Login / Signup

MSCs rescue impaired wound healing in a murine LAD1 model by adaptive responses to low TGF-β1 levels.

Dongsheng JiangKarmveer SinghJana MuschhammerSusanne SchatzAnca SindrilaruEvgenia MakrantonakiYu QiMeinhard WlaschekKarin Scharffetter-Kochanek
Published in: EMBO reports (2020)
Mutations in the CD18 gene encoding the common β-chain of β2 integrins result in impaired wound healing in humans and mice suffering from leukocyte adhesion deficiency syndrome type 1 (LAD1). Transplantation of adipose tissue-derived mesenchymal stem cells (MSCs) restores normal healing of CD18-/- wounds by restoring the decreased TGF-β1 concentrations. TGF-β1 released from MSCs leads to enhanced myofibroblast differentiation, wound contraction, and vessel formation. We uncover that MSCs are equipped with a sensing mechanism for TGF-β1 concentrations at wound sites. Low TGF-β1 concentrations as occurring in CD18-/- wounds induce TGF-β1 release from MSCs, whereas high TGF-β1 concentrations suppress TGF-β1 production. This regulation depends on TGF-β receptor sensing and is relayed to microRNA-21 (miR-21), which subsequently suppresses the translation of Smad7, the negative regulator of TGF-β1 signaling. Inactivation of TGF-β receptor, or overexpression or silencing of miR-21 or Smad7, abrogates TGF-β1 sensing, and thus prevents the adaptive MSC responses required for tissue repair.
Keyphrases