Skip to main content

Endocrine Disruptors (Xenoestrogens): An Overview

  • Chapter
  • First Online:
Plastics in Dentistry and Estrogenicity

Abstract

In the last decades, a large number of natural and synthetic chemicals have been identified as interfering with the endocrine system; they are collectively termed endocrine-disrupting chemicals (EDCs) or endocrine disruptors. According to the working definition of the Environmental Protection Agency (EPA), an endocrine disruptor is “an exogenous agent that interferes with the synthesis, release, transport, metabolism, binding, action, or elimination of natural hormones in the body responsible for the maintenance of homeostasis, reproduction, regulation of developmental processes and/or behavior”[1]. Endocrine disruptors comprise more than 100.000 synthetic chemical compounds that belong to different classes. A subset of the endocrine disruptors, including synthetic estrogens, natural products, commercial chemicals, industrial compounds, or by-products among which plastics, are known as environmental estrogens or xenoestrogens; they confer estrogenic potential (“estrogenicity”) translated as affinity to the estrogen receptors (ER) (α or β), thus ability to activate expression of estrogen-dependent genes or stimulation of cell proliferation of ER-competent cells [2].

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 84.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 129.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 109.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Kavlock RJ, Daston GP, DeRosa C, Fenner-Crisp P, Gray LE, Kaattari S, Lucier G, Luster M, Mac MJ, Maczka C, Miller R, Moore J, Rolland R, Scott G, Sheehan DM, Sinks T, Tilson HA (1996) Research needs for the risk assessment of health and environmental effects of endocrine disruptors: a report of the USEPA-sponsored workshop. Environ Health Perspect 104(Suppl 4):715–740

    PubMed  Google Scholar 

  2. Katzenellnbogen JA (1995) The structural pervasiveness of estrogenic activity. Environ Health Perspect 103(suppl 7):99–101

    Google Scholar 

  3. Hammond GL (1995) Potential functions of plasma steroid-binding proteins. Trends Endocrinol Metab 6:298–304

    PubMed  Google Scholar 

  4. Apter D, Bolton NJ, Hammond GL, Vihko R (1984) Serum sex hormone-binding globulin during puberty in girls and in different types of adolescent menstrual cycles. Acta Endocrinol (Copenh) 107:413–419

    Google Scholar 

  5. Belgorosky A, Rivarola MA (1986) Progressive decrease in serum sex hormone-binding globulin from infancy to late prepuberty in boys. J Clin Endocrinol Metab 63:510–512

    PubMed  Google Scholar 

  6. Hammond GL, Langley MS, Robinson PA, Nummi S, Lund L (1984) Serum steroid binding protein concentrations, distribution of progestogens, and bioavailability of testosterone during treatment with contraceptives containing desogestrel or levonorgestrel. Fertil Steril 42:44–51

    PubMed  Google Scholar 

  7. Colborn T (2004) Endocrine disruption overview: are males at risk? Adv Exp Med Biol 545:189–201

    PubMed  Google Scholar 

  8. Kinlay M, Plant JA, Bell JN, Voulvoulis N (2008) Endocrine disrupting pesticides: implications for risk assessment. Environ Int 34:168–183

    Google Scholar 

  9. Golden RJ, Noller KL, Titus-Ernstoff L, Kaufman RH, Mittendorf R, Stillman R, Reese EA (1998) Environmental endocrine modulators and human health: an assessment of the biological evidence. Crit Rev Toxicol 28:109–227

    PubMed  Google Scholar 

  10. Toppari J, Larsen JC, Christiansen P, Giwercman A, Grandjean P, Guillette LJ Jr, Jégou B, Jensen TK, Jouannet P, Keiding N, Leffers H, McLachlan JA, Meyer O, Müller J, Rajpert-De Meyts E, Scheike T, Sharpe R, Sumpter J, Skakkebaek NE (1996) Male reproductive health and environmental xenoestrogens. Environ Health Perspect 104(Suppl 4):741–803

    PubMed  Google Scholar 

  11. Colborn T, Dumanoski D, Myers JP (1995) Our stolen future. Penguin, New York

    Google Scholar 

  12. van Larebeke N, Hens L, Schepens P, Covaci A, Baeyens J, Everaert K, Bernheim JL, Vlietinck R, De Poorter G (2001) The Belgian PCB and dioxin incident of January–June 1999: exposure data and potential impact on health. Environ Health Perspect 109:265–273

    PubMed  Google Scholar 

  13. Carpenter DO, Arcaro KF, Bush B, Niemi WD, Pang S, Vakharia DD (1998) Human health and chemical mixtures: an overview. Environ Health Perspect 106(Suppl 6):1263–1270

    PubMed  Google Scholar 

  14. Damstra T, Barlow S, Bergman A, Kavlock R, Van der Kraak G (eds) (2002) International Programme on Chemical Safety: global assessment of the state-of-the-science on endocrine disruptors. WHO, Geneva. http://www.who.int/pcs/emerg_site/edc/global_edc_TOC.htm. Accessed 20 Oct 2013

    Google Scholar 

  15. Welshons WV, Nagel SC, vom Saal FS (2006) Large effects from small exposures. III. Endocrine mechanisms mediating effects of bisphenol A at levels of human exposure. Endocrinology 147(6 Suppl):S56–S69

    PubMed  Google Scholar 

  16. Brotons JA, Olea-Serrano MF, Villalobos M, Pedraza V, Olea N (1995) Xenoestrogens released from lacquer coatings in food cans. Environ Health Perspect 103:608–612

    PubMed  Google Scholar 

  17. Muncke J (2009) Exposure to endocrine disrupting compounds via the food chain: is packaging a relevant source? Sci Total Environ 407:4549–4559

    PubMed  Google Scholar 

  18. Vandenberg LN, Hauser R, Marcus M, Olea N, Welshons WV (2007) Human exposure to bisphenol A (BPA). Reprod Toxicol 24:139–177

    PubMed  Google Scholar 

  19. Grund S, Higley E, Schönenberger R, Suter MJF, Giesy JP, Braunbeck T, Hecker M, Hollert H (2010) The endocrine disrupting potential of sediments from the Upper Danube River (Germany) as revealed by in vitro bioassays and chemical analysis. Environ Sci Pollut Res 18:446–460

    Google Scholar 

  20. Rodgers-Gray TP, Jobling S, Kelly C, Morris S, Brighty G, Waldock MJ, Sumpter JP, Tyler CR (2001) Exposure of juvenile roach (Rutilus rutilus) to treated sewage effluent induces dose-dependent and persistent disruption in gonadal duct development. Environ Sci Technol 35:462–470

    PubMed  Google Scholar 

  21. Ternes TA, Stumpf M, Mueller J, Haberer K, Wilken RD, Servos M (1999) Behavior and occurrence of estrogens in municipal sewage treatment plants–I. Investigations in Germany, Canada and Brazil. Sci Total Environ 225:81–90

    PubMed  Google Scholar 

  22. Jobling S, Williams R, Johnson A, Taylor A, Gross-Sorokin M, Nolan M, Tyler CR, van Aerle R, Santos E, Brighty G (2006) Predicted exposures to steroid estrogens in U.K. rivers correlate with widespread sexual disruption in wild fish populations. Environ Health Perspect 114(Suppl 1):32–39

    PubMed  Google Scholar 

  23. Keiter S, Rastall A, Kosmehl T, Wurm K, Erdinger L, Braunbeck T, Hollert H (2006) Ecotoxicological assessment of sediment, suspended matter and water samples in the upper Danube river – a pilot study in search for the causes for the decline of fish catches. Environ Sci Pollut Res 13:308–319

    Google Scholar 

  24. Gaido K, Dohme L, Wang F, Chen I, Blankvoort B, Ramamoorthy K, Safe S (1998) Comparative estrogenic activity of wine extracts and organochlorine pesticide residues in food. Environ Health Perspect 106(Suppl 6):1347–1351

    PubMed  Google Scholar 

  25. Borchers A, Teuber SS, Keen CL, Gershwin ME (2010) Food safety. Clin Rev Allergy Immunol 39:95–141

    PubMed  Google Scholar 

  26. Judd N, Griffith WC, Faustman EM (2004) Contribution of PCB exposure from fish consumption to total dioxin-like dietary exposure. Regul Toxicol Pharmacol 40:125–135

    PubMed  Google Scholar 

  27. Grimvall E, Rylander L, Nilsson-Ehle P, Nilsson U, Strömberg U, Hagmar L, Ostman C (1997) Monitoring of polychlorinated biphenyls in human blood plasma: methodological developments and influence of age, lactation, and fish consumption. Arch Environ Contam Toxicol 32:329–336

    PubMed  Google Scholar 

  28. Onn Wong K, Woon Leo L, Seah HL (2005) Dietary exposure assessment of infants to bisphenol A from the use of polycarbonate baby milk bottles. Food Addit Contam 22:280–288

    PubMed  Google Scholar 

  29. Pinto B, Reali D (2009) Screening of estrogen-like activity of mineral water stored in PET bottles. Int J Hyg Environ Health 212:228–232

    PubMed  Google Scholar 

  30. Wagner M, Oehlmann J (2009) Endocrine disruptors in bottled mineral water: total estrogenic burden and migration from plastic bottles. Environ Sci Pollut Res Int 16:278–286

    PubMed  Google Scholar 

  31. Witorsch RJ (2002) Endocrine disruptors: can biological effects and environmental risks be predicted? Regul Toxicol Pharmacol 3:118–130

    Google Scholar 

  32. Guo YL, Ryan JJ, Lau BP, Yu ML, Hsu CC (1997) Blood serum levels of PCBs and PCDFs in Yucheng women 14 years after exposure to a toxic rice oil. Arch Environ Contam Toxicol 33:104–108

    PubMed  Google Scholar 

  33. Milbrath MO, Wenger Y, Chang CW, Emond C, Garabrant D, Gillespie BW, Jolliet O (2009) Apparent half-lives of dioxins, furans, and polychlorinated biphenyls as a function of age, body fat, smoking status, and breast-feeding. Environ Health Perspect 117:417–425

    PubMed  Google Scholar 

  34. Garritano S, Pinto B, Calderisi M, Cirillo T, Amodio-Cocchieri R, Reali D (2006) Estrogen-like activity of seafood related to environmental chemical contaminants. Environ Health 30:5–9

    Google Scholar 

  35. Kogevinas M (2001) Human health effects of dioxins: cancer, reproductive and endocrine system effects. Hum Reprod Update 7:331–339

    PubMed  Google Scholar 

  36. Giguere V, Yang N, Segui P, Evans RM (1988) Identification of a new class of steroid hormone receptors. Nature 331:91–94

    PubMed  Google Scholar 

  37. Enmark E, Pelto-Huikko M, Grandien K, Lagercrantz S, Lagercrantz J, Fried G, Nordenskjöld M, Gustafsson JA (1997) Human estrogen receptor b–gene structure, chromosomal localization, expression pattern. J Clin Endocrinol Metab 82:4258–4265

    PubMed  Google Scholar 

  38. Arts J, Kuiper GG, Janssen JM, Gustafsson JA, Löwik CW, Pols HA, van Leeuwen JP (1997) Differential expression of estrogen receptor α and β mRNA during differentiation of human osteoblast SV-HFO cells. Endocrinology 138:5067–5070

    PubMed  Google Scholar 

  39. Kuiper GGJM, Carlsson B, Grandien K, Enmark E, Haggblad J, Nilsson S, Gustafsson J-A (1997) Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors α and β. Endocrinology 3:863–870

    Google Scholar 

  40. Kuiper GGJM, Shughrue PJ, Pelto-Huikko M, Merchenthaler I, Gustafsson J-A (1998) The estrogen receptor β subtype: a novel mediator of estrogen action in neuroendocrine systems. Front Neuroendocrinol 19:253–286

    PubMed  Google Scholar 

  41. Lindner V, Kim SK, Karas RH, Kuiper GGJM, Gustafsson J-A, Mendelsohn ME (1998) Increased expression of estrogen receptor-β mRNA in male blood vessels after vascular injury. Circ Res 83:224–229

    PubMed  Google Scholar 

  42. Osterlund M, Kuiper GJM, Gustafsson J-A, Hurd YL (1998) Differential distribution and regulation of estrogen receptor α and β mRNA within the female rat brain. Mol Brain Res 54:175–180

    PubMed  Google Scholar 

  43. Matthews J, Gustafsson JA (2003) Estrogen signaling: a subtle balance between ERα and Erβ. Mol Interv 3:281–292

    PubMed  Google Scholar 

  44. Moore JT, McKee DD, Slentz-Kesler K, Moore LB, Jones SA, Horne EL, Su JL, Kliewer SA, Lehmann JM, Willson TM (1998) Cloning and characterization of human estrogen receptor β isoforms. Biochem Biophys Res Commun 247:75–78

    PubMed  Google Scholar 

  45. Spencer TE, Jenster G, Burcin MM, Allis CD, Zhou J, Mizzen CA et al (1997) Steroid receptor coactivator-1 is a histone acetyltransferase. Nature 389:194–198

    PubMed  Google Scholar 

  46. McKenna NJ, Lanz RB, O’Malley BW (1999) Nuclear receptor coregulators: cellular and molecular biology. Endocr Rev 20:321–344

    PubMed  Google Scholar 

  47. Klein-Hitpass L, Tsai SY, Greene GL, Clark JH, Tsai MJ, O’Malley BW (1989) Specific binding of estrogen receptor to the estrogen response element. Mol Cell Biol 9:43–49

    PubMed  Google Scholar 

  48. Vanacker JM, Pettersson K, Gustafsson JA, Laudet V (1999) Transcriptional targets shared by estrogen receptor-related receptors (ERRs) and estrogen receptor (ER)alpha but not by ER beta. EMBO J 18:4270–4279

    PubMed  Google Scholar 

  49. Chen JD, Evans RM (1995) A transcriptional co-repressor that interacts with nuclear hormone receptors. Nature 377:454–457

    PubMed  Google Scholar 

  50. Heery DM, Kalkhoven E, Hoare S, Parker MG (1997) A signature motif in transcriptional co-activators mediates binding to nuclear receptors. Nature 387:733–736

    PubMed  Google Scholar 

  51. Nagy L, Kao HY, Chakravarti D, Lin RJ, Hassig CA, Ayer D (1997) Nuclear receptor repression mediated by a complex containing SMRT, mSin3A, and histone deacetylase. Cell 89:373–380

