1932

Abstract

While definitions vary, endocrine-disrupting chemicals (EDCs) have two fundamental features: their disruption of hormone function and their contribution to disease and disability. The unique vulnerability of children to low-level EDC exposures has eroded the notion that only the dose makes the thing a poison, requiring a paradigm shift in scientific and policy practice. In this review, we discuss the unique vulnerability of children as early as fetal life and provide an overview of epidemiological studies on programming effects of EDCs on neuronal, metabolic, and immune pathways as well as on endocrine, reproductive, and renal systems. Building on this accumulating evidence, we dispel and address existing myths about the health effects of EDCs with examples from child health research. Finally, we provide a list of effective actions to reduce exposure and subsequent harm that are applicable to individuals, communities, and policy-makers.

Keyword(s): childhoodDOHaDEDCprenatalprogramming
Loading

Article metrics loading...

/content/journals/10.1146/annurev-pharmtox-021921-093352
2022-01-06
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/pharmtox/62/1/annurev-pharmtox-021921-093352.html?itemId=/content/journals/10.1146/annurev-pharmtox-021921-093352&mimeType=html&fmt=ahah

Literature Cited

  1. 1. 
    Kassotis CD, Vandenberg LN, Demeneix BA, Porta M, Slama R, Trasande L. 2020. Endocrine-disrupting chemicals: economic, regulatory, and policy implications. Lancet Diabetes Endocrinol. 8:719–30
    [Google Scholar]
  2. 2. 
    Gore AC, Chappell VA, Fenton SE, Flaws JA, Nadal A et al. 2015. EDC-2: the Endocrine Society's second scientific statement on endocrine-disrupting chemicals. Endocr. Rev. 36:E1–150
    [Google Scholar]
  3. 3. 
    Ragusa A, Svelato A, Santacroce C, Catalano P, Notarstefano V et al. 2021. Plasticenta: first evidence of microplastics in human placenta. Environ. Int. 146:106274
    [Google Scholar]
  4. 4. 
    Nahar MS, Liao C, Kannan K, Dolinoy DC 2013. Fetal liver bisphenol A concentrations and biotransformation gene expression reveal variable exposure and altered capacity for metabolism in humans. J. Biochem. Mol. Toxicol. 27:116–23
    [Google Scholar]
  5. 5. 
    Nahar MS, Liao C, Kannan K, Harris C, Dolinoy DC 2015. In utero bisphenol A concentration, metabolism, and global DNA methylation across matched placenta, kidney, and liver in the human fetus. Chemosphere 124:54–60
    [Google Scholar]
  6. 6. 
    Lehmann GM, LaKind JS, Davis MH, Hines EP, Marchitti SA et al. 2018. Environmental chemicals in breast milk and formula: exposure and risk assessment implications. Environ. Health Perspect. 126:96001
    [Google Scholar]
  7. 7. 
    Calatayud Arroyo M, García Barrera T, Callejón Leblic B, Arias Borrego A, Collado MC 2021. A review of the impact of xenobiotics from dietary sources on infant health: early life exposures and the role of the microbiota. Environ. Pollut. 269:115994
    [Google Scholar]
  8. 8. 
    Choi J, Knudsen LE, Mizrak S, Joas A 2017. Identification of exposure to environmental chemicals in children and older adults using human biomonitoring data sorted by age: results from a literature review. Int. J. Hyg. Environ. Health 220:282–98
    [Google Scholar]
  9. 9. 
    Fischer D, Hooper K, Athanasiadou M, Athanassiadis I, Bergman A 2006. Children show highest levels of polybrominated diphenyl ethers in a California family of four: a case study. Environ. Health Perspect. 114:1581–84
    [Google Scholar]
  10. 10. 
    Johnson-Restrepo B, Kannan K. 2009. An assessment of sources and pathways of human exposure to polybrominated diphenyl ethers in the United States. Chemosphere 76:542–48
    [Google Scholar]
  11. 11. 
    Wu Q, Kannan K 2019. Perfluoroalkyl substances (PFASs) in foodstuffs and human dietary exposure. Advances in the Determination of Xenobiotics in Foods B Gomara, ML Marina 258–311 Oak Park, IL: Bentham Sci. Publ.
    [Google Scholar]
  12. 12. 
    Tran TM, Kannan K. 2015. Occurrence of phthalate diesters in particulate and vapor phases in indoor air and implications for human exposure in Albany, New York, USA. Arch. Environ. Contam. Toxicol. 68:489–99
    [Google Scholar]
  13. 13. 
    Liao C, Kannan K. 2013. Concentrations and profiles of bisphenol A and other bisphenol analogues in foodstuffs from the United States and their implications for human exposure. J. Agric. Food. Chem. 61:4655–62
    [Google Scholar]
  14. 14. 
    Zhu H, Kannan K. 2019. Occurrence of melamine and its derivatives in breast milk from the United States and its implications for exposure in infants. Environ. Sci. Technol. 53:7859–65
    [Google Scholar]
  15. 15. 
    Asimakopoulos AG, Elangovan M, Kannan K. 2016. Migration of parabens, bisphenols, benzophenone-type UV filters, triclosan, and triclocarban from teethers and its implications for infant exposure. Environ. Sci. Technol. 50:13539–47
    [Google Scholar]
  16. 16. 
