, World Health Organization, Obesity and Overweight, Fact sheet 311, WHO, issue.fs311, 2014.

L. Trasande, R. Zoeller, U. Hass, A. Kortenkamp, P. Grandjean et al., Burden of disease and costs of exposure to endocrine disrupting chemicals in the European Union: an updated analysis, Andrology, vol.100, issue.4, pp.565-572, 2016.
DOI : 10.1210/jc.2015-1622

URL : https://hal.archives-ouvertes.fr/hal-01500574

B. Neel and R. Sargis, The Paradox of Progress: Environmental Disruption of Metabolism and the Diabetes Epidemic, Diabetes, vol.60, issue.7, pp.1838-1848, 2011.
DOI : 10.2337/db11-0153

P. Baillie-hamilton, Chemical Toxins: A Hypothesis to Explain the Global Obesity Epidemic, The Journal of Alternative and Complementary Medicine, vol.8, issue.2, pp.185-192, 2002.
DOI : 10.1089/107555302317371479

J. Grier, Ban of DDT and subsequent recovery of Reproduction in bald eagles, Science, vol.218, issue.4578, pp.1232-1235, 1982.
DOI : 10.1126/science.7146905

A. Herbst, H. Ulfelder, and D. Poskanzer, Adenocarcinoma of the vagina. Association of maternal stilbestrol therapy with tumor appearance in young women, American Journal of Obstetrics and Gynecology, vol.181, issue.6, pp.878-881, 1971.
DOI : 10.1016/S0002-9378(99)70411-4

T. Colborn, F. Saal, and A. Soto, Developmental effects of endocrine-disrupting chemicals in wildlife and humans, Environmental Health Perspectives, vol.101, issue.5, pp.378-384, 1993.
DOI : 10.1289/ehp.93101378

R. Newbold, B. Bullock, M. Lachlan, and J. , Exposure to Diethylstilbestrol During Pregnancy Permanently Alters the Ovary and Oviduct, PMID: 6850046] 9 WHO. Global assessment of the state of the science of endocrine disruptors, pp.735-744, 1983.
DOI : 10.1095/biolreprod28.3.735

J. Dorne, Metabolism, variability and risk assessment, Toxicology, vol.268, issue.3, pp.156-164, 2010.
DOI : 10.1016/j.tox.2009.11.004

, PMID, 19932147.

L. Vandenberg, T. Colborn, T. Hayes, J. Heindel, D. Jacobs et al., Hormones and Endocrine-Disrupting Chemicals: Low-Dose Effects and Nonmonotonic Dose Responses, Endocrine Reviews, vol.33, issue.3, pp.378-455, 2012.
DOI : 10.1210/er.2011-1050

D. Barker, J. Eriksson, T. Forsen, and O. C. , Fetal origins of adult disease: strength of effects and biological basis, International Journal of Epidemiology, vol.31, issue.6, pp.1235-1239, 2002.
DOI : 10.1016/S0140-6736(00)03553-4

R. Barouki, P. Gluckman, P. Grandjean, M. Hanson, and J. Heindel, Developmental origins of non-communicable disease: Implications for research and public health, Environmental Health, vol.127, issue.3, p.42, 2012.
DOI : 10.1016/j.jsbmb.2011.08.007

URL : https://hal.archives-ouvertes.fr/inserm-00712586

F. Mauvais-jarvis, Sex differences in metabolic homeostasis, diabetes, and obesity, Biology of Sex Differences, vol.67, issue.1, p.14, 2015.
DOI : 10.1016/j.diabres.2004.05.001

M. Brown and J. Goldstein, Selective versus Total Insulin Resistance: A Pathogenic Paradox, Cell Metabolism, vol.7, issue.2, pp.95-96, 2008.
DOI : 10.1016/j.cmet.2007.12.009

S. Guo, Insulin signaling, resistance, and the metabolic syndrome: insights from mouse models into disease mechanisms, Journal of Endocrinology, vol.378, issue.9786, pp.1-23, 2014.
DOI : 10.1016/S0140-6736(11)61027-1

M. Cave, H. Clair, J. Hardesty, K. Falkner, W. Feng et al., Nuclear receptors and nonalcoholic fatty liver disease 1 1This article is part of a Special Issue entitled: Xenobiotic nuclear receptors: New Tricks for An Old Dog, edited by Dr. Wen Xie., Biochimica et Biophysica Acta (BBA) - Gene Regulatory Mechanisms, vol.1859, issue.9, pp.1083-1099, 2016.
DOI : 10.1016/j.bbagrm.2016.03.002

C. Casals-casas and B. Desvergne, Endocrine Disruptors: From Endocrine to Metabolic Disruption, Annual Review of Physiology, vol.73, issue.1, pp.135-162, 2011.
DOI : 10.1146/annurev-physiol-012110-142200

T. Wada, J. Gao, and W. Xie, PXR and CAR in energy metabolism. Trends in endocrinology and metabolism, TEM, vol.20, issue.6, pp.273-279, 2009.

