H. Leridon and R. Slama, The impact of a decline in fecundity and of pregnancy postponement on final number of children and demand for assisted reproduction technology, Hum Reprod, vol.23, pp.1312-1319, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00270754

J. Toppari, J. C. Larsen, P. Christiansen, A. Giwercman, and P. Grandjean, Male reproductive health and environmental xenoestrogens, Environ Health Perspect, vol.104, pp.741-803, 1996.

R. M. Sharpe and N. E. Skakkebaek, Testicular dysgenesis syndrome: mechanistic insights and potential new downstream effects, Fertil Steril, vol.89, pp.33-38, 2008.

N. E. Skakkebaek, E. Rajpert-de-meyts, and K. M. Main, Testicular dysgenesis syndrome: an increasingly common developmental disorder with environmental aspects, Hum Reprod, vol.16, pp.972-978, 2001.

L. Gaspari, F. Paris, C. Jandel, N. Kalfa, and M. Orsini, Prenatal environmental risk factors for genital malformations in a population of 1442 French male newborns: a nested case-control study, Hum Reprod, vol.26, pp.3155-3162, 2011.

R. Habert, V. Muczynski, A. Lehraiki, R. Lambrot, and C. Lecureuil, Adverse effects of endocrine disruptors on the foetal testis development: focus on the phthalates, Folia Histochem Cytobiol, vol.47, pp.67-74, 2009.

H. M. Scott, J. I. Mason, and R. M. Sharpe, Steroidogenesis in the fetal testis and its susceptibility to disruption by exogenous compounds, Endocr Rev, vol.30, pp.883-925, 2009.

J. Toppari, H. E. Virtanen, K. M. Main, and N. E. Skakkebaek, Cryptorchidism and hypospadias as a sign of testicular dysgenesis syndrome (TDS): environmental connection, Birth Defects Res A Clin Mol Teratol, vol.88, pp.910-919, 2010.

K. Bay, K. M. Main, J. Toppari, N. E. Skakkebaek, and M. , Testicular descent: INSL3, testosterone, genes and the intrauterine milieu, Nat Rev Urol, vol.8, pp.187-196, 2011.

R. Ivell and R. Anand-ivell, Biology of insulin-like factor 3 in human reproduction, Hum Reprod Update, vol.15, pp.463-476, 2009.

S. Van-den-driesche, H. M. Scott, D. J. Macleod, M. Fisken, and M. Walker, Relative importance of prenatal and postnatal androgen action in determining growth of the penis and anogenital distance in the rat before, during and after puberty, Int J Androl, vol.34, pp.578-586, 2011.

M. Welsh, P. T. Saunders, M. Fisken, H. M. Scott, and G. R. Hutchison, Identification in rats of a programming window for reproductive tract masculinization, disruption of which leads to hypospadias and cryptorchidism, J Clin Invest, vol.118, pp.1479-1490, 2008.

K. M. Main, N. E. Skakkebaek, H. E. Virtanen, and J. Toppari, Genital anomalies in boys and the environment, Best Pract Res Clin Endocrinol Metab, vol.24, pp.279-289, 2010.

A. M. Soto and C. Sonnenschein, Environmental causes of cancer: endocrine disruptors as carcinogens, Nat Rev Endocrinol, vol.6, pp.363-370, 2010.

C. E. Talsness, A. J. Andrade, S. N. Kuriyama, J. A. Taylor, and F. S. Vom-saal, Components of plastic: experimental studies in animals and relevance for human health, Philos Trans R Soc Lond B Biol Sci, vol.364, pp.2079-2096, 2009.

R. W. Tyl, Basic exploratory research versus guideline-compliant studies used for hazard evaluation and risk assessment: bisphenol A as a case study, Environ Health Perspect, vol.117, pp.1644-1651, 2009.

L. N. Vandenberg, M. V. Maffini, C. Sonnenschein, B. S. Rubin, and A. M. Soto, Bisphenol-A and the great divide: a review of controversies in the field of endocrine disruption, Endocr Rev, vol.30, pp.75-95, 2009.

L. N. Vandenberg, R. Hauser, M. Marcus, N. Olea, and W. V. Welshons, Human exposure to bisphenol A (BPA), Reprod Toxicol, vol.24, pp.139-177, 2007.

M. Tanaka, S. Nakaya, M. Katayama, H. Leffers, and S. Nozawa, Effect of prenatal exposure to bisphenol A on the serum testosterone concentration of rats at birth, Hum Exp Toxicol, vol.25, pp.369-373, 2006.

R. Thuillier, G. Manku, Y. Wang, and M. Culty, Changes in MAPK pathway in neonatal and adult testis following fetal estrogen exposure and effects on rat testicular cells, Microsc Res Tech, vol.72, pp.773-786, 2009.