    PubMed  Google Scholar 

  52. Lomvardas S, Thanos D (2002) Opening chromatin. Mol Cell 9:209–211

    PubMed  Google Scholar 

  53. Johansson L, Thomsen JS, Damdimopoulos AE, Spyrou G, Gustafsson J-A, Treuter E (1999) The orphan nuclear receptor SHP inhibits agonist-dependent transcriptional activity of estrogen receptors ERα and ERβ. J Biol Chem 247:345–353

    Google Scholar 

  54. Seol W, Hanstein B, Brown M, Moore DD (1998) Inhibition of estrogen receptor action by the orphan receptor SHP (Short Heterodimer Partner). Mol Endocrinol 12:1551–1557

    PubMed  Google Scholar 

  55. Brown M, Moore DD (1998) Inhibition of estrogen receptor action by the orphan receptor SHP (Short Heterodimer Partner). Mol Endocrinol 12:1551–1557

    PubMed  Google Scholar 

  56. Warnmark A, Almlof T, Leers J, Gustafsson JA, Treuter E (2001) Differential recruitment of the mammalian mediator subunit TRAP220 by estrogen receptors ERalpha and ERbeta. J Biol Chem 276:23397–23404

    PubMed  Google Scholar 

  57. Wong CW, Komm B, Cheskis BJ (2001) Structure-function evaluation of ERalpha and Erbeta interplay with SRC family coactivators. ER selective ligands. Biochemistry 40:6756–6765

    PubMed  Google Scholar 

  58. Graham JD, Bain DL, Richer JK, Jackson TA, Tung L, Horwitz KB (2000) Nuclear receptor conformation, coregulators, and tamoxifen-resistant breast cancer. Steroids 65:579–584

    PubMed  Google Scholar 

  59. Smith CL, Nawaz Z, O’Malley BW (1997) Coactivator and corepressor regulation of the agonist/antagonist activity of the mixed antiestrogen, 4-hydroxytamoxifen. Mol Endocrinol 11:657–666

    PubMed  Google Scholar 

  60. Zhang X, Jeyakumar M, Petukhov S, Bagchi MK (1998) A nuclear receptor corepressor modulates transcriptional activity of antagonist-occupied steroid hormone receptor. Mol Endocrinol 12:513–524

    PubMed  Google Scholar 

  61. Bramlett KS, Wu Y, Burris TP (2001) Ligands specify coactivator nuclear receptor (NR) box affinity for estrogen receptor subtypes. Mol Endocrinol 15:909–922

    PubMed  Google Scholar 

  62. Kraichely DM, Sun J, Katzenellenbogen JA, Katzenellenbogen BS (2000) Conformational changes and coactivator recruitment by novel ligands for estrogen receptor alpha and estrogen receptor-beta: correlations with biological character and distinct differences among SRC coactivator family members. Endocrinology 141:3534–3545

    PubMed  Google Scholar 

  63. Ko L, Cardona GR, Iwasaki T, Bramlett KS, Burris TP, Chin WW (2002) Ser-884 adjacent to the LXXLL motif of coactivator TRBP defines selectivity for ERs and TRs. Mol Endocrinol 16:128–140

    PubMed  Google Scholar 

  64. Pettersson K, Grandien K, Kuiper GGJM, Gustafsson J-A (1997) Mouse estrogen receptor beta forms estrogen response element binding heterodimers with estrogen receptor alpha. Mol Endocrinol 11:1486–1496

    PubMed  Google Scholar 

  65. Chang EC, Frasor J, Komm B, Katzenellenbogen BS (2006) Impact of estrogen receptor beta on gene networks regulated by estrogen receptor alpha in breast cancer cells. Endocrinology 147:4831–4842

    PubMed  Google Scholar 

  66. Liu MM, Albanese C, Anderson CM, Hilty K, Webb P, Uht RM (2002) Opposing action of estrogen receptors alpha and beta on cyclin D1 gene expression. J Biol Chem 277:24353–24360

    PubMed  Google Scholar 

  67. Pettersson K, Delaunay F, Gustafsson JA (2000) Estrogen receptor beta acts as a dominant regulator of estrogen signaling. Oncogene 19:4970–4978

    PubMed  Google Scholar 

  68. Monroe DG, Secreto FJ, Subramaniam M, Getz BJ, Khosla S, Spelsberg TC (2005) Estrogen receptor alpha and beta heterodimers exert unique effects on estrogen- and tamoxifen-dependent gene expression in human U2OS osteosarcoma cells. Mol Endocrinol 19:1555–1568

    PubMed  Google Scholar 

  69. Stossi F, Barnett DH, Frasor J, Komm B, Lyttle CR, Katzenellenbogen BS (2004) Transcriptional profiling of estrogen-regulated gene expression via estrogen receptor (ER) alpha or ER beta in human osteosarcoma cells: distinct and common target genes for these receptors. Endocrinology 145:3473–3486

    PubMed  Google Scholar 

  70. Kushner PJ, Agard DA, Greene GL, Scanlan TS, Shiau AK, Uht RM, Webb P (2000) Estrogen receptor pathways to AP-1. J Steroid Biochem Mol Biol 74:311–317

    PubMed  Google Scholar 

  71. Safe S (2001) Transcriptional activation of genes by 17 b-estradiol through estrogen receptor-Sp1 interactions. Vitam Horm 62:231–252

    PubMed  Google Scholar 

  72. Gaub MP, Bellard M, Scheuer I, Chambon P, Sassone-Corsi P (1990) Activation of the ovalbumin gene by the estrogen receptor involves the fos–jun complex. Cell 63:1267–1276

    PubMed  Google Scholar 

  73. Li C, Briggs MR, Ahlborn TE, Kraemer FB, Liu J (2001) Requirement of Sp1 and estrogen receptor alpha interaction in 17beta estradiol-mediated transcriptional activation of the low density lipoprotein receptor gene expression. Endocrinology 142:1546–1553

    PubMed  Google Scholar 

  74. Schmitt M, Bausero P, Simoni P, Queuche D, Geoffroy V, Marschal C, Kempf J, Quirin-Stricker C (1995) Positive and negative effects of nuclear receptors on transcription activation by AP-1 of the human choline acetyl-transferase proximal promoter. J Neurosci Res 40:152–164

    PubMed  Google Scholar 

  75. Umayahara Y, Kawamori R, Watada H, Imano E, Iwama N, Morishima T, Yamasaki Y, Kajimoto Y, Kamada T (1994) Estrogen regulation of the insulin-like growth factor I gene transcription involves an AP-1 enhancer. J Biol Chem 269:16433–16442

    PubMed  Google Scholar 

  76. Aronica SM, Katzenellenbogen BS (1993) Stimulation of estrogen receptor mediated transcription and alteration in the phosphorylation state of the rat uterine estrogen receptor by estrogen, cyclic adenosine monophosphate, and insulin-like growth factor-I. Mol Endocrinol 7:743–752

    PubMed  Google Scholar 

  77. Ignar-Trowbridge DM, Nelson KG, Bidwell MC, Curtis SW, Washburn TF, McLachlan JA, Korach KS (1992) Coupling of dual signaling pathways: epidermal growth factor action involves the estrogen receptor. Proc Natl Acad Sci U S A 89:4658–4662

    PubMed  Google Scholar 

  78. Kato S, Endoh H, Masuhiro Y, Kitamoto T, Uchiyama S, Sasaki H, Masushige S, Gotoh Y, Nishida E, Kawashima H, Metzger D, Chambon P (1995) Activation of the estrogen receptor through phosphorylation by mitogen-activated protein kinase. Science 270:1491–1494

    PubMed  Google Scholar 

  79. Bunone G, Briand PA, Miksicek RJ, Picard D (1996) Activation of the unliganded estrogen receptor by EGF involves the MAP kinase pathway and direct phosphorylation. EMBO J 15:2174–2183

    PubMed  Google Scholar 

  80. Gottlicher M, Heck S, Herrlich P (1998) Transcriptional cross-talk, the second mode of steroid hormone receptor action. J Mol Med 76:480–489

    PubMed  Google Scholar 

  81. O’Lone R, Frith MC, Karlsson EK, Hansen U (2004) Genomic targets of nuclear estrogen receptors. Mol Endocrinol 18:1859–1875

    PubMed  Google Scholar 

  82. Porter W, Saville B, Hoivik D, Safe S (1997) Functional synergy between the transcription factor Sp1 and the estrogen receptor. Mol Endocrinol 11:1569–1580

    PubMed  Google Scholar 

  83. Stein B, Yang MX (1995) Repression of the interleukin-6 promoter by estrogen receptor is mediated by NF-kappa B and C/EBP beta. Mol Cell Biol 15:4971–4979

    PubMed  Google Scholar 

  84. Razandi M, Pedram A, Greene GL, Levin ER (1999) Cell membrane and nuclear estrogen receptors (ERs) originate from a single transcript: studies of ERalpha and ERbeta expressed in Chinese hamster ovary cells. Mol Endocrinol 13:307–319

    PubMed  Google Scholar 

  85. Improta-Brears T, Whorton AR, Codazzi F, York JD, Meyer T, McDonnell DP (1999) Estrogen-induced activation of mitogen-activated protein kinase requires mobilization of intracellular calcium. Proc Natl Acad Sci U S A 96:4686–4691

    PubMed  Google Scholar 

  86. Ahmad S, Singh N, Glazer RI (1999) Role of AKT1 in 17beta estradiol- and insulin-like growth factor I (IGF-I)-dependent proliferation and prevention of apoptosis in MCF-7 breast carcinoma cells. Biochem Pharmacol 58:425–430

    PubMed  Google Scholar 

  87. Castoria G, Migliaccio A, Bilancio A, Di Domenico M, deFalco A, Lombardi M, Fiorentino R et al (2001) PI3-kinase in concert with Src promotes the S-phase entry of oestradiol-stimulated MCF-7 cells. EMBO J 20:6050–6059

    PubMed  Google Scholar 

  88. Chen Z, Yuhanna IS, Galcheva-Gargova Z, Karas RH, Mendelsohn ME, Shaul PW (1999) Estrogen receptor alpha mediates the nongenomic activation of endothelial nitric oxide synthase by estrogen. J Clin Invest 103:401–406

    PubMed  Google Scholar 

  89. Jessop HL, Sjoberg M, Cheng MZ, Zaman G, Wheeler- Jones CP, Lanyon LE (2001) Mechanical strain and estrogen activate estrogen receptor a in bone cells. J Bone Miner Res 16:1045–1055

    PubMed  Google Scholar 

  90. Kousteni S, Bellido T, Plotkin LI, O’Brien CA, Bodenner DL, Han L, Han K, DiGregorio GB, Katzenellenbogen JA, Katzenellenbogen BS, Roberson PK, Weinstein RS, Jilka RL, Manolagas SC (2001) Nongenotropic, sex-nonspecific signaling through the estrogen or androgen receptors: dissociation from transcriptional activity. Cell 104:719–730

    PubMed  Google Scholar 

  91. Migliaccio A, Di Domenico M, Castoria G, de Falco A, Bontempo P, Nola E, Auricchio F (1996) Tyrosine kinase/p21ras/MAP-kinase pathway activation by estradiol-receptor complex in MCF-7 cells. EMBO J 15:1292–1300

    PubMed  Google Scholar 

  92. Watters JJ, Campbell JS, Cunningham MJ, Krebs EG, Dorsa DM (1997) Rapid membrane effects of steroids in neuroblastoma cells: effects of estrogen on mitogen activated protein kinase signalling cascade and c-fos immediate early gene transcription. Endocrinology 138:4030–4033

    PubMed  Google Scholar 

  93. Ordonez-Moran P, Munoz A (2009) Nuclear receptors genomic and non-genomic effects converge. Cell Cycle 8:1675–1680

    PubMed  Google Scholar 

  94. Pappas TC, Gametchu B, Watson CS (1995) Membrane estrogen receptors identified by multiple antibody labeling and impeded-ligand binding. FASEB J 9:404–410

    PubMed  Google Scholar 

  95. Ding Q, Gros R, Limbird L, Chorazyczewski J, Feldman RD (2009) Estradiol-mediated ERK phosphorylation and apoptosis in vascular smooth muscle cells requires GPR 30. Am J Physiol Cell Physiol 297:C1178–C1187

    PubMed  Google Scholar 

  96. Filardo EJ, Quinn JA, Bland KI, Frackleton AR (2000) Estrogen-induced activation of Erk-1 and Erk-2 requires the G protein-coupled receptor homolog, GPR30, and occurs via trans-activation of the epidermal growth factor receptor through release of HB-EGF. Mol Endocrinol 14:1649–1660

    PubMed  Google Scholar 

  97. Migliaccio A, Piccolo D, Castoria G, Di Domenico M, Bilancio A, Lombardi M, Gong W, Beato M, Auricchio F (1998) Activation of the Src/p21ras/Erk pathway by progesterone receptor via cross-talk with estrogen receptor. EMBO J 17:2008–2018

    PubMed  Google Scholar 

  98. Razandi M, Pedram A, Park ST, Levin ER, Oh P, Schnitzer J (2003) Proximal events in signaling by plasma membrane estrogen receptors ERs associate with and regulate the production of caveolin: implications for signaling and cellular actions. J Biol Chem 278:2701–2712

    PubMed  Google Scholar 

  99. Simoncini T, Hafezi-Moghadam A, Brazil DP, Ley K, Chin WW, Liao JK (2000) Interaction of oestrogen receptor with the regulatory subunit of phosphatidylinositol-3-OH kinase. Nature 407:538–541

    PubMed  Google Scholar 

  100. Song RX, McPherson RA, Adam L, Bao Y, Shupnik M, Kumar R, Santen RJ (2002) Linkage of rapid estrogen action to MAPK activation by ERalpha-Shc association and Shc pathway activation. Mol Endocrinol 16:116–127

    PubMed  Google Scholar 

  101. Wyckoff MH, Chambliss KL, Mineo C, Yuhanna IS, Mendelsohn ME, Mumby SM, Shaul PW (2001) Plasma membrane estrogen receptors are coupled to endothelial nitric-oxide synthase through Gs(i). J Biol Chem 276:27071–27076