J. He, Q. Cheng, and W. Xie, Minireview: Nuclear Receptor-Controlled Steroid Hormone Synthesis and Metabolism, Molecular Endocrinology, vol.24, issue.1, pp.11-21, 2010.
DOI : 10.1210/me.2009-0212

, 21

H. Swanson, T. Wada, W. Xie, B. Renga, A. Zampella et al., Role of nuclear receptors in lipid dysfunction and obesity-related diseases. Drug metabolism and disposition: the biological fate of chemicals 2013, pp.1-11

E. Swedenborg, J. Ruegg, S. Makela, and I. Pongratz, Endocrine disruptive chemicals: mechanisms of action and involvement in metabolic disorders, Journal of Molecular Endocrinology, vol.43, issue.1, pp.1-10, 2009.
DOI : 10.1677/JME-08-0132

D. Rebourcet, F. Odet, A. Verot, E. Combe, E. Meugnier et al., -dioxin on male reproductive function: identification of Ccl5 as a potential marker, International Journal of Andrology, vol.52, issue.Suppl, pp.413-424, 2010.
DOI : 10.1016/S0041-008X(96)80010-X

L. Merrill, M. Emond, C. Kim, M. Antignac, J. et al., Toxicological function of adipose tissue: focus on persistent organic pollutants, Environmental health perspectives, vol.121, issue.2, pp.162-169, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01549161

P. Tontonoz and B. Spiegelman, Fat and Beyond: The Diverse Biology of PPAR??, Annual Review of Biochemistry, vol.77, issue.1, pp.289-312, 2008.
DOI : 10.1146/annurev.biochem.77.061307.091829

A. Foryst-ludwig, M. Clemenz, S. Hohmann, M. Hartge, C. Sprang et al., Metabolic Actions of Estrogen Receptor Beta (ER??) are Mediated by a Negative Cross-Talk with PPAR??, PLoS Genetics, vol.10, issue.6, p.1000108, 2008.
DOI : 10.1371/journal.pgen.1000108.s005

J. Pascussi, S. Gerbal-chaloin, C. Duret, M. Daujat-chavanieu, M. Vilarem et al., The Tangle of Nuclear Receptors that Controls Xenobiotic Metabolism and Transport: Crosstalk and Consequences, Annual Review of Pharmacology and Toxicology, vol.48, issue.1, pp.1-32, 2008.
DOI : 10.1146/annurev.pharmtox.47.120505.105349

URL : https://hal.archives-ouvertes.fr/inserm-00162144

F. Ohtake, K. Takeyama, T. Matsumoto, H. Kitagawa, Y. Yamamoto et al., Modulation of oestrogen receptor signalling by association with the activated dioxin receptor, Nature, vol.71, issue.6939, pp.545-550, 2003.
DOI : 10.1016/S0015-0282(98)00437-3

S. Kliewer, J. Lehmann, and T. Willson, Orphan Nuclear Receptors: Shifting Endocrinology into Reverse, Science, vol.284, issue.5415, pp.757-760, 1999.
DOI : 10.1126/science.284.5415.757

D. Torre, S. Mitro, N. Fontana, R. Gomaraschi, M. Favari et al., An Essential Role for Liver ERalpha in Coupling Hepatic Metabolism to the Reproductive Cycle, Cell reports, vol.15, issue.2, pp.360-371, 2016.

J. Heindel, B. Blumberg, M. Cave, R. Machtinger, A. Mantovani et al., Vom Saal F. Metabolism disrupting chemicals and metabolic disorders, Reprod Toxicol, 2016.

L. Magueresse-battistoni, B. Vidal, H. Naville, and D. , Lifelong consumption of low-dosed food pollutants and metabolic health, Journal of Epidemiology and Community Health, vol.33, issue.(Suppl 3), pp.512-515, 2015.
DOI : 10.1377/hlthaff.2013.0686

J. Michalek and M. Pavuk, Diabetes and Cancer in Veterans of Operation Ranch Hand After Adjustment for Calendar Period, Days of Spraying, and Time Spent in Southeast Asia, Journal of Occupational and Environmental Medicine, vol.50, issue.3, pp.330-340, 2008.
DOI : 10.1097/JOM.0b013e31815f889b

P. Bertazzi, D. Consonni, S. Bachetti, M. Rubagotti, A. Baccarelli et al., Health Effects of Dioxin Exposure: A 20-Year Mortality Study, American Journal of Epidemiology, vol.153, issue.11, pp.1031-1044, 2001.
DOI : 10.1093/aje/153.11.1031

D. Lee, M. Porta, D. Jacobs, J. Vandenberg, and L. , Chlorinated Persistent Organic Pollutants, Obesity, and Type 2 Diabetes, Endocrine Reviews, vol.35, issue.4, pp.557-601, 2014.
DOI : 10.1210/er.2013-1084