    Liao C, Kannan K. 2014. A survey of alkylphenols, bisphenols, and triclosan in personal care products from China and the United States. Arch. Environ. Contam. Toxicol. 67:50–59
    [Google Scholar]
  17. 17. 
    Guo Y, Wang L, Kannan K 2014. Phthalates and parabens in personal care products from China: concentrations and human exposure. Arch. Environ. Contam. Toxicol. 66:113–19
    [Google Scholar]
  18. 18. 
    Zhu H, Kannan K. 2020. Occurrence and distribution of melamine and its derivatives in surface water, drinking water, precipitation, wastewater, and swimming pool water. Environ. Pollut. 258:113743
    [Google Scholar]
  19. 19. 
    Gillman MW, Blaisdell CJ. 2018. Environmental Influences on Child Health Outcomes, a research program of the NIH. Curr. Opin. Pediatr. 30:260–62
    [Google Scholar]
  20. 20. 
    Vrijheid M, Slama R, Robinson O, Chatzi L, Coen M et al. 2014. The human early-life exposome (HELIX): project rationale and design. Environ. Health Perspect. 122:535–44
    [Google Scholar]
  21. 21. 
    Jaddoe VWV, Felix JF, Andersen A-MN, Charles M-A, Chatzi L et al. 2020. The LifeCycle Project-EU Child Cohort Network: a federated analysis infrastructure and harmonized data of more than 250,000 children and parents. Eur. J. Epidemiol. 35:709–24
    [Google Scholar]
  22. 22. 
    Zoeller TR, Dowling AL, Herzig CT, Iannacone EA, Gauger KJ, Bansal R. 2002. Thyroid hormone, brain development, and the environment. Environ. Health Perspect. 110:355–61
    [Google Scholar]
  23. 23. 
    McEwen BS, Milner TA. 2017. Understanding the broad influence of sex hormones and sex differences in the brain. . J. Neurosci. Res. 95:24–39
    [Google Scholar]
  24. 24. 
    Ponsonby AL, Symeonides C, Vuillermin P, Mueller J, Sly PD, Saffery R 2016. Epigenetic regulation of neurodevelopmental genes in response to in utero exposure to phthalate plastic chemicals: How can we delineate causal effects?. Neurotoxicology 55:92–101
    [Google Scholar]
  25. 25. 
    Bennett D, Bellinger DC, Birnbaum LS, Bradman A, Chen A et al. 2016. Project TENDR: targeting environmental neuro-developmental risks. The TENDR consensus statement. Environ. Health Perspect. 124:A118–22
    [Google Scholar]
  26. 26. 
    Luo J, Xiao J, Gao Y, Ramlau-Hansen CH, Toft G et al. 2020. Prenatal exposure to perfluoroalkyl substances and behavioral difficulties in childhood at 7 and 11 years. Environ. Res. 191:110111
    [Google Scholar]
  27. 27. 
    Lenters V, Iszatt N, Forns J, Čechová E, Kočan A et al. 2019. Early-life exposure to persistent organic pollutants (OCPs, PBDEs, PCBs, PFASs) and attention-deficit/hyperactivity disorder: A multi-pollutant analysis of a Norwegian birth cohort. Environ. Int. 125:33–42
    [Google Scholar]
  28. 28. 
    Ghassabian A, Bell EM, Ma WL, Sundaram R, Kannan K et al. 2018. Concentrations of perfluoroalkyl substances and bisphenol A in newborn dried blood spots and the association with child behavior. Environ. Pollut. 243:1629–36
    [Google Scholar]
  29. 29. 
    Weng JC, Hong CI, Tasi JD, Shen CY, Su PH, Wang SL. 2020. The association between prenatal endocrine-disrupting chemical exposure and altered resting-state brain fMRI in teenagers. Brain Struct. Funct. 225:1669–84
    [Google Scholar]
  30. 30. 
    Liew Z, Ritz B, Bach CC, Asarnow RF, Bech BH et al. 2018. Prenatal exposure to perfluoroalkyl substances and IQ scores at age 5; a study in the Danish national birth cohort. Environ. Health Perspect. 126:067004
    [Google Scholar]
  31. 31. 
    Liew Z, Olsen J, Cui X, Ritz B, Arah OA. 2015. Bias from conditioning on live birth in pregnancy cohorts: an illustration based on neurodevelopment in children after prenatal exposure to organic pollutants. Int. J. Epidemiol. 44:345–54
    [Google Scholar]
  32. 32. 
    Preston EV, McClean MD, Claus Henn B, Stapleton HM, Braverman LE et al. 2017. Associations between urinary diphenyl phosphate and thyroid function. Environ. Int. 101:158–64
    [Google Scholar]
  33. 33. 
    Hill KL, Hamers T, Kamstra JH, Willmore WG, Letcher RJ. 2018. Organophosphate triesters and selected metabolites enhance binding of thyroxine to human transthyretin in vitro. Toxicol. Lett. 285:87–93
    [Google Scholar]
  34. 34. 
    Hart R, Doherty DA, Frederiksen H, Keelan JA, Hickey M et al. 2014. The influence of antenatal exposure to phthalates on subsequent female reproductive development in adolescence: a pilot study. Reproduction 147:379–90
    [Google Scholar]
  35. 35. 
