EDC-2: The Endocrine Society's Second Scientific Statement on Endocrine-Disrupting Chemicals
Tóm tắt
Từ khóa
Tài liệu tham khảo
Diamanti-Kandarakis, 2009, Endocrine-disrupting chemicals: an Endocrine Society Scientific Statement, Endocr Rev, 30, 293, 10.1210/er.2009-0002
Gore, 2015, Executive summary to EDC-2: The Endocrine Society's second scientific statement on endocrine-disrupting chemicals, Endocr Rev, 36, 10.1210/er.2015-1093
Zoeller, 2012, Endocrine-disrupting chemicals and public health protection: a statement of principles from the Endocrine Society, Endocrinology, 153, 4097, 10.1210/en.2012-1422
Dodds, 1936, Synthetic oestrogenic agents without the phenanthrene nucleus, Nature, 137, 996, 10.1038/137996a0
vom Saal, 2014, Evidence that bisphenol A (BPA) can be accurately measured without contamination in human serum and urine, and that BPA causes numerous hazards from multiple routes of exposure, Mol Cell Endocrinol, 398, 101, 10.1016/j.mce.2014.09.028
Meeker, 2011, Relationship between urinary phthalate and bisphenol A concentrations and serum thyroid measures in U.S. adults and adolescents from the National Health and Nutrition Examination Survey (NHANES) 2007–2008, Environ Health Perspect, 119, 1396, 10.1289/ehp.1103582
Calafat, 2008, Exposure of the U.S. population to bisphenol A and 4-tertiary-octylphenol: 2003–2004, Environ Health Perspect, 116, 39, 10.1289/ehp.10753
Völkel, 2002, Metabolism and kinetics of bisphenol A in humans at low doses following oral administration, Chem Res Toxicol, 15, 1281, 10.1021/tx025548t
Ng, 2014, Global, regional, and national prevalence of overweight and obesity in children and adults during 1980–2013: a systematic analysis for the Global Burden of Disease Study 2013, Lancet, 384, 766, 10.1016/S0140-6736(14)60460-8
Churchwell, 2014, Comparison of life-stage-dependent internal dosimetry for bisphenol A, ethinyl estradiol, a reference estrogen, and endogenous estradiol to test an estrogenic mode of action in Sprague Dawley rats, Toxicol Sci, 139, 4, 10.1093/toxsci/kfu021
Vandenberg, 2014, A round robin approach to the analysis of bisphenol A (BPA) in human blood samples, Environ Health, 13, 25, 10.1186/1476-069X-13-25
Patterson, 2013, Concurrent determination of bisphenol A pharmacokinetics in maternal and fetal rhesus monkeys, Toxicol Appl Pharmacol, 267, 41, 10.1016/j.taap.2012.12.006
Gerona, 2013, Bisphenol-A (BPA), BPA glucuronide, and BPA sulfate in midgestation umbilical cord serum in a northern and central California population, Environ Sci Technol, 47, 12477, 10.1021/es402764d
Liao, 2012, Determination of free and conjugated forms of bisphenol A in human urine and serum by liquid chromatography-tandem mass spectrometry, Environ Sci Technol, 46, 5003, 10.1021/es300115a
Veiga-Lopez, 2015, Impact of gestational bisphenol A on oxidative stress and free fatty acids: human association and interspecies animal testing studies, Endocrinology, 156, 911, 10.1210/en.2014-1863
Teeguarden, 2013, Are typical human serum BPA concentrations measurable and sufficient to be estrogenic in the general population?, Food Chem Toxicol, 62, 949, 10.1016/j.fct.2013.08.001
Nahar, 2013, Fetal liver bisphenol A concentrations and biotransformation gene expression reveal variable exposure and altered capacity for metabolism in humans, J Biochem Mol Toxicol, 27, 116, 10.1002/jbt.21459
Wu, 2012, The public health threat of phthalate-tainted foodstuffs in Taiwan: the policies the government implemented and the lessons we learned, Environ Int, 44, 75, 10.1016/j.envint.2012.01.014
Wu, 2014, Findings of 2731 suspected phthalate-tainted foodstuffs during the 2011 phthalates incident in Taiwan, J Formos Med Assoc, 113, 600, 10.1016/j.jfma.2014.02.010
U.S. Environmental Protection Agency, 2007, Phthalates: TEACH Chemical Summary
Hines, 2009, Concentrations of phthalate metabolites in milk, urine, saliva, and serum of lactating North Carolina women, Environ Health Perspect, 117, 86, 10.1289/ehp.11610
Fromme, 2011, Phthalates and their metabolites in breast milk–results from the Bavarian Monitoring of Breast Milk (BAMBI), Environ Int, 37, 715, 10.1016/j.envint.2011.02.008
Hannon, 2015, The effects of phthalates on the ovary, Front Endocrinol (Lausanne), 6, 8, 10.3389/fendo.2015.00008
Gianessi, 1998, Benefits of triazine herbicides, Triazine Herbicides: Risk Assessment, 1, 10.1021/bk-1998-0683.ch001
Agency for Toxic Substances and Disease Registry, 2003, Toxicological Profile for Atrazine
Solomon, 2013, Ecological risk assessment of atrazine in North American surface waters, Environ Toxicol Chem, 32, 10, 10.1002/etc.2050
Boucher, 2014, Domain-specific effects of prenatal exposure to PCBs, mercury, and lead on infant cognition: results from the Environmental Contaminants and Child Development Study in Nunavik, Environ Health Perspect, 122, 310, 10.1289/ehp.1206323
Jurewicz, 2013, Chemical exposure early in life and the neurodevelopment of children–an overview of current epidemiological evidence, Ann Agric Environ Med, 20, 465
Doi, 2013, Prenatal exposure to a polychlorinated biphenyl (PCB) congener influences fixation duration on biological motion at 4-months-old: a preliminary study, PLoS One, 8, e59196, 10.1371/journal.pone.0059196
Herrick, 2004, An unrecognized source of PCB contamination in schools and other buildings, Environ Health Perspect, 112, 1051, 10.1289/ehp.6912
Lauby-Secretan, 2013, Carcinogenicity of polychlorinated biphenyls and polybrominated biphenyls, Lancet Oncol, 14, 287, 10.1016/S1470-2045(13)70104-9
Soto, 1995, The E-SCREEN assay as a tool to identify estrogens: an update on estrogenic environmental pollutants, Environ Health Perspect, 103, 113
Portigal, 2002, Polychlorinated biphenyls interfere with androgen-induced transcriptional activation and hormone binding, Toxicol Appl Pharmacol, 179, 185, 10.1006/taap.2002.9371
Agency for Toxic Substances and Disease Registry, 2004, Toxicological Profile for Polybrominated Biphenyls and Polybrominated Diphenyl Ethers (PBBs and PBDEs)
Zota, 2011, Polybrominated diphenyl ethers, hydroxylated polybrominated diphenyl ethers, and measures of thyroid function in second trimester pregnant women in California, Environ Sci Technol, 45, 7896, 10.1021/es200422b
Costa, 2007, Developmental neurotoxicity of polybrominated diphenyl ether (PBDE) flame retardants, Neurotoxicology, 28, 1047, 10.1016/j.neuro.2007.08.007
U.S. Environmental Protection Agency, 2010, An Exposure Assessment of Polybrominated Diphenyl Ethers
Knower, 2014, Endocrine disruption of the epigenome: a breast cancer link, Endocr Relat Cancer, 21, T33, 10.1530/ERC-13-0513
National Toxicology Program, 2011, Report on Carcinogens, 12th Edition, iii
McGlynn, 2008, Persistent organochlorine pesticides and risk of testicular germ cell tumors, J Natl Cancer Inst, 100, 663, 10.1093/jnci/djn101
Hardell, 2004, Adipose tissue concentrations of p,p'-DDE and the risk for endometrial cancer, Gynecol Oncol, 95, 706, 10.1016/j.ygyno.2004.08.022
Porta, 2008, Differences in serum concentrations of organochlorine compounds by occupational social class in pancreatic cancer, Environ Res, 108, 370, 10.1016/j.envres.2008.06.010
Codru, 2007, Diabetes in relation to serum levels of polychlorinated biphenyls and chlorinated pesticides in adult Native Americans, Environ Health Perspect, 115, 1442, 10.1289/ehp.10315
Safe, 1997, Organochlorine exposure and risk for breast cancer, Prog Clin Biol Res, 396, 133
Wolff, 1993, Blood levels of organochlorine residues and risk of breast cancer, J Natl Cancer Inst, 85, 648, 10.1093/jnci/85.8.648
Xu, 2014, Bisphenol A regulates the estrogen receptor α signaling in developing hippocampus of male rats through estrogen receptor, Hippocampus, 24, 1570, 10.1002/hipo.22336
Martinez-Arguelles, 2014, In utero exposure to the endocrine disruptor di-(2-ethylhexyl) phthalate induces long-term changes in gene expression in the adult male adrenal gland, Endocrinology, 155, 1667, 10.1210/en.2013-1921
World Health Organization, 2014, Global Status Report on Noncommunicable Diseases
World Health Organization, 2013, Fact Sheets: Noncommunicable Diseases
Sladek, 2007, A genome-wide association study identifies novel risk loci for type 2 diabetes, Nature, 445, 881, 10.1038/nature05616
Fall, 2014, Genome-wide association studies of obesity and metabolic syndrome, Mol Cell Endocrinol, 382, 740, 10.1016/j.mce.2012.08.018
Vaxillaire, 2014, Type 2 diabetes-related genetic risk scores associated with variations in fasting plasma glucose and development of impaired glucose homeostasis in the prospective DESIR study, Diabetologia, 57, 1601, 10.1007/s00125-014-3277-x
Barouki, 2012, Developmental origins of non-communicable disease: implications for research and public health, Environ Health, 11, 42, 10.1186/1476-069X-11-42
Nadal, 2009, The pancreatic β-cell as a target of estrogens and xenoestrogens: implications for blood glucose homeostasis and diabetes, Mol Cell Endocrinol, 304, 63, 10.1016/j.mce.2009.02.016
Grandjean, 2008, Late insights into early origins of disease, Basic Clin Pharmacol Toxicol, 102, 94, 10.1111/j.1742-7843.2007.00167.x
Barker, 1997, Fetal undernutrition and disease in later life, Rev Reprod, 2, 105, 10.1530/ror.0.0020105
Maffini, 2010, Development and maturation of the normal female reproductive system: breast, Environmental Impacts on Reproductive Health and Fertility, 36
Newbold, 2010, Developmental exposures and implications for early and latent disease, Environmental Impacts on Reproductive Health and Fertility, 93, 10.1017/CBO9780511674686.008
Schug, 2011, Endocrine disrupting chemicals and disease susceptibility, J Steroid Biochem Mol Biol, 127, 204, 10.1016/j.jsbmb.2011.08.007
Gorski, 2002, Hypothalamic imprinting by gonadal steroid hormones, Adv Exp Med Biol, 511, 57, 10.1007/978-1-4615-0621-8_5
Collman, 2011, Developmental basis of disease: environmental impacts, J Dev Orig Health Dis, 2, 49, 10.1017/S2040174411000031
Barker, 1990, The fetal and infant origins of adult disease, BMJ, 301, 1111, 10.1136/bmj.301.6761.1111
Barker, 2004, The developmental origins of adult disease, J Am Coll Nutr, 23, 588S, 10.1080/07315724.2004.10719428
Ravelli, 1976, Obesity in young men after famine exposure in utero and early infancy, N Engl J Med, 295, 349, 10.1056/NEJM197608122950701
Roseboom, 2001, Effects of prenatal exposure to the Dutch famine on adult disease in later life: an overview, Mol Cell Endocrinol, 185, 93, 10.1016/S0303-7207(01)00721-3
Hilakivi-Clarke, 2013, Exposures to synthetic estrogens at different times during the life, and their effect on breast cancer risk, J Mammary Gland Biol Neoplasia, 18, 25, 10.1007/s10911-013-9274-8
Dieckmann, 1953, Does the administration of diethylstilbestrol during pregnancy have therapeutic value?, Am J Obstet Gynecol, 66, 1062, 10.1016/S0002-9378(16)38617-3
Herbst, 1971, Adenocarcinoma of the vagina. Association of maternal stilbestrol therapy with tumor appearance in young women, N Engl J Med, 284, 878, 10.1056/NEJM197104222841604
Troisi, 2007, Cancer risk in women prenatally exposed to diethylstilbestrol, Int J Cancer, 121, 356, 10.1002/ijc.22631
Verloop, 2010, Cancer risk in DES daughters, Cancer Causes Control, 21, 999, 10.1007/s10552-010-9526-5
Herbst, 1976, Summary of the changes in the human female genital tract as a consequence of maternal diethylstilbestrol therapy, J Toxicol Environ Health Suppl, 1, 13
Edelman, 1986, Urogenital tract changes in female offspring exposed to DES, DES/Diethylstilbestrol—New Perspectives, 69
Troisi, 2013, Medical conditions among adult offspring prenatally exposed to diethylstilbestrol, Epidemiology, 24, 430, 10.1097/EDE.0b013e318289bdf7
Harris, 2012, Diethylstilboestrol–a long-term legacy, Maturitas, 72, 108, 10.1016/j.maturitas.2012.03.002
Titus-Ernstoff, 2008, Offspring of women exposed in utero to diethylstilbestrol (DES): a preliminary report of benign and malignant pathology in the third generation, Epidemiology, 19, 251, 10.1097/EDE.0b013e318163152a
Christensen, 2011, Epigenomics in environmental health, Front Genet, 2, 84, 10.3389/fgene.2011.00084
Rissman, 2014, Minireview: transgenerational epigenetic inheritance: focus on endocrine disrupting compounds, Endocrinology, 155, 2770, 10.1210/en.2014-1123
Ho, 2012, Environmental epigenetics and its implication on disease risk and health outcomes, Ilar J, 53, 289, 10.1093/ilar.53.3-4.289
Greally, 2013, In vitro and in vivo testing methods of epigenomic endpoints for evaluating endocrine disruptors, Altex, 30, 445, 10.14573/altex.2013.4.445
Uzumcu, 2012, Epigenetic mechanisms in the actions of endocrine-disrupting chemicals: gonadal effects and role in female reproduction, Reprod Domest Anim, 47, 338, 10.1111/j.1439-0531.2012.02096.x
Jirtle, 2007, Environmental epigenomics and disease susceptibility, Nat Rev Genet, 8, 253, 10.1038/nrg2045
Lister, 2013, Global epigenomic reconfiguration during mammalian brain development, Science, 341, 1237905, 10.1126/science.1237905
Lister, 2009, Human DNA methylomes at base resolution show widespread epigenomic differences, Nature, 462, 315, 10.1038/nature08514
Bogdanovic, 2009, DNA methylation and methyl-CpG binding proteins: developmental requirements and function, Chromosoma, 118, 549, 10.1007/s00412-009-0221-9
Tiwari, 2008, PcG proteins, DNA methylation, and gene repression by chromatin looping, PLoS Biol, 6, 2911, 10.1371/journal.pbio.0060306
Cosgrove, 2004, Regulated nucleosome mobility and the histone code, Nat Struct Mol Biol, 11, 1037, 10.1038/nsmb851
Clapier, 2009, The biology of chromatin remodeling complexes, Annu Rev Biochem, 78, 273, 10.1146/annurev.biochem.77.062706.153223
Berdasco, 2010, Aberrant epigenetic landscape in cancer: how cellular identity goes awry, Dev Cell, 19, 698, 10.1016/j.devcel.2010.10.005
Cannell, 2008, How do microRNAs regulate gene expression?, Biochem Soc Trans, 36, 1224, 10.1042/BST0361224
Gore, 2014, Implications of prenatal steroid perturbations for neurodevelopment, behavior, and autism, Endocr Rev, 35, 961, 10.1210/er.2013-1122
Skinner, 2010, Epigenetic transgenerational actions of environmental factors in disease etiology, Trends Endocrinol Metab, 21, 214, 10.1016/j.tem.2009.12.007
Walker, 2011, Transgenerational neuroendocrine disruption of reproduction, Nat Rev Endocrinol, 7, 197, 10.1038/nrendo.2010.215
Crews, 2006, Epigenetics, evolution, endocrine disruption, health, and disease, Endocrinology, 147, S4, 10.1210/en.2005-1122
Skinner, 2008, What is an epigenetic transgenerational phenotype? F3 or F2, Reprod Toxicol, 25, 2, 10.1016/j.reprotox.2007.09.001
Bromer, 2010, Bisphenol-A exposure in utero leads to epigenetic alterations in the developmental programming of uterine estrogen response, FASEB J, 24, 2273, 10.1096/fj.09-140533
Newbold, 2006, Adverse effects of the model environmental estrogen diethylstilbestrol are transmitted to subsequent generations, Endocrinology, 147, S11, 10.1210/en.2005-1164
Newbold, 2004, Lessons learned from perinatal exposure to diethylstilbestrol, Toxicol Appl Pharmacol, 199, 142, 10.1016/j.taap.2003.11.033
Newbold, 2008, Prenatal exposure to diethylstilbestrol (DES), Fertil Steril, 89, e55, 10.1016/j.fertnstert.2008.01.062
Dolinoy, 2007, Maternal nutrient supplementation counteracts bisphenol A-induced DNA hypomethylation in early development, Proc Natl Acad Sci USA, 104, 13056, 10.1073/pnas.0703739104
Ho, 2006, Developmental exposure to estradiol and bisphenol A increases susceptibility to prostate carcinogenesis and epigenetically regulates phosphodiesterase type 4 variant 4, Cancer Res, 66, 5624, 10.1158/0008-5472.CAN-06-0516
Tang, 2012, Neonatal exposure to estradiol/bisphenol A alters promoter methylation and expression of Nsbp1 and Hpcal1 genes and transcriptional programs of Dnmt3a/b and Mbd2/4 in the rat prostate gland throughout life, Endocrinology, 153, 42, 10.1210/en.2011-1308
Susiarjo, 2013, Bisphenol A exposure disrupts genomic imprinting in the mouse, PLoS Genet, 9, e1003401, 10.1371/journal.pgen.1003401
Anderson, 2012, Epigenetic responses following maternal dietary exposure to physiologically relevant levels of bisphenol A, Environ Mol Mutagen, 53, 334, 10.1002/em.21692
Kim, 2013, Bisphenol A-associated epigenomic changes in prepubescent girls: a cross-sectional study in Gharbiah, Egypt, Environ Health, 12, 33, 10.1186/1476-069X-12-33
Salian, 2009, Perinatal exposure of rats to bisphenol A affects the fertility of male offspring, Life Sci, 85, 742, 10.1016/j.lfs.2009.10.004
Wolstenholme, 2013, Transgenerational effects of prenatal bisphenol A on social recognition, Horm Behav, 64, 833, 10.1016/j.yhbeh.2013.09.007
Crews, 2012, Epigenetic transgenerational inheritance of altered stress responses, Proc Natl Acad Sci USA, 109, 9143, 10.1073/pnas.1118514109
Crews, 2007, Transgenerational epigenetic imprints on mate preference, Proc Natl Acad Sci USA, 104, 5942, 10.1073/pnas.0610410104
Gillette, 2014, Sexually dimorphic effects of ancestral exposure to vinclozolin on stress reactivity in rats, Endocrinology, 155, 3853, 10.1210/en.2014-1253
Guerrero-Bosagna, 2010, Epigenetic transgenerational actions of vinclozolin on promoter regions of the sperm epigenome, PLoS One, 5, e13100, 10.1371/journal.pone.0013100
Anway, 2005, Epigenetic transgenerational actions of endocrine disruptors and male fertility, Science, 308, 1466, 10.1126/science.1108190
Guerrero-Bosagna, 2012, Epigenetic transgenerational inheritance of vinclozolin induced mouse adult onset disease and associated sperm epigenome biomarkers, Reprod Toxicol, 34, 694, 10.1016/j.reprotox.2012.09.005
Nilsson, 2012, Environmentally induced epigenetic transgenerational inheritance of ovarian disease, PLoS One, 7, e36129, 10.1371/journal.pone.0036129
Skinner, 2013, Environmentally induced transgenerational epigenetic reprogramming of primordial germ cells and the subsequent germ line, PLoS One, 8, e66318, 10.1371/journal.pone.0066318
Li, 2014, Exposure to diethylhexyl phthalate (DEHP) results in a heritable modification of imprint genes DNA methylation in mouse oocytes, Mol Biol Rep, 41, 1227, 10.1007/s11033-013-2967-7
Doyle, 2013, Transgenerational effects of di-(2-ethylhexyl) phthalate on testicular germ cell associations and spermatogonial stem cells in mice, Biol Reprod, 88, 112, 10.1095/biolreprod.112.106104
Wu, 2010, Dynamic epigenetic changes involved in testicular toxicity induced by di-2-(ethylhexyl) phthalate in mice, Basic Clin Pharmacol Toxicol, 106, 118, 10.1111/j.1742-7843.2009.00483.x
Song, 2010, Environmental neurotoxic pesticide increases histone acetylation to promote apoptosis in dopaminergic neuronal cells: relevance to epigenetic mechanisms of neurodegeneration, Mol Pharmacol, 77, 621, 10.1124/mol.109.062174
Song, 2011, Paraquat induces epigenetic changes by promoting histone acetylation in cell culture models of dopaminergic degeneration, Neurotoxicology, 32, 586, 10.1016/j.neuro.2011.05.018
Kuo, 2008, Genotoxicity of low dose N-nitroso propoxur to human gastric cells, Food Chem Toxicol, 46, 1619, 10.1016/j.fct.2008.01.014
Warita, 2010, Direct effects of diethylstilbestrol on the gene expression of the cholesterol side-chain cleavage enzyme (P450scc) in testicular Leydig cells, Life Sci, 87, 281, 10.1016/j.lfs.2010.06.020
Avissar-Whiting, 2010, Bisphenol A exposure leads to specific microRNA alterations in placental cells, Reprod Toxicol, 29, 401, 10.1016/j.reprotox.2010.04.004
Veiga-Lopez, 2013, Developmental programming: gestational bisphenol-A treatment alters trajectory of fetal ovarian gene expression, Endocrinology, 154, 1873, 10.1210/en.2012-2129
Kovanecz, 2014, Oral bisphenol A (BPA) given to rats at moderate doses is associated with erectile dysfunction, cavernosal lipofibrosis and alterations of global gene transcription, Int J Impot Res, 26, 67, 10.1038/ijir.2013.37
Tilghman, 2012, Endocrine disruptor regulation of microRNA expression in breast carcinoma cells, PLoS One, 7, e32754, 10.1371/journal.pone.0032754
Walker, 2012, Developmental reprogramming of cancer susceptibility, Nat Rev Cancer, 12, 479, 10.1038/nrc3220
McLachlan, 2001, Gene imprinting in developmental toxicology: a possible interface between physiology and pathology, Toxicol Lett, 120, 161, 10.1016/S0378-4274(01)00295-8
Vandenberg, 2012, Hormones and endocrine-disrupting chemicals: low-dose effects and nonmonotonic dose responses, Endocr Rev, 33, 378, 10.1210/er.2011-1050
Kovacs, 2012, Textbook of Endocrine Physiology
Geck, 1997, Expression of novel genes linked to the androgen-induced, proliferative shutoff in prostate cancer cells, J Steroid Biochem Mol Biol, 63, 211, 10.1016/S0960-0760(97)00122-2
Sonnenschein, 1989, Negative controls of cell proliferation: human prostate cancer cells and androgens, Cancer Res, 49, 3474
Welshons, 2003, Large effects from small exposures. I. Mechanisms for endocrine-disrupting chemicals with estrogenic activity, Environ Health Perspect, 111, 994, 10.1289/ehp.5494
Alonso-Magdalena, 2010, Bisphenol A exposure during pregnancy disrupts glucose homeostasis in mothers and adult male offspring, Environ Health Perspect, 118, 1243, 10.1289/ehp.1001993
Wei, 2011, Perinatal exposure to bisphenol A at reference dose predisposes offspring to metabolic syndrome in adult rats on a high-fat diet, Endocrinology, 152, 3049, 10.1210/en.2011-0045
Angle, 2013, Metabolic disruption in male mice due to fetal exposure to low but not high doses of bisphenol A (BPA): evidence for effects on body weight, food intake, adipocytes, leptin, adiponectin, insulin and glucose regulation, Reprod Toxicol, 42, 256, 10.1016/j.reprotox.2013.07.017
Hines, 2009, Phenotypic dichotomy following developmental exposure to perfluorooctanoic acid (PFOA) in female CD-1 mice: low doses induce elevated serum leptin and insulin, and overweight in mid-life, Mol Cell Endocrinol, 304, 97, 10.1016/j.mce.2009.02.021
Hao, 2012, The endocrine disruptor mono-(2-ethylhexyl) phthalate promotes adipocyte differentiation and induces obesity in mice, Biosci Rep, 32, 619, 10.1042/BSR20120042
Alonso-Magdalena, 2008, Pancreatic insulin content regulation by the estrogen receptor ER α, PLoS One, 3, e2069, 10.1371/journal.pone.0002069
vom Saal, 1997, Prostate enlargement in mice due to fetal exposure to low doses of estradiol or diethylstilbestrol and opposite effects at high doses, Proc Natl Acad Sci USA, 94, 2056, 10.1073/pnas.94.5.2056
Ziv-Gal, 2015, The effects of in utero bisphenol A exposure on reproductive capacity in several generations of mice, Toxicol Appl Pharmacol, 284, 354, 10.1016/j.taap.2015.03.003
Lichtenstein, 2000, Environmental and heritable factors in the causation of cancer–analyses of cohorts of twins from Sweden, Denmark, and Finland, N Engl J Med, 343, 78, 10.1056/NEJM200007133430201
Mocarelli, 1986, Clinical laboratory manifestations of exposure to dioxin in children. A six-year study of the effects of an environmental disaster near Seveso, Italy, JAMA, 256, 2687, 10.1001/jama.1986.03380190057025
Warner, 2011, Dioxin exposure and cancer risk in the Seveso Women's Health Study, Environ Health Perspect, 119, 1700, 10.1289/ehp.1103720
Warner, 2013, Diabetes, metabolic syndrome, and obesity in relation to serum dioxin concentrations: the Seveso Women's Health Study, Environ Health Perspect, 121, 906, 10.1289/ehp.1206113
Mocarelli, 2011, Perinatal exposure to low doses of dioxin can permanently impair human semen quality, Environ Health Perspect, 119, 713, 10.1289/ehp.1002134
Eskenazi, 2010, Serum dioxin concentrations and time to pregnancy, Epidemiology, 21, 224, 10.1097/EDE.0b013e3181cb8b95
Kim, 2003, Impact of Agent Orange exposure among Korean Vietnam veterans, Ind Health, 41, 149, 10.2486/indhealth.41.149
Agency for Toxic Substances and Disease Registry. 2014. Camp Lejeune, NC. Available at: http://www.atsdr.cdc.gov/sites/lejeune/index.html.
