V. Pelekanou and G. Leclercq, Recent insights into the effect of natural and environmental estrogens on mammary development and carcinogenesis, The International Journal of Developmental Biology, vol.55, issue.7-8-9, pp.869-878, 2011.
DOI : 10.1387/ijdb.113369vp

R. J. Kavlock, G. P. Daston, and C. Derosa, Research needs for the risk assessment of health and environmental effects of endocrine disruptors: a report of the U.S. EPA-sponsored workshop, Environmental Health Perspectives, vol.104, issue.Suppl 4, pp.715-740, 1996.
DOI : 10.1289/ehp.96104s4715

E. K. Shanle and W. Xu, Endocrine Disrupting Chemicals Targeting Estrogen Receptor Signaling: Identification and Mechanisms of Action, Chemical Research in Toxicology, vol.24, issue.1, pp.6-19, 2011.
DOI : 10.1021/tx100231n

T. Lóránd, E. Vigh, and J. Garai, Hormonal Action of Plant Derived and Anthropogenic Non-Steroidal Estrogenic Compounds: Phytoestrogens and Xenoestrogens, Current Medicinal Chemistry, vol.17, issue.30, pp.3542-3574, 2010.
DOI : 10.2174/092986710792927813

A. Kortenkamp and R. Altenburger, Synergisms with mixtures of xenoestrogens: A reevaluation using the method of isoboles, Science of The Total Environment, vol.221, issue.1, pp.59-73, 1998.
DOI : 10.1016/S0048-9697(98)00261-7

D. W. Singleton and S. A. Khan, Xenoestrogen exposure and mechanisms of endocrine disruption, Frontiers in Bioscience, vol.8, issue.6, pp.110-118, 2003.
DOI : 10.2741/1010

J. M. Harrington, G. F. Stein, R. O. Rivera, and A. V. De-morales, The Occupational Hazards of Formulating Oral Contraceptives ??? A Survey of Plant Employees, Archives of Environmental Health: An International Journal, vol.26, issue.1, pp.12-15, 1978.
DOI : 10.1080/00039896.1978.10667301

G. H. Degen and H. M. Bolt, Endocrine disruptors: update on xenoestrogens, International Archives of Occupational and Environmental Health, vol.73, issue.7, pp.433-441, 2000.
DOI : 10.1007/s004200000163

R. R. Newbold, Prenatal exposure to diethylstilbestrol (DES), Fertility and Sterility, vol.89, issue.2, pp.55-56, 2008.
DOI : 10.1016/j.fertnstert.2008.01.062

L. Multigner, J. R. Ndong, and A. Giusti, Chlordecone Exposure and Risk of Prostate Cancer, Journal of Clinical Oncology, vol.28, issue.21, pp.3457-3462, 2010.
DOI : 10.1200/JCO.2009.27.2153

URL : https://hal.archives-ouvertes.fr/inserm-00518099

Z. H. Liu, Y. Kanjo, and S. Mizutani, A review of phytoestrogens: Their occurrence and fate in the environment, Water Research, vol.44, issue.2, pp.567-577, 2010.
DOI : 10.1016/j.watres.2009.03.025

R. M. Evans, The steroid and thyroid hormone receptor superfamily, Science, vol.240, issue.4854, pp.889-895, 1988.
DOI : 10.1126/science.3283939

A. C. Pike, Lessons learnt from structural studies of the oestrogen receptor, Best Practice & Research Clinical Endocrinology & Metabolism, vol.20, issue.1, pp.1-14, 2006.
DOI : 10.1016/j.beem.2005.09.002

A. Boudot, G. Kerdivel, and D. Habauzit, Differential Estrogen-Regulation of CXCL12 Chemokine Receptors, CXCR4 and CXCR7, Contributes to the Growth Effect of Estrogens in Breast Cancer Cells, PLoS ONE, vol.132, issue.6, 2011.
DOI : 10.1371/journal.pone.0020898.s002

URL : https://hal.archives-ouvertes.fr/hal-00605459

L. Cicatiello, C. Scafoglio, and L. Altucci, A genomic view of estrogen actions in human breast cancer cells by expression profiling of the hormone-responsive transcriptome, Journal of Molecular Endocrinology, vol.32, issue.3, pp.719-775, 2004.
DOI : 10.1677/jme.0.0320719

G. Kerdivel, A. Boudot, and F. Pakdel, Estrogen represses CXCR7 gene expression by inhibiting the recruitment of NF??B transcription factor at the CXCR7 promoter in breast cancer cells, Biochemical and Biophysical Research Communications, vol.431, issue.4, pp.729-733, 2013.
DOI : 10.1016/j.bbrc.2013.01.050

S. Nilsson, S. Mäkelä, and E. Treuter, Mechanisms of estrogen action, Physiological Reviews, vol.81, issue.4, pp.1535-1565, 2001.

P. , L. Rosa, V. Pesiri, G. Leclercq, M. Marino et al., Palmitoylation regulates 17í µí»½-estradiol-induced estrogen receptor-í µí»¼ degradation and transcriptional activity, Molecular Endocrinology, vol.26, pp.762-774, 2012.