    Lin LC, Wang SL, Chang YC, Huang PC, Cheng JT et al. 2011. Associations between maternal phthalate exposure and cord sex hormones in human infants. Chemosphere 83:1192–99
    [Google Scholar]
  36. 36. 
    Sun D, Zhou L, Wang S, Liu T, Zhu J et al. 2018. Effect of di-(2-ethylhexyl) phthalate on the hypothalamus-pituitary-thyroid axis in adolescent rat. Endocr. J. 65:261–68
    [Google Scholar]
  37. 37. 
    Castorina R, Bradman A, Stapleton HM, Butt C, Avery D et al. 2017. Current-use flame retardants: maternal exposure and neurodevelopment in children of the CHAMACOS cohort. Chemosphere 189:574–80
    [Google Scholar]
  38. 38. 
    Yao Y, Li M, Pan L, Duan Y, Duan X et al. 2020. Exposure to organophosphate ester flame retardants and plasticizers during pregnancy: thyroid endocrine disruption and mediation role of oxidative stress. Environ. Int. 146:106215
    [Google Scholar]
  39. 39. 
    Radke EG, Braun JM, Nachman RM, Cooper GS. 2020. Phthalate exposure and neurodevelopment: a systematic review and meta-analysis of human epidemiological evidence. Environ. Int. 137:105408
    [Google Scholar]
  40. 40. 
    Zhang Q, Chen XZ, Huang X, Wang M, Wu J 2019. The association between prenatal exposure to phthalates and cognition and neurobehavior of children—evidence from birth cohorts. Neurotoxicology 73:199–212
    [Google Scholar]
  41. 41. 
    Shoaff JR, Calafat AM, Schantz SL, Korrick SA. 2019. Endocrine disrupting chemical exposure and maladaptive behavior during adolescence. Environ. Res. 172:231–41
    [Google Scholar]
  42. 42. 
    Schulz KM, Sisk CL. 2016. The organizing actions of adolescent gonadal steroid hormones on brain and behavioral development. Neurosci. Biobehav. Rev. 70:148–58
    [Google Scholar]
  43. 43. 
    Barker DJ, Osmond C, Forsen TJ, Kajantie E, Eriksson JG 2005. Trajectories of growth among children who have coronary events as adults. N. Engl. J. Med. 353:1802–9
    [Google Scholar]
  44. 44. 
    Philips EM, Jaddoe VW, Trasande L. 2017. Effects of early exposure to phthalates and bisphenols on cardiometabolic outcomes in pregnancy and childhood. Reprod. Toxicol. 68:105–18
    [Google Scholar]
  45. 45. 
    Chen M-Y, Ike M, Fujita M 2002. Acute toxicity, mutagenicity, and estrogenicity of bisphenol-A and other bisphenols. Environ. Toxicol. 17:80–86
    [Google Scholar]
  46. 46. 
    Chin KY, Pang KL, Mark-Lee WF. 2018. A review on the effects of bisphenol A and its derivatives on skeletal health. Int. J. Med. Sci. 15:1043–50
    [Google Scholar]
  47. 47. 
    Xin F, Smith LM, Susiarjo M, Bartolomei MS, Jepsen KJ 2018. Endocrine-disrupting chemicals, epigenetics, and skeletal system dysfunction: exploration of links using bisphenol A as a model system. Environ. Epigenet. 4:dvy002
    [Google Scholar]
  48. 48. 
    Hugo ER, Brandebourg TD, Woo JG, Loftus J, Alexander JW, Ben-Jonathan N 2008. Bisphenol A at environmentally relevant doses inhibits adiponectin release from human adipose tissue explants and adipocytes. Environ. Health Perspect. 116:1642–47
    [Google Scholar]
  49. 49. 
    Desvergne B, Feige JN, Casals-Casas C. 2009. PPAR-mediated activity of phthalates: a link to the obesity epidemic?. Mol. Cell. Endocrinol. 304:43–48
    [Google Scholar]
  50. 50. 
    Gourlay T, Samartzis I, Stefanou D, Taylor K. 2003. Inflammatory response of rat and human neutrophils exposed to di-(2-ethyl-hexyl)-phthalate-plasticized polyvinyl chloride. Artif. Organs 27:256–60
    [Google Scholar]
  51. 51. 
    Desdoits-Lethimonier C, Albert O, Le Bizec B, Perdu E, Zalko D et al. 2012. Human testis steroidogenesis is inhibited by phthalates. Hum. Reprod. 27:1451–59
    [Google Scholar]
  52. 52. 
    Woodward MJ, Obsekov V, Jacobson MH, Kahn LG, Trasande L. 2020. Phthalates and sex steroid hormones among men from NHANES, 2013–2016. J. Clin. Endocrinol. Metab. 105:e1225–34
    [Google Scholar]
  53. 53. 
    Kahn LG, Philippat C, Nakayama SF, Slama R, Trasande L. 2020. Endocrine-disrupting chemicals: implications for human health. Lancet Diabetes Endocrinol. 8:703–18
    [Google Scholar]
  54. 54. 
    Bowman A, Peterson KE, Dolinoy DC, Meeker JD, Sánchez BN et al. 2019. Phthalate exposures, DNA methylation and adiposity in Mexican children through adolescence. Front. Public Health 7:162
    [Google Scholar]
  55. 55. 