Barry, 2013, Perfluorooctanoic acid (PFOA) exposures and incident cancers among adults living near a chemical plant, Environ Health Perspect, 121, 1313, 10.1289/ehp.1306615
Bucher, 2002, The National Toxicology Program rodent bioassay: designs, interpretations, and scientific contributions, Ann NY Acad Sci, 982, 198, 10.1111/j.1749-6632.2002.tb04934.x
Rudel, 2007, Chemicals causing mammary gland tumors in animals signal new directions for epidemiology, chemicals testing, and risk assessment for breast cancer prevention, Cancer, 109, 2635, 10.1002/cncr.22653
Macon, 2013, Endocrine disruptors and the breast: early life effects and later life disease, J Mammary Gland Biol Neoplasia, 18, 43, 10.1007/s10911-013-9275-7
Tice, 2013, Improving the human hazard characterization of chemicals: a Tox21 update, Environ Health Perspect, 121, 756, 10.1289/ehp.1205784
Kavlock, 2012, Update on EPA's ToxCast program: providing high throughput decision support tools for chemical risk management, Chem Res Toxicol, 25, 1287, 10.1021/tx3000939
Schwarzman, Screening for chemical contributions to breast cancer risk: a case study for chemical safety evaluation [published online June 2, 2015], Environ Health Perspect
Myers, 2015, Estrogenic and anti-estrogenic activity of off-the-shelf hair and skin care products, J Expo Sci Environ Epidemiol, 25, 271, 10.1038/jes.2014.32
World Health Organization, 2015, Obesity and Overweight Fact Sheet No. 311
Finkelstein, 2009, Annual medical spending attributable to obesity: payer-and service-specific estimates, Health Aff (Millwood), 28, w822, 10.1377/hlthaff.28.5.w822
Baillie-Hamilton, 2002, Chemical toxins: a hypothesis to explain the global obesity epidemic, J Altern Complement Med, 8, 185, 10.1089/107555302317371479
Heindel, 2003, Endocrine disruptors and the obesity epidemic, Toxicol Sci, 76, 247, 10.1093/toxsci/kfg255
Newbold, 2005, Developmental exposure to estrogenic compounds and obesity, Birth Defects Res A Clin Mol Teratol, 73, 478, 10.1002/bdra.20147
Grün, 2006, Environmental obesogens: organotins and endocrine disruption via nuclear receptor signaling, Endocrinology, 147, S50, 10.1210/en.2005-1129
Enan, 1992, 2,3,7,8-Tetrachlorodibenzo-p-dioxin causes reduction of glucose transporting activities in the plasma membranes of adipose tissue and pancreas from the guinea pig, J Biol Chem, 267, 19785, 10.1016/S0021-9258(19)88622-2
Quesada, 2002, Low doses of the endocrine disruptor bisphenol-A and the native hormone 17β-estradiol rapidly activate transcription factor CREB, FASEB J, 16, 1671, 10.1096/fj.02-0313fje
Nadal, 2000, Nongenomic actions of estrogens and xenoestrogens by binding at a plasma membrane receptor unrelated to estrogen receptor α and estrogen receptor β, Proc Natl Acad Sci USA, 97, 11603, 10.1073/pnas.97.21.11603
Yau, 1977, The inhibitory effect of DDT on insulin secretion in mice, Toxicol Appl Pharmacol, 39, 81, 10.1016/0041-008X(77)90179-X
Alonso-Magdalena, 2006, The estrogenic effect of bisphenol A disrupts pancreatic β-cell function in vivo and induces insulin resistance, Environ Health Perspect, 114, 106, 10.1289/ehp.8451
Lin, 2011, Associations between maternal phthalate exposure and cord sex hormones in human infants, Chemosphere, 83, 1192, 10.1016/j.chemosphere.2010.12.079
Lee, 2006, PEGylated glucagon-like peptide-1 displays preserved effects on insulin release in isolated pancreatic islets and improved biological activity in db/db mice, Diabetologia, 49, 1608, 10.1007/s00125-006-0234-3
Lang, 2008, Association of urinary bisphenol A concentration with medical disorders and laboratory abnormalities in adults, JAMA, 300, 1303, 10.1001/jama.300.11.1303
WHO Expert Consultation, 2004, Appropriate body-mass index for Asian populations and its implications for policy and intervention strategies, Lancet, 363, 157, 10.1016/S0140-6736(03)15268-3
Segal, 2002, Twins and virtual twins: bases of relative body weight revisited, Int J Obes Relat Metab Disord, 26, 437, 10.1038/sj.ijo.0801941
Speakman, 2011, Set points, settling points and some alternative models: theoretical options to understand how genes and environments combine to regulate body adiposity, Dis Model Mech, 4, 733, 10.1242/dmm.008698
Klimentidis, 2011, Canaries in the coal mine: a cross-species analysis of the plurality of obesity epidemics, Proc Biol Sci, 278, 1626, 10.1098/rspb.2010.1890
Dirtu, 2013, Organohalogenated contaminants in domestic cats' plasma in relation to spontaneous acromegaly and type 2 diabetes mellitus: a clue for endocrine disruption in humans?, Environ Int, 57–58, 60, 10.1016/j.envint.2013.04.004
Gray, 2007, Adipose tissue expandability in the maintenance of metabolic homeostasis, Nutr Rev, 65, S7, 10.1301/nr.2007.jun.S7-S12
Virtue, 2010, Adipose tissue expandability, lipotoxicity and the metabolic syndrome–an allostatic perspective, Biochim Biophys Acta, 1801, 338, 10.1016/j.bbalip.2009.12.006
Grün, 2006, Endocrine-disrupting organotin compounds are potent inducers of adipogenesis in vertebrates, Mol Endocrinol, 20, 2141, 10.1210/me.2005-0367
Janesick, 2012, Obesogens, stem cells and the developmental programming of obesity, Int J Androl, 35, 437, 10.1111/j.1365-2605.2012.01247.x
Janesick, 2011, Endocrine disrupting chemicals and the developmental programming of adipogenesis and obesity, Birth Defects Res C Embryo Today, 93, 34, 10.1002/bdrc.20197
Casals-Casas, 2011, Endocrine disruptors: from endocrine to metabolic disruption, Annu Rev Physiol, 73, 135, 10.1146/annurev-physiol-012110-142200
Thayer, 2012, Role of environmental chemicals in diabetes and obesity: a National Toxicology Program workshop review, Environ Health Perspect, 120, 779, 10.1289/ehp.1104597
Behl, 2013, Evaluation of the association between maternal smoking, childhood obesity, and metabolic disorders: a National Toxicology Program workshop review, Environ Health Perspect, 121, 170, 10.1289/ehp.1205404
Maull, 2012, Evaluation of the association between arsenic and diabetes: a National Toxicology Program workshop review, Environ Health Perspect, 120, 1658, 10.1289/ehp.1104579
Taylor, 2013, Evaluation of the association between persistent organic pollutants (POPs) and diabetes in epidemiological studies: a National Toxicology Program workshop review, Environ Health Perspect, 121, 774, 10.1289/ehp.1205502
Carwile, 2011, Urinary bisphenol A and obesity: NHANES 2003–2006, Environ Res, 111, 825, 10.1016/j.envres.2011.05.014
Shankar, 2012, Urinary bisphenol A levels and measures of obesity: results from the National Health and Nutrition Examination Survey 2003–2008, ISRN Endocrinol, 2012, 965243, 10.5402/2012/965243
Trasande, 2012, Association between urinary bisphenol A concentration and obesity prevalence in children and adolescents, JAMA, 308, 1113, 10.1001/2012.jama.11461
Bhandari, 2013, Urinary bisphenol A and obesity in U.S. children, Am J Epidemiol, 177, 1263, 10.1093/aje/kws391
Wang, 2012, Urinary bisphenol A (BPA) concentration associates with obesity and insulin resistance, J Clin Endocrinol Metab, 97, E223, 10.1210/jc.2011-1989
Harley, 2013, Prenatal and postnatal bisphenol A exposure and body mass index in childhood in the CHAMACOS cohort, Environ Health Perspect, 121, 514, 10.1289/ehp.1205548
Stahlhut, 2007, Concentrations of urinary phthalate metabolites are associated with increased waist circumference and insulin resistance in adult U.S. males, Environ Health Perspect, 115, 876, 10.1289/ehp.9882
Hatch, 2008, Association of urinary phthalate metabolite concentrations with body mass index and waist circumference: a cross-sectional study of NHANES data, 1999–2002, Environ Health, 7, 27, 10.1186/1476-069X-7-27
Lind, 2012, Serum concentrations of phthalate metabolites are related to abdominal fat distribution two years later in elderly women, Environ Health, 11, 21, 10.1186/1476-069X-11-21
Wang, 2013, Urinary phthalate metabolites are associated with body mass index and waist circumference in Chinese school children, PLoS One, 8, e56800, 10.1371/journal.pone.0056800
Teitelbaum, 2012, Associations between phthalate metabolite urinary concentrations and body size measures in New York City children, Environ Res, 112, 186, 10.1016/j.envres.2011.12.006
Trasande, 2013, Race/ethnicity-specific associations of urinary phthalates with childhood body mass in a nationally representative sample, Environ Health Perspect, 121, 501, 10.1289/ehp.1205526
Elobeid, 2010, Endocrine disruptors and obesity: an examination of selected persistent organic pollutants in the NHANES 1999–2002 data, Int J Environ Res Public Health, 7, 2988, 10.3390/ijerph7072988
Halldorsson, 2012, Prenatal exposure to perfluorooctanoate and risk of overweight at 20 years of age: a prospective cohort study, Environ Health Perspect, 120, 668, 10.1289/ehp.1104034
Lee, 2014, Chlorinated persistent organic pollutants, obesity, and type 2 diabetes, Endocr Rev, 35, 557, 10.1210/er.2013-1084
Kanayama, 2005, Organotin compounds promote adipocyte differentiation as agonists of the peroxisome proliferator-activated receptor γ/retinoid X receptor pathway, Mol Pharmacol, 67, 766, 10.1124/mol.104.008409
Tontonoz, 2008, Fat and beyond: the diverse biology of PPARγ, Annu Rev Biochem, 77, 289, 10.1146/annurev.biochem.77.061307.091829
Janesick, 2014, Transgenerational inheritance of prenatal obesogen exposure, Mol Cell Endocrinol, 398, 31, 10.1016/j.mce.2014.09.002
Hiromori, 2009, Structure-dependent activation of peroxisome proliferator-activated receptor (PPAR) γ by organotin compounds, Chem Biol Interact, 180, 238, 10.1016/j.cbi.2009.03.006
Hurst, 2003, Activation of PPARα and PPARγ by environmental phthalate monoesters, Toxicol Sci, 74, 297, 10.1093/toxsci/kfg145
Hectors, 2013, Evaluation of the INS-1 832/13 cell line as a β-cell based screening system to assess pollutant effects on β-cell function, PLoS One, 8, e60030, 10.1371/journal.pone.0060030
Hu, 2013, Effects of parabens on adipocyte differentiation, Toxicol Sci, 131, 56, 10.1093/toxsci/kfs262
Hao, 2012, The endocrine disruptor 4-nonylphenol promotes adipocyte differentiation and induces obesity in mice, Cell Physiol Biochem, 30, 382, 10.1159/000339032
Pereira-Fernandes, 2013, Evaluation of a screening system for obesogenic compounds: screening of endocrine disrupting compounds and evaluation of the PPAR dependency of the effect, PLoS One, 8, e77481, 10.1371/journal.pone.0077481
Biemann, 2012, Endocrine disrupting chemicals affect the adipogenic differentiation of mesenchymal stem cells in distinct ontogenetic windows, Biochem Biophys Res Commun, 417, 747, 10.1016/j.bbrc.2011.12.028
Li, 2012, Triflumizole is an obesogen in mice that acts through peroxisome proliferator activated receptor γ (PPARγ), Environ Health Perspect, 120, 1720, 10.1289/ehp.1205383
Zhang, 2012, Fetal exposure to bisphenol A affects the primordial follicle formation by inhibiting the meiotic progression of oocytes, Mol Biol Rep, 39, 5651, 10.1007/s11033-011-1372-3
Masuno, 2005, Bisphenol A accelerates terminal differentiation of 3T3–L1 cells into adipocytes through the phosphatidylinositol 3-kinase pathway, Toxicol Sci, 84, 319, 10.1093/toxsci/kfi088
Valentino, 2013, Bisphenol-A impairs insulin action and up-regulates inflammatory pathways in human subcutaneous adipocytes and 3T3–L1 cells, PLoS One, 8, e82099, 10.1371/journal.pone.0082099
Ohlstein, 2014, Bisphenol A enhances adipogenic differentiation of human adipose stromal/stem cells, J Mol Endocrinol, 53, 345, 10.1530/JME-14-0052
Chamorro-García, 2012, Bisphenol A diglycidyl ether induces adipogenic differentiation of multipotent stromal stem cells through a peroxisome proliferator-activated receptor γ-independent mechanism, Environ Health Perspect, 120, 984, 10.1289/ehp.1205063
Wang, 2013, The environmental obesogen bisphenol A promotes adipogenesis by increasing the amount of 11β-hydroxysteroid dehydrogenase type 1 in the adipose tissue of children, Int J Obes (Lond), 37, 999, 10.1038/ijo.2012.173
Sargis, 2010, Environmental endocrine disruptors promote adipogenesis in the 3T3–L1 cell line through glucocorticoid receptor activation, Obesity (Silver Spring), 18, 1283, 10.1038/oby.2009.419
Neel, 2013, The endocrine disrupting chemical tolylfluanid alters adipocyte metabolism via glucocorticoid receptor activation, Mol Endocrinol, 27, 394, 10.1210/me.2012-1270
Arsenescu, 2008, Polychlorinated biphenyl-77 induces adipocyte differentiation and proinflammatory adipokines and promotes obesity and atherosclerosis, Environ Health Perspect, 116, 761, 10.1289/ehp.10554
Kamstra, 2014, Transcriptional and epigenetic mechanisms underlying enhanced in vitro adipocyte differentiation by the brominated flame retardant BDE-47, Environ Sci Technol, 48, 4110, 10.1021/es405524b
Regnier, 2014, Adipocytes under assault: environmental disruption of adipose physiology, Biochim Biophys Acta, 1842, 520, 10.1016/j.bbadis.2013.05.028
La Merrill, 2013, Toxicological function of adipose tissue: focus on persistent organic pollutants, Environ Health Perspect, 121, 162, 10.1289/ehp.1205485
Heine, 2000, Increased adipose tissue in male and female estrogen receptor-α knockout mice, Proc Natl Acad Sci USA, 97, 12729, 10.1073/pnas.97.23.12729
Park, 2011, Genetic rescue of nonclassical ERα signaling normalizes energy balance in obese Erα-null mutant mice, J Clin Invest, 121, 604, 10.1172/JCI41702
Barros, 2011, Estrogen receptors and the metabolic network, Cell Metab, 14, 289, 10.1016/j.cmet.2011.08.005
Ropero, 2008, The role of estrogen receptors in the control of energy and glucose homeostasis, Steroids, 73, 874, 10.1016/j.steroids.2007.12.018
Newbold, 2010, Impact of environmental endocrine disrupting chemicals on the development of obesity, Hormones (Athens), 9, 206, 10.14310/horm.2002.1271
Akingbemi, 2004, Inhibition of testicular steroidogenesis by the xenoestrogen bisphenol A is associated with reduced pituitary luteinizing hormone secretion and decreased steroidogenic enzyme gene expression in rat Leydig cells, Endocrinology, 145, 592, 10.1210/en.2003-1174
Miyawaki, 2007, Perinatal and postnatal exposure to bisphenol A increases adipose tissue mass and serum cholesterol level in mice, J Atheroscler Thromb, 14, 245, 10.5551/jat.E486
Patisaul, 2008, Neonatal exposure to endocrine active compounds or an ERβ agonist increases adult anxiety and aggression in gonadally intact male rats, Horm Behav, 53, 580, 10.1016/j.yhbeh.2008.01.008
Rubin, 2009, Bisphenol A: perinatal exposure and body weight, Mol Cell Endocrinol, 304, 55, 10.1016/j.mce.2009.02.023
Somm, 2009, Perinatal exposure to bisphenol A alters early adipogenesis in the rat, Environ Health Perspect, 117, 1549, 10.1289/ehp.11342
García-Arevalo, 2014, Exposure to bisphenol-A during pregnancy partially mimics the effects of a high-fat diet altering glucose homeostasis and gene expression in adult male mice, PLoS One, 9, e100214, 10.1371/journal.pone.0100214
Vom Saal, 2012, The estrogenic endocrine disrupting chemical bisphenol A (BPA) and obesity, Mol Cell Endocrinol, 354, 74, 10.1016/j.mce.2012.01.001
Batista, 2012, Short-term treatment with bisphenol-A leads to metabolic abnormalities in adult male mice, PLoS One, 7, e33814, 10.1371/journal.pone.0033814
Mackay, 2013, Organizational effects of perinatal exposure to bisphenol-A and diethylstilbestrol on arcuate nucleus circuitry controlling food intake and energy expenditure in male and female CD-1 mice, Endocrinology, 154, 1465, 10.1210/en.2012-2044
Ryan, 2010, Perinatal exposure to bisphenol-A and the development of metabolic syndrome in CD-1 mice, Endocrinology, 151, 2603, 10.1210/en.2009-1218
Ryan, 2006, Developmental exposure to environmental estrogens alters anxiety and spatial memory in female mice, Horm Behav, 50, 85, 10.1016/j.yhbeh.2006.01.007
Nakamura, 2012, Prenatal and lactational exposure to low-doses of bisphenol A alters adult mice behavior, Brain Dev, 34, 57, 10.1016/j.braindev.2010.12.011
van Esterik, 2014, Programming of metabolic effects in C57BL/6JxFVB mice by exposure to bisphenol A during gestation and lactation, Toxicology, 321, 40, 10.1016/j.tox.2014.04.001
Tyl, 2002, Three-generation reproductive toxicity study of dietary bisphenol A in CD Sprague-Dawley rats, Toxicol Sci, 68, 121, 10.1093/toxsci/68.1.121
Kirchner, 2010, Prenatal exposure to the environmental obesogen tributyltin predisposes multipotent stem cells to become adipocytes, Mol Endocrinol, 24, 526, 10.1210/me.2009-0261
Decherf, 2010, Disruption of thyroid hormone-dependent hypothalamic set-points by environmental contaminants, Mol Cell Endocrinol, 323, 172, 10.1016/j.mce.2010.04.010
Decherf, 2011, The obesogen hypothesis: a shift of focus from the periphery to the hypothalamus, J Toxicol Environ Health B Crit Rev, 14, 423, 10.1080/10937404.2011.578561
Riu, 2014, Halogenated bisphenol-A analogs act as obesogens in zebrafish larvae (Danio rerio), Toxicol Sci, 139, 48, 10.1093/toxsci/kfu036
Takacs, 2007, Activation of mouse and human peroxisome proliferator-activated receptors (α, β/δ, γ) by perfluorooctanoic acid and perfluorooctane sulfonate, Toxicol Sci, 95, 108, 10.1093/toxsci/kfl135
Vitalone, 2010, Long-term effects of developmental exposure to low doses of PCB 126 and methylmercury, Toxicol Lett, 197, 38, 10.1016/j.toxlet.2010.04.024
Slotkin, 2011, Does early-life exposure to organophosphate insecticides lead to prediabetes and obesity?, Reprod Toxicol, 31, 297, 10.1016/j.reprotox.2010.07.012
Zhu, 2008, Effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin administration and high-fat diet on the body weight and hepatic estrogen metabolism in female C3H/HeN mice, Toxicol Appl Pharmacol, 226, 107, 10.1016/j.taap.2007.08.018
La Merrill, 2014, Perinatal exposure of mice to the pesticide DDT impairs energy expenditure and metabolism in adult female offspring, PLoS One, 9, e103337, 10.1371/journal.pone.0103337
Feil, 2011, Epigenetics and the environment: emerging patterns and implications, Nat Rev Genet, 13, 97, 10.1038/nrg3142
Manikkam, 2013, Plastics derived endocrine disruptors (BPA, DEHP and DBP) induce epigenetic transgenerational inheritance of obesity, reproductive disease and sperm epimutations, PLoS One, 8, e55387, 10.1371/journal.pone.0055387
Skinner, 2013, Ancestral dichlorodiphenyltrichloroethane (DDT) exposure promotes epigenetic transgenerational inheritance of obesity, BMC Med, 11, 228, 10.1186/1741-7015-11-228
American Diabetes Association, 2004, Diagnosis and classification of diabetes mellitus, Diabetes Care, 27, S5, 10.2337/diacare.27.2007.S5
Lazar, 2005, How obesity causes diabetes: not a tall tale, Science, 307, 373, 10.1126/science.1104342
Virtue, 2008, It's not how fat you are, it's what you do with it that counts, PLoS Biol, 6, e237, 10.1371/journal.pbio.0060237
Alonso-Magdalena, 2011, Endocrine disruptors in the etiology of type 2 diabetes mellitus, Nat Rev Endocrinol, 7, 346, 10.1038/nrendo.2011.56
Ruderman, 1998, The metabolically obese, normal-weight individual revisited, Diabetes, 47, 699, 10.2337/diabetes.47.5.699
Yoon, 2006, Epidemic obesity and type 2 diabetes in Asia, Lancet, 368, 1681, 10.1016/S0140-6736(06)69703-1
Chen, 2012, The worldwide epidemiology of type 2 diabetes mellitus–present and future perspectives, Nat Rev Endocrinol, 8, 228, 10.1038/nrendo.2011.183
Kuo, 2013, Environmental chemicals and type 2 diabetes: an updated systematic review of the epidemiologic evidence, Curr Diab Rep, 13, 831, 10.1007/s11892-013-0432-6
Magliano, 2014, Persistent organic pollutants and diabetes: a review of the epidemiological evidence, Diabetes Metab, 40, 1, 10.1016/j.diabet.2013.09.006
Rignell-Hydbom, 2009, Exposure to p,p'-DDE: a risk factor for type 2 diabetes, PLoS One, 4, e7503, 10.1371/journal.pone.0007503
Turyk, 2009, Organochlorine exposure and incidence of diabetes in a cohort of Great Lakes sport fish consumers, Environ Health Perspect, 117, 1076, 10.1289/ehp.0800281
Lee, 2010, Low dose of some persistent organic pollutants predicts type 2 diabetes: a nested case-control study, Environ Health Perspect, 118, 1235, 10.1289/ehp.0901480
Lee, 2011, Polychlorinated biphenyls and organochlorine pesticides in plasma predict development of type 2 diabetes in the elderly: the prospective investigation of the vasculature in Uppsala Seniors (PIVUS) study, Diabetes Care, 34, 1778, 10.2337/dc10-2116
Wang, 2008, Increased risk of diabetes and polychlorinated biphenyls and dioxins: a 24-year follow-up study of the Yucheng cohort, Diabetes Care, 31, 1574, 10.2337/dc07-2449
Wu, 2013, Persistent organic pollutants and type 2 diabetes: a prospective analysis in the Nurses' Health Study and meta-analysis, Environ Health Perspect, 121, 153, 10.1289/ehp.1205248
Shankar, 2011, Relationship between urinary bisphenol A levels and diabetes mellitus, J Clin Endocrinol Metab, 96, 3822, 10.1210/jc.2011-1682
Beydoun, 2014, Sex differences in the association of urinary bisphenol-A concentration with selected indices of glucose homeostasis among U.S. adults, Ann Epidemiol, 24, 90, 10.1016/j.annepidem.2013.07.014
Melzer, 2010, Association of urinary bisphenol A concentration with heart disease: evidence from NHANES 2003/06, PLoS One, 5, e8673, 10.1371/journal.pone.0008673
Silver, 2011, Urinary bisphenol A and type-2 diabetes in U.S. adults: data from NHANES 2003–2008, PLoS One, 6, e26868, 10.1371/journal.pone.0026868
Sun, 2014, Association of urinary concentrations of bisphenol A and phthalate metabolites with risk of type 2 diabetes: a prospective investigation in the Nurses' Health Study (NHS) and NHSII cohorts, Environ Health Perspect, 122, 616, 10.1289/ehp.1307201
LaKind, 2012, Use of NHANES data to link chemical exposures to chronic diseases: a cautionary tale, PLoS One, 7, e51086, 10.1371/journal.pone.0051086
Ruzzin, 2010, Persistent organic pollutant exposure leads to insulin resistance syndrome, Environ Health Perspect, 118, 465, 10.1289/ehp.0901321
Novelli, 2005, 2,3,7,8-Tetrachlorodibenzo-p-dioxin-induced impairment of glucose-stimulated insulin secretion in isolated rat pancreatic islets, Toxicol Lett, 156, 307, 10.1016/j.toxlet.2004.12.004
Kurita, 2009, Aryl hydrocarbon receptor-mediated effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on glucose-stimulated insulin secretion in mice, J Appl Toxicol, 29, 689, 10.1002/jat.1459
Wang, 2011, Aryl hydrocarbon receptor deficiency enhances insulin sensitivity and reduces PPAR-α pathway activity in mice, Environ Health Perspect, 119, 1739, 10.1289/ehp.1103593
Kim, 2012, Inflammatory pathway genes belong to major targets of persistent organic pollutants in adipose cells, Environ Health Perspect, 120, 508, 10.1289/ehp.1104282
Hardy, 2012, What causes the insulin resistance underlying obesity?, Curr Opin Endocrinol Diabetes Obes, 19, 81, 10.1097/MED.0b013e3283514e13
Hugo, 2008, Bisphenol A at environmentally relevant doses inhibits adiponectin release from human adipose tissue explants and adipocytes, Environ Health Perspect, 116, 1642, 10.1289/ehp.11537
Grasselli, 2013, Direct effects of bisphenol A on lipid homeostasis in rat hepatoma cells, Chemosphere, 91, 1123, 10.1016/j.chemosphere.2013.01.016
Soriano, 2012, Rapid insulinotropic action of low doses of bisphenol-A on mouse and human islets of Langerhans: role of estrogen receptor β, PLoS One, 7, e31109, 10.1371/journal.pone.0031109
Alonso-Magdalena, 2005, Low doses of bisphenol A and diethylstilbestrol impair Ca2+ signals in pancreatic α-cells through a nonclassical membrane estrogen receptor within intact islets of Langerhans, Environ Health Perspect, 113, 969, 10.1289/ehp.8002
Alonso-Magdalena, 2012, Bisphenol-A acts as a potent estrogen via non-classical estrogen triggered pathways, Mol Cell Endocrinol, 355, 201, 10.1016/j.mce.2011.12.012
Díaz-Villaseñor, 2006, Sodium arsenite impairs insulin secretion and transcription in pancreatic β-cells, Toxicol Appl Pharmacol, 214, 30, 10.1016/j.taap.2005.11.015
Douillet, 2013, Methylated trivalent arsenicals are potent inhibitors of glucose stimulated insulin secretion by murine pancreatic islets, Toxicol Appl Pharmacol, 267, 11, 10.1016/j.taap.2012.12.007
Chen, 2010, Inorganic mercury causes pancreatic β-cell death via the oxidative stress-induced apoptotic and necrotic pathways, Toxicol Appl Pharmacol, 243, 323, 10.1016/j.taap.2009.11.024
Chen, 2009, Heavy metals, islet function and diabetes development, Islets, 1, 169, 10.4161/isl.1.3.9262
Hectors, 2011, Environmental pollutants and type 2 diabetes: a review of mechanisms that can disrupt β cell function, Diabetologia, 54, 1273, 10.1007/s00125-011-2109-5
Neel, 2011, The paradox of progress: environmental disruption of metabolism and the diabetes epidemic, Diabetes, 60, 1838, 10.2337/db11-0153
Hectors, 2013, Insulin resistance and environmental pollutants: experimental evidence and future perspectives, Environ Health Perspect, 121, 1273, 10.1289/ehp.1307082
Ibrahim, 2011, Chronic consumption of farmed salmon containing persistent organic pollutants causes insulin resistance and obesity in mice, PLoS One, 6, e25170, 10.1371/journal.pone.0025170
Wan, 2014, Perinatal exposure to perfluorooctane sulfonate affects glucose metabolism in adult offspring, PLoS One, 9, e87137, 10.1371/journal.pone.0087137
Yang, 2014, Environmentally relevant levels of bisphenol A may accelerate the development of type II diabetes mellitus in adolescent Otsuka Long Evans Tokushima Fatty rats, J Toxicol Environ Health Sci, 6, 41, 10.1007/s13530-014-0186-9
Marmugi, 2012, Low doses of bisphenol A induce gene expression related to lipid synthesis and trigger triglyceride accumulation in adult mouse liver, Hepatology, 55, 395, 10.1002/hep.24685
Perreault, 2013, Bisphenol A impairs hepatic glucose sensing in C57BL/6 male mice, PLoS One, 8, e69991, 10.1371/journal.pone.0069991
Zuo, 2014, Chronic exposure to tributyltin chloride induces pancreatic islet cell apoptosis and disrupts glucose homeostasis in male mice, Environ Sci Technol, 48, 5179, 10.1021/es404729p
Cabaton, 2013, Effects of low doses of bisphenol A on the metabolome of perinatally exposed CD-1 mice, Environ Health Perspect, 121, 586, 10.1289/ehp.1205588
Ma, 2013, Hepatic DNA methylation modifications in early development of rats resulting from perinatal BPA exposure contribute to insulin resistance in adulthood, Diabetologia, 56, 2059, 10.1007/s00125-013-2944-7
Li, 2014, F0 maternal BPA exposure induced glucose intolerance of F2 generation through DNA methylation change in Gck, Toxicol Lett, 228, 192, 10.1016/j.toxlet.2014.04.012
Lin, 2011, Developmental exposure to di(2-ethylhexyl) phthalate impairs endocrine pancreas and leads to long-term adverse effects on glucose homeostasis in the rat, Am J Physiol Endocrinol Metab, 301, E527, 10.1152/ajpendo.00233.2011
Naville, 2013, Low-dose food contaminants trigger sex-specific, hepatic metabolic changes in the progeny of obese mice, FASEB J, 27, 3860, 10.1096/fj.13-231670
Eizirik, 2009, The role of inflammation in insulitis and β-cell loss in type 1 diabetes, Nat Rev Endocrinol, 5, 219, 10.1038/nrendo.2009.21
Tuomi, 2005, Type 1 and type 2 diabetes: what do they have in common?, Diabetes, 54, S40, 10.2337/diabetes.54.suppl_2.S40
Fourlanos, 2008, The accelerator hypothesis and increasing incidence of type 1 diabetes, Curr Opin Endocrinol Diabetes Obes, 15, 321, 10.1097/MED.0b013e3283073a5a
Howard, 2011, Environmental pollutants and β cell function: relevance for type 1 and gestational diabetes, Diabetologia, 54, 3168, 10.1007/s00125-011-2318-y
Howard, 2012, What is the role of human contamination by environmental chemicals in the development of type 1 diabetes?, J Epidemiol Community Health, 66, 479, 10.1136/jech.2011.133694
Bodin, 2014, Transmaternal bisphenol A exposure accelerates diabetes type 1 development in NOD mice, Toxicol Sci, 137, 311, 10.1093/toxsci/kft242
Humblet, 2008, Dioxins and cardiovascular disease mortality, Environ Health Perspect, 116, 1443, 10.1289/ehp.11579
Min, 2011, Potential role for organochlorine pesticides in the prevalence of peripheral arterial diseases in obese persons: results from the National Health and Nutrition Examination Survey 1999–2004, Atherosclerosis, 218, 200, 10.1016/j.atherosclerosis.2011.04.044
Wu, 2011, Activation of aryl hydrocarbon receptor induces vascular inflammation and promotes atherosclerosis in apolipoprotein E−/− mice, Arterioscler Thromb Vasc Biol, 31, 1260, 10.1161/ATVBAHA.110.220202
Aragon, 2008, Perinatal 2,3,7,8-tetrachlorodibenzo-p-dioxin exposure sensitizes offspring to angiotensin II-induced hypertension, Cardiovasc Toxicol, 8, 145, 10.1007/s12012-008-9023-1
La Merrill, 2013, Prenatal exposure to the pesticide DDT and hypertension diagnosed in women before age 50: a longitudinal birth cohort study, Environ Health Perspect, 121, 594, 10.1289/ehp.1205921
Siddiqui, 2002, Association of maternal blood pressure and hemoglobin level with organochlorines in human milk, Hum Exp Toxicol, 21, 1, 10.1191/0960327102ht198oa
Yan, 2011, Bisphenol A and 17β-estradiol promote arrhythmia in the female heart via alteration of calcium handling, PLoS One, 6, e25455, 10.1371/journal.pone.0025455
Kim, 2013, Acute cardiovascular toxicity of sterilizers, PHMG, and PGH: severe inflammation in human cells and heart failure in zebrafish, Cardiovasc Toxicol, 13, 148, 10.1007/s12012-012-9193-8
Saura, 2014, Oral administration of bisphenol A induces high blood pressure through angiotensin II/CaMKII-dependent uncoupling of eNOS, FASEB J, 28, 4719, 10.1096/fj.14-252460
Posnack, 2014, Bisphenol A exposure and cardiac electrical conduction in excised rat hearts, Environ Health Perspect, 122, 384, 10.1289/ehp.1206157
Melzer, 2012, Urinary bisphenol A concentration and risk of future coronary artery disease in apparently healthy men and women, Circulation, 125, 1482, 10.1161/CIRCULATIONAHA.111.069153