    PubMed  Google Scholar 

  102. Tremblay A, Tremblay GB, Labrie F, Giguere V (1999) Ligand-independent recruitment of SRC-1 to estrogen receptor beta through phosphorylation of activation function AF-1. Mol Cell 3:513–519

    PubMed  Google Scholar 

  103. Singleton DW, Feng Y, Burd CJ, Khan SA (2003) Non-genomic activity and subsequent c-fos induction by estrogen receptor ligands are not sufficient to promote deoxyribonucleic acid synthesis in human endometrial adenocarcinoma cells. Endocrinology 144:121–128

    PubMed  Google Scholar 

  104. Stancel GM, Boettger-Tong HL, Chiappetta C, Hyder SM, Kirkland JL, Murthy L, Loose-Mitchell DS (1995) Toxicity of endogenous and environmental estrogens: what is the role of elemental interactions? Environ Health Perspect 103(Suppl 7):29–33

    PubMed  Google Scholar 

  105. Kuiper GGJM, Lemmen JG, Carlsson B, Corton JC, Safe SH, van der Saag PT, van der Bur B, Gustafsson J-A (1998) Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor beta. Endocrinology 139:4252–4263

    PubMed  Google Scholar 

  106. Gulden M, Turan A, Seibert H (1997) Texte 46/97 Substanzen mit endokriner Wirkung in Oberflachengewassern Umweltbundesamt (German Environmental Agency) Berichtsnummer UBA–FB 97-068, ISSN 0722-186X

    Google Scholar 

  107. Jorgensen M, Vendelbo B, Skakkebaek NE, Leffers H (2000) Assaying estrogenicity by quantitating the expression levels of endogenous estrogen-regulated genes. Environ Health Perspect 108:403–412

    PubMed  Google Scholar 

  108. Diel P, Olff S, Schmidt S, Michna H (2002) Effects of the environmental estrogens bisphenol A, o, p′-DDT, p-tert-octylphenol and coumestrol on apoptosis induction cell proliferation and the expression of estrogen sensitive molecular parameters in the human breast cancer cell line MCF-7. J Steroid Biochem Mol Biol 80:61–70

    PubMed  Google Scholar 

  109. Soto AM, Sonnenschein C, Chung KL, Fernandez MF, Olea N, Serrano FO (1995) The E-SCREEN assay as a tool to identify estrogens: an update on estrogenic environmental pollutants. Environ Health Perspect 103(Suppl 7):113–122

    PubMed  Google Scholar 

  110. Kelce WR, Stone CR, Laws SC, Gray LE, Kemppainen JA, Wilson EM (1995) Persistent DDT metabolite p, p′-DDE is a potent androgen receptor antagonist. Nature 375:581–585

    PubMed  Google Scholar 

  111. Payne J, Scholze M, Kortenkamp A (2001) Mixtures of four organochlorines enhance human breast cancer cell proliferation. Environ Health Perspect 109:391–397

    PubMed  Google Scholar 

  112. Bulayeva NN, Watson CS (2004) Xenoestrogen-induced ERK- and ERK-2 activation via multiple membrane-initiated signaling pathways. Environ Health Perspect 112:1481–1487

    PubMed  Google Scholar 

  113. Ruehlmann DO, Steinert JR, Valverde MA, Jacob R, Mann GE (1998) Environmental estrogenic pollutants induce acute vascular relaxation by inhibiting L-type Ca2+ channels in smooth muscle cells. FASEB J 12:613–619

    PubMed  Google Scholar 

  114. Nadal A, Ropero AB, Laribi O, Maillet M, Fuentes E, Soria B (2000) Nongenomic actions of estrogens and xenoestrogens by binding at a plasma membrane receptor unrelated to estrogen receptor alpha and estrogen receptor beta. Proc Natl Acad Sci U S A 97:11603–11608

    PubMed  Google Scholar 

  115. Zivadinovic D, Gametchu B, Watson CS (2005) Membrane estrogen receptor-alpha levels in MCF-7 breast cancer cells predict cAMP and proliferation responses. Breast Cancer Res 7:R101–R112

    PubMed  Google Scholar 

  116. Zivadinovic D, Watson CS (2005) Membrane estrogen receptor-alpha levels predict estrogen-induced ERK1/2 activation in MCF-7 cells. Breast Cancer Res 7:R130–R144

    PubMed  Google Scholar 

  117. Narita S, Goldblum RM, Watson CS, Brooks EG, Estes DM, Curran EM, Midoro-Horiuti T (2007) Environmental estrogens induce mast cell degranulation and enhance IgE-mediated release of allergic mediators. Environ Health Perspect 115:48–52

    PubMed  Google Scholar 

  118. Alyea RA, Watson CS (2009) Nongenomic mechanisms of physiological estrogen-mediated dopamine efflux. BMC Neurosci 10:59–67

    PubMed  Google Scholar 

  119. Otto C, Fuchs I, Altmann H, Klewer M, Schwarz G, Bohlmann R, Nguyen D, Zorn L, Vonk R, Prelle K, Osterman T, Malmström C, Fritzemeier KH (2008) In vivo characterization of estrogen receptor modulators with reduced genomic versus nongenomic activity in vitro. J Steroid Biochem Mol Biol 111:95–100

    PubMed  Google Scholar 

  120. Silva E, Kabil A, Kortenkamp A (2010) Cross-talk between non-genomic and genomic signalling pathways–distinct effect profiles of environmental estrogens. Toxicol Appl Pharmacol 245:160–170

    PubMed  Google Scholar 

  121. Kochukov MY, Jeng YJ, Watson CS (2009) Alkylphenol xenoestrogens with varying carbon chain lengths differentially and potently activate signaling and functional responses in GH3/B6/F10 somatomammotropes. Environ Health Perspect 117:723–730

    PubMed  Google Scholar 

  122. Kwack SJ, Kwon O, Kim HS, Kim SS, Kim SH, Sohn KH, Lee RD, Park CH, Jeung EB, An BS, Park KL (2002) Comparative evaluation of alkylphenolic compounds on estrogenic activity in vitro and in vivo. J Toxicol Environ Health A 65:419–431

    PubMed  Google Scholar 

  123. Tabira Y, Nakai M, Asai D, Yakabe Y, Tahara Y, Shinmyozu T, Noguchi M, Takatsuki M, Shimohigashi Y (1999) Structural requirements of para-alkylphenols to bind to estrogen receptor. Eur J Biochem 262:240–245

    PubMed  Google Scholar 

  124. Norman AW, Mizwicki MT, Norman DPG (2004) Steroid-hormone rapid actions, membrane receptors and a conformational ensemble model. Nat Rev Drug Discov 3:27–34

    PubMed  Google Scholar 

  125. Coosen R, van Velsen FL (1989) Effects of the beta-isomer of hexachlorocyclohexane on estrogen-sensitive human mammary tumor cells. Toxicol Appl Pharmacol 101:310–318

    PubMed  Google Scholar 

  126. Steinmetz R, Young PC, Caperell-Grant A, Gize EA, Madhukar BV, Ben Jonathan N, Bigsby RM (1996) Novel estrogenic action of the pesticide residue beta-hexachlorocyclohexane in human breast cancer cells. Cancer Res 56:5403–5409

    PubMed  Google Scholar 

  127. Silva E, Scholze M, Kortenkamp A (2007) Activity of xenoestrogens at nanomolar concentrations in the E-Screen assay. Environ Health Perspect 115(Suppl 1):91–97

    PubMed  Google Scholar 

  128. Safe S, Wang F, Porter W, Duan R, McDougal A (1998) Ah receptor agonists as endocrine disruptors: antiestrogenic activity and mechanisms. Toxicol Lett 102–103:343–347

    PubMed  Google Scholar 

  129. Gu YZ, Hogenesch JB, Bradfield CA (2000) The PAS superfamily: sensors of environmental and developmental signals. Annu Rev Pharmacol Toxicol 40:519–561

    PubMed  Google Scholar 

  130. Reyes H, Reisz-Porszasz S, Hankinson O (1992) Identification of the Ah receptor nuclear translocator protein (Arnt) as a component of the DNA binding form of the Ah receptor. Science 256:1193–1195

    PubMed  Google Scholar 

  131. Lahvis GP, Bradfield CA (1998) Ahr null alleles: distinctive or different? Biochem Pharmacol 56:781–787

    PubMed  Google Scholar 

  132. Baba T, Mimura J, Nakamura N, Harada N, Yamamoto M, Morohashi K, Fujii-Kuriyama Y (2005) Intrinsic function of the aryl hydrocarbon (dioxin) receptor as a key factor in female reproduction. Mol Cell Biol 25:10040–10051

    PubMed  Google Scholar 

  133. Kociba RJ, Keyes DG, Beyer JE, Carreon RM, Wade CE, Dittenber DA, Kalnins RP, Frauson LE, Park CN, Barnard SD, Hummel RA, Humiston CG (1978) Results of a two-year chronic toxicity and oncogenicity study of 2,3,7,8-tetrachlorodibenzo-p-dioxin in rats. Toxicol Appl Pharmacol 46:279–303

    PubMed  Google Scholar 

  134. Holcomb M, Safe S (1994) Inhibition of 7,12-dimethylbenzanthracene-induced rat mammary tumor growth by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Cancer Lett 82:43–47

    PubMed  Google Scholar 

  135. Wihlén B, Ahmed S, Inzunza J, Matthews J (2009) Estrogen receptor subtype- and promoter-specific modulation of aryl hydrocarbon receptor-dependent transcription. Mol Cancer Res 7:977–986

    PubMed  Google Scholar 

  136. Safe S, Wormke M (2003) Inhibitory aryl hydrocarbon receptor-estrogen receptor alpha cross-talk and mechanisms of action. Chem Res Toxicol 16:807–816

    PubMed  Google Scholar 

  137. Astroff B, Safe S (1988) Comparative antiestrogenic activities of 2,3,7,8-tetrachlorodibenzo-p-dioxin and 6-methyl-1,3,8-trichlorodibenzofuran in the female rat. Toxicol Appl Pharmacol 95:435–443

    PubMed  Google Scholar 

  138. Astroff B, Safe S (1990) 2,3,7,8-Tetrachlorodibenzo-p-dioxin as an antiestrogen: effect on rat uterine peroxidase activity. Biochem Pharmacol 39:485–488

    PubMed  Google Scholar 

  139. Zacharewski T, Harris M, Safe S (1991) Evidence for the mechanism of action of the 2,3,7,8- tetrachlorodibenzo-p-dioxin-mediated decrease of nuclear estrogen receptor levels in wild-type and mutant mouse Hepa 1c1c7 cells. Biochem Pharmacol 41:1931–1939

    PubMed  Google Scholar 

  140. White TE, Gasiewicz TA (1993) The human estrogen receptor structural gene contains a DNA sequence that binds activated mouse and human Ah receptors: a possible mechanism of estrogen receptor regulation by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Biochem Biophys Res Commun 193:956–962

    PubMed  Google Scholar 

  141. Kang KS, Wilson MR, Hayashi T, Chang CC, Trosko JE (1996) Inhibition of gap junctional intercellular communication in normal human breast epithelial cells after treatment with pesticides, PCBs, and PBBs, alone or in mixtures. Environ Health Perspect 104:192–200

    PubMed  Google Scholar 

  142. Kharat I, Saatcioglu F (1996) Antiestrogenic effects of 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin are mediated by direct transcriptional interference with the liganded estrogen receptor. Cross-talk between aryl hydrocarbon- and estrogen-mediated signalling. J Biol Chem 271:10533–10537

    PubMed  Google Scholar 

  143. Jeon MS, Esser C (2000) The murine IL-2 promoter contains distal regulatory elements responsive to the Ah receptor, a member of the evolutionarily conserved bHLH-PAS transcription factor family. J Immunol 165:6975–6983

    PubMed  Google Scholar 

  144. Silverston AE, Frazier DE Jr, Fiore NC, Soults JA, Gasiewicz TA (1994) Dexamethasone, beta-estradiol and 2,3,7,8-tetrachlorodibenzo-p-dioxin elicit thymic atrophy through different cellular targets. Toxicol Appl Pharmacol 126:248–259

    Google Scholar 

  145. Kincade PW, Medina KL, Smithson G (1994) Sex hormones as negative regulators of lymphopoiesis. Immunol Rev 137:119–134

    PubMed  Google Scholar 

  146. McMurray RW, Suwannaroj S, Ndebele K, Jenkins JK (2001) Differential effects of sex steroids on T and B lymphocytes: modulation of cell cycling, apoptosis and bcl-2. Pathobiology 69:44–58

    PubMed  Google Scholar 

  147. Nebert DW, Russell DW (2002) Clinical importance of the cytochromes P450. Lancet 360:1155–1162

    PubMed  Google Scholar 

  148. Kawajiri K, Fujii-Kuriyama Y (2007) Cytochrome P450 gene regulation and physiological functions mediated by the arylhydrocarbon receptor. Arch Biochem Biophys 464:207–212

    PubMed  Google Scholar 

  149. Yueh MF, Huang YH, Hiller A, Chen S, Nguyen N, Tukey RH (2003) Involvement of the xenobiotic response element (XRE) in Ah receptor-mediated induction of human UDP-glucuronosyltransferase1A1. J Biol Chem 278:15001–15006

    PubMed  Google Scholar 

  150. Dere E, Boverhof DR, Burgoon LD, Zacharewski TR (2006) In vivo–in vitro toxicogenomic comparison of TCDD-elicited gene expression in Hepa1c1c7 mouse hepatoma cells and C57BL/6 hepatic tissue. BMC Genomics 7:80–97

    PubMed  Google Scholar 

  151. Daujat M, Clair P, Astier C, Fabre I, Pineau T, Yerle M, Gellin J, Maurel P (1991) Induction, regulation and messenger half-life of cytochromes P450 IA1, IA2 and IIIA6 in primary cultures of rabbit hepatocytes. CYP1A1, 1A2 and 3A6 chromosome location in the rabbit and evidence that post-transcriptional control of gene IA2 does not involve mRNA stabilization. Eur J Biochem 200:501–510

    PubMed  Google Scholar 

  152. Nebert DW (1989) The Ah locus: genetic differences in toxicity, cancer, mutation, and birth defects. Crit Rev Toxicol 20:153–174