A. Pedram, M. Razandi, M. Aitkenhead, C. C. Hughes, and E. R. Levin, Integration of the Non-genomic and Genomic Actions of Estrogen. MEMBRANE-INITIATED SIGNALING BY STEROID TO TRANSCRIPTION AND CELL BIOLOGY, Journal of Biological Chemistry, vol.277, issue.52, pp.50768-50775, 2002.
DOI : 10.1074/jbc.M210106200

S. Safe and K. Kim, Non-classical genomic estrogen receptor (ER)/specificity protein and ER/activating protein-1 signaling pathways, Journal of Molecular Endocrinology, vol.41, issue.5, pp.5-6, 2008.
DOI : 10.1677/JME-08-0103

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2582054

L. Celik, J. D. Lund, and B. Schiøtt, Exploring Interactions of Endocrine-Disrupting Compounds with Different Conformations of the Human Estrogen Receptor ?? Ligand Binding Domain: A Molecular Docking Study, Chemical Research in Toxicology, vol.21, issue.11, pp.2195-2206, 2008.
DOI : 10.1021/tx800278d

G. Kerdivel, R. L. Guevel, D. Habauzit, F. Brion, S. Ait-aissa et al., Estrogenic Potency of Benzophenone UV Filters in Breast Cancer Cells: Proliferative and Transcriptional Activity Substantiated by Docking Analysis, PLoS ONE, vol.81, issue.1, 2013.
DOI : 10.1371/journal.pone.0060567.t002

URL : https://hal.archives-ouvertes.fr/inserm-00838323

J. R. Gosden, P. G. Middleton, and D. Rout, Localization of the human oestrogen receptor gene to chromosome 6q24→q27 by in situ hybridization, Cytogenetic and Genome Research, vol.43, issue.3-4, pp.3-4, 1986.
DOI : 10.1159/000132325

E. Enmark, M. Pelto-huikko, and K. Grandien, Human estrogen receptor í µí»½-gene structure, chromosomal localization, and expression pattern, Journal of Clinical Endocrinology and Metabolism, vol.82, issue.12, pp.4258-4265, 1997.

G. Flouriot, C. Griffin, M. Kenealy, V. Sonntag-buck, and F. Gannon, Differentially expressed messenger RNA isoforms of the human estrogen receptor-í µí»¼ gene are generated by alternative splicing and promoter usage, Molecular Endocrinology, vol.12, issue.12, pp.1939-1954, 1998.

Y. Okuda, S. Hirata, N. Watanabe, T. Shoda, J. Kato et al., Novel Splicing Events of Untranslated First Exons in Human Estrogen Receptor Alpha(ER.ALPHA.) Gene., Endocrine Journal, vol.50, issue.1, pp.97-104, 2003.
DOI : 10.1507/endocrj.50.97

G. Flouriot, H. Brand, and S. Denger, Identification of a new isoform of the human estrogen receptor-alpha (hER-alpha) that is encoded by distinct transcripts and that is able to repress hER-alpha activation function 1, The EMBO Journal, vol.19, issue.17, pp.4688-4700, 2000.
DOI : 10.1093/emboj/19.17.4688

G. Penot, C. Le-péron, and Y. Mérot, The Human Estrogen Receptor-?? Isoform hER??46 Antagonizes the Proliferative Influence of hER??66 in MCF7 Breast Cancer Cells, Endocrinology, vol.146, issue.12, pp.5474-5484, 2005.
DOI : 10.1210/en.2005-0866

Y. W. Zhao, X. Zhang, P. Shen, B. W. Loggie, Y. Chang et al., Identification, cloning, and expression of human estrogen receptor-í µí»¼36, a novel variant of human estrogen receptorí µí»¼66, Biochemical and Biophysical Research Communications, vol.336, issue.4, pp.1023-1027, 2005.

H. Shi, H. Shigeta, N. Yang, K. Fu, G. O-'brian et al., Human Estrogen Receptor-Like 1 (ESRL1) Gene: Genomic Organization, Chromosomal Localization, and Promoter Characterization, Genomics, vol.44, issue.1, pp.52-60, 1997.
DOI : 10.1006/geno.1997.4850

P. T. Saunders, Oestrogen receptor beta (ER beta), Reviews of Reproduction, vol.3, issue.3, pp.164-171, 1998.
DOI : 10.1530/ror.0.0030164

J. F. Couse and K. S. Korach, Estrogen Receptor Null Mice: What Have We Learned and Where Will They Lead Us?, Endocrine Reviews, vol.20, issue.3, pp.358-417, 1999.
DOI : 10.1210/edrv.20.3.0370

G. Delbès, C. Levacher, C. Duquenne, C. Racine, P. Pakarinen et al., Endogenous Estrogens Inhibit Mouse Fetal Leydig Cell Development via Estrogen Receptor ??, Endocrinology, vol.146, issue.5, pp.2454-2461, 2005.
DOI : 10.1210/en.2004-1540

M. E. Wilson, J. M. Westberry, and A. L. Trout, Estrogen receptor-alpha gene expression in the cortex: Sex differences during development and in adulthood, Hormones and Behavior, vol.59, issue.3, pp.353-357, 2011.
DOI : 10.1016/j.yhbeh.2010.08.004

X. Fan, H. J. Kim, M. Warner, and J. A. Gustafsson, Estrogen receptor beta is essential for sprouting of nociceptive primary afferents and for morphogenesis and maintenance of the dorsal horn interneurons, Proceedings of the National Academy of Sciences, vol.104, issue.34, pp.13696-13701, 2007.
DOI : 10.1073/pnas.0705936104

