University of Illinois at Chicago
Browse
42.full.pdf (1.35 MB)

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 RatProstate Gland Throughout Life

Download (1.35 MB)
journal contribution
posted on 2013-11-22, 00:00 authored by Wan-yee Tang, Lisa M. Morey, Yuk Yin Cheung, Lynn Birch, Gail S. Prins, Shuk-mei Ho
Evidence supporting an early origin of prostate cancer is growing. We demonstrated previously that brief exposure of neonatal rats to estradiol or bisphenol A elevated their risk of developing precancerous lesions in the prostate upon androgen-supported treatment with estradiol as adults. Epigenetic reprogramming may be a mechanism underlying this inductive event in early life, because we observed overexpression of phosphodiesterase 4D variant 4 (Pde4d4) through induction of hypomethylation of its promoter. This epigenetic mark was invisible in early life (postnatal d 10), becoming apparent only after sexual maturation. Here, we asked whether other estrogen-reprogrammable epigenetic marks have similar or different patterns in gene methylation changes throughout life. We found that hypomethylation of the promoter of nucleosome binding protein-1 (Nsbp1), unlike Pde4d4, is an early and permanent epigenetic mark of neonatal exposure to estradiol/bisphenol A that persists throughout life, unaffected by events during adulthood. In contrast, hippocalcin-like 1 (Hpcal1) is a highly plastic epigenetic mark whose hypermethylation depends on both type of early-life exposure and adult-life events. Four of the eight genes involved in DNA methylation/demethylation showed early and persistent overexpression that was not a function of DNA methylation at their promoters, including genes encoding de novo DNA methyltransferases (Dnmt3a/b) and methyl-CpG binding domain proteins (Mbd2/4) that have demethylating activities. Their lifelong aberrant expression implicates them in early-life reprogramming and prostate carcinogenesis during adulthood. We speculate that the distinctly different fate of early-life epigenetic marks during adulthood reflects the complex nature of lifelong editing of early-life epigenetic reprogramming.

Funding

This work is in part supported by National Institutes of Health Grants CA136023 (to G.S.P.), DK40890 (to G.S.P.), ES015584 (to G.S.P. and S.M.H.), ES018758 (to G.S.P. and S.M.H.), CA112532 (to S.M.H.), ES018789 (to S.M.H.), ES006096 (to S.M.H.), ES019480 (to S.M.H.), ES020988 (to S.M.H.), and ES016817 (to W.-y.T.) and by the Department of Veterans Affairs Award BX000675 (to S.M.H.).

History

Publisher Statement

Post print version of article may differ from published version. The definitive version is available through Endocrine Society at DOI: 10.1210/en.2011-1308.

Publisher

Endocrine Society

Language

  • en_US

issn

0013-7227

Issue date

2012-01-01

Usage metrics

    Categories

    No categories selected

    Keywords

    Exports

    RefWorks
    BibTeX
    Ref. manager
    Endnote
    DataCite
    NLM
    DC