RECK (reversion-inducing cysteine-rich protein with Kazal motifs) regulates migration, differentiation and Wnt/beta-catenin signaling in human mesenchymal stem cells

Christian Mahl, Virginia Egea, Remco T. A. Megens, Thomas Pitsch, Donato Santovito, Christian Weber, Christian Ries*

*Corresponding author for this work

Research output: Contribution to journalArticleAcademicpeer-review

Abstract

The membrane-anchored glycoprotein RECK (reversion-inducing cysteine-rich protein with Kazal motifs) inhibits expression and activity of certain matrix metalloproteinases (MMPs), thereby suppressing tumor cell metastasis. However, RECK's role in physiological cell function is largely unknown. Human mesenchymal stem cells (hMSCs) are able to differentiate into various cell types and represent promising tools in multiple clinical applications including the regeneration of injured tissues by endogenous or transplanted hMSCs. RNA interference of RECK in hMSCs revealed that endogenous RECK suppresses the transcription and biosynthesis of tissue inhibitor of metalloproteinases (TIMP)-2 but does not influence the expression of MMP-2, MMP-9, membrane type (MT)1-MMP and TIMP-1 in these cells. Knockdown of RECK in hMSCs promoted monolayer regeneration and chemotactic migration of hMSCs, as demonstrated by scratch wound and chemotaxis assay analyses. Moreover, expression of endogenous RECK was upregulated upon osteogenic differentiation and diminished after adipogenic differentiation of hMSCs. RECK depletion in hMSCs reduced their capacity to differentiate into the osteogenic lineage whereas adipogenesis was increased, demonstrating that RECK functions as a master switch between both pathways. Furthermore, knockdown of RECK in hMSCs attenuated the Wnt/beta-catenin signaling pathway as indicated by reduced stability and impaired transcriptional activity of beta-catenin. The latter was determined by analysis of the beta-catenin target genes Dickkopf1 (DKK1), axis inhibition protein 2 (AXIN2), runt-related transcription factor 2 (RUNX2) and a luciferase-based beta-catenin-activated reporter (BAR) assay. Our findings demonstrate that RECK is a regulator of hMSC functions suggesting that modulation of RECK may improve the development of hMSC-based therapeutical approaches in regenerative medicine.
Original languageEnglish
Pages (from-to)1489-1501
JournalCellular and Molecular Life Sciences
Volume73
Issue number7
DOIs
Publication statusPublished - Apr 2016

Keywords

  • RECK
  • hMSC
  • Chemotactic migration
  • Osteogenic differentiation
  • Canonical Wnt/beta-catenin signaling

Cite this