Hostname: page-component-76fb5796d-x4r87 Total loading time: 0 Render date: 2024-04-26T06:38:30.038Z Has data issue: false hasContentIssue false

Perinatal exposures to phthalates and phthalate mixtures result in sex-specific effects on body weight, organ weights and intracisternal A-particle (IAP) DNA methylation in weanling mice

Published online by Cambridge University Press:  11 July 2018

K. Neier
Affiliation:
Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
D. Cheatham
Affiliation:
Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
L. D. Bedrosian
Affiliation:
Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
D. C. Dolinoy*
Affiliation:
Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
*
Address for correspondence: Dana C. Dolinoy, Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA. E-mail: ddolinoy@umich.edu

Abstract

Developmental exposure to phthalates has been implicated as a risk for obesity; however, epidemiological studies have yielded conflicting results and mechanisms are poorly understood. An additional layer of complexity in epidemiological studies is that humans are exposed to mixtures of many different phthalates. Here, we utilize an established mouse model of perinatal exposure to investigate the effects of three phthalates, diethylhexyl phthalate (DEHP), diisononyl phthalate (DINP) and dibutyl phthalate (DBP), on body weight and organ weights in weanling mice. In addition to individual phthalate exposures, we employed two mixture exposures: DEHP+DINP and DEHP+DINP+DBP. Phthalates were administered through phytoestrogen-free chow at the following exposure levels: 25 mg DEHP/kg chow, 25 mg DBP/kg chow and 75 mg DINP/kg chow. The viable yellow agouti (Avy) mouse strain, along with measurement of tail DNA methylation, was used as a biosensor to examine effects of phthalates and phthalate mixtures on the DNA methylome. We found that female and male mice perinatally exposed to DINP alone had increased body weights at postnatal day 21 (PND21), and that exposure to mixtures did not exaggerate these effects. Females exposed to DINP and DEHP+DINP had increased relative liver weights at PND21, and females exposed to a mixture of DEHP+DINP+DBP had increased relative gonadal fat weight. Phthalate-exposed Avy/a offspring exhibited altered coat color distributions and altered DNA methylation at intracisternal A-particles (IAPs), repetitive elements in the mouse genome. These findings provide evidence that developmental exposures to phthalates influence body weight and organ weight changes in early life, and are associated with altered DNA methylation at IAPs.

Type
Original Article
Copyright
© Cambridge University Press and the International Society for Developmental Origins of Health and Disease 2018. 

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

Footnotes

These authors contributed equally to this work.