URL : https://academic.oup.com/edrv/article-pdf/35/4/557/8863257/edrv0557.pdf

M. Gauthier, R. Rabasa-lhoret, D. Prud-'homme, A. Karelis, D. Geng et al., The Metabolically Healthy But Obese Phenotype Is Associated With Lower Plasma Levels of Persistent Organic Pollutants as Compared to the Metabolically Abnormal Obese Phenotype, The Journal of Clinical Endocrinology & Metabolism, vol.99, issue.6, pp.1061-10662013, 2014.
DOI : 10.1210/jc.2013-3935

R. Newbold, E. Padilla-banks, R. Snyder, T. Phillips, and W. Jefferson, Developmental exposure to endocrine disruptors and the obesity epidemic, Reproductive Toxicology, vol.23, issue.3, pp.290-296, 2007.
DOI : 10.1016/j.reprotox.2006.12.010

P. Heine, J. Taylor, G. Iwamoto, D. Lubahn, and P. Cooke, Increased adipose tissue in male and female estrogen receptor-alpha knockout mice, Proceedings of the National Academy of Sciences, vol.84, issue.12, pp.12729-12734, 2000.
DOI : 10.1210/jc.84.12.4677

R. Chamorro-garcia, M. Sahu, R. Abbey, J. Laude, N. Pham et al., Transgenerational Inheritance of Increased Fat Depot Size, Stem Cell Reprogramming, and Hepatic Steatosis Elicited by Prenatal Exposure to the Obesogen Tributyltin in Mice, Environmental Health Perspectives, vol.121, issue.3, pp.359-366, 2013.
DOI : 10.1289/ehp.1205701

A. Janesick, G. Dimastrogiovanni, L. Vanek, C. Boulos, R. Chamorro-garcia et al.,

, On the Utility of ToxCast and ToxPi as Methods for Identifying New Obesogens, Environmental health perspectives, vol.124, issue.8, pp.1214-1226, 2016.

A. Pereira-fernandes, C. Vanparys, L. Vergauwen, D. Knapen, P. Jorens et al.,

, Toxicogenomics in the 3T3-L1 cell line, a new approach for screening of obesogenic compounds Toxicological sciences : an official journal of the Society of, Toxicology, vol.140, issue.2, pp.352-363, 2014.

J. Ruzzin, R. Petersen, E. Meugnier, L. Madsen, E. Lock et al., Persistent Organic Pollutant Exposure Leads to Insulin Resistance Syndrome, Environmental Health Perspectives, vol.118, issue.4, pp.465-471, 2010.
DOI : 10.1289/ehp.0901321.s1

URL : https://hal.archives-ouvertes.fr/hal-01982018

R. Vella, N. Pillon, B. Zarrouki, M. Croze, L. Koppe et al., Ozone Exposure Triggers Insulin Resistance Through Muscle c-Jun N-Terminal Kinase Activation, Diabetes, vol.64, issue.3, pp.1011-1024, 2015.
DOI : 10.2337/db13-1181

URL : https://hal.archives-ouvertes.fr/hal-01850613

A. Gore, V. Chappell, S. Fenton, J. Flaws, A. Nadal et al.,

, The Endocrine Society's Second Scientific Statement on Endocrine-Disrupting Chemicals, Endocrine reviews, vol.36, issue.6, pp.1-150, 2015.

C. Richter, L. Birnbaum, F. Farabollini, R. Newbold, B. Rubin et al., In vivo effects of bisphenol A in laboratory rodent studies, Reproductive Toxicology, vol.24, issue.2, pp.199-224, 2007.
DOI : 10.1016/j.reprotox.2007.06.004

A. Pereira-fernandes, H. Demaegdt, K. Vandermeiren, T. Hectors, P. Jorens et al., Evaluation of a Screening System for Obesogenic Compounds: Screening of Endocrine Disrupting Compounds and Evaluation of the PPAR Dependency of the Effect, PLoS ONE, vol.81, issue.10, p.77481, 2013.
DOI : 10.1371/journal.pone.0077481.t001

J. Wei, X. Sun, Y. Chen, Y. Li, L. Song et al., Perinatal exposure to bisphenol A exacerbates nonalcoholic steatohepatitis-like phenotype in male rat offspring fed on a high-fat diet, Journal of Endocrinology, vol.152, issue.8, pp.313-325, 2014.
DOI : 10.1210/en.2011-0045

M. Garcia-arevalo, P. Alonso-magdalena, J. Servitja, T. Boronat, B. Merin et al.,

G. Gomez, A. Novials, I. Quesada, A. Nadal, . Maternal et al., Endocrinology, vol.2016, p.20161390

M. Garcia-arevalo, P. Alonso-magdalena, R. D. Santos, J. Quesada, I. Carneiro et al., Exposure to Bisphenol-A during Pregnancy Partially Mimics the Effects of a High-Fat Diet Altering Glucose Homeostasis and Gene Expression in Adult Male Mice, PLoS ONE, vol.122, issue.255, p.100214, 2014.
DOI : 10.1371/journal.pone.0100214.s004

P. Alonso-magdalena, I. Quesada, and A. Nadal, Endocrine disruptors in the etiology

, diabetes mellitus, Nature reviews Endocrinology, vol.7, issue.6, pp.346-353, 2011.