    Yang TC, Peterson KE, Meeker JD, Sánchez BN, Zhang Z et al. 2017. Bisphenol A and phthalates in utero and in childhood: association with child BMI z-score and adiposity. Environ. Res. 156:326–33
    [Google Scholar]
  56. 56. 
    Valvi D, Casas M, Mendez MA, Ballesteros-Gomez A, Luque N et al. 2013. Prenatal bisphenol a urine concentrations and early rapid growth and overweight risk in the offspring. Epidemiology 24:791–99
    [Google Scholar]
  57. 57. 
    Buckley JP, Engel SM, Braun JM, Whyatt RM, Daniels JL et al. 2016. Prenatal phthalate exposures and body mass index among 4- to 7-year-old children: a pooled analysis. Epidemiology 27:449–58
    [Google Scholar]
  58. 58. 
    Harley KG, Berger K, Rauch S, Kogut K, Claus Henn B et al. 2017. Association of prenatal urinary phthalate metabolite concentrations and childhood BMI and obesity. Pediatr. Res. 82:405–15
    [Google Scholar]
  59. 59. 
    Hoepner LA, Whyatt RM, Widen EM, Hassoun A, Oberfield SE et al. 2016. Bisphenol A and adiposity in an inner-city birth cohort. Environ. Health Perspect. 124:1644–50
    [Google Scholar]
  60. 60. 
    Braun JM, Li N, Arbuckle TE, Dodds L, Massarelli I et al. 2019. Association between gestational urinary bisphenol A concentrations and adiposity in young children: the MIREC study. Environ. Res. 172:454–61
    [Google Scholar]
  61. 61. 
    Janesick A, Blumberg B. 2011. Endocrine disrupting chemicals and the developmental programming of adipogenesis and obesity. Birth Defects Res. C Embryo Today 93:34–50
    [Google Scholar]
  62. 62. 
    Steenland K, Barry V, Savitz D 2018. Serum perfluorooctanoic acid and birthweight: an updated meta-analysis with bias analysis. Epidemiology 29:765–76
    [Google Scholar]
  63. 63. 
    Starling AP, Adgate JL, Hamman RF, Kechris K, Calafat AM, Dabelea D. 2019. Prenatal exposure to per- and polyfluoroalkyl substances and infant growth and adiposity: the Healthy Start study. Environ. Int. 131:104983
    [Google Scholar]
  64. 64. 
    Halldorsson TI, Rytter D, Haug LS, Bech BH, Danielsen I et al. 2012. Prenatal exposure to perfluorooctanoate and risk of overweight at 20 years of age: a prospective cohort study. Environ. Health Perspect. 120:668–73
    [Google Scholar]
  65. 65. 
    Braun JM, Chen A, Romano ME, Calafat AM, Webster GM et al. 2016. Prenatal perfluoroalkyl substance exposure and child adiposity at 8 years of age: the HOME study. Obesity 24:231–37
    [Google Scholar]
  66. 66. 
    Mora AM, Oken E, Rifas-Shiman SL, Webster TF, Gillman MW et al. 2017. Prenatal exposure to perfluoroalkyl substances and adiposity in early and mid-childhood. Environ. Health Perspect. 125:467–73
    [Google Scholar]
  67. 67. 
    Chen Q, Zhang X, Zhao Y, Lu W, Wu J et al. 2019. Prenatal exposure to perfluorobutanesulfonic acid and childhood adiposity: a prospective birth cohort study in Shanghai, China. Chemosphere 226:17–23
    [Google Scholar]
  68. 68. 
    Liu P, Yang F, Wang Y, Yuan Z 2018. Perfluorooctanoic acid (PFOA) exposure in early life increases risk of childhood adiposity: a meta-analysis of prospective cohort studies. Int. J. Environ. Res. Public Health 15:2070
    [Google Scholar]
  69. 69. 
    Nowak K, Jabłońska E, Ratajczak-Wrona W. 2019. Immunomodulatory effects of synthetic endocrine disrupting chemicals on the development and functions of human immune cells. Environ. Int. 125:350–64
    [Google Scholar]
  70. 70. 
    Granum B, Haug LS, Namork E, Stolevik SB, Thomsen C et al. 2013. Pre-natal exposure to perfluoroalkyl substances may be associated with altered vaccine antibody levels and immune-related health outcomes in early childhood. J. Immunotoxicol. 10:373–79
    [Google Scholar]
  71. 71. 
    Goudarzi H, Miyashita C, Okada E, Kashino I, Chen CJ et al. 2017. Prenatal exposure to perfluoroalkyl acids and prevalence of infectious diseases up to 4 years of age. Environ. Int. 104:132–8
    [Google Scholar]
  72. 72. 
    Fei C, McLaughlin JK, Lipworth L, Olsen J 2010. Prenatal exposure to PFOA and PFOS and risk of hospitalization for infectious diseases in early childhood. Environ. Res. 110:773–77
    [Google Scholar]
  73. 73. 
    Grandjean P, Andersen E, Budtz-Jorgensen E, Nielsen F, Molbak K, Weihe P. 2012. Serum vaccine antibody concentrations in children exposed to perfluorinated compounds. JAMA 307:391–97
    [Google Scholar]
  74. 74. 