References

1. Haugen, AC, Schug, TT, Collman, G, Heindel, JJ. Evolution of DOHaD: the impact of environmental health sciences. J Dev Orig Health Dis. 2015; 6, 5564.Google Scholar
2. Schettler, T. Human exposure to phthalates via consumer products. Int J Androl. 2006; 29, 134139.Google Scholar
3. Heudorf, U, Mersch-Sundermann, V, Angerer, J. Phthalates: toxicology and exposure. Int J Hyg Environ Health. 2007; 210, 623634.Google Scholar
4. Foster, P. Disruption of reproductive development in male rat offspring following in utero exposure to phthalate esters. Int J Androl. 2006; 29, 140147.Google Scholar
5. Hsieh, MH, Breyer, BN, Eisenberg, ML, Baskin, LS. Associations among hypospadias, cryptorchidism, anogenital distance, and endocrine disruption. Curr Urol Rep. 2008; 9, 137142.Google Scholar
6. Hannon, PR, Flaws, JA. The effects of phthalates on the ovary. Front Endocrinol (Lausanne). 2015; 6, 8.Google Scholar
7. Hao, C, Cheng, X, Guo, J, Xia, H, Ma, X. Perinatal exposure to diethyl-hexyl-phthalate induces obesity in mice. Front Biosci (Elite Ed). 2013; 5, 725733.Google Scholar
8. Yang, TC, Peterson, KE, Meeker, JD, et al. Bisphenol A and phthalates in utero and in childhood: association with child BMI z-score and adiposity. Environ Res. 2017; 156, 326333.Google Scholar
9. Rajesh, P, Balasubramanian, K. Phthalate exposure in utero causes epigenetic changes and impairs insulin signalling. J Endocrinol. 2014; 223, 4766.Google Scholar
10. Strakovsky, RS, Lezmi, S, Shkoda, I, et al. In utero growth restriction and catch-up adipogenesis after developmental di (2-ethylhexyl) phthalate exposure cause glucose intolerance in adult male rats following a high-fat dietary challenge. J Nutr Biochem. 2015; 26, 12081220.Google Scholar
11. Buckley, JP, Engel, SM, Braun, JM, et al. Prenatal phthalate exposures and body mass index among 4- to 7-year-old children: a pooled analysis. Epidemiology. 2016; 27, 449458.Google Scholar
12. Shoaff, J, Papandonatos, GD, Calafat, AM, et al. Early-life phthalate exposure and adiposity at 8 years of age. Environ Health Perspect. 2017; 125, 97008.Google Scholar
13. Braun, JM, Gennings, C, Hauser, R, Webster, TF. What can epidemiological studies tell us about the impact of chemical mixtures on human health? Environ Health Perspect. 2016; 124, A6A9.Google Scholar
14. Braun, JM, Just, AC, Williams, PL, et al. Personal care product use and urinary phthalate metabolite and paraben concentrations during pregnancy among women from a fertility clinic. J Expo Sci Environ Epidemiol. 2014; 24, 459466.Google Scholar
15. Carlson, KR, Garland, SE. Estimated phthalate exposure and risk to pregnant women and women of reproductive age as assessed using four NHANES biomonitoring data sets (2005/2006, 2007/2008, 2009/2010, 2011/2012). Consumer Product Safety Comission Directorate for Hazard Identification and Reduction. 2015.Google Scholar
16. Reik, W, Dean, W, Walter, J. Epigenetic reprogramming in mammalian development. Science. 2001; 293, 10891093.Google Scholar
17. Jirtle, RL, Skinner, MK. Environmental epigenomics and disease susceptibility. Nat Rev Genet. 2007; 8, 253262.Google Scholar
18. Dolinoy, DC, Das, R, Weidman, JR, Jirtle, RL. Metastable epialleles, imprinting, and the fetal origins of adult diseases. Pediatr Res. 2007; 61(Pt 2), 30R37R.Google Scholar
19. Faulk, C, Dolinoy, DC. Timing is everything: the when and how of environmentally induced changes in the epigenome of animals. Epigenetics. 2011; 6, 791797.Google Scholar
20. Zhang, X-F, Zhang, L-J, Li, L, et al. Diethylhexyl phthalate exposure impairs follicular development and affects oocyte maturation in the mouse. Environ Mol Mutagen. 2013; 54, 354361.Google Scholar
21. Martinez-Arguelles, DB, Papadopoulos, V. Identification of hot spots of DNA methylation in the adult male adrenal in response to in utero exposure to the ubiquitous endocrine disruptor plasticizer di-(2-ethylhexyl) phthalate. Endocrinology. 2015; 156, 124133.Google Scholar
22. Huen, K, Calafat, AM, Bradman, A, et al. Maternal phthalate exposure during pregnancy is associated with DNA methylation of LINE-1 and Alu repetitive elements in Mexican-American children. Environ Res. 2016; 148, 5562.Google Scholar
23. Solomon, O, Yousefi, P, Huen, K, et al. Prenatal phthalate exposure and altered patterns of DNA methylation in cord blood. Environ Mol Mutagen. 2017; 58, 398410.Google Scholar
24. Wu, H, Estill, MS, Shershebnev, A, et al. Preconception urinary phthalate concentrations and sperm DNA methylation profiles among men undergoing IVF treatment: a cross-sectional study. Hum Reprod. 2017; 32, 21592169.Google Scholar
25. Waterland, RA, Jirtle, RL. Early nutrition, epigenetic changes at transposons and imprinted genes, and enhanced susceptibility to adult chronic diseases. Nutrition. 2004; 20, 6368.Google Scholar