    PubMed  Google Scholar 

  153. Whitlock JP Jr (1999) Induction of cytochrome P4501A1. Annu Rev Pharmacol Toxicol 39:103–125

    PubMed  Google Scholar 

  154. Ambrosone CB, Freudenheim JL, Graham S, Marshall JR, Vena JE, Brasure JR, Laughlin R, Nemoto T, Michalek AM, Harrington A et al (1995) Cytochrome P4501A1 and glutathione S-transferase (M1) genetic polymorphisms and postmenopausal breast cancer risk. Cancer Res 55:3483–3485

    PubMed  Google Scholar 

  155. Johnson MR, Carter G, Grint C, Lightman SL (1993) Relationship between ovarian steroids, gonadotrophins and relaxin during the menstrual cycle. Acta Endocrinol (Copenh) 129:121–125

    Google Scholar 

  156. Chen M, Hsu I, Wolfe A, Radovick S, Huang KH, Yu SQ, Chang CS, Messing EM, Yeh SY (2009) Defects of prostate development and reproductive system in the estrogen receptor-alpha null male mice. Endocrinology 150:251–259

    PubMed  Google Scholar 

  157. Hess RA, Bunick D, Bahr J (2001) Oestrogen, its receptors and function in the male reproductive tract – a review. Mol Cell Endocrinol 178:29–38

    PubMed  Google Scholar 

  158. McCarthy MM, Schwarz JM, Wright CL, Dean SL (2008) Mechanisms mediating oestradiol modulation of the developing brain. J Neuroendocrinol 20:777–783

    PubMed  Google Scholar 

  159. McCarthy MM (2008) Estradiol and the developing brain. Physiol Rev 88:91–124

    PubMed  Google Scholar 

  160. Cooke PS, Naaz A (2004) Role of estrogens in adipocyte development and function. Exp Biol Med 229:1127–1135

    Google Scholar 

  161. Törnwall J, Carey AB, Fox RI, Fox HS (1999) Estrogen in autoimmunity: expression of estrogen receptors in thymic and autoimmune T cells. J Gend Specif Med 2:33–40

    PubMed  Google Scholar 

  162. Vandenput L, Ohlsson C (2009) Estrogens as regulators of bone health in men. Nat Rev Endocrinol 5:437–443

    PubMed  Google Scholar 

  163. Verthelyi D (2001) Sex hormones as immunomodulators in health and disease. Int Immunopharmacol 1:983–993

    PubMed  Google Scholar 

  164. Kristiansson P, Wang JX (2001) Reproductive hormones and blood pressure during pregnancy. Hum Reprod 161:13–17

    Google Scholar 

  165. Niino M, Hirotani M, Fukazawa T, Kikuchi S, Sasaki H (2009) Estrogens as potential therapeutic agents in multiple sclerosis. Cent Nerv Syst Agents Med Chem 9:87–94

    PubMed  Google Scholar 

  166. Miller IN, Cronin-Golomb A (2010) Gender differences in Parkinson’s disease: clinical characteristics and cognition. Mov Disord 25:2695–2703

    PubMed  Google Scholar 

  167. Hogan AM, Collins D, Baird AW, Winter DC (2009) Estrogen and gastrointestinal malignancy. Mol Cell Endocrinol 307:19–24

    PubMed  Google Scholar 

  168. Harvey PW, Rush KC, Kockburn A (1999) Endocrine hormonal toxicology: an integrated mechanistic and target systems approach. In: Harvey PW, Rush KC, Cockburn A (eds) Endocrine and hormonal toxicology. Wiley, Chichester, pp 3–11

    Google Scholar 

  169. Shore LS, Cuneah O, Shemesh M (2010) Effects of environmental estrogens on animals in Israel: implications for effects on humans. Rev Environ Health 25:337–343

    PubMed  Google Scholar 

  170. Lemos MF, van Gestel CA, Soares AM (2010) Developmental toxicity of endocrine disrupters bisphenol A and vinclozolin in a terrestrial isopod. Arch Environ Contam Toxicol 59:274–281

    PubMed  Google Scholar 

  171. Fisher JS (2004) Are all EDC effects mediated via steroid hormone receptors? Toxicology 205:33–41

    PubMed  Google Scholar 

  172. Zheng SJ, Tian HJ, Cao J, Gao YQ (2010) Exposure to di(n-butyl)phthalate and benzo(a)pyrene alters IL-1β secretion and subset expression of testicular macrophages, resulting in decreased testosterone production in rats. Toxicol Appl Pharmacol 248:28–37

    PubMed  Google Scholar 

  173. Homberger E, Reggiani G, Sambeth J, Wipf HK (1979) The Seveso accident: its nature, extent and consequences. Ann Occup Hyg 22:327–367

    PubMed  Google Scholar 

  174. Johnson MH, Everitt BJ (2004) Essential reproduction, 5th edn. Blackwell Publishing, Williston

    Google Scholar 

  175. Knobil E, Neill JD (2006) The physiology of reproduction. Elsevier Amsterdam, Boston

    Google Scholar 

  176. Herbison AE (1998) Multimodal influence of estrogen upon gonadotropin-releasing hormone neurons. Endocr Rev 19:302–330

    PubMed  Google Scholar 

  177. Herbison AE, Pape JR (2001) New evidence for estrogen receptors in gonadotropin-releasing hormone neurons. Front Neuroendocrinol 22:292–308

    PubMed  Google Scholar 

  178. Gu Q, Korach KS, Moss RL (1999) Rapid action of 17b-estradiol on kainate-induced currents in hippocampal neurons lacking intracellular estrogen receptors. Endocrinology 140:660–666

    PubMed  Google Scholar 

  179. Abraham IM, Todman MG, Korach KS, Herbison AE (2004) Critical in vivo roles for classical estrogen receptors in rapid estrogen actions on intracellular signalling in mouse brain. Endocrinology 145:3055–3061

    PubMed  Google Scholar 

  180. Rasier G, Parent AS, Gérard A, Denooz R, Lebrethon MC, Charlier C, Bourguignon JP (2008) Mechanisms of interaction of endocrine-disrupting chemicals with glutamate-evoked secretion of gonadotropin-releasing hormone. Toxicol Sci 102:33–41

    PubMed  Google Scholar 

  181. Lee PA, Guo SS, Kulin HE (2001) Age of puberty: data from the United States of America. APMIS 109:81–88

    PubMed  Google Scholar 

  182. Buck Louis GM, Gray LE Jr, Marcus M, Ojeda SR, Pescovitz OH, Witchel SF (2008) Environmental factors and puberty timing: expert panel research needs. Pediatrics 121(Suppl 3):S192–S207

    PubMed  Google Scholar 

  183. Rasier G, Toppari J, Parent AS, Bourguignon JP (2006) Female sexual maturation and reproduction after prepubertal exposure to estrogens and endocrine disrupting chemicals: a review of rodent and human data. Mol Cell Endocrinol 254–255:187–201

    PubMed  Google Scholar 

  184. Gerhard I, Frick A, Monga B, Runnebaum B (1999) Pentachlorophenol exposure in women with gynecological and endocrine dysfunction. Environ Res 80:383–388

    PubMed  Google Scholar 

  185. Mendola P, Buck GM, Sever LE, Zielezny M, Vena JE (1997) Consumption of PCB-contaminated freshwater fish and shortened menstrual cycle length. Am J Epidemiol 146:955–960

    PubMed  Google Scholar 

  186. Windham GC, Lee D, Mitchell P, Anderson M, Petreas M, Lasley B (2005) Exposure to organochlorine compounds and effects on ovarian function. Epidemiology 16:182–190

    PubMed  Google Scholar 

  187. Cummings AM, Hedge JM, Birnbaum LS (1999) Effect of prenatal exposure to TCDD on the promotion of endometriotic lesion growth by TCDD in adult female rats and mice. Toxicol Sci 52:45–49

    PubMed  Google Scholar 

  188. Johnson KL, Cummings AM, Birnbaum LS (1997) Promotion of endometriosis in mice by polychlorinated dibenzo-p-dioxins, dibenzofurans, and biphenyls. Environ Health Perspect 105:750–755

    PubMed  Google Scholar 

  189. Rier SE, Martin DC, Bowman RE, Dmowski WP, Becker JL (1993) Endometriosis in rhesus monkeys (Macaca mulatta) following chronic exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Fundam Appl Toxicol 21:433–441

    PubMed  Google Scholar 

  190. Lang DS, Becker S, Devlin RB, Koren HS (1998) Cell-specific differences in the susceptibility of potential cellular targets of human origin derived from blood and lung following treatment with 2,3,7,8- tetrachlorodibenzo-p-dioxin (TCDD). Cell Biol Toxicol 14:23–38

    PubMed  Google Scholar 

  191. Mayani A, Barel S, Soback S, Almagor M (1997) Dioxin concentrations in women with endometriosis. Hum Reprod 12:373–375

    PubMed  Google Scholar 

  192. Tsukino H, Hanaoka T, Sasaki H, Motoyama H, Hiroshima M, Tanaka T, Kabuto M, Niskar AS, Rubin C, Patterson DG Jr, Turner W, Needham L, Tsugane S (2005) Associations between serum levels of selected organochlorine compounds and endometriosis in infertile Japanese women. Environ Res 99:118–125

    PubMed  Google Scholar 

  193. Knox SS, Jackson T, Javins B, Frisbee SJ, Shankar A, Ducatman AM (2011) Implications of early menopause in women exposed to perfluorocarbons. J Clin Endocrinol Metab 96:1747–1753

    PubMed  Google Scholar 

  194. Bermudez DS, Gray LE Jr, Wilson VS (2010) Modeling the interaction of binary and ternary mixtures of estradiol with bisphenol A and bisphenol AF in an in vitro estrogen-mediated transcriptional activation assay (T47D-KBluc). Toxicol Sci 116:477–487

    PubMed  Google Scholar 

  195. Crain DA, Guillette LJ Jr, Rooney AA, Pickford DB (1997) Alterations in steroidogenesis in alligators (Alligator mississippiensis) exposed naturally and experimentally to environmental contaminants. Environ Health Perspect 105:528–533

    PubMed  Google Scholar 

  196. Hayes TB, Case P, Chui S, Chung D, Haeffele C, Haston K, Lee M, Mai VP, Marjuoa Y, Parker J, Tsui M (2006) Pesticide mixtures, endocrine disruption, and amphibian declines: are we underestimating the impact? Environ Health Perspect 114(Suppl 1):40–50

    PubMed  Google Scholar 

  197. Axmon A, Rylander L, Strömberg U, Hagmar L (2000) Miscarriages and stillbirths in women with a high intake of fish contaminated with persistent organochlorine compounds. Int Arch Occup Environ Health 73:204–208

    PubMed  Google Scholar 

  198. De Celis R, Pedrón-Nuevo N, Feria-Velasco A (1996) Toxicology of male reproduction in animals and humans. Arch Androl 37:201–218

    PubMed  Google Scholar 

  199. Jarrell J, Gocmen A, Foster W, Brant R, Chan S, Sevcik M (1998) Evaluation of reproductive outcomes in women inadvertently exposed to hexachlorobenzene in southeastern Turkey in the 1950s. Reprod Toxicol 12:469–476

    PubMed  Google Scholar 

  200. Pathak R, Mustafa M, Ahmed RS, Tripathi AK, Guleria K, Banerjee BD (2010) Association between recurrent miscarriages and organochlorine pesticide levels. Clin Biochem 43:131–135

    PubMed  Google Scholar 

  201. Willis WO, de Peyster A, Molgaard CA, Walker C, MacKendrick T (1993) Pregnancy outcome among women exposed to pesticides through work or residence in an agricultural area. J Occup Med 35:943–949

    PubMed  Google Scholar 

  202. Harris CA, Hamilton PB, Runnalls TJ, Vinciotti V, Henshaw A, Hodgson D, Coe TS, Jobling S, Tyler CR, Sumpter JP (2011) The consequences of feminization in breeding groups of wild fish. Environ Health Perspect 119:306–311

    PubMed  Google Scholar 

  203. Davis DL, Gottlieb MB, Stampnitzky JR (1998) Reduced ratio of male to female births in several industrial countries: a sentinel health indicator? JAMA 279:1018–1023

    PubMed  Google Scholar 

  204. Mocarelli P, Gerthoux PM, Ferrari E, Patterson DG Jr, Kieszak SM, Brambilla P, Vincoli N, Signorini S et al (2000) Paternal concentrations of dioxin and sex ratio of offspring. Lancet 35:1858–1863

    Google Scholar 

  205. Taylor HS (2008) Endocrine disruptors affect developmental programming of HOX gene expression. Fertil Steril 89(2 Suppl):e57–e58

    PubMed  Google Scholar 

  206. Fei X, Chung H, Taylor HS (2005) Methoxychlor disrupts uterine Hoxa10 gene expression. Endocrinology 146:3445–3451

    PubMed  Google Scholar 

  207. Filigheddu N, Sampietro S, Chianale F, Porporato PE, Gaggianesi M, Gregnanin I, Rainero E, Ferrara M, Perego B, Riboni F, Baldanzi G, Graziani A, Surico N (2011) Diacylglycerol kinase α mediates 17-β-estradiol-induced proliferation, motility, and anchorage-independent growth of Hec-1A endometrial cancer cell line through the G protein-coupled estrogen receptor GPR30. Cell Signal 23:1988–1996

    PubMed  Google Scholar 

  208. Cavalieri EL, Stack DE, Devanesan PD, Todorovic R, Dwivedy I, Higginbotham S, Johansson SL, Patil KD, Gross ML, Gooden JK, Ramanathan R, Cerny RL, Rogan EG (1997) Molecular origin of cancer: catechol estrogen-3,4-quinones as endogenous tumor initiators. Proc Natl Acad Sci 94:10937–10942

    PubMed  Google Scholar 

  209. O’Donnell L, Robertson KM, Jones ME, Simpson ER (2001) Estrogen and spermatogenesis. Endocr Rev 22:289–318

    PubMed  Google Scholar 

  210. Shapiro E, Huang H, Masch RJ, McFadden DE, Wu XR, Ostrer H (2005) Immunolocalization of androgen receptor and estrogen receptors alpha and beta in human fetal testis and epididymis. J Urol 174:1695–1698

    PubMed  Google Scholar 

  211. Taylor RA, Cowin P, Couse JF, Korach KS, Risbridger GP (2006) 17beta-estradiol induces apoptosis in the developing rodent prostate independently of ERalpha or ERbeta. Endocrinology 147:191–200