P. Alonso-magdalena, I. Quesada, and A. Nadal, Prenatal Exposure to BPA and Offspring Outcomes, Dose-Response, vol.122, issue.2
DOI : 10.1210/en.2012-1422

T. Diabesogenic-behavior and . Bpa, Dose-response : a publication of, International Hormesis Society, vol.13, issue.2, p.1559325815590395, 2015.

B. Angle, R. Do, D. Ponzi, R. Stahlhut, B. Drury et al., Metabolic disruption in male mice due to fetal exposure to low but not high doses of bisphenol A (BPA): Evidence for effects on body weight, food intake, adipocytes, leptin, adiponectin, insulin and glucose regulation, Reproductive Toxicology, vol.42, pp.256-268, 2013.
DOI : 10.1016/j.reprotox.2013.07.017

A. Marmugi, S. Ducheix, F. Lasserre, A. Polizzi, A. Paris et al., Low doses of bisphenol a induce gene expression related to lipid synthesis and trigger triglyceride accumulation in adult mouse liver, Hepatology, vol.18, issue.Pt 1, pp.395-407, 2012.
DOI : 10.1016/j.jnutbio.2006.12.006

URL : https://hal.archives-ouvertes.fr/hal-01019497

E. Hugo, T. Brandebourg, J. Woo, J. Loftus, J. Alexander et al., Bisphenol A at Environmentally Relevant Doses Inhibits Adiponectin Release from Human Adipose Tissue Explants and Adipocytes, Environmental Health Perspectives, vol.116, issue.12, pp.1642-1647, 2008.
DOI : 10.1289/ehp.11537

URL : https://ehp.niehs.nih.gov/wp-content/uploads/116/12/ehp.11537.pdf

C. Menale, A. Grandone, C. Nicolucci, G. Cirillo, S. Crispi et al., Bisphenol A is associated with insulin resistance and modulates adiponectin and resistin gene expression in obese children Pediatric obesity, 2016.
DOI : 10.1111/ijpo.12154

R. Valentino, D. Esposito, V. Passaretti, F. Liotti, A. Cabaro et al., Bisphenol-A Impairs Insulin Action and Up-Regulates Inflammatory Pathways in Human Subcutaneous Adipocytes and 3T3-L1 Cells, PLoS ONE, vol.78, issue.5, p.82099, 2013.
DOI : 10.1371/journal.pone.0082099.t002

URL : https://doi.org/10.1371/journal.pone.0082099

N. Chevalier and P. Fenichel, Bisphenol A: Targeting metabolic tissues Reviews in endocrine & metabolic disorders 2015, pp.299-309
DOI : 10.1007/s11154-016-9333-8

K. Thayer, J. Heindel, J. Bucher, and M. Gallo, Role of Environmental Chemicals in Diabetes and Obesity: A National Toxicology Program Workshop Review, Environmental Health Perspectives, vol.120, issue.6, pp.779-789, 2012.
DOI : 10.1289/ehp.1104597

D. Naville, C. Pinteur, N. Vega, Y. Menade, M. Vigier et al., Low-dose food contaminants trigger sex-specific, hepatic metabolic changes in the progeny of obese mice, The FASEB Journal, vol.64, issue.9, pp.3860-3870, 2013.
DOI : 10.1186/1476-069X-11-42

URL : https://hal.archives-ouvertes.fr/inserm-00833778

D. Naville, E. Labaronne, N. Vega, C. Pinteur, E. Canet-soulas et al., Metabolic Outcome of Female Mice Exposed to a Mixture of Low-Dose Pollutants in a Diet-Induced Obesity Model, PLOS ONE, vol.4, issue.9???10, pp.124015-61, 2015.
DOI : 10.1371/journal.pone.0124015.s006

A. Kortenkamp, Low dose mixture effects of endocrine disrupters and their implications for regulatory thresholds in chemical risk assessment, Current Opinion in Pharmacology, vol.19, pp.105-111, 2014.
DOI : 10.1016/j.coph.2014.08.006

E. Silva, N. Rajapakse, and A. Kortenkamp, Something from ???Nothing??? ??? Eight Weak Estrogenic Chemicals Combined at Concentrations below NOECs Produce Significant Mixture Effects, Environmental Science & Technology, vol.36, issue.8, pp.1751-1756, 2002.
DOI : 10.1021/es0101227

F. Orton, S. Ermler, S. Kugathas, E. Rosivatz, M. Scholze et al., Mixture effects at very low doses with combinations of anti-androgenic pesticides, antioxidants, industrial pollutant and chemicals used in personal care products (EFSA) EFSA. Cumulative risk assessment of pesticides to human health: the way forward, Toxicology and applied pharmacology EFSA Scientific Colloquium Summary Report, vol.278, issue.3, pp.201-208, 2006.

