P. Germain, P. Chambon, G. Eichele, R. M. Evans, M. A. Lazar et al., International union of pharmacology. LXIII. Retinoid X receptors, Pharmacol Rev, vol.58, pp.760-72, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00187925

H. Gronemeyer, J. A. Gustafsson, and V. Laudet, Principles for modulation of the nuclear receptor superfamily, Nat Rev Drug Discov, vol.3, pp.950-64, 2004.

W. Bourguet, P. Germain, and H. Gronemeyer, Nuclear receptor ligand-binding domains: three-dimensional structures, molecular interactions and pharmacological implications, Trends Pharmacol Sci, vol.21, pp.381-389, 2000.
DOI : 10.1016/s0165-6147(00)01548-0

A. C. Pike, Lessons learnt from structural studies of the oestrogen receptor, Best Pract Res Clin Endocrinol Metab, vol.20, pp.1-14, 2006.

J. P. Renaud and D. Moras, Structural studies on nuclear receptors, Cell Mol Life Sci, vol.57, pp.1748-69, 2000.
DOI : 10.1007/pl00000656

A. Benecke, P. Chambon, and H. Gronemeyer, Synergy between estrogen receptor alpha activation functions AF1 and AF2 mediated by transcription intermediary factor TIF2, EMBO Rep, vol.1, pp.151-158, 2000.
DOI : 10.1093/embo-reports/kvd028

URL : http://embor.embopress.org/content/1/2/151.full.pdf

M. Bommer, A. Benecke, H. Gronemeyer, R. , and C. , TIF2 mediates the synergy between RARalpha 1 activation functions AF-1 and AF-2, J Biol Chem, vol.277, pp.37961-37967, 2002.

E. M. Wilson, Analysis of interdomain interactions of the androgen receptor, Methods Mol Biol, vol.776, pp.113-142, 2011.

E. Diamanti-kandarakis, J. P. Bourguignon, L. C. Giudice, R. Hauser, G. S. Prins et al., Endocrine-disrupting chemicals: an endocrine society scientific statement, Endocr Rev, vol.30, pp.293-342, 2009.
DOI : 10.1210/er.2009-0002

URL : https://academic.oup.com/edrv/article-pdf/30/4/293/10334449/edrv0293.pdf

A. K. Hotchkiss, C. V. Rider, C. R. Blystone, V. S. Wilson, P. C. Hartig et al., Fifteen years after "Wingspread" -environmental endocrine disrupters and human and wildlife health: where we are today and where we need to go, Toxicol Sci, vol.105, pp.235-59, 2008.

E. Swedenborg, J. Ruegg, S. Makela, and I. Pongratz, Endocrine disruptive chemicals: mechanisms of action and involvement in metabolic disorders, J Mol Endocrinol, vol.43, pp.1-10, 2009.

M. M. Tabb and B. Blumberg, New modes of action for endocrine-disrupting chemicals, Mol Endocrinol, vol.20, pp.475-82, 2006.

J. Janosek, K. Hilscherova, L. Blaha, and I. Holoubek, Environmental xenobiotics and nuclear receptors -interactions, effects and in vitro assessment, Toxicol In vitro, vol.20, pp.18-37, 2006.

J. Toppari, Environmental endocrine disrupters, Sex Dev, vol.2, pp.260-267, 2008.
DOI : 10.1159/000152042

J. F. Couse and K. S. Korach, Estrogen receptor null mice: what have we learned and where will they lead us?, Endocr Rev, vol.20, pp.358-417, 1999.
DOI : 10.1210/edrv.20.3.0370

URL : https://academic.oup.com/edrv/article-pdf/20/3/358/8864683/edrv0358.pdf

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 beta, Endocrinology, vol.139, pp.4252-63, 1998.

M. M. Liu, C. Albanese, C. M. Anderson, K. Hilty, P. Webb et al., Opposing action of estrogen receptors alpha and beta on cyclin D1 gene expression, J Biol Chem, vol.277, pp.24353-60, 2002.

