10 research outputs found

    Developmental exposures to common environmental contaminants, DEHP and lead, alter adult brain and blood hydroxymethylation in mice

    Get PDF
    Introduction: The developing epigenome changes rapidly, potentially making it more sensitive to toxicant exposures. DNA modifications, including methylation and hydroxymethylation, are important parts of the epigenome that may be affected by environmental exposures. However, most studies do not differentiate between these two DNA modifications, possibly masking significant effects.Methods: To investigate the relationship between DNA hydroxymethylation and developmental exposure to common contaminants, a collaborative, NIEHS-sponsored consortium, TaRGET II, initiated longitudinal mouse studies of developmental exposure to human-relevant levels of the phthalate plasticizer di(2-ethylhexyl) phthalate (DEHP), and the metal lead (Pb). Exposures to 25 mg DEHP/kg of food (approximately 5 mg DEHP/kg body weight) or 32 ppm Pb-acetate in drinking water were administered to nulliparous adult female mice. Exposure began 2 weeks before breeding and continued throughout pregnancy and lactation, until offspring were 21 days old. At 5 months, perinatally exposed offspring blood and cortex tissue were collected, for a total of 25 male mice and 17 female mice (n = 5–7 per tissue and exposure). DNA was extracted and hydroxymethylation was measured using hydroxymethylated DNA immunoprecipitation sequencing (hMeDIP-seq). Differential peak and pathway analysis was conducted comparing across exposure groups, tissue types, and animal sex, using an FDR cutoff of 0.15.Results: DEHP-exposed females had two genomic regions with lower hydroxymethylation in blood and no differences in cortex hydroxymethylation. For DEHP-exposed males, ten regions in blood (six higher and four lower) and 246 regions (242 higher and four lower) and four pathways in cortex were identified. Pb-exposed females had no statistically significant differences in blood or cortex hydroxymethylation compared to controls. Pb-exposed males, however, had 385 regions (all higher) and six pathways altered in cortex, but no differential hydroxymethylation was identified in blood.Discussion: Overall, perinatal exposure to human-relevant levels of two common toxicants showed differences in adult DNA hydroxymethylation that was specific to sex, exposure type, and tissue, but male cortex was most susceptible to hydroxymethylation differences by exposure. Future assessments should focus on understanding if these findings indicate potential biomarkers of exposure or are related to functional long-term health effects

    Developmental Exposures to Phthalates and Phthalate Mixtures and Life-Course Metabolic Outcomes: Using a Mouse Model to Inform Human Studies and Elucidate Mechanisms

    Full text link
    Nearly 40 percent of US adults and 20 percent of US children are obese. Given obesity’s multiple dangerous comorbidities, this presents a significant concern for public health. A growing body of evidence suggests that exposures to environmental chemicals may be contributing to the obesity epidemic. Such chemicals have been termed “obesogens” and among them are phthalates, endocrine disrupting chemicals (EDCs) that are present in food packaging, children’s toys, and personal care products. Exposures to phthalates during development have been linked to adverse metabolic health outcomes in both animal and human studies, but findings from human studies are less consistent. One possible reason is humans are co-exposed to many phthalates, and these mixture exposures are difficult to interpret. Additionally, the vast majority of animal studies to date have focused on examining metabolic impacts of diethylhexyl phthalate (DEHP), despite the recent introduction of newer phthalates on the market to replace it, including diisononyl phthalate (DINP). Furthermore, mechanisms linking developmental exposures and later-life health outcomes, such as epigenetic reprogramming via DNA methylation, are still poorly understood. The overall objective of this dissertation was to utilize an animal model of perinatal phthalate exposures to investigate long-term metabolic impacts in a manner that would inform human studies and infer underlying mechanisms. We incorporated exposures to three individual phthalates (DEHP, DINP, and dibutyl phthalate (DBP)), as well as two phthalate mixtures (DEHP+DINP and DEHP+DINP+DBP). We then took phenotypic and molecular measurements on the offspring at two time points: at weaning on postnatal day 21 (PND21) at the end of the exposure period and at 10 months of age, >9 months after exposure had ceased. In Aim 1, we investigated early-life metabolic phenotypes by measuring body weight and relative liver weights and examined biomarkers of whole-genome DNA methylation alterations at PND21. In Aim 2, we evaluated metabolic phenotypes longitudinally at two and eight months of age to determine whether developmental exposures to phthalates influenced metabolism across the life course. Finally, in Aim 3, we measured the transcriptome and DNA methylation in liver and white adipose tissue (WAT) at both PND21 and 10 months to elucidate a molecular mechanism. We found that developmental exposures to individual phthalates and phthalate mixtures were associated with increased body weights in males and females in early postnatal life. Females, but not males, perinatally exposed to DINP-only and a mixture of DEHP+DINP also had increased relative liver weights at PND21. We also observed a sex-specific effect on tail DNA methylation at repetitive elements in mice exposed to individual phthalates and phthalate mixtures, indicating a sexually dimorphic effect on the epigenome. Developmental exposures to DEHP-only and DINP-only resulted in increased body fat percentage and glucose intolerance, respectively, across the life course. However, we did not observe longitudinal adverse metabolic impacts in mice perinatally exposed to phthalate mixtures, suggesting a potential adaptive response in these mice. In females perinatally exposed to DINP, we identified several persistently up-regulated PPAR target genes in the liver that could lead to increased fatty acid synthesis. Fatty acid synthase (Fasn) also exhibited increased promoter region DNA methylation at both PND21 and 10 months of age, implicating a role for epigenetic reprogramming. Taken together, the work here demonstrates short-term and long-term metabolic impacts following perinatal exposures to phthalates, and presents a new potential mechanism describing the underlying biology in the liver.PHDToxicologyUniversity of Michigan, Horace H. Rackham School of Graduate Studieshttps://deepblue.lib.umich.edu/bitstream/2027.42/151581/1/kneier_1.pd