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, Nature Communications, vol.320, issue.1, p.8089, 2015.
DOI : 10.1107/S0907444909042073

URL : https://hal.archives-ouvertes.fr/hal-01838034

D. Sarigiannis and U. Hansen, Considering the cumulative risk of mixtures of chemicals ??? A challenge for policy makers, Environmental Health, vol.11, issue.Suppl 1, p.18, 2012.
DOI : 10.1504/IJRAM.2009.030697

J. Suen, C. Hung, H. Yu, and S. Huang, Alkylphenols???potential modulators of the allergic response, The Kaohsiung Journal of Medical Sciences, vol.28, issue.7, pp.43-48, 2012.
DOI : 10.1016/j.kjms.2012.05.009

URL : https://doi.org/10.1016/j.kjms.2012.05.009

E. Bonefeld-jorgensen, M. Long, M. Hofmeister, and A. Vinggaard, Endocrine-Disrupting Potential of Bisphenol A, Bisphenol A Dimethacrylate, 4-n-Nonylphenol, and 4-n-Octylphenol in Vitro: New Data and a Brief Review, Environmental Health Perspectives, vol.115, issue.S-1, pp.69-76, 2007.
DOI : 10.1289/ehp.9368

K. Schafer and S. Kegley, Persistent toxic chemicals in the US food supply, Journal of Epidemiology & Community Health, vol.56, issue.11, pp.813-817, 2002.
DOI : 10.1136/jech.56.11.813

URL : http://jech.bmj.com/content/jech/56/11/813.full.pdf

M. Angrish, B. Mets, A. Jones, and T. Zacharewski, Dietary Fat Is a Lipid Source in 2,3,7,8-Tetrachlorodibenzo-??-Dioxin (TCDD)-Elicited Hepatic Steatosis in C57BL/6 Mice, Toxicological Sciences, vol.26, issue.Pt 22, pp.377-386, 2012.
DOI : 10.1128/MCB.26.9.3455-3467.2006

URL : https://academic.oup.com/toxsci/article-pdf/128/2/377/16683892/kfs155.pdf

C. Duval, F. Teixeira-clerc, A. Leblanc, S. Touch, C. Emond et al., Chronic Exposure to Low Doses of Dioxin Promotes Liver Fibrosis Development in the C57BL6, J Diet-Induced Obesity Mouse Model. Environmental health perspectives, p.72, 2016.

V. Arsenescu, R. Arsenescu, V. King, H. Swanson, and L. Cassis, Polychlorinated Biphenyl-77 Induces Adipocyte Differentiation and Proinflammatory Adipokines and Promotes Obesity and Atherosclerosis, Environmental Health Perspectives, vol.116, issue.6, pp.761-768, 2008.
DOI : 10.1289/ehp.10554

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2430232/pdf/ehp0116-000761.pdf

X. Liu, G. Yu, Z. Cao, B. Wang, J. Huang et al., Occurrence of organophosphorus flame retardants on skin wipes: Insight into human exposure from dermal absorption, Environment International, vol.98, 2016.
DOI : 10.1016/j.envint.2016.10.021

A. Hoppe and G. Carey, Polybrominated Diphenyl Ethers as Endocrine Disruptors of Adipocyte Metabolism**, Obesity, vol.93, issue.12, pp.2942-2950, 2007.
DOI : 10.1172/JCI200319451

URL : http://onlinelibrary.wiley.com/doi/10.1038/oby.2007.351/pdf

A. Sarkar, D. Ray, A. Shrivastava, and S. Sarker, Molecular Biomarkers: Their significance and application in marine pollution monitoring, Ecotoxicology, vol.73, issue.4, pp.333-340, 2006.
DOI : 10.1016/j.scitotenv.2004.05.008

T. Kanayama, N. Kobayashi, S. Mamiya, T. Nakanishi, and J. Nishikawa, Organotin Compounds Promote Adipocyte Differentiation as Agonists of the Peroxisome Proliferator-Activated Receptor ??/Retinoid X Receptor Pathway, Molecular Pharmacology, vol.67, issue.3, pp.766-774, 2005.
DOI : 10.1124/mol.104.008409

Z. Zuo, S. Chen, T. Wu, J. Zhang, Y. Su et al., Tributyltin causes obesity and hepatic steatosis in male mice, Environmental Toxicology, vol.230, issue.1, pp.79-85, 2011.
DOI : 10.1016/j.tox.2006.10.026

W. Volkel, M. Kiranoglu, and H. Fromme, Determination of free and total bisphenol A in human urine to assess daily uptake as a basis for a valid risk assessment, Toxicology Letters, vol.179, issue.3, pp.155-162, 2008.
DOI : 10.1016/j.toxlet.2008.05.002

A. Krishnan, P. Stathis, S. Permuth, L. Tokes, and D. Feldman, Bisphenol-A: an estrogenic substance is released from polycarbonate flasks during autoclaving., Endocrinology, vol.132, issue.6, pp.2279-2286, 1993.
DOI : 10.1210/endo.132.6.8504731

J. Boucher, S. Ahmed, and E. Atlas, Bisphenol S Induces Adipogenesis in Primary Human Preadipocytes From Female Donors, Endocrinology, vol.157, issue.4, pp.1397-1407, 2016.
DOI : 10.1210/en.2015-1872

A. Brantsaeter, T. Ydersbond, J. Hoppin, M. Haugen, and H. Meltzer, Organic Food in the Diet: Exposure and Health Implications. Annual review of public health, 2016.