    PubMed  Google Scholar 

  212. Delbès G, Levacher C, Habert R (2006) Estrogen effects on fetal and neonatal testicular development. Reproduction 132:527–538

    PubMed  Google Scholar 

  213. Guillette LJ Jr, Gross TS, Masson GR, Matter JM, Percival HF, Woodward AR (1994) Developmental abnormalities of the gonad and abnormal sex hormone concentrations in juvenile alligators from contaminated and control lakes in Florida. Environ Health Perspect 102:680–688

    PubMed  Google Scholar 

  214. Kholkute SD, Rodriguez J, Dukelow WR (1994) Reproductive toxicity of Aroclor-1254: effects on oocyte, spermatozoa, in vitro fertilization, and embryo development in the mouse. Reprod Toxicol 8:487–493

    PubMed  Google Scholar 

  215. Silvestroni L, Palleschi S (1999) Effects of organochlorine xenobiotics on human spermatozoa. Chemosphere 39:1249–1252

    PubMed  Google Scholar 

  216. Barber LB, Lee KE, Swackhamer DL, Schoenfuss HL (2007) Reproductive responses of male fat head minnows exposed to wastewater treatment plant effluent, effluent treated with XAD8 resin, and an environmentally relevant mixture of alkylphenol compounds. Aquat Toxicol 82:36–46

    PubMed  Google Scholar 

  217. McLachlan JA (1977) Prenatal exposure to diethylstilbestrol in mice: toxicological studies. J Toxicol Environ Health 2:527–537

    PubMed  Google Scholar 

  218. McLachlan JA, Newbold RR, Burow ME, Li SF (2001) From malformations to molecular mechanisms in the male: three decades of research on endocrine disrupters. APMIS 109:263–272

    PubMed  Google Scholar 

  219. Penny R (1982) Pediatrics-epitomes of progress: the effect of des on male offspring. West J Med 136:329–330

    PubMed  Google Scholar 

  220. Adamopoulos DA, Pappa A, Nicopoulou S, Andreou E, Karamertzanis M, Michopoulos J, Deligianni V, Simou M (1996) Seminal volume and total sperm number trends in men attending subfertility clinics in the greater Athens area during the period 1977–1993. Hum Reprod 11:1936–1941

    PubMed  Google Scholar 

  221. Sultan C, Balaguer P, Terouanne B, Georget V, Paris F, Jeandel C, Lumbroso S, Nicolas J (2001) Environmental xenoestrogens, antiandrogens and disorders of male sexual differentiation. Mol Cell Endocrinol 178:99–105

    PubMed  Google Scholar 

  222. Luconi M, Bonaccorsi L, Forti G, Baldi E (2001) Effects of estrogenic compounds on human spermatozoa: evidence for interaction with a nongenomic receptor for estrogen on human sperm membrane. Mol Cell Endocrinol 178:39–45

    PubMed  Google Scholar 

  223. Comhaire FH, Mahmoud AM, Schoonjans F (2007) Sperm quality, birth rates and the environment in Flanders (Belgium). Reprod Toxicol 23:133–137

    PubMed  Google Scholar 

  224. Irvine S, Cawood E, Richardson D, MacDonald E, Aitken J (1996) Evidence of deteriorating semen quality in the United Kingdom: birth cohort study in 577 men in Scotland over 11 years. BMJ 312:467–471

    PubMed  Google Scholar 

  225. Slutsky M, Levin JL, Levy BS (1999) Azoospermia and oligospermia among a large cohort of DBCP applicators in 12 countries. Int J Occup Environ Health 5:116–122

    PubMed  Google Scholar 

  226. Egeland GM, Sweeney MH, Fingerhut MA, Wille KK, Schnorr TM, Halperin WE (1994) Total serum testosterone and gonadotropins in workers exposed to dioxin. Am J Epidemiol 139:272–281

    PubMed  Google Scholar 

  227. Giwercman AH, Rignell-Hydbom A, Toft G, Rylander L, Hagmar L, Lindh C, Pedersen HS, Ludwicki JK, Lesovoy V, Shvets M, Spano M, Manicardi GC, Bizzaro D, Bonefeld-Jorgensen EC, Bonde JP (2006) Reproductive hormone levels in men exposed to persistent organohalogen pollutants: a study of inuit and three European cohorts. Environ Health Perspect 114:1348–1353

    PubMed  Google Scholar 

  228. Swan SH, Brazil C, Drobnis EZ, Liu F, Kruse RL, Hatch M, Redmon JB, Wang C, Overstreet JW, Study for Future Families Research Group (2003) Geographic differences in semen quality of fertile U.S. males. Environ Health Perspect 111:414–420

    PubMed  Google Scholar 

  229. Kristensen P, Irgens LM, Andersen A, Bye AS, Sundheim L (1997) Birth defects among offspring of Norwegian farmers, 1967–1991. Epidemiology 8:537–544

    PubMed  Google Scholar 

  230. Weidner IS, Møller H, Jensen TK, Skakkebaek NE (1998) Cryptorchidism and hypospadias in sons of gardeners and farmers. Environ Health Perspect 106:793–796

    PubMed  Google Scholar 

  231. Adami HO, Bergström R, Möhner M, Zatoński W, Storm H, Ekbom A, Tretli S, Teppo L, Ziegler H, Rahu M et al (1994) Testicular cancer in nine northern European countries. Int J Cancer 59:33–38

    PubMed  Google Scholar 

  232. Kalfa N, Philibert P, Baskin LS, Sultan C (2011) Hypospadias: interactions between environment and genetics. Mol Cell Endocrinol 335:89–95

    PubMed  Google Scholar 

  233. Huyghe E, Matsuda T, Thonneau P (2003) Increasing incidence of testicular cancer worldwide: a review. J Urol 170:5–11

    PubMed  Google Scholar 

  234. Sharpe RM, Skakkebaek NE (1993) Are oestrogens involved in falling sperm counts and disorders of the male reproductive tract? Lancet 2341:1392–1395

    Google Scholar 

  235. Skakkebæk NE, Rajpert-De Meyts E, Malm KE (2001) Testicular dysgenesis syndrome: an increasingly common developmental disorder with environmental aspects. Hum Reprod 16:972–978

    PubMed  Google Scholar 

  236. McGlynn KA, Quraishi SM, Graubard BI, Weber JP, Rubertone MV, Erickson RL (2008) Persistent organochlorine pesticides and risk of testicular germ cell tumors. J Natl Cancer Inst 100:663–671

    PubMed  Google Scholar 

  237. Moline JM, Golden AL, Bar-Chama N, Smith E, Rauch ME, Chapin RE, Perreault SD, Schrader SM, Suk WA, Landrigan PJ (2000) Exposure to hazardous substances and male reproductive health: a research framework. Environ Health Perspect 108:803–813

    PubMed  Google Scholar 

  238. Witorsch RJ (2002) Low-dose in utero effects of xenoestrogens in mice and their relevance to humans: an analytical review of the literature. Food Chem Toxicol 407:905–912

    Google Scholar 

  239. Giwercman A, Giwercman YL (2011) Environmental factors and testicular function. Best Pract Res Clin Endocrinol Metab 25:391–402

    PubMed  Google Scholar 

  240. Hardell L, van Bavel B, Lindström G, Carlberg M, Dreifaldt AC, Wijkström H, Starkhammar H, Eriksson M, Hallquist A, Kolmert T (2003) Increased concentrations of polychlorinated biphenyls, hexachlorobenzene, and chlordanes in mothers of men with testicular cancer. Environ Health Perspect 11:930–934

    Google Scholar 

  241. Hontz AE, Li SA, Lingle WL, Negron V, Bruzek A, Salisbury JL, Li JJ (2007) Aurora A and B overexpression and centrosome amplification in early estrogen-induced tumor foci in the Syrian hamster kidney: implications for chromosomal instability, aneuploidy, and neoplasia. Cancer Res 67:2957–2963

    PubMed  Google Scholar 

  242. Cavalieri E, Frenkel K, Liehr JG, Rogan E, Roy D (2000) Estrogens as endogenous genotoxic agents – DNA adducts and mutations. J Natl Cancer Inst Monogr 27:75–93

    PubMed  Google Scholar 

  243. Quick EL, Parry EM, Parry JM (2008) Do oestrogens induce chromosome specific aneuploidy in vitro, similar to the pattern of aneuploidy seen in breast cancer? Mutat Res 651:46–55

    PubMed  Google Scholar 

  244. Jobling S, Reynolds T, White R, Parker MG, Sumpter JP (1995) A variety of environmentally persistent chemicals, including some phthalate plasticizers, are weakly estrogenic. Environ Health Perspect 103:582–587

    PubMed  Google Scholar 

  245. Goto N, Hiyoshi H, Ito I, Tsuchiya M, Nakajima Y, Yanagisawa J (2011) Estrogen and antiestrogens alter breast cancer invasiveness by modulating the transforming growth factor-β signaling pathway. Cancer Sci 102:1501–1508

    PubMed  Google Scholar 

  246. Goodman A, Schorge J, Greene MF (2011) The long-term effects of in utero exposures–the DES story. N Engl J Med 36422:2083–2084

    Google Scholar 

  247. Beral V (2003) Breast cancer and hormone-replacement therapy in the Million Women Study. Lancet 362:419–427

    PubMed  Google Scholar 

  248. Bernstein L, Ross RK, Pike MC, Brown JB, Henderson BE (1990) Hormone levels in older women: a study of post-menopausal breast cancer patients and healthy population controls. Br J Cancer 61:298–302

    PubMed  Google Scholar 

  249. Henderson BE, Feigelson HS (2000) Hormonal carcinogenesis. Carcinogenesis 21:427–433

    PubMed  Google Scholar 

  250. Li CI, Malone KE, Porter PL, Weiss NS, Tang MT, Cushing-Haugen KL, Daling JR (2003) Relationship between long durations and different regimens of hormone therapy and risk of breast cancer. JAMA 289:3254–3263

    PubMed  Google Scholar 

  251. Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, Jackson RD, Beresford SA, Howard BV, Johnson KC, Kotchen JM, Ockene J, Writing Group for the Women’s Health Initiative Investigators (2002) Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women’s Health Initiative randomized controlled trial. JAMA 288:321–333

    PubMed  Google Scholar 

  252. Fussell KC, Udasin RG, Smith PJ, Gallo MA, Laskin JD (2011) Catechol metabolites of endogenous estrogens induce redox cycling and generate reactive oxygen species in breast epithelial cells. Carcinogenesis 32:1285–1293

    PubMed  Google Scholar 

  253. Bocchinfuso WP, Korach KS (1997) Mammary gland development and tumorigenesis in estrogen receptor knockout mice. J Mammary Gland Biol Neoplasia 2:323–334

    PubMed  Google Scholar 

  254. Deroo BJ, Korach KS (2006) Exposure to environmental chemicals that are able to disrupt the hormonal equilibrium might represent another risk factor for this disease: estrogen receptors and human disease. J Clin Invest 116:561–570

    PubMed  Google Scholar 

  255. Yaich L, Dupont WD, Cavener DR, Parl FF (1992) Analysis of the PvuII restriction fragment-length polymorphism and exon structure of the estrogen receptor gene in breast cancer and peripheral blood. Cancer Res 52:77–83

    PubMed  Google Scholar 

  256. Levitz M, Banerjee S, Raju U, Toniolo PG, Shore RE, Nachtigall LE (1997) Sex hormone-binding globulin in estrogen-dependent cancer and estrogen replacement therapy. Ann N Y Acad Sci 828:358–365

    PubMed  Google Scholar 

  257. Zeleniuch-Jacquotte A, Shore RE, Koenig KL, Akhmedkhanov A, Afanasyeva Y, Kato I, Kim MY, Rinaldi S, Kaaks R, Toniolo P (2004) Postmenopausal levels of oestrogen, androgen, and SHBG and breast cancer: long-term results of a prospective study. Br J Cancer 90:153–159

    PubMed  Google Scholar 

  258. Henderson AK, Rosen D, Miller GL, Figgs LW, Zahm SH, Sieber SM, Rothman N, Humphrey HE, Sinks T (1995) Breast cancer among women exposed to polybrominated biphenyls. Epidemiology 6:544–546

    PubMed  Google Scholar 

  259. Safe SH (2000) Endocrine disruptors and human health-is there a problem? An update. Environ Health Perspect 108:487–493

    PubMed  Google Scholar 

  260. Kettles MK, Browning SR, Prince TS, Horstman SW (1997) Triazine herbicide exposure and breast cancer incidence: an ecologic study of Kentucky counties. Environ Health Perspect 10511:1222–1227

    Google Scholar 

  261. Wolff MS, Toniolo PG, Lee EW, Rivera M, Dubin N (1993) Blood levels of organochlorine residues and risk of breast cancer. J Natl Cancer Inst 85:648–652

    PubMed  Google Scholar 

  262. Høyer AP, Grandjean P, Jørgensen T, Brock JW, Hartvig HB (1998) Organochlorine exposure and risk of breast cancer. Lancet 352:1816–1820

    PubMed  Google Scholar 

  263. Unger M, Kiaer H, Blichert-Toft M, Olsen J, Clausen J (1984) Organochlorine compounds in human breast fat from deceased with and without breast cancer and in a biopsy material from newly diagnosed patients undergoing breast surgery. Environ Res 34:24–28

    PubMed  Google Scholar 

  264. Bertazzi A, Pesatori AC, Consonni D, Tironi A, Landi MT, Zocchetti C (1993) Cancer incidence in a population accidentally exposed to 2,3,7,8-tetrachlorodibenzo-para-dioxin. Epidemiology 4:398–406

    PubMed  Google Scholar 

  265. La Merrill M, Harper R, Birnbaum LS, Cardiff RD, Threadgill DW (2010) Maternal dioxin exposure combined with a diet high in fat increases mammary cancer incidence in mice. Environ Health Perspect 118:596–601

    PubMed  Google Scholar 

  266. Fenton SE, Hamm JT, Birnbaum LS, Youngblood GL (2002) Persistent abnormalities in the rat mammary gland following gestational and lactational exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Toxicol Sci 67:63–74