    Placenta and fetal brain share a neurodevelopmental disorder DNA methylation profile in a mouse model of prenatal PCB exposure.

    No full text
    Polychlorinated biphenyls (PCBs) are developmental neurotoxicants implicated as environmental risk factors for neurodevelopmental disorders (NDDs). Here, we report the effects of prenatal exposure to a human-relevant mixture of PCBs on the DNA methylation profiles of mouse placenta and fetal brain. Thousands of differentially methylated regions (DMRs) distinguish placenta and fetal brain from PCB-exposed mice from sex-matched vehicle controls. In both placenta and fetal brain, PCB-associated DMRs are enriched for functions related to neurodevelopment and cellular signaling and enriched within regions of bivalent chromatin. The placenta and brain PCB DMRs overlap significantly and map to a shared subset of genes enriched for Wnt signaling, Slit/Robo signaling, and genes differentially expressed in NDD models. The consensus PCB DMRs also significantly overlap with DMRs from human NDD brain and placenta. These results demonstrate that PCB-exposed placenta contains a subset of DMRs that overlap fetal brain DMRs relevant to an NDD

    Tissue and sex-specific programming of DNA methylation by perinatal lead exposure: implications for environmental epigenetics studies

    No full text
    Early developmental environment can influence long-term health through reprogramming of the epigenome. Human environmental epigenetics studies rely on surrogate tissues, such as blood, to assess the effects of environment on disease-relevant but inaccessible target tissues. However, the extent to which environment-induced epigenetic changes are conserved between these tissues is unclear. A better understanding of this conservation is imperative for effective design and interpretation of human environmental epigenetics studies. The Toxicant Exposures and Responses by Genomic and Epigenomic Regulators of Transcription (TaRGET II) consortium was established by the National Institute of Environmental Health Sciences to address the utility of surrogate tissues as proxies for toxicant-induced epigenetic changes in target tissues. We and others have recently reported that perinatal exposure to lead (Pb) is associated with adverse metabolic outcomes. Here, we investigated the sex-specific effects of perinatal exposure to a human environmentally relevant level of Pb on DNA methylation in paired liver and blood samples from adult mice using enhanced reduced-representation bisulphite sequencing. Although Pb exposure ceased at 3 weeks of age, we observed thousands of sex-specific differentially methylated cytosines in the blood and liver of Pb-exposed animals at 5 months of age, including 44 genomically imprinted loci. We observed significant tissue overlap in the genes mapping to differentially methylated cytosines. A small but significant subset of Pb-altered genes exhibit basal sex differences in gene expression in the mouse liver. Collectively, these data identify potential molecular targets for Pb-induced metabolic diseases, and inform the design of more robust human environmental epigenomics studies

    Sex disparate gut microbiome and metabolome perturbations precede disease progression in a mouse model of Rett syndrome.

    No full text
    Rett syndrome (RTT) is a regressive neurodevelopmental disorder in girls, characterized by multisystem complications including gut dysbiosis and altered metabolism. While RTT is known to be caused by mutations in the X-linked gene MECP2, the intermediate molecular pathways of progressive disease phenotypes are unknown. Mecp2 deficient rodents used to model RTT pathophysiology in most prior studies have been male. Thus, we utilized a patient-relevant mouse model of RTT to longitudinally profile the gut microbiome and metabolome across disease progression in both sexes. Fecal metabolites were altered in Mecp2e1 mutant females before onset of neuromotor phenotypes and correlated with lipid deficiencies in brain, results not observed in males. Females also displayed altered gut microbial communities and an inflammatory profile that were more consistent with RTT patients than males. These findings identify new molecular pathways of RTT disease progression and demonstrate the relevance of further study in female Mecp2 animal models
    corecore