Cited time in webofscience Cited time in scopus

Glucosylceramide synthase regulates adipo‐osteogenic differentiation through synergistic activation of PPARγ with GlcCer

Title
Glucosylceramide synthase regulates adipo‐osteogenic differentiation through synergistic activation of PPARγ with GlcCer
Author(s)
Jang, Hyun‐JunLim, SeyoungKim, Jung‐MinYoon, SoraLee, Chae YoungHwang, Hyeon‐JeongShin, Jeong WooShin, Kyeong JinKim, Hye YunPark, Kwang IlNam, DouguLee, Ja YilYea, KyungmooHirabayashi, YoshioLee, Yu JinChae, Young ChanSuh, Pann‐GhillChoi, Jang Hyun
DGIST Authors
Jang, Hyun‐JunLim, SeyoungKim, Jung‐MinYoon, SoraLee, Chae YoungHwang, Hyeon‐JeongShin, Jeong WooShin, Kyeong JinKim, Hye YunPark, Kwang IlNam, DouguLee, Ja YilYea, KyungmooHirabayashi, YoshioLee, Yu JinChae, Young ChanSuh, Pann‐GhillChoi, Jang Hyun
Issued Date
2020-01
Type
Article
Article Type
Article
Author Keywords
adipogenesisglucosylceramideglucosylceramide synthasemesenchymal stem cellsosteogenesisPDMPperoxisome proliferator-activated receptor gamma
Keywords
GAUCHER-DISEASEFATE DECISIONCELLSINHIBITIONMETABOLISMOSTEOBLASTCERAMIDEMARKERSMOUSE
ISSN
0892-6638
Abstract
Dysregulation of the adipo-osteogenic differentiation balance of mesenchymal stem cells (MSCs), which are common progenitor cells of adipocytes and osteoblasts, has been associated with many pathophysiologic diseases, such as obesity, osteopenia, and osteoporosis. Growing evidence suggests that lipid metabolism is crucial for maintaining stem cell homeostasis and cell differentiation; however, the detailed underlying mechanisms are largely unknown. Here, we demonstrate that glucosylceramide (GlcCer) and its synthase, glucosylceramide synthase (GCS), are key determinants of MSC differentiation into adipocytes or osteoblasts. GCS expression was increased during adipogenesis and decreased during osteogenesis. Targeting GCS using RNA interference or a chemical inhibitor enhanced osteogenesis and inhibited adipogenesis by controlling the transcriptional activity of peroxisome proliferator-activated receptor γ (PPARγ). Treatment with GlcCer sufficiently rescued adipogenesis and inhibited osteogenesis in GCS knockdown MSCs. Mechanistically, GlcCer interacted directly with PPARγ through A/B domain and synergistically enhanced rosiglitazone-induced PPARγ activation without changing PPARγ expression, thereby treatment with exogenous GlcCer increased adipogenesis and inhibited osteogenesis. Animal studies demonstrated that inhibiting GCS reduced adipocyte formation in white adipose tissues under normal chow diet and high-fat diet feeding and accelerated bone repair in a calvarial defect model. Taken together, our findings identify a novel lipid metabolic regulator for the control of MSC differentiation and may have important therapeutic implications. © 2019 Federation of American Societies for Experimental Biology.
URI
http://hdl.handle.net/20.500.11750/10977
DOI
10.1096/fj.201901437r
Publisher
Federation of American Societies for Experimental Biology
Related Researcher
  • 예경무 Yea, Kyungmoo
  • Research Interests Antibody; Engineering; Phage Display; Therapeutics; Immune; Exosome; Translational; Cytokine
Files in This Item:

There are no files associated with this item.

Appears in Collections:
Department of New Biology Protein Engineering Lab 1. Journal Articles

qrcode

  • twitter
  • facebook
  • mendeley

Items in Repository are protected by copyright, with all rights reserved, unless otherwise indicated.

BROWSE