parallel mTORC1-Dependent and Independent Pathways, Cell Metab, vol.14, pp.21-32, 2011.

C. R. Yellaturu, X. Deng, L. M. Cagen, H. G. Wilcox, C. M. Mansbach et al., Insulin Enhances Post-translational Processing of Nascent SREBP-1c by Promoting Its Phosphorylation and Association with COPII Vesicles, J. Biol. Chem, vol.284, pp.7518-7532, 2009.

N. Yokomori, K. Nishio, K. Aida, and M. Negishi, Transcriptional regulation by HNF-4 of the steroid 15alpha-hydroxylase P450 (Cyp2a-4) gene in mouse liver, J. Steroid Biochem. Mol. Biol, vol.62, pp.307-314, 1997.

K. Yoshinari, K. Kobayashi, R. Moore, T. Kawamoto, and M. Negishi, Identification of the nuclear receptor CAR:HSP90 complex in mouse liver and recruitment of protein phosphatase 2A in response to phenobarbital, FEBS Lett, vol.548, pp.17-20, 2003.

A. Yu, K. Fukamachi, K. W. Krausz, C. Cheung, and F. J. Gonzalez, Potential role for human cytochrome P450 3A4 in estradiol homeostasis, Endocrinology, vol.146, pp.2911-2919, 2005.

Y. Zhai, T. Wada, B. Zhang, S. Khadem, S. Ren et al., A functional cross-talk between liver X receptor-? and constitutive androstane receptor links lipogenesis and xenobiotic responses, Mol. Pharmacol, vol.78, pp.666-674, 2010.

D. Zhang, Z. Liu, C. S. Choi, L. Tian, R. Kibbey et al., , 2007.

, Mitochondrial dysfunction due to long-chain Acyl-CoA dehydrogenase deficiency causes hepatic steatosis and hepatic insulin resistance, Proc. Natl. Acad. Sci, vol.104, pp.17075-17080

J. Zhang, W. Huang, S. S. Chua, P. Wei, and D. D. Moore, Modulation of acetaminophen-induced hepatotoxicity by the xenobiotic receptor CAR, Science, vol.298, pp.422-424, 2002.

J. Zhang, W. Huang, M. Qatanani, R. M. Evans, and D. D. Moore, The Constitutive Androstane Receptor and Pregnane X Receptor Function Coordinately to Prevent Bile Acid-induced Hepatotoxicity, J. Biol. Chem, vol.279, pp.49517-49522, 2004.

J. Zhang, W. Huang, M. Qatanani, R. M. Evans, and D. D. Moore, The constitutive androstane receptor and pregnane X receptor function coordinately to prevent bile acid-induced hepatotoxicity, J. Biol. Chem, vol.279, pp.49517-49522, 2004.

J. Zhang, J. Zhang, R. Liu, J. Gan, J. Liu et al., Endocrine-Disrupting Effects of Pesticides through Interference with Human Glucocorticoid Receptor, Environ. Sci. Technol, vol.50, pp.435-443, 2016.

X. Zhang, Z. Shi, J. Lyv, X. He, N. A. Englert et al., Pyrene is a Novel Constitutive Androstane Receptor (CAR) Activator and Causes Hepatotoxicity by CAR, Toxicol. Sci, vol.147, pp.436-445, 2015.

J. Zhou, Y. Zhai, Y. Mu, H. Gong, H. Uppal et al., A Novel Pregnane X Receptor-mediated and Sterol Regulatory Element-binding Protein-independent Lipogenic Pathway, J. Biol. Chem, vol.281, pp.15013-15020, 2006.

L. Zhu, S. S. Baker, C. Gill, W. Liu, R. Alkhouri et al., Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: A connection between endogenous alcohol and NASH, Hepatology, vol.57, pp.601-609, 2013.

B. L. Zwicker, A. , and L. B. , Transport and biological activities of bile acids, Int. J. Biochem. Cell Biol, vol.45, pp.1389-1398, 2013.

, NURSA -Nuclear Receptor Signaling Atlas

, Base de données -Eurostat