Bae, 2012, Associations of bisphenol A exposure with heart rate variability and blood pressure, Hypertension, 60, 786, 10.1161/HYPERTENSIONAHA.112.197715
Bae, 2015, Exposure to bisphenol A from drinking canned beverages increases blood pressure: randomized crossover trial, Hypertension, 65, 313, 10.1161/HYPERTENSIONAHA.114.04261
Mehran, 2012, Hyperinsulinemia drives diet-induced obesity independently of brain insulin production, Cell Metab, 16, 723, 10.1016/j.cmet.2012.10.019
Sharpe, 2013, Obesogens and obesity–an alternative view?, Obesity (Silver Spring), 21, 1081, 10.1002/oby.20373
Meeker, 2012, Exposure to environmental endocrine disruptors and child development, Arch Pediatr Adolesc Med, 166, 952, 10.1001/archpediatrics.2012.241
Martino-Andrade, 2010, Reproductive toxicity of phthalate esters, Mol Nutr Food Res, 54, 148, 10.1002/mnfr.200800312
Ashiru, 2009, Fertility and occupational hazards: review of the literature, Afr J Reprod Health, 13, 159
Weinhold, 2012, More chemicals show epigenetic effects across generations, Environ Health Perspect, 120, A228, 10.1289/ehp.120-a228
Bellelis, 2011, Environmental factors and endometriosis, Rev Assoc Med Bras, 57, 448, 10.1016/S0104-4230(11)70093-8
Kay, 2013, Reproductive and developmental effects of phthalate diesters in females, Crit Rev Toxicol, 43, 200, 10.3109/10408444.2013.766149
Balabanič, 2011, Negative impact of endocrine-disrupting compounds on human reproductive health, Reprod Fertil Dev, 23, 403, 10.1071/RD09300
Craig, 2011, Endocrine-disrupting chemicals in ovarian function: effects on steroidogenesis, metabolism and nuclear receptor signaling, Reproduction, 142, 633, 10.1530/REP-11-0136
Kadhel, 2012, Organochlorine pollutants and female fertility: a systematic review focusing on in vitro fertilization studies, Reprod Sci, 19, 1246, 10.1177/1933719112446077
Peretz, 2014, Bisphenol A and reproductive health: update of experimental and human evidence, 2007–2013, Environ Health Perspect, 122, 775, 10.1289/ehp.1307728
Caserta, 2014, Bisphenol A and the female reproductive tract: an overview of recent laboratory evidence and epidemiological studies, Reprod Biol Endocrinol, 12, 37, 10.1186/1477-7827-12-37
Rivera, 2011, Neonatal exposure to bisphenol A or diethylstilbestrol alters the ovarian follicular dynamics in the lamb, Reprod Toxicol, 32, 304, 10.1016/j.reprotox.2011.06.118
Hunt, 2012, Bisphenol A alters early oogenesis and follicle formation in the fetal ovary of the rhesus monkey, Proc Natl Acad Sci USA, 109, 17525, 10.1073/pnas.1207854109
Wang, 2014, In utero bisphenol A exposure disrupts germ cell nest breakdown and reduces fertility with age in the mouse, Toxicol Appl Pharmacol, 276, 157, 10.1016/j.taap.2014.02.009
Weinberger, 2014, Effects of maternal exposure to phthalates and bisphenol A during pregnancy on gestational age, J Matern Fetal Neonatal Med, 27, 323, 10.3109/14767058.2013.815718
Lawson, 2011, Gene expression in the fetal mouse ovary is altered by exposure to low doses of bisphenol A, Biol Reprod, 84, 79, 10.1095/biolreprod.110.084814
Zhang, 2012, Bisphenol A exposure modifies DNA methylation of imprint genes in mouse fetal germ cells, Mol Biol Rep, 39, 8621, 10.1007/s11033-012-1716-7
Brieño-Enríquez, 2011, Human meiotic progression and recombination are affected by bisphenol A exposure during in vitro human oocyte development, Hum Reprod, 26, 2807, 10.1093/humrep/der249
Brieño-Enríquez, 2012, Gene expression is altered after bisphenol A exposure in human fetal oocytes in vitro, Mol Hum Reprod, 18, 171, 10.1093/molehr/gar074
Trapphoff, 2013, Chronic exposure to a low concentration of bisphenol A during follicle culture affects the epigenetic status of germinal vesicles and metaphase II oocytes, Fertil Steril, 100, 1758, 10.1016/j.fertnstert.2013.08.021
Muczynski, 2012, Cellular and molecular effect of MEHP involving LXRα in human fetal testis and ovary, PLoS One, 7, e48266, 10.1371/journal.pone.0048266
Bonilla, 2010, Deregulation of the Sod1 and Nd1 genes in mouse fetal oocytes exposed to mono-(2-ethylhexyl) phthalate (MEHP), Reprod Toxicol, 30, 387, 10.1016/j.reprotox.2010.04.008
Zhang, 2014, Di-(2-ethylhexyl) phthalate and bisphenol A exposure impairs mouse primordial follicle assembly in vitro, Environ Mol Mutagen, 55, 343, 10.1002/em.21847
Sobinoff, 2010, Adding insult to injury: effects of xenobiotic-induced preantral ovotoxicity on ovarian development and oocyte fusibility, Toxicol Sci, 118, 653, 10.1093/toxsci/kfq272
Takeda, 2011, 2,3,7,8-Tetrachlorodibenzo-p-dioxin potentially attenuates the gene expression of pituitary gonadotropin β-subunits in a fetal age-specific fashion: a comparative study using cultured pituitaries, J Toxicol Sci, 36, 221, 10.2131/jts.36.221
Rodríguez, 2010, Neonatal exposure to bisphenol A reduces the pool of primordial follicles in the rat ovary, Reprod Toxicol, 30, 550, 10.1016/j.reprotox.2010.07.008
Li, 2014, Prepubertal bisphenol A exposure interferes with ovarian follicle development and its relevant gene expression, Reprod Toxicol, 44, 33, 10.1016/j.reprotox.2013.09.002
Delclos, 2014, Toxicity evaluation of bisphenol A administered by gavage to Sprague Dawley rats from gestation day 6 through postnatal day 90, Toxicol Sci, 139, 174, 10.1093/toxsci/kfu022
Peretz, 2011, Bisphenol A impairs follicle growth, inhibits steroidogenesis, and downregulates rate-limiting enzymes in the estradiol biosynthesis pathway, Toxicol Sci, 119, 209, 10.1093/toxsci/kfq319
Peretz, 2012, Bisphenol A inhibits follicle growth and induces atresia in cultured mouse antral follicles independently of the genomic estrogenic pathway, Biol Reprod, 87, 63, 10.1095/biolreprod.112.101899
Ziv-Gal, 2013, Bisphenol A inhibits cultured mouse ovarian follicle growth partially via the aryl hydrocarbon receptor signaling pathway, Reprod Toxicol, 42, 58, 10.1016/j.reprotox.2013.07.022
Peretz, 2013, Mouse strain does not influence the overall effects of bisphenol A-induced toxicity in adult antral follicles, Biol Reprod, 89, 108, 10.1095/biolreprod.113.111864
Pollock, 2014, Triclosan exacerbates the presence of 14C-bisphenol A in tissues of female and male mice, Toxicol Appl Pharmacol, 278, 116, 10.1016/j.taap.2014.04.017
Popa, 2014, Influence of Genista tinctoria L. or methylparaben on subchronic toxicity of bisphenol A in rats, Biomed Environ Sci, 27, 85
Chen, 2012, Benzyl butyl phthalate induces necrosis by AhR mediation of CYP1B1 expression in human granulosa cells, Reprod Toxicol, 33, 67, 10.1016/j.reprotox.2011.11.004
Li, 2012, Di-(2-ethylhexyl) phthalate reduces progesterone levels and induces apoptosis of ovarian granulosa cell in adult female ICR mice, Environ Toxicol Pharmacol, 34, 869, 10.1016/j.etap.2012.08.013
Hannon, 2014, Daily exposure to di(2-ethylhexyl) phthalate alters estrous cyclicity and accelerates primordial follicle recruitment potentially via dysregulation of the phosphatidylinositol 3-kinase signaling pathway in adult mice, Biol Reprod, 90, 136, 10.1095/biolreprod.114.119032
Xu, 2010, Ovotoxicity and PPAR-mediated aromatase downregulation in female Sprague-Dawley rats following combined oral exposure to benzo[a]pyrene and di-(2-ethylhexyl) phthalate, Toxicol Lett, 199, 323, 10.1016/j.toxlet.2010.09.015
Gupta, 2010, Di-(2-ethylhexyl) phthalate and mono-(2-ethylhexyl) phthalate inhibit growth and reduce estradiol levels of antral follicles in vitro, Toxicol Appl Pharmacol, 242, 224, 10.1016/j.taap.2009.10.011
Inada, 2012, Evaluation of ovarian toxicity of mono-(2-ethylhexyl) phthalate (MEHP) using cultured rat ovarian follicles, J Toxicol Sci, 37, 483, 10.2131/jts.37.483
Wang, 2012, Mono-(2-ethylhexyl) phthalate induces oxidative stress and inhibits growth of mouse ovarian antral follicles, Biol Reprod, 87, 152
Wang, 2012, Di (2-ethylhexyl) phthalate inhibits growth of mouse ovarian antral follicles through an oxidative stress pathway, Toxicol Appl Pharmacol, 258, 288, 10.1016/j.taap.2011.11.008
Craig, 2014, Co-treatment of mouse antral follicles with 17β-estradiol interferes with mono-2-ethylhexyl phthalate (MEHP)-induced atresia and altered apoptosis gene expression, Reprod Toxicol, 45, 45, 10.1016/j.reprotox.2014.01.002
Ambruosi, 2011, In vitro acute exposure to DEHP affects oocyte meiotic maturation, energy and oxidative stress parameters in a large animal model, PLoS One, 6, e27452, 10.1371/journal.pone.0027452
Craig, 2013, Di-n-butyl phthalate disrupts the expression of genes involved in cell cycle and apoptotic pathways in mouse ovarian antral follicles, Biol Reprod, 88, 23, 10.1095/biolreprod.112.105122
Aoyama, 2012, Two-generation reproduction toxicity study in rats with methoxychlor, Congenit Anom (Kyoto), 52, 28, 10.1111/j.1741-4520.2011.00344.x
Aoyama, 2014, Reproductive toxicities of methoxychlor based on estrogenic properties of the compound and its estrogenic metabolite, hydroxyphenyltrichloroethane, Vitam Horm, 94, 193, 10.1016/B978-0-12-800095-3.00007-9
Basavarajappa, 2012, Methoxychlor inhibits growth and induces atresia through the aryl hydrocarbon receptor pathway in mouse ovarian antral follicles, Reprod Toxicol, 34, 16, 10.1016/j.reprotox.2012.03.007
Gupta, 2009, Methoxychlor inhibits growth of antral follicles by altering cell cycle regulators, Toxicol Appl Pharmacol, 240, 1, 10.1016/j.taap.2009.07.007
Paulose, 2011, Increased sensitivity of estrogen receptor α overexpressing antral follicles to methoxychlor and its metabolites, Toxicol Sci, 120, 447, 10.1093/toxsci/kfr011
Miller, 2005, Methoxychlor directly affects ovarian antral follicle growth and atresia through Bcl-2- and Bax-mediated pathways, Toxicol Sci, 88, 213, 10.1093/toxsci/kfi276
Paulose, 2012, Methoxychlor-induced ovarian follicle toxicity in mice: dose and exposure duration-dependent effects, Birth Defects Res B Dev Reprod Toxicol, 95, 219, 10.1002/bdrb.21007
Borgeest, 2004, Methoxychlor-induced atresia in the mouse involves Bcl-2 family members, but not gonadotropins or estradiol, Biol Reprod, 70, 1828, 10.1095/biolreprod.103.022889
Gupta, 2007, Methoxychlor and its metabolites inhibit growth and induce atresia of baboon antral follicles, Toxicol Pathol, 35, 649, 10.1080/01926230701459960
Paulose, 2012, Estrogen receptor α overexpressing mouse antral follicles are sensitive to atresia induced by methoxychlor and its metabolites, Reprod Toxicol, 33, 353, 10.1016/j.reprotox.2012.01.007
Tomic, 2006, Methoxychlor induces atresia of antral follicles in ERα-overexpressing mice, Toxicol Sci, 93, 196, 10.1093/toxsci/kfl040
Basavarajappa, 2012, Methoxychlor induces atresia by altering Bcl2 factors and inducing caspase activity in mouse ovarian antral follicles in vitro, Reprod Toxicol, 34, 545, 10.1016/j.reprotox.2012.08.007
Gupta, 2006, Methoxychlor causes mitochondrial dysfunction and oxidative damage in the mouse ovary, Toxicol Appl Pharmacol, 216, 436, 10.1016/j.taap.2006.06.013
Gupta, 2006, Methoxychlor inhibits growth and induces atresia of antral follicles through an oxidative stress pathway, Toxicol Sci, 93, 382, 10.1093/toxsci/kfl052
Miller, 2006, Methoxychlor metabolites may cause ovarian toxicity through estrogen-regulated pathways, Toxicol Sci, 93, 180, 10.1093/toxsci/kfl034
Harvey, 2009, Effect of the methoxychlor metabolite HPTE on the rat ovarian granulosa cell transcriptome in vitro, Toxicol Sci, 110, 95, 10.1093/toxsci/kfp089
Craig, 2013, Pregnenolone co-treatment partially restores steroidogenesis, but does not prevent growth inhibition and increased atresia in mouse ovarian antral follicles treated with mono-hydroxy methoxychlor, Toxicol Appl Pharmacol, 272, 780, 10.1016/j.taap.2013.08.002
Koç, 2009, Dose-dependent effects of endosulfan and malathion on adult Wistar albino rat ovaries, Pak J Biol Sci, 12, 498, 10.3923/pjbs.2009.498.503
Nandi, 2011, Chlorpyrifos and endosulfan affect buffalo oocyte maturation, fertilization, and embryo development in vitro directly and through cumulus cells, Environ Toxicol, 26, 57, 10.1002/tox.20529
Sangha, 2013, Cypermethrin induced pathological and biochemical changes in reproductive organs of female rats, J Environ Biol, 34, 99
Shanthalatha, 2012, Effect of methomyl formulation, a carbamate pesticide on ovarian follicular development and fertility in albino mice, J Environ Biol, 33, 33
Kapoor, 2011, Toxicological impact of technical imidacloprid on ovarian morphology, hormones and antioxidant enzymes in female rats, Food Chem Toxicol, 49, 3086, 10.1016/j.fct.2011.09.009
Guerra, 2011, In utero and lactational exposure to fenvalerate disrupts reproductive function in female rats, Reprod Toxicol, 32, 298, 10.1016/j.reprotox.2011.08.002
Fei, 2010, Fenvalerate inhibits the growth of primary cultured rat preantral ovarian follicles, Toxicology, 267, 1, 10.1016/j.tox.2009.10.022
Cecconi, 2013, Effects of trifluralin on the mouse ovary, Environ Toxicol, 28, 201, 10.1002/tox.20711
Liu, 2011, Disrupting effects of bifenthrin on ovulatory gene expression and prostaglandin synthesis in rat ovarian granulosa cells, Toxicology, 282, 47, 10.1016/j.tox.2011.01.007
Grassi, 2011, Assessment of female reproductive endpoints in Sprague-Dawley rats developmentally exposed to Diuron: potential ovary toxicity, Birth Defects Res B Dev Reprod Toxicol, 92, 478, 10.1002/bdrb.20317
Pochettino, 2013, Oxidative stress in ventral prostate, ovary, and breast by 2,4-dichlorophenoxyacetic acid in pre- and postnatal exposed rats, Environ Toxicol, 28, 1, 10.1002/tox.20690
Karavan, 2012, Effects of estrogenic compounds on neonatal oocyte development, Reprod Toxicol, 34, 51, 10.1016/j.reprotox.2012.02.005
Kim, 2009, Effects of diethylstilbestrol on programmed oocyte death and induction of polyovular follicles in neonatal mouse ovaries, Biol Reprod, 81, 1002, 10.1095/biolreprod.108.070599
Alwis, 2011, Neonatal diethylstilbestrol exposure disrupts female reproductive tract structure/function via both direct and indirect mechanisms in the hamster, Reprod Toxicol, 32, 472, 10.1016/j.reprotox.2011.09.006
Petro, 2012, Endocrine-disrupting chemicals in human follicular fluid impair in vitro oocyte developmental competence, Hum Reprod, 27, 1025, 10.1093/humrep/der448
Tischkau, 2011, Circadian clock disruption in the mouse ovary in response to 2,3,7,8-tetrachlorodibenzo-p-dioxin, Toxicol Lett, 201, 116, 10.1016/j.toxlet.2010.12.013
Jung, 2010, Attenuation of cell cycle progression by 2,3,7,8-tetrachlorodibenzo-p-dioxin eliciting ovulatory blockade in gonadotropin-primed immature rats, Endocr J, 57, 863, 10.1507/endocrj.K10E-220
Chen, 2009, Proteomic analysis of the rat ovary following chronic low-dose exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), J Toxicol Environ Health A, 72, 717, 10.1080/15287390902841136
Yoshizawa, 2009, Reproductive lesions in female Harlan Sprague-Dawley rats following two-year oral treatment with dioxin and dioxin-like compounds, Toxicol Pathol, 37, 921, 10.1177/0192623309351721
Magre, 2012, Gender differences in transcriptional signature of developing rat testes and ovaries following embryonic exposure to 2,3,7,8-TCDD, PLoS One, 7, e40306, 10.1371/journal.pone.0040306
Mlynarczuk, 2009, The influence of polychlorinated biphenyls (PCBs), dichlorodiphenyltrichloroethane (DDT) and its metabolite-dichlorodiphenyldichloroethylene (DDE) on mRNA expression for NP-I/OT and PGA, involved in oxytocin synthesis in bovine granulosa and luteal cells, Reprod Toxicol, 28, 354, 10.1016/j.reprotox.2009.04.007
Pocar, 2012, Effects of polychlorinated biphenyls in CD-1 mice: reproductive toxicity and intergenerational transmission, Toxicol Sci, 126, 213, 10.1093/toxsci/kfr327
Kraugerud, 2012, In utero and lactational exposure to PCB 118 and PCB 153 alter ovarian follicular dynamics and GnRH-induced luteinizing hormone secretion in female lambs, Environ Toxicol, 27, 623, 10.1002/tox.20679
Mlynarczuk, 2013, The effect of PCB126, 77, and 153 on the intracellular mobilization of Ca+2 in bovine granulosa and luteal cells after FSH and LH surge in vitro, Pol J Vet Sci, 16, 417, 10.2478/pjvs-2013-0059
Paro, 2012, The fungicide mancozeb induces toxic effects on mammalian granulosa cells, Toxicol Appl Pharmacol, 260, 155, 10.1016/j.taap.2012.02.005
Vo, 2010, Potential estrogenic effect(s) of parabens at the prepubertal stage of a postnatal female rat model, Reprod Toxicol, 29, 306, 10.1016/j.reprotox.2010.01.013
Lee, 2012, Tributyltin increases the expression of apoptosis- and adipogenesis-related genes in rat ovaries, Clin Exp Reprod Med, 39, 15, 10.5653/cerm.2012.39.1.15
Jefferson, 2010, Adult ovarian function can be affected by high levels of soy, J Nutr, 140, 2322S, 10.3945/jn.110.123802
Zama, 2013, Targeted genome-wide methylation and gene expression analyses reveal signaling pathways involved in ovarian dysfunction after developmental EDC exposure in rats, Biol Reprod, 88, 52, 10.1095/biolreprod.112.104802
Zama, 2009, Fetal and neonatal exposure to the endocrine disruptor methoxychlor causes epigenetic alterations in adult ovarian genes, Endocrinology, 150, 4681, 10.1210/en.2009-0499
Manikkam, 2012, Pesticide and insect repellent mixture (permethrin and DEET) induces epigenetic transgenerational inheritance of disease and sperm epimutations, Reprod Toxicol, 34, 708, 10.1016/j.reprotox.2012.08.010
Manikkam, 2012, Dioxin (TCDD) induces epigenetic transgenerational inheritance of adult onset disease and sperm epimutations, PLoS One, 7, e46249, 10.1371/journal.pone.0046249
Ehrlich, 2012, Urinary bisphenol A concentrations and early reproductive health outcomes among women undergoing IVF, Hum Reprod, 27, 3583, 10.1093/humrep/des328
Mok-Lin, 2010, Urinary bisphenol A concentrations and ovarian response among women undergoing IVF, Int J Androl, 33, 385, 10.1111/j.1365-2605.2009.01014.x
Bloom, 2011, Bisphenol A exposure reduces the estradiol response to gonadotropin stimulation during in vitro fertilization, Fertil Steril, 96, 672, 10.1016/j.fertnstert.2011.06.063
Ehrlich, 2013, Urinary bisphenol A concentrations and cytochrome P450 19 A1 (Cyp19) gene expression in ovarian granulosa cells: an in vivo human study, Reprod Toxicol, 42, 18, 10.1016/j.reprotox.2013.06.071
Lee, 2014, Changes in steroid metabolism among girls with precocious puberty may not be associated with urinary levels of bisphenol A, Reprod Toxicol, 44, 1, 10.1016/j.reprotox.2013.03.008
Xi, 2011, Effect of perinatal and postnatal bisphenol A exposure to the regulatory circuits at the hypothalamus-pituitary-gonadal axis of CD-1 mice, Reprod Toxicol, 31, 409, 10.1016/j.reprotox.2010.12.002
Fernández, 2010, Neonatal exposure to bisphenol A and reproductive and endocrine alterations resembling the polycystic ovarian syndrome in adult rats, Environ Health Perspect, 118, 1217, 10.1289/ehp.0901257
Tan, 2013, Bisphenol A differentially activates protein kinase C isoforms in murine placental tissue, Toxicol Appl Pharmacol, 269, 163, 10.1016/j.taap.2013.03.016
Lee, 2013, Bisphenol A exposure during adulthood causes augmentation of follicular atresia and luteal regression by decreasing 17β-estradiol synthesis via downregulation of aromatase in rat ovary, Environ Health Perspect, 121, 663, 10.1289/ehp.1205823
Peretz, 2013, Bisphenol A down-regulates rate-limiting Cyp11a1 to acutely inhibit steroidogenesis in cultured mouse antral follicles, Toxicol Appl Pharmacol, 271, 249, 10.1016/j.taap.2013.04.028
Zhou, 2008, Effect of bisphenol A on steroid hormone production in rat ovarian theca-interstitial and granulosa cells, Mol Cell Endocrinol, 283, 12, 10.1016/j.mce.2007.10.010
Grasselli, 2010, Bisphenol A disrupts granulosa cell function, Domest Anim Endocrinol, 39, 34, 10.1016/j.domaniend.2010.01.004
Hart, 2014, The influence of antenatal exposure to phthalates on subsequent female reproductive development in adolescence: a pilot study, Reproduction, 147, 379, 10.1530/REP-13-0331
Svechnikova, 2007, The influence of di-(2-ethylhexyl) phthalate on steroidogenesis by the ovarian granulosa cells of immature female rats, J Endocrinol, 194, 603, 10.1677/JOE-07-0238
Liu, 2014, Effects of di-(2-ethylhexyl) phthalate on the hypothalamus-pituitary-ovarian axis in adult female rats, Reprod Toxicol, 46, 141, 10.1016/j.reprotox.2014.03.006
Martinez-Arguelles, 2011, In utero exposure to the antiandrogen di-(2-ethylhexyl) phthalate decreases adrenal aldosterone production in the adult rat, Biol Reprod, 85, 51, 10.1095/biolreprod.110.089920
Moyer, 2012, Reproductive effects in F1 adult females exposed in utero to moderate to high doses of mono-2-ethylhexylphthalate (MEHP), Reprod Toxicol, 34, 43, 10.1016/j.reprotox.2012.02.006
Herreros, 2013, Toxicokinetics of di(2-ethylhexyl) phthalate (DEHP) and its effects on luteal function in sheep, Reprod Biol, 13, 66, 10.1016/j.repbio.2013.01.177
Hannon, 2015, Mono(2-ethylhexyl) phthalate accelerates early folliculogenesis and inhibits steroidogenesis in cultured mouse whole ovaries and antral follicles, Biol Reprod, 92, 120, 10.1095/biolreprod.115.129148
Mlynarcíková, 2009, Effects of selected endocrine disruptors on meiotic maturation, cumulus expansion, synthesis of hyaluronan and progesterone by porcine oocyte-cumulus complexes, Toxicol In Vitro, 23, 371, 10.1016/j.tiv.2008.12.017
Lenie, 2009, Steroidogenesis-disrupting compounds can be effectively studied for major fertility-related endpoints using in vitro cultured mouse follicles, Toxicol Lett, 185, 143, 10.1016/j.toxlet.2008.12.015
Reinsberg, 2009, Effect of mono-(2-ethylhexyl) phthalate on steroid production of human granulosa cells, Toxicol Appl Pharmacol, 239, 116, 10.1016/j.taap.2009.05.022
Ohno, 2009, Mono-(2-ethylhexyl) phthalate induces NR4A subfamily and GIOT-1 gene expression, and suppresses CYP19 expression in human granulosa-like tumor cell line KGN, Toxicol Lett, 191, 353, 10.1016/j.toxlet.2009.10.004
Gunnarsson, 2008, Mono-(2-ethylhexyl) phthalate stimulates basal steroidogenesis by a cAMP-independent mechanism in mouse gonadal cells of both sexes, Reproduction, 135, 693, 10.1530/REP-07-0460
Luderer, 2013, Effects of gestational and lactational exposure to heptachlor epoxide on age at puberty and reproductive function in men and women, Environ Res, 121, 84, 10.1016/j.envres.2012.11.001
Basavarajappa, 2011, Methoxychlor reduces estradiol levels by altering steroidogenesis and metabolism in mouse antral follicles in vitro, Toxicol Appl Pharmacol, 253, 161, 10.1016/j.taap.2011.04.007
Akgul, 2011, The methoxychlor metabolite, HPTE, inhibits rat luteal cell progesterone production, Reprod Toxicol, 32, 77, 10.1016/j.reprotox.2011.05.013
Craig, 2010, Mono-hydroxy methoxychlor alters levels of key sex steroids and steroidogenic enzymes in cultured mouse antral follicles, Toxicol Appl Pharmacol, 249, 107, 10.1016/j.taap.2010.09.001
Gill, 2011, Toxic effects of cypermethrin and methamidophos on bovine corpus luteal cells and progesterone production, Exp Toxicol Pathol, 63, 131, 10.1016/j.etp.2009.10.007
Gregoraszczuk, 2011, Differential accumulation of HCBz and PeCBz in porcine ovarian follicles and their opposing actions on steroid secretion and CYP11, CYP17, 17β-HSD and CYP19 protein expression. A tissue culture approach, Reprod Toxicol, 31, 494, 10.1016/j.reprotox.2011.01.006
Liu, 2011, Enantioselective endocrine-disrupting effects of bifenthrin on hormone synthesis in rat ovarian cells, Toxicology, 290, 42, 10.1016/j.tox.2011.08.016
Taketa, 2011, Differential stimulation pathways of progesterone secretion from newly formed corpora lutea in rats treated with ethylene glycol monomethyl ether, sulpiride, or atrazine, Toxicol Sci, 121, 267, 10.1093/toxsci/kfr062
Quignot, 2012, Characterization of endocrine-disrupting chemicals based on hormonal balance disruption in male and female adult rats, Reprod Toxicol, 33, 339, 10.1016/j.reprotox.2012.01.004
Tinfo, 2011, Understanding the effects of atrazine on steroidogenesis in rat granulosa and H295R adrenal cortical carcinoma cells, Reprod Toxicol, 31, 184, 10.1016/j.reprotox.2010.11.005
Basini, 2012, Atrazine disrupts steroidogenesis, VEGF and NO production in swine granulosa cells, Ecotoxicol Environ Saf, 85, 59, 10.1016/j.ecoenv.2012.08.027
Fa, 2013, Involvement of ERK1/2 signaling pathway in atrazine action on FSH-stimulated LHR and CYP19A1 expression in rat granulosa cells, Toxicol Appl Pharmacol, 270, 1, 10.1016/j.taap.2013.03.031
Su, 2012, The effect of in utero exposure to dioxins and polychlorinated biphenyls on reproductive development in eight year-old children, Environ Int, 39, 181, 10.1016/j.envint.2011.09.009
Huang, 2011, Three-generation experiment showed female C57BL/6J mice drink drainage canal water containing low level of TCDD-like activity causing high pup mortality, J Toxicol Sci, 36, 713, 10.2131/jts.36.713
Karman, 2012, 2,3,7,8-Tetrachlorodibenzo-p-dioxin activates the aryl hydrocarbon receptor and alters sex steroid hormone secretion without affecting growth of mouse antral follicles in vitro, Toxicol Appl Pharmacol, 261, 88, 10.1016/j.taap.2012.03.015
Karman, 2012, Dioxin exposure reduces the steroidogenic capacity of mouse antral follicles mainly at the level of HSD17B1 without altering atresia, Toxicol Appl Pharmacol, 264, 1, 10.1016/j.taap.2012.07.031
Maranghi, 2013, Dietary exposure of juvenile female mice to polyhalogenated seafood contaminants (HBCD, BDE-47, PCB-153, TCDD): comparative assessment of effects in potential target tissues, Food Chem Toxicol, 56, 443, 10.1016/j.fct.2013.02.056
Sechman, 2014, Effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on secretion of steroids and STAR, HSD3B and CYP19A1 mRNA expression in chicken ovarian follicles, Toxicol Lett, 225, 264, 10.1016/j.toxlet.2013.12.021
Sechman, 2011, Effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on steroid concentrations in blood and gonads of chicken embryo, Toxicol Lett, 205, 190, 10.1016/j.toxlet.2011.06.004
Signorile, 2010, Pre-natal exposure of mice to bisphenol A elicits an endometriosis-like phenotype in female offspring, Gen Comp Endocrinol, 168, 318, 10.1016/j.ygcen.2010.03.030
Signorile, 2012, Endocrine disruptors in utero cause ovarian damages linked to endometriosis, Front Biosci (Elite Ed), 4, 1724, 10.2741/e493
Kendig, 2012, Estrogen-like disruptive effects of dietary exposure to bisphenol A or 17α-ethinyl estradiol in CD1 mice, Int J Toxicol, 31, 537, 10.1177/1091581812463254
Yu, 2010, Estrogen receptor α and β expressions in hypothalamus-pituitary-ovary axis in rats exposed lactationally to soy isoflavones and bisphenol A, Biomed Environ Sci, 23, 357, 10.1016/S0895-3988(10)60076-1
Calhoun, 2014, Bisphenol A exposure alters developmental gene expression in the fetal rhesus macaque uterus, PLoS One, 9, e85894, 10.1371/journal.pone.0085894
Aldad, 2011, Bisphenol-A exposure alters endometrial progesterone receptor expression in the nonhuman primate, Fertil Steril, 96, 175, 10.1016/j.fertnstert.2011.04.010
Berger, 2010, Bisphenol-A exposure during the period of blastocyst implantation alters uterine morphology and perturbs measures of estrogen and progesterone receptor expression in mice, Reprod Toxicol, 30, 393, 10.1016/j.reprotox.2010.06.006
Varayoud, 2008, Developmental exposure to Bisphenol A impairs the uterine response to ovarian steroids in the adult, Endocrinology, 149, 5848, 10.1210/en.2008-0651
Bosquiazzo, 2010, Effects of neonatal exposure to bisphenol A on steroid regulation of vascular endothelial growth factor expression and endothelial cell proliferation in the adult rat uterus, Biol Reprod, 82, 86, 10.1095/biolreprod.109.078543
Mendoza-Rodríguez, 2011, Administration of bisphenol A to dams during perinatal period modifies molecular and morphological reproductive parameters of the offspring, Reprod Toxicol, 31, 177, 10.1016/j.reprotox.2010.10.013
Aghajanova, 2011, Effect of bisphenol A on human endometrial stromal fibroblasts in vitro, Reprod Biomed Online, 22, 249, 10.1016/j.rbmo.2010.12.007
Bredhult, 2009, Gene expression analysis of human endometrial endothelial cells exposed to bisphenol A, Reprod Toxicol, 28, 18, 10.1016/j.reprotox.2009.03.006
Kendziorski, 2012, Strain specific induction of pyometra and differences in immune responsiveness in mice exposed to 17α-ethinyl estradiol or the endocrine disrupting chemical bisphenol A, Reprod Toxicol, 34, 22, 10.1016/j.reprotox.2012.03.001
Hewitt, 2011, Estrogenic activity of bisphenol A and 2,2-bis(p-hydroxyphenyl)-1,1,1-trichloroethane (HPTE) demonstrated in mouse uterine gene profiles, Environ Health Perspect, 119, 63, 10.1289/ehp.1002347
An, 2013, Effects of estrogen and estrogenic compounds, 4-tert-octylphenol, and bisphenol A on the uterine contraction and contraction-associated proteins in rats, Mol Cell Endocrinol, 375, 27, 10.1016/j.mce.2013.04.025
Hiyama, 2011, Bisphenol-A (BPA) affects reproductive formation across generations in mice, J Vet Med Sci, 73, 1211, 10.1292/jvms.11-0135
Okuda, 2010, In vivo estrogenic potential of 4-methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene, an active metabolite of bisphenol A, in uterus of ovariectomized rat, Toxicol Lett, 197, 7, 10.1016/j.toxlet.2010.04.017
Toft, 2012, Association between pregnancy loss and urinary phthalate levels around the time of conception, Environ Health Perspect, 120, 458, 10.1289/ehp.1103552
Zhang, 2011, Estrogen agonist/antagonist properties of dibenzyl phthalate (DBzP) based on in vitro and in vivo assays, Toxicol Lett, 207, 7, 10.1016/j.toxlet.2011.08.017
Wang, 2010, Effect of phthalate esters on the secretion of prostaglandins (F2α and E2) and oxytocin in cultured bovine ovarian and endometrial cells, Domest Anim Endocrinol, 39, 131, 10.1016/j.domaniend.2010.03.002