D. D. Heidrich, S. Steckelbroeck, and D. Klingmuller, Inhibition of human cytochrome P450 aromatase activity by butyltins, Steroids, vol.66, issue.10, pp.763-769, 2001.
DOI : 10.1016/S0039-128X(01)00108-8

M. H. Kester, S. Bulduk, and D. , Potent Inhibition of Estrogen Sulfotransferase by Hydroxylated PCB Metabolites: A Novel Pathway Explaining the Estrogenic Activity of PCBs, Endocrinology, vol.141, issue.5, pp.1897-1900, 2000.
DOI : 10.1210/endo.141.5.7530

H. Déchaud, C. Ravard, F. Claustrat, A. B. De-laperrì-ere, and M. Pugeat, Xenoestrogen interaction with human sex hormone-binding globulin (hSHBG)1, Steroids, vol.64, issue.5, pp.328-334, 1999.
DOI : 10.1016/S0039-128X(98)00114-7

E. J. Routledge, R. White, M. G. Parker, and J. P. Sumpter, Differential Effects of Xenoestrogens on Coactivator Recruitment by Estrogen Receptor (ER) ?? and ER??, Journal of Biological Chemistry, vol.275, issue.46, pp.35986-35993, 2000.
DOI : 10.1074/jbc.M006777200

S. Jagadeesh, S. Kyo, and P. P. Banerjee, Genistein Represses Telomerase Activity via Both Transcriptional and Posttranslational Mechanisms in Human Prostate Cancer Cells, Cancer Research, vol.66, issue.4, pp.2107-2115, 2006.
DOI : 10.1158/0008-5472.CAN-05-2494

P. H. Watson, R. T. Pon, and R. P. Shiu, Inhibition of cmyc expression by phosphorothioate antisense oligonucleotide identifies a critical role for c-myc in the growth of human breast cancer, Cancer Research, vol.51, issue.15, pp.3996-4000, 1991.

F. Martin, J. A. Reig, and B. Soria, Secretagogue-induced [Ca2+]i changes in single rat pancreatic islets and correlation with simultaneously measured insulin release, Journal of Molecular Endocrinology, vol.15, issue.2, pp.177-185, 1995.
DOI : 10.1677/jme.0.0150177

X. Li, S. Zhang, and S. Safe, Activation of kinase pathways in MCF-7 cells by 17??-estradiol and structurally diverse estrogenic compounds, The Journal of Steroid Biochemistry and Molecular Biology, vol.98, issue.2-3, pp.122-132, 2006.
DOI : 10.1016/j.jsbmb.2005.08.018

H. Cho and B. S. Katzenellenbogen, Synergistic activation of estrogen receptor-mediated transcription by estradiol and protein kinase activators., Molecular Endocrinology, vol.7, issue.3, pp.441-452, 1993.
DOI : 10.1210/mend.7.3.7683375

F. Ohtake, K. I. Takeyama, and T. Matsumoto, Modulation of oestrogen receptor signalling by association with the activated dioxin receptor, Nature, vol.423, issue.6939, pp.545-550, 2003.
DOI : 10.1038/nature01606

S. Safe and M. Wormke, Inhibitory Aryl Hydrocarbon Receptor???Estrogen Receptor ?? Cross-Talk and Mechanisms of Action, Chemical Research in Toxicology, vol.16, issue.7, pp.807-816, 2003.
DOI : 10.1021/tx034036r

S. Khan, R. Barhoumi, R. Burghardt, S. Liu, K. Kim et al., Molecular Mechanism of Inhibitory Aryl Hydrocarbon Receptor???Estrogen Receptor/Sp1 Cross Talk in Breast Cancer Cells, Molecular Endocrinology, vol.20, issue.9, pp.2199-2214, 2006.
DOI : 10.1210/me.2006-0100

J. Matthews, B. Wihlén, J. Thomsen, and J. A. Gustafsson, Aryl Hydrocarbon Receptor-Mediated Transcription: Ligand-Dependent Recruitment of Estrogen Receptor ?? to 2,3,7,8-Tetrachlorodibenzo- p-Dioxin-Responsive Promoters, Molecular and Cellular Biology, vol.25, issue.13, pp.5317-5328, 2005.
DOI : 10.1128/MCB.25.13.5317-5328.2005

P. Pocar, B. Fischer, T. Klonisch, and S. Hombach-klonisch, Molecular interactions of the aryl hydrocarbon receptor and its biological and toxicological relevance for reproduction, Reproduction, vol.129, issue.4, pp.379-389, 2005.
DOI : 10.1530/rep.1.00294

M. Wormke, M. Stoner, and B. Saville, The Aryl Hydrocarbon Receptor Mediates Degradation of Estrogen Receptor ?? through Activation of Proteasomes, Molecular and Cellular Biology, vol.23, issue.6, pp.1843-1855, 2003.
DOI : 10.1128/MCB.23.6.1843-1855.2003

R. , L. Guével, F. G. Petit, P. L. Goff, R. Métivier et al., Inhibition of rainbow trout (Oncorhynchus mykiss) estrogen receptor activity by cadmium, Biology of Reproduction, vol.63, issue.1, pp.259-266, 2000.