T. J. Murray, M. V. Maffini, A. A. Ucci, C. Sonnenschein, and A. M. Soto, Induction of mammary gland ductal hyperplasias and carcinoma in situ following fetal bisphenol A exposure, Reprod Toxicol, vol.23, pp.383-390, 2007.

K. L. Howdeshell, J. Furr, C. R. Lambright, V. S. Wilson, and B. C. Ryan, Gestational and lactational exposure to ethinyl estradiol, but not bisphenol A, decreases androgen-dependent reproductive organ weights and epididymal sperm abundance in the male long evans hooded rat, Toxicol Sci, vol.102, pp.371-382, 2008.

K. Kobayashi, M. Miyagawa, R. S. Wang, S. Sekiguchi, and M. Suda, Effects of in utero and lactational exposure to bisphenol A on somatic growth and anogenital distance in F1 rat offspring, Ind Health, vol.40, pp.375-381, 2002.

R. W. Tyl, C. B. Myers, M. C. Marr, B. F. Thomas, and A. R. Keimowitz, Threegeneration reproductive toxicity study of dietary bisphenol A in CD Sprague-Dawley rats, Toxicol Sci, vol.68, pp.121-146, 2002.

M. Miao, W. Yuan, Y. He, Z. Zhou, and J. Wang, In utero exposure to bisphenol-A and anogenital distance of male offspring, Birth Defects Res A Clin Mol Teratol, vol.91, pp.867-872, 2011.

P. Fenichel, H. Dechaux, C. Harthe, J. Gal, and P. Ferrari, Unconjugated bisphenol A cord blood levels in boys with descended or undescended testes, Hum Reprod, vol.27, pp.983-990, 2012.

R. Habert, I. Devif, M. N. Gangnerau, and L. Lecerf, Ontogenesis of the in vitro response of rat testis to gonadotropin-releasing hormone, Mol Cell Endocrinol, vol.82, pp.199-206, 1991.

L. Lecerf, V. Rouiller-fabre, C. Levacher, C. Gautier, and J. M. Saez, Stimulatory effect of follicle-stimulating hormone on basal and luteinizing hormone-stimulated testosterone secretions by the fetal rat testis in vitro, Endocrinology, vol.133, pp.2313-2318, 1993.

G. Livera, V. Rouiller-fabre, P. Durand, and R. Habert, Multiple effects of retinoids on the development of Sertoli, germ, and Leydig cells of fetal and neonatal rat testis in culture, Biol Reprod, vol.62, pp.1303-1314, 2000.
URL : https://hal.archives-ouvertes.fr/hal-02689320

R. Olaso, C. Pairault, B. Boulogne, P. Durand, and R. Habert, Transforming growth factor beta1 and beta2 reduce the number of gonocytes by increasing apoptosis, Endocrinology, vol.139, pp.733-740, 1998.
URL : https://hal.archives-ouvertes.fr/hal-02685794

G. Livera, G. Delbes, C. Pairault, V. Rouiller-fabre, and R. Habert, Organotypic culture, a powerful model for studying rat and mouse fetal testis development, Cell Tissue Res, vol.324, pp.507-521, 2006.

R. Lambrot, H. Coffigny, C. Pairault, A. C. Donnadieu, and R. Frydman, Use of organ culture to study the human fetal testis development: effect of retinoic acid, J Clin Endocrinol Metab, vol.91, pp.2696-2703, 2006.

R. Lambrot, G. Livera, H. Coffigny, C. Pairault, and R. Frydman, A new method for toxicity assays on human and mouse fetal testis, Biochimie, vol.88, pp.1831-1835, 2006.

G. Delbes, C. Duquenne, J. Szenker, J. Taccoen, and R. Habert, Developmental changes in testicular sensitivity to estrogens throughout fetal and neonatal life, Toxicol Sci, vol.99, pp.234-243, 2007.

J. Lassurguere, G. Livera, R. Habert, and B. Jegou, Time-and dose-related effects of estradiol and diethylstilbestrol on the morphology and function of the fetal rat testis in culture, Toxicol Sci, vol.73, pp.160-169, 2003.

F. Chauvigne, A. Menuet, L. Lesne, M. C. Chagnon, and C. Chevrier, Time-and dose-related effects of di-(2-ethylhexyl) phthalate and its main metabolites on the function of the rat fetal testis in vitro, Environ Health Perspect, vol.117, pp.515-521, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00669512

R. Lambrot, V. Muczynski, C. Lecureuil, G. Angenard, and H. Coffigny, Phthalates impair germ cell development in the human fetal testis in vitro without change in testosterone production, Environ Health Perspect, vol.117, pp.32-37, 2009.