K. Pettersson, F. Delaunay, and J. A. Gustafsson, Estrogen receptor beta acts as a dominant regulator of estrogen signaling, Oncogene, vol.19, pp.4970-4978, 2000.

M. K. Lindberg, S. Movérare, S. Skrtic, H. Gao, K. Dahlman-wright et al., Estrogen receptor (ER)-beta reduces ERalpha-regulated gene transcription, supporting a "ying yang" relationship between ERalpha and ERbeta in mice, Mol Endocrinol, vol.17, pp.203-211, 2003.

S. Ogawa, V. Eng, J. Taylor, D. B. Lubahn, K. S. Korach et al., Roles of estrogen receptor-alpha gene expression in reproduction-related behaviors in female mice, Endocrinology, vol.139, pp.5070-81, 1998.

Z. Weihua, S. Saji, S. Mäkinen, G. Cheng, E. V. Jensen et al., Estrogen receptor (ER) beta, a modulator of ERalpha in the uterus, Proc Natl Acad Sci U S A, vol.97, pp.5936-5977, 2000.

G. H. Veeneman, Non-steroidal subtype selective estrogens, Curr Med Chem, vol.12, pp.1077-136, 2005.
DOI : 10.2174/0929867053764662

P. Balaguer, F. François, F. Comunale, H. Fenet, A. M. Boussioux et al., Reporter cell lines to study the estrogenic effects of xenoestrogens, Sci Total Environ, vol.233, pp.47-56, 1999.

A. Docquier, A. Garcia, J. Savatier, A. Boulahtouf, S. Bonnet et al., Negative regulation of estrogen signaling by ER? and RIP140 in ovarian cancer cells, Mol Endocrinol, vol.27, pp.1429-1470, 2013.

M. Kojima, K. Fukunaga, M. Sasaki, M. Nakamura, M. Tsuji et al., Evaluation of estrogenic activities of pesticides using an in vitro reporter gene assay, Int J Environ Health Res, vol.15, pp.271-80, 2005.

V. S. Wilson, K. Bobseine, and L. E. Gray, Development and characterization of a cell line that stably expresses an estrogen-responsive luciferase reporter for the detection of estrogen receptor agonist and antagonists, Toxicol Sci, vol.81, pp.69-77, 2004.

T. Barkhem, B. Carlsson, Y. Nilsson, E. Enmark, J. Gustafsson et al., Differential response of estrogen receptor alpha and estrogen receptor beta to partial estrogen agonists/antagonists, Mol Pharmacol, vol.54, pp.105-117, 1998.

E. K. Shanle, J. R. Hawse, and W. Xu, Generation of stable reporter breast cancer cell lines for the identification of ER subtype selective ligands, Biochem Pharmacol, vol.82, pp.1940-1949, 2011.

B. Van-der-burg, R. Winter, M. Weimer, P. Berckmans, G. Suzuki et al., Optimization and prevalidation of the in vitro ERalpha CALUX method to test estrogenic and antiestrogenic activity of compounds, Reprod Toxicol, vol.30, pp.73-80, 2010.

A. Escande, A. Pillon, N. Servant, J. P. Cravedi, F. Larrea et al., Evaluation of ligand selectivity using reporter cell lines stably expressing estrogen receptor alpha or beta, Biochem Pharmacol, vol.71, pp.1459-69, 2006.

J. M. Molina-molina, M. Real, I. Jimenez-diaz, H. Belhassen, A. Hedhili et al., Assessment of estrogenic and anti-androgenic activities of the mycotoxin zearalenone and its metabolites using in vitro receptor-specific bioassays, Food Chem Toxicol, vol.74, pp.233-242, 2014.

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.1306-1319, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-01076600

E. Gomez, A. Pillon, H. Fenet, D. Rosain, M. J. Duchesne et al., Estrogenic activity of cosmetic components in reporter cell lines: parabens, UV screens, and musks, J Toxicol Environ Health A, vol.68, pp.239-51, 2005.

