Secretion of SDF‐1α by bone marrow‐derived stromal cells enhances skin wound healing of C57BL/6 mice exposed to ionizing radiation

Patients treated for cancer therapy using ionizing radiation (IR) have delayed tissue repair and regeneration. The mechanisms mediating these defects remain largely unknown at present, thus limiting the development of therapeutic approaches. Using a wound healing model, we here investigate the mecha...

Full description

Saved in:
Bibliographic Details
Published inJournal of cellular and molecular medicine Vol. 14; no. 6b; pp. 1594 - 1604
Main Authors Landry, Yannick, Lê, Oanh, Mace, Kimberly A., Restivo, Terry E., Beauséjour, Christian M.
Format Journal Article
LanguageEnglish
Published Oxford, UK Blackwell Publishing Ltd 01.06.2010
John Wiley & Sons, Inc
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Patients treated for cancer therapy using ionizing radiation (IR) have delayed tissue repair and regeneration. The mechanisms mediating these defects remain largely unknown at present, thus limiting the development of therapeutic approaches. Using a wound healing model, we here investigate the mechanisms by which IR exposure limits skin regeneration. Our data show that induction of the stromal cell‐derived growth factor 1α (SDF‐1α) is severely impaired in the wounded skin of irradiated, compared to non‐irradiated, mice. Hence, we evaluated the potential of bone marrow‐derived multipotent stromal cells (MSCs), which secrete high levels of SDF‐1α, to improve skin regeneration in irradiated mice. Injection of MSCs into the wound margin led to remarkable enhancement of skin healing in mice exposed to IR. Injection of irradiated MSCs into the wound periphery of non‐irradiated mice delayed wound closure, also suggesting an important role for the stromal microenvironment in skin repair. The beneficial actions of MSCs were mainly paracrine, as the cells did not differentiate into keratinocytes. Specific knockdown of SDF‐1α expression led to drastically reduced efficiency of MSCs in improving wound closure, indicating that SDF‐1α secretion by MSCs is largely responsible for their beneficial action. We also found that one mechanism by which SDF‐1α enhances wound closure likely involves increased skin vascularization. Our findings collectively indicate that SDF‐1α is an important deregulated cytokine in irradiated wounded skin, and that the decline in tissue regeneration potential following IR can be reversed, given adequate microenvironmental support
ISSN:1582-1838
1582-4934
DOI:10.1111/j.1582-4934.2009.00887.x