    Watkins DJ, Ferguson KK, Anzalota Del Toro LV, Alshawabkeh AN, Cordero JF, Meeker JD 2015. Associations between urinary phenol and paraben concentrations and markers of oxidative stress and inflammation among pregnant women in Puerto Rico. Int. J. Hyg. Environ. Health 218:212–19
    [Google Scholar]
  75. 75. 
    Kelley AS, Banker M, Goodrich JM, Dolinoy DC, Burant C et al. 2019. Early pregnancy exposure to endocrine disrupting chemical mixtures are associated with inflammatory changes in maternal and neonatal circulation. Sci. Rep. 9:5422
    [Google Scholar]
  76. 76. 
    Yang SN, Hsieh CC, Kuo HF, Lee MS, Huang MY et al. 2014. The effects of environmental toxins on allergic inflammation. Allergy Asthma Immunol. Res. 6:478–84
    [Google Scholar]
  77. 77. 
    Casas M, Gascon M. 2020. Prenatal exposure to endocrine-disrupting chemicals and asthma and allergic diseases. J. Investig. Allergol. Clin. Immunol. 30:215–28
    [Google Scholar]
  78. 78. 
    Rogers JA, Metz L, Yong VW. 2013. Review: endocrine disrupting chemicals and immune responses: a focus on bisphenol-A and its potential mechanisms. Mol. Immunol. 53:421–30
    [Google Scholar]
  79. 79. 
    Thorup J, McLachlan R, Cortes D, Nation TR, Balic A et al. 2010. What is new in cryptorchidism and hypospadias—a critical review on the testicular dysgenesis hypothesis. J. Pediatr. Surg. 45:2074–86
    [Google Scholar]
  80. 80. 
    Xing JS, Bai ZM. 2018. Is testicular dysgenesis syndrome a genetic, endocrine, or environmental disease, or an unexplained reproductive disorder?. Life Sci 194:120–29
    [Google Scholar]
  81. 81. 
    Yu X, Nassar N, Mastroiacovo P, Canfield M, Groisman B et al. 2019. Hypospadias prevalence and trends in international birth defect surveillance systems, 1980–2010. Eur. Urol. 76:482–90
    [Google Scholar]
  82. 82. 
    Surveill. Epidemiol. End Results Program 2018. Cancer stat facts: testicular cancer Rep., Natl. Cancer Inst. Bethesda, MD: https://seer.cancer.gov/statfacts/html/testis.html
  83. 83. 
    Bray F, Richiardi L, Ekbom A, Pukkala E, Cuninkova M, Møller H 2006. Trends in testicular cancer incidence and mortality in 22 European countries: continuing increases in incidence and declines in mortality. Int. J. Cancer 118:3099–111
    [Google Scholar]
  84. 84. 
    Sathyanarayana S, Grady R, Barrett ES, Redmon B, Nguyen RHN et al. 2016. First trimester phthalate exposure and male newborn genital anomalies. Environ. Res. 151:777–82
    [Google Scholar]
  85. 85. 
    Fisher BG, Thankamony A, Mendiola J, Petry CJ, Frederiksen H et al. 2020. Maternal serum concentrations of bisphenol A and propyl paraben in early pregnancy are associated with male infant genital development. Hum. Reprod. 35:913–28
    [Google Scholar]
  86. 86. 
    Sunman B, Yurdakök K, Kocer-Gumusel B, Özyüncü Ö, Akbıyık F et al. 2019. Prenatal bisphenol A and phthalate exposure are risk factors for male reproductive system development and cord blood sex hormone levels. Reprod. Toxicol. 87:146–55
    [Google Scholar]
  87. 87. 
    Barrett ES, Parlett LE, Sathyanarayana S, Redmon JB, Nguyen RH, Swan SH. 2016. Prenatal stress as a modifier of associations between phthalate exposure and reproductive development: results from a multicentre pregnancy cohort study. Paediatr. Perinat. Epidemiol. 30:105–14
    [Google Scholar]
  88. 88. 
    Bornehag CG, Carlstedt F, Jönsson BA, Lindh CH, Jensen TK et al. 2015. Prenatal phthalate exposures and anogenital distance in Swedish boys. Environ. Health Perspect. 123:101–7
    [Google Scholar]
  89. 89. 
    Muerköster AP, Frederiksen H, Juul A, Andersson AM, Jensen RC et al. 2020. Maternal phthalate exposure associated with decreased testosterone/LH ratio in male offspring during mini-puberty. Odense Child Cohort. Environ. Int 144:106025
    [Google Scholar]
  90. 90. 
    Hart RJ, Frederiksen H, Doherty DA, Keelan JA, Skakkebaek NE et al. 2018. The possible impact of antenatal exposure to ubiquitous phthalates upon male reproductive function at 20years of age. Front. Endocrinol 9:288
    [Google Scholar]
  91. 91. 
    Jensen TK, Frederiksen H, Kyhl HB, Lassen TH, Swan SH et al. 2016. Prenatal exposure to phthalates and anogenital distance in male infants from a low-exposed Danish cohort (2010–2012). Environ. Health Perspect. 124:1107–13
    [Google Scholar]
  92. 92. 