T. Ugranli, E. Gungormus, P. Kavcar, E. Demircioglu, M. Odabasi et al., POPs in a major conurbation in Turkey: ambient air concentrations, seasonal variation, inhalation and dermal exposure, and associated carcinogenic risks. Environmental science and pollution research international 2016, pp.22500-22512

G. Howell, C. Mulligan, E. Meek, and J. Chambers, Effect of chronic p , p ???-dichlorodiphenyldichloroethylene (DDE) exposure on high fat diet-induced alterations in glucose and lipid metabolism in male C57BL/6H mice, Toxicology, vol.328, pp.112-122, 2015.
DOI : 10.1016/j.tox.2014.12.017

L. Merrill, M. Karey, E. Moshier, E. Lindtner, C. et al., Perinatal Exposure of Mice to the Pesticide DDT Impairs Energy Expenditure and Metabolism in Adult Female Offspring, PLoS ONE, vol.117, issue.7, p.103337, 2014.
DOI : 10.1371/journal.pone.0103337.s011

S. Lim, S. Ahn, I. Song, M. Chung, H. Jang et al., Chronic Exposure to the Herbicide, Atrazine, Causes Mitochondrial Dysfunction and Insulin Resistance, PLoS ONE, vol.105, issue.4, p.5186, 2009.
DOI : 10.1371/journal.pone.0005186.s001

H. Koch, M. Lorber, K. Christensen, C. Palmke, S. Koslitz et al., Identifying sources of phthalate exposure with human biomonitoring: Results of a 48h fasting study with urine collection and personal activity patterns, International Journal of Hygiene and Environmental Health, vol.216, issue.6, pp.672-681, 2013.
DOI : 10.1016/j.ijheh.2012.12.002

E. Mylchreest, R. Cattley, and P. Foster, -butyl) Phthalate: An Antiandrogenic Mechanism?, Toxicological Sciences, vol.43, issue.1, pp.47-60, 1998.
DOI : 10.1093/toxsci/43.1.47

M. Manikkam, R. Tracey, C. Guerrero-bosagna, and M. Skinner, Plastics Derived Endocrine Disruptors (BPA, DEHP and DBP) Induce Epigenetic Transgenerational Inheritance of Obesity, Reproductive Disease and Sperm Epimutations, PLoS ONE, vol.60, issue.255, p.55387, 2013.
DOI : 10.1371/journal.pone.0055387.s007

C. Hao, X. Cheng, H. Xia, and X. Ma, The endocrine disruptor mono-(2-ethylhexyl) phthalate promotes adipocyte differentiation and induces obesity in mice, Bioscience Reports, vol.278, issue.6, pp.619-629, 2012.
DOI : 10.1074/jbc.M210062200

A. Liem, P. Furst, and C. Rappe, Exposure of populations to dioxins and related compounds, Food Additives and Contaminants, vol.30, issue.4, pp.241-259, 2000.
DOI : 10.1289/ehp.98106775

B. Wahlang, K. Falkner, B. Gregory, D. Ansert, D. Young et al.,

M. Cave, Polychlorinated biphenyl 153 is a diet-dependent obesogen that worsens nonalcoholic fatty liver disease in male C57BL6/J mice, The Journal of nutritional biochemistry, vol.24, issue.9, pp.1587-1595, 2013.

C. Pope, R. Burnett, G. Thurston, M. Thun, E. Calle et al.,

, Cardiovascular mortality and long-term exposure to particulate air pollution: epidemiological evidence of general pathophysiological pathways of disease, Circulation, vol.109, issue.1, pp.71-77, 2004.

N. Telang, M. Katdare, H. Bradlow, and M. Osborne, Estradiol metabolism: an endocrine biomarker for modulation of human mammary carcinogenesis, Environmental Health Perspectives, vol.105, issue.Suppl 3, pp.559-564, 1997.
DOI : 10.1289/ehp.97105s3559

P. Irigaray, V. Ogier, S. Jacquenet, V. Notet, P. Sibille et al.,

. Benzo, adrenergic stimulation of adipose tissue lipolysis and causes weight gain in mice. A novel molecular mechanism of toxicity for a common food pollutant, The FEBS journal, vol.273, issue.7, pp.1362-1372, 2006.

