Clinical and Translational Medicine (Feb 2022)

Melatonin enhances osteoblastogenesis of senescent bone marrow stromal cells through NSD2‐mediated chromatin remodelling

  • Ying Xie,
  • Na Han,
  • Feng Li,
  • Lijuan Wang,
  • Gerui Liu,
  • Meilin Hu,
  • Sheng Wang,
  • Xuelei Wei,
  • Jing Guo,
  • Hongmei Jiang,
  • Jingjing Wang,
  • Xin Li,
  • Yixuan Wang,
  • Jingya Wang,
  • Xiyun Bian,
  • Zhongjiao Zhu,
  • Hui Zhang,
  • Chunhua Liu,
  • Xiaozhi Liu,
  • Zhiqiang Liu

DOI
https://doi.org/10.1002/ctm2.746
Journal volume & issue
Vol. 12, no. 2
pp. n/a – n/a

Abstract

Read online

Abstract Background Aging‐associated osteoporosis is frequently seen in the elderly in clinic, but efficient managements are limited because of unclear nosogenesis. The current study aims to investigate the role of melatonin on senescent bone marrow stromal cells (BMSCs) and the underlying regulating mechanism. Methods Melatonin levels were tested by ELISA. Gene expression profiles were performed by RNA‐sequencing, enrichment of H3K36me2 on gene promoters was analyzed by Chromatin Immunoprecipitation Sequencing (ChIP‐seq), and chromatin accessibility was determined by Assay for Transposase‐Accessible Chromatin with high‐throughput sequencing (ATAC‐seq). Osteogenesis of BMSCs in vitro was measured by Alizarin Red and Alkaline Phosphatase staining, and in vivo effects of melatonin was assessed by histological staining and micro computed tomography (micro‐CT) scan. Correlation of NSD2 expression and severity of senile osteoporosis patients were analyzed by Pearson correlation. Results Melatonin levels were decreased during aging in human bone marrow, accompanied by downregulation of the histone methyltransferase nuclear receptor binding SET domain protein 2 (NSD2) expression in the senescent BMSCs. Melatonin stimulated the expression of NSD2 through MT1/2‐mediated signaling pathways, resulting in the rebalancing of H3K36me2 and H3K27me3 modifications to increase chromatin accessibility of the osteogenic genes, runt‐related transcription factor 2 (RUNX2) and bone gamma‐carboxyglutamate protein (BGLAP). Melatonin promoted osteogenesis of BMSCs in vitro, and alleviates osteoporosis progression in the aging mice. In clinic, severity of senile osteoporosis (SOP) was negatively correlated with melatonin level in bone marrow, as well as NSD2 expression in BMSCs. Similarly, melatonin remarkably enhanced osteogenic differentiation of BMSCs derived from SOP patients in vitro. Conclusions Collectively, our study dissects previously unreported mechanistic insights into the epigenetic regulating machinery of melatonin in meliorating osteogenic differentiation of senescent BMSC, and provides evidence for application of melatonin in preventing aging‐associated bone loss.

Keywords