We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research Article

Early-life lead exposure results in dose- and sex-specific effects on weight and epigenetic gene regulation in weanling mice

    Christopher Faulk

    Department of Environmental Health Sciences, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109-2200, USA

    ,
    Amanda Barks

    Department of Environmental Health Sciences, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109-2200, USA

    ,
    Kevin Liu

    Department of Environmental Health Sciences, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109-2200, USA

    ,
    Jaclyn M Goodrich

    Department of Environmental Health Sciences, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109-2200, USA

    &
    Dana C Dolinoy

    * Author for correspondence

    Department of Environmental Health Sciences, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109-2200, USA.

    Published Online:https://doi.org/10.2217/epi.13.49

    Aims: Epidemiological and animal data suggest that the development of adult chronic conditions is influenced by early-life exposure-induced changes to the epigenome. This study investigates the effects of perinatal lead (Pb) exposure on DNA methylation and bodyweight in weanling mice. Materials & methods: Viable yellow agouti (Avy) mouse dams were exposed to 0, 2.1, 16 and 32 ppm Pb acetate before conception through weaning. Epigenetic effects were evaluated by scoring coat color of Avy/a offspring and quantitative bisulfite sequencing of two retrotransposon-driven (Avy and CDK5 activator-binding protein intracisternal A particle element) and two imprinted (Igf2 and Igf2r) loci in tail DNA. Results: Maternal blood Pb levels were below the limit of detection in controls, and 4.1, 25.1 and 32.1 µg/dl for each dose, respectively. Pb exposure was associated with a trend of increased wean bodyweight in males (p = 0.03) and altered coat color in Avy/a offspring. DNA methylation at Avy and the CDK5 activator-binding protein intracisternal A-particle element was significantly different from controls following a cubic trend (p = 0.04; p = 0.01), with male-specific effects at the Avy locus. Imprinted genes did not shift in methylation across exposures. Conclusion: Dose- and sex-specific responses in bodyweight and DNA methylation indicate that Pb acts on the epigenome in a locus-specific fashion, dependent on the genomic feature hosting the CpG site of interest, and that sex is a factor in epigenetic response.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    References