M. D. Johnson, N. Kenney, and A. Stoica, Cadmium mimics the in vivo effects of estrogen in the uterus and mammary gland, Nature Medicine, vol.9, issue.8, pp.1081-1084, 2003.
DOI : 10.1038/nm902

E. Silva, M. J. Lopez-espinosa, J. M. Molina-molina, M. Fernández, N. Olea et al., Lack of activity of cadmium in in vitro estrogenicity assays, Toxicology and Applied Pharmacology, vol.216, issue.1, pp.20-28, 2006.
DOI : 10.1016/j.taap.2006.04.002

M. Takiguchi and S. Yoshihara, New aspects of cadmium as endocrine disruptor, Environmental Sciences, vol.13, issue.2, pp.107-116, 2006.

A. Stoica, B. S. Katzenellenbogen, and M. B. Martin, Activation of estrogen receptor-í µí»¼ by the heavy metal cadmium, Molecular Endocrinology, vol.14, issue.4, pp.545-553, 2000.

C. Byrne, S. D. Divekar, G. B. Storchan, D. A. Parodi, and M. B. Martin, Metals and Breast Cancer, Journal of Mammary Gland Biology and Neoplasia, vol.144, issue.Suppl 1, pp.63-73, 2013.
DOI : 10.1007/s10911-013-9273-9

S. D. Divekar, G. B. Storchan, and K. Sperle, The Role of Calcium in the Activation of Estrogen Receptor-Alpha, Cancer Research, vol.71, issue.5, pp.1658-1668, 2011.
DOI : 10.1158/0008-5472.CAN-10-1899

L. J. Guillette-jr, T. S. Gross, G. R. Masson, J. M. Matter, H. F. Percival et al., Developmental abnormalities of the gonad and abnormal sex hormone concentrations in juvenile alligators from contaminated and control lakes in Florida, Environmental Health Perspectives, vol.102, issue.8, pp.680-688, 1994.
DOI : 10.1289/ehp.94102680

W. R. Kelce, C. R. Stone, S. C. Laws, L. E. Gray, J. A. Kemppainen et al., Persistent DDT metabolite p,p'???DDE is a potent androgen receptor antagonist, Nature, vol.375, issue.6532, pp.581-585, 1995.
DOI : 10.1038/375581a0

K. Yamasaki, H. Okuda, T. Takeuchi, and Y. Minobe, Effects of in utero through lactational exposure to dicyclohexyl phthalate and p,p???-DDE in Sprague???Dawley rats, Toxicology Letters, vol.189, issue.1, pp.14-20, 2009.
DOI : 10.1016/j.toxlet.2009.04.023

A. Thiel, S. Guth, S. Böhm, and G. Eisenbrand, Dicofol degradation to p,p???-dichlorobenzophenone ??? A potential antiandrogen, Toxicology, vol.282, issue.3, pp.88-93, 2011.
DOI : 10.1016/j.tox.2011.01.016

A. Adamsson, V. Salonen, J. Paranko, and J. Toppari, Effects of maternal exposure to di-isononylphthalate (DINP) and 1,1-dichloro-2,2-bis(p-chlorophenyl)ethylene (p,p???-DDE) on steroidogenesis in the fetal rat testis and adrenal gland, Reproductive Toxicology, vol.28, issue.1, pp.66-74, 2009.
DOI : 10.1016/j.reprotox.2009.03.002

C. G. Campbell, S. E. Borglin, F. B. Green, A. Grayson, E. Wozei et al., Biologically directed environmental monitoring, fate, and transport of estrogenic endocrine disrupting compounds in water: A review, Chemosphere, vol.65, issue.8, pp.1265-1280, 2006.
DOI : 10.1016/j.chemosphere.2006.08.003

S. Rodriguez-mozaz, M. J. De-alda, and D. Barceló, Picogram per Liter Level Determination of Estrogens in Natural Waters and Waterworks by a Fully Automated On-Line Solid-Phase Extraction-Liquid Chromatography-Electrospray Tandem Mass Spectrometry Method, Analytical Chemistry, vol.76, issue.23, pp.6998-7006, 2005.
DOI : 10.1021/ac049051v

A. Peñalver, E. Pocurull, F. Borrull, and R. M. Marcé, Method based on solid-phase microextraction???high-performance liquid chromatography with UV and electrochemical detection to determine estrogenic compounds in water samples, Journal of Chromatography A, vol.964, issue.1-2, pp.153-160, 2002.
DOI : 10.1016/S0021-9673(02)00694-5

J. Carpinteiro, J. B. Quintana, I. Rodríguez, A. M. Carro, R. A. Lorenzo et al., Applicability of solid-phase microextraction followed by on-fiber silylation for the determination of estrogens in water samples by gas chromatography???tandem mass spectrometry, Journal of Chromatography A, vol.1056, issue.1-2, pp.179-185, 2004.
DOI : 10.1016/S0021-9673(04)01082-9

F. Bianchi, M. Mattarozzi, and M. Careri, An SPME???GC???MS method using an octadecyl silica fibre for the determination of the potential angiogenesis modulators 17??-estradiol and 2-methoxyestradiol in culture media, Analytical and Bioanalytical Chemistry, vol.283, issue.1197, pp.2639-2645, 2010.
DOI : 10.1007/s00216-010-3508-z