S. Banzhaf, M. Filipovic, J. Lewis, C. Sparrenbom, and R. Barthel, A review of contamination of surface-, ground-, and drinking water in Sweden by perfluoroalkyl and polyfluoroalkyl substances (PFASs), Ambio, vol.524, issue.525, 2016.
DOI : 10.1016/j.scitotenv.2015.03.111

E. Hines, S. White, J. Stanko, E. Gibbs-flournoy, C. Lau et al., Phenotypic dichotomy following developmental exposure to perfluorooctanoic acid (PFOA) in female CD-1 mice: Low doses induce elevated serum leptin and insulin, and overweight in mid-life???, Molecular and Cellular Endocrinology, vol.304, issue.1-2, pp.97-105, 2009.
DOI : 10.1016/j.mce.2009.02.021

URL : https://zenodo.org/record/1259213/files/article.pdf

W. Fan, T. Yanase, M. Nomura, T. Okabe, K. Goto et al.,

, Androgen receptor null male mice develop late-onset obesity caused by decreased energy expenditure and lipolytic activity but show normal insulin sensitivity with high adiponectin secretion, Diabetes, vol.54, issue.4, pp.1000-1008, 2005.

M. Byerly, A. Salayta, M. Swanson, R. Kwon, K. Peterson et al., Estrogen-related receptor ?? deletion modulates whole-body energy balance via estrogen-related receptor ?? and attenuates neuropeptide Y gene expression, European Journal of Neuroscience, vol.15, issue.Suppl 1, pp.1033-1047, 2013.
DOI : 10.1016/j.bmcl.2005.01.025

URL : http://europepmc.org/articles/pmc3618562?pdf=render

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, Journal of Biological Chemistry, vol.34, issue.38, pp.25984-25992, 2009.
DOI : 10.2337/diab.34.9.911

URL : http://www.jbc.org/content/284/38/25984.full.pdf

C. Wang, C. Xu, S. Krager, K. Bottum, D. Liao et al., Aryl Hydrocarbon Receptor Deficiency Enhances Insulin Sensitivity and Reduces PPAR-?? Pathway Activity in Mice, Environmental Health Perspectives, vol.119, issue.12, pp.1739-1744, 2011.
DOI : 10.1289/ehp.1103593

URL : https://ehp.niehs.nih.gov/doi/pdf/10.1289/ehp.1103593

C. Xu, C. Wang, Z. Zhang, C. Jaeger, S. Krager et al., Aryl hydrocarbon receptor deficiency protects mice from diet-induced adiposity and metabolic disorders through increased energy expenditure, International Journal of Obesity, vol.15, issue.8, pp.1300-1309
DOI : 10.3390/ijms150711700

URL : http://europepmc.org/articles/pmc4526411?pdf=render

M. Guerre-millo, C. Rouault, P. Poulain, J. Andre, V. Poitout et al., PPAR-??-Null Mice Are Protected From High-Fat Diet-Induced Insulin Resistance, Diabetes, vol.50, issue.12, pp.2809-2814, 2001.
DOI : 10.2337/diabetes.50.12.2809

URL : http://diabetes.diabetesjournals.org/content/50/12/2809.full.pdf

J. He, J. Gao, M. Xu, S. Ren, M. Stefanovic-racic et al., PXR Ablation Alleviates Diet-Induced and Genetic Obesity and Insulin Resistance in Mice, Diabetes, vol.62, issue.6, pp.1876-1887, 2013.
DOI : 10.2337/db12-1039

URL : http://diabetes.diabetesjournals.org/content/diabetes/62/6/1876.full.pdf

P. Thomas and J. Dong, Binding and activation of the seven-transmembrane estrogen receptor GPR30 by environmental estrogens: A potential novel mechanism of endocrine disruption, The Journal of Steroid Biochemistry and Molecular Biology, vol.102, issue.1-5, pp.175-179, 2006.
DOI : 10.1016/j.jsbmb.2006.09.017

H. Lee, S. Chattopadhyay, E. Gong, R. Ahn, and K. Lee, Antiandrogenic Effects of Bisphenol A and Nonylphenol on the Function of Androgen Receptor, Toxicological Sciences, vol.75, issue.1, pp.40-46, 2003.
DOI : 10.1093/toxsci/kfg150

R. Sargis, D. Johnson, R. Choudhury, and M. Brady, Environmental Endocrine Disruptors Promote Adipogenesis in the 3T3-L1 Cell Line through Glucocorticoid Receptor Activation, Obesity, vol.43, issue.7, pp.1283-1288, 2010.
DOI : 10.1210/jcem-67-6-1122

URL : http://europepmc.org/articles/pmc3957336?pdf=render

M. Rehan, E. Ahmad, I. Sheikh, A. Abuzenadah, G. Damanhouri et al., Androgen and Progesterone Receptors Are Targets for Bisphenol A (BPA), 4-Methyl-2,4-bis-(P-Hydroxyphenyl)Pent-1-Ene???A Potent Metabolite of BPA, and 4-Tert-Octylphenol: A Computational Insight, PLOS ONE, vol.78, issue.9, p.138438, 2015.
DOI : 10.1371/journal.pone.0138438.t008

K. Moriyama, T. Tagami, T. Akamizu, T. Usui, M. Saijo et al., Thyroid Hormone Action Is Disrupted by Bisphenol A as an Antagonist, The Journal of Clinical Endocrinology & Metabolism, vol.87, issue.11, pp.5185-5190, 2002.
DOI : 10.1210/jc.2002-020209

