Login / Signup

SMURF2 phosphorylation at Thr249 modifies glioma stemness and tumorigenicity by regulating TGF-β receptor stability.

Manami HiraiwaKazuya FukasawaTakashi IezakiHemragul SabitTetsuhiro HorieKazuya TokumuraSayuki IwahashiMisato MurataMasaki KobayashiAkane SuzukiGyujin ParkKatsuyuki KanedaTomoki TodoAtsushi HiraoMitsutoshi NakadaEiichi Hinoi
Published in: Communications biology (2022)
Glioma stem cells (GSCs) contribute to the pathogenesis of glioblastoma, the most malignant form of glioma. The implication and underlying mechanisms of SMAD specific E3 ubiquitin protein ligase 2 (SMURF2) on the GSC phenotypes remain unknown. We previously demonstrated that SMURF2 phosphorylation at Thr 249 (SMURF2 Thr249 ) activates its E3 ubiquitin ligase activity. Here, we demonstrate that SMURF2 Thr249 phosphorylation plays an essential role in maintaining GSC stemness and tumorigenicity. SMURF2 silencing augmented the self-renewal potential and tumorigenicity of patient-derived GSCs. The SMURF2 Thr249 phosphorylation level was low in human glioblastoma pathology specimens. Introduction of the SMURF2 T249A mutant resulted in increased stemness and tumorigenicity of GSCs, recapitulating the SMURF2 silencing. Moreover, the inactivation of SMURF2 Thr249 phosphorylation increases TGF-β receptor (TGFBR) protein stability. Indeed, TGFBR1 knockdown markedly counteracted the GSC phenotypes by SMURF2 T249A mutant. These findings highlight the importance of SMURF2 Thr249 phosphorylation in maintaining GSC phenotypes, thereby demonstrating a potential target for GSC-directed therapy.
Keyphrases
  • stem cells
  • transforming growth factor
  • endothelial cells
  • risk assessment
  • binding protein
  • cell therapy
  • amino acid
  • signaling pathway
  • protein protein
  • wild type