J. M. Molina-molina, A. Escande, A. Pillon, E. Gomez, F. Pakdel et al., Profiling of benzophenone derivatives using fish and human estrogen receptor-specific in vitro bioassays, Toxicol Appl Pharmacol, vol.232, pp.384-95, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00319498

V. Delfosse, M. Grimaldi, J. L. Pons, A. Boulahtouf, A. Le-maire et al., Structural and mechanistic insights into bisphenols action provide guidelines for risk assessment and discovery of bisphenol A substitutes, Proc Natl Acad Sci U S A, vol.109, pp.14930-14935, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00726779

J. M. Molina-molina, E. Amaya, M. Grimaldi, J. M. Sáenz, M. Real et al., In vitro study on the agonistic and antagonistic activities of bisphenol-S and other bisphenol-A congeners and derivatives via nuclear receptors, Toxicol Appl Pharmacol, vol.272, pp.127-163, 2013.

A. Riu, A. Le-maire, M. Grimaldi, M. Audebert, A. Hillenweck et al., Characterization of novel ligands of ER?, ER?, and PPAR?: the case of halogenated bisphenol A and their conjugated metabolites, Toxicol Sci, vol.122, pp.372-82, 2011.

T. Matsumoto, M. Sakari, M. Okada, A. Yokoyama, S. Takahashi et al., The androgen receptor in health and disease, Annu Rev Physiol, vol.75, pp.201-225, 2013.

B. He, J. A. Kemppainen, and E. M. Wilson, FXXLF and WXXLF sequences mediate the NH2-terminal interaction with the ligand binding domain of the androgen receptor, J Biol Chem, vol.275, pp.22986-94, 2000.

V. S. Wilson, K. Bobseine, C. R. Lambright, and L. E. Gray, A novel cell line, MDA-kb2, that stably expresses an androgen-and glucocorticoid-responsive reporter for the detection of hormone receptor agonists and antagonists, Toxicol Sci, vol.66, pp.69-81, 2002.

B. Térouanne, B. Tahiri, V. Georget, C. Belon, N. Poujol et al., A stable prostatic bioluminescent cell line to investigate androgen and antiandrogen effects, Mol Cell Endocrinol, vol.160, pp.251-259, 2000.

F. Paris, N. Servant, B. Térouanne, and C. Sultan, Evaluation of androgenic bioactivity in human serum by recombinant cell line: preliminary results, Mol Cell Endocrinol, vol.198, pp.123-132, 2002.

S. Aït-aïssa, S. Laskowski, N. Laville, J. M. Porcher, and F. Brion, Anti-androgenic activities of environmental pesticides in the MDA-kb2 reporter cell line, Toxicol In vitro, vol.24, pp.1979-85, 2010.

J. M. Molina-molina, A. Hillenweck, I. Jouanin, D. Zalko, J. P. Cravedi et al., Steroid receptor profiling of vinclozolin and its primary metabolites, Toxicol Appl Pharmacol, vol.216, pp.44-54, 2006.

V. Christen, P. Crettaz, A. Oberli-schrämmli, and K. Fent, Antiandrogenic activity of phthalate mixtures: validity of concentration addition, Toxicol Appl Pharmacol, vol.259, pp.169-76, 2012.

V. Christen, P. Crettaz, and K. Fent, Additive and synergistic antiandrogenic activities of mixtures of azol fungicides and vinclozolin, Toxicol Appl Pharmacol, vol.279, pp.455-66, 2014.

V. Bellet, G. Hernandez-raquet, S. Dagnino, L. Seree, P. Pardon et al., Occurrence of androgens in sewage treatment plants influents is associated with antagonist activities on other steroid receptors, Water Res, vol.46, pp.1912-1934, 2012.
URL : https://hal.archives-ouvertes.fr/ineris-00963355

N. Creusot, S. Aït-aïssa, N. Tapie, P. Pardon, F. Brion et al., Identification of synthetic steroids in river water downstream from pharmaceutical manufacture discharges based on a bioanalytical approach and passive sampling, Environ Sci Technol, vol.48, pp.18-24, 2010.
URL : https://hal.archives-ouvertes.fr/ineris-01855483

C. Sultan, P. Balaguer, B. Terouanne, V. Georget, F. Paris et al., Environmental xenoestrogens, antiandrogens and disorders of male sexual differentiation, Mol Cell Endocrinol, vol.178, pp.99-105, 2001.