, 109

P. Darnerud, Brominated flame retardants as possible endocrine disrupters, International Journal of Andrology, vol.68, issue.2, pp.152-160, 2008.
DOI : 10.1093/toxsci/66.1.105

C. Hurst and D. Waxman, Activation of PPAR?? and PPAR?? by Environmental Phthalate Monoesters, Toxicological Sciences, vol.274, issue.5, pp.297-308, 2003.
DOI : 10.1074/jbc.274.5.2672

X. Cheng and C. Klaassen, Perfluorocarboxylic Acids Induce Cytochrome P450 Enzymes in Mouse Liver through Activation of PPAR-?? and CAR Transcription Factors, Toxicological Sciences, vol.23, issue.1, pp.29-36, 2008.
DOI : 10.1089/1044549041562267

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, Toxicology and Applied Pharmacology, vol.217, issue.3, pp.235-244, 2006.
DOI : 10.1016/j.taap.2006.08.011

J. Feige, L. Gelman, D. Rossi, V. Zoete, R. Metivier et al., The Endocrine Disruptor Monoethyl-hexyl-phthalate Is a Selective Peroxisome Proliferator-activated Receptor ?? Modulator That Promotes Adipogenesis, Journal of Biological Chemistry, vol.54, issue.26, pp.19152-19166, 2007.
DOI : 10.1210/me.2005-0367

URL : https://hal.archives-ouvertes.fr/hal-00177041

L. Li, Q. Wang, Y. Zhang, Y. Niu, X. Yao et al., The Molecular Mechanism of Bisphenol A (BPA) as an Endocrine Disruptor by Interacting with Nuclear Receptors: Insights from Molecular Dynamics (MD) Simulations, PLOS ONE, vol.98, issue.3, p.120330, 2015.
DOI : 10.1371/journal.pone.0120330.t002

C. Hsu, J. Zhao, R. Huang, J. Hsieh, J. Hamm et al., Quantitative High-Throughput Profiling of Environmental Chemicals and Drugs that Modulate Farnesoid X Receptor, Scientific Reports, vol.4, issue.1, pp.6437-25257666, 2014.
DOI : 10.1038/srep05664

URL : https://www.nature.com/articles/srep06437.pdf

A. Eveillard, L. Mselli-lakhal, A. Mogha, F. Lasserre, A. Polizzi et al., Di-(2-ethylhexyl)-phthalate (DEHP) activates the constitutive androstane receptor (CAR): A novel signalling pathway sensitive to phthalates, Biochemical Pharmacology, vol.77, issue.11, pp.1735-1746, 2009.
DOI : 10.1016/j.bcp.2009.02.023

URL : https://hal.archives-ouvertes.fr/hal-00493499

J. Dekeyser, E. Laurenzana, E. Peterson, T. Chen, and C. Omiecinski, Selective Phthalate Activation of Naturally Occurring Human Constitutive Androstane Receptor Splice Variants and the Pregnane X Receptor, Toxicological Sciences, vol.16, issue.2, pp.381-391, 2011.
DOI : 10.1016/j.molcel.2004.11.042

M. Mozzicafreddo, M. Cuccioloni, L. Bonfili, V. Cecarini, F. Palermo et al., Environmental pollutants directly affect the liver X receptor alpha activity: Kinetic and thermodynamic characterization of binding, The Journal of Steroid Biochemistry and Molecular Biology, vol.152, pp.1-7, 2015.
DOI : 10.1016/j.jsbmb.2015.04.011

A. Takeshita, N. Koibuchi, J. Oka, M. Taguchi, Y. Shishiba et al., Bisphenol-A, an environmental estrogen, activates the human orphan nuclear receptor, steroid and xenobiotic receptor-mediated transcription, European Journal of Endocrinology, vol.145, issue.4, pp.513-517, 2001.
DOI : 10.1530/eje.0.1450513

M. Milnes, A. Garcia, E. Grossman, F. Grun, J. Shiotsugu et al.,

H. Watanabe, T. Iguchi, and B. Blumberg, Activation of steroid and xenobiotic receptor (SXR, NR1I2) and its orthologs in laboratory, toxicologic, and genome model species, Environmental health perspectives, vol.116, issue.7, pp.880-885, 2008.

M. Denison and S. Heath-pagliuso, The Ah Receptor: A Regulator of the Biochemical and Toxicological Actions of Structurally Diverse Chemicals, Bulletin of Environmental Contamination and Toxicology, vol.61, issue.5, pp.557-568, 1998.
DOI : 10.1007/PL00002973

S. Sato, H. Shirakawa, S. Tomita, Y. Ohsaki, K. Haketa et al., Low-dose dioxins alter gene expression related to cholesterol biosynthesis, lipogenesis, and glucose metabolism through the aryl hydrocarbon receptor-mediated pathway in mouse liver, Toxicology and Applied Pharmacology, vol.229, issue.1, pp.10-19, 2008.
DOI : 10.1016/j.taap.2007.12.029