Chung, 2013, Uterotrophic and Hershberger assays for endocrine disruption properties of plastic food contact materials polypropylene (PP) and polyethylene terephthalate (PET), J Toxicol Environ Health A, 76, 624, 10.1080/15287394.2013.801767
Yu, 2013, Maternal exposure to the mixture of organophosphorus pesticides induces reproductive dysfunction in the offspring, Environ Toxicol, 28, 507, 10.1002/tox.20741
Uslu, 2013, Evaluation of estrogenic effects of polychlorinated biphenyls and organochlorinated pesticides using immature rat uterotrophic assay, Hum Exp Toxicol, 32, 476, 10.1177/0960327112472999
Mlynarczuk, 2010, Effect of environmental pollutants on oxytocin synthesis and secretion from corpus luteum and on contractions of uterus from pregnant cows, Toxicol Appl Pharmacol, 247, 243, 10.1016/j.taap.2010.07.003
Chang, 2010, Deleterious effects of arsenic, benomyl and carbendazim on human endometrial cell proliferation in vitro, Taiwan J Obstet Gynecol, 49, 449, 10.1016/S1028-4559(10)60097-6
Kwekel, 2013, o-p'-DDT-mediated uterotrophy and gene expression in immature C57BL/6 mice and Sprague-Dawley rats, Toxicol Appl Pharmacol, 273, 532, 10.1016/j.taap.2013.09.024
Milesi, 2012, Neonatal exposure to low doses of endosulfan disrupts the expression of proteins regulating uterine development and differentiation, Reprod Toxicol, 33, 85, 10.1016/j.reprotox.2011.12.003
Undeger, 2010, Effect of the herbicide pendimethalin on rat uterine weight and gene expression and in silico receptor binding analysis, Food Chem Toxicol, 48, 502, 10.1016/j.fct.2009.11.001
Laffin, 2010, The pyrethroid metabolites 3-phenoxybenzoic acid and 3-phenoxybenzyl alcohol do not exhibit estrogenic activity in the MCF-7 human breast carcinoma cell line or Sprague-Dawley rats, Toxicology, 267, 39, 10.1016/j.tox.2009.10.003
Yoshida, 2011, Delayed adverse effects of neonatal exposure to diethylstilbestrol and their dose dependency in female rats, Toxicol Pathol, 39, 823, 10.1177/0192623311413785
Yin, 2012, Neonatal diethylstilbestrol exposure alters the metabolic profile of uterine epithelial cells, Dis Model Mech, 5, 870, 10.1242/dmm.009076
Jefferson, 2013, Persistently altered epigenetic marks in the mouse uterus after neonatal estrogen exposure, Mol Endocrinol, 27, 1666, 10.1210/me.2013-1211
Hayashi, 2011, WNTs in the neonatal mouse uterus: potential regulation of endometrial gland development, Biol Reprod, 84, 308, 10.1095/biolreprod.110.088161
Yamashita, 2013, Mechanisms of angiogenic suppression in uteri exposed to diethylstilbestrol neonatally in the mouse, Biol Reprod, 88, 116, 10.1095/biolreprod.112.106443
Hong, 2010, Histopathological and gene expression analysis of mice exposed to diethylstilbestrol, Toxicol Mech Methods, 20, 105, 10.3109/15376510903572631
Simmons, 2010, Response of adult mouse uterus to early disruption of estrogen receptor-α signaling is influenced by Krüppel-like factor 9, J Endocrinol, 205, 147, 10.1677/JOE-09-0474
Bredfeldt, 2010, Xenoestrogen-induced regulation of EZH2 and histone methylation via estrogen receptor signaling to PI3K/AKT, Mol Endocrinol, 24, 993, 10.1210/me.2009-0438
Bosquiazzo, 2013, Perinatal exposure to diethylstilbestrol alters the functional differentiation of the adult rat uterus, J Steroid Biochem Mol Biol, 138, 1, 10.1016/j.jsbmb.2013.02.011
Wrobel, 2009, Involvement of prostaglandin F2α in the adverse effect of PCB 77 on the force of contractions of bovine myometrium, Toxicology, 262, 224, 10.1016/j.tox.2009.06.012
Burns, 2013, A single gestational exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin disrupts the adult uterine response to estradiol in mice, Toxicol Sci, 136, 514, 10.1093/toxsci/kft208
Tsang, 2012, 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) suppresses spheroids attachment on endometrial epithelial cells through the down-regulation of the Wnt-signaling pathway, Reprod Toxicol, 33, 60, 10.1016/j.reprotox.2011.11.002
Resuehr, 2012, Progesterone-dependent regulation of endometrial cannabinoid receptor type 1 (CB1-R) expression is disrupted in women with endometriosis and in isolated stromal cells exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), Fertil Steril, 98, 948, 10.1016/j.fertnstert.2012.06.009
Louis, 2013, The effect of triclosan on the uterotrophic response to extended doses of ethinyl estradiol in the weanling rat, Reprod Toxicol, 36, 71, 10.1016/j.reprotox.2012.12.001
Dixon, 2012, Histopathologic changes in the uterus, cervix and vagina of immature CD-1 mice exposed to low doses of perfluorooctanoic acid (PFOA) in a uterotrophic assay, Reprod Toxicol, 33, 506, 10.1016/j.reprotox.2011.10.011
Shaw, 2009, Estrogenicity of parabens revisited: impact of parabens on early pregnancy and an uterotrophic assay in mice, Reprod Toxicol, 28, 26, 10.1016/j.reprotox.2009.03.003
Smith, 2012, Higher incidence of clear cell adenocarcinoma of the cervix and vagina among women born between 1947 and 1971 in the United States, Cancer Causes Control, 23, 207, 10.1007/s10552-011-9855-z
Laronda, 2012, The development of cervical and vaginal adenosis as a result of diethylstilbestrol exposure in utero, Differentiation, 84, 252, 10.1016/j.diff.2012.05.004
Laronda, 2013, Diethylstilbestrol induces vaginal adenosis by disrupting SMAD/RUNX1-mediated cell fate decision in the Müllerian duct epithelium, Dev Biol, 381, 5, 10.1016/j.ydbio.2013.06.024
Katoh, 2013, Epithelial-stromal interactions in the mouse vagina exposed neonatally to diethylstilbestrol, In Vivo, 27, 333
Nakamura, 2012, Wnt family genes and their modulation in the ovary-independent and persistent vaginal epithelial cell proliferation and keratinization induced by neonatal diethylstilbestrol exposure in mice, Toxicology, 296, 13, 10.1016/j.tox.2012.02.010
Nakamura, 2012, p21 and Notch signalings in the persistently altered vagina induced by neonatal diethylstilbestrol exposure in mice, J Vet Med Sci, 74, 1589, 10.1292/jvms.12-0182
Matsuda, 2014, Activated vitamin D3 and pro-activated vitamin D3 attenuate induction of permanent changes caused by neonatal estrogen exposure in the mouse vagina, J Reprod Dev, 60, 274, 10.1262/jrd.2014-015
Brannick, 2012, Prenatal exposure to low doses of bisphenol A increases pituitary proliferation and gonadotroph number in female mice offspring at birth, Biol Reprod, 87, 82, 10.1095/biolreprod.112.100636
Fernández, 2009, Neonatal exposure to bisphenol A alters reproductive parameters and gonadotropin releasing hormone signaling in female rats, Environ Health Perspect, 117, 757, 10.1289/ehp.0800267
Jeng, 2010, Combinations of physiologic estrogens with xenoestrogens alter calcium and kinase responses, prolactin release, and membrane estrogen receptor trafficking in rat pituitary cells, Environ Health, 9, 61, 10.1186/1476-069X-9-61
Carbone, 2012, Different effects by sex on hypothalamic-pituitary axis of prepubertal offspring rats produced by in utero and lactational exposure to di-(2-ethylhexyl) phthalate (DEHP), Neurotoxicology, 33, 78, 10.1016/j.neuro.2011.11.009
Fraites, 2009, Characterization of the hypothalamic-pituitary-adrenal axis response to atrazine and metabolites in the female rat, Toxicol Sci, 112, 88, 10.1093/toxsci/kfp194
Foradori, 2011, The differential effect of atrazine on luteinizing hormone release in adrenalectomized adult female Wistar rats, Biol Reprod, 85, 684, 10.1095/biolreprod.111.092452
Goldman, 2013, Atrazine-induced elevation or attenuation of the LH surge in the ovariectomized, estrogen-primed female rat: role of adrenal progesterone, Reproduction, 146, 305, 10.1530/REP-13-0011
Ishikawa, 2014, Effects of diethylstilbestrol on luteinizing hormone-producing cells in the mouse anterior pituitary, Exp Biol Med (Maywood), 239, 311, 10.1177/1535370213519722
Kakuta, 2012, Involvement of gonadotropins in the induction of hypertrophy-hyperplasia in the interstitial tissues of ovaries in neonatally diethylstilbestrol-treated mice, Reprod Toxicol, 33, 35, 10.1016/j.reprotox.2011.10.013
Takeda, 2009, Maternal exposure to dioxin disrupts gonadotropin production in fetal rats and imprints defects in sexual behavior, J Pharmacol Exp Ther, 329, 1091, 10.1124/jpet.109.151282
Koga, 2012, Restoration of dioxin-induced damage to fetal steroidogenesis and gonadotropin formation by maternal co-treatment with α-lipoic acid, PLoS One, 7, e40322, 10.1371/journal.pone.0040322
Takeda, 2012, Dioxin silences gonadotropin expression in perinatal pups by inducing histone deacetylases: a new insight into the mechanism for the imprinting of sexual immaturity by dioxin, J Biol Chem, 287, 18440, 10.1074/jbc.M111.335158
Takeda, 2011, The effect of maternal exposure to dioxin on fetal steroidogenesis in the steroidogenic organs [in Japanese], Fukuoka Igaku Zasshi, 102, 159
Jablonska, 2011, In vitro effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on ovarian, pituitary, and pineal function in pigs, Theriogenology, 76, 921, 10.1016/j.theriogenology.2011.04.023
Cao, 2011, Aryl hydrocarbon receptor activation in lactotropes and gonadotropes interferes with estradiol-dependent and -independent preprolactin, glycoprotein α and luteinizing hormone β gene expression, Mol Cell Endocrinol, 333, 151, 10.1016/j.mce.2010.12.027
Isling, 2014, Late-life effects on rat reproductive system after developmental exposure to mixtures of endocrine disrupters, Reproduction, 147, 465, 10.1530/REP-13-0448
Durmaz, 2014, Urinary bisphenol A levels in girls with idiopathic central precocious puberty, J Clin Res Pediatr Endocrinol, 6, 16, 10.4274/Jcrpe.1220
Wolff, 2010, Investigation of relationships between urinary biomarkers of phytoestrogens, phthalates, and phenols and pubertal stages in girls, Environ Health Perspect, 118, 1039, 10.1289/ehp.0901690
Wolff, 2008, Environmental exposures and puberty in inner-city girls, Environ Res, 107, 393, 10.1016/j.envres.2008.03.006
Lee, 2014, Prenatal bisphenol A and birth outcomes: MOCEH (Mothers and Children's Environmental Health) study, Int J Hyg Environ Health, 217, 328, 10.1016/j.ijheh.2013.07.005
Ryan, 2010, In utero and lactational exposure to bisphenol A, in contrast to ethinyl estradiol, does not alter sexually dimorphic behavior, puberty, fertility, and anatomy of female LE rats, Toxicol Sci, 114, 133, 10.1093/toxsci/kfp266
Adewale, 2009, Neonatal bisphenol-A exposure alters rat reproductive development and ovarian morphology without impairing activation of gonadotropin-releasing hormone neurons, Biol Reprod, 81, 690, 10.1095/biolreprod.109.078261
Nah, 2011, Effects of early prepubertal exposure to bisphenol A on the onset of puberty, ovarian weights, and estrous cycle in female mice, Clin Exp Reprod Med, 38, 75, 10.5653/cerm.2011.38.2.75
Chakraborty, 2012, Relationships between urinary biomarkers of phytoestrogens, phthalates, phenols, and pubertal stages in girls, Adolesc Health Med Ther, 3, 17, 10.2147/AHMT.S15947
Jurewicz, 2011, Exposure to phthalates: reproductive outcome and children health. A review of epidemiological studies, Int J Occup Med Environ Health, 24, 115, 10.2478/s13382-011-0022-2
Chen, 2013, Phthalates may promote female puberty by increasing kisspeptin activity, Hum Reprod, 28, 2765, 10.1093/humrep/det325
Wolff, 2014, Phthalate exposure and pubertal development in a longitudinal study of US girls, Hum Reprod, 29, 1558, 10.1093/humrep/deu081
Frederiksen, 2012, High urinary phthalate concentration associated with delayed pubarche in girls, Int J Androl, 35, 216, 10.1111/j.1365-2605.2012.01260.x
Lomenick, 2010, Phthalate exposure and precocious puberty in females, J Pediatr, 156, 221, 10.1016/j.jpeds.2009.09.047
Mouritsen, 2013, Urinary phthalates from 168 girls and boys measured twice a year during a 5-year period: associations with adrenal androgen levels and puberty, J Clin Endocrinol Metab, 98, 3755, 10.1210/jc.2013-1284
Ahmad, Assessment of estrogenic potential of di-n-butyl phthalate and butyl benzyl phthalate in vivo [published online July 5, 2013], Toxicol Ind Health
Hu, 2013, Short-term neonatal/prepubertal exposure of dibutyl phthalate (DBP) advanced pubertal timing and affected hypothalamic kisspeptin/GPR54 expression differently in female rats, Toxicology, 314, 65, 10.1016/j.tox.2013.09.007
Ding, 2010, Effects of in utero exposure to di(2-ethylhexyl) phthalate on sexual development in female offspring[in Chinese], Zhonghua Yu Fang Yi Xue Za Zhi, 44, 150
Ozen, 2014, Agricultural pesticides and precocious puberty, Vitam Horm, 94, 27, 10.1016/B978-0-12-800095-3.00002-X
Ozen, 2012, Effects of pesticides used in agriculture on the development of precocious puberty, Environ Monit Assess, 184, 4223, 10.1007/s10661-011-2257-6
Deng, 2012, Effects of growth environments and two environmental endocrine disruptors on children with idiopathic precocious puberty, Eur J Endocrinol, 166, 803, 10.1530/EJE-11-0876
Davis, 2011, The effects of prenatal exposure to atrazine on pubertal and postnatal reproductive indices in the female rat, Reprod Toxicol, 32, 43, 10.1016/j.reprotox.2011.04.004
Zorrilla, 2010, The effects of simazine, a chlorotriazine herbicide, on pubertal development in the female Wistar rat, Reprod Toxicol, 29, 393, 10.1016/j.reprotox.2010.03.010
Rollerova, 2011, Neonatal exposure to herbicide acetochlor alters pubertal development in female Wistar rats, Toxicol Mech Methods, 21, 406, 10.3109/15376516.2010.551554
Manikkam, 2012, Transgenerational actions of environmental compounds on reproductive disease and identification of epigenetic biomarkers of ancestral exposures, PLoS One, 7, e31901, 10.1371/journal.pone.0031901
Si, 2012, Perinatal exposure to low doses of tributyltin chloride advances puberty and affects patterns of estrous cyclicity in female mice, Environ Toxicol, 27, 662, 10.1002/tox.21756
Patisaul, 2013, Accumulation and endocrine disrupting effects of the flame retardant mixture Firemaster550 in rats: an exploratory assessment, J Biochem Mol Toxicol, 27, 124, 10.1002/jbt.21439
Colciago, 2009, Chronic treatment with polychlorinated biphenyls (PCB) during pregnancy and lactation in the rat. Part 2: effects on reproductive parameters, on sex behavior, on memory retention and on hypothalamic expression of aromatase and 5α-reductases in the offspring, Toxicol Appl Pharmacol, 239, 46, 10.1016/j.taap.2009.04.023
Dickerson, 2011, Endocrine disruption of brain sexual differentiation by developmental PCB exposure, Endocrinology, 152, 581, 10.1210/en.2010-1103
Kristensen, 2013, Long-term effects of prenatal exposure to perfluoroalkyl substances on female reproduction, Hum Reprod, 28, 3337, 10.1093/humrep/det382
Lopez-Espinosa, 2011, Association of perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS) with age of puberty among children living near a chemical plant, Environ Sci Technol, 45, 8160, 10.1021/es1038694
Tucker, 2015, The mammary gland is a sensitive pubertal target in CD-1 and C57Bl/6 mice following perinatal perfluorooctanoic acid (PFOA) exposure, Reprod Toxicol, 54, 26, 10.1016/j.reprotox.2014.12.002
Buttke, 2012, Exposures to endocrine-disrupting chemicals and age of menarche in adolescent girls in NHANES (2003–2008), Environ Health Perspect, 120, 1613, 10.1289/ehp.1104748
Monje, 2010, Exposure of neonatal female rats to bisphenol A disrupts hypothalamic LHRH pre-mRNA processing and estrogen receptor α expression in nuclei controlling estrous cyclicity, Reprod Toxicol, 30, 625, 10.1016/j.reprotox.2010.08.004
Tyl, 2008, Two-generation reproductive toxicity study of dietary bisphenol A in CD-1 (Swiss) mice, Toxicol Sci, 104, 362, 10.1093/toxsci/kfn084
Buck Louis, 2011, Persistent organochlorine pollutants and menstrual cycle characteristics, Chemosphere, 85, 1742, 10.1016/j.chemosphere.2011.09.027
Cragin, 2011, Menstrual cycle characteristics and reproductive hormone levels in women exposed to atrazine in drinking water, Environ Res, 111, 1293, 10.1016/j.envres.2011.09.009
Buck Louis, 2014, Urinary bisphenol A, phthalates, and couple fecundity: the Longitudinal Investigation of Fertility and the Environment (LIFE) Study, Fertil Steril, 101, 1359, 10.1016/j.fertnstert.2014.01.022
Ehrlich, 2012, Urinary bisphenol A concentrations and implantation failure among women undergoing in vitro fertilization, Environ Health Perspect, 120, 978, 10.1289/ehp.1104307
Lathi, 2014, Conjugated bisphenol A in maternal serum in relation to miscarriage risk, Fertil Steril, 102, 123, 10.1016/j.fertnstert.2014.03.024
Caserta, 2013, The influence of endocrine disruptors in a selected population of infertile women, Gynecol Endocrinol, 29, 444, 10.3109/09513590.2012.758702
Krotz, 2012, Phthalates and bisphenol do not accumulate in human follicular fluid, J Assist Reprod Genet, 29, 773, 10.1007/s10815-012-9775-1
Berger, 2008, Impact of acute bisphenol-A exposure upon intrauterine implantation of fertilized ova and urinary levels of progesterone and 17β-estradiol, Reprod Toxicol, 26, 94, 10.1016/j.reprotox.2008.06.007
Xiao, 2011, Preimplantation exposure to bisphenol A (BPA) affects embryo transport, preimplantation embryo development, and uterine receptivity in mice, Reprod Toxicol, 32, 434
Varayoud, 2011, Neonatal exposure to bisphenol A alters rat uterine implantation-associated gene expression and reduces the number of implantation sites, Endocrinology, 152, 1101, 10.1210/en.2009-1037
Tiwari, 2013, Mutagenic effect of bisphenol A on adult rat male germ cells and their fertility, Reprod Toxicol, 40, 60, 10.1016/j.reprotox.2013.05.013
Varayoud, 2014, Long-lasting effects of neonatal bisphenol A exposure on the implantation process, Vitam Horm, 94, 253, 10.1016/B978-0-12-800095-3.00010-9
Tranfo, 2012, Urinary phthalate monoesters concentration in couples with infertility problems, Toxicol Lett, 213, 15, 10.1016/j.toxlet.2011.11.033
Schmidt, 2012, Effects of di(2-ethylhexyl) phthalate (DEHP) on female fertility and adipogenesis in C3H/N mice, Environ Health Perspect, 120, 1123, 10.1289/ehp.1104016
Niermann, 2015, Prenatal exposure to di-(2-ethylhexyl) phthalate (DEHP) affects reproductive outcomes in female mice, Reprod Toxicol, 53, 23, 10.1016/j.reprotox.2015.02.013
Mahalingaiah, 2012, Association of hexachlorobenzene (HCB), dichlorodiphenyltrichloroethane (DDT), and dichlorodiphenyldichloroethylene (DDE) with in vitro fertilization (IVF) outcomes, Environ Health Perspect, 120, 316, 10.1289/ehp.1103696
Blanco-Muñoz, 2013, Association between PON1 genetic polymorphisms and miscarriage in Mexican women exposed to pesticides, Sci Total Environ, 449, 302, 10.1016/j.scitotenv.2013.01.034
Toft, 2010, Fetal loss and maternal serum levels of 2,2′,4,4′,5,5′-hexachlorbiphenyl (CB-153) and 1,1-dichloro-2,2-bis(p-chlorophenyl)ethylene (p,p'-DDE) exposure: a cohort study in Greenland and two European populations, Environ Health, 9, 22, 10.1186/1476-069X-9-22
Naidoo, 2011, Spontaneous miscarriages and infant deaths among female farmers in rural South Africa, Scand J Work Environ Health, 37, 227, 10.5271/sjweh.3133
Bastos, 2013, Organochlorine compound levels in fertile and infertile women from Rio de Janeiro, Brazil, Arq Bras Endocrinol Metabol, 57, 346, 10.1590/S0004-27302013000500003
Al-Saleh, 2009, Outcome of in-vitro fertilization treatment and DDT levels in serum and follicular fluid, Med Sci Monit, 15, BR320
Hougaard, 2009, Infertility among women working in horticulture. A follow-up study in the Danish Occupational Hospitalization Register, Fertil Steril, 91, 1385, 10.1016/j.fertnstert.2008.04.036
Jirsová, 2010, Effect of polychlorinated biphenyls (PCBs) and 1,1,1-trichloro-2,2,-bis (4-chlorophenyl)-ethane (DDT) in follicular fluid on the results of in vitro fertilization-embryo transfer (IVF-ET) programs, Fertil Steril, 93, 1831, 10.1016/j.fertnstert.2008.12.063
Tiemann, 2008, In vivo and in vitro effects of the organochlorine pesticides DDT, TCPM, methoxychlor, and lindane on the female reproductive tract of mammals: a review, Reprod Toxicol, 25, 316, 10.1016/j.reprotox.2008.03.002
Yang, 2009, Bisphenol A exposure is associated with oxidative stress and inflammation in postmenopausal women, Environ Res, 109, 797, 10.1016/j.envres.2009.04.014
Souter, 2013, The association of bisphenol-A urinary concentrations with antral follicle counts and other measures of ovarian reserve in women undergoing infertility treatments, Reprod Toxicol, 42, 224, 10.1016/j.reprotox.2013.09.008
Grindler, 2015, Persistent organic pollutants and early menopause in U.S. women, PLoS One, 10, e0116057, 10.1371/journal.pone.0116057
Gore, 2011, Early life exposure to endocrine-disrupting chemicals causes lifelong molecular reprogramming of the hypothalamus and premature reproductive aging, Mol Endocrinol, 25, 2157, 10.1210/me.2011-1210
Hoover, 2011, Adverse health outcomes in women exposed in utero to diethylstilbestrol, N Engl J Med, 365, 1304, 10.1056/NEJMoa1013961
Jablonska, 2010, Temporal and anatomical sensitivities to the aryl hydrocarbon receptor agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin leading to premature acyclicity with age in rats, Int J Androl, 33, 405, 10.1111/j.1365-2605.2009.01031.x
Walker, 2013, Disruption of reproductive aging in female and male rats by gestational exposure to estrogenic endocrine disruptors, Endocrinology, 154, 2129, 10.1210/en.2012-2123
Smith, 2013, Urinary paraben concentrations and ovarian aging among women from a fertility center, Environ Health Perspect, 121, 1299, 10.1289/ehp.1205350
Rutkowska, 2014, Bisphenol A (BPA) and its potential role in the pathogenesis of the polycystic ovary syndrome (PCOS), Gynecol Endocrinol, 30, 260, 10.3109/09513590.2013.871517
Kandaraki, 2011, Endocrine disruptors and polycystic ovary syndrome (PCOS): elevated serum levels of bisphenol A in women with PCOS, J Clin Endocrinol Metab, 96, E480, 10.1210/jc.2010-1658
Li, 2011, Effects of eight environmental endocrine disruptors on insulin resistance in patients with polycystic ovary syndrome: a preliminary investigation [in Chinese], Nan Fang Yi Ke Da Xue Xue Bao, 31, 1753
Newbold, 2009, Prenatal exposure to bisphenol A at environmentally relevant doses adversely affects the murine female reproductive tract later in life, Environ Health Perspect, 117, 879, 10.1289/ehp.0800045
Patisaul, 2014, Soy but not bisphenol A (BPA) induces hallmarks of polycystic ovary syndrome (PCOS) and related metabolic co-morbidities in rats, Reprod Toxicol, 49C, 209, 10.1016/j.reprotox.2014.09.003
Abbott, 2013, Nonhuman primate models of polycystic ovary syndrome, Mol Cell Endocrinol, 373, 21, 10.1016/j.mce.2013.01.013
Rae, 2013, The pancreas is altered by in utero androgen exposure: implications for clinical conditions such as polycystic ovary syndrome (PCOS), PLoS One, 8, e56263, 10.1371/journal.pone.0056263
Padmanabhan, 2013, Sheep models of polycystic ovary syndrome phenotype, Mol Cell Endocrinol, 373, 8, 10.1016/j.mce.2012.10.005
Cobellis, 2009, Measurement of bisphenol A and bisphenol B levels in human blood sera from healthy and endometriotic women, Biomed Chromatogr, 23, 1186, 10.1002/bmc.1241
Kim, 2011, Increased plasma levels of phthalate esters in women with advanced-stage endometriosis: a prospective case-control study, Fertil Steril, 95, 357, 10.1016/j.fertnstert.2010.07.1059
Huang, 2010, Association between phthalate exposure and glutathione S-transferase M1 polymorphism in adenomyosis, leiomyoma and endometriosis, Hum Reprod, 25, 986, 10.1093/humrep/deq015
Weuve, 2010, Association of exposure to phthalates with endometriosis and uterine leiomyomata: findings from NHANES, 1999–2004, Environ Health Perspect, 118, 825, 10.1289/ehp.0901543
Buck Louis, 2013, Bisphenol A and phthalates and endometriosis: the Endometriosis: Natural History, Diagnosis and Outcomes Study, Fertil Steril, 100, 162, 10.1016/j.fertnstert.2013.03.026
Upson, 2013, Phthalates and risk of endometriosis, Environ Res, 126, 91, 10.1016/j.envres.2013.07.003
Itoh, 2009, Urinary phthalate monoesters and endometriosis in infertile Japanese women, Sci Total Environ, 408, 37, 10.1016/j.scitotenv.2009.09.012
Kim, 2010, Increased viability of endometrial cells by in vitro treatment with di-(2-ethylhexyl) phthalate, Fertil Steril, 94, 2413, 10.1016/j.fertnstert.2010.04.027
Upson, 2013, Organochlorine pesticides and risk of endometriosis: findings from a population-based case-control study, Environ Health Perspect, 121, 1319, 10.1289/ehp.1306648
Cooney, 2010, Organochlorine pesticides and endometriosis, Reprod Toxicol, 30, 365, 10.1016/j.reprotox.2010.05.011
Herington, 2011, Immune interactions in endometriosis, Expert Rev Clin Immunol, 7, 611, 10.1586/eci.11.53
Bruner-Tran, 2010, Dioxin and endometrial progesterone resistance, Semin Reprod Med, 28, 59, 10.1055/s-0029-1242995
Wang, 2010, Abnormal regulation of chemokine TECK and its receptor CCR9 in the endometriotic milieu is involved in pathogenesis of endometriosis by way of enhancing invasiveness of endometrial stromal cells, Cell Mol Immunol, 7, 51, 10.1038/cmi.2009.102
Li, 2011, CD82 gene suppression in endometrial stromal cells leads to increase of the cell invasiveness in the endometriotic milieu, J Mol Endocrinol, 47, 195, 10.1530/JME-10-0165
Wang, 2010, The high level of RANTES in the ectopic milieu recruits macrophages and induces their tolerance in progression of endometriosis, J Mol Endocrinol, 45, 291, 10.1677/JME-09-0177
Porpora, 2009, Endometriosis and organochlorinated environmental pollutants: a case-control study on Italian women of reproductive age, Environ Health Perspect, 117, 1070, 10.1289/ehp.0800273
Vichi, 2012, Glutathione transferase polymorphisms and risk of endometriosis associated with polychlorinated biphenyls exposure in Italian women: a gene-environment interaction, Fertil Steril, 97, 1143, 10.1016/j.fertnstert.2012.02.027
Martínez-Zamora, 2015, Increased levels of dioxin-like substances in adipose tissue in patients with deep infiltrating endometriosis, Hum Reprod, 30, 1059, 10.1093/humrep/dev026
Niskar, 2009, Serum dioxins, polychlorinated biphenyls, and endometriosis: a case-control study in Atlanta, Chemosphere, 74, 944, 10.1016/j.chemosphere.2008.10.005
Buck Louis, 2012, Persistent lipophilic environmental chemicals and endometriosis: the ENDO Study, Environ Health Perspect, 120, 811, 10.1289/ehp.1104432
Trabert, 2010, Non-dioxin-like polychlorinated biphenyls and risk of endometriosis, Environ Health Perspect, 118, 1280, 10.1289/ehp.0901444
Shen, 2013, Measurement of phenolic environmental estrogens in women with uterine leiomyoma, PLoS One, 8, e79838, 10.1371/journal.pone.0079838
Zhou, 2013, Measurement of phenolic environmental estrogens in human urine samples by HPLC-MS/MS and primary discussion the possible linkage with uterine leiomyoma, J Chromatogr B Analyt Technol Biomed Life Sci, 938, 80, 10.1016/j.jchromb.2013.08.032
Han, 2011, Bisphenol-A concentrations from leiomyoma patients by LC/MS, Toxicol Res, 27, 49, 10.5487/TR.2011.27.1.049
Shen, 2014, An evidence in vitro for the influence of bisphenol A on uterine leiomyoma, Eur J Obstet Gynecol Reprod Biol, 178, 80, 10.1016/j.ejogrb.2014.03.052
Wang, 2013, Bisphenol A at environmentally relevant doses induces cyclooxygenase-2 expression and promotes invasion of human mesenchymal stem cells derived from uterine myoma tissue, Taiwan J Obstet Gynecol, 52, 246, 10.1016/j.tjog.2013.04.016
Gao, 2010, An endocrine-disrupting chemical, fenvalerate, induces cell cycle progression and collagen type I expression in human uterine leiomyoma and myometrial cells, Toxicol Lett, 196, 133, 10.1016/j.toxlet.2010.03.004
Gao, 2012, An essential role of p27 downregulation in fenvalerate-induced cell growth in human uterine leiomyoma and smooth muscle cells, Am J Physiol Endocrinol Metab, 303, E1025, 10.1152/ajpendo.00107.2012
D'Aloisio, 2012, Early-life exposures and early-onset uterine leiomyomata in black women in the Sister Study, Environ Health Perspect, 120, 406, 10.1289/ehp.1103620
D'Aloisio, 2010, Association of intrauterine and early-life exposures with diagnosis of uterine leiomyomata by 35 years of age in the Sister Study, Environ Health Perspect, 118, 375, 10.1289/ehp.0901423
Mahalingaiah, 2014, Prenatal diethylstilbestrol exposure and risk of uterine leiomyomata in the Nurses' Health Study II, Am J Epidemiol, 179, 186, 10.1093/aje/kwt250
Trabert, 2015, Persistent organic pollutants (POPs) and fibroids: results from the ENDO study, J Expo Sci Environ Epidemiol, 25, 278, 10.1038/jes.2014.31
Lambertino, 2011, Uterine leiomyomata in a cohort of Great Lakes sport fish consumers, Environ Res, 111, 565, 10.1016/j.envres.2011.01.006
Cantonwine, 2010, Bisphenol A exposure in Mexico City and risk of prematurity: a pilot nested case control study, Environ Health, 9, 62, 10.1186/1476-069X-9-62
Cabaton, 2011, Perinatal exposure to environmentally relevant levels of bisphenol A decreases fertility and fecundity in CD-1 mice, Environ Health Perspect, 119, 547, 10.1289/ehp.1002559
Kobayashi, 2012, Lack of effects for dietary exposure of bisphenol A during in utero and lactational periods on reproductive development in rat offspring, J Toxicol Sci, 37, 565, 10.2131/jts.37.565
Adibi, 2009, Maternal urinary metabolites of di-(2-ethylhexyl) phthalate in relation to the timing of labor in a US multicenter pregnancy cohort study, Am J Epidemiol, 169, 1015, 10.1093/aje/kwp001
Meeker, 2009, Urinary phthalate metabolites in relation to preterm birth in Mexico City, Environ Health Perspect, 117, 1587, 10.1289/ehp.0800522
Whyatt, 2009, Prenatal di(2-ethylhexyl)phthalate exposure and length of gestation among an inner-city cohort, Pediatrics, 124, e1213, 10.1542/peds.2009-0325
Ferguson, 2014, Environmental phthalate exposure and preterm birth, JAMA Pediatr, 168, 61, 10.1001/jamapediatrics.2013.3699