    • Barker D. Intrauterine programming of coronary heart disease and stroke. Acta Paediatra Suppl.423,178–182 (1997).
    • Barker D. Programming the baby. In: Mothers, Babies, and Disease in Later Life. Barker D (Ed.). BMJ Publishing, London, UK (1994).▪ Developmental origins of adult disease are clearly presented and human health implications laid forth.
    • Hu H, Tellez-Rojo MM, Bellinger D et al. Fetal lead exposure at each stage of pregnancy as a predictor of infant mental development. Environ. Health Perspect.114(11),1730–1735 (2006).
    • Bellinger DC. Teratogen update: lead and pregnancy. Birth Defects Res. A Clin. Mol. Teratol.73(6),409–420 (2005).
    • Lanphear BP, Hornung R, Khoury J et al. Low-level environmental lead exposure and children’s intellectual function: an international pooled analysis. Environ. Health Perspect.113(7),894–899 (2005).
    • Navas-Acien A, Guallar E, Silbergeld EK, Rothenberg SJ. Lead exposure and cardiovascular disease – a systematic review. Environ. Health Perspect.115(3),472–482 (2007).
    • Wong GP, Ng TL, Martin TR, Farquharson DF. Effects of low-level lead exposure in utero.Obstet. Gynecol. Surv.47(5),285–289 (1992).
    • Agency for Toxic Substances and Disease Registry. Toxicological Profile for Lead. Agency for Toxic Substances and Disease Registry, GA, USA (2007).
    • Gilbert SG, Weiss B. A rationale for lowering the blood lead action level from 10 to 2 microg/dl. Neurotoxicology27(5),693–701 (2006).
    • 10  Snyder JE, Filipov NM, Parsons PJ, Lawrence DA. The efficiency of maternal transfer of lead and its influence on plasma IgE and splenic cellularity of mice. Toxicol. Sci.57(1),87–94 (2000).
    • 11  Leasure JL, Giddabasappa A, Chaney S et al. Low-level human equivalent gestational lead exposure produces sex-specific motor and coordination abnormalities and late-onset obesity in year-old mice. Environ. Health Perspect.116(3),355–361 (2008).▪▪ Notable for its careful study of low levels of gestational lead exposure in mice and the effects in male and female offspring at 1 year of life.
    • 12  Iavicoli I, Carelli G, Stanek EJ, Castellino N, Li Z, Calabrese EJ. Low doses of dietary lead are associated with a profound reduction in the time to the onset of puberty in female mice. Reprod. Toxicol.22(4),586–590 (2006).
    • 13  Faulk C, Dolinoy DC. Timing is everything: the when and how of environmentally induced changes in the epigenome of animals. Epigenetics6(7),791–797 (2011).
    • 14  Lim U, Song MA. Dietary and lifestyle factors of DNA methylation. Methods Mol. Biol.863,359–376 (2012).
    • 15  Anderson OS, Nahar MS, Faulk C et al. Epigenetic responses following maternal dietary exposure to physiologically relevant levels of bisphenol A. Environ. Mol. Mutagen.53(5),334–342 (2012).
    • 16  Koedrith P, Kim H, Weon JI, Seo YR. Toxicogenomic approaches for understanding molecular mechanisms of heavy metal mutagenicity and carcinogenicity. Int. J. Hyg. Environ. Health216(5),587–598 (2013).
    • 17  Dolinoy DC. The agouti mouse model: an epigenetic biosensor for nutritional and environmental alterations on the fetal epigenome. Nutr. Rev.66(Suppl. 1),S7–S11 (2008).
    • 18  Waterland RA, Dolinoy DC, Lin JR, Smith CA, Shi X, Tahiliani KG. Maternal methyl supplements increase offspring DNA methylation at axin fused. Genesis44(9),401–406 (2006).
    • 19  Dolinoy DC, Weidman JR, Waterland RA, Jirtle RL. Maternal genistein alters coat color and protects avy mouse offspring from obesity by modifying the fetal epigenome. Environ. Health Perspect.114(4),567–572 (2006).
    • 20  Rakyan VK, Blewitt ME, Druker R, Preis JI, Whitelaw E. Metastable epialleles in mammals. Trends Genet.18(7),348–351 (2002).
    • 21  Hackett JA, Surani MA. DNA methylation dynamics during the mammalian life cycle. Philos. Trans. R Soc. Lond. B Biol. Sci.368(1609),20110328 (2013).
