Abstract

Glucocorticoid provokes bone mass loss and fatty marrow, accelerating osteoporosis development. Bromodomain protein BRD4, an acetyl–histone-binding chromatin reader, regulates stem cell and tissue homeostasis. We uncovered that glucocorticoid inhibited acetyl Lys-9 at the histone 3 (H3K9ac)-binding Runx2 promoter and decreased osteogenic differentiation, whereas bromodomain protein 4 (BRD4) and adipocyte formation were upregulated in bone-marrow mesenchymal progenitor cells. BRD4 knockdown improved H3K9ac occupation at the Runx2 promoter and osteogenesis, but attenuated glucocorticoid-mediated adipocyte formation together with the unaffected H3K9ac-binding PPARγ2 promoter. BRD4 regulated epigenome related to fatty acid metabolism and the forkhead box P1 (Foxp1) pathway, which occupied the PPARγ2 promoter to modulate glucocorticoid-induced adipocytic activity. In vivo, BRD4 inhibitor JQ-1 treatment mitigated methylprednisolone-induced suppression of bone mass, trabecular microstructure, mineral acquisition, and osteogenic differentiation. Foxp1 signaling, marrow fat, and adipocyte formation in glucocorticoid-treated skeleton were reversed upon JQ-1 treatment. Taken together, glucocorticoid-induced H3K9 hypoacetylation augmented BRD4 action to Foxp1, which steered mesenchymal progenitor cells toward adipocytes at the cost of osteogenic differentiation in osteoporotic skeletons. BRD4 inhibition slowed bone mass loss and marrow adiposity. Collective investigations convey a new epigenetic insight into acetyl histone reader BRD4 control of osteogenesis and adipogenesis in skeleton, and highlight the remedial effects of the BRD4 inhibitor on glucocorticoid-induced osteoporosis.

Highlights

  • Physiological glucocorticoid is indispensable in sustaining differentiation capacity of osteogenic cells, underpinning bone mineral accretion to maintain skeletal tissue health [1] and bone regeneration [2]

  • We investigated whether H3K9 acetylation affected osteoblast differentiation or adipocyte formation in glucocorticoid-treated osteoprogenitor cells

  • Bone-marrow mesenchymal stem cells were incubated in osteogenic medium with and without 1 μM dexamethasone, an in vitro model mimicking glucocorticoid-induced bone loss

Read more

Summary

Introduction

Physiological glucocorticoid is indispensable in sustaining differentiation capacity of osteogenic cells, underpinning bone mineral accretion to maintain skeletal tissue health [1] and bone regeneration [2]. The development of mineral acquisition loss and fatty marrow hints that osteogenic and adipogenic programs in mesenchymal progenitor cells are dysregulated in the osteoporotic bone microenvironment. Increasing evidence has revealed that epigenetic pathways modulate gene transcription without altering genomic sequences, regulating osteogenesis of mesenchymal stem cells, bone tissue metabolism, and osteoporosis development [9]. Acetyltransferase p300/CBP-associated factor (PACF) hyperacetylation of lysine 9 at histone 3 (H3K9) increases bone morphogenetic protein gene expression, upregulating osteogenic differentiation of mesenchymal stem cells [11].

Objectives
Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call