Adibi, 2010, Transcriptional biomarkers of steroidogenesis and trophoblast differentiation in the placenta in relation to prenatal phthalate exposure, Environ Health Perspect, 118, 291, 10.1289/ehp.0900788
Ferguson, 2013, Environmental contaminant exposures and preterm birth: a comprehensive review, J Toxicol Environ Health B Crit Rev, 16, 69, 10.1080/10937404.2013.775048
Stillerman, 2008, Environmental exposures and adverse pregnancy outcomes: a review of the science, Reprod Sci, 15, 631, 10.1177/1933719108322436
Mustafa, 2013, Gene-environment interaction in preterm delivery with special reference to organochlorine pesticides, Mol Hum Reprod, 19, 35, 10.1093/molehr/gas039
Rinsky, 2012, Atrazine exposure in public drinking water and preterm birth, Public Health Rep, 127, 72, 10.1177/003335491212700108
Kadhel, 2014, Chlordecone exposure, length of gestation, and risk of preterm birth, Am J Epidemiol, 179, 536, 10.1093/aje/kwt313
Cremonese, 2012, Pesticide exposure and adverse pregnancy events, Southern Brazil, 1996–2000 [in Portuguese], Cad Saude Publica, 28, 1263, 10.1590/S0102-311X2012000700005
Basterrechea, 2014, Prenatal exposure to hexachlorobenzene (HCB) and reproductive effects in a multicentre birth cohort in Spain, Sci Total Environ, 466–467, 770, 10.1016/j.scitotenv.2013.07.053
Ochoa-Acuña, 2009, Drinking-water herbicide exposure in Indiana and prevalence of small-for-gestational-age and preterm delivery, Environ Health Perspect, 117, 1619, 10.1289/ehp.0900784
Ridano, 2012, Chlorpyrifos modifies the expression of genes involved in human placental function, Reprod Toxicol, 33, 331, 10.1016/j.reprotox.2012.01.003
Bruner-Tran, 2011, Developmental exposure to TCDD reduces fertility and negatively affects pregnancy outcomes across multiple generations, Reprod Toxicol, 31, 344, 10.1016/j.reprotox.2010.10.003
Peltier, 2013, 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) enhances placental inflammation, J Reprod Immunol, 98, 10, 10.1016/j.jri.2013.02.005
Ding, 2011, Developmental dioxin exposure of either parent is associated with an increased risk of preterm birth in adult mice, Reprod Toxicol, 31, 351, 10.1016/j.reprotox.2010.11.003
Ranjit, 2010, Bisphenol-A and disparities in birth outcomes: a review and directions for future research, J Perinatol, 30, 2, 10.1038/jp.2009.90
Snijder, 2013, Fetal growth and prenatal exposure to bisphenol A: the generation R study, Environ Health Perspect, 121, 393, 10.1289/ehp.1205296
Burdorf, 2011, The effects of work-related maternal risk factors on time to pregnancy, preterm birth and birth weight: the Generation R Study, Occup Environ Med, 68, 197, 10.1136/oem.2009.046516
Snijder, 2012, Occupational exposure to chemical substances and time to pregnancy: a systematic review, Hum Reprod Update, 18, 284, 10.1093/humupd/dms005
Suzuki, 2012, Foetal exposure to phthalate esters and anogenital distance in male newborns, Int J Androl, 35, 236, 10.1111/j.1365-2605.2011.01190.x
Huang, 2009, Association between prenatal exposure to phthalates and the health of newborns, Environ Int, 35, 14, 10.1016/j.envint.2008.05.012
Philippat, 2012, Exposure to phthalates and phenols during pregnancy and offspring size at birth, Environ Health Perspect, 120, 464, 10.1289/ehp.1103634
Suzuki, 2010, Prenatal exposure to phthalate esters and PAHs and birth outcomes, Environ Int, 36, 699, 10.1016/j.envint.2010.05.003
Pocar, 2012, Exposure to di(2-ethyl-hexyl) phthalate (DEHP) in utero and during lactation causes long-term pituitary-gonadal axis disruption in male and female mouse offspring, Endocrinology, 153, 937, 10.1210/en.2011-1450
Wang, 2011, Study on the effect of di-(2-ethylhexyl) phthalate on pregnant rats and the protection of zinc against it in pregnancy [in Chinese], Zhonghua Fu Chan Ke Za Zhi, 46, 928
Takai, 2009, Collaborative work on evaluation of ovarian toxicity. 10) Two- or four-week repeated dose studies and fertility study of di-(2-ethylhexyl) phthalate (DEHP) in female rats, J Toxicol Sci, 34, SP111, 10.2131/jts.34.S111
Chen, 2010, Perinatal exposure to di-(2-ethylhexyl) phthalate leads to restricted growth and delayed lung maturation in newborn rats, J Perinat Med, 38, 515, 10.1515/jpm.2010.083
Guerra, 2010, Reproductive development and function of female rats exposed to di-eta-butyl-phthalate (DBP) in utero and during lactation, Reprod Toxicol, 29, 99, 10.1016/j.reprotox.2009.10.005
Gemmill, 2013, Residential proximity to methyl bromide use and birth outcomes in an agricultural population in California, Environ Health Perspect, 121, 737, 10.1289/ehp.1205682
Harley, 2011, Association of organophosphate pesticide exposure and paraoxonase with birth outcome in Mexican-American women, PLoS One, 6, e23923, 10.1371/journal.pone.0023923
Migeot, 2013, Drinking-water exposure to a mixture of nitrate and low-dose atrazine metabolites and small-for-gestational age (SGA) babies: a historic cohort study, Environ Res, 122, 58, 10.1016/j.envres.2012.12.007
Sathyanarayana, 2010, Maternal pesticide use and birth weight in the Agricultural Health Study, J Agromedicine, 15, 127, 10.1080/10599241003622699
Chevrier, 2011, Urinary biomarkers of prenatal atrazine exposure and adverse birth outcomes in the PELAGIE birth cohort, Environ Health Perspect, 119, 1034, 10.1289/ehp.1002775
Barr, 2010, Pesticide concentrations in maternal and umbilical cord sera and their relation to birth outcomes in a population of pregnant women and newborns in New Jersey, Sci Total Environ, 408, 790, 10.1016/j.scitotenv.2009.10.007
Acosta-Maldonado, 2009, Effects of exposure to pesticides during pregnancy on placental maturity and weight of newborns: a cross-sectional pilot study in women from the Chihuahua State, Mexico, Hum Exp Toxicol, 28, 451, 10.1177/0960327109107045
Wang, 2012, Organophosphate pesticide exposure and perinatal outcomes in Shanghai, China, Environ Int, 42, 100, 10.1016/j.envint.2011.04.015
Pathak, 2010, Organochlorine pesticide residue levels and oxidative stress in preterm delivery cases, Hum Exp Toxicol, 29, 351, 10.1177/0748233710363334
Bulgaroni, 2013, Environmental pesticide exposure modulates cytokines, arginase and ornithine decarboxylase expression in human placenta, Reprod Toxicol, 39, 23, 10.1016/j.reprotox.2013.03.010
Pathak, 2011, Intra uterine growth retardation: association with organochlorine pesticide residue levels and oxidative stress markers, Reprod Toxicol, 31, 534, 10.1016/j.reprotox.2011.02.007
Bergonzi, 2011, Persistent organochlorine compounds in fetal and maternal tissues: evaluation of their potential influence on several indicators of fetal growth and health, Sci Total Environ, 409, 2888, 10.1016/j.scitotenv.2011.04.031
Wojtyniak, 2010, Association of maternal serum concentrations of 2,2′, 4,4′5,5′-hexachlorobiphenyl (CB-153) and 1,1-dichloro-2,2-bis (p-chlorophenyl)-ethylene (p,p'-DDE) levels with birth weight, gestational age and preterm births in Inuit and European populations, Environ Health, 9, 56, 10.1186/1476-069X-9-56
Al-Saleh, 2012, Levels of DDT and its metabolites in placenta, maternal and cord blood and their potential influence on neonatal anthropometric measures, Sci Total Environ, 416, 62, 10.1016/j.scitotenv.2011.11.020
Lemos, 2011, Response of blastocyst-endometrium interactions in albino rats to sublethal doses of biological and synthetic insecticides, Food Chem Toxicol, 49, 2541, 10.1016/j.fct.2011.06.063
Padmanabhan, 2010, Developmental reprogramming of reproductive and metabolic dysfunction in sheep: native steroids vs. environmental steroid receptor modulators, Int J Androl, 33, 394, 10.1111/j.1365-2605.2009.01024.x
Lam, 2014, The Navigation Guide-evidence-based medicine meets environmental health: integration of animal and human evidence for PFOA effects on fetal growth, Environ Health Perspect, 122, 1040, 10.1289/ehp.1307923
Johnson, 2014, The Navigation Guide-evidence-based medicine meets environmental health: systematic review of human evidence for PFOA effects on fetal growth, Environ Health Perspect, 122, 1028, 10.1289/ehp.1307893
Koustas, 2014, The Navigation Guide-evidence-based medicine meets environmental health: systematic review of nonhuman evidence for PFOA effects on fetal growth, Environ Health Perspect, 122, 1015, 10.1289/ehp.1307177
Konishi, 2009, Prenatal exposure to PCDDs/PCDFs and dioxin-like PCBs in relation to birth weight, Environ Res, 109, 906, 10.1016/j.envres.2009.07.010
Eskenazi, 2013, In utero and childhood polybrominated diphenyl ether (PBDE) exposures and neurodevelopment in the CHAMACOS Study, Environ Health Perspect, 121, 257, 10.1289/ehp.1205597
Tawara, 2009, Effects of maternal dioxin exposure on newborn size at birth among Japanese mother-infant pairs, Environ Health Prev Med, 14, 88, 10.1007/s12199-008-0061-x
Vafeiadi, 2014, Persistent organic pollutants exposure during pregnancy, maternal gestational weight gain, and birth outcomes in the mother-child cohort in Crete, Greece (RHEA study), Environ Int, 64, 116, 10.1016/j.envint.2013.12.015
Dallaire, 2013, Exposure to organochlorines and mercury through fish and marine mammal consumption: associations with growth and duration of gestation among Inuit newborns, Environ Int, 54, 85, 10.1016/j.envint.2013.01.013
Govarts, 2012, Birth weight and prenatal exposure to polychlorinated biphenyls (PCBs) and dichlorodiphenyldichloroethylene (DDE): a meta-analysis within 12 European birth cohorts, Environ Health Perspect, 120, 162, 10.1289/ehp.1103767
Wesselink, 2014, Maternal dioxin exposure and pregnancy outcomes over 30 years of follow-up in Seveso, Environ Int, 63, 143, 10.1016/j.envint.2013.11.005
Petroff, 2011, The aryl hydrocarbon receptor agonist 2,3,7,8-tetrachloro-dibenzo-p-dioxin (TCDD) alters early embryonic development in a rat IVF exposure model, Reprod Toxicol, 32, 286, 10.1016/j.reprotox.2011.07.005
Hotchkiss, 2008, Fifteen years after “Wingspread”–environmental endocrine disrupters and human and wildlife health: where we are today and where we need to go, Toxicol Sci, 105, 235, 10.1093/toxsci/kfn030
Carlsen, 1992, Evidence for decreasing quality of semen during past 50 years, BMJ, 305, 609, 10.1136/bmj.305.6854.609
Sharpe, 1993, Are oestrogens involved in falling sperm counts and disorders of the male reproductive tract?, Lancet, 341, 1392, 10.1016/0140-6736(93)90953-E
Toppari, 1996, Male reproductive health and environmental xenoestrogens, Environ Health Perspect, 104, 741
Fisher, 2003, Human 'testicular dysgenesis syndrome': a possible model using in-utero exposure of the rat to dibutyl phthalate, Hum Reprod, 18, 1383, 10.1093/humrep/deg273
Scott, 2008, Relationship between androgen action in the “male programming window,” fetal Sertoli cell number, and adult testis size in the rat, Endocrinology, 149, 5280, 10.1210/en.2008-0413
Dean, 2013, Clinical review: anogenital distance or digit length ratio as measures of fetal androgen exposure: relationship to male reproductive development and its disorders, J Clin Endocrinol Metab, 98, 2230, 10.1210/jc.2012-4057
Toppari, 2010, Trends in puberty timing in humans and environmental modifiers, Mol Cell Endocrinol, 324, 39, 10.1016/j.mce.2010.03.011
Skakkebaek, 2001, Testicular dysgenesis syndrome: an increasingly common developmental disorder with environmental aspects, Hum Reprod, 16, 972, 10.1093/humrep/16.5.972
Dalgaard, 2012, A genome-wide association study of men with symptoms of testicular dysgenesis syndrome and its network biology interpretation, J Med Genet, 49, 58, 10.1136/jmedgenet-2011-100174
Christiansen, 2012, Mixtures of endocrine disrupting contaminants modelled on human high end exposures: an exploratory study in rats, Int J Androl, 35, 303, 10.1111/j.1365-2605.2011.01242.x
Christiansen, 2009, Synergistic disruption of external male sex organ development by a mixture of four antiandrogens, Environ Health Perspect, 117, 1839, 10.1289/ehp.0900689
Rider, 2010, Cumulative effects of in utero administration of mixtures of reproductive toxicants that disrupt common target tissues via diverse mechanisms of toxicity, Int J Androl, 33, 443, 10.1111/j.1365-2605.2009.01049.x
Toppari, 2010, Cryptorchidism and hypospadias as a sign of testicular dysgenesis syndrome (TDS): environmental connection, Birth Defects Res A Clin Mol Teratol, 88, 910, 10.1002/bdra.20707
Rocheleau, 2009, Pesticides and hypospadias: a meta-analysis, J Pediatr Urol, 5, 17, 10.1016/j.jpurol.2008.08.006
Morales-Suárez-Varela, 2011, Parental occupational exposure to endocrine disrupting chemicals and male genital malformations: a study in the Danish National Birth Cohort study, Environ Health, 10, 3, 10.1186/1476-069X-10-3
Rocheleau, 2011, Maternal occupational pesticide exposure and risk of hypospadias in the National Birth Defects Prevention Study, Birth Defects Res A Clin Mol Teratol, 91, 927, 10.1002/bdra.22860
Nassar, 2010, Parental occupational exposure to potential endocrine disrupting chemicals and risk of hypospadias in infants, Occup Environ Med, 67, 585, 10.1136/oem.2009.048272
Giordano, 2010, Maternal exposures to endocrine disrupting chemicals and hypospadias in offspring, Birth Defects Res A Clin Mol Teratol, 88, 241, 10.1002/bdra.20657
Carmichael, 2010, Hypospadias and halogenated organic pollutant levels in maternal mid-pregnancy serum samples, Chemosphere, 80, 641, 10.1016/j.chemosphere.2010.04.055
Longnecker, 2002, Maternal serum level of 1,1-dichloro-2,2-bis(p-chlorophenyl)ethylene and risk of cryptorchidism, hypospadias, and polythelia among male offspring, Am J Epidemiol, 155, 313, 10.1093/aje/155.4.313
Pierik, 2007, Maternal pregnancy serum level of heptachlor epoxide, hexachlorobenzene, and β-hexachlorocyclohexane and risk of cryptorchidism in offspring, Environ Res, 105, 364, 10.1016/j.envres.2007.04.005
Trabert, 2012, Maternal pregnancy levels of trans-nonachlor and oxychlordane and prevalence of cryptorchidism and hypospadias in boys, Environ Health Perspect, 120, 478, 10.1289/ehp.1103936
McGlynn, 2009, Maternal pregnancy levels of polychlorinated biphenyls and risk of hypospadias and cryptorchidism in male offspring, Environ Health Perspect, 117, 1472, 10.1289/ehp.0800389
Bhatia, 2005, Organochlorine pesticides and male genital anomalies in the child health and development studies, Environ Health Perspect, 113, 220, 10.1289/ehp.7382
Small, 2009, Maternal exposure to a brominated flame retardant and genitourinary conditions in male offspring, Environ Health Perspect, 117, 1175, 10.1289/ehp.0800058
Carmichael, 2003, Hypospadias in California: trends and descriptive epidemiology, Epidemiology, 14, 701, 10.1097/01.ede.0000091603.43531.d0
Hass, 2007, Combined exposure to anti-androgens exacerbates disruption of sexual differentiation in the rat, Environ Health Perspect, 115, 122, 10.1289/ehp.9360
Axelstad, 2014, Mixtures of endocrine-disrupting contaminants induce adverse developmental effects in preweaning rats, Reproduction, 147, 489, 10.1530/REP-13-0447
Virtanen, 2008, Epidemiology and pathogenesis of cryptorchidism, Hum Reprod Update, 14, 49, 10.1093/humupd/dmm027
Acerini, 2009, The descriptive epidemiology of congenital and acquired cryptorchidism in a UK infant cohort, Arch Dis Child, 94, 868, 10.1136/adc.2008.150219
Jensen, 2010, Cryptorchidism concordance in monozygotic and dizygotic twin brothers, full brothers, and half-brothers, Fertil Steril, 93, 124, 10.1016/j.fertnstert.2008.09.041
Boisen, 2004, Difference in prevalence of congenital cryptorchidism in infants between two Nordic countries, Lancet, 363, 1264, 10.1016/S0140-6736(04)15998-9
Damgaard, 2007, Cryptorchidism and maternal alcohol consumption during pregnancy, Environ Health Perspect, 115, 272, 10.1289/ehp.9608
Virtanen, 2006, Mild gestational diabetes as a risk factor for congenital cryptorchidism, J Clin Endocrinol Metab, 91, 4862, 10.1210/jc.2006-1420
Thorup, 2006, The incidence of bilateral cryptorchidism is increased and the fertility potential is reduced in sons born to mothers who have smoked during pregnancy, J Urol, 176, 734, 10.1016/j.juro.2006.03.042
Damgaard, 2008, Risk factors for congenital cryptorchidism in a prospective birth cohort study, PLoS One, 3, e3051, 10.1371/journal.pone.0003051
Kristensen, 2011, Intrauterine exposure to mild analgesics is a risk factor for development of male reproductive disorders in human and rat, Hum Reprod, 26, 235, 10.1093/humrep/deq323
Hosie, 2000, Is there a correlation between organochlorine compounds and undescended testes?, Eur J Pediatr Surg, 10, 304, 10.1055/s-2008-1072381
Damgaard, 2006, Persistent pesticides in human breast milk and cryptorchidism, Environ Health Perspect, 114, 1133, 10.1289/ehp.8741
Fernandez, 2007, Human exposure to endocrine-disrupting chemicals and prenatal risk factors for cryptorchidism and hypospadias: a nested case-control study, Environ Health Perspect, 115, 8, 10.1289/ehp.9351
Krysiak-Baltyn, 2012, Association between chemical pattern in breast milk and congenital cryptorchidism: modelling of complex human exposures, Int J Androl, 35, 294, 10.1111/j.1365-2605.2012.01268.x
Main, 2007, Flame retardants in placenta and breast milk and cryptorchidism in newborn boys, Environ Health Perspect, 115, 1519, 10.1289/ehp.9924
Virtanen, 2012, Associations between congenital cryptorchidism in newborn boys and levels of dioxins and PCBs in placenta, Int J Androl, 35, 283, 10.1111/j.1365-2605.2011.01233.x
Rantakokko, 2013, Association of placenta organotin concentrations with congenital cryptorchidism and reproductive hormone levels in 280 newborn boys from Denmark and Finland, Hum Reprod, 28, 1647, 10.1093/humrep/det040
Vesterholm Jensen, 2014, No association between exposure to perfluorinated compounds and congenital cryptorchidism: a nested case-control study among 215 boys from Denmark and Finland, Reproduction, 147, 411, 10.1530/REP-13-0444
Brucker-Davis, 2008, Cryptorchidism at birth in Nice area (France) is associated with higher prenatal exposure to PCBs and DDE, as assessed by colostrum concentrations, Hum Reprod, 23, 1708, 10.1093/humrep/den186
Swan, 2008, Environmental phthalate exposure in relation to reproductive outcomes and other health endpoints in humans, Environ Res, 108, 177, 10.1016/j.envres.2008.08.007
Main, 2006, Human breast milk contamination with phthalates and alterations of endogenous reproductive hormones in infants three months of age, Environ Health Perspect, 114, 270, 10.1289/ehp.8075
Emmen, 2000, Involvement of insulin-like factor 3 (Insl3) in diethylstilbestrol-induced cryptorchidism, Endocrinology, 141, 846, 10.1210/endo.141.2.7379
Kortenkamp, 2014, Mind the gap: can we explain declining male reproductive health with known antiandrogens?, Reproduction, 147, 515, 10.1530/REP-13-0440
Rajpert-De Meyts, 2006, Developmental model for the pathogenesis of testicular carcinoma in situ: genetic and environmental aspects, Hum Reprod Update, 12, 303, 10.1093/humupd/dmk006
McGlynn, 2005, Increasing incidence of testicular germ cell tumors among black men in the United States, J Clin Oncol, 23, 5757, 10.1200/JCO.2005.08.227
Purdue, 2005, International patterns and trends in testis cancer incidence, Int J Cancer, 115, 822, 10.1002/ijc.20931
Hemminki, 2002, Cancer risks in first-generation immigrants to Sweden, Int J Cancer, 99, 218, 10.1002/ijc.10322
Beranger, 2013, Occupational and environmental exposures associated with testicular germ cell tumours: systematic review of prenatal and life-long exposures, PLoS One, 8, e77130, 10.1371/journal.pone.0077130
Hardell, 2006, In utero exposure to persistent organic pollutants in relation to testicular cancer risk, Int J Androl, 29, 228, 10.1111/j.1365-2605.2005.00622.x
Hardell, 2003, Increased concentrations of polychlorinated biphenyls, hexachlorobenzene, and chlordanes in mothers of men with testicular cancer, Environ Health Perspect, 111, 930, 10.1289/ehp.5816
Hardell, 2004, Concentrations of polychlorinated biphenyls in blood and the risk for testicular cancer, Int J Androl, 27, 282, 10.1111/j.1365-2605.2004.00489.x
Biggs, 2008, Serum organochlorine pesticide residues and risk of testicular germ cell carcinoma: a population-based case-control study, Cancer Epidemiol Biomarkers Prev, 17, 2012, 10.1158/1055-9965.EPI-08-0032
Purdue, 2009, Prediagnostic serum concentrations of organochlorine compounds and risk of testicular germ cell tumors, Environ Health Perspect, 117, 1514, 10.1289/ehp.0800359
Giannandrea, 2011, Pesticide exposure and serum organochlorine residuals among testicular cancer patients and healthy controls, J Environ Sci Health B, 46, 780
Cook, 2011, Organochlorine compounds and testicular dysgenesis syndrome: human data, Int J Androl, 34, e68, 10.1111/j.1365-2605.2011.01171.x
Giannandrea, 2013, Effect of endogenous and exogenous hormones on testicular cancer: the epidemiological evidence, Int J Dev Biol, 57, 255, 10.1387/ijdb.130015fg
Sharpe, 2008, Testicular dysgenesis syndrome: mechanistic insights and potential new downstream effects, Fertil Steril, 89, e33, 10.1016/j.fertnstert.2007.12.026
Ravnborg, 2011, Prenatal and adult exposures to smoking are associated with adverse effects on reproductive hormones, semen quality, final height and body mass index, Hum Reprod, 26, 1000, 10.1093/humrep/der011
Virtanen, 2012, Prenatal exposure to smoking and male reproductive health, Curr Opin Endocrinol Diabetes Obes, 19, 228, 10.1097/MED.0b013e3283537cb8
Vested, 2014, Persistent organic pollutants and male reproductive health, Asian J Androl, 16, 71, 10.4103/1008-682X.122345
Haugen, 2011, Differences in serum levels of CB-153 and p,p'-DDE, and reproductive parameters between men living south and north in Norway, Reprod Toxicol, 32, 261, 10.1016/j.reprotox.2011.06.072
Toft, 2006, Semen quality and exposure to persistent organochlorine pollutants, Epidemiology, 17, 450, 10.1097/01.ede.0000221769.41028.d2
Dallinga, 2002, Decreased human semen quality and organochlorine compounds in blood, Hum Reprod, 17, 1973, 10.1093/humrep/17.8.1973
Hauser, 2003, Lack of an association between environmental exposure to polychlorinated biphenyls and p,p'-DDE and DNA damage in human sperm measured using the neutral comet assay, Hum Reprod, 18, 2525, 10.1093/humrep/deg508
Rozati, 2002, Role of environmental estrogens in the deterioration of male factor fertility, Fertil Steril, 78, 1187, 10.1016/S0015-0282(02)04389-3
Bonde, 2008, Fertility and markers of male reproductive function in Inuit and European populations spanning large contrasts in blood levels of persistent organochlorines, Environ Health Perspect, 116, 269, 10.1289/ehp.10700
Rignell-Hydbom, 2004, Exposure to CB-153 and p,p'-DDE and male reproductive function, Hum Reprod, 19, 2066, 10.1093/humrep/deh362
Richthoff, 2003, Serum levels of 2,2′,4,4′,5,5′-hexachlorobiphenyl (CB-153) in relation to markers of reproductive function in young males from the general Swedish population, Environ Health Perspect, 111, 409, 10.1289/ehp.5767
Hauser, 2003, The relationship between human semen parameters and environmental exposure to polychlorinated biphenyls and p,p'-DDE, Environ Health Perspect, 111, 1505, 10.1289/ehp.6175
Stronati, 2006, Relationships between sperm DNA fragmentation, sperm apoptotic markers and serum levels of CB-153 and p,p'-DDE in European and Inuit populations, Reproduction, 132, 949, 10.1530/rep.1.01034
Rignell-Hydbom, 2005, Exposure to PCBs and p,p'-DDE and human sperm chromatin integrity, Environ Health Perspect, 113, 175, 10.1289/ehp.7252
Spanò, 2005, Exposure to PCB and p, p'-DDE in European and Inuit populations: impact on human sperm chromatin integrity, Hum Reprod, 20, 3488, 10.1093/humrep/dei297
Aneck-Hahn, 2007, Impaired semen quality associated with environmental DDT exposure in young men living in a malaria area in the Limpopo Province, South Africa, J Androl, 28, 423, 10.2164/jandrol.106.001701
De Jager, 2006, Reduced seminal parameters associated with environmental DDT exposure and p,p'-DDE concentrations in men in Chiapas, Mexico: a cross-sectional study, J Androl, 27, 16, 10.2164/jandrol.05121
Guo, 2000, Semen quality after prenatal exposure to polychlorinated biphenyls and dibenzofurans, Lancet, 356, 1240, 10.1016/S0140-6736(00)02792-6
Akutsu, 2008, Polybrominated diphenyl ethers in human serum and sperm quality, Bull Environ Contam Toxicol, 80, 345, 10.1007/s00128-008-9370-4
Abdelouahab, 2011, Polybrominated diphenyl ethers and sperm quality, Reprod Toxicol, 31, 546, 10.1016/j.reprotox.2011.02.005
Joensen, 2013, PFOS (perfluorooctanesulfonate) in serum is negatively associated with testosterone levels, but not with semen quality, in healthy men, Hum Reprod, 28, 599, 10.1093/humrep/des425
Joensen, 2009, Do perfluoroalkyl compounds impair human semen quality?, Environ Health Perspect, 117, 923, 10.1289/ehp.0800517
Toft, 2012, Exposure to perfluorinated compounds and human semen quality in Arctic and European populations, Hum Reprod, 27, 2532, 10.1093/humrep/des185
Raymer, 2012, Concentrations of perfluorooctane sulfonate (PFOS) and perfluorooctanoate (PFOA) and their associations with human semen quality measurements, Reprod Toxicol, 33, 419, 10.1016/j.reprotox.2011.05.024
Specht, 2012, Sperm DNA integrity in relation to exposure to environmental perfluoroalkyl substances - a study of spouses of pregnant women in three geographical regions, Reprod Toxicol, 33, 577, 10.1016/j.reprotox.2012.02.008
Vested, 2013, Associations of in utero exposure to perfluorinated alkyl acids with human semen quality and reproductive hormones in adult men, Environ Health Perspect, 121, 453, 10.1289/ehp.1205118
American Cancer Society, 2014, Cancer Facts & Figures 2014
Jobling, 2012, Evidence for endocrine disruption in humans and wildlife: endocrine disruptors and hormone-related cancers, State of the Science of Endocrine Disrupting Chemicals – 2012, 126
Fenton, 2006, Endocrine-disrupting compounds and mammary gland development: early exposure and later life consequences, Endocrinology, 147, S18, 10.1210/en.2005-1131
Rudel, 2011, Environmental exposures and mammary gland development: state of the science, public health implications, and research recommendations, Environ Health Perspect, 119, 1053, 10.1289/ehp.1002864
Russo, 1987, Development of the human mammary gland, The Mammary Gland: Development, Regulation and Function, 67, 10.1007/978-1-4899-5043-7_3
Russo, 1978, Developmental stage of the rat mammary gland as determinant of its susceptibility to 7,12-dimethylbenz[a]anthracene, J Natl Cancer Inst, 61, 1439
Russo, 1996, Experimentally induced mammary tumors in rats, Breast Cancer Res Treat, 39, 7, 10.1007/BF01806074
Barkan, 2010, Extracellular matrix: a gatekeeper in the transition from dormancy to metastatic growth, Eur J Cancer, 46, 1181, 10.1016/j.ejca.2010.02.027
Van den Berg, 2006, The 2005 World Health Organization reevaluation of human and mammalian toxic equivalency factors for dioxins and dioxin-like compounds, Toxicol Sci, 93, 223, 10.1093/toxsci/kfl055
Safe, 1998, Ah receptor agonists as endocrine disruptors: antiestrogenic activity and mechanisms, Toxicol Lett, 102–103, 343, 10.1016/S0378-4274(98)00331-2
Gray, 1995, In utero 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) alters reproductive morphology and function in female rat offspring, Toxicol Appl Pharmacol, 133, 285, 10.1006/taap.1995.1153
Gray, 1997, In utero exposure to low doses of 2,3,7,8-tetrachlorodibenzo-p-dioxin alters reproductive development of female Long Evans hooded rat offspring, Toxicol Appl Pharmacol, 146, 237, 10.1006/taap.1997.8222
Brown, 1998, Prenatal TCDD and predisposition to mammary cancer in the rat, Carcinogenesis, 19, 1623, 10.1093/carcin/19.9.1623
Chaffin, 1996, In utero and lactational exposure of female Holtzman rats to 2,3,7,8-tetrachlorodibenzo-p-dioxin: modulation of the estrogen signal, Biol Reprod, 55, 62, 10.1095/biolreprod55.1.62
Den Hond, 2002, Sexual maturation in relation to polychlorinated aromatic hydrocarbons: Sharpe and Skakkebaek's hypothesis revisited, Environ Health Perspect, 110, 771, 10.1289/ehp.02110771
Leijs, 2008, Delayed initiation of breast development in girls with higher prenatal dioxin exposure; a longitudinal cohort study, Chemosphere, 73, 999, 10.1016/j.chemosphere.2008.05.053
Bertazzi, 1997, Dioxin exposure and cancer risk: a 15-year mortality study after the “Seveso accident.”, Epidemiology, 8, 646
Bertazzi, 1989, Ten-year mortality study of the population involved in the Seveso incident in 1976, Am J Epidemiol, 129, 1187, 10.1093/oxfordjournals.aje.a115240
Pesatori, 2009, Cancer incidence in the population exposed to dioxin after the “Seveso accident”: twenty years of follow-up, Environ Health, 8, 39, 10.1186/1476-069X-8-39
Warner, 2002, Serum dioxin concentrations and breast cancer risk in the Seveso Women's Health Study, Environ Health Perspect, 110, 625, 10.1289/ehp.02110625
Revich, 2001, Dioxin exposure and public health in Chapaevsk, Russia, Chemosphere, 43, 951, 10.1016/S0045-6535(00)00456-2
Fenton, 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, 10.1093/toxsci/67.1.63
Lewis, 2001, In utero and lactational treatment with 2,3,7,8-tetrachlorodibenzo-p-dioxin impairs mammary gland differentiation but does not block the response to exogenous estrogen in the postpubertal female rat, Toxicol Sci, 62, 46, 10.1093/toxsci/62.1.46
Desaulniers, 2001, Modulatory effects of neonatal exposure to TCDD, or a mixture of PCBs, p,p'-DDT, and p-p'-DDE, on methylnitrosourea-induced mammary tumor development in the rat, Environ Health Perspect, 109, 739
Karmaus, 2005, Maternal concentration of dichlorodiphenyl dichloroethylene (DDE) and initiation and duration of breast feeding, Paediatr Perinat Epidemiol, 19, 388, 10.1111/j.1365-3016.2005.00658.x
