Your browser doesn't support javascript.
loading
Role of RHEB in Regulating Differentiation Fate of Mesenchymal Stem Cells for Cartilage and Bone Regeneration.
Ashraf, Sajjad; Han, In-Bo; Park, Hansoo; Lee, Soo-Hong.
  • Ashraf S; Department of Biomedical Science, CHA University, Seongnam-si 463-400, Korea. sajjadashraf471@gmail.com.
  • Han IB; School of Integrative Engineering, Chung-Ang University, Seoul 156-863, Korea. sajjadashraf471@gmail.com.
  • Park H; Department of Neurosurgery, Bundang Medical Center, CHA University, Seongnam-si 463-400, Korea. hanib@cha.ac.kr.
  • Lee SH; School of Integrative Engineering, Chung-Ang University, Seoul 156-863, Korea. heyshoo@cau.ac.kr.
Int J Mol Sci ; 18(4)2017 Apr 24.
Article en En | MEDLINE | ID: mdl-28441755
ABSTRACT
Advances in mesenchymal stem cells (MSCs) and cell replacement therapies are promising approaches to treat cartilage and bone defects since substantial differentiation capacities of MSCs match the demands of tissue regeneration. Our understanding of the dynamic process requiring indispensable differentiation of MSCs remains limited. Herein, we describe the role of RHEB (Ras homolog enriched in brain) regulating gene signature for differentiation of human adipose derived mesenchymal stem cells (ASCs) into chondrogenic, osteogenic, and adipogenic lineages. RHEB-overexpression increases the proliferation of the ASCs. RHEB enhances the chondrogenic differentiation of ASCs in 3D culture via upregulation of SOX9 with concomitant increase in glycosaminoglycans (GAGs), and type II collagen (COL2). RHEB increases the osteogenesis via upregulation of runt related transcription factor 2 (RUNX2) with an increase in the calcium and phosphate contents. RHEB also increases the expression of osteogenic markers, osteonectin and osteopontin. RHEB knockdown ASCs were incapable of expressing sufficient SRY (Sex determining region Y)-box 9 (SOX9) and RUNX2, and therefore had decreased chondrogenic and osteogenic differentiation. RHEB-overexpression impaired ASCs differentiation into adipogenic lineage, through downregulation of CCAAT/enhancer binding protein beta (C/EBPß). Conversely, RHEB knockdown abolished the negative regulation of adipogenesis. We demonstrate that RHEB is a novel regulator, with a critical role in ASCs lineage determination, and RHEB-modulated ASCs may be useful as a cell therapy for cartilage and bone defect treatments.
Asunto(s)
Palabras clave

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Regeneración / Huesos / Cartílago / Células Madre Mesenquimatosas / Proteína Homóloga de Ras Enriquecida en el Cerebro Límite: Female / Humans / Middle aged Idioma: En Año: 2017 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Regeneración / Huesos / Cartílago / Células Madre Mesenquimatosas / Proteína Homóloga de Ras Enriquecida en el Cerebro Límite: Female / Humans / Middle aged Idioma: En Año: 2017 Tipo del documento: Article