Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Gene Expr Patterns ; 28: 1-11, 2018 06.
Article in English | MEDLINE | ID: mdl-29339137

ABSTRACT

Embryo culture and assisted reproductive technologies have been associated with a disproportionately high number of epigenetic abnormalities in the resulting offspring. However, the mechanisms by which these techniques influence the epigenome remain poorly defined. In this study, we evaluated the capacity of oxygen concentration to influence the transcriptional control of a selection of key enzymes regulating chromatin structure. In mouse embryonic stem cells, oxygen concentrations modulated the transcriptional regulation of the TET family of enzymes, as well as the de novo methyltransferase Dnmt3a. These transcriptional changes were associated with alterations in the control of multiple imprinted genes, including H19, Igf2, Igf2r, and Peg3. Similarly, exposure of in vitro produced bovine embryos to atmospheric oxygen concentrations was associated with disruptions in the transcriptional regulation of TET1, TET3, and DNMT3a, along with the DNA methyltransferase co-factor HELLS. In addition, exposure to high oxygen was associated with alterations in the abundance of transcripts encoding members of the Polycomb repressor complex (EED and EZH2), the histone methyltransferase SETDB1 and multiple histone demethylases (KDM1A, KDM4B, and KDM4C). These disruptions were accompanied by a reduction in embryo viability and suppression of the pluripotency genes NANOG and SOX2. These experiments demonstrate that oxygen has the capacity to modulate the transcriptional control of chromatin modifying genes involved in the establishment and maintenance of both pluripotency and genomic imprinting.


Subject(s)
Biomarkers/metabolism , Chromatin/metabolism , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Genomic Imprinting , Oxygen/metabolism , Animals , Cattle , Cells, Cultured , Chromatin/chemistry , Chromatin/genetics , Chromatin Assembly and Disassembly , DNA Methylation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Stem Cells/cytology , Epigenesis, Genetic , Gene Expression Profiling , Mice , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Epigenetics ; 12(10): 841-853, 2017.
Article in English | MEDLINE | ID: mdl-28816587

ABSTRACT

The preconception environment is a significant modifier of dysgenesis and the development of environmentally-induced disease. To date, fetal alcohol spectrum disorders (FASDs) have been exclusively associated with maternal exposures, yet emerging evidence suggests male-inherited alterations in the developmental program of sperm may be relevant to the growth-restriction phenotypes of this condition. Using a mouse model of voluntary consumption, we find chronic preconception male ethanol exposure associates with fetal growth restriction, decreased placental efficiency, abnormalities in cholesterol trafficking, sex-specific alterations in the genetic pathways regulating hepatic fibrosis, and disruptions in the regulation of imprinted genes. Alterations in the DNA methylation profiles of imprinted loci have been identified in clinical studies of alcoholic sperm, suggesting the legacy of paternal drinking may transmit via heritable disruptions in the regulation of imprinted genes. However, the capacity of sperm-inherited changes in DNA methylation to broadly transmit environmentally-induced phenotypes remains unconfirmed. Using bisulphite mutagenesis and second-generation deep sequencing, we find no evidence to suggest that these phenotypes or any of the associated transcriptional changes are linked to alterations in the sperm-inherited DNA methylation profile. These observations are consistent with recent studies examining the male transmission of diet-induced phenotypes and emphasize the importance of epigenetic mechanisms of paternal inheritance beyond DNA methylation. This study challenges the singular importance of maternal alcohol exposures and suggests paternal alcohol abuse is a significant, yet overlooked epidemiological factor complicit in the genesis of alcohol-induced growth defects, and may provide mechanistic insight into the failure of FASD children to thrive postnatally.


Subject(s)
DNA Methylation/genetics , Fetal Alcohol Spectrum Disorders/genetics , Fetal Growth Retardation/genetics , Prenatal Exposure Delayed Effects/genetics , Alcohol Drinking/adverse effects , Animals , Child , DNA Methylation/drug effects , Disease Models, Animal , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/genetics , Ethanol/toxicity , Female , Fetal Alcohol Spectrum Disorders/pathology , Fetal Growth Retardation/chemically induced , Fetal Growth Retardation/pathology , Humans , Male , Mice , Phenotype , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/pathology , Spermatozoa/drug effects , Spermatozoa/pathology
3.
Alcohol ; 60: 121-133, 2017 05.
Article in English | MEDLINE | ID: mdl-28433419

ABSTRACT

Alterations to chromatin structure induced by environmental insults have become an attractive explanation for the persistence of exposure effects into subsequent life stages. However, a growing body of work examining the epigenetic impact that alcohol and other drugs of abuse exert consistently notes a disconnection between induced changes in chromatin structure and patterns of gene transcription. Thus, an important question is whether perturbations in the 'histone code' induced by prenatal exposures to alcohol implicitly subvert gene expression, or whether the hierarchy of cellular signaling networks driving development is such that they retain control over the transcriptional program. To address this question, we examined the impact of ethanol exposure in mouse embryonic stem cells cultured under 2i conditions, where the transcriptional program is rigidly enforced through the use of small molecule inhibitors. We find that ethanol-induced changes in post-translational histone modifications are dose-dependent, unique to the chromatin modification under investigation, and that the extent and direction of the change differ between the period of exposure and the recovery phase. Similar to in vivo models, we find post-translational modifications affecting histone 3 lysine 9 are the most profoundly impacted, with the signature of exposure persisting long after alcohol has been removed. These changes in chromatin structure associate with dose-dependent alterations in the levels of transcripts encoding Dnmt1, Uhrf1, Tet1, Tet2, Tet3, and Polycomb complex members Eed and Ezh2. However, in this model, ethanol-induced changes to the chromatin template do not consistently associate with changes in gene transcription, impede the process of differentiation, or affect the acquisition of monoallelic patterns of expression for the imprinted gene Igf2R. These findings question the inferred universal relevance of epigenetic changes induced by drugs of abuse and suggest that changes in chromatin structure cannot unequivocally explain dysgenesis in isolation.


Subject(s)
Chromatin Assembly and Disassembly/drug effects , Ethanol/toxicity , Gene Expression Regulation, Developmental/drug effects , Mouse Embryonic Stem Cells/drug effects , Transcription, Genetic/drug effects , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Female , Fetal Alcohol Spectrum Disorders/genetics , Fetal Alcohol Spectrum Disorders/metabolism , Fetal Alcohol Spectrum Disorders/pathology , Genomic Imprinting/drug effects , Histones/genetics , Histones/metabolism , Lysine , Mice , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/pathology , Nucleic Acid Conformation , Pregnancy , Prenatal Exposure Delayed Effects , Protein Conformation , Structure-Activity Relationship , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...