    PubMed  Google Scholar 

  267. Høyer AP, Gerdes AM, Jørgensen T, Rank F, Hartvig HB (2002) Organochlorines, p53 mutations in relation to breast cancer risk and survival. A Danish cohort-nested case-controls study. Breast Cancer Res Treat 71:59–65

    PubMed  Google Scholar 

  268. Badawi AF, Cavalieri EL, Rogan EG (2001) Role of human cytochrome P450 1A1, 1A2, 1B1, and 3A4 in the 2-, 4-, and 16alpha-hydroxylation of 17beta-estradiol. Metabolism 50:1001–1003

    PubMed  Google Scholar 

  269. Cavalieri EL, Kumar S, Todorovic R, Higginbotham S, Badawi AF, Rogan EG (2001) Imbalance of estrogen homeostasis in kidney and liver of hamsters treated with estradiol: implications for estrogen-induced initiation of renal tumors. Chem Res Toxicol 14:1041–1050

    PubMed  Google Scholar 

  270. Yager JD (2000) Endogenous estrogens as carcinogens through metabolic activation. J Natl Cancer Inst Monogr 27:67–73

    PubMed  Google Scholar 

  271. Safe S (2000) Molecular biology of the Ah receptor and its role in carcinogenesis. Toxicol Lett 120:1–7

    Google Scholar 

  272. Darbre PD, Charles AK (2010) Environmental oestrogens and breast cancer: evidence for combined involvement of dietary, household and cosmetic xenoestrogens. Anticancer Res 30:815–827

    PubMed  Google Scholar 

  273. Wang Y, Liang H, Tussing L, Braunschweig C, Caballero B, Flay B (2007) Obesity and related risk factors among low socio-economic status minority students in Chicago. Public Health Nutr 10:927–938

    PubMed  Google Scholar 

  274. Cooke PS, Heine PA, Taylor JA, Lubahn DB (2001) The role of estrogen and estrogen receptor-alpha in male adipose tissue. Mol Cell Endocrinol 178:147–154

    PubMed  Google Scholar 

  275. Couse JF, Korach KS (1999) Estrogen receptor null mice: what have we learned and where will they lead us? Endocr Rev 20:358–417

    PubMed  Google Scholar 

  276. Naaz A, Zakroczymski M, Heine P (2002) Effect of ovariectomy on adipose tissue of mice in the absence of estrogen receptor alpha (ERalpha): a potential role for estrogen receptor beta (ERbeta). Horm Metab Res 34:758–763

    PubMed  Google Scholar 

  277. Penza M, Montani C, Romani A, Vignolini P, Pampaloni B, Tanini A, Brandi ML, Alonso-Magdalena P, Nadal A, Ottobrini L, Parolini O, Bignotti E, Calza S, Maggi A, Grigolato PG, Di Lorenzo D (2006) Genistein affects adipose tissue deposition in a dose-dependent and gender-specific manner. Endocrinology 147:5740–5751

    PubMed  Google Scholar 

  278. Liang YQ, Akishita M, Kim S, Ako J, Hashimoto M, Iijima K, Ohike Y, Watanabe T, Sudoh N, Toba K, Yoshizumi M, Ouchi Y (2002) Estrogen receptor beta is involved in the anorectic action of estrogen. Int J Obes Relat Metab Disord 26:1103–1109

    PubMed  Google Scholar 

  279. Musatov S, Chen W, Pfaff DW, Mobbs CV, Yang XJ, Clegg DJ, Kaplitt MG, Ogawa S (2007) Silencing of estrogen receptor alpha in the ventromedial nucleus of hypothalamus leads to metabolic syndrome. Proc Natl Acad Sci U S A 104:2501–2506

    PubMed  Google Scholar 

  280. Newbold RR, Padilla-Banks E, Snyder RJ, Jefferson WN (2007) Perinatal exposure to environmental estrogens and the development of obesity. Mol Nutr Food Res 51:912–917

    PubMed  Google Scholar 

  281. Newbold RR, Padilla-Banks E, Snyder RJ, Phillips TM, Jefferson WN (2007) Developmental exposure to endocrine disruptors and the obesity epidemic. Reprod Toxicol 23:290–296

    PubMed  Google Scholar 

  282. Newbold RR, Jefferson WN, Grissom SF, Padilla-Banks E, Snyder RJ, Lobenhofer EK (2007) Developmental exposure to diethylstilbestrol alters uterine gene expression that may be associated with uterine neoplasia later in life. Mol Carcinog 46:783–796

    PubMed  Google Scholar 

  283. McAllister EJ, Dhurandhar NV, Keith SW, Aronne LJ, Barger J, Baskin M, Benca RM, Biggio J, Boggiano MM, Eisenmann JC, Elobeid M, Fontaine KR, Gluckman P, Hanlon EC, Katzmarzyk P, Pietrobelli A, Redden DT, Ruden DM, Wang C, Waterland RA, Wright SM, Allison DB (2009) Ten putative contributors to the obesity epidemic. Crit Rev Food Sci Nutr 49:868–913

    PubMed  Google Scholar 

  284. Collins S (2005) Overview of clinical perspectives and mechanisms of obesity. Birth Defects Res A Clin Mol Teratol 73:470–471

    PubMed  Google Scholar 

  285. Baillie-Hamilton PF (2002) Chemical toxins: a hypothesis to explain the global obesity epidemic. J Altern Complement Med 8:185–192

    PubMed  Google Scholar 

  286. Heindel JJ (2003) Endocrine disruptors and the obesity epidemic. Toxicol Sci 76:247–249

    PubMed  Google Scholar 

  287. Newbold RR (2010) Impact of environmental endocrine disrupting chemicals on the development of obesity. Hormones (Athens) 93:206–217

    Google Scholar 

  288. Goncharov A, Haase RF, Santiago-Rivera A, Santiago-Rivera A, Morse G, Akwesasne Task Force on the Environment, McCaffrey RJ, Rej R, Carpenter DO (2008) High serum PCBs are associated with elevation of serum lipids and cardiovascular disease in a Native American population. Environ Res 106:226–239

    PubMed  Google Scholar 

  289. Lee DH, Lee IK, Jin SH, Steffes M, Jacobs DR Jr (2007) Association between serum concentrations of persistent organic pollutants and insulin resistance among nondiabetic adults: results from the National Health and Nutrition Examination Survey 1999–2002. Diabetes Care 30:622–628

    PubMed  Google Scholar 

  290. Lee DH, Steffes MW, Jacobs DR Jr (2008) Can persistent organic pollutants explain the association between serum gamma-glutamyltransferase and type 2 diabetes? Diabetologia 51:402–407

    PubMed  Google Scholar 

  291. Pelletier C, Doucet E, Imbeault P, Tremblay A (2002) Associations between weight loss-induced changes in plasma organochlorine concentrations, serum T(3) concentration, and resting metabolic rate. Toxicol Sci 67:46–51

    PubMed  Google Scholar 

  292. Gladen BC, Ragan NB, Rogan WJ (2000) Pubertal growth and development and prenatal and lactational exposure to polychlorinated biphenyls and dichlorodiphenyl dichloroethene. J Pediatr 136:490–496

    PubMed  Google Scholar 

  293. Vasiliu O, Cameron L, Gardiner J, Deguire P, Karmaus W (2006) Polybrominated biphenyls, polychlorinated biphenyls, body weight, and incidence of adult-onset diabetes mellitus. Epidemiology 17:352–359

    PubMed  Google Scholar 

  294. World Health Organization (2005) Welcome to the diabetes program 2005. http://www.who.int/diabetes/en/

  295. Tchernof A, Calles-Escandon J, Sites CK et al (1998) Menopause, central body fatness and insulin resistance: effects of hormone-replacement therapy. Coron Artery Dis 9:503–511

    PubMed  Google Scholar 

  296. Bryzgalova G, Lundholm L, Portwood N, Gustafsson JA, Khan A, Efendic S, Dahlman-Wright K (2008) Mechanisms of antidiabetogenic and body weight-lowering effects of estrogen in high-fat diet-fed mice. Am J Physiol Endocrinol Metab 295:E904–E912

    PubMed  Google Scholar 

  297. Salpeter SR, Walsh JM, Ormiston TM, Greyber E, Buckley NS, Salpeter EE (2006) Meta-analysis: effect of hormone-replacement therapy on components of the metabolic syndrome in postmenopausal women. Diabetes Obes Metab 8:538–554

    PubMed  Google Scholar 

  298. Barros RP, Machado UF, Gustafsson JA (2006) Estrogen receptors: new players in diabetes mellitus. Trends Mol Med 12:425–431

    PubMed  Google Scholar 

  299. Nadal A, Alonso-Magdalena P, Soriano S, Quesada I, Ropero AB (2009) The pancreatic beta-cell as a target of estrogens and xenoestrogens: implications for blood glucose homeostasis and diabetes. Mol Cell Endocrinol 304:63–68

    PubMed  Google Scholar 

  300. Alonso-Magdalena P, Ropero AB, Carrera MP, Cederroth CR, Baquié M, Gauthier BR, Nef S, Stefani E, Nadal A (2008) Pancreatic insulin content regulation by the estrogen receptor ER alpha. PLoS One 3:e2069

    PubMed  Google Scholar 

  301. Le May C, Chu K, Hu M, Ortega CS, Simpson ER, Korach KS, Tsai MJ, Mauvais-Jarvis F (2006) Estrogens protect pancreatic beta-cells from apoptosis and prevent insulin-deficient diabetes mellitus in mice. Proc Natl Acad Sci U S A 103:9232–9237

    PubMed  Google Scholar 

  302. Soriano S, Ropero AB, Alonso-Magdalena P, Ripoll C, Quesada I, Gassner B, Kuhn M, Gustafsson JA, Nadal A (2009) Rapid regulation of K(ATP) channel activity by 17{beta}-estradiol in pancreatic beta}-cells involves the estrogen receptor {beta and the atrial natriuretic peptide receptor. Mol Endocrinol 23:1973–1982

    PubMed  Google Scholar 

  303. Barros RP, Machado UF, Warner M, Gustafsson JA (2006) Muscle GLUT4 regulation by estrogen receptors ERbeta and ERalpha. Proc Natl Acad Sci U S A 103:1605–1608

    PubMed  Google Scholar 

  304. Lang IA, Galloway TS, Scarlett A, Henley WE, Depledge M, Wallace RB, Melzer D (2008) Association of urinary bisphenol A concentration with medical disorders and laboratory abnormalities in adults. JAMA 300:1303–1310

    PubMed  Google Scholar 

  305. vom Saal FS, Myers JP (2008) Bisphenol A and risk of metabolic disorders. JAMA 300:1353–1355

    Google Scholar 

  306. Lee DH, Lee IK, Porta M, Steffes M, Jacobs DR Jr (2007) Relationship between serum concentrations of persistent organic pollutants and the prevalence of metabolic syndrome among non-diabetic adults: results from the National Health and Nutrition Examination Survey 1999–2002. Diabetologia 50:1841–1851

    PubMed  Google Scholar 

  307. Lee DH, Lim JS, Song K, Boo Y, Jacobs DR Jr (2006) Graded associations of blood lead and urinary cadmium concentrations with oxidative-stress-related markers in the U.S. population: results from the third National Health and Nutrition Examination Survey. Environ Health Perspect 114:350–354

    PubMed  Google Scholar 

  308. Sakurai K, Kawazuma M, Adachi T, Harigaya T, Saito Y, Hashimoto N, Mori C (2004) Bisphenol A affects glucose transport in mouse 3T3-F442A adipocytes. Br J Pharmacol 141:209–214

    PubMed  Google Scholar 

  309. Masuno H, Iwanami J, Kidani T, Sakayama K, Honda K (2005) Bisphenol A accelerates terminal differentiation of 3T3-L1 cells into adipocytes through the phosphatidylinositol 3-kinase pathway. Toxicol Sci 84:319–327

    PubMed  Google Scholar 

  310. Masuno H, Kidani T, Sekiya K, Sakayama K, Shiosaka T, Yamamoto H, Honda K (2002) Bisphenol A in combination with insulin can accelerate the conversion of 3T3-L1 fibroblasts to adipocytes. J Lipid Res 43:676–684

    PubMed  Google Scholar 

  311. Hugo ER, Brandebourg TD, Woo JG, Loftus J, Alexander JW, Ben-Jonathan N (2008) Bisphenol A at environmentally relevant doses inhibits adiponectin release from human adipose tissue explants and adipocytes. Environ Health Perspect 116:1642–1647

    PubMed  Google Scholar 

  312. Amantea D, Russo R, Bagetta G, Corasaniti MT (2005) From clinical evidence to molecular mechanisms underlying neuroprotection afforded by estrogens. Pharmacol Res 52:119–132

    PubMed  Google Scholar 

  313. Ardelt AA, McCullough LD, Korach KS, Wang MM, Munzenmaier DH, Hurn PD (2005) Estradiol regulates angiopoietin-1 mRNA expression through estrogen receptor-alpha in a rodent experimental stroke model. Stroke 36:337–341

    PubMed  Google Scholar 

  314. Shulman LM (2002) Is there a connection between estrogen and Parkinson’s disease? Parkinsonism Relat Disord 8:289–295

    PubMed  Google Scholar 

  315. Gillies GE, Murray HE, Dexter D, McArthur S (2004) Sex dimorphisms in the neuroprotective effects of estrogen in an animal model of Parkinson’s disease. Pharmacol Biochem Behav 78:513–522

    PubMed  Google Scholar 

  316. Quesada A, Micevych PE (2004) Estrogen interacts with the IGF-1 system to protect nigros-triatal dopamine and maintain motoric behavior after 6-hydroxdopamine lesions. J Neurosci Res 75:107–116

    PubMed  Google Scholar 

  317. Luine VN (1985) Estradiol increases choline acetyltransferase activity in specific basal forebrain nuclei and projection areas of female rats. Exp Neurol 89:484–490

    PubMed  Google Scholar 

  318. Behl C, Widmann M, Trapp T, Holsboer F (1995) 17-beta estradiol protects neurons from oxidative stress-induced cell death in vitro. Biochem Biophys Res Commun 216:473–482

    PubMed  Google Scholar 

  319. Pinkerton JV, Henderson VW (2005) Estrogen and cognition with a focus on Alzheimer’s disease. Semin Reprod Med 23:172–179

