4.7 Article

Gli3 Regulation of Myogenesis Is Necessary for Ischemia-Induced Angiogenesis

Journal

CIRCULATION RESEARCH
Volume 113, Issue 10, Pages 1148-+

Publisher

LIPPINCOTT WILLIAMS & WILKINS
DOI: 10.1161/CIRCRESAHA.113.301546

Keywords

angiogenesis, pathological; hedgehogs; ischemia; muscle, skeletal; regeneration

Funding

  1. Fondation de la Recherche Medicale, program on cardiovascular aging [DCV20070409258]
  2. Conseil Regional d'Aquitaine (action inter-regionale Aquitaine-Midi Pyrenees)
  3. Communaute de Travail des Pyrenees
  4. ANR program [ANR-07-PHYSIO-010-02]
  5. National League against Cancer
  6. CHU de Bordeaux
  7. National Institute of Health [HL093439, HL113541]

Ask authors/readers for more resources

Rationale: A better understanding of the mechanism underlying skeletal muscle repair is required to develop therapies that promote tissue regeneration in adults. Hedgehog signaling has been shown previously to be involved in myogenesis and angiogenesis: 2 crucial processes for muscle development and regeneration. Objective: The objective of this study was to identify the role of the hedgehog transcription factor Gli3 in the cross-talk between angiogenesis and myogenesis in adults. Methods and Results: Using conditional knockout mice, we found that Gli3 deficiency in endothelial cells did not affect ischemic muscle repair, whereas in myocytes, Gli3 deficiency resulted in severely delayed ischemia-induced myogenesis. Moreover, angiogenesis was also significantly impaired in HSA-Cre(ERT2); Gli3(Flox/Flox) mice, demonstrating that impaired myogenesis indirectly affects ischemia-induced angiogenesis. The role of Gli3 in myocytes was then further investigated. We found that Gli3 promotes myoblast differentiation through myogenic factor 5 regulation. In addition, we found that Gli3 regulates several proangiogenic factors, including thymidine phosphorylase and angiopoietin-1 both in vitro and in vivo, which indirectly promote endothelial cell proliferation and arteriole formation. In addition, we found that Gli3 is upregulated in proliferating myoblasts by the cell cycle-associated transcription factor E2F1. Conclusions: This study shows for the first time that Gli3-regulated postnatal myogenesis is necessary for muscle repair-associated angiogenesis. Most importantly, it implies that myogenesis drives angiogenesis in the setting of skeletal muscle repair and identifies Gli3 as a potential target for regenerative medicine.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available