26. Qin, C, Wang, Z, Shang, J, et al. Intracisternal A-particle genes: distribution in the mouse genome, active subtypes, and potential roles as species-specific mediators of susceptibility to cancer. Mol Carcinog. 2010; 49, 5467.Google Scholar
27. Faulk, C, Barks, A, Dolinoy, DC. Phylogenetic and DNA methylation analysis reveal novel regions of variable methylation in the mouse IAP class of transposons. BMC Genomics. 2013; 14, 48.Google Scholar
28. Dolinoy, DC. The agouti mouse model: an epigenetic biosensor for nutritional and environmental alterations on the fetal epigenome. Nutr Rev. 2008; 66(Suppl 1), S7–11.Google Scholar
29. Dolinoy, DC, Huang, D, Jirtle, RL. Maternal nutrient supplementation counteracts bisphenol A-induced DNA hypomethylation in early development. Proc Natl Acad Sci U S A. 2007; 104, 1305613061.Google Scholar
30. Dolinoy, DC, Jirtle, RL. Environmental epigenomics in human health and disease. Environ Mol Mutagen. 2008; 49, 48.Google Scholar
31. Dolinoy, DC, Weinhouse, C, Jones, TR, Rozek, LS, Jirtle, RL. Variable histone modifications at the A(vy) metastable epiallele. Epigenetics. 2010; 5, 637644.Google Scholar
32. Waterland, RA, Jirtle, RL. Transposable elements: targets for early nutritional effects on epigenetic gene regulation. Mol Cell Biol. 2003; 23, 52935300.Google Scholar
33. Weinhouse, C, Anderson, OS, Bergin, IL, et al. Dose-dependent incidence of hepatic tumors in adult mice following perinatal exposure to bisphenol A. Environ Health Perspect. 2014; 122, 485491.Google Scholar
34. Miltenberger, RJ, Mynatt, RL, Wilkinson, JE, Woychik, RP. The role of the agouti gene in the yellow obese syndrome. J Nutr. 1997; 127, 1902S1907S.Google Scholar
35. Vandenberg, LN, Welshons, WV, Vom Saal, FS, Toutain, P-L, Myers, JP. Should oral gavage be abandoned in toxicity testing of endocrine disruptors? Environ Health. 2014; 13, 46.Google Scholar
36. Schmidt, J-S, Schaedlich, K, Fiandanese, N, Pocar, P, Fischer, B. Effects of di(2-ethylhexyl) phthalate (DEHP) on female fertility and adipogenesis in C3H/N mice. Environ Health Perspect. 2012; 120, 11231129.Google Scholar
37. Mylchreest, E, Wallace, DG, Cattley, RC, Foster, PM. Dose-dependent alterations in androgen-regulated male reproductive development in rats exposed to Di(n-butyl) phthalate during late gestation. Toxicol Sci. 2000; 55, 143151.Google Scholar
38. de Jesus, MM, Negrin, AC, Taboga, SR, Pinto-Fochi, ME, Góes, RM. Histopathological alterations in the prostates of Mongolian gerbils exposed to a high-fat diet and di-n-butyl phthalate individually or in combination. Reprod Toxicol. 2015; 52, 2639.Google Scholar
39. Hannas, BR, Lambright, CS, Furr, J, et al. Dose-response assessment of fetal testosterone production and gene expression levels in rat testes following in utero exposure to diethylhexyl phthalate, diisobutyl phthalate, diisoheptyl phthalate, and diisononyl phthalate. Toxicol Sci. 2011; 123, 206216.Google Scholar
40. Calafat, AM, Brock, JW, Silva, MJ, et al. Urinary and amniotic fluid levels of phthalate monoesters in rats after the oral administration of di(2-ethylhexyl) phthalate and di-n-butyl phthalate. Toxicology. 2006; 217, 2230.Google Scholar
41. Lorber, M, Calafat, AM. Dose reconstruction of di(2-ethylhexyl) phthalate using a simple pharmacokinetic model. Environ Health Perspect. 2012; 120, 17051710.Google Scholar
42. Wittassek, M, Angerer, J, Kolossa-Gehring, M, et al. Fetal exposure to phthalates – a pilot study. Int J Hyg Environ Health. 2009; 212, 492498.Google Scholar
43. Wittassek, M, Angerer, J. Phthalates: metabolism and exposure. Int J Androl. 2008; 31, 131138.Google Scholar
44. Chang, J-W, Lee, C-C, Pan, W-H, et al. Estimated daily intake and cumulative risk assessment of phthalates in the general Taiwanese after the 2011 DEHP food scandal. Sci Rep. 2017; 7, 45009.Google Scholar
45. Silva, MJ, Reidy, JA, Herbert, AR, et al. Detection of phthalate metabolites in human amniotic fluid. Bull Environ Contam Toxicol. 2004; 72, 12261231.Google Scholar
46. Huang, P-C, Tsai, C-H, Liang, W-Y, et al. Early phthalates exposure in pregnant women is associated with alteration of thyroid hormones. PLoS One. 2016; 11, e0159398.Google Scholar
47. Huang, P, Kuo, P, Chou, Y, Lin, S, Lee, C. Association between prenatal exposure to phthalates and the health of newborns. Environ Int. 2009; 35, 1420.Google Scholar
48. Grunau, C, Clark, SJ, Rosenthal, A. Bisulfite genomic sequencing: systematic investigation of critical experimental parameters. Nucleic Acids Res. 2001; 29, E65E65.Google Scholar
49. Waterman, SJ, Keller, LH, Trimmer, GW, et al. Two-generation reproduction study in rats given di-isononyl phthalate in the diet. Reprod Toxicol. 2000; 14, 2136.Google Scholar