    PubMed  Google Scholar 

  320. Morinaga A, Ono K, Takasaki J, Ikeda T, Hirohata M, Yamada M (2011) Effects of sex hormones on Alzheimer’s disease-associated β-amyloid oligomer formation in vitro. Exp Neurol 228:298–302

    PubMed  Google Scholar 

  321. Janicki SC, Schupf N (2010) Hormonal influences on cognition and risk for Alzheimer’s disease. Curr Neurol Neurosci Rep 10:359–366

    PubMed  Google Scholar 

  322. Nicoletti A, Arabia G, Pugliese P, Nicoletti G, Torchia G, Condino F, Morgante L, Quattrone A, Zappia M (2007) Hormonal replacement therapy in women with Parkinson disease and levodopa-induced dyskinesia: a crossover trial. Clin Neuropharmacol 30:276–280

    PubMed  Google Scholar 

  323. Saunders-Pullman R (2003) Estrogens and Parkinson disease: neuroprotective, symptomatic, neither, or both? Endocrine 21:81–87

    PubMed  Google Scholar 

  324. Sherwin BB (2003) Estrogen and cognitive functioning in women. Endocr Rev 24:133–151

    PubMed  Google Scholar 

  325. Lebrun CE, van der Schouw YT, de Jong FH, Pols HA, Grobbee DE, Lamberts SW (2005) Endogenous oestrogens are related to cognition in healthy elderly women. Clin Endocrinol (Oxf) 63:50–55

    Google Scholar 

  326. Schupf N, Pang D, Patel BN, Silverman W, Schubert R, Lai F, Kline JK, Stern Y, Ferin M, Tycko B, Mayeux R (2003) Onset of dementia is associated with age at menopause in women with Down’s syndrome. Ann Neurol 54:433–438

    PubMed  Google Scholar 

  327. Bloch M, Daly RC, Rubinow DR (2003) Endocrine factors in the etiology of postpartum depression. Compr Psychiatry 44:234–246

    PubMed  Google Scholar 

  328. Farage MA, Osborn TW, MacLean AB (2008) Cognitive, sensory, and emotional changes associated with the menstrual cycle: a review. Arch Gynecol Obstet 278:299–307

    PubMed  Google Scholar 

  329. Hampson E (1990) Variations in sex-related cognitive abilities across the menstrual cycle. Brain Cogn 14:26–43

    PubMed  Google Scholar 

  330. Lewinsohn PM, Rohde P, Seeley JR (1998) Major depressive disorder in older adolescents: prevalence, risk factors, and clinical implications. Clin Psychol Rev 18:765–794

    PubMed  Google Scholar 

  331. Steiner M, Dunn E, Born L (2003) Hormones and mood: from menarche to menopause and beyond. J Affect Disord 74:67–83

    PubMed  Google Scholar 

  332. Rubinow DR, Schmidt PJ (2006) Gonadal steroid regulation of mood: the lessons of premenstrual syndrome. Front Neuroendocrinol 27:210–216

    PubMed  Google Scholar 

  333. Dubal DB, Zhu H, Yu J, Rau SW, Shughrue PJ, Merchenthaler I, Kindy MS, Wise PM (2001) Estrogen receptor alpha, not beta, is a critical link in estradiol-mediated protection against brain injury. Proc Natl Acad Sci U S A 98:1952–1957

    PubMed  Google Scholar 

  334. Panzica GC, Viglietti-Panzica C, Mura E, Quinn MJ Jr, Lavoie E, Palanza P, Ottinger MA (2007) Effects of xenoestrogens on the differentiation of behaviorally-relevant neural circuits. Front Neuroendocrinol 28:179–200

    PubMed  Google Scholar 

  335. Shinomiya N, Shinomiya M (2003) Dichlorodiphenyltrichloroethane suppresses neurite outgrowth and induces apoptosis in PC12 pheochromocytoma cells. Toxicol Lett 137:175–183

    PubMed  Google Scholar 

  336. Petersen SL, Krishnan S, Hudgens ED (2006) The aryl hydrocarbon receptor pathway and sexual differentiation of neuroendocrine functions. Endocrinology 147:S33–S42

    PubMed  Google Scholar 

  337. Stump DG, Beck MJ, Radovsky A, Garman RH, Freshwater LL, Sheets LP, Marty MS, Waechter JM Jr, Dimond SS, Van Miller JP, Shiotsuka RN, Beyer D, Chappelle AH, Hentges SG (2010) Developmental neurotoxicity study of dietary bisphenol A in Sprague–Dawley rats. Toxicol Sci 115:167–182

    PubMed  Google Scholar 

  338. Seo BW, Sparks AJ, Medora K, Amin S, Schantz SL (1999) Learning and memory in rats gestationally and lactationally exposed to 2,3,7,8-tetrachlo-rodibenzo-p-dioxin (TCDD). Neurotoxicol Teratol 21:231–239

    PubMed  Google Scholar 

  339. Hojo R, Stern S, Zareba G, Markowski VP, Cox C, Kost JT, Weiss B (2002) Sexually dimorphic behavioral responses to prenatal dioxin exposure. Environ Health Perspect 110:247–254

    PubMed  Google Scholar 

  340. Holene E, Nafstad I, Skaare JU, Krogh H, Sagvolden T (1999) Behavioural effects in female rats of postnatal exposure to sub-toxic doses of polychlorinated biphenyl congener 153. Acta Paediatr Suppl 88:55–63

    PubMed  Google Scholar 

  341. Masuo Y, Morita M, Oka S, Ishido M (2004) Motor hyperactivity caused by a deficit in dopaminergic neurons and the effects of endocrine disruptors: a study inspired by the physiological roles of PACAP in the brain. Regul Pept 123:225–234

    PubMed  Google Scholar 

  342. Johnson BL, DeRosa CT (1995) Chemical mixtures released from hazardous waste sites: implications for health risk assessment. Toxicology 105:145–156

    PubMed  Google Scholar 

  343. Guo YL, Lambert GH, Hsu CC, Hsu MML (2004) Yucheng: health effects of prenatal exposure to polychlorinated biphenyls and dibenzofurans. Int Arc Occup Environ Health 77:153–158

    Google Scholar 

  344. Jacobson SW, Fein GG, Jacobson JL, Schwartz PM, Dowler JK (1985) The effect of intrauterine PCB exposure on visual recognition memory. Child Dev 564:853–860

    Google Scholar 

  345. Patandin S, Lanting CI, Mulder PG, Boersma ER, Sauer PJ, Weisglas-Kuperus N (1999) Effects of environmental exposure to polychlorinated biphenyls and dioxins on cognitive abilities in Dutch children at 42 months of age. J Pediatr 1341:33–41

    Google Scholar 

  346. Winneke G, Bucholski A, Heinzow B, Krämer U, Schmidt E, Walkowiak J, Wiener JA, Steingrüber HJ (1998) Developmental neurotoxicity of polychlorinated biphenyls (PCBs): cognitive and psychomotor functions in 7-month old children. Toxicol Lett 102–103:423–428

    PubMed  Google Scholar 

  347. Boucher O, Muckle G, Bastien CH (2009) Prenatal exposure to polychlorinated biphenyls: a neuropsychologic analysis. Environ Health Perspect 1171:7–16

    Google Scholar 

  348. Schantz SL, Widholm JJ, Rice DC (2003) Effects of PCB exposure on neuropsychological function in children. Environ Health Perspect 111:357–376

    PubMed  Google Scholar 

  349. Cicchetti DV, Kaufman AS, Sparrow SS (2004) The relationship between prenatal and postnatal exposure to polychlorinated biphenyls (PCBs) and cognitive, neuropsychological and behavioral deficits: a critical appraisal. Psychol Sch 41:589–624

    Google Scholar 

  350. Ross G (2004) The public health implications of polychlorinated biphenyls (PCBs) in the environment. Ecotoxicol Environ Saf 59:275–291

    PubMed  Google Scholar 

  351. Tanida T, Warita K, Ishihara K, Fukui S, Mitsuhashi T, Sugawara T, Tabuchi Y, Nanmori T, Qi WM, Inamoto T, Yokoyama T, Kitagawa H, Hoshi N (2009) Fetal and neonatal exposure to three typical environmental chemicals with different mechanisms of action: mixed exposure to phenol, phthalate, and dioxin cancels the effects of sole exposure on mouse midbrain dopaminergic nuclei. Toxicol Lett 189:40–47

    PubMed  Google Scholar 

  352. Schuurs AH, Verheul HAJ (1990) Effects of gender and sex steroids on the immune response. J Steroid Biochem 35:157–172

    PubMed  Google Scholar 

  353. Tsokos GC, Kammer GM (2000) Molecular aberrations in human systemic lupus erythematosus. Mol Med Today 6:418–424

    PubMed  Google Scholar 

  354. Cerillo G, Rees A, Manchanda N, Reilly C, Brogan I I, White A, Needham M (1998) The oestrogen receptor regulates NFκB and AP-1 activity in a cell-specific manner. J Steroid Biochem Mol Biol 67:79–88

    PubMed  Google Scholar 

  355. McKay LI, Cidlowski JA (1999) Molecular control of immune/inflammatory responses: interactions between nuclear factor-κB and steroid receptor-signaling pathways. Endocr Rev 20:435–459

    PubMed  Google Scholar 

  356. Baeuerle PA, Henkel T (1994) Function and activation of NF- κB in the immune system. Annu Rev Immunol 12:141–179

    PubMed  Google Scholar 

  357. Liu J, Beller DI (2003) Distinct pathways for NF-κB regulation are associated with aberrant macrophage IL-12 production in lupus- and diabetes-prone mouse strains. J Immunol 9:4489–4496

    Google Scholar 

  358. Katsiari CG, Tsokos GC (2006) Transcriptional repression of interleukin-2 in human systemic lupus erythematosus. Autoimmun Rev 5:118–121

    PubMed  Google Scholar 

  359. Dean JH, Cornacoff JB, Haley PJ, Hincks JR (1994) The integration of immunotoxicology in drug discovery and development: investigative and in vitro possibilities. Toxicol In Vitro 8:939–944

    PubMed  Google Scholar 

  360. Koppe J, de Boer P (2001) Immunotoxicity by dioxins and PCBs in the perinatal period. In: Nikolopoulou-Stamati P, Hens L, Howard CV (eds) Endocrine disruptors environmental health and policies. Kluwer Academic Publishers, Dordrecht

    Google Scholar 

  361. Lahvis GP, Wells RS, Kuehl DW, Stewart JL, Rhinehart HL, Via CS (1995) Decreased lymphocyte responses in free-ranging bottlenose dolphins (Tursiops truncatus) are associated with increased concentrations of PCBs and DDT in peripheral blood. Environ Health Perspect 103(Suppl 4):67–72

    PubMed  Google Scholar 

  362. Smialowicz RJ, DeVito MJ, Williams WC, Birnbaum LS (2008) Relative potency based on hepatic enzyme induction predicts immunosuppressive effects of a mixture of PCDDS/PCDFS and PCBS. Toxicol Appl Pharmacol 227:477–484

    PubMed  Google Scholar 

  363. Pesatori AC, Zocchetti C, Guercilena S, Consonni D, Turrini D, Bertazzi PA (1998) Dioxin exposure and non-malignant health effects: a mortality study. Occup Environ Med 55:126–131

    PubMed  Google Scholar 

  364. Nagayama J, Tsuji H, Iida T, Hirakawa H, Matsueda T, Ohki M (2001) Effects of contamination level of dioxins and related chemicals on thyroid hormone and immune response systems in patients with “Yusho”. Chemosphere 43:1005–1010

    PubMed  Google Scholar 

  365. Nakanishi Y, Shigematsu N, Kurita Y, Matsuba K, Kanegae H, Ishimaru S, Kawazoe Y (1985) Respiratory involvement and immune status in Yusho patients. Environ Health Perspect 59:31–36

    PubMed  Google Scholar 

  366. Aoki Y (2001) Polychlorinated biphenyls, polychlorinated dibenzo-p-dioxins, and polychlorinated dibenzofurans as endocrine disrupters–what we have learned from Yusho disease. Environ Res 86:2–11

    PubMed  Google Scholar 

  367. Weisglas-Kuperus N, Patandin S, Berbers GA, Sas TC, Mulder PG, Sauer PJ, Hooijkaas H (2000) Immunologic effects of background exposure to polychlorinated biphenyls and dioxins in Dutch preschool children. Environ Health Perspect 108:1203–1207

    PubMed  Google Scholar 

  368. Leijs MM, Koppe JG, Olie K, van Aalderen WM, de Voogt P, ten Tusscher GW (2009) Effects of dioxins, PCBs, and PBDEs on immunology and hematology in adolescents. Environ Sci Technol 43:7946–7955

    PubMed  Google Scholar 

  369. ten Tusscher GW, Koppe JG (2004) Perinatal dioxin exposure and later effects–a review. Chemosphere 54:1329–1336

    PubMed  Google Scholar 

  370. Bornehag CG, Nanberg E (2010) Phthalate exposure and asthma in children. Int J Androl 33:333–345

    PubMed  Google Scholar 

  371. Langer P (2010) The impacts of organochlorines and other persistent pollutants on thyroid and metabolic health. Front Neuroendocrinol 314:497–518

    Google Scholar 

  372. Iida T, Hirakawa H, Matsueda T, Takenaka S, Nagayama J (1999) Polychlorinated dibenzo-p-dioxins and related compounds: the blood levels of young Japanese women. Chemosphere 3815:3497–3502

    Google Scholar 

  373. Lane NE (2006) Epidemiology, etiology, and diagnosis of osteoporosis. Am J Obstet Gynecol 194(Suppl 2):S3–S11

    PubMed  Google Scholar 

  374. North American Menopause Society (2006) Management of osteoporosis in postmenopausal women: 2006 position statement of The North American Menopause Society. Menopause 13:340–367

    Google Scholar 

  375. Bord S, Horner A, Beavan S, Compston J (2001) Estrogen receptors alpha and beta are differentially expressed in developing human bone. J Clin Endocrinol Metab 86:2309–2314

    PubMed  Google Scholar 

  376. Braidman IP, Hainey L, Batra G, Selby PL, Saunders PT, Hoyland JA (2001) Localization of estrogen receptor beta protein expression in adult human bone. J Bone Miner Res 16:214–220

