Article ID Journal Published Year Pages File Type
2093447 Stem Cell Reports 2016 13 Pages PDF
Abstract

•p38α deletion in Prx1+ BM-MSCs led to osteoporosis and cartilage anomaly•p38α controls proliferation and tri-lineage differentiation of Prx1+ BM-MSCs•p38α regulates osteoclastogenesis through OPG production by BM-MSCs•The BM-MSC p38-OPG axis participates in estrogen deficiency-induced osteoporosis

SummaryBone marrow-derived mesenchymal stromal cells (BM-MSCs) are capable of differentiating into osteoblasts, chondrocytes, and adipocytes. Skewed differentiation of BM-MSCs contributes to the pathogenesis of osteoporosis. Yet how BM-MSC lineage commitment is regulated remains unclear. We show that ablation of p38α in Prx1+ BM-MSCs produced osteoporotic phenotypes, growth plate defects, and increased bone marrow fat, secondary to biased BM-MSC differentiation from osteoblast/chondrocyte to adipocyte and increased osteoclastogenesis and bone resorption. p38α regulates BM-MSC osteogenic commitment through TAK1-NF-κB signaling and osteoclastogenesis through osteoprotegerin (OPG) production by BM-MSCs. Estrogen activates p38α to maintain OPG expression in BM-MSCs to preserve the bone. Ablation of p38α in BM-MSCs positive for Dermo1, a later BM-MSC marker, only affected osteogenic differentiation. Thus, p38α mitogen-activated protein kinase (MAPK) in Prx1+ BM-MSCs acts to preserve the bone by promoting osteogenic lineage commitment and sustaining OPG production. This study thus unravels previously unidentified roles for p38α MAPK in skeletal development and bone remodeling.

Related Topics
Life Sciences Biochemistry, Genetics and Molecular Biology Biotechnology
Authors
, , , , , , , , , , , , , ,