J. M. Nogueira, Novel sorption-based methodologies for static microextraction analysis: A review on SBSE and related techniques, Analytica Chimica Acta, vol.757, pp.1-10, 2012.
DOI : 10.1016/j.aca.2012.10.033

H. G. Mol, S. Sunarto, and O. M. Steijger, Determination of endocrine disruptors in water after derivatization with N-methyl-N-(tert.-butyldimethyltrifluoroacetamide) using gas chromatography with mass spectrometric detection, Journal of Chromatography A, vol.879, issue.1, pp.97-112, 2000.
DOI : 10.1016/S0021-9673(00)00124-2

M. Farré, L. Kantiani, M. Petrovic, S. Pérez, and D. Barceló, Achievements and future trends in the analysis of emerging organic contaminants in environmental samples by mass spectrometry and bioanalytical techniques, Journal of Chromatography A, vol.1259, pp.86-99, 2012.
DOI : 10.1016/j.chroma.2012.07.024

S. Biau, S. Bayle, P. D. Barbara, and B. Roig, The chick embryo: an animal model for detection of the effects of hormonal compounds, Analytical and Bioanalytical Chemistry, vol.107, issue.3, pp.1397-1403, 2007.
DOI : 10.1007/s00216-006-0870-y

W. D. Isenhower-jr, R. R. Newbold, R. C. Cefalo, K. S. Korach, and J. A. Mclachlan, Absence of estrogenic activity in some drugs commonly used during pregnancy, Biological Research in Pregnancy and Perinatology, vol.7, issue.1, pp.6-10, 1986.

E. Padilla-banks, W. N. Jefferson, and R. R. Newbold, The Immature Mouse Is a Suitable Model for Detection of Estrogenicity in the Uterotropic Bioassay, Environmental Health Perspectives, vol.109, issue.8, pp.821-826, 2001.
DOI : 10.1289/ehp.01109821

J. Kanno, L. Onyon, J. Haseman, P. Fenner-crisp, J. Ashby et al., The OECD Program to Validate the Rat Uterotrophic Bioassay to Screen Compounds for in Vivo Estrogenic Responses: Phase 1, Environmental Health Perspectives, vol.109, issue.8, pp.785-794, 2001.
DOI : 10.1289/ehp.01109785

G. Flouriot, F. Pakdel, B. Ducouret, and Y. Valotaire, Influence of xenobiotics on rainbow trout liver estrogen receptor and vitellogenin gene expression, Journal of Molecular Endocrinology, vol.15, issue.2, pp.143-151, 1995.
DOI : 10.1677/jme.0.0150143

C. R. Tyler, J. P. Sumpter, and P. R. Witthames, The Dynamics of Oocyte Growth during Vitellogenesis in the Rainbow Trout (Oncorhynchus Mykiss), Biology of Reproduction, vol.43, issue.2, pp.202-209, 1990.
DOI : 10.1095/biolreprod43.2.202

F. R. Knudsen, A. E. Schou, and M. L. Wiborg, Increase of Plasma Vitellogenin Concentration in Rainbow Trout ( Oncorhynchus mykiss ) Exposed to Effluents from Oil Refinery Treatment Works and Municipal Sewage, Bulletin of Environmental Contamination and Toxicology, vol.59, issue.5, pp.802-806, 1997.
DOI : 10.1007/s001289900552

F. Brion, Y. Le-page, and B. Piccini, Screening Estrogenic Activities of Chemicals or Mixtures In Vivo Using Transgenic (cyp19a1b-GFP) Zebrafish Embryos, PLoS ONE, vol.21, issue.5, 2012.
DOI : 10.1371/journal.pone.0036069.t002

URL : https://hal.archives-ouvertes.fr/hal-00877371

P. Ciana, G. Di-luccio, and S. Belcredito, Profiling of Estrogen Receptor Activity, Molecular Endocrinology, vol.15, issue.7, pp.1104-1113, 2001.
DOI : 10.1210/mend.15.7.0658

O. Lee, A. Takesono, M. Tada, C. R. Tyler, and T. Kudoh, Biosensor Zebrafish Provide New Insights into Potential Health Effects of Environmental Estrogens, Environmental Health Perspectives, vol.120, issue.7, pp.990-996, 2012.
DOI : 10.1289/ehp.1104433

H. Chen, J. Hu, and J. Yang, Generation of a fluorescent transgenic zebrafish for detection of environmental estrogens, Aquatic Toxicology, vol.96, issue.1, pp.53-61, 2010.
DOI : 10.1016/j.aquatox.2009.09.015

A. M. Soto, C. Sonnenschein, K. L. Chung, M. F. Fernandez, N. Olea et al., The E-SCREEN assay as a tool to identify estrogens: an update on estrogenic environmental pollutants, Environmental Health Perspectives, vol.103, issue.Suppl 7, pp.113-122, 1995.
DOI : 10.1289/ehp.95103s7113

P. Balaguer, F. François, and F. Comunale, Reporter cell lines to study the estrogenic effects of xenoestrogens, Science of The Total Environment, vol.233, issue.1-3, pp.1-3, 1999.
DOI : 10.1016/S0048-9697(99)00178-3

D. Habauzit, A. Boudot, G. Kerdivel, G. Flouriot, and F. Pakdel, Development and validation of a test for environmental estrogens: Checking xeno-estrogen activity by CXCL12 secretion in BREAST CANCER CELL LINES (CXCL-test), Environmental Toxicology, vol.31, issue.Suppl 7, pp.495-503, 2010.
DOI : 10.1002/tox.20594