A. Lehraiki, C. Racine, A. Krust, R. Habert, and C. Levacher, Phthalates impair germ cell number in the mouse fetal testis by an androgen-and estrogenindependent mechanism, Toxicol Sci, vol.111, pp.372-382, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-00438655

V. Muczynski, J. P. Cravedi, A. Lehraiki, C. Levacher, and D. Moison, Effect of mono-(2-ethylhexyl) phthalate on human and mouse fetal testis: In vitro and in vivo approaches, Toxicol Appl Pharmacol, vol.261, pp.97-104, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00776675

G. Angenard, V. Muczynski, H. Coffigny, C. Pairault, and C. Duquenne, Cadmium increases human fetal germ cell apoptosis, Environ Health Perspect, vol.118, pp.331-337, 2010.

D. M. Kristensen, U. Hass, L. Lesne, G. Lottrup, and P. R. Jacobsen, Intrauterine exposure to mild analgesics is a risk factor for development of male reproductive disorders in human and rat, Hum Reprod, vol.26, pp.235-244, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00682685

A. Lehraiki, C. Chamaillard, A. Krust, R. Habert, and C. Levacher, Genistein impairs early testosterone production in fetal mouse testis via estrogen receptor alpha, Toxicol In Vitro, vol.25, pp.1542-1547, 2011.

S. Dupont, A. Krust, A. Gansmuller, A. Dierich, and P. Chambon, Effect of single and compound knockouts of estrogen receptors alpha (ERalpha) and beta (ERbeta) on mouse reproductive phenotypes, Development, vol.127, pp.4277-4291, 2000.

L. Evtouchenko, L. Studer, C. Spenger, E. Dreher, and R. W. Seiler, A mathematical model for the estimation of human embryonic and fetal age, Cell Transplant, vol.5, pp.453-464, 1996.

X. Zhang, H. Chang, S. Wiseman, Y. He, and E. Higley, Bisphenol A disrupts steroidogenesis in human H295R cells, Toxicol Sci, vol.121, pp.320-327, 2011.

M. Z. Lacroix, S. Puel, S. H. Collet, T. Corbel, and N. Picard-hagen, Simultaneous quantification of bisphenol A and its glucuronide metabolite (BPA-G) in plasma and urine: applicability to toxicokinetic investigations, Talanta, vol.85, pp.2053-2059, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02649171

R. Habert and R. Picon, Testosterone, dihydrotestosterone and estradiol-17 beta levels in maternal and fetal plasma and in fetal testes in the rat, J Steroid Biochem, vol.21, pp.193-198, 1984.

G. Delbes, C. Levacher, C. Duquenne, C. Racine, and P. Pakarinen, Endogenous estrogens inhibit mouse fetal Leydig cell development via estrogen receptor alpha, Endocrinology, vol.146, pp.2454-2461, 2005.

M. Z. Lacroix, S. Puel, S. H. Collet, T. Corbel, and N. Picard-hagen, Simultaneous quantification of bisphenol A and its glucuronide metabolite (BPA-G) in plasma and urine: applicability to toxicokinetic investigations, Talanta, vol.85, pp.2053-2059, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02649171

L. N. Vandenberg, I. Chahoud, J. J. Heindel, V. Padmanabhan, and F. J. Paumgartten, Urinary, circulating, and tissue biomonitoring studies indicate widespread exposure to bisphenol A, Environ Health Perspect, vol.118, pp.1055-1070, 2010.

J. G. Teeguarden, A. M. Calafat, X. Ye, D. R. Doerge, and M. I. Churchwell, Twenty-four hour human urine and serum profiles of bisphenol a during highdietary exposure, Toxicol Sci, vol.123, pp.48-57, 2011.

Y. Ikezuki, O. Tsutsumi, Y. Takai, Y. Kamei, and Y. Taketani, Determination of bisphenol A concentrations in human biological fluids reveals significant early prenatal exposure, Hum Reprod, vol.17, pp.2839-2841, 2002.

F. S. Vom-saal, G. S. Prins, and W. V. Welshons, Report of Very Low Real World Exposure to Bisphenol A is Unwarranted Based on a Lack of Data and Flawed Assumptions, Toxicol Sci, 2011.

N. J. Cabaton, P. R. Wadia, B. S. Rubin, D. Zalko, and C. M. Schaeberle, Perinatal exposure to environmentally relevant levels of bisphenol A decreases fertility and fecundity in CD-1 mice, Environ Health Perspect, vol.119, pp.547-552, 2010.
URL : https://hal.archives-ouvertes.fr/hal-02649514

T. Hanaoka, N. Kawamura, K. Hara, and S. Tsugane, Urinary bisphenol A and plasma hormone concentrations in male workers exposed to bisphenol A diglycidyl ether and mixed organic solvents, Occup Environ Med, vol.59, pp.625-628, 2002.