    PubMed  Google Scholar 

  377. Windahl SH, Vidal O, Andersson G, Gustafsson JA, Ohlsson C (1999) Increased cortical bone mineral content but unchanged trabecular bone mineral density in female ERbeta (-/-) mice. J Clin Invest 104:895–901

    PubMed  Google Scholar 

  378. McDougall KE, Perry MJ, Gibson RL, Colley SM, Korach KS, Tobias JH (2003) Estrogen receptor-alpha dependency of estrogen’s stimulatory action on cancellous bone formation in male mice. Endocrinology 144:1994–1999

    PubMed  Google Scholar 

  379. Windahl SH, Andersson G, Gustafsson JA (2002) Elucidation of estrogen receptor function in bone with the use of mouse models. Trends Endocrinol Metab 13:195–200

    PubMed  Google Scholar 

  380. Smith EP, Boyd J, Frank GR, Takahashi H, Cohen RM, Specker B, Williams TC, Lubahn DB, Korach KS (1994) Estrogen resistance caused by a mutation in the estrogen-receptor gene in a man. N Engl J Med 331:1056–1061

    PubMed  Google Scholar 

  381. Alveblom AK, Rylander L, Johnell O, Hagmar L (2003) Incidence of hospitalized osteoporotic fractures in cohorts with high dietary intake of persistent organochlorine compounds. Int Arch Occup Environ Health 76:246–248

    PubMed  Google Scholar 

  382. Beard J, Marshall S, Jong K, Newton R, Tripplett-McBride T, Humphries B, Bronks R (2000) 1,1,1-trichloro-2,2-bis (p-chlorophenyl)-ethane (DDT) and reduced bone mineral density. Arch Environ Health 55:177–180

    PubMed  Google Scholar 

  383. Côté S, Ayotte P, Dodin S, Blanchet C, Mulvad G, Petersen HS, Gingras S, Dewailly E (2006) Plasma organochlorine concentrations and bone ultrasound measurements: a cross-sectional study in peri-and postmenopausal Inuit women from Greenland. Environ Health 21:5–33

    Google Scholar 

  384. Wang SL, Lin CY, Guo YL, Lin LY, Chou WL, Chang LW (2004) Infant exposure to polychlorinated dibenzo-p-dioxins, dibenzofurans and biphenyls (PCDD/Fs, PCBs)–correlation between prenatal and postnatal exposure. Chemosphere 54:1459–1473

    PubMed  Google Scholar 

  385. Tan J, Li QQ, Loganath A, Chong YS, Xiao M, Obbard JP (2008) Multivariate data analyses of persistent organic pollutants in maternal adipose tissue in Singapore. Environ Sci Technol 42:2681–2687

    PubMed  Google Scholar 

  386. Braun JM, Hauser R (2011) Bisphenol A and children’s health. Curr Opin Pediatr 23:233–239

    PubMed  Google Scholar 

  387. Steinhardt GF (2004) Endocrine disruption and hypospadias. Adv Exp Med Biol 545:203–215

    PubMed  Google Scholar 

  388. Palanza P, Gioiosa L, vom Saal FS, Parmigiani S (2008) Effects of developmental exposure to bisphenol A on brain and behavior in mice. Environ Res 108:150–157

    PubMed  Google Scholar 

  389. Schönfelder G, Friedrich K, Paul M, Chahoud I (2004) Developmental effects of prenatal exposure to bisphenol a on the uterus of rat offspring. Neoplasia 6:584–594

    PubMed  Google Scholar 

  390. Fernández M, Bourguignon N, Lux-Lantos V, Libertun C (2010) Neonatal exposure to bisphenol a and reproductive and endocrine alterations resembling the polycystic ovarian syndrome in adult rats. Environ Health Perspect 118:1217–1222

    PubMed  Google Scholar 

  391. vom Saal FS, Akingbemi BT, Belcher SM, Birnbaum LS, Crain DA, Eriksen M, Farabollini F, Guillette LJ Jr, Hauser R, Heindel JJ, Ho SM, Hunt PA, Iguchi T, Jobling S, Kanno J, Keri RA, Knudsen KE, Laufer H, LeBlanc GA, Marcus M, McLachlan JA, Myers JP, Nadal A, Newbold RR, Olea N, Prins GS, Richter CA, Rubin BS, Sonnenschein C, Soto AM, Talsness CE, Vandenbergh JG, Vandenberg LN, Walser-Kuntz DR, Watson CS, Welshons WV, Wetherill Y, Zoeller RT (2007) Chapel Hill bisphenol A expert panel consensus statement: integration of mechanisms, effects in animals and potential to impact human health at current levels of exposure. Reprod Toxicol 24:131–138

    Google Scholar 

  392. Golub MS, Wu KL, Kaufman FL, Li LH, Moran-Messen F, Zeise L, Alexeeff GV, Donald JM (2010) Bisphenol A: developmental toxicity from early prenatal exposure. Birth Defects Res B Dev Reprod Toxicol 89:441–466

    PubMed  Google Scholar 

  393. Welshons WV, Nagel SC, Thayer KA, Judy BM, vom Saal FS (1999) Low-dose bioactivity of xenoestrogens in animals: fetal exposure to low doses of methoxychlor and other xenoestrogens increases adult prostate size in mice. Toxicol Ind Health 15:12–25

    PubMed  Google Scholar 

  394. Baker VA (2001) Endocrine disrupters–testing strategies to assess human hazard. Toxicol In Vitro 15:413–419

    PubMed  Google Scholar 

  395. Longnecker MP, Klebanoff MA, Zhou H, Brock JW (2001) Association between maternal serum concentration of the DDT metabolite DDE and preterm and small-for-gestational-age babies at birth. Lancet 358:110–114

    PubMed  Google Scholar 

  396. Colborn T (2004) Neurodevelopment and endocrine disruption. Environ Health Perspect 112:944–949

    PubMed  Google Scholar 

  397. Schreiber J (1997) Transport of organic chemicals to breast milk: tetrachloroethene case study. In: Kacew S, Lambert G (eds) Environmental toxicology and pharmacology of human development. Taylor & Francis, Washington, DC

    Google Scholar 

  398. Iida T, Hirakawa H, Matsueda T, Takenaka S, Nagayama J (1999) Polychlorinated dibenzo-p-dioxins and related compounds in breast milk of Japanese primiparas and multiparas. Chemosphere 38:2461–2466

    PubMed  Google Scholar 

  399. Walkowiak J, Wiener JA, Fastabend A, Heinzow B, Krämer U, Schmidt E, Steingrüber HJ, Wundram S, Winneke GO (2001) Environmental exposure to polychlorinated biphenyls and quality of the home environment: effects on psychodevelopment in early childhood. Lancet 358:1602–1607

    PubMed  Google Scholar 

  400. Schlumpf M, Kypke K, Wittassek M, Angerer J, Mascher H, Mascher D, Vökt C, Birchler M, Lichtensteiger W (2010) Exposure patterns of UV filters, fragrances, parabens, phthalates, organochlor pesticides, PBDEs, and PCBs in human milk: correlation of UV filters with use of cosmetics. Chemosphere 81:1171–1183

    PubMed  Google Scholar 

  401. Hooper K, McDonald TA (2000) The PBDEs: an emerging environmental challenge and another reason for breast-milk monitoring programs. Environ Health Perspect 108:387–392

    PubMed  Google Scholar 

  402. Howdeshell KL, Wilson VS, Furr J, Lambright CR, Rider CV, Blystone CR, Hotchkiss AK, Gray LE Jr (2008) A mixture of five phthalate esters inhibits fetal testicular testosterone production in the Sprague–Dawley rat in a cumulative, dose-additive manner. Toxicol Sci 105:153–165

    PubMed  Google Scholar 

  403. Harvey PW, Everett DJ (2006) Regulation of endocrine-disrupting chemicals: critical overview and deficiencies in toxicology and risk assessment for human health. Best Pract Res Clin Endocrinol Metab 20:145–165

    PubMed  Google Scholar 

  404. Bignert A, Olsson M, Persson W, Jensen S, Zakrisson S, Litzén K, Eriksson U, Häggberg L, Alsberg T (1998) Temporal trends of organochlorines in Northern Europe, 1967–1995. Relation to global fractionation, leakage from sediments and international measures. Environ Pollut 99:177–198

    PubMed  Google Scholar 

  405. Rubin BS, Murray MK, Damassa DA, King JC, Soto AM (2001) Perinatal exposure to low doses of bisphenol A affects body weight, patterns of estrous cyclicity, and plasma LH levels. Environ Health Perspect 109:675–680

    PubMed  Google Scholar 

  406. Palmer JR, Anderson D, Helmrich SP, Herbst AL (2000) Risk factors for diethylstilbestrol-associated clear cell adenocarcinoma. Obstet Gynecol 95:814–820

    Google Scholar 

  407. Sheehan DM, Willingham E, Gaylor D, Bergeron JM, Crews D (1999) No threshold dose for estradiol-induced sex reversal of turtle embryos: how little is too much? Environ Health Perspect 107:155–159

    PubMed  Google Scholar 

  408. Laporte JR (1978) Multinationals and health: reflections on the Seveso catastrophe. Int J Health Serv 8:619–632

    PubMed  Google Scholar 

  409. Myers JP, Zoeller RT, vom Saal FS (2009) A clash of old and new scientific concepts in toxicity, with important implications for public health. Environ Health Perspect 117:1652–1655

    PubMed  Google Scholar 

  410. Watson CS, Jeng YJ, Kochukov MY (2010) Nongenomic signaling pathways of estrogen toxicity. Toxicol Sci 115:1–11

    PubMed  Google Scholar 

  411. Koo HJ, Lee BM (2004) Estimated exposure to phthalates in cosmetics and risk assessment. J Toxicol Environ Health A 67:1901–1914

    PubMed  Google Scholar 

  412. Yang SH, Morgan AA, Nguyen HP, Moore H, Figard BJ, Schug KA (2011) Quantitative determination of bisphenol A from human saliva using bulk derivatization and trap-and-elute liquid chromatography coupled to electrospray ionization mass spectrometry. Environ Toxicol Chem 30:1243–1251

    PubMed  Google Scholar 

  413. Brouwers MM, Besselink H, Bretveld RW, Anzion R, Scheepers PT, Brouwer A, Roeleveld N (2011) Estrogenic and androgenic activities in total plasma measured with reporter-gene bioassays: relevant exposure measures for endocrine disruptors in epidemiologic studies? Environ Int 37:557–564

    PubMed  Google Scholar 

  414. Patel CJ, Butte AJ (2010) Predicting environmental chemical factors associated with disease-related gene expression data. BMC Med Genomics 6:3–17

    Google Scholar 

  415. Hubal EA, Sheldon LS, Zufall MJ, Burke JM, Thomas KW (2000) The challenge of assessing children’s residential exposure to pesticides. J Expo Anal Environ Epidemiol 10:638–649

    PubMed  Google Scholar 

  416. Bonefeld-Jørgensen EC, Andersen HR, Rasmussen TH, Vinggaard AM (2001) Effect of highly bioaccumulated polychlorinated biphenyl congeners on estrogen and androgen receptor activity. Toxicology 158:141–153

    PubMed  Google Scholar 

  417. Fang H, Tong W, Perkins R, Soto AM, Prechtl NV, Sheehan DM (2002) Quantitative comparisons of in vitro assays for estrogenic activities. Environ Health Perspect 108:723–729

    Google Scholar 

  418. Legler J, Zeinstra LM, Schuitemaker F, Lanser PH, Bogerd J, Brouwer A, Vethaak AD, De Voogt P, Murk AJ, Van der Burg B (2002) Comparison of in vivo and in vitro reporter gene assays for short-term screening of estrogenic activity. Environ Sci Technol 36:4410–4415

    PubMed  Google Scholar 

  419. Hong H, Tong W, Fang H, Shi L, Xie Q, Wu J, Perkins R, Walker JD, Branham W, Sheehan DM (2002) Prediction of estrogen receptor binding for 58,000 chemicals using an integrated system of a tree-based model with structural alerts. Environ Health Perspect 110:29–36

    PubMed  Google Scholar 

  420. Bolger R, Wiese TE, Ervin K, Nestich S, Checovich W (1998) Rapid screening of environmental chemicals for estrogen receptor binding capacity. Environ Health Perspect 106:551–557

    PubMed  Google Scholar 

  421. Soto AM, Justicia H, Wray JW, Sonnenschein C (1991) p-Nonyl-phenol: an estrogenic xenobiotic released from “modified” polystyrene. Environ Health Perspect 92:167–173

    PubMed  Google Scholar 

  422. Jeltsch JM, Roberts M, Schatz C, Garnier JM, Brown AM, Chambon P (1987) Structure of the human oestrogen-responsive gene pS2. Nucleic Acids Res 15:1401–1414

    PubMed  Google Scholar 

  423. EDSTAC (1998) Endocrine disruptor screening and testing advisory committee final report U.S. Environmental Protection Agency. http://www.epa.gov/endo/pubs/edspoverview/finalrpt.htm

  424. Charles GD, Kan HL, Schisler MR, Bhaskar Gollapudi B, Sue Marty M (2005) A comparison of in vitro and in vivo EDSTAC test battery results for detecting antiandrogenic activity. Toxicol Appl Pharmacol 202:108–120

    PubMed  Google Scholar 

  425. Folmar LC, Hemmer MJ, Denslow ND, Kroll K, Chen J, Cheek A, Richman H, Meredith H, Grau EG (2002) A comparison of the estrogenic potencies of estradiol, ethynylestradiol, diethylstilbestrol, nonylphenol and methoxychlor in vivo and in vitro. Aquat Toxicol 60:101–110

    PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Charis Liapi .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2014 Springer-Verlag Berlin Heidelberg

About this chapter

Cite this chapter

Dimogerontas, G., Liapi, C. (2014). Endocrine Disruptors (Xenoestrogens): An Overview. In: Eliades, T., Eliades, G. (eds) Plastics in Dentistry and Estrogenicity. Springer, Berlin, Heidelberg. https://doi.org/10.1007/978-3-642-29687-1_1

Download citation

  • DOI: https://doi.org/10.1007/978-3-642-29687-1_1

  • Published:

  • Publisher Name: Springer, Berlin, Heidelberg

  • Print ISBN: 978-3-642-29686-4

  • Online ISBN: 978-3-642-29687-1

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics