Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters











Database
Language
Publication year range
1.
FASEB J ; 33(2): 2719-2731, 2019 02.
Article in English | MEDLINE | ID: mdl-30303739

ABSTRACT

Remodeling of the gut microbiota is implicated in various metabolic and inflammatory diseases of the gastrointestinal tract. We hypothesized that the gut microbiota affects the DNA methylation profile of intestinal epithelial cells (IECs) which could, in turn, alter intestinal function. In this study, we used mass spectrometry and methylated DNA capture to respectively investigate global and genome-wide DNA methylation of intestinal epithelial cells from germ-free (GF) and conventionally raised mice. In colonic IECs from GF mice, DNA was markedly hypermethylated. This was associated with a dramatic loss of ten-eleven-translocation activity, a lower DNA methyltransferase activity and lower circulating levels of the 1-carbon metabolite, folate. At the gene level, we found an enrichment for differentially methylated regions proximal to genes regulating the cytotoxicity of NK cells (false-discovery rate < 8.9E-6), notably genes regulating the cross-talk between NK cells and target cells, such as members of the NK group 2 member D ligand superfamily Raet. This distinct epigenetic signature was associated with a marked decrease in Raet1 expression and a loss of CD56+/CD45+ cells in the intestine of GF mice. Thus, our results indicate that altered activity of methylation-modifying enzymes in GF mice influences the IEC epigenome and modulates the crosstalk between IECs and NK cells. Epigenetic reprogramming of IECs may modulate intestinal function in diseases associated with altered gut microbiota.-Poupeau, A., Garde, C., Sulek, K., Citirikkaya, K., Treebak, J. T., Arumugam, M., Simar, D., Olofsson, L. E., Bäckhed, F., Barrès, R. Genes controlling the activation of natural killer lymphocytes are epigenetically remodeled in intestinal cells from germ-free mice.


Subject(s)
Biomarkers/analysis , Epigenesis, Genetic , Epithelial Cells/immunology , Gastrointestinal Microbiome , Gene Expression Regulation , Germ-Free Life , Killer Cells, Natural/immunology , Animals , DNA Methylation , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Female , Intestines/cytology , Intestines/microbiology , Intestines/physiology , Killer Cells, Natural/metabolism , Killer Cells, Natural/microbiology , Male , Mice
2.
Diabetes ; 65(12): 3573-3584, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27650856

ABSTRACT

Exposure to ionizing radiation increases the risk of chronic metabolic disorders such as insulin resistance and type 2 diabetes later in life. We hypothesized that irradiation reprograms the epigenome of metabolic progenitor cells, which could account for impaired metabolism after cancer treatment. C57Bl/6 mice were treated with a single dose of irradiation and subjected to high-fat diet (HFD). RNA sequencing and reduced representation bisulfite sequencing were used to create transcriptomic and epigenomic profiles of preadipocytes and skeletal muscle satellite cells collected from irradiated mice. Mice subjected to total body irradiation showed alterations in glucose metabolism and, when challenged with HFD, marked hyperinsulinemia. Insulin signaling was chronically disrupted in skeletal muscle and adipose progenitor cells collected from irradiated mice and differentiated in culture. Epigenomic profiling of skeletal muscle and adipose progenitor cells from irradiated animals revealed substantial DNA methylation changes, notably for genes regulating the cell cycle, glucose/lipid metabolism, and expression of epigenetic modifiers. Our results show that total body irradiation alters intracellular signaling and epigenetic pathways regulating cell proliferation and differentiation of skeletal muscle and adipose progenitor cells and provide a possible mechanism by which irradiation used in cancer treatment increases the risk for metabolic disease later in life.


Subject(s)
Diet, High-Fat/adverse effects , Muscle, Skeletal/metabolism , Radiation, Ionizing , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Body Weight/drug effects , Body Weight/radiation effects , Cells, Cultured , Computational Biology , Epigenomics , Immunoblotting , Insulin Resistance/radiation effects , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , Reactive Oxygen Species , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA
SELECTION OF CITATIONS
SEARCH DETAIL