V. Giguère, To ERR in the estrogen pathway, Trends Endocrinol Metab, vol.13, pp.220-225, 2002.

A. L. Bookout, Y. Jeong, M. Downes, R. T. Yu, R. M. Evans et al., Anatomical profiling of nuclear receptor expression reveals a hierarchical transcriptional network, Cell, vol.126, pp.789-99, 2006.

E. Audet-walsh and V. Giguére, The multiple universes of estrogen-related receptor ? and ? in metabolic control and related diseases, Acta Pharmacol Sin, vol.36, pp.51-61, 2015.

P. F. Baillie-hamilton, Chemical toxins: a hypothesis to explain the global obesity epidemic, J Altern Complement Med, vol.8, pp.185-92, 2002.

N. Ijichi, T. Shigekawa, K. Ikeda, K. Horie-inoue, T. Fujimura et al., Estrogen-related receptor ? modulates cell proliferation and estrogen signaling in breast cancer, J Steroid Biochem Mol Biol, vol.123, pp.1-7, 2011.
DOI : 10.1016/j.jsbmb.2010.09.002

D. Lu, Y. Kiriyama, K. Y. Lee, and V. Giguere, Transcriptional regulation of the estrogen-inducible pS2 breast cancer marker gene by the ERR family of orphan nuclear receptors, Cancer Res, vol.61, pp.6755-61, 2001.

J. M. Vanacker, K. Pettersson, J. A. Gustafsson, and V. Laudet, Transcriptional targets shared by estrogen receptor-related receptors (ERRs) and estrogen receptor (ER) alpha, but not by ERbeta, EMBO J, vol.18, pp.4270-4279, 1999.
DOI : 10.1093/emboj/18.15.4270

URL : http://emboj.embopress.org/content/18/15/4270.full.pdf

E. A. Ariazi, G. M. Clark, and J. E. Mertz, Estrogen-related receptor alpha and estrogenrelated receptor gamma associate with unfavorable and favorable biomarkers, respectively, in human breast cancer, Cancer Res, vol.62, pp.6510-6518, 2002.

S. Yu, X. Wang, C. F. Ng, S. Chen, and F. L. Chan, ERRgamma suppresses cell proliferation and tumor growth of androgen-sensitive and androgeninsensitive prostate cancer cells and its implication as a therapeutic target for prostate cancer, Cancer Res, vol.67, pp.4904-4918, 2007.

H. Greschik, J. M. Wurtz, S. Sanglier, W. Bourguet, A. Van-dorsselaer et al., Structural and functional evidence for ligand-independent transcriptional activation by the estrogen-related receptor 3, Mol Cell, vol.9, pp.303-316, 2002.

J. Li, M. Ma, and Z. Wang, In vitro profiling of endocrine disrupting effects of phenols, Toxicol In vitro, vol.24, pp.201-208, 2010.

H. Okada, T. Tokunaga, X. Liu, S. Takayanagi, A. Matsushima et al., Direct evidence revealing structural elements essential for the high binding ability of bisphenol A to human estrogen-related receptor-gamma, Environ Health Perspect, vol.116, pp.32-40, 2008.

M. Suetsugi, L. Su, K. Karlsberg, Y. C. Yuan, and S. Chen, Flavone and isoflavone phytoestrogens are agonists of estrogen related receptors, Mol Cancer Res, vol.1, pp.981-91, 2003.

C. Yang and S. Chen, Two organochlorine pesticides, toxaphene and chlordane, are antagonists for estrogen related receptor alpha-1 orphan receptor, Cancer Res, vol.59, pp.4519-4543, 1999.

