4.6 Article

Epigenetic Mechanisms Mediating Cell State Transitions in Chondrocytes

Journal

JOURNAL OF BONE AND MINERAL RESEARCH
Volume 36, Issue 5, Pages 968-985

Publisher

WILEY
DOI: 10.1002/jbmr.4263

Keywords

EPIGENETICS; GENETIC RESEARCH; CHONDROCYTE AND CARTILAGE BIOLOGY; GROWTH PLATE; CELL; TISSUE SIGNALING; TRANSCRIPTION FACTORS; DEVELOPMENTAL MODELING; BONE MODELING AND REMODELING

Funding

  1. DFG [FOR 2407, Vo620/14, GRK 1431]

Ask authors/readers for more resources

Research has shown that specific differentiation steps during endochondral ossification are regulated by specific epigenetic modifications. Differences in gene expression and enhancer profiles between cell types are significant, with metabolic pathways playing a key role in cell differentiation.
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. (c) 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).

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available