Rogan, 1987, Polychlorinated biphenyls (PCBs) and dichlorodiphenyl dichloroethene (DDE) in human milk: effects on growth, morbidity, and duration of lactation, Am J Public Health, 77, 1294, 10.2105/AJPH.77.10.1294
Gladen, 1995, DDE and shortened duration of lactation in a northern Mexican town, Am J Public Health, 85, 504, 10.2105/AJPH.85.4.504
Cupul-Uicab, 2008, DDE, a degradation product of DDT, and duration of lactation in a highly exposed area of Mexico, Environ Health Perspect, 116, 179, 10.1289/ehp.10550
Weldon, 2010, Serum persistent organic pollutants and duration of lactation among Mexican-American women, J Environ Public Health, 2010, 861757, 10.1155/2010/861757
Brody, 2007, Environmental pollutants and breast cancer: epidemiologic studies, Cancer, 109, 2667, 10.1002/cncr.22655
Ingber, 2013, DDT/DDE and breast cancer: a meta-analysis, Regul Toxicol Pharmacol, 67, 421, 10.1016/j.yrtph.2013.08.021
Cohn, 2007, DDT and breast cancer in young women: new data on the significance of age at exposure, Environ Health Perspect, 115, 1406, 10.1289/ehp.10260
White, 2013, Exposure to fogger trucks and breast cancer incidence in the Long Island Breast Cancer Study Project: a case-control study, Environ Health, 12, 24, 10.1186/1476-069X-12-24
Boada, 2012, Complex organochlorine pesticide mixtures as determinant factor for breast cancer risk: a population-based case-control study in the Canary Islands (Spain), Environ Health, 11, 28, 10.1186/1476-069X-11-28
Brown, 1995, Xenoestrogens alter mammary gland differentiation and cell proliferation in the rat, Environ Health Perspect, 103, 708
Kornbrust, 1986, Effects of 1,1-dichloro-2,2-bis[p-chlorophenyl]ethylene (DDE) on lactation in rats, J Toxicol Environ Health, 17, 23, 10.1080/15287398609530799
Johnson, 2012, Accelerated mammary tumor onset in a HER2/Neu mouse model exposed to DDT metabolites locally delivered to the mammary gland, Environ Health Perspect, 120, 1170, 10.1289/ehp.1104327
Fielden, 2002, Normal mammary gland morphology in pubertal female mice following in utero and lactational exposure to genistein at levels comparable to human dietary exposure, Toxicol Lett, 133, 181, 10.1016/S0378-4274(02)00154-6
Hovey, 2005, Effects of neonatal exposure to diethylstilbestrol, tamoxifen, and toremifene on the BALB/c mouse mammary gland, Biol Reprod, 72, 423, 10.1095/biolreprod.104.029769
Tomooka, 1982, Growth of mouse mammary glands after neonatal sex hormone treatment, J Natl Cancer Inst, 69, 1347
Browning, 1958, Effect of feeding low levels of diethylstilbestrol on gestation and lactation of rats, J Nutr, 66, 321, 10.1093/jn/66.3.321
Rothschild, 1987, Transplacental effects of diethylstilbestrol on mammary development and tumorigenesis in female ACI rats, Cancer Res, 47, 4508
Kawaguchi, 2009, Effects of fetal exposure to diethylstilbestrol on mammary tumorigenesis in rats, J Vet Med Sci, 71, 1599, 10.1292/jvms.001599
Ninomiya, 2007, Effects of neonatally administered diethylstilbestrol on induction of mammary carcinomas induced by 7, 12-dimethylbenz(a)anthracene in female rats, Toxicol Pathol, 35, 813, 10.1080/01926230701584205
Doherty, 2010, In utero exposure to diethylstilbestrol (DES) or bisphenol-A (BPA) increases EZH2 expression in the mammary gland: an epigenetic mechanism linking endocrine disruptors to breast cancer, Horm Cancer, 1, 146, 10.1007/s12672-010-0015-9
Lamartiniere, 1992, Neonatal diethylstilbestrol prevents spontaneously developing mammary tumors, Hormonal Carcinogenesis, 305, 10.1007/978-1-4613-9208-8_46
Olsen, 2007, Half-life of serum elimination of perfluorooctanesulfonate, perfluorohexanesulfonate, and perfluorooctanoate in retired fluorochemical production workers, Environ Health Perspect, 115, 1298, 10.1289/ehp.10009
Benninghoff, 2011, Estrogen-like activity of perfluoroalkyl acids in vivo and interaction with human and rainbow trout estrogen receptors in vitro, Toxicol Sci, 120, 42, 10.1093/toxsci/kfq379
Henry, 2013, Comparison of in vitro cytotoxicity, estrogenicity and anti-estrogenicity of triclosan, perfluorooctane sulfonate and perfluorooctanoic acid, J Appl Toxicol, 33, 265, 10.1002/jat.1736
Knox, 2011, Implications of early menopause in women exposed to perfluorocarbons, J Clin Endocrinol Metab, 96, 1747, 10.1210/jc.2010-2401
Keane, Scientific and regulatory policy committee (SRPC) points to consider: histopathology evaluation of the pubertal development and thyroid function assay (OPPTS 890.1450, OPPTS 890.1500) in rats to screen for endocrine disruptors [published online May 6, 2015], Toxicol Pathol
Bonefeld-Jorgensen, 2011, Perfluorinated compounds are related to breast cancer risk in Greenlandic Inuit: a case control study, Environ Health, 10, 88, 10.1186/1476-069X-10-88
Kale, 2015, Breastfeeding versus formula-feeding and girls' pubertal development, Matern Child Health J, 19, 519, 10.1007/s10995-014-1533-9
White, 2007, Gestational PFOA exposure of mice is associated with altered mammary gland development in dams and female offspring, Toxicol Sci, 96, 133, 10.1093/toxsci/kfl177
Lau, 2006, Effects of perfluorooctanoic acid exposure during pregnancy in the mouse, Toxicol Sci, 90, 510, 10.1093/toxsci/kfj105
White, 2009, Effects of perfluorooctanoic acid on mouse mammary gland development and differentiation resulting from cross-foster and restricted gestational exposures, Reprod Toxicol, 27, 289, 10.1016/j.reprotox.2008.11.054
Provenzano, 2008, Collagen density promotes mammary tumor initiation and progression, BMC Med, 6, 11, 10.1186/1741-7015-6-11
White, 2011, Gestational and chronic low-dose PFOA exposures and mammary gland growth and differentiation in three generations of CD-1 mice, Environ Health Perspect, 119, 1070, 10.1289/ehp.1002741
Macon, 2011, Prenatal perfluorooctanoic acid exposure in CD-1 mice: low-dose developmental effects and internal dosimetry, Toxicol Sci, 122, 134, 10.1093/toxsci/kfr076
Zhao, 2012, Perfluorooctanoic acid effects on ovaries mediate its inhibition of peripubertal mammary gland development in Balb/c and C57Bl/6 mice, Reprod Toxicol, 33, 563, 10.1016/j.reprotox.2012.02.004
Albrecht, 2013, A species difference in the peroxisome proliferator-activated receptor α-dependent response to the developmental effects of perfluorooctanoic acid, Toxicol Sci, 131, 568, 10.1093/toxsci/kfs318
Sengupta, 2013, Molecular mechanism of action of bisphenol and bisphenol A mediated by oestrogen receptor α in growth and apoptosis of breast cancer cells, Br J Pharmacol, 169, 167, 10.1111/bph.12122
Pupo, 2012, Bisphenol A induces gene expression changes and proliferative effects through GPER in breast cancer cells and cancer-associated fibroblasts, Environ Health Perspect, 120, 1177, 10.1289/ehp.1104526
Takayanagi, 2006, Endocrine disruptor bisphenol A strongly binds to human estrogen-related receptor γ (ERRγ) with high constitutive activity, Toxicol Lett, 167, 95, 10.1016/j.toxlet.2006.08.012
Krishnan, 1993, Bisphenol-A: an estrogenic substance is released from polycarbonate flasks during autoclaving, Endocrinology, 132, 2279, 10.1210/endo.132.6.8504731
Rochester, 2013, Bisphenol A and human health: a review of the literature, Reprod Toxicol, 42, 132, 10.1016/j.reprotox.2013.08.008
Markey, 2001, In utero exposure to bisphenol A alters the development and tissue organization of the mouse mammary gland, Biol Reprod, 65, 1215, 10.1093/biolreprod/65.4.1215
Muñoz-de-Toro, 2005, Perinatal exposure to bisphenol-A alters peripubertal mammary gland development in mice, Endocrinology, 146, 4138, 10.1210/en.2005-0340
Acevedo, 2013, Perinatally administered bisphenol A as a potential mammary gland carcinogen in rats, Environ Health Perspect, 121, 1040, 10.1289/ehp.1306734
Jenkins, 2009, Oral exposure to bisphenol A increases dimethylbenzanthracene-induced mammary cancer in rats, Environ Health Perspect, 117, 910, 10.1289/ehp.11751
Weber Lozada, 2011, Bisphenol A increases mammary cancer risk in two distinct mouse models of breast cancer, Biol Reprod, 85, 490, 10.1095/biolreprod.110.090431
Vandenberg, 2013, The male mammary gland: a target for the xenoestrogen bisphenol A, Reprod Toxicol, 37, 15, 10.1016/j.reprotox.2013.01.002
Kass, 2015, Prenatal bisphenol A exposure delays the development of the male rat mammary gland, Reprod Toxicol, 54, 37, 10.1016/j.reprotox.2014.02.001
Tharp, 2012, Bisphenol A alters the development of the rhesus monkey mammary gland, Proc Natl Acad Sci USA, 109, 8190, 10.1073/pnas.1120488109
Colón, 2000, Identification of phthalate esters in the serum of young Puerto Rican girls with premature breast development, Environ Health Perspect, 108, 895
López-Carrillo, 2010, Exposure to phthalates and breast cancer risk in northern Mexico, Environ Health Perspect, 118, 539, 10.1289/ehp.0901091
Jobling, 1995, A variety of environmentally persistent chemicals, including some phthalate plasticizers, are weakly estrogenic, Environ Health Perspect, 103, 582, 10.1289/ehp.95103582
Marsman, 1995, NTP technical report on the toxicity studies of dibutyl phthalate (CAS no. 84-74-2) administered in feed to F344/N rats and B6C3F1 mice, Toxic Rep Ser, 30, 1
Lee, 2004, Diverse developmental toxicity of di-n-butyl phthalate in both sexes of rat offspring after maternal exposure during the period from late gestation through lactation, Toxicology, 203, 221, 10.1016/j.tox.2004.06.013
Moral, 2011, In utero exposure to butyl benzyl phthalate induces modifications in the morphology and the gene expression profile of the mammary gland: an experimental study in rats, Environ Health, 10, 5, 10.1186/1476-069X-10-5
Moral, 2007, The plasticizer butyl benzyl phthalate induces genomic changes in rat mammary gland after neonatal/prepubertal exposure, BMC Genom, 8, 453, 10.1186/1471-2164-8-453
Kettles, 1997, Triazine herbicide exposure and breast cancer incidence: an ecologic study of Kentucky counties, Environ Health Perspect, 105, 1222, 10.1289/ehp.971051222
Muir, 2004, Breast cancer incidence and its possible spatial association with pesticide application in two counties of England, Public Health, 118, 513, 10.1016/j.puhe.2003.12.019
Hopenhayn-Rich, 2002, Regional assessment of atrazine exposure and incidence of breast and ovarian cancers in Kentucky, Arch Environ Contam Toxicol, 42, 127, 10.1007/s002440010300
McElroy, 2007, Risk of breast cancer for women living in rural areas from adult exposure to atrazine from well water in Wisconsin, J Expo Sci Environ Epidemiol, 17, 207, 10.1038/sj.jes.7500511
Rayner, 2004, Exposure parameters necessary for delayed puberty and mammary gland development in Long-Evans rats exposed in utero to atrazine, Toxicol Appl Pharmacol, 195, 23, 10.1016/j.taap.2003.11.005
Enoch, 2007, Mammary gland development as a sensitive end point after acute prenatal exposure to an atrazine metabolite mixture in female Long-Evans rats, Environ Health Perspect, 115, 541, 10.1289/ehp.9612
Hovey, 2011, Quantitative assessment of mammary gland development in female Long Evans rats following in utero exposure to atrazine, Toxicol Sci, 119, 380, 10.1093/toxsci/kfq337
Gammon, 2005, A risk assessment of atrazine use in California: human health and ecological aspects, Pest Manag Sci, 61, 331, 10.1002/ps.1000
Eldridge, 1999, The mammary tumor response in triazine-treated female rats: a threshold-mediated interaction with strain and species-specific reproductive senescence, Steroids, 64, 672, 10.1016/S0039-128X(99)00051-3
Wetzel, 1994, Chronic effects of atrazine on estrus and mammary tumor formation in female Sprague-Dawley and Fischer 344 rats, J Toxicol Environ Health, 43, 169, 10.1080/15287399409531913
Fukamachi, 2004, Possible enhancing effects of atrazine and nonylphenol on 7,12-dimethylbenz[a]anthracene-induced mammary tumor development in human c-Ha-ras proto-oncogene transgenic rats, Cancer Sci, 95, 404, 10.1111/j.1349-7006.2004.tb03223.x
Di Cristofano, 2007, Endometrial carcinoma, Annu Rev Pathol, 2, 57, 10.1146/annurev.pathol.2.010506.091905
Allen, 2008, Endogenous sex hormones and endometrial cancer risk in women in the European Prospective Investigation into Cancer and Nutrition (EPIC), Endocr Relat Cancer, 15, 485, 10.1677/ERC-07-0064
Sturgeon, 1998, Serum concentrations of organochlorine compounds and endometrial cancer risk (United States), Cancer Causes Control, 9, 417, 10.1023/A:1008823802393
Kogevinas, 1997, Cancer mortality in workers exposed to phenoxy herbicides, chlorophenols, and dioxins. An expanded and updated international cohort study, Am J Epidemiol, 145, 1061, 10.1093/oxfordjournals.aje.a009069
Hiroi, 2004, Differences in serum bisphenol A concentrations in premenopausal normal women and women with endometrial hyperplasia, Endocr J, 51, 595, 10.1507/endocrj.51.595
Gibson, 2014, Endocrine disruption of oestrogen action and female reproductive tract cancers, Endocr Relat Cancer, 21, T13, 10.1530/ERC-13-0342
Hall, 2013, Endocrine disrupting chemicals promote the growth of ovarian cancer cells via the ER-CXCL12-CXCR4 signaling axis, Mol Carcinog, 52, 715, 10.1002/mc.21913
Beral, 2007, Ovarian cancer and hormone replacement therapy in the Million Women Study, Lancet, 369, 1703, 10.1016/S0140-6736(07)60534-0
Halon, 2011, Estrogen receptor α expression in ovarian cancer predicts longer overall survival, Pathol Oncol Res, 17, 511, 10.1007/s12253-010-9340-0
Donna, 1989, Triazine herbicides and ovarian epithelial neoplasms, Scand J Work Environ Health, 15, 47, 10.5271/sjweh.1882
Young, 2005, Triazine herbicides and epithelial ovarian cancer risk in central California, J Occup Environ Med, 47, 1148, 10.1097/01.jom.0000177044.43959.e8
Alavanja, 2005, Cancer incidence in the Agricultural Health Study, Scand J Work Environ Health, 31, 39
Vieira, 2013, Perfluorooctanoic acid exposure and cancer outcomes in a contaminated community: a geographic analysis, Environ Health Perspect, 121, 318, 10.1289/ehp.1205829
Davis, 2000, Ovarian tumors in rats induced by chronic 2,3,7,8-tetrachlorodibenzo-p-dioxin treatment, Cancer Res, 60, 5414
Cook, 2005, Interaction between genetic susceptibility and early-life environmental exposure determines tumor-suppressor-gene penetrance, Proc Natl Acad Sci USA, 102, 8644, 10.1073/pnas.0503218102
Cook, 2007, Identification of a sensitive period for developmental programming that increases risk for uterine leiomyoma in Eker rats, Reprod Sci, 14, 121, 10.1177/1933719106298401
Kabbarah, 2005, Diethylstilbestrol effects and lymphomagenesis in Mlh1-deficient mice, Int J Cancer, 115, 666, 10.1002/ijc.20918
White, 2011, Endocrine disrupting properties of perfluorooctanoic acid, J Steroid Biochem Mol Biol, 127, 16, 10.1016/j.jsbmb.2011.03.011
Paulose, 2015, Estrogens in the wrong place at the wrong time: fetal BPA exposure and mammary cancer, Reprod Toxicol, 54, 58, 10.1016/j.reprotox.2014.09.012
Prins, 2015, Accessory sex glands in the male, Physiology of Reproduction, Fourth Edition, 773
Goffin, 2011, Prolactin regulation of the prostate gland: a female player in a male game, Nat Rev Urol, 8, 597, 10.1038/nrurol.2011.143
Reiter, 1995, Growth hormone directly affects the function of the different lobes of the rat prostate, Endocrinology, 136, 3338, 10.1210/endo.136.8.7628369
Colao, 1999, Effect of growth hormone (GH) and insulin-like growth factor I on prostate diseases: an ultrasonographic and endocrine study in acromegaly, GH deficiency, and healthy subjects, J Clin Endocrinol Metab, 84, 1986, 10.1210/jcem.84.6.5776
Chen, 2008, Targeting the androgen receptor pathway in prostate cancer, Curr Opin Pharmacol, 8, 440, 10.1016/j.coph.2008.07.005
Knudsen, 2011, Outsmarting androgen receptor: creative approaches for targeting aberrant androgen signaling in advanced prostate cancer, Expert Rev Endocrinol Metab, 6, 483, 10.1586/eem.11.33
Green, 2012, Androgen action and metabolism in prostate cancer, Mol Cell Endocrinol, 360, 3, 10.1016/j.mce.2011.09.046
Bosland, 2005, The role of estrogens in prostate carcinogenesis: a rationale for chemoprevention, Rev Urol, 7, S4
Wang, 2001, A human prostatic epithelial model of hormonal carcinogenesis, Cancer Res, 61, 6064
Hu, 2011, Estrogen-initiated transformation of prostate epithelium derived from normal human prostate stem-progenitor cells, Endocrinology, 152, 2150, 10.1210/en.2010-1377
Coffey, 1990, Clinical and experimental studies of benign prostatic hyperplasia, Urol Clin North Am, 17, 461, 10.1016/S0094-0143(21)00960-5
Prins, 2013, Prostate stem cells, hormones and development, Stem Cells and Prostate Cancer, 1, 10.1007/978-1-4614-6498-3_1
Driscoll, 1980, Effects of prenatal maternal estrogen on the male urogenital system, Obstet Gynecol, 56, 537
Palmer, 2009, Urogenital abnormalities in men exposed to diethylstilbestrol in utero: a cohort study, Environ Health, 8, 37, 10.1186/1476-069X-8-37
Bostwick, 2000, Prostatic intraepithelial neoplasia: the preinvasive stage of prostate cancer. Overview of the prostate committee report, Scand J Urol Nephrol Suppl, 205, 1, 10.1080/003655900750169257-1
Alavanja, 2003, Use of agricultural pesticides and prostate cancer risk in the Agricultural Health Study cohort, Am J Epidemiol, 157, 800, 10.1093/aje/kwg040
Barry, 2012, Methyl bromide exposure and cancer risk in the Agricultural Health Study, Cancer Causes Control, 23, 807, 10.1007/s10552-012-9949-2
Koutros, 2013, Risk of total and aggressive prostate cancer and pesticide use in the Agricultural Health Study, Am J Epidemiol, 177, 59, 10.1093/aje/kws225
Christensen, 2010, Coumaphos exposure and incident cancer among male participants in the Agricultural Health Study (AHS), Environ Health Perspect, 118, 92, 10.1289/ehp.0800446
Koutros, 2013, Genetic susceptibility loci, pesticide exposure and prostate cancer risk, PLoS One, 8, e58195, 10.1371/journal.pone.0058195
Usmani, 2006, Inhibition of the human liver microsomal and human cytochrome P450 1A2 and 3A4 metabolism of estradiol by deployment-related and other chemicals, Drug Metab Dispos, 34, 1606, 10.1124/dmd.106.010439
Usmani, 2003, Inhibition and activation of the human liver microsomal and human cytochrome P450 3A4 metabolism of testosterone by deployment-related chemicals, Drug Metab Dispos, 31, 384, 10.1124/dmd.31.4.384
Uzun, 2009, Malathion-induced testicular toxicity in male rats and the protective effect of vitamins C and E, Food Chem Toxicol, 47, 1903, 10.1016/j.fct.2009.05.001
Laville, 2006, Modulation of aromatase activity and mRNA by various selected pesticides in the human choriocarcinoma JEG-3 cell line, Toxicology, 228, 98, 10.1016/j.tox.2006.08.021
Band, 2011, Prostate cancer risk and exposure to pesticides in British Columbia farmers, Prostate, 71, 168, 10.1002/pros.21232
Cockburn, 2011, Prostate cancer and ambient pesticide exposure in agriculturally intensive areas in California, Am J Epidemiol, 173, 1280, 10.1093/aje/kwr003
Xu, 2010, Associations of serum concentrations of organochlorine pesticides with breast cancer and prostate cancer in U.S. adults, Environ Health Perspect, 118, 60, 10.1289/ehp.0900919
Barry, 2012, Genetic variation in nucleotide excision repair pathway genes, pesticide exposure and prostate cancer risk, Carcinogenesis, 33, 331, 10.1093/carcin/bgr258
Barry, 2011, Genetic variation in base excision repair pathway genes, pesticide exposure, and prostate cancer risk, Environ Health Perspect, 119, 1726, 10.1289/ehp.1103454
Quignot, 2012, A comparison of two human cell lines and two rat gonadal cell primary cultures as in vitro screening tools for aromatase modulation, Toxicol In Vitro, 26, 107, 10.1016/j.tiv.2011.11.004
Koutros, 2011, Xenobiotic-metabolizing gene variants, pesticide use, and the risk of prostate cancer, Pharmacogenet Genomics, 21, 615, 10.1097/FPC.0b013e3283493a57
Bonner, 2010, Occupational exposure to terbufos and the incidence of cancer in the Agricultural Health Study, Cancer Causes Control, 21, 871, 10.1007/s10552-010-9514-9
Karami, 2013, Pesticide exposure and inherited variants in vitamin D pathway genes in relation to prostate cancer, Cancer Epidemiol Biomarkers Prev, 22, 1557, 10.1158/1055-9965.EPI-12-1454
Krishnan, 2003, The role of vitamin D in prostate cancer, Recent Results Cancer Res, 164, 205, 10.1007/978-3-642-55580-0_15
Koutros, 2010, Pesticide use modifies the association between genetic variants on chromosome 8q24 and prostate cancer, Cancer Res, 70, 9224, 10.1158/0008-5472.CAN-10-1078
Institute of Medicine, 2014, Veterans and Agent Orange: Update 2012
Akhtar, 2004, Cancer in US Air Force veterans of the Vietnam War, J Occup Environ Med, 46, 123, 10.1097/01.jom.0000111603.84316.0f
Chamie, 2008, Agent Orange exposure, Vietnam War veterans, and the risk of prostate cancer, Cancer, 113, 2464, 10.1002/cncr.23695
Ansbaugh, 2013, Agent Orange as a risk factor for high-grade prostate cancer, Cancer, 119, 2399, 10.1002/cncr.27941
Shah, 2009, Exposure to Agent Orange is a significant predictor of prostate-specific antigen (PSA)-based recurrence and a rapid PSA doubling time after radical prostatectomy, BJU Int, 103, 1168, 10.1111/j.1464-410X.2009.08405.x
Leng, 2014, 2,3,7,8-Tetrachlorodibezo-p-dioxin exposure and prostate cancer: a meta-analysis of cohort studies, Public Health, 128, 207, 10.1016/j.puhe.2013.10.006
Shen, 1992, Protein-DNA interactions at a dioxin-responsive enhancer. Mutational analysis of the DNA-binding site for the liganded Ah receptor, J Biol Chem, 267, 6815, 10.1016/S0021-9258(19)50499-9
Probst, 1993, Role of the aryl hydrocarbon receptor nuclear translocator protein in aryl hydrocarbon (dioxin) receptor action, Mol Pharmacol, 44, 511
Puga, 2009, The aryl hydrocarbon receptor cross-talks with multiple signal transduction pathways, Biochem Pharmacol, 77, 713, 10.1016/j.bcp.2008.08.031
Matsumura, 2009, The significance of the nongenomic pathway in mediating inflammatory signaling of the dioxin-activated Ah receptor to cause toxic effects, Biochem Pharmacol, 77, 608, 10.1016/j.bcp.2008.10.013
Hu, 2012, Actions of estrogens and endocrine disrupting chemicals on human prostate stem/progenitor cells and prostate cancer risk, Mol Cell Endocrinol, 354, 63, 10.1016/j.mce.2011.08.032
Kollara, 2010, Four and a half LIM domain 2 alters the impact of aryl hydrocarbon receptor on androgen receptor transcriptional activity, J Steroid Biochem Mol Biol, 118, 51, 10.1016/j.jsbmb.2009.09.017
Vezina, 2009, AHR signaling in prostate growth, morphogenesis, and disease, Biochem Pharmacol, 77, 566, 10.1016/j.bcp.2008.09.039
Seachrist, 2015, A review of the carcinogenic potential of bisphenol A, Reprod Toxicol
Tarapore, 2014, Exposure to bisphenol A correlates with early-onset prostate cancer and promotes centrosome amplification and anchorage-independent growth in vitro, PLoS One, 9, e90332, 10.1371/journal.pone.0090332
Prins, 2011, Serum bisphenol A pharmacokinetics and prostate neoplastic responses following oral and subcutaneous exposures in neonatal Sprague-Dawley rats, Reprod Toxicol, 31, 1, 10.1016/j.reprotox.2010.09.009
Prins, 2014, Bisphenol A promotes human prostate stem-progenitor cell self-renewal and increases in vivo carcinogenesis in human prostate epithelium, Endocrinology, 155, 805, 10.1210/en.2013-1955
Ho, 2015, Exposure of human prostaspheres to bisphenol A epigenetically regulates SNORD family non-coding RNAs via histone modification, Endocrinology, 156, 3984, 10.1210/en.2015-1067
Calderon-Gierszal, 2015, Directed differentiation of human embryonic stem cells into prostate organoids in vitro and its perturbation by low-dose bisphenol A exposure, PLoS One, 10, e0133238, 10.1371/journal.pone.0133238
Tomasetti, 2015, Cancer etiology. Variation in cancer risk among tissues can be explained by the number of stem cell divisions, Science, 347, 78, 10.1126/science.1260825
Wetherill, 2006, Bisphenol A facilitates bypass of androgen ablation therapy in prostate cancer, Mol Cancer Ther, 5, 3181, 10.1158/1535-7163.MCT-06-0272
Wetherill, 2007, In vitro molecular mechanisms of bisphenol A action, Reprod Toxicol, 24, 178, 10.1016/j.reprotox.2007.05.010
Derouiche, 2013, Bisphenol A stimulates human prostate cancer cell migration via remodelling of calcium signalling, Springerplus, 2, 54, 10.1186/2193-1801-2-54
De Flora, 2011, Upregulation of clusterin in prostate and DNA damage in spermatozoa from bisphenol A-treated rats and formation of DNA adducts in cultured human prostatic cells, Toxicol Sci, 122, 45, 10.1093/toxsci/kfr096
Wong, 2015, Identification of secretaglobin Scgb2a1 as a target for developmental reprogramming by BPA in the rat prostate, Epigenetics, 10, 127, 10.1080/15592294.2015.1009768
Brandt, 2014, Indole-3-carbinol attenuates the deleterious gestational effects of bisphenol A exposure on the prostate gland of male F1 rats, Reprod Toxicol, 43, 56, 10.1016/j.reprotox.2013.11.001
Prins, 2001, Estrogen imprinting of the developing prostate gland is mediated through stromal estrogen receptor α: studies with αERKO and βERKO mice, Cancer Res, 61, 6089
Richter, 2007, Estradiol and bisphenol A stimulate androgen receptor and estrogen receptor gene expression in fetal mouse prostate mesenchyme cells, Environ Health Perspect, 115, 902, 10.1289/ehp.9804
Arase, 2011, Endocrine disrupter bisphenol A increases in situ estrogen production in the mouse urogenital sinus, Biol Reprod, 84, 734, 10.1095/biolreprod.110.087502
Selvakumar, 2011, Differential expression of androgen and estrogen receptors in PCB (Aroclor 1254)-exposed rat ventral prostate: impact of alpha-tocopherol, Exp Toxicol Pathol, 63, 105, 10.1016/j.etp.2009.10.003
Castro, 2013, Bisphenol A exposure during adulthood alters expression of aromatase and 5α-reductase isozymes in rat prostate, PLoS One, 8, e55905, 10.1371/journal.pone.0055905
Ruder, 2014, Mortality among 24,865 workers exposed to polychlorinated biphenyls (PCBs) in three electrical capacitor manufacturing plants: a ten-year update, Int J Hyg Environ Health, 217, 176, 10.1016/j.ijheh.2013.04.006
Sawada, 2010, Plasma organochlorines and subsequent risk of prostate cancer in Japanese men: a nested case-control study, Environ Health Perspect, 118, 659, 10.1289/ehp.0901214
Aronson, 2010, Plasma organochlorine levels and prostate cancer risk, J Expo Sci Environ Epidemiol, 20, 434, 10.1038/jes.2009.33
Endo, 2003, Effects of single non-ortho, mono-ortho, and di-ortho chlorinated biphenyls on cell functions and proliferation of the human prostatic carcinoma cell line, LNCaP, Reprod Toxicol, 17, 229, 10.1016/S0890-6238(02)00126-0
Zhu, 2013, A new player in environmentally induced oxidative stress: polychlorinated biphenyl congener, 3,3′-dichlorobiphenyl (PCB11), Toxicol Sci, 136, 39, 10.1093/toxsci/kft186
Zhu, 2009, Polychlorinated-biphenyl-induced oxidative stress and cytotoxicity can be mitigated by antioxidants after exposure, Free Radic Biol Med, 47, 1762, 10.1016/j.freeradbiomed.2009.09.024
Cowin, 2010, Vinclozolin exposure in utero induces postpubertal prostatitis and reduces sperm production via a reversible hormone-regulated mechanism, Endocrinology, 151, 783, 10.1210/en.2009-0982
Kavlock, 2005, Mode of action: inhibition of androgen receptor function–vinclozolin-induced malformations in reproductive development, Crit Rev Toxicol, 35, 721, 10.1080/10408440591007377
Yu, 2004, Reproductive disorders in pubertal and adult phase of the male rats exposed to vinclozolin during puberty, J Vet Med Sci, 66, 847, 10.1292/jvms.66.847
Cowin, 2008, Early-onset endocrine disruptor-induced prostatitis in the rat, Environ Health Perspect, 116, 923, 10.1289/ehp.11239
Doolan, 2014, An update on occupation and prostate cancer, Asian Pac J Cancer Prev, 15, 501, 10.7314/APJCP.2014.15.2.501
Yang, 2008, Does arsenic exposure increase the risk for prostate cancer?, J Toxicol Environ Health A, 71, 1559, 10.1080/15287390802392065
Davey, 2007, Arsenic as an endocrine disruptor: effects of arsenic on estrogen receptor-mediated gene expression in vivo and in cell culture, Toxicol Sci, 98, 75, 10.1093/toxsci/kfm013
Davey, 2008, Arsenic as an endocrine disruptor: arsenic disrupts retinoic acid receptor-and thyroid hormone receptor-mediated gene regulation and thyroid hormone-mediated amphibian tail metamorphosis, Environ Health Perspect, 116, 165, 10.1289/ehp.10131
Zhou, 2011, Arsenite interacts selectively with zinc finger proteins containing C3H1 or C4 motifs, J Biol Chem, 286, 22855, 10.1074/jbc.M111.232926
García-Esquinas, 2013, Arsenic exposure and cancer mortality in a US-based prospective cohort: the Strong Heart Study, Cancer Epidemiol Biomarkers Prev, 22, 1944, 10.1158/1055-9965.EPI-13-0234-T
Tokar, 2010, Cancer in experimental animals exposed to arsenic and arsenic compounds, Crit Rev Toxicol, 40, 912, 10.3109/10408444.2010.506641
Benbrahim-Tallaa, 2008, Inorganic arsenic and human prostate cancer, Environ Health Perspect, 116, 158, 10.1289/ehp.10423
Treas, 2013, Chronic exposure to arsenic, estrogen, and their combination causes increased growth and transformation in human prostate epithelial cells potentially by hypermethylation-mediated silencing of MLH1, Prostate, 73, 1660, 10.1002/pros.22701
Ngalame, 2014, Aberrant microRNA expression likely controls RAS oncogene activation during malignant transformation of human prostate epithelial and stem cells by arsenic, Toxicol Sci, 138, 268, 10.1093/toxsci/kfu002
Tokar, 2010, Arsenic-specific stem cell selection during malignant transformation, J Natl Cancer Inst, 102, 638, 10.1093/jnci/djq093
Xu, 2012, Arsenic-transformed malignant prostate epithelia can convert noncontiguous normal stem cells into an oncogenic phenotype, Environ Health Perspect, 120, 865, 10.1289/ehp.1204987
Tokar, 2011, Arsenic, stem cells, and the developmental basis of adult cancer, Toxicol Sci, 120, S192, 10.1093/toxsci/kfq342
Kortenkamp, 2011, Are cadmium and other heavy metal compounds acting as endocrine disrupters?, Met Ions Life Sci, 8, 305