    Wenzel AG, Bloom MS, Butts CD, Wineland RJ, Brock JW et al. 2018. Influence of race on prenatal phthalate exposure and anogenital measurements among boys and girls. Environ. Int. 110:61–70
    [Google Scholar]
  93. 93. 
    Wineland RJ, Bloom MS, Cruze L, Butts CD, Wenzel AG et al. 2019. In utero effects of maternal phthalate exposure on male genital development. Prenat. Diagn. 39:209–18
    [Google Scholar]
  94. 94. 
    Giannandrea F, Fargnoli S 2017. Environmental factors affecting growth and occurrence of testicular cancer in childhood: an overview of the current epidemiological evidence. Children 4:1
    [Google Scholar]
  95. 95. 
    Cathey A, Watkins DJ, Sánchez BN, Tamayo-Ortiz M, Solano-Gonzalez M et al. 2020. Onset and tempo of sexual maturation is differentially associated with gestational phthalate exposure between boys and girls in a Mexico City birth cohort. Environ. Int. 136:105469
    [Google Scholar]
  96. 96. 
    Buck Louis G, Cooney MA, Peterson CM 2011. Ovarian dysgenesis syndrome. J. Dev. Orig. Health Dis. 2:25–35
    [Google Scholar]
  97. 97. 
    Rudel RA, Fenton SE, Ackerman JM, Euling SY, Makris SL. 2011. Environmental exposures and mammary gland development: state of the science, public health implications, and research recommendations. Environ. Health Perspect. 119:1053–61
    [Google Scholar]
  98. 98. 
    Cohn BA, Wolff MS, Cirillo PM, Sholtz RI. 2007. DDT and breast cancer in young women: new data on the significance of age at exposure. Environ. Health Perspect. 115:1406–14
    [Google Scholar]
  99. 99. 
    Kahn LG, Harley KG, Siegel EL, Zhu Y, Factor-Litvak P et al. 2021. Persistent organic pollutants and couple fecundability: a systematic review. Hum. Reprod. Update 27:2339–66
    [Google Scholar]
  100. 100. 
    Carignan CC, Minguez-Alarcon L, Butt CM, Williams PL, Meeker JD et al. 2017. Urinary concentrations of organophosphate flame retardant metabolites and pregnancy outcomes among women undergoing in vitro fertilization. Environ. Health Perspect. 125:087018
    [Google Scholar]
  101. 101. 
    Brenner BM, Garcia DL, Anderson S 1988. Glomeruli and blood pressure. Less of one, more the other?. Am. J. Hypertens. 1:335–47
    [Google Scholar]
  102. 102. 
    Luyckx VA, Brenner BM. 2015. Birth weight, malnutrition and kidney-associated outcomes—a global concern. Nat. Rev. Nephrol. 11:135–49
    [Google Scholar]
  103. 103. 
    Ashari S, Karami M, Shokrzadeh M, Ghandadi M, Ghassemi-Barghi N et al. 2020. The implication of mitochondrial dysfunction and mitochondrial oxidative damage in di (2-ethylhexyl) phthalate induced nephrotoxicity in both in vivo and in vitro models. Toxicol. Mech. Methods 30:427–37
    [Google Scholar]
  104. 104. 
    Vasconcelos AL, Silva MJ, Louro H. 2019. In vitro exposure to the next-generation plasticizer diisononyl cyclohexane-1,2-dicarboxylate (DINCH): cytotoxicity and genotoxicity assessment in human cells. J. Toxicol. Environ. Health A 82:526–36
    [Google Scholar]
  105. 105. 
    Stanifer JW, Stapleton HM, Souma T, Wittmer A, Zhao X, Boulware LE. 2018. Perfluorinated chemicals as emerging environmental threats to kidney health: a scoping review. Clin. J. Am. Soc. Nephrol. 13:1479–92
    [Google Scholar]
  106. 106. 
    Zheng LY, Sanders AP, Saland JM, Wright RO, Arora M. 2017. Environmental exposures and pediatric kidney function and disease: a systematic review. Environ. Res. 158:625–48
    [Google Scholar]
  107. 107. 
    Blake BE, Fenton SE. 2020. Early life exposure to per- and polyfluoroalkyl substances (PFAS) and latent health outcomes: a review including the placenta as a target tissue and possible driver of peri- and postnatal effects. Toxicology 443:152565
    [Google Scholar]
  108. 108. 
    Jacobson MH, Wu Y, Liu M, Attina TM, Naidu M et al. 2020. Serially assessed bisphenol A and phthalate exposure and association with kidney function in children with chronic kidney disease in the US and Canada: a longitudinal cohort study. PLOS Med 17:e1003384
    [Google Scholar]
  109. 109. 
    Malits J, Attina TM, Karthikraj R, Kannan K, Naidu M et al. 2018. Renal function and exposure to bisphenol A and phthalates in children with chronic kidney disease. Environ. Res. 167:575–82
    [Google Scholar]
  110. 110. 
    Watkins DJ, Josson J, Elston B, Bartell SM, Shin HM et al. 2013. Exposure to perfluoroalkyl acids and markers of kidney function among children and adolescents living near a chemical plant. Environ. Health Perspect. 121:625–30
    [Google Scholar]
  111. 111. 
