Your browser doesn't support javascript.
loading
Fighting Against Promoter DNA Hyper-Methylation: Protective Histone Modification Profiles of Stress-Resistant Intestinal Stem Cells.
Thalheim, Torsten; Hopp, Lydia; Herberg, Maria; Siebert, Susann; Kerner, Christiane; Quaas, Marianne; Schweiger, Michal R; Aust, Gabriela; Galle, Joerg.
Afiliação
  • Thalheim T; Interdisciplinary Center for Bioinformatics (IZBI), Leipzig University, 04107 Leipzig, Germany.
  • Hopp L; Interdisciplinary Center for Bioinformatics (IZBI), Leipzig University, 04107 Leipzig, Germany.
  • Herberg M; Interdisciplinary Center for Bioinformatics (IZBI), Leipzig University, 04107 Leipzig, Germany.
  • Siebert S; Laboratory for Translational Epigenetics and Tumor Genetics, University Hospital Cologne, 50391 Cologne, Germany.
  • Kerner C; Center for Molecular Medicine Cologne, CMMC, 50391 Cologne, Germany.
  • Quaas M; Department of Surgery, Research Laboratories, Leipzig University, 04103 Leipzig, Germany.
  • Schweiger MR; Department of Surgery, Research Laboratories, Leipzig University, 04103 Leipzig, Germany.
  • Aust G; Laboratory for Translational Epigenetics and Tumor Genetics, University Hospital Cologne, 50391 Cologne, Germany.
  • Galle J; Center for Molecular Medicine Cologne, CMMC, 50391 Cologne, Germany.
Int J Mol Sci ; 21(6)2020 Mar 12.
Article em En | MEDLINE | ID: mdl-32178409
ABSTRACT
Aberrant DNA methylation in stem cells is a hallmark of aging and tumor development. Recently, we have suggested that promoter DNA hyper-methylation originates in DNA repair and that even successful DNA repair might confer this kind of epigenetic long-term change. Here, we ask for interrelations between promoter DNA methylation and histone modification changes observed in the intestine weeks after irradiation and/or following Msh2 loss. We focus on H3K4me3 recruitment to the promoter of H3K27me3 target genes. By RNA- and histone ChIP-sequencing, we demonstrate that this recruitment occurs without changes of the average gene transcription and does not involve H3K9me3. Applying a mathematical model of epigenetic regulation of transcription, we show that the recruitment can be explained by stronger DNA binding of H3K4me3 and H3K27me3 histone methyl-transferases as a consequence of lower DNA methylation. This scenario implicates stable transcription despite of H3K4me3 recruitment, in agreement with our RNA-seq data. Following several kinds of stress, including moderate irradiation, stress-sensitive intestinal stem cell (ISCs) are known to become replaced by more resistant populations. Our simulation results suggest that the stress-resistant ISCs are largely protected against promoter hyper-methylation of H3K27me3 target genes.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Células-Tronco / DNA / Histonas / Regiões Promotoras Genéticas / Metilação de DNA / Intestinos Limite: Animals Idioma: En Ano de publicação: 2020 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Células-Tronco / DNA / Histonas / Regiões Promotoras Genéticas / Metilação de DNA / Intestinos Limite: Animals Idioma: En Ano de publicação: 2020 Tipo de documento: Article