J. Wang, F. Fang, Z. Huang, Y. Wang, and C. Wong, Kaempferol is an estrogenrelated receptor alpha and gamma inverse agonist, FEBS Lett, vol.5, pp.83643-83650, 2009.
DOI : 10.1016/j.febslet.2009.01.030

URL : https://febs.onlinelibrary.wiley.com/doi/pdf/10.1016/j.febslet.2009.01.030

J. Berger and D. E. Moller, The mechanisms of action of PPARs, Annu Rev Med, vol.53, pp.409-444, 2002.

L. Michalik, J. Auwerx, J. P. Berger, V. K. Chatterjee, C. K. Glass et al., International union of pharmacology. LXI. Peroxisome proliferator-activated receptors, Pharmacol Rev, vol.58, pp.726-767, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00188041

T. Varga, Z. Czimmerer, and L. Nagy, PPARs are a unique set of fatty acid regulated transcription factors controlling both lipid metabolism and inflammation, Biochim Biophys Acta, vol.1812, pp.1007-1029, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00706534

P. Tontonoz and B. M. Spiegelman, Fat and beyond: the diverse biology of PPARgamma, Annu Rev Biochem, vol.77, pp.289-312, 2008.

M. Ahmadian, J. M. Suh, N. Hah, C. Liddle, A. R. Atkins et al., PPAR? signaling and metabolism: the good, the bad and the future, Nat Med, vol.19, pp.557-66, 2013.

B. Desvergne and W. Wahli, Peroxisome proliferator-activated receptors: nuclear control of metabolism, Endocr Rev, vol.20, pp.649-88, 1999.

A. Le-maire, M. Grimaldi, D. Roecklin, S. Dagnino, V. Vivat-hannah et al., Activation of RXR-PPAR heterodimers by organotin environmental endocrine disruptors, EMBO Rep, vol.10, pp.367-73, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-00375675

A. Riu, M. Grimaldi, A. Le-maire, G. Bey, K. Phillips et al., Peroxisome proliferator-activated receptor ? is a target for halogenated analogs of bisphenol A, Environ Health Perspect, vol.119, pp.1227-1259, 2011.

A. Janesick and B. Blumberg, Minireview: PPARgamma as the target of obesogens, J Steroid Biochem Mol Biol, vol.127, pp.4-8, 2011.

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

A. Janesick and B. Blumberg, Obesogens, stem cells and the developmental programming of obesity, Int J Androl, vol.35, pp.437-485, 2012.

L. Gijsbers, H. Y. Man, S. K. Kloet, L. H. De-haan, J. Keijer et al., Stable reporter cell lines for peroxisome proliferator-activated receptor ? (PPAR?)-mediated modulation of gene expression, Anal Biochem, vol.414, pp.77-83, 2011.

M. Seimandi, G. Lemaire, A. Pillon, A. Perrin, I. Carlavan et al., Differential responses of PPARalpha, PPARdelta, and PPARgamma reporter cell lines to selective PPAR synthetic ligands, Anal Biochem, vol.344, pp.8-15, 2005.

T. R. Zoeller, A. L. Dowling, C. T. Herzig, E. A. Iannacone, K. J. Gauger et al., Thyroid hormone, brain development, and the environment, Environ Health Perspect, vol.110, pp.355-61, 2002.

J. Bernal, Thyroid hormone receptors in brain development and function, Nat Clin Pract Endocrinol Metab, vol.3, pp.249-59, 2007.

K. M. Crofton, Thyroid disrupting chemicals: mechanisms and mixtures, Int J Androl, vol.31, pp.209-232, 2008.
DOI : 10.1111/j.1365-2605.2007.00857.x

J. Freitas, P. Cano, C. Craig-veit, M. L. Goodson, J. D. Furlow et al., Detection of thyroid hormone receptor disruptors by a novel stable in vitro reporter gene assay, Toxicol In vitro, vol.25, pp.257-66, 2011.

M. L. Jugan, M. Lévy-bimbot, M. Pomérance, S. Tamisier-karolak, J. P. Blondeau et al., A new bioluminescent cellular assay to measure the transcriptional effects of chemicals that modulate the alpha-1 thyroid hormone receptor, Toxicol In vitro, vol.21, pp.1197-205, 2007.