    • 22  Downing C, Johnson TE, Larson C et al. Subtle decreases in DNA methylation and gene expression at the mouse Igf2 locus following prenatal alcohol exposure: effects of a methyl-supplemented diet. Alcohol45(1),65–71 (2011).
    • 23  McKay JA, Williams EA, Mathers JC. Effect of maternal and post-weaning folate supply on gene-specific DNA methylation in the small intestine of weaning and adult apc and wild type mice. Front. Genet.2,23 (2011).
    • 24  Ding GL, Wang FF, Shu J et al. Transgenerational glucose intolerance with Igf2/H19 epigenetic alterations in mouse islet induced by intrauterine hyperglycemia. Diabetes61(5),1133–1142 (2012).
    • 25  Zhang XF, Zhang LJ, Feng YN et al. Bisphenol A exposure modifies DNA methylation of imprint genes in mouse fetal germ cells. Mol. Biol. Rep.39(9),8621–8628 (2012).
    • 26  Somm E, Stouder C, Paoloni-Giacobino A. Effect of developmental dioxin exposure on methylation and expression of specific imprinted genes in mice. Reprod. Toxicol.35,150–155 (2013).
    • 27  Susiarjo M, Sasson I, Mesaros C, Bartolomei MS. Bisphenol A exposure disrupts genomic imprinting in the mouse. PLoS Genet.9(4),e1003401 (2013).
    • 28  Gallou-Kabani C, Gabory A, Tost J et al. Sex- and diet-specific changes of imprinted gene expression and DNA methylation in mouse placenta under a high-fat diet. PLoS One5(12),e14398 (2010).
    • 29  Waterland RA, Jirtle RL. Transposable elements: targets for early nutritional effects on epigenetic gene regulation. Mol. Cell Biol.23(15),5293–5300 (2003).
    • 30  Green MR, Sambrook J. Molecular Cloning: A Laboratory Manual (4th Edition). Cold Spring Harbor Laboratory Press, NY, USA (2012).
    • 31  Grunau C, Clark SJ, Rosenthal A. Bisulfite genomic sequencing: systematic investigation of critical experimental parameters. Nucleic Acids Res.29(13),E65–E65 (2001).
    • 32  Druker R. Complex patterns of transcription at the insertion site of a retrotransposon in the mouse. Nucleic Acids Res.32(19),5800–5808 (2004).
    • 33  Waterland RA, Lin JR, Smith CA, Jirtle RL. Post-weaning diet affects genomic imprinting at the insulin-like growth factor 2 (Igf2) locus. Hum. Mol. Genet.15(5),705–716 (2006).
    • 34  Fauque P, Ripoche MA, Tost J et al. Modulation of imprinted gene network in placenta results in normal development of in vitro manipulated mouse embryos. Hum. Mol. Genet.19(9),1779–1790 (2010).
    • 35  Miltenberger RJ, Mynatt RL, Wilkinson JE, Woychik RP. The role of the agouti gene in the yellow obese syndrome. J. Nutr.127(9),1902S–1907S (1997).
    • 36  Rossi-George A, Virgolini MB, Weston D, Cory-Slechta DA. Alterations in glucocorticoid negative feedback following maternal Pb, prenatal stress and the combination: a potential biological unifying mechanism for their corresponding disease profiles. Toxicol. Appl. Pharmacol.234(1),117–127 (2009).
    • 37  Ronis MJ, Badger TM, Shema SJ et al. Endocrine mechanisms underlying the growth effects of developmental lead exposure in the rat. J. Toxicol. Environ. Health A54(2),101–120 (1998).
    • 38  Pine M, Hiney J, Dearth R, Bratton J, Les Dees WL. IGF-1 administration to prepubertal female rats can overcome delayed puberty caused by maternal Pb exposure. Reprod. Toxicol.21,104–109 (2006).
    • 39  Dearth RK, Hiney JK, Srivastava V, Burdick SB, Bratton GR, Dees WL. Effects of lead (Pb) exposure during gestation and lactation on female pubertal development in the rat. Reprod. Toxicol.16(4),343–352 (2002).
    • 40  McGivern RF, Sokol RZ, Berman NG. Prenatal lead exposure in the rat during the third week of gestation: long-term behavioral, physiological, and anatomical effects associated with reproduction. Toxicol. Appl. Pharmacol.110(2),206–215 (1991).
    • 41  Petrusz P, Weaver CM, Grant LD, Mushak P, Krigman MR. Lead poisoning and reproduction: effects on pituitary and serum gonadotropins in neonatal rats. Environ. Res.19(2),383–391 (1979).
    • 42  Gorbel F, Boujelbene M, Makni-Ayadi F et al. Cytotoxic effects of lead on the endocrine and exocrine sexual function of pubescent male and female rats. Demonstration of apoptotic activity. C R Biol.325(9),927–940 (2002).