Mullins, 2012, Environmental exposures and prostate cancer, Urol Oncol, 30, 216, 10.1016/j.urolonc.2011.11.014
García-Esquinas, 2014, Cadmium exposure and cancer mortality in a prospective cohort: the Strong Heart Study, Environ Health Perspect, 122, 363, 10.1289/ehp.1306587
Cheung, 2014, Association between urinary cadmium and all cause, all cancer and prostate cancer specific mortalities for men: an analysis of National Health and Nutrition Examination Survey (NHANES III) data, Asian Pac J Cancer Prev, 15, 483, 10.7314/APJCP.2014.15.1.483
Julin, 2012, Dietary cadmium exposure and prostate cancer incidence: a population-based prospective cohort study, Br J Cancer, 107, 895, 10.1038/bjc.2012.311
Prajapati, 2014, A single low dose of cadmium exposure induces benign prostate hyperplasia like condition in rat: A novel benign prostate hyperplasia rodent model, Exp Biol Med (Maywood), 239, 829, 10.1177/1535370214536118
Lacorte, 2011, Early changes induced by short-term low-dose cadmium exposure in rat ventral and dorsolateral prostates, Microsc Res Tech, 74, 988, 10.1002/jemt.20985
Xu, 2013, Recruitment of normal stem cells to an oncogenic phenotype by noncontiguous carcinogen-transformed epithelia depends on the transforming carcinogen, Environ Health Perspect, 121, 944, 10.1289/ehp.1306714
Andersen, 2002, Narrow individual variations in serum T(4) and T(3) in normal subjects: a clue to the understanding of subclinical thyroid disease, J Clin Endocrinol Metab, 87, 1068, 10.1210/jcem.87.3.8165
Andersen, 2003, Biologic variation is important for interpretation of thyroid function tests, Thyroid, 13, 1069, 10.1089/105072503770867237
Fekete, 2014, Central regulation of hypothalamic-pituitary-thyroid axis under physiological and pathophysiological conditions, Endocr Rev, 35, 159, 10.1210/er.2013-1087
Gilbert, 2011, Impact of low-level thyroid hormone disruption induced by propylthiouracil on brain development and function, Toxicol Sci, 124, 432, 10.1093/toxsci/kfr244
Kampf-Lassin, 2013, Acute downregulation of type II and type III iodothyronine deiodinases by photoperiod in peripubertal male and female Siberian hamsters, Gen Comp Endocrinol, 193, 72, 10.1016/j.ygcen.2013.07.010
Zoeller, 2010, Environmental chemicals targeting thyroid, Hormones (Athens), 9, 28, 10.14310/horm.2002.1250
Bansal, 2014, Polybrominated diphenyl ether (DE-71) interferes with thyroid hormone action independent of effects on circulating levels of thyroid hormone in male rats, Endocrinology, 155, 4104, 10.1210/en.2014-1154
Triggiani, 2009, Role of iodine, selenium and other micronutrients in thyroid function and disorders, Endocr Metab Immune Disord Drug Targets, 9, 277, 10.2174/187153009789044392
Braverman, 2004, Werner and Ingbar's the Thyroid: A Fundamental and Clinical Text
Zimmermann, 2012, The effects of iodine deficiency in pregnancy and infancy, Paediatr Perinat Epidemiol, 26, 108, 10.1111/j.1365-3016.2012.01275.x
Burniat, 2012, Iodotyrosine deiodinase defect identified via genome-wide approach, J Clin Endocrinol Metab, 97, E1276, 10.1210/jc.2011-3314
Dumitrescu, 2011, Inherited defects of thyroid hormone metabolism, Ann Endocrinol (Paris), 72, 95, 10.1016/j.ando.2011.03.011
Szabo, 2009, Effects of perinatal PBDE exposure on hepatic phase I, phase II, phase III, and deiodinase 1 gene expression involved in thyroid hormone metabolism in male rat pups, Toxicol Sci, 107, 27, 10.1093/toxsci/kfn230
Council on Environmental Health, 2014, Iodine deficiency, pollutant chemicals, and the thyroid: new information on an old problem, Pediatrics, 133, 1163, 10.1542/peds.2014-0900
Leung, 2014, Environmental perchlorate exposure: potential adverse thyroid effects, Curr Opin Endocrinol Diabetes Obes, 21, 372, 10.1097/MED.0000000000000090
Taylor, 2014, Maternal perchlorate levels in women with borderline thyroid function during pregnancy and the cognitive development of their offspring: data from the Controlled Antenatal Thyroid Study, J Clin Endocrinol Metab, 99, 4291, 10.1210/jc.2014-1901
Köhrle, 2008, Environment and endocrinology: the case of thyroidology, Ann Endocrinol (Paris), 69, 116, 10.1016/j.ando.2008.02.008
Zoeller, 2007, Current and potential rodent screens and tests for thyroid toxicants, Crit Rev Toxicol, 37, 55, 10.1080/10408440601123461
Paul, 2014, Development of a thyroperoxidase inhibition assay for high-throughput screening, Chem Res Toxicol, 27, 387, 10.1021/tx400310w
Song, 2012, Changes in thyroid peroxidase activity in response to various chemicals, J Environ Monit, 14, 2121, 10.1039/c2em30106g
Howdeshell, 2002, A model of the development of the brain as a construct of the thyroid system, Environ Health Perspect, 110, 337, 10.1289/ehp.02110s3337
Montaño, 2012, New approaches to assess the transthyretin binding capacity of bioactivated thyroid hormone disruptors, Toxicol Sci, 130, 94, 10.1093/toxsci/kfs228
Cao, 2011, In vitro fluorescence displacement investigation of thyroxine transport disruption by bisphenol A, J Environ Sci (China), 23, 315, 10.1016/S1001-0742(10)60408-1
Gutleb, 2010, In vitro assay shows that PCB metabolites completely saturate thyroid hormone transport capacity in blood of wild polar bears (Ursus maritimus), Environ Sci Technol, 44, 3149, 10.1021/es903029j
Cao, 2010, Structure-based investigation on the binding interaction of hydroxylated polybrominated diphenyl ethers with thyroxine transport proteins, Toxicology, 277, 20, 10.1016/j.tox.2010.08.012
Weiss, 2009, Competitive binding of poly- and perfluorinated compounds to the thyroid hormone transport protein transthyretin, Toxicol Sci, 109, 206, 10.1093/toxsci/kfp055
Marchesini, 2008, Biosensor discovery of thyroxine transport disrupting chemicals, Toxicol Appl Pharmacol, 232, 150, 10.1016/j.taap.2008.06.014
Moreno, 2010, Genetics and phenomics of hypothyroidism and goiter due to iodotyrosine deiodinase (DEHAL1) gene mutations, Mol Cell Endocrinol, 322, 91, 10.1016/j.mce.2010.03.010
Shimizu, 2014, Iodotyrosine deiodinase, a novel target of environmental halogenated chemicals for disruption of the thyroid hormone system in mammals, Biol Pharm Bull, 37, 1430, 10.1248/bpb.b14-00240
Shimizu, 2013, Structure-activity relationships of 44 halogenated compounds for iodotyrosine deiodinase-inhibitory activity, Toxicology, 314, 22, 10.1016/j.tox.2013.08.017
Roques, 2013, The nuclear receptors pregnane X receptor and constitutive androstane receptor contribute to the impact of fipronil on hepatic gene expression linked to thyroid hormone metabolism, Biochem Pharmacol, 86, 997, 10.1016/j.bcp.2013.08.012
Brucker-Davis, 1998, Effects of environmental synthetic chemicals on thyroid function, Thyroid, 8, 827, 10.1089/thy.1998.8.827
Greer, 2002, Health effects assessment for environmental perchlorate contamination: the dose response for inhibition of thyroidal radioiodine uptake in humans, Environ Health Perspect, 110, 927, 10.1289/ehp.02110927
Blount, 2006, Urinary perchlorate and thyroid hormone levels in adolescent and adult men and women living in the United States, Environ Health Perspect, 114, 1865, 10.1289/ehp.9466
Steinmaus, 2007, Impact of smoking and thiocyanate on perchlorate and thyroid hormone associations in the 2001–2002 National Health and Nutrition Examination Survey, Environ Health Perspect, 115, 1333, 10.1289/ehp.10300
Steinmaus, 2013, Combined effects of perchlorate, thiocyanate, and iodine on thyroid function in the National Health and Nutrition Examination Survey 2007–08, Environ Res, 123, 17, 10.1016/j.envres.2013.01.005
Zoeller, 2004, Timing of thyroid hormone action in the developing brain: clinical observations and experimental findings, J Neuroendocrinol, 16, 809, 10.1111/j.1365-2826.2004.01243.x
Pearce, 2007, Breast milk iodine and perchlorate concentrations in lactating Boston-area women, J Clin Endocrinol Metab, 92, 1673, 10.1210/jc.2006-2738
Ginsberg, 2007, Evaluation of the U.S. EPA/OSWER preliminary remediation goal for perchlorate in groundwater: focus on exposure to nursing infants, Environ Health Perspect, 115, 361, 10.1289/ehp.9533
Lawrence, 2001, Low dose perchlorate (3 mg daily) and thyroid function, Thyroid, 11, 295, 10.1089/105072501750159796
Lawrence, 2000, The effect of short-term low-dose perchlorate on various aspects of thyroid function, Thyroid, 10, 659, 10.1089/10507250050137734
Gilbert, 2008, Developmental exposure to perchlorate alters synaptic transmission in hippocampus of the adult rat, Environ Health Perspect, 116, 752, 10.1289/ehp.11089
Schantz, 2003, Effects of PCB exposure on neuropsychological function in children, Environ Health Perspect, 111, 357, 10.1289/ehp.5461
Hagmar, 2003, Polychlorinated biphenyls and thyroid status in humans: a review, Thyroid, 13, 1021, 10.1089/105072503770867192
El Majidi, 2014, Systematic analysis of the relationship between standardized biological levels of polychlorinated biphenyls and thyroid function in pregnant women and newborns, Chemosphere, 98, 1, 10.1016/j.chemosphere.2013.10.006
Giera, 2013, Effects and predicted consequences of persistent and bioactive organic pollutants on thyroid function, Effects of Persistent and Bioactive Organic Pollutants on Human Health, 203, 10.1002/9781118679654.ch9
Erickson, 1986, Analytical Chemistry of PCBs
Zoeller, 2000, Developmental exposure to polychlorinated biphenyls exerts thyroid hormone-like effects on the expression of RC3/neurogranin and myelin basic protein messenger ribonucleic acids in the developing rat brain, Endocrinology, 141, 181, 10.1210/endo.141.1.7273
Londono, 2010, Hydroxylated PCB induces Ca(2+) oscillations and alterations of membrane potential in cultured cortical cells, J Appl Toxicol, 30, 334, 10.1002/jat.1501
Miyazaki, 2008, Identification of the functional domain of thyroid hormone receptor responsible for polychlorinated biphenyl-mediated suppression of its action in vitro, Environ Health Perspect, 116, 1231, 10.1289/ehp.11176
Gauger, 2007, Polychlorinated biphenyls 105 and 118 form thyroid hormone receptor agonists after cytochrome P4501A1 activation in rat pituitary GH3 cells, Environ Health Perspect, 115, 1623, 10.1289/ehp.10328
Giera, 2011, Individual polychlorinated biphenyl (PCB) congeners produce tissue- and gene-specific effects on thyroid hormone signaling during development, Endocrinology, 152, 2909, 10.1210/en.2010-1490
Wadzinski, 2014, Endocrine disruption in human placenta: expression of the dioxin-inducible enzyme, CYP1A1, is correlated with that of thyroid hormone-regulated genes, J Clin Endocrinol Metab, 99, E2735, 10.1210/jc.2014-2629
Hofmann, 2009, Interference of endocrine disrupters with thyroid hormone receptor-dependent transactivation, Toxicol Sci, 110, 125, 10.1093/toxsci/kfp086
Frederiksen, 2009, Human internal and external exposure to PBDEs–a review of levels and sources, Int J Hyg Environ Health, 212, 109, 10.1016/j.ijheh.2008.04.005
Costa, 2014, A mechanistic view of polybrominated diphenyl ether (PBDE) developmental neurotoxicity, Toxicol Lett, 230, 282, 10.1016/j.toxlet.2013.11.011
Herbstman, 2010, Prenatal exposure to PBDEs and neurodevelopment, Environ Health Perspect, 118, 712, 10.1289/ehp.0901340
Suvorov, 2009, Perinatal exposure to low-dose BDE-47, an emergent environmental contaminant, causes hyperactivity in rat offspring, Neonatology, 95, 203, 10.1159/000155651
Rice, 2007, Developmental delays and locomotor activity in the C57BL6/J mouse following neonatal exposure to the fully-brominated PBDE, decabromodiphenyl ether, Neurotoxicol Teratol, 29, 511, 10.1016/j.ntt.2007.03.061
Viberg, 2006, Neonatal exposure to higher brominated diphenyl ethers, hepta-, octa-, or nonabromodiphenyl ether, impairs spontaneous behavior and learning and memory functions of adult mice, Toxicol Sci, 92, 211, 10.1093/toxsci/kfj196
Eriksson, 2006, Polybrominated diphenyl ethers, a group of brominated flame retardants, can interact with polychlorinated biphenyls in enhancing developmental neurobehavioral defects, Toxicol Sci, 94, 302, 10.1093/toxsci/kfl109
Gascon, 2012, Polybrominated diphenyl ethers (PBDEs) in breast milk and neuropsychological development in infants, Environ Health Perspect, 120, 1760, 10.1289/ehp.1205266
Gascon, 2011, Effects of pre and postnatal exposure to low levels of polybromodiphenyl ethers on neurodevelopment and thyroid hormone levels at 4 years of age, Environ Int, 37, 605, 10.1016/j.envint.2010.12.005
Kim, 2011, Para- and ortho-substitutions are key determinants of polybrominated diphenyl ether activity toward ryanodine receptors and neurotoxicity, Environ Health Perspect, 119, 519, 10.1289/ehp.1002728
Kuriyama, 2007, Developmental exposure to low-dose PBDE-99: tissue distribution and thyroid hormone levels, Toxicology, 242, 80, 10.1016/j.tox.2007.09.011
Ellis-Hutchings, 2006, Polybrominated diphenyl ether (PBDE)-induced alterations in vitamin A and thyroid hormone concentrations in the rat during lactation and early postnatal development, Toxicol Appl Pharmacol, 215, 135, 10.1016/j.taap.2006.02.008
Hallgren, 2001, Effects of polybrominated diphenyl ethers (PBDEs) and polychlorinated biphenyls (PCBs) on thyroid hormone and vitamin A levels in rats and mice, Arch Toxicol, 75, 200, 10.1007/s002040000208
Ren, 2013, Hydroxylated polybrominated diphenyl ethers exhibit different activities on thyroid hormone receptors depending on their degree of bromination, Toxicol Appl Pharmacol, 268, 256, 10.1016/j.taap.2013.01.026
Li, 2010, Hormone activity of hydroxylated polybrominated diphenyl ethers on human thyroid receptor-β: in vitro and in silico investigations, Environ Health Perspect, 118, 602, 10.1289/ehp.0901457
Ibhazehiebo, 2011, Disruption of thyroid hormone receptor-mediated transcription and thyroid hormone-induced Purkinje cell dendrite arborization by polybrominated diphenyl ethers, Environ Health Perspect, 119, 168, 10.1289/ehp.1002065
Nakamura, 2013, Cell type-dependent agonist/antagonist activities of polybrominated diphenyl ethers, Toxicol Lett, 223, 192, 10.1016/j.toxlet.2013.09.007
Dingemans, 2011, Neurotoxicity of brominated flame retardants: (in)direct effects of parent and hydroxylated polybrominated diphenyl ethers on the (developing) nervous system, Environ Health Perspect, 119, 900, 10.1289/ehp.1003035
Huang, 2007, Associations between urinary phthalate monoesters and thyroid hormones in pregnant women, Hum Reprod, 22, 2715, 10.1093/humrep/dem205
Janjua, 2007, Systemic uptake of diethyl phthalate, dibutyl phthalate, and butyl paraben following whole-body topical application and reproductive and thyroid hormone levels in humans, Environ Sci Technol, 41, 5564, 10.1021/es0628755
Meeker, 2007, Di(2-ethylhexyl) phthalate metabolites may alter thyroid hormone levels in men, Environ Health Perspect, 115, 1029, 10.1289/ehp.9852
Dirtu, 2013, Phthalate metabolites in obese individuals undergoing weight loss: urinary levels and estimation of the phthalates daily intake, Environ Int, 59, 344, 10.1016/j.envint.2013.06.023
Wu, 2013, Intake of phthalate-tainted foods alters thyroid functions in Taiwanese children, PLoS One, 8, e55005, 10.1371/journal.pone.0055005
National Toxicology Program, 1983, NTP carcinogenesis bioassay of diallyl phthalate (CAS no. 131-17-9) in B6C3F1 mice (gavage study), Natl Toxicol Program Tech Rep Ser, 242, 1
Hinton, 1986, Effects of phthalic acid esters on the liver and thyroid, Environ Health Perspect, 70, 195, 10.1289/ehp.8670195
O'Connor, 2002, Evaluation of a 15-day screening assay using intact male rats for identifying antiandrogens, Toxicol Sci, 69, 92, 10.1093/toxsci/69.1.92
Wenzel, 2005, Modulation of iodide uptake by dialkyl phthalate plasticisers in FRTL-5 rat thyroid follicular cells, Mol Cell Endocrinol, 244, 63, 10.1016/j.mce.2005.02.008
Shimada, 2004, Characteristics of 3,5,3′-triiodothyronine (T3)-uptake system of tadpole red blood cells: effect of endocrine-disrupting chemicals on cellular T3 response, J Endocrinol, 183, 627, 10.1677/joe.1.05893
Ibhazehiebo, 2011, Thyroid hormone receptor-mediated transcription is suppressed by low dose phthalate, Niger J Physiol Sci, 26, 143
Ghisari, 2009, Effects of plasticizers and their mixtures on estrogen receptor and thyroid hormone functions, Toxicol Lett, 189, 67, 10.1016/j.toxlet.2009.05.004
Kim, 2014, Tetrabromobisphenol A and hexabromocyclododecane flame retardants in infant-mother paired serum samples, and their relationships with thyroid hormones and environmental factors, Environ Pollut, 184, 193, 10.1016/j.envpol.2013.08.034
Wang, 2013, Urinary bisphenol A concentration and thyroid function in Chinese adults, Epidemiology, 24, 295, 10.1097/EDE.0b013e318280e02f
Sriphrapradang, 2013, Association between bisphenol A and abnormal free thyroxine level in men, Endocrine, 44, 441, 10.1007/s12020-013-9889-y
Teeguarden, 2005, Evaluation of oral and intravenous route pharmacokinetics, plasma protein binding, and uterine tissue dose metrics of bisphenol A: a physiologically based pharmacokinetic approach, Toxicol Sci, 85, 823, 10.1093/toxsci/kfi135
Domoradzki, 2004, Age and dose dependency of the pharmacokinetics and metabolism of bisphenol A in neonatal Sprague-Dawley rats following oral administration, Toxicol Sci, 77, 230, 10.1093/toxsci/kfh054
Moriyama, 2002, Thyroid hormone action is disrupted by bisphenol A as an antagonist, J Clin Endocrinol Metab, 87, 5185, 10.1210/jc.2002-020209
Kitamura, 2002, Thyroid hormonal activity of the flame retardants tetrabromobisphenol A and tetrachlorobisphenol A, Biochem Biophys Res Commun, 293, 554, 10.1016/S0006-291X(02)00262-0
Freitas, 2011, Detection of thyroid hormone receptor disruptors by a novel stable in vitro reporter gene assay, Toxicol In Vitro, 25, 257, 10.1016/j.tiv.2010.08.013
Sun, 2009, Anti-thyroid hormone activity of bisphenol A, tetrabromobisphenol A and tetrachlorobisphenol A in an improved reporter gene assay, Toxicol In Vitro, 23, 950, 10.1016/j.tiv.2009.05.004
Sheng, 2012, Low concentrations of bisphenol A suppress thyroid hormone receptor transcription through a nongenomic mechanism, Toxicol Appl Pharmacol, 259, 133, 10.1016/j.taap.2011.12.018
Zoeller, 2005, Bisphenol-A, an environmental contaminant that acts as a thyroid hormone receptor antagonist in vitro, increases serum thyroxine, and alters RC3/neurogranin expression in the developing rat brain, Endocrinology, 146, 607, 10.1210/en.2004-1018
Siesser, 2005, Hyperactivity, impaired learning on a vigilance task, and a differential response to methylphenidate in the TRβPV knock-in mouse, Psychopharmacology (Berl), 181, 653, 10.1007/s00213-005-0024-5
McDonald, 1998, Hyperactivity and learning deficits in transgenic mice bearing a human mutant thyroid hormone β1 receptor gene, Learn Mem, 5, 289, 10.1101/lm.5.4.289
Weiss, 1997, Behavioral effects of liothyronine (L-T3) in children with attention deficit hyperactivity disorder in the presence and absence of resistance to thyroid hormone, Thyroid, 7, 389, 10.1089/thy.1997.7.389
Hauser, 1993, Attention deficit-hyperactivity disorder in people with generalized resistance to thyroid hormone, N Engl J Med, 328, 997, 10.1056/NEJM199304083281403
Gore, 2008, Developmental programming and endocrine disruptor effects on reproductive neuroendocrine systems, Front Neuroendocrinol, 29, 358, 10.1016/j.yfrne.2008.02.002
Segner, 2013, Impact of environmental estrogens on Yfish considering the diversity of estrogen signaling, Gen Comp Endocrinol, 191, 190, 10.1016/j.ygcen.2013.05.015
Le Page, 2011, Neuroendocrine effects of endocrine disruptors in teleost fish, J Toxicol Environ Health B Crit Rev, 14, 370, 10.1080/10937404.2011.578558
Rempel, 2008, Effects of environmental estrogens and antiandrogens on endocrine function, gene regulation, and health in fish, Int Rev Cell Mol Biol, 267, 207, 10.1016/S1937-6448(08)00605-9
Kinch, 2015, Low-dose exposure to bisphenol A and replacement bisphenol S induces precocious hypothalamic neurogenesis in embryonic zebrafish, Proc Natl Acad Sci USA, 112, 1475, 10.1073/pnas.1417731112
Picot, 2014, Vulnerability of the neural circuitry underlying sexual behavior to chronic adult exposure to oral bisphenol A in male mice, Endocrinology, 155, 502, 10.1210/en.2013-1639
Chen, 2014, Sex differences in the adult HPA axis and affective behaviors are altered by perinatal exposure to a low dose of bisphenol A, Brain Res, 1571, 12, 10.1016/j.brainres.2014.05.010
Cao, 2013, Prenatal bisphenol A exposure alters sex-specific estrogen receptor expression in the neonatal rat hypothalamus and amygdala, Toxicol Sci, 133, 157, 10.1093/toxsci/kft035
Rebuli, 2014, Investigation of the effects of subchronic low dose oral exposure to bisphenol A (BPA) and ethinyl estradiol (EE) on estrogen receptor expression in the juvenile and adult female rat hypothalamus, Toxicol Sci, 140, 190, 10.1093/toxsci/kfu074
Kundakovic, 2013, Sex-specific epigenetic disruption and behavioral changes following low-dose in utero bisphenol A exposure, Proc Natl Acad Sci USA, 110, 9956, 10.1073/pnas.1214056110
Naulé, 2014, Neuroendocrine and behavioral effects of maternal exposure to oral bisphenol A in female mice, J Endocrinol, 220, 375, 10.1530/JOE-13-0607
Ramos, 2003, Bisphenol A induces both transient and permanent histofunctional alterations of the hypothalamic-pituitary-gonadal axis in prenatally exposed male rats, Endocrinology, 144, 3206, 10.1210/en.2002-0198
Mahoney, 2010, Developmental programming: impact of fetal exposure to endocrine-disrupting chemicals on gonadotropin-releasing hormone and estrogen receptor mRNA in sheep hypothalamus, Toxicol Appl Pharmacol, 247, 98, 10.1016/j.taap.2010.05.017
Panagiotidou, 2014, Perinatal exposure to low-dose bisphenol A affects the neuroendocrine stress response in rats, J Endocrinol, 220, 207, 10.1530/JOE-13-0416
Poimenova, 2010, Corticosterone-regulated actions in the rat brain are affected by perinatal exposure to low dose of bisphenol A, Neuroscience, 167, 741, 10.1016/j.neuroscience.2010.02.051
Faass, 2013, Developmental effects of perinatal exposure to PBDE and PCB on gene expression in sexually dimorphic rat brain regions and female sexual behavior, Gen Comp Endocrinol, 188, 232, 10.1016/j.ygcen.2013.04.008
Compagnone, 1995, Expression of the steroidogenic enzyme P450scc in the central and peripheral nervous systems during rodent embryogenesis, Endocrinology, 136, 2689, 10.1210/endo.136.6.7750493
Furukawa, 1998, Steroidogenic acute regulatory protein (StAR) transcripts constitutively expressed in the adult rat central nervous system: colocalization of StAR, cytochrome P-450SCC (CYP XIA1), and 3β-hydroxysteroid dehydrogenase in the rat brain, J Neurochem, 71, 2231, 10.1046/j.1471-4159.1998.71062231.x
Strömstedt, 1995, Messenger RNAs encoding steroidogenic enzymes are expressed in rodent brain, Brain Res Mol Brain Res, 34, 75, 10.1016/0169-328X(95)00140-N
Zwain, 1999, Dehydroepiandrosterone: biosynthesis and metabolism in the brain, Endocrinology, 140, 880, 10.1210/endo.140.2.6528
Do Rego, 2009, Neurosteroid biosynthesis: enzymatic pathways and neuroendocrine regulation by neurotransmitters and neuropeptides, Front Neuroendocrinol, 30, 259, 10.1016/j.yfrne.2009.05.006
Balthazart, 2011, Sex differences in brain aromatase activity: genomic and non-genomic controls, Front Endocrinol (Lausanne), 2, 34, 10.3389/fendo.2011.00034
McCarthy, 2012, Aromatase and sexual differentiation of the rodent brain; the old, the new, and the unexpected, Brain Aromatase, Estrogens and Behavior, 315, 10.1093/acprof:oso/9780199841196.003.0016
Wu, 2009, Estrogen masculinizes neural pathways and sex-specific behaviors, Cell, 139, 61, 10.1016/j.cell.2009.07.036
Celotti, 1997, Steroid metabolism in the mammalian brain: 5α-reduction and aromatization, Brain Res Bull, 44, 365, 10.1016/S0361-9230(97)00216-5
Hammer, 2004, Transcriptional regulation of P450scc gene expression in the embryonic rodent nervous system, Endocrinology, 145, 901, 10.1210/en.2003-0125
Taves, 2011, Extra-adrenal glucocorticoids and mineralocorticoids: evidence for local synthesis, regulation, and function, Am J Physiol Endocrinol Metab, 301, E11, 10.1152/ajpendo.00100.2011
Colciago, 2006, Prenatal Aroclor 1254 exposure and brain sexual differentiation: effect on the expression of testosterone metabolizing enzymes and androgen receptors in the hypothalamus of male and female rats, Reprod Toxicol, 22, 738, 10.1016/j.reprotox.2006.07.002
Desaulniers, 2005, Comparisons of brain, uterus, and liver mRNA expression for cytochrome p450s, DNA methyltransferase-1, and catechol-O-methyltransferase in prepubertal female Sprague-Dawley rats exposed to a mixture of aryl hydrocarbon receptor agonists, Toxicol Sci, 86, 175, 10.1093/toxsci/kfi178
Auger, 2011, Epigenetic turn ons and turn offs: chromatin reorganization and brain differentiation, Endocrinology, 152, 349, 10.1210/en.2010-0793
Kurian, 2010, Sex differences in epigenetic regulation of the estrogen receptor-α promoter within the developing preoptic area, Endocrinology, 151, 2297, 10.1210/en.2009-0649
Nugent, 2011, Hormonally mediated epigenetic changes to steroid receptors in the developing brain: implications for sexual differentiation, Horm Behav, 59, 338, 10.1016/j.yhbeh.2010.08.009
Gagnidze, 2013, Hormone-dependent chromatin modifications related to sexually differentiated behaviors, Multiple Origins of Sex Differences in Brain: Neuroendocrine Functions and their Pathologies, 1, 10.1007/978-3-642-33721-5_1
Tsai, 2009, Sex differences in histone modifications in the neonatal mouse brain, Epigenetics, 4, 47, 10.4161/epi.4.1.7288
Murray, 2009, Epigenetic control of sexual differentiation of the bed nucleus of the stria terminalis, Endocrinology, 150, 4241, 10.1210/en.2009-0458
Murray, 2011, Effects of neonatal treatment with valproic acid on vasopressin immunoreactivity and olfactory behaviour in mice, J Neuroendocrinol, 23, 906, 10.1111/j.1365-2826.2011.02196.x
Matsuda, 2011, Histone deacetylation during brain development is essential for permanent masculinization of sexual behavior, Endocrinology, 152, 2760, 10.1210/en.2011-0193
Yaoi, 2008, Genome-wide analysis of epigenomic alterations in fetal mouse forebrain after exposure to low doses of bisphenol A, Biochem Biophys Res Commun, 376, 563, 10.1016/j.bbrc.2008.09.028
Wolstenholme, 2012, Gestational exposure to bisphenol A produces transgenerational changes in behaviors and gene expression, Endocrinology, 153, 3828, 10.1210/en.2012-1195
Zhou, 2013, Persistent overexpression of DNA methyltransferase 1 attenuating GABAergic inhibition in basolateral amygdala accounts for anxiety in rat offspring exposed perinatally to low-dose bisphenol A, J Psychiatr Res, 47, 1535, 10.1016/j.jpsychires.2013.05.013
Walker, 2014, Dynamic postnatal developmental and sex-specific neuroendocrine effects of prenatal polychlorinated biphenyls in rats, Mol Endocrinol, 28, 99, 10.1210/me.2013-1270
Yeo, 2013, Bisphenol A delays the perinatal chloride shift in cortical neurons by epigenetic effects on the Kcc2 promoter, Proc Natl Acad Sci USA, 110, 4315, 10.1073/pnas.1300959110
Guida, 2014, Histone deacetylase 4 promotes ubiquitin-dependent proteasomal degradation of Sp3 in SH-SY5Y cells treated with di(2-ethylhexyl)phthalate (DEHP), determining neuronal death, Toxicol Appl Pharmacol, 280, 190, 10.1016/j.taap.2014.07.014
Lindén, 2010, Dioxins, the aryl hydrocarbon receptor and the central regulation of energy balance, Front Neuroendocrinol, 31, 452, 10.1016/j.yfrne.2010.07.002
Kirilov, 2013, Dependence of fertility on kisspeptin-Gpr54 signaling at the GnRH neuron, Nat Commun, 4, 2492, 10.1038/ncomms3492
Seminara, 2003, The GPR54 gene as a regulator of puberty, N Engl J Med, 349, 1614, 10.1056/NEJMoa035322
de Roux, 2003, Hypogonadotropic hypogonadism due to loss of function of the KiSS1-derived peptide receptor GPR54, Proc Natl Acad Sci USA, 100, 10972, 10.1073/pnas.1834399100
Castellano, 2010, Kisspeptins: bridging energy homeostasis and reproduction, Brain Res, 1364, 129, 10.1016/j.brainres.2010.08.057
Bai, 2011, Increase of anteroventral periventricular kisspeptin neurons and generation of E2-induced LH-surge system in male rats exposed perinatally to environmental dose of bisphenol-A, Endocrinology, 152, 1562, 10.1210/en.2010-1042
Patisaul, 2009, Impact of neonatal exposure to the ERα agonist CD-1, bisphenol-A or phytoestrogens on hypothalamic kisspeptin fiber density in male and female rats, Neurotoxicology, 30, 350, 10.1016/j.neuro.2009.02.010
Losa-Ward, 2012, Disrupted organization of RFamide pathways in the hypothalamus is associated with advanced puberty in female rats neonatally exposed to bisphenol A, Biol Reprod, 87, 28, 10.1095/biolreprod.112.100826
Abi Salloum, 2013, Developmental programming: impact of prenatal exposure to bisphenol-A and methoxychlor on steroid feedbacks in sheep, Toxicol Appl Pharmacol, 268, 300, 10.1016/j.taap.2013.02.011
Dickerson, 2011, Prenatal PCBs disrupt early neuroendocrine development of the rat hypothalamus, Toxicol Appl Pharmacol, 252, 36, 10.1016/j.taap.2011.01.012
Bellingham, 2009, Exposure to a complex cocktail of environmental endocrine-disrupting compounds disturbs the kisspeptin/GPR54 system in ovine hypothalamus and pituitary gland, Environ Health Perspect, 117, 1556, 10.1289/ehp.0900699
Harvey, 2007, Adrenal toxicology: a strategy for assessment of functional toxicity to the adrenal cortex and steroidogenesis, J Appl Toxicol, 27, 103, 10.1002/jat.1221
Hotchkiss, 2012, Atrazine does not induce pica behavior at doses that increase hypothalamic-pituitary-adrenal axis activation and cause conditioned taste avoidance, Neurotoxicol Teratol, 34, 295, 10.1016/j.ntt.2012.03.001