50. Masutomi, N, Shibutani, M, Takagi, H, et al. Impact of dietary exposure to methoxychlor, genistein, or diisononyl phthalate during the perinatal period on the development of the rat endocrine/reproductive systems in later life. Toxicology. 2003; 192, 149170.Google Scholar
51. Vandenberg, LN, Colborn, T, Hayes, TB, et al. Hormones and endocrine-disrupting chemicals: low-dose effects and nonmonotonic dose responses. Endocr Rev. 2012; 33, 378455.Google Scholar
52. Pocar, P, Fiandanese, N, Secchi, C, et al. 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. 2012; 153, 937948.Google Scholar
53. Jiang, J-T, Xu, H-L, Zhu, Y-P, et al. Reduced Fgf10/Fgfr2 and androgen receptor (AR) in anorectal malformations male rats induced by di- n -butyl phthalate (DBP): A study on the local and systemic toxicology of DBP. Toxicology. 2015; 338, 7785.Google Scholar
54. Okayama, Y, Wakui, S, Wempe, MF, et al. In utero exposure to Di(n -butyl)phthalate induces morphological and biochemical changes in rats postpuberty. Toxicol Pathol. 2017; 45, 526535.Google Scholar
55. Oshida, K, Vasani, N, Thomas, RS, et al. Identification of modulators of the nuclear receptor peroxisome proliferator-activated receptor α (PPARα) in a mouse liver gene expression compendium. PLoS One. 2015; 10, e0112655.Google Scholar
56. Sarath Josh, MK, Pradeep, S, Vijayalekshmi Amma, KS, et al. Phthalates efficiently bind to human peroxisome proliferator activated receptor and retinoid X receptor α, β, γ subtypes: an in silico approach. J Appl Toxicol. 2014; 34, 754765.Google Scholar
57. Hayashi, Y, Ito, Y, Yamagishi, N, et al. Hepatic peroxisome proliferator-activated receptor α may have an important role in the toxic effects of di(2-ethylhexyl)phthalate on offspring of mice. Toxicology. 2011; 289, 110.Google Scholar
58. Laughter, AR, Dunn, CS, Swanson, CL, et al. Role of the peroxisome proliferator-activated receptor α (PPARα) in responses to trichloroethylene and metabolites, trichloroacetate and dichloroacetate in mouse liver. Toxicology. 2004; 203, 8398.Google Scholar
59. Wassenaar, PNH, Legler, J. Systematic review and meta-analysis of early life exposure to di(2-ethylhexyl) phthalate and obesity related outcomes in rodents. Chemosphere. 2017; 188, 174181.Google Scholar
60. Anderson, OS, Nahar, MS, Faulk, C, et al. Epigenetic responses following maternal dietary exposure to physiologically relevant levels of bisphenol A. Environ Mol Mutagen. 2012; 53, 334342.Google Scholar
61. Faulk, C, Barks, A, Liu, K, Goodrich, JM, Dolinoy, DC. Early-life lead exposure results in dose- and sex-specific effects on weight and epigenetic gene regulation in weanling mice. Epigenomics. 2013; 5, 487500.Google Scholar
62. Breton, CV, Marsit, CJ, Faustman, E, et al. Small-magnitude effect sizes in epigenetic end points are important in children’s environmental health studies: the Children’s Environmental Health and Disease Prevention Research Center’s Epigenetics Working Group. Environ Health Perspect. 2017; 125, 511526.Google Scholar
63. Montrose, L, Faulk, C, Francis, J, Dolinoy, DC. Perinatal lead (Pb) exposure results in sex and tissue-dependent adult DNA methylation alterations in murine IAP transposons. Environ Mol Mutagen. 2017; 58, 540550.Google Scholar
64. National Academies of Sciences Engineering and Medicine. Application of systematic review methods in an overall strategy for evaluating low-dose toxicity from endocrine active chemicals. 2017. The National Academies Press: Washington, DC.Google Scholar
Supplementary material: File

Neier et al. supplementary material

Neier et al. supplementary material 1

Download Neier et al. supplementary material(File)
File 15.5 KB
Supplementary material: Image

Neier et al. supplementary material

Neier et al. supplementary material 2

Download Neier et al. supplementary material(Image)
Image 784.5 KB
Supplementary material: Image

Neier et al. supplementary material

Neier et al. supplementary material 3

Download Neier et al. supplementary material(Image)
Image 537.4 KB
Supplementary material: Image

Neier et al. supplementary material

Neier et al. supplementary material 4

Download Neier et al. supplementary material(Image)
Image 2.6 MB
Supplementary material: Image

Neier et al. supplementary material

Neier et al. supplementary material 5

Download Neier et al. supplementary material(Image)
Image 520.6 KB
Supplementary material: File

Neier et al. supplementary material

Neier et al. supplementary material 6

Download Neier et al. supplementary material(File)
File 14 KB
Supplementary material: File

Neier et al. supplementary material

Neier et al. supplementary material 7

Download Neier et al. supplementary material(File)
File 46.1 KB