Metabolomics,Unknown,Transcriptomics,Genomics,Proteomics

Dataset Information

0

Genome-wide methylation maps for Proliferating and Senescent cells


ABSTRACT: Altered DNA methylation and associated destabilization of genome integrity and function is a hallmark of cancer. Replicative senescence imposes a limit on proliferative potential that all cancer cells must bypass. Compared to proliferating cells, senescent cells exhibit marked chromatin re-organization. Here we show by whole-genome single-nucleotide bisulfite sequencing that replicative senescent human cells exhibit widespread alterations in their DNA methylome. These changes are linked to mislocalization of the maintenance DNA methyltransferase (DNMT1) in cells approaching senescence, altered replication-coupled DNA methylation and de-repression of repetitive satellite sequences. Deficiency of DNMT1 triggers chromatin changes characteristic of senescence and expression of satellite sequences. Most importantly, but paradoxically, gains and losses of methylation in replicative senescence are similar to those in cancer, and this M-bM-^@M-^XreprogrammedM-bM-^@M-^Y methylation landscape is largely retained when cells escape or bypass senescence. In sum, altered regulation of DNMT1 in cells approaching replicative senescence contributes to changes in chromatin structure and function. Consequently, if senescent cells escape the proliferative barrier, they already harbor epigenetic changes likely to promote malignancy. Examination of methylation status in IMR90 cells

ORGANISM(S): Homo sapiens

SUBMITTER: Tony McBryan 

PROVIDER: E-GEOD-48580 | biostudies-arrayexpress |

REPOSITORIES: biostudies-arrayexpress

altmetric image

Publications


Altered DNA methylation and associated destabilization of genome integrity and function is a hallmark of cancer. Replicative senescence is a tumour suppressor process that imposes a limit on the proliferative potential of normal cells that all cancer cells must bypass. Here we show by whole-genome single-nucleotide bisulfite sequencing that replicative senescent human cells exhibit widespread DNA hypomethylation and focal hypermethylation. Hypomethylation occurs preferentially at gene-poor, late  ...[more]

Similar Datasets

2013-11-25 | GSE48580 | GEO
2017-01-20 | GSE80322 | GEO
2017-02-12 | GSE81797 | GEO
2017-02-12 | GSE81788 | GEO
2017-02-12 | GSE81786 | GEO
2017-02-12 | GSE90942 | GEO
2018-01-31 | GSE91070 | GEO
2018-01-31 | GSE91069 | GEO
2022-11-15 | MTBLS4568 | MetaboLights
2023-03-20 | GSE227160 | GEO