    Kataria A, Trachtman H, Malaga-Dieguez L, Trasande L. 2015. Association between perfluoroalkyl acids and kidney function in a cross-sectional study of adolescents. Environ. Health 14:89
    [Google Scholar]
  112. 112. 
    Geiger SD, Xiao J, Shankar A 2013. Positive association between perfluoroalkyl chemicals and hyperuricemia in children. Am. J. Epidemiol. 177:1255–62
    [Google Scholar]
  113. 113. 
    Trasande L, Attina TM, Trachtman H. 2013. Bisphenol A exposure is associated with low-grade urinary albumin excretion in children of the United States. Kidney Int 83:741–48
    [Google Scholar]
  114. 114. 
    Trasande L, Sathyanarayana S, Trachtman H. 2014. Dietary phthalates and low-grade albuminuria in US children and adolescents. Clin. J. Am. Soc. Nephrol. 9:100–9
    [Google Scholar]
  115. 115. 
    Tsai HJ, Chen BH, Wu CF, Wang SL, Huang PC et al. 2016. Intake of phthalate-tainted foods and microalbuminuria in children: the 2011 Taiwan food scandal. Environ. Int 89–90:129–37
    [Google Scholar]
  116. 116. 
    Vernet C, Philippat C, Agier L, Calafat AM, Ye X et al. 2019. An empirical validation of the within-subject biospecimens pooling approach to minimize exposure misclassification in biomarker-based studies. Epidemiology 30:756–67
    [Google Scholar]
  117. 117. 
    Hamra GB, Buckley JP. 2018. Environmental exposure mixtures: questions and methods to address them. Curr. Epidemiol. Rep. 5:160–65
    [Google Scholar]
  118. 118. 
    Carvalho CF, Menezes-Filho JA, de Matos VP, Bessa JR, Coelho-Santos J et al. 2014. Elevated airborne manganese and low executive function in school-aged children in Brazil. Neurotoxicology 45:301–8
    [Google Scholar]
  119. 119. 
    Bradford Hill A 1965. The environment and disease: association or causation?. Proc. R. Soc. Med. 58:295–300
    [Google Scholar]
  120. 120. 
    IPCS (Int. Programme Chem. Saf.) 2004. IPCS risk assessment terminology Harmon. Proj. Doc. 1, World Health Organ. Geneva, Switz:.
  121. 121. 
    Woodruff TJ, Zeise L, Axelrad DA, Guyton KZ, Janssen S et al. 2008. Meeting report: moving upstream—evaluating adverse upstream end points for improved risk assessment and decision-making. Environ. Health Perspect. 116:1568–75
    [Google Scholar]
  122. 122. 
    Vandenberg LN, Hunt PA, Gore AC. 2019. Endocrine disruptors and the future of toxicology testing—lessons from CLARITY-BPA. Nat. Rev. Endocrinol. 15:366–74
    [Google Scholar]
  123. 123. 
    Diamanti-Kandarakis E, Bourguignon JP, Giudice LC, Hauser R, Prins GS et al. 2009. Endocrine-disrupting chemicals: an Endocrine Society scientific statement. Endocr. Rev. 30:293–342
    [Google Scholar]
  124. 124. 
    Maffini MV, Alger HM, Bongard ED, Neltner TG. 2011. Enhancing FDA's evaluation of science to ensure chemicals added to human food are safe: workshop proceedings. Comp. Rev. Food Sci. Food Saf. 10:321–41
    [Google Scholar]
  125. 125. 
    Mendiola J, Stahlhut RW, Jorgensen N, Liu F, Swan SH. 2011. Shorter anogenital distance predicts poorer semen quality in young men in Rochester, New York. Environ. Health Perspect. 119:958–63
    [Google Scholar]
  126. 126. 
    US FDA (US Food Drug Admin.) 2019. Listing of color additives exempt from certification; soy leghemoglobin. Fed. Regist. 84:244
    [Google Scholar]
  127. 127. 
    Gore AC, Martien KM, Gagnidze K, Pfaff D 2014. Implications of prenatal steroid perturbations for neurodevelopment, behavior, and autism. Endocr. Rev. 35:961–91
    [Google Scholar]
  128. 128. 
    Heindel JJ, Vandenberg LN. 2015. Developmental origins of health and disease: a paradigm for understanding disease etiology and prevention. Curr. Opin. Pediatr. 27:248–53
    [Google Scholar]
  129. 129. 
    Vandenberg LN, Colborn T, Hayes TB 2013. Regulatory decisions on endocrine disrupting chemicals should be based on the principles of endocrinology. Reprod. Toxicol. 38:1–15
    [Google Scholar]
  130. 130. 
    Wallen K. 2009. The organizational hypothesis: reflections on the 50th anniversary of the publication of Phoenix, Goy, Gerall, and Young 1959. Horm. Behav. 55:561–65
    [Google Scholar]
  131. 131. 
    Miller MD, Crofton KM, Rice DC, Zoeller RT 2009. Thyroid-disrupting chemicals: interpreting upstream biomarkers of adverse outcomes. Environ. Health Perspect. 117:1033–41
    [Google Scholar]
  132. 132. 
    Calafat AM, Longnecker MP, Koch HM, Swan SH, Hauser R et al. 2015. Optimal exposure biomarkers for nonpersistent chemicals in environmental epidemiology. Environ. Health Perspect. 123:A166–68
    [Google Scholar]
  133. 133. 