URL : https://hal.archives-ouvertes.fr/hal-00592335

J. C. Swart, E. J. Pool, and J. H. Van-wyk, The implementation of a battery of in vivo and in vitro bioassays to assess river water for estrogenic endocrine disrupting chemicals, Ecotoxicology and Environmental Safety, vol.74, issue.1, pp.138-143, 2011.
DOI : 10.1016/j.ecoenv.2010.09.006

J. Legler, C. E. Van-den-brink, and A. Brouwer, Development of a stably transfected estrogen receptor-mediated luciferase reporter gene assay in the human T47D breast cancer cell line, Toxicological Sciences, vol.48, issue.1, pp.55-66, 1999.
DOI : 10.1093/toxsci/48.1.55

S. F. Arnold, M. K. Robinson, A. C. Notides, L. J. Guillette, and J. A. Mclachan, A yeast estrogen screen for examining the relative exposure of cells to natural and xenoestrogens, Environmental Health Perspectives, vol.104, issue.5, pp.544-548, 1996.
DOI : 10.1289/ehp.96104544

F. Petit, P. L. Goff, J. P. Cravédi, Y. Valotaire, and F. Pakdel, Two complementary bioassays for screening the estrogenic potency of xenobiotics: recombinant yeast for trout estrogen receptor and trout hepatocyte cultures, Journal of Molecular Endocrinology, vol.19, issue.3, pp.321-335, 1997.
DOI : 10.1677/jme.0.0190321

F. Petit, Y. Valotaire, and F. Pakdel, Differential Functional Activities of Rainbow Trout and Human Estrogen Receptors Expressed in the Yeast Saccharomyces cerevisiae, European Journal of Biochemistry, vol.268, issue.2, pp.584-592, 1995.
DOI : 10.1146/annurev.genet.19.1.209

Y. , L. Page, M. Scholze, O. Kah, and F. Pakdel, Assessment of xenoestrogens using three distinct estrogen receptors and the zebrafish brain aromatase gene in a highly responsive glial cell system, Environmental Health Perspectives, vol.114, issue.5, pp.752-758, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00068400

H. R. Andersen, A. M. Andersson, and S. F. Arnold, Comparison of Short-Term Estrogenicity Tests for Identification of Hormone-Disrupting Chemicals, Environmental Health Perspectives, vol.107, issue.Suppl 1, pp.89-108, 1999.
DOI : 10.1289/ehp.99107s189

N. Fujimoto, H. Honda, and S. Kitamura, Effects of environmental estrogenic chemicals on AP1 mediated transcription with estrogen receptors ?? and ??, The Journal of Steroid Biochemistry and Molecular Biology, vol.88, issue.1, pp.53-59, 2004.
DOI : 10.1016/j.jsbmb.2003.10.006

D. Habauzit, G. Flouriot, F. Pakdel, and C. Saligaut, Effects of Estrogens and Endocrine-Disrupting Chemicals on Cell Differentiation???Survival???Proliferation in Brain: Contributions of Neuronal Cell Lines, Journal of Toxicology and Environmental Health, Part B, vol.117, issue.5-7, pp.5-7, 2011.
DOI : 10.1016/j.brainres.2007.06.092

URL : https://hal.archives-ouvertes.fr/hal-00750272

M. D. Shelby, R. R. Newbold, D. B. Tully, K. Chae, and V. L. Davis, Assessing environmental chemicals for estrogenicity using a combination of in vitro and in vivo assays, Environmental Health Perspectives, vol.104, issue.12, pp.1296-1300, 1996.
DOI : 10.1289/ehp.961041296

L. C. , C. Jr, and C. Lyons, Electrode systems for continuous monitoring in cardiovascular surgery, Annals of the New York Academy of Sciences, vol.102, pp.29-45, 1962.

S. Rodriguez-mozaz, M. P. Marco, M. J. De-alda, and D. Barceló, Biosensors for environmental monitoring of endocrine disruptors: a review article, Analytical and Bioanalytical Chemistry, vol.378, issue.3, pp.588-598, 2004.

P. D. Ursi, E. Salvi, P. Fossa, L. Milanesi, and E. Rovida, Modelling the interaction of steroid receptors with endocrine disrupting chemicals, BMC Bioinformatics, vol.6, 2005.

H. Kanso, L. Barthelmebs, N. Inguimbert, and T. Noguer, Immunosensors for Estradiol and Ethinylestradiol Based on New Synthetic Estrogen Derivatives: Application to Wastewater Analysis, Analytical Chemistry, vol.85, issue.4, pp.2397-2404, 2013.
DOI : 10.1021/ac303406c

M. Usami, K. Mitsunaga, and Y. Ohno, Estrogen receptor binding assay of chemicals with a surface plasmon resonance biosensor, The Journal of Steroid Biochemistry and Molecular Biology, vol.81, issue.1, pp.47-55, 2002.
DOI : 10.1016/S0960-0760(02)00046-8

M. Seifert, S. Haindl, and B. Hock, Development of an enzyme linked receptor assay (ELRA) for estrogens and xenoestrogens, Analytica Chimica Acta, vol.386, issue.3, pp.191-199, 1999.
DOI : 10.1016/S0003-2670(99)00044-6

R. L. Rich, L. R. Hoth, and K. F. Geoghegan, Kinetic analysis of estrogen receptor/ligand interactions, Proceedings of the National Academy of Sciences, vol.99, issue.13, pp.8562-8567, 2002.
DOI : 10.1073/pnas.142288199

P. Fechner, F. Pröll, M. Carlquist, and G. Proll, An advanced biosensor for the prediction of estrogenic effects of endocrinedisrupting chemicals on the estrogen receptor alpha, Analytical and Bioanalytical Chemistry, vol.393, pp.6-7, 2009.