    • 43  Ronis MJ, Gandy J, Badger T. Endocrine mechanisms underlying reproductive toxicity in the developing rat chronically exposed to dietary lead. J. Toxicol. Environ. Health A54(2),77–99 (1998).
    • 44  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 Genomics14(1),48 (2013).
    • 45  Bernal AJ, Dolinoy DC, Huang D, Skaar DA, Weinhouse C, Jirtle RL. Adaptive radiation-induced epigenetic alterations mitigated by antioxidants. FASEB J.27(2),665–671 (2013).▪ Hormetic response was observed following low-dose gestational radiation exposure, correlating with increased DNA methylation.
    • 46  Li G, Kohorst JJ, Zhang W et al. Early postnatal nutrition determines adult physical activity and energy expenditure in female mice. Diabetes62(8),2773–2783 (2013).
    • 47  Patel BB, Raad M, Sebag IA, Chalifour LE. Lifelong exposure to bisphenol A alters cardiac structure/function, protein expression, and DNA methylation in adult mice. Toxicol. Sci.133(1),174–185 (2013).
    • 48  Sterrenburg L, Gaszner B, Boerrigter J et al. Chronic stress induces sex-specific alterations in methylation and expression of corticotropin-releasing factor gene in the rat. PLoS One6(11),e28128 (2011).
    • 49  Schneider JS, Kidd S, Anderson DW. Influence of developmental lead exposure on expression of DNA methyltransferases and methyl cytosine-binding proteins in hippocampus. Toxicol. Lett.217(1),75–81 (2012).
    • 50  Thompson SL, Konfortova G, Gregory RI, Reik W, Dean W, Feil R. Environmental effects on genomic imprinting in mammals. Toxicol. Lett.120(1–3),143–150 (2001).
    • 51  Heijmans BT, Tobi EW, Stein AD et al. Persistent epigenetic differences associated with prenatal exposure to famine in humans. Proc. Natl Acad. Sci. USA105(44),17046–17049 (2008).
    • 52  Roberts CT, Owens JA, Sferruzzi-Perri AN. Distinct actions of insulin-like growth factors (IGFs) on placental development and fetal growth: lessons from mice and guinea pigs. Placenta29(Suppl. A),S42–S47 (2008).
    • 53  Li YY, Chen T, Wan Y, Xu SQ. Lead exposure in pheochromocytoma cells induces persistent changes in amyloid precursor protein gene methylation patterns. Environ. Toxicol.27(8),495–502 (2012).▪▪ Gene-specific and global DNA hypomethylation were observed in this study, and may be associated with lead-induced neurodegeneration.
    • 54  Park SK, Elmarsafawy S, Mukherjee B et al. Cumulative lead exposure and age-related hearing loss: the VA normative aging study. Hear. Res269(1–2),48–55 (2010).
    • 55  Schwartz BS, Hu H. Adult lead exposure: time for change. Environ. Health Perspect.115(3),451–454 (2007).
    • 56  Tokar EJ, Diwan BA, Waalkes MP. Early life inorganic lead exposure induces testicular teratoma and renal and urinary bladder preneoplasia in adult metallothionein-knockout mice but not in wild type mice. Toxicology276(1),5–10 (2010).
    • 57  Gonzalez-Cossio T, Peterson KE, Sanin LH et al. Decrease in birth weight in relation to maternal bone-lead burden. Pediatrics100(5),856–862 (1997).
    • 58  Lee MG, Chun OK, Song WO. Determinants of the blood lead level of US women of reproductive age. J. Am. Coll. Nutr.24(1),1–9 (2005).
    • 59  Bernard SM, McGeehin MA. Prevalence of blood lead levels > or = 5 micro g/dl among US children 1 to 5 years of age and socioeconomic and demographic factors associated with blood of lead levels 5 to 10 micro g/dl, Third National Health and Nutrition Examination Survey, 1988–1994. Pediatrics112(6 Pt 1),1308–1313 (2003).
    • 60  Kaminen-Ahola N, Ahola A, Flatscher-Bader T et al. Postnatal growth restriction and gene expression changes in a mouse model of fetal alcohol syndrome. Birth Defects Res. Part A Clin. Mol. Teratol.88(10),818–826 (2010).
    • 61  Dolinoy DC, Weidman JR, Waterland RA, Jirtle RL. Maternal genistein alters coat color and protects Avy mouse offspring from obesity by modifying the fetal epigenome. Environ. Health Perspect.114(4),567–572 (2006).