    Vandenberg LN. 2019. Low dose effects challenge the evaluation of endocrine disrupting chemicals. Trends. Food. Sci. Technol. 84:58–61
    [Google Scholar]
  134. 134. 
    Woodruff TJ, Zota AR, Schwartz JM. 2011. Environmental chemicals in pregnant women in the United States: NHANES 2003–2004. Environ. Health Perspect. 119:878–85
    [Google Scholar]
  135. 135. 
    NASEM (Natl. Acad. Sci. Eng. Med.) 2017. Application of systematic review methods in an overall strategy for evaluating low-dose toxicity from endocrine active chemicals Rep., Natl. Acad. Sci. Eng. Med. Washington, DC:
  136. 136. 
    Vandenberg LN, Colborn T, Hayes TB, Heindel JJ, Jacobs DR Jr. et al. 2012. Hormones and endocrine-disrupting chemicals: low-dose effects and nonmonotonic dose responses. Endocr. Rev. 33:378–455
    [Google Scholar]
  137. 137. 
    Demeneix B, Vandenberg LN, Ivell R, Zoeller RT. 2020. Thresholds and endocrine disruptors: an endocrine society policy perspective. J. Endocr. Soc. 4:bvaa085
    [Google Scholar]
  138. 138. 
    Bradman A, Quirós-Alcalá L, Castorina R, Aguilar Schall R, Camacho J et al. 2015. Effect of organic diet intervention on pesticide exposures in young children living in low-income urban and agricultural communities. Environ. Health Perspect. 123:1086–93
    [Google Scholar]
  139. 139. 
    Harley KG, Kogut K, Madrigal DS, Cardenas M, Vera IA et al. 2016. Reducing phthalate, paraben, and phenol exposure from personal care products in adolescent girls: findings from the HERMOSA intervention study. Environ. Health Perspect. 124:1600–7
    [Google Scholar]
  140. 140. 
    Rudel RA, Gray JM, Engel CL, Rawsthorne TW, Dodson RE et al. 2011. Food packaging and bisphenol A and bis(2-ethyhexyl) phthalate exposure: findings from a dietary intervention. Environ. Health Perspect. 119:914–20
    [Google Scholar]
  141. 141. 
    Young AS, Hauser R, James-Todd TM, Coull BA, Zhu H et al. 2021. Impact of “healthier” materials interventions on dust concentrations of per- and polyfluoroalkyl substances, polybrominated diphenyl ethers, and organophosphate esters. Environ. Int. 150:106151
    [Google Scholar]
  142. 142. 
    Attina TM, Hauser R, Sathyanarayana S, Hunt PA, Bourguignon J-P et al. 2016. Exposure to endocrine-disrupting chemicals in the USA: a population-based disease burden and cost analysis. Lancet Diabetes Endocrinol. 4:996–1003
    [Google Scholar]
  143. 143. 
    Trasande L, Zoeller RT, Hass U, Kortenkamp A, Grandjean P et al. 2015. Estimating burden and disease costs of exposure to endocrine-disrupting chemicals in the European union. J. Clin. Endocrinol. Metab. 100:1245–55
    [Google Scholar]
  144. 144. 
    Trasande L. 2014. Further limiting bisphenol A in food uses could provide health and economic benefits. Health Aff 33:316–23
    [Google Scholar]
  145. 145. 
    Yang J, Zhao Y, Li M, Du M, Li X, Li Y 2019. A review of a class of emerging contaminants: the classification, distribution, intensity of consumption, synthesis routes, environmental effects and expectation of pollution abatement to organophosphate flame retardants (OPFRs). Int. J. Mol. Sci. 20:2874
    [Google Scholar]
  146. 146. 
    Kunikane H, Watanabe K, Fukuoka M, Saijo N, Furuse K et al. 2001. Double-blind randomized control trial of the effect of recombinant human erythropoietin on chemotherapy-induced anemia in patients with non-small cell lung cancer. Int. J. Clin. Oncol. 6:296–301
    [Google Scholar]
  147. 147. 
    Goulson D. 2013. Review: an overview of the environmental risks posed by neonicotinoid insecticides. J. Appl. Ecol. 50:977–87
    [Google Scholar]
  148. 148. 
    Brendel S, Fetter É, Staude C, Vierke L, Biegel-Engler A. 2018. Short-chain perfluoroalkyl acids: environmental concerns and a regulatory strategy under REACH. Environ. Sci. Eur. 30:9
    [Google Scholar]
  149. 149. 
    Xu H, Zhou Q, Zhang J, Chen X, Zhao H et al. 2020. Exposure to elevated per- and polyfluoroalkyl substances in early pregnancy is related to increased risk of gestational diabetes mellitus: a nested case-control study in Shanghai, China. Environ. Int. 143:105952
    [Google Scholar]
  150. 150. 
    La Merrill MA, Vandenberg LN, Smith MT, Goodson W, Browne P et al. 2020. Consensus on the key characteristics of endocrine-disrupting chemicals as a basis for hazard identification. Nat. Rev. Endocrinol. 16:45–57
    [Google Scholar]
/content/journals/10.1146/annurev-pharmtox-021921-093352
Loading
/content/journals/10.1146/annurev-pharmtox-021921-093352
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error