R. Paulmurugan and S. S. Gambhir, An intramolecular folding sensor for imaging estrogen receptor-ligand interactions, Proceedings of the National Academy of Sciences, vol.103, issue.43, pp.15883-15888, 2006.
DOI : 10.1073/pnas.0607385103

S. B. Kim, M. Sato, and H. Tao, Molecular Tension-Indexed Bioluminescent Probe for Determining Protein???Protein Interactions, Bioconjugate Chemistry, vol.20, issue.12, pp.2324-2330, 2009.
DOI : 10.1021/bc900330w

B. K. Sung, Y. Umezawa, K. A. Kanno, and H. Tao, An integrated-molecule-format multicolor probe for monitoring multiple activities of a bioactive small molecule, ACS Chemical Biology, vol.3, issue.6, pp.359-372, 2008.

E. Jisa, E. Dornstauder, S. Ogawa, S. Inoue, M. Muramatsu et al., Transcriptional activities of estrogen receptor alpha and beta in yeast properties of raloxifene11Abbreviations: ERE, estrogen response element; E2, 17??-estradiol; RAL, raloxifene; ER??, estrogen receptor ??; ER??, estrogen receptor ??; and SERM, selective estrogen receptor modulator., Biochemical Pharmacology, vol.62, issue.7, pp.953-961, 2001.
DOI : 10.1016/S0006-2952(01)00731-6

A. Berthier, C. Elie-caille, E. Lesniewska, R. Delage-mourroux, and W. Boireau, Label-free sensing and atomic force spectroscopy for the characterization of protein-DNA and protein-protein interactions: application to estrogen receptors, Journal of Molecular Recognition, vol.33, issue.15, pp.429-435, 2011.
DOI : 10.1002/jmr.1106

B. J. Cheskis, S. Karathanasis, and C. R. Lyttle, Estrogen receptor ligands modulate its interaction with DNA, The Journal of Biological Chemistry, vol.272, issue.17, pp.11384-11391, 1997.

D. Habauzit, J. Armengaud, B. Roig, and J. Chopineau, Determination of estrogen presence in water by SPR using estrogen receptor dimerization, Analytical and Bioanalytical Chemistry, vol.1160, issue.Suppl 7, pp.873-883, 2008.
DOI : 10.1007/s00216-007-1725-x

]. D. Habauzit, J. Chopineau, and B. Roig, SPR-based biosensors: a tool for biodetection of hormonal compounds, Analytical and Bioanalytical Chemistry, vol.205, issue.7, pp.1215-1223, 2007.
DOI : 10.1007/s00216-006-0958-4

W. Y. Peh, E. Reimhult, F. T. Huey, J. S. Thomsen, and X. Su, Understanding Ligand Binding Effects on the Conformation of Estrogen Receptor ??-DNA Complexes: A Combinational Quartz Crystal Microbalance with Dissipation and Surface Plasmon Resonance Study, Biophysical Journal, vol.92, issue.12, pp.4415-4423, 2007.
DOI : 10.1529/biophysj.106.099382

A. Bouter, N. Buisine, and A. L. Grand, Control of vitellogenin genes expression by sequences derived from transposable elements in rainbow trout, Biochimica et Biophysica Acta (BBA) - Gene Regulatory Mechanisms, vol.1799, issue.8, pp.546-554, 2010.
DOI : 10.1016/j.bbagrm.2010.07.003

URL : https://hal.archives-ouvertes.fr/inserm-00511709

M. Szatkowski-ozers, J. J. Hill, K. Ervin, C. A. Royer, and J. Gorski, The dissociation rate of estrogen receptor ?? from the consensus estrogen response element1These studies were supported, in part, by the College of Agricultural and Life Sciences, University of Wisconsin and by National Institutes of Health Grants HD07259 and HD08192 to J.G.1, Molecular and Cellular Endocrinology, vol.175, issue.1-2, pp.101-109, 2001.
DOI : 10.1016/S0303-7207(01)00395-1

M. S. Ozers, J. J. Hill, and K. Ervin, Equilibrium Binding of Estrogen Receptor with DNA Using Fluorescence Anisotropy, Journal of Biological Chemistry, vol.272, issue.48, pp.30405-30411, 1997.
DOI : 10.1074/jbc.272.48.30405

A. Tamrazi, K. E. Carlson, J. R. Daniels, K. M. Hurth, and J. A. Katzenellenbogen, Estrogen Receptor Dimerization: Ligand Binding Regulates Dimer Affinity and DimerDissociation Rate, Molecular Endocrinology, vol.16, issue.12, pp.2706-2719, 2002.
DOI : 10.1210/me.2002-0250