Riffle, 2014, Novel molecular events associated with altered steroidogenesis induced by exposure to atrazine in the intact and castrate male rat, Reprod Toxicol, 47, 59, 10.1016/j.reprotox.2014.05.008
Jašarevic, 2011, Disruption of adult expression of sexually selected traits by developmental exposure to bisphenol A, Proc Natl Acad Sci USA, 108, 11715, 10.1073/pnas.1107958108
Meserve, 1992, Influence of maternal ingestion of Aroclor 1254 (PCB) or FireMaster BP-6 (PBB) on unstimulated and stimulated corticosterone levels in young rats, Bull Environ Contam Toxicol, 48, 715, 10.1007/BF00195992
Reilly, 2015, The effects of prenatal PCBs on adult social behavior in rats, Horm Behav, 73, 47, 10.1016/j.yhbeh.2015.06.002
Zimmer, 2009, Altered stress-induced cortisol levels in goats exposed to polychlorinated biphenyls (PCB 126 and PCB 153) during fetal and postnatal development, J Toxicol Environ Health A, 72, 164, 10.1080/15287390802539004
Cameron, 2008, Epigenetic programming of phenotypic variations in reproductive strategies in the rat through maternal care, J Neuroendocrinol, 20, 795, 10.1111/j.1365-2826.2008.01725.x
Donaldson, 2008, Oxytocin, vasopressin, and the neurogenetics of sociality, Science, 322, 900, 10.1126/science.1158668
Wolstenholme, 2011, Gestational exposure to low dose bisphenol A alters social behavior in juvenile mice, PLoS One, 6, e25448, 10.1371/journal.pone.0025448
Sullivan, 2014, A novel model for neuroendocrine toxicology: neurobehavioral effects of BPA exposure in a prosocial species, the prairie vole (Microtus ochrogaster), Endocrinology, 155, 3867, 10.1210/en.2014-1379
Patisaul, 2012, Anxiogenic effects of developmental bisphenol A exposure are associated with gene expression changes in the juvenile rat amygdala and mitigated by soy, PLoS One, 7, e43890, 10.1371/journal.pone.0043890
Kodavanti, 2010, Neuroendocrine actions of organohalogens: thyroid hormones, arginine vasopressin, and neuroplasticity, Front Neuroendocrinol, 31, 479, 10.1016/j.yfrne.2010.06.005
Kreiss, 1985, Studies on populations exposed to polychlorinated biphenyls, Environ Health Perspect, 60, 193, 10.1289/ehp.8560193
Kreiss, 1981, Association of blood pressure and polychlorinated biphenyl levels, JAMA, 245, 2505, 10.1001/jama.1981.03310490023017
Shah, 2011, Altered cardiovascular reactivity and osmoregulation during hyperosmotic stress in adult rats developmentally exposed to polybrominated diphenyl ethers (PBDEs), Toxicol Appl Pharmacol, 256, 103, 10.1016/j.taap.2011.07.014
Engell, 2006, Perinatal exposure to endocrine disrupting compounds alters behavior and brain in the female pine vole, Neurotoxicol Teratol, 28, 103, 10.1016/j.ntt.2005.10.002
Tait, 2009, Long-term effects on hypothalamic neuropeptides after developmental exposure to chlorpyrifos in mice, Environ Health Perspect, 117, 112, 10.1289/ehp.11696
Venerosi, 2012, Sex dimorphic behaviors as markers of neuroendocrine disruption by environmental chemicals: the case of chlorpyrifos, Neurotoxicology, 33, 1420, 10.1016/j.neuro.2012.08.009
Landrigan, 2010, What causes autism? Exploring the environmental contribution, Curr Opin Pediatr, 22, 219, 10.1097/MOP.0b013e328336eb9a
Landrigan, 2012, A research strategy to discover the environmental causes of autism and neurodevelopmental disabilities, Environ Health Perspect, 120, a258, 10.1289/ehp.1104285
Jacobson, 1996, Intellectual impairment in children exposed to polychlorinated biphenyls in utero, N Engl J Med, 335, 783, 10.1056/NEJM199609123351104
Patandin, 1999, Effects of environmental exposure to polychlorinated biphenyls and dioxins on cognitive abilities in Dutch children at 42 months of age, J Pediatr, 134, 33, 10.1016/S0022-3476(99)70369-0
Darvill, 2000, Prenatal exposure to PCBs and infant performance on the Fagan test of infant intelligence, Neurotoxicology, 21, 1029
Koopman-Esseboom, 1996, Effects of polychlorinated biphenyl/dioxin exposure and feeding type on infants' mental and psychomotor development, Pediatrics, 97, 700, 10.1542/peds.97.5.700
Bellanger, 2015, Neurobehavioral deficits, diseases, and associated costs of exposure to endocrine-disrupting chemicals in the European Union, J Clin Endocrinol Metab, 100, 1256, 10.1210/jc.2014-4323
Harley, 2013, Prenatal and early childhood bisphenol A concentrations and behavior in school-aged children, Environ Res, 126, 43, 10.1016/j.envres.2013.06.004
Braun, 2011, Impact of early-life bisphenol A exposure on behavior and executive function in children, Pediatrics, 128, 873, 10.1542/peds.2011-1335
Kim, 2011, Prenatal exposure to phthalates and infant development at 6 months: prospective Mothers and Children's Environmental Health (MOCEH) study, Environ Health Perspect, 119, 1495, 10.1289/ehp.1003178
Swan, 2010, Prenatal phthalate exposure and reduced masculine play in boys, Int J Androl, 33, 259, 10.1111/j.1365-2605.2009.01019.x
Engel, 2010, Prenatal phthalate exposure is associated with childhood behavior and executive functioning, Environ Health Perspect, 118, 565, 10.1289/ehp.0901470
Chen, 2014, Prenatal polybrominated diphenyl ether exposures and neurodevelopment in U.S. children through 5 years of age: the HOME study, Environ Health Perspect, 122, 856, 10.1289/ehp.1307562
Fitzgerald, 2012, Polybrominated diphenyl ethers (PBDEs), polychlorinated biphenyls (PCBs) and neuropsychological status among older adults in New York, Neurotoxicology, 33, 8, 10.1016/j.neuro.2011.10.011
Boucher, 2012, Response inhibition and error monitoring during a visual go/no-go task in Inuit children exposed to lead, polychlorinated biphenyls, and methylmercury, Environ Health Perspect, 120, 608, 10.1289/ehp.1103828
Rauh, 2011, Seven-year neurodevelopmental scores and prenatal exposure to chlorpyrifos, a common agricultural pesticide, Environ Health Perspect, 119, 1196, 10.1289/ehp.1003160
Bouchard, 2011, Prenatal exposure to organophosphate pesticides and IQ in 7-year-old children, Environ Health Perspect, 119, 1189, 10.1289/ehp.1003185
Engel, 2011, Prenatal exposure to organophosphates, paraoxonase 1, and cognitive development in childhood, Environ Health Perspect, 119, 1182, 10.1289/ehp.1003183
Shelton, 2014, Neurodevelopmental disorders and prenatal residential proximity to agricultural pesticides: the CHARGE Study, Environ Health Perspect, 122, 1103, 10.1289/ehp.1307044
Braun, 2014, Gestational exposure to endocrine-disrupting chemicals and reciprocal social, repetitive, and stereotypic behaviors in 4- and 5-year-old children: the HOME Study, Environ Health Perspect, 122, 513, 10.1289/ehp.1307261
Jones, 2011, Pre- and postnatal bisphenol A treatment results in persistent deficits in the sexual behavior of male rats, but not female rats, in adulthood, Horm Behav, 59, 246, 10.1016/j.yhbeh.2010.12.006
Ferguson, 2014, Developmental treatment with ethinyl estradiol, but not bisphenol A, causes alterations in sexually dimorphic behaviors in male and female Sprague Dawley rats, Toxicol Sci, 140, 374, 10.1093/toxsci/kfu077
Monje, 2009, Neonatal exposure to bisphenol A alters estrogen-dependent mechanisms governing sexual behavior in the adult female rat, Reprod Toxicol, 28, 435, 10.1016/j.reprotox.2009.06.012
Decatanzaro, 2013, Perturbation of male sexual behavior in mice (Mus musculus) within a discrete range of perinatal bisphenol-A doses in the context of a high- or low-phytoestrogen diet, Food Chem Toxicol, 55, 164, 10.1016/j.fct.2012.12.046
Steinberg, 2007, The effects of prenatal PCBs on adult female paced mating reproductive behaviors in rats, Horm Behav, 51, 364, 10.1016/j.yhbeh.2006.12.004
Cummings, 2008, Exposure to PCB 77 affects partner preference but not sexual behavior in the female rat, Physiol Behav, 95, 471, 10.1016/j.physbeh.2008.07.016
Faass, 2009, Female sexual behavior, estrous cycle and gene expression in sexually dimorphic brain regions after pre- and postnatal exposure to endocrine active UV filters, Neurotoxicology, 30, 249, 10.1016/j.neuro.2008.12.008
Tian, 2010, Prenatal and postnatal exposure to bisphenol A induces anxiolytic behaviors and cognitive deficits in mice, Synapse, 64, 432, 10.1002/syn.20746
Wang, 2014, Changes in memory and synaptic plasticity induced in male rats after maternal exposure to bisphenol A, Toxicology, 322, 51, 10.1016/j.tox.2014.05.001
Kuwahara, 2013, Perinatal exposure to low-dose bisphenol A impairs spatial learning and memory in male rats, J Pharmacol Sci, 123, 132, 10.1254/jphs.13093FP
Fan, 2013, Does preconception paternal exposure to a physiologically relevant level of bisphenol A alter spatial memory in an adult rat?, Horm Behav, 64, 598, 10.1016/j.yhbeh.2013.08.014
Stump, 2010, Developmental neurotoxicity study of dietary bisphenol A in Sprague-Dawley rats, Toxicol Sci, 115, 167, 10.1093/toxsci/kfq025
Sadowski, 2014, Effects of perinatal bisphenol A exposure during early development on radial arm maze behavior in adult male and female rats, Neurotoxicol Teratol, 42, 17, 10.1016/j.ntt.2014.01.002
Williams, 2013, Effects of developmental bisphenol A exposure on reproductive-related behaviors in California mice (Peromyscus californicus): a monogamous animal model, PLoS One, 8, e55698, 10.1371/journal.pone.0055698
Xu, 2015, Sex-specific effects of long-term exposure to bisphenol-A on anxiety- and depression-like behaviors in adult mice, Chemosphere, 120, 258, 10.1016/j.chemosphere.2014.07.021
Matsuda, 2012, Effects of perinatal exposure to low dose of bisphenol A on anxiety like behavior and dopamine metabolites in brain, Prog Neuropsychopharmacol Biol Psychiatry, 39, 273, 10.1016/j.pnpbp.2012.06.016
Xu, 2012, Gestational and lactational exposure to bisphenol-A affects anxiety- and depression-like behaviors in mice, Horm Behav, 62, 480, 10.1016/j.yhbeh.2012.08.005
Jašarevic, 2013, Sex and dose-dependent effects of developmental exposure to bisphenol A on anxiety and spatial learning in deer mice (Peromyscus maniculatus bairdii) offspring, Horm Behav, 63, 180, 10.1016/j.yhbeh.2012.09.009
Jones, 2012, Perinatal BPA exposure demasculinizes males in measures of affect but has no effect on water maze learning in adulthood, Horm Behav, 61, 605, 10.1016/j.yhbeh.2012.02.011
Negishi, 2014, Altered social interactions in male juvenile cynomolgus monkeys prenatally exposed to bisphenol A, Neurotoxicol Teratol, 44, 46, 10.1016/j.ntt.2014.05.004
Belloni, 2011, Early exposure to low doses of atrazine affects behavior in juvenile and adult CD1 mice, Toxicology, 279, 19, 10.1016/j.tox.2010.07.002
Piedrafita, 2008, Developmental exposure to polychlorinated biphenyls or methylmercury, but not to its combination, impairs the glutamate-nitric oxide-cyclic GMP pathway and learning in 3-month-old rats, Neuroscience, 154, 1408, 10.1016/j.neuroscience.2008.05.013
Elnar, 2012, Neurodevelopmental and behavioral toxicity via lactational exposure to the sum of six indicator non-dioxin-like-polychlorinated biphenyls (∑6NDL-PCBs) in mice, Toxicology, 299, 44, 10.1016/j.tox.2012.05.004
Jolous-Jamshidi, 2010, Perinatal exposure to polychlorinated biphenyls alters social behaviors in rats, Toxicol Lett, 199, 136, 10.1016/j.toxlet.2010.08.015
Carbone, 2013, Antiandrogenic effect of perinatal exposure to the endocrine disruptor di-(2-ethylhexyl) phthalate increases anxiety-like behavior in male rats during sexual maturation, Horm Behav, 63, 692, 10.1016/j.yhbeh.2013.01.006
Lin, 2014, Gestational and lactational exposure to atrazine via the drinking water causes specific behavioral deficits and selectively alters monoaminergic systems in C57BL/6 mouse dams, juvenile and adult offspring, Toxicol Sci, 141, 90, 10.1093/toxsci/kfu107
Anway, 2008, Comparative anti-androgenic actions of vinclozolin and flutamide on transgenerational adult onset disease and spermatogenesis, Reprod Toxicol, 26, 100, 10.1016/j.reprotox.2008.07.008
Nilsson, 2008, Transgenerational epigenetic effects of the endocrine disruptor vinclozolin on pregnancies and female adult onset disease, Reproduction, 135, 713, 10.1530/REP-07-0542
Guerrero-Bosagna, 2013, Environmentally induced epigenetic transgenerational inheritance of altered Sertoli cell transcriptome and epigenome: molecular etiology of male infertility, PLoS One, 8, e59922, 10.1371/journal.pone.0059922
Tracey, 2013, Hydrocarbons (jet fuel JP-8) induce epigenetic transgenerational inheritance of obesity, reproductive disease and sperm epimutations, Reprod Toxicol, 36, 104, 10.1016/j.reprotox.2012.11.011
Schneider, 2008, Vinclozolin–the lack of a transgenerational effect after oral maternal exposure during organogenesis, Reprod Toxicol, 25, 352, 10.1016/j.reprotox.2008.04.001
Stouder, 2010, Transgenerational effects of the endocrine disruptor vinclozolin on the methylation pattern of imprinted genes in the mouse sperm, Reproduction, 139, 373, 10.1530/REP-09-0340
Skinner, 2008, Transgenerational epigenetic programming of the brain transcriptome and anxiety behavior, PLoS One, 3, e3745, 10.1371/journal.pone.0003745
Dietert, 2015, The sum of our parts, The Scientist, 29, 45
vom Saal, 1981, Variation in phenotype due to random intrauterine positioning of male and female fetuses in rodents, J Reprod Fertil, 62, 633, 10.1530/jrf.0.0620633
de Medeiros, 2010, Deconstructing early life experiences: distinguishing the contributions of prenatal and postnatal factors to adult male sexual behavior in the rat, Psychol Sci, 21, 1494, 10.1177/0956797610382122
Vom Saal, 2014, Bisphenol A (BPA) pharmacokinetics with daily oral bolus or continuous exposure via silastic capsules in pregnant rhesus monkeys: relevance for human exposures, Reprod Toxicol, 45, 105, 10.1016/j.reprotox.2014.01.007
Saffarini, 2015, Developmental exposure to estrogen alters differentiation and epigenetic programming in a human fetal prostate xenograft model, PLoS One, 10, e0122290, 10.1371/journal.pone.0122290
Kalfa, 2011, Prevalence of hypospadias in grandsons of women exposed to diethylstilbestrol during pregnancy: a multigenerational national cohort study, Fertil Steril, 95, 2574, 10.1016/j.fertnstert.2011.02.047
Eladak, 2015, A new chapter in the bisphenol A story: bisphenol S and bisphenol F are not safe alternatives to this compound, Fertil Steril, 103, 11, 10.1016/j.fertnstert.2014.11.005
Viñas, 2013, Bisphenol S disrupts estradiol-induced nongenomic signaling in a rat pituitary cell line: effects on cell functions, Environ Health Perspect, 121, 352, 10.1289/ehp.1205826
Crews, 2014, Transgenerational epigenetics: current controversies and debates, Transgenerational Epigenetics, 371, 10.1016/B978-0-12-405944-3.00026-X
Stotland, 2014, Counseling patients on preventing prenatal environmental exposures–a mixed-methods study of obstetricians, PLoS One, 9, e98771, 10.1371/journal.pone.0098771
Schug, 2013, Designing endocrine disruption out of the next generation of chemicals, Green Chem, 15, 181, 10.1039/C2GC35055F
Lamb, 2014, Critical comments on the WHO-UNEP State of the Science of Endocrine Disrupting Chemicals - 2012, Regul Toxicol Pharmacol, 69, 22, 10.1016/j.yrtph.2014.02.002
Bergman, Manufacturing doubt about endocrine disrupter science - A rebuttal of industry-sponsored critical comments on the UNEP/WHO report “State of the Science of Endocrine Disrupting Chemicals 2012” [published online July 31, 2015], Regul Toxicol Pharmacol
Fredslund, 2012, Breast cancer in the Arctic–changes over the past decades, Int J Circumpolar Health, 71, 19155, 10.3402/ijch.v71i0.19155
Kuiper, 1998, Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor β, Endocrinology, 139, 4252, 10.1210/endo.139.10.6216
Matsushima, 2007, Structural evidence for endocrine disruptor bisphenol A binding to human nuclear receptor ERR γ, J Biochem, 142, 517, 10.1093/jb/mvm158
Lee, 2003, Antiandrogenic effects of bisphenol A and nonylphenol on the function of androgen receptor, Toxicol Sci, 75, 40, 10.1093/toxsci/kfg150
Ashby, 2004, Gene expression changes in the immature rat uterus: effects of uterotrophic and sub-uterotrophic doses of bisphenol A, Toxicol Sci, 82, 458, 10.1093/toxsci/kfh283
Sui, 2012, Bisphenol A and its analogues activate human pregnane X receptor, Environ Health Perspect, 120, 399, 10.1289/ehp.1104426
Viñas, 2013, Mixtures of xenoestrogens disrupt estradiol-induced non-genomic signaling and downstream functions in pituitary cells, Environ Health, 12, 26, 10.1186/1476-069X-12-26
Greathouse, 2012, Environmental estrogens differentially engage the histone methyltransferase EZH2 to increase risk of uterine tumorigenesis, Mol Cancer Res, 10, 546, 10.1158/1541-7786.MCR-11-0605
Ge, 2014, Involvement of activating ERK1/2 through G protein coupled receptor 30 and estrogen receptor α/β in low doses of bisphenol A promoting growth of Sertoli TM4 cells, Toxicol Lett, 226, 81, 10.1016/j.toxlet.2014.01.035
Sheng, 2011, Low concentrations of bisphenol A induce mouse spermatogonial cell proliferation by G protein-coupled receptor 30 and estrogen receptor-α, Environ Health Perspect, 119, 1775, 10.1289/ehp.1103781
Dong, 2011, Bisphenol A induces a rapid activation of Erk1/2 through GPR30 in human breast cancer cells, Environ Pollut, 159, 212, 10.1016/j.envpol.2010.09.004
Liang, 2014, Cellular mechanism of the nonmonotonic dose response of bisphenol A in rat cardiac myocytes, Environ Health Perspect, 122, 601, 10.1289/ehp.1307491
Tanabe, 2006, Rapid Ca(2+) signaling induced by bisphenol A in cultured rat hippocampal neurons, Neuro Endocrinol Lett, 27, 97
Vandenberg, 2008, Perinatal exposure to the xenoestrogen bisphenol-A induces mammary intraductal hyperplasias in adult CD-1 mice, Reprod Toxicol, 26, 210, 10.1016/j.reprotox.2008.09.015
Wadia, 2013, Low-dose BPA exposure alters the mesenchymal and epithelial transcriptomes of the mouse fetal mammary gland, PLoS One, 8, e63902, 10.1371/journal.pone.0063902
Moral, 2008, Effect of prenatal exposure to the endocrine disruptor bisphenol A on mammary gland morphology and gene expression signature, J Endocrinol, 196, 101, 10.1677/JOE-07-0056
Klotz, 1996, Identification of environmental chemicals with estrogenic activity using a combination of in vitro assays, Environ Health Perspect, 104, 1084, 10.1289/ehp.961041084
Kelce, 1995, Persistent DDT metabolite p,p'-DDE is a potent androgen receptor antagonist, Nature, 375, 581, 10.1038/375581a0
Korach, 1978, Estrogenic activity in vivo and in vitro of some diethylstilbestrol metabolites and analogs, Proc Natl Acad Sci USA, 75, 468, 10.1073/pnas.75.1.468
Fang, 2003, Study of 202 natural, synthetic, and environmental chemicals for binding to the androgen receptor, Chem Res Toxicol, 16, 1338, 10.1021/tx030011g
Tremblay, 2001, Diethylstilbestrol regulates trophoblast stem cell differentiation as a ligand of orphan nuclear receptor ERR β, Genes Dev, 15, 833, 10.1101/gad.873401
Li, 2006, Activation of kinase pathways in MCF-7 cells by 17β-estradiol and structurally diverse estrogenic compounds, J Steroid Biochem Mol Biol, 98, 122, 10.1016/j.jsbmb.2005.08.018
Bulayeva, 2004, Xenoestrogen-induced ERK-1 and ERK-2 activation via multiple membrane-initiated signaling pathways, Environ Health Perspect, 112, 1481, 10.1289/ehp.7175
Bromer, 2009, Hypermethylation of homeobox A10 by in utero diethylstilbestrol exposure: an epigenetic mechanism for altered developmental programming, Endocrinology, 150, 3376, 10.1210/en.2009-0071
Li, 1997, Developmental exposure to diethylstilbestrol elicits demethylation of estrogen-responsive lactoferrin gene in mouse uterus, Cancer Res, 57, 4356
Ohtake, 2003, Modulation of oestrogen receptor signalling by association with the activated dioxin receptor, Nature, 423, 545, 10.1038/nature01606
Kester, 2000, Potent inhibition of estrogen sulfotransferase by hydroxylated PCB metabolites: a novel pathway explaining the estrogenic activity of PCBs, Endocrinology, 141, 1897, 10.1210/endo.141.5.7530
Heneweer, 2005, Inhibition of aromatase activity by methyl sulfonyl PCB metabolites in primary culture of human mammary fibroblasts, Toxicol Appl Pharmacol, 202, 50, 10.1016/j.taap.2004.06.006
You, 2006, 4-Hydroxy-PCB106 acts as a direct thyroid hormone receptor agonist in rat GH3 cells, Mol Cell Endocrinol, 257–258, 26, 10.1016/j.mce.2006.06.009
Maloney, 1999, Trans-activation of PPARα and PPARγ by structurally diverse environmental chemicals, Toxicol Appl Pharmacol, 161, 209, 10.1006/taap.1999.8809
Pylkkänen, 1993, Prostatic dysplasia associated with increased expression of C-MYC in neonatally estrogenized mice, J Urol, 149, 1593, 10.1016/S0022-5347(17)36458-3
Fenton, 2012, Perinatal environmental exposures affect mammary development, function, and cancer risk in adulthood, Annu Rev Pharmacol Toxicol, 52, 455, 10.1146/annurev-pharmtox-010611-134659
Newbold, 2009, Environmental estrogens and obesity, Mol Cell Endocrinol, 304, 84, 10.1016/j.mce.2009.02.024
Hao, 2012, The endocrine disruptor diethylstilbestrol induces adipocyte differentiation and promotes obesity in mice, Toxicol Appl Pharmacol, 263, 102, 10.1016/j.taap.2012.06.003
Wahlang, 2013, Polychlorinated biphenyl 153 is a diet-dependent obesogen that worsens nonalcoholic fatty liver disease in male C57BL6/J mice, J Nutr Biochem, 24, 1587, 10.1016/j.jnutbio.2013.01.009
Zuo, 2011, Tributyltin causes obesity and hepatic steatosis in male mice, Environ Toxicol, 26, 79, 10.1002/tox.20531
Ishida, 2005, 2,3,7,8-Tetrachlorodibenzo-p-dioxin-induced change in intestinal function and pathology: evidence for the involvement of arylhydrocarbon receptor-mediated alteration of glucose transportation, Toxicol Appl Pharmacol, 205, 89, 10.1016/j.taap.2004.09.014
Weber, 1991, Reduced activities of key enzymes of gluconeogenesis as possible cause of acute toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in rats, Toxicology, 66, 133, 10.1016/0300-483X(91)90214-L
Pocock, 2002, Effects of perinatal octylphenol on ultrasound vocalization, behavior and reproductive physiology in rats, Physiol Behav, 76, 645, 10.1016/S0031-9384(02)00788-6
Fisher, 1999, Effect of neonatal exposure to estrogenic compounds on development of the excurrent ducts of the rat testis through puberty to adulthood, Environ Health Perspect, 107, 397, 10.1289/ehp.99107397
de Jager, 1999, The effect of p-nonylphenol on the fertility potential of male rats after gestational, lactational and direct exposure, Andrologia, 31, 107
Wolf, 1999, Administration of potentially antiandrogenic pesticides (procymidone, linuron, iprodione, chlozolinate, p,p'-DDE, and ketoconazole) and toxic substances (dibutyl- and diethylhexyl phthalate, PCB 169, and ethane dimethane sulphonate) during sexual differentiation produces diverse profiles of reproductive malformations in the male rat, Toxicol Ind Health, 15, 94, 10.1177/074823379901500109
You, 1998, Impaired male sexual development in perinatal Sprague-Dawley and Long-Evans hooded rats exposed in utero and lactationally to p,p'-DDE, Toxicol Sci, 45, 162
Gray, 1999, The estrogenic and antiandrogenic pesticide methoxychlor alters the reproductive tract and behavior without affecting pituitary size or LH and prolactin secretion in male rats, Toxicol Ind Health, 15, 37, 10.1191/074823399678846655
Guillette, 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, 10.1289/ehp.94102680
McLachlan, 1977, Toxicologic comparisons of experimental and clinical exposure to diethylstilbestrol during gestation, Adv Sex Horm Res, 3, 309
Lewis, 2003, The effects of the phytoestrogen genistein on the postnatal development of the rat, Toxicol Sci, 71, 74, 10.1093/toxsci/71.1.74
McKinnell, 2001, Suppression of androgen action and the induction of gross abnormalities of the reproductive tract in male rats treated neonatally with diethylstilbestrol, J Androl, 22, 323, 10.1002/j.1939-4640.2001.tb02186.x
Rivas, 2002, Induction of reproductive tract developmental abnormalities in the male rat by lowering androgen production or action in combination with a low dose of diethylstilbestrol: evidence for importance of the androgen-estrogen balance, Endocrinology, 143, 4797, 10.1210/en.2002-220531
Yamamoto, 2003, Effects of maternal exposure to diethylstilbestrol on the development of the reproductive system and thyroid function in male and female rat offspring, J Toxicol Sci, 28, 385, 10.2131/jts.28.385
Faber, 1991, The effect of neonatal exposure to DES and o,p'-DDT on pituitary responsiveness to GnRH in adult castrated rats, Reprod Toxicol, 5, 363, 10.1016/0890-6238(91)90095-W
Gray, 1994, Developmental effects of an environmental antiandrogen: the fungicide vinclozolin alters sex differentiation of the male rat, Toxicol Appl Pharmacol, 129, 46, 10.1006/taap.1994.1227
Hellwig, 2000, Pre- and postnatal oral toxicity of vinclozolin in Wistar and Long-Evans rats, Regul Toxicol Pharmacol, 32, 42, 10.1006/rtph.2000.1400
Elzeinova, 2008, Effect of low dose of vinclozolin on reproductive tract development and sperm parameters in CD1 outbred mice, Reprod Toxicol, 26, 231, 10.1016/j.reprotox.2008.09.007
Vinggaard, 1999, Rapid and sensitive reporter gene assays for detection of antiandrogenic and estrogenic effects of environmental chemicals, Toxicol Appl Pharmacol, 155, 150, 10.1006/taap.1998.8598
Ostby, 1999, The fungicide procymidone alters sexual differentiation in the male rat by acting as an androgen-receptor antagonist in vivo and in vitro, Toxicol Ind Health, 15, 80, 10.1177/074823379901500108
Colbert, 2005, Perinatal exposure to low levels of the environmental antiandrogen vinclozolin alters sex-differentiated social play and sexual behaviors in the rat, Environ Health Perspect, 113, 700, 10.1289/ehp.7509
Mably, 1992, In utero and lactational exposure of male rats to 2,3,7,8-tetrachlorodibenzo-p-dioxin. 3. Effects on spermatogenesis and reproductive capability, Toxicol Appl Pharmacol, 114, 118, 10.1016/0041-008X(92)90103-Y
Ohsako, 2001, Maternal exposure to a low dose of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) suppressed the development of reproductive organs of male rats: dose-dependent increase of mRNA levels of 5α-reductase type 2 in contrast to decrease of androgen receptor in the pubertal ventral prostate, Toxicol Sci, 60, 132, 10.1093/toxsci/60.1.132
Simanainen, 2004, Pattern of male reproductive system effects after in utero and lactational 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure in three differentially TCDD-sensitive rat lines, Toxicol Sci, 80, 101, 10.1093/toxsci/kfh142
McIntyre, 2001, Androgen-mediated development in male rat offspring exposed to flutamide in utero: permanence and correlation of early postnatal changes in anogenital distance and nipple retention with malformations in androgen-dependent tissues, Toxicol Sci, 62, 236, 10.1093/toxsci/62.2.236
Durrer, 2007, Estrogen sensitivity of target genes and expression of nuclear receptor co-regulators in rat prostate after pre- and postnatal exposure to the ultraviolet filter 4-methylbenzylidene camphor, Environ Health Perspect, 115, 42, 10.1289/ehp.9134
Faqi, 1998, Effects on developmental landmarks and reproductive capability of 3,3′,4,4′-tetrachlorobiphenyl and 3,3′,4,4′,5-pentachlorobiphenyl in offspring of rats exposed during pregnancy, Hum Exp Toxicol, 17, 365
Kuriyama, 2004, In utero exposure to low-dose 2,3′,4,4′,5-pentachlorobiphenyl (PCB 118) impairs male fertility and alters neurobehavior in rat offspring, Toxicology, 202, 185, 10.1016/j.tox.2004.05.006
Barlow, 2004, Male reproductive tract lesions at 6, 12, and 18 months of age following in utero exposure to di(n-butyl) phthalate, Toxicol Pathol, 32, 79, 10.1080/01926230490265894
Borch, 2004, Steroidogenesis in fetal male rats is reduced by DEHP and DINP, but endocrine effects of DEHP are not modulated by DEHA in fetal, prepubertal and adult male rats, Reprod Toxicol, 18, 53, 10.1016/j.reprotox.2003.10.011
Gray, 2000, Perinatal exposure to the phthalates DEHP, BBP, and DINP, but not DEP, DMP, or DOTP, alters sexual differentiation of the male rat, Toxicol Sci, 58, 350, 10.1093/toxsci/58.2.350
Mylchreest, 1999, Disruption of androgen-regulated male reproductive development by di(n-butyl) phthalate during late gestation in rats is different from flutamide, Toxicol Appl Pharmacol, 156, 81, 10.1006/taap.1999.8643
Mylchreest, 2000, Dose-dependent alterations in androgen-regulated male reproductive development in rats exposed to di(n-butyl) phthalate during late gestation, Toxicol Sci, 55, 143, 10.1093/toxsci/55.1.143
Parks, 2000, The plasticizer diethylhexyl phthalate induces malformations by decreasing fetal testosterone synthesis during sexual differentiation in the male rat, Toxicol Sci, 58, 339, 10.1093/toxsci/58.2.339
Andrade, 2006, A dose-response study following in utero and lactational exposure to di-(2-ethylhexyl)-phthalate (DEHP): non-monotonic dose-response and low dose effects on rat brain aromatase activity, Toxicology, 227, 185, 10.1016/j.tox.2006.07.022
Adeeko, 2003, Effects of in utero tributyltin chloride exposure in the rat on pregnancy outcome, Toxicol Sci, 74, 407, 10.1093/toxsci/kfg131
Kishta, 2007, In utero exposure to tributyltin chloride differentially alters male and female fetal gonad morphology and gene expression profiles in the Sprague-Dawley rat, Reprod Toxicol, 23, 1, 10.1016/j.reprotox.2006.08.014
Toppari, 2012, Endocrine Disrupters and Child Health. Possible Developmental Early Effects of Endocrine Disrupters on Child Health
Prins, 2010, Early-life estrogens and prostate cancer in an animal model, J Dev Orig Health Dis, 1, 365, 10.1017/S2040174410000577
Calderon-Gierszal, Directed differentiation of human embryonic stem cells (hESC) to prostate: novel models that verify bisphenol A effects on human prostate development
Arai, 1983, Long-term effects of perinatal exposure to sex steroids and diethylstilbestrol on the reproductive system of male mammals, Int Rev Cytol, 84, 235, 10.1016/S0074-7696(08)61019-0