Your browser doesn't support javascript.
loading
Metabolic programming determines the lineage-differentiation fate of murine bone marrow stromal progenitor cells.
Tencerova, Michaela; Rendina-Ruedy, Elizabeth; Neess, Ditte; Færgeman, Nils; Figeac, Florence; Ali, Dalia; Danielsen, Morten; Haakonsson, Anders; Rosen, Clifford J; Kassem, Moustapha.
Affiliation
  • Tencerova M; 1Department of Molecular Endocrinology, University of Southern Denmark and Odense University Hospital, 5000 Odense, Denmark.
  • Rendina-Ruedy E; 2Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME 04074 USA.
  • Neess D; 3Villum Center for Analytical Biosciences, Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark.
  • Færgeman N; 3Villum Center for Analytical Biosciences, Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark.
  • Figeac F; 1Department of Molecular Endocrinology, University of Southern Denmark and Odense University Hospital, 5000 Odense, Denmark.
  • Ali D; 1Department of Molecular Endocrinology, University of Southern Denmark and Odense University Hospital, 5000 Odense, Denmark.
  • Danielsen M; MSOmics, 2950 Copenhagen, Denmark.
  • Haakonsson A; 1Department of Molecular Endocrinology, University of Southern Denmark and Odense University Hospital, 5000 Odense, Denmark.
  • Rosen CJ; 2Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME 04074 USA.
  • Kassem M; 1Department of Molecular Endocrinology, University of Southern Denmark and Odense University Hospital, 5000 Odense, Denmark.
Bone Res ; 7: 35, 2019.
Article in En | MEDLINE | ID: mdl-31754546
ABSTRACT
Enhanced bone marrow adipogenesis and impaired osteoblastogenesis have been observed in obesity, suggesting that the metabolic microenvironment regulates bone marrow adipocyte and osteoblast progenitor differentiation fate. To determine the molecular mechanisms, we studied two immortalized murine cell lines of adipocyte or osteoblast progenitors (BMSCsadipo and BMSCsosteo, respectively) under basal and adipogenic culture conditions. At baseline, BMSCsadipo, and BMSCsosteo exhibit a distinct metabolic program evidenced by the presence of specific global gene expression, cellular bioenergetics, and metabolomic signatures that are dependent on insulin signaling and glycolysis in BMSCsosteo versus oxidative phosphorylation in BMSCsadipo. To test the flexibility of the metabolic program, we treated BMSCsadipo with parathyroid hormone, S961 (an inhibitor of insulin signaling) and oligomycin (an inhibitor of oxidative phosphorylation). The treatment induced significant changes in cellular bioenergetics that were associated with decreased adipocytic differentiation. Similarly, 12 weeks of a high-fat diet in mice led to the expansion of adipocyte progenitors, enhanced adipocyte differentiation and insulin signaling in cultured BMSCs. Our data demonstrate that BMSC progenitors possess a distinct metabolic program and are poised to respond to exogenous metabolic cues that regulate their differentiation fate.
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Bone Res Year: 2019 Document type: Article Affiliation country: Denmark

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Bone Res Year: 2019 Document type: Article Affiliation country: Denmark
...