Your browser doesn't support javascript.
loading
Epigenetic Mechanisms Mediating Cell State Transitions in Chondrocytes.
Wuelling, Manuela; Neu, Christoph; Thiesen, Andrea M; Kitanovski, Simo; Cao, Yingying; Lange, Anja; Westendorf, Astrid M; Hoffmann, Daniel; Vortkamp, Andrea.
Afiliação
  • Wuelling M; Developmental Biology, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany.
  • Neu C; Developmental Biology, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany.
  • Thiesen AM; Developmental Biology, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany.
  • Kitanovski S; Bioinformatics and Computational Biophysics, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany.
  • Cao Y; Bioinformatics and Computational Biophysics, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany.
  • Lange A; Bioinformatics and Computational Biophysics, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany.
  • Westendorf AM; Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany.
  • Hoffmann D; Bioinformatics and Computational Biophysics, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany.
  • Vortkamp A; Developmental Biology, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany.
J Bone Miner Res ; 36(5): 968-985, 2021 05.
Article em En | MEDLINE | ID: mdl-33534175
ABSTRACT
Epigenetic modifications play critical roles in regulating cell lineage differentiation, but the epigenetic mechanisms guiding specific differentiation steps within a cell lineage have rarely been investigated. To decipher such mechanisms, we used the defined transition from proliferating (PC) into hypertrophic chondrocytes (HC) during endochondral ossification as a model. We established a map of activating and repressive histone modifications for each cell type. ChromHMM state transition analysis and Pareto-based integration of differential levels of mRNA and epigenetic marks revealed that differentiation-associated gene repression is initiated by the addition of H3K27me3 to promoters still carrying substantial levels of activating marks. Moreover, the integrative analysis identified genes specifically expressed in cells undergoing the transition into hypertrophy. Investigation of enhancer profiles detected surprising differences in enhancer number, location, and transcription factor binding sites between the two closely related cell types. Furthermore, cell type-specific upregulation of gene expression was associated with increased numbers of H3K27ac peaks. Pathway analysis identified PC-specific enhancers associated with chondrogenic genes, whereas HC-specific enhancers mainly control metabolic pathways linking epigenetic signature to biological functions. Since HC-specific enhancers show a higher conservation in postnatal tissues, the switch to metabolic pathways seems to be a hallmark of differentiated tissues. Surprisingly, the analysis of H3K27ac levels at super-enhancers revealed a rapid adaption of H3K27ac occupancy to changes in gene expression, supporting the importance of enhancer modulation for acute alterations in gene expression. © 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Condrócitos / Epigênese Genética Tipo de estudo: Prognostic_studies Idioma: En Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Condrócitos / Epigênese Genética Tipo de estudo: Prognostic_studies Idioma: En Ano de publicação: 2021 Tipo de documento: Article