J. D. Meeker, A. M. Calafat, and R. Hauser, Urinary bisphenol A concentrations in relation to serum thyroid and reproductive hormone levels in men from an infertility clinic, Environ Sci Technol, vol.44, pp.1458-1463, 2010.

J. Mendiola, N. Jorgensen, A. M. Andersson, A. M. Calafat, and X. Ye, Are environmental levels of bisphenol a associated with reproductive function in fertile men?, Environ Health Perspect, vol.118, pp.1286-1291, 2010.

T. Galloway, R. Cipelli, J. Guralnik, L. Ferrucci, and S. Bandinelli, Daily bisphenol A excretion and associations with sex hormone concentrations: results from the InCHIANTI adult population study, Environ Health Perspect, vol.118, pp.1603-1608, 2010.

L. Ye, B. Zhao, G. Hu, Y. Chu, and R. S. Ge, Inhibition of human and rat testicular steroidogenic enzyme activities by bisphenol A, Toxicol Lett, vol.207, pp.137-142, 2011.

C. S. Mazur, J. F. Kenneke, J. K. Hess-wilson, and J. C. Lipscomb, Differences between human and rat intestinal and hepatic bisphenol A glucuronidation and the influence of alamethicin on in vitro kinetic measurements, Drug Metab Dispos, vol.38, pp.2232-2238, 2010.

J. A. Taylor, V. Saal, F. S. Welshons, W. V. Drury, B. Rottinghaus et al., Similarity of bisphenol A pharmacokinetics in rhesus monkeys and mice: relevance for human exposure, Environ Health Perspect, vol.119, pp.422-430, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01137161

D. Nakamura, Y. Yanagiba, Z. Duan, Y. Ito, and A. Okamura, Bisphenol A may cause testosterone reduction by adversely affecting both testis and pituitary systems similar to estradiol, Toxicol Lett, vol.194, pp.16-25, 2010.

E. P. Murono, R. C. Derk, and J. H. De-leon, Differential effects of octylphenol, 17beta-estradiol, endosulfan, or bisphenol A on the steroidogenic competence of cultured adult rat Leydig cells, Reprod Toxicol, vol.15, pp.551-560, 2001.

B. T. Akingbemi, C. M. Sottas, A. I. Koulova, G. R. Klinefelter, and M. P. Hardy, Inhibition of testicular steroidogenesis by the xenoestrogen bisphenol A is associated with reduced pituitary luteinizing hormone secretion and decreased steroidogenic enzyme gene expression in rat Leydig cells, Endocrinology, vol.145, pp.592-603, 2004.

T. L. Gaskell, L. L. Robinson, N. P. Groome, R. A. Anderson, and P. T. Saunders, Differential expression of two estrogen receptor beta isoforms in the human fetal testis during the second trimester of pregnancy, J Clin Endocrinol Metab, vol.88, pp.424-432, 2003.

G. G. Kuiper, J. G. Lemmen, B. Carlsson, J. C. Corton, and S. H. Safe, Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor beta, Endocrinology, vol.139, pp.4252-4263, 1998.

A. Bouskine, M. Nebout, F. Brucker-davis, M. Benahmed, and P. Fenichel, Low doses of bisphenol A promote human seminoma cell proliferation by activating PKA and PKG via a membrane G-protein-coupled estrogen receptor, Environ Health Perspect, vol.117, pp.1053-1058, 2009.

N. Chevalier, A. Vega, A. Bouskine, B. Siddeek, and J. F. Michiels, 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.

S. Takayanagi, T. Tokunaga, X. Liu, H. Okada, and A. Matsushima, Endocrine disruptor bisphenol A strongly binds to human estrogen-related receptor gamma (ERRgamma) with high constitutive activity, Toxicol Lett, vol.167, pp.95-105, 2006.

D. F. Swaab, Sexual differentiation of the brain and behavior, Best Pract Res Clin Endocrinol Metab, vol.21, pp.431-444, 2007.

J. Merlet, C. Racine, E. Moreau, S. G. Moreno, and R. Habert, Male fetal germ cells are targets for androgens that physiologically inhibit their proliferation, Proc Natl Acad Sci U S A, vol.104, pp.3615-3620, 2007.

S. Salian, T. Doshi, and G. Vanage, Perinatal exposure of rats to Bisphenol A affects fertility of male offspring-an overview, Reprod Toxicol, vol.31, pp.359-362, 2011.