J. B. Fini, A. Riu, L. Debrauwer, A. Hillenweck, L. Mével et al., Parallel biotransformation of tetrabromobisphenol A in Xenopus laevis and mammals: Xenopus as a model for endocrine perturbation studies, Toxicol Sci, vol.125, pp.359-67, 2012.

J. Orans, D. G. Teotico, and M. R. Redinbo, The nuclear xenobiotic receptor pregnane X receptor: recent insights and new challenges, Mol Endocrinol, vol.19, pp.2891-900, 2005.

A. Di-masi, D. Marinis, E. Ascenzi, P. Marino, and M. , Nuclear receptors CAR and PXR: molecular, functional, and biomedical aspects, Mol Aspects Med, vol.30, pp.297-343, 2009.

E. Mikamo, S. Harada, J. Nishikawa, and T. Nishihara, Endocrine disruptors induce cytochrome P450 by affecting transcriptional regulation via pregnane X receptor, Toxicol Appl Pharmacol, vol.193, pp.66-72, 2003.

Y. Sui, S. H. Park, R. N. Helsley, M. Sunkara, F. J. Gonzalez et al., Bisphenol A increases atherosclerosis in pregnane X receptor-humanized ApoE deficient mice, J Am Heart Assoc, vol.3, p.492, 2014.

N. K. Chaturvedi, S. Kumar, S. Negi, and R. K. Tyagi, Endocrine disruptors provoke differential modulatory responses on androgen receptor and pregnane and xenobiotic receptor: potential implications in metabolic disorders, Mol Cell Biochem, vol.345, pp.291-308, 2010.

M. Banerjee, D. Robbins, and T. Chen, Targeting xenobiotic receptors PXR and CAR in human diseases, Drug Discov Today, 2014.

H. Wang, M. Venkatesh, H. Li, R. Goetz, S. Mukherjee et al., Pregnane X receptor activation induces FGF19-dependent tumor aggressiveness in humans and mice, J Clin Invest, vol.121, pp.3220-3252, 2011.

J. M. Lehmann, D. D. Mckee, M. A. Watson, T. M. Willson, J. T. Moore et al., The human orphan nuclear receptor PXR is activated by compounds that regulate CYP3A4 gene expression and cause drug interactions, J Clin Invest, vol.102, pp.1016-1039, 1998.

T. W. Synold, I. Dussault, and B. M. Forman, The orphan nuclear receptor SXR coordinately regulates drug metabolism and efflux, Nat Med, vol.7, pp.584-90, 2001.

R. E. Watkins, J. M. Maglich, L. B. Moore, G. B. Wisely, S. M. Noble et al., A crystal structure of human PXR in complex with the St. John's wort compound hyperforin, Biochemistry, vol.42, pp.1430-1438, 2003.

J. M. Wentworth, M. Agostini, J. Love, J. W. Schwabe, and V. K. Chatterjee, St John's wort, a herbal antidepressant, activates the steroid X receptor, J Endocrinol, vol.166, pp.11-17, 2000.

G. Lemaire, G. De-sousa, and R. Rahmani, A PXR reporter gene assay in a stable cell culture system: CYP3A4 and CYP2B6 induction by pesticides, Biochem Pharmacol, vol.68, pp.2347-58, 2004.

M. Ratajewski, I. Grzelak, K. Wisniewska, K. Ryba, M. Gorzkiewicz et al., Screening of a chemical library reveals novel PXRactivating pharmacologic compounds, Toxicol Lett, vol.232, pp.193-202, 2014.

J. Raucy, L. Warfe, M. F. Yueh, and S. W. Allen, A cell-based reporter gene assay for determining induction of CYP3A4 in a high-volume system, J Pharmacol Exp Ther, vol.303, pp.412-435, 2002.