E. Swedenborg and I. Pongratz, AhR and ARNT modulate ER signaling, Toxicology, vol.268, issue.3, pp.132-138, 2010.
DOI : 10.1016/j.tox.2009.09.007

R. R. Newbold, R. B. Hanson, W. N. Jefferson, B. C. Bullock, J. Haseman et al., Increased tumors but uncompromised fertility in the female descendants of mice exposed developmentally to diethylstilbestrol, Carcinogenesis, vol.19, issue.9, pp.1655-1663, 1998.
DOI : 10.1093/carcin/19.9.1655

R. L. Jirtle and M. K. Skinner, Environmental epigenomics and disease susceptibility, Nature Reviews Genetics, vol.403, issue.4, pp.253-262, 2007.
DOI : 10.1038/nrg2045

M. D. Anway, A. S. Cupp, N. Uzumcu, and M. K. Skinner, Epigenetic Transgenerational Actions of Endocrine Disruptors and Male Fertility, Science, vol.308, issue.5727, pp.1466-1469, 2005.
DOI : 10.1126/science.1108190

M. D. Anway, C. Leathers, and M. K. Skinner, Endocrine Disruptor Vinclozolin Induced Epigenetic Transgenerational Adult-Onset Disease, Endocrinology, vol.147, issue.12, pp.5515-5523, 2006.
DOI : 10.1210/en.2006-0640

H. S. Chang, M. D. Anway, S. S. Rekow, and M. K. Skinner, Transgenerational Epigenetic Imprinting of the Male Germline by Endocrine Disruptor Exposure during Gonadal Sex Determination, Endocrinology, vol.147, issue.12, pp.5524-5541, 2006.
DOI : 10.1210/en.2006-0987

R. Kavlock and A. Cummings, Mode of Action: Inhibition of Androgen Receptor Function???Vinclozolin-Induced Malformations in Reproductive Development, Critical Reviews in Toxicology, vol.52, issue.1, pp.721-726, 2005.
DOI : 10.1074/jbc.270.34.19998

M. Manikkam, C. Guerrero-bosagna, R. Tracey, M. M. Haque, and M. K. Skinner, Transgenerational Actions of Environmental Compounds on Reproductive Disease and Identification of Epigenetic Biomarkers of Ancestral Exposures, PLoS ONE, vol.23, issue.2, 2012.
DOI : 10.1371/journal.pone.0031901.s009

J. M. Molina-molina, A. Hillenweck, and I. Jouanin, Steroid receptor profiling of vinclozolin and its primary metabolites, Toxicology and Applied Pharmacology, vol.216, issue.1, pp.44-54, 2006.
DOI : 10.1016/j.taap.2006.04.005

E. Nilsson, G. Larsen, M. Manikkam, C. Guerrero-bosagna, M. I. Savenkova et al., Environmentally induced International Journal of Endocrinology epigenetic transgenerational inheritance of ovarian disease, PLoS ONE, vol.7, issue.10, 2012.

C. M. Revankar, D. F. Cimino, L. A. Sklar, J. B. Arterburn, and E. R. Prossnitz, A Transmembrane Intracellular Estrogen Receptor Mediates Rapid Cell Signaling, Science, vol.307, issue.5715, pp.1625-1630, 2005.
DOI : 10.1126/science.1106943

E. J. Filardo, J. A. Quinn, A. R. Frackelton, and K. I. Bland, Estrogen Action Via the G Protein-Coupled Receptor, GPR30: Stimulation of Adenylyl Cyclase and cAMP-Mediated Attenuation of the Epidermal Growth Factor Receptor-to-MAPK Signaling Axis, Molecular Endocrinology, vol.16, issue.1, pp.70-84, 2002.
DOI : 10.1210/mend.16.1.0758

M. Maggiolini, A. Vivacqua, and G. Fasanella, The G Protein-coupled Receptor GPR30 Mediates c-fos Up-regulation by 17??-Estradiol and Phytoestrogens in Breast Cancer Cells, Journal of Biological Chemistry, vol.279, issue.26, pp.27008-27016, 2004.
DOI : 10.1074/jbc.M403588200

A. Vivacqua, D. Bonofiglio, and L. Albanito, 17beta-Estradiol, Genistein, and 4-Hydroxytamoxifen Induce the Proliferation of Thyroid Cancer Cells through the G Protein-Coupled Receptor GPR30, Molecular Pharmacology, vol.70, issue.4, pp.1414-1423, 2006.
DOI : 10.1124/mol.106.026344

P. Thomas and J. Dong, Binding and activation of the seventransmembrane estrogen receptor GPR30 by environmental estrogens: a potential novel mechanism of endocrine disruption, Journal of Steroid Biochemistry and Molecular Biology, vol.102, pp.1-5, 2006.

C. S. Watson, R. A. Alyea, Y. J. Jeng, and M. Y. Kochukov, Nongenomic actions of low concentration estrogens and xenoestrogens on multiple tissues, Molecular and Cellular Endocrinology, vol.274, issue.1-2, pp.1-7, 2007.
DOI : 10.1016/j.mce.2007.05.011

J. Tiefenbach, P. R. Moll, and M. R. Nelson, A live zebrafishbased screening system for human nuclear receptor ligand and cofactor discovery Article ID e9797, 2010. Submit your manuscripts at http://www.hindawi, PloS ONE, vol.5, issue.3, 2013.