M. Sekimoto, S. Sano, T. Hosaka, K. Nemoto, and M. Degawa, Establishment of a stable human cell line, HPL-A3, for use in reporter gene assays of cytochrome P450 3A inducers, Biol Pharm Bull, vol.35, pp.677-85, 2012.

G. Lemaire, W. Mnif, P. Mauvais, P. Balaguer, and R. Rahmani, Activation of alphaand beta-estrogen receptors by persistent pesticides in reporter cell lines, Life Sci, vol.79, pp.1160-1169, 2006.

G. Lemaire, W. Mnif, J. M. 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.501-510, 2006.

W. Mnif, J. M. Pascussi, A. Pillon, A. Escande, A. Bartegi et al., Estrogens and antiestrogens activate hPXR, Toxicol Lett, vol.170, pp.19-29, 2007.

N. Creusot, S. Kinani, P. Balaguer, N. Tapie, K. Lemenach et al., Evaluation of an hPXR reporter gene assay for the detection of aquatic emerging pollutants: screening of chemicals and application to water samples, Anal Bioanal Chem, vol.396, pp.569-83, 2010.
URL : https://hal.archives-ouvertes.fr/ineris-00453470

D. J. Mangelsdorf and R. M. Evans, The RXR heterodimers and orphan receptors, Cell, vol.83, pp.841-50, 1995.
DOI : 10.1016/0092-8674(95)90200-7

URL : https://doi.org/10.1016/0092-8674(95)90200-7

P. Germain, J. Iyer, C. Zechel, and H. Gronemeyer, Co-regulator recruitment and the mechanism of retinoic acid receptor synergy, Nature, vol.415, pp.187-92, 2002.

R. A. Heyman, D. J. Mangelsdorf, J. A. Dyck, R. B. Stein, G. Eichele et al., 9-cis retinoic acid is a high affinity ligand for the retinoid X receptor, Cell, vol.68, pp.397-406, 1992.

A. M. De-urquiza, S. Liu, M. Sjoberg, R. H. Zetterstrom, W. Griffiths et al., Docosahexaenoic acid, a ligand for the retinoid X receptor in mouse brain, Science, vol.290, pp.2140-2144, 2000.

V. Nahoum, E. Pérez, P. Germain, F. Rodríguez-barrios, F. Manzo et al., Modulators of the structural dynamics of the retinoid X receptor to reveal receptor function, Proc Natl Acad Sci U S A, vol.104, pp.17323-17331, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00188372

A. Ropero, A. B. Soriano, S. García-arévalo, M. Ripoll, C. Fuentes et al., Bisphenol-A acts as a potent estrogen via non-classical estrogen triggered pathways, Mol Cell Endocrinol, vol.355, pp.201-208, 2012.

H. Ajj, A. Chesnel, S. Pinel, F. Plenat, S. Flament et al., An alkylphenol mix promotes seminoma derived cell proliferation through an ERalpha36-mediated mechanism, PLoS One, vol.8, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00840880

N. Chevalier, A. Vega, A. Bouskine, B. Siddeek, J. F. Michiels et al., GPR30, the non-classical membrane G protein related estrogen receptor, is overexpressed in human seminoma and promotes seminoma cell proliferation, PLoS One, vol.7, p.34672, 2012.

A. Kortenkamp, Low dose mixture effects of endocrine disrupters and their implications for regulatory thresholds in chemical risk assessment, Curr Opin Pharmacol, vol.19, pp.105-116, 2014.

D. Montes-grajales and J. Olivero-verbel, Computer-aided identification of novel protein targets of bisphenol A, Toxicol Lett, vol.222, pp.312-332, 2013.

F. M. Mcrobb, I. Kufareva, and R. Abagyan, In silico identification and pharmacological evaluation of novel endocrine disrupting chemicals that act via the ligand-binding domain of the estrogen receptor ?, Toxicol Sci, vol.141, pp.188-97, 2014.

A. Vuorinen, A. Odermatt, and D. Schuster, In silico methods in the discovery of endocrine disrupting chemicals, J Steroid Biochem Mol Biol, vol.137, pp.18-26, 2013.