Abstract

Introduction: Resident c-Kit + cardiac progenitor cells (CPCs) differentiation into cardiomyocytes in vivo is controversial. Using lineage tracing and gene ablation techniques we documented that the Wnt signaling modulator Sfrp2 substantially increased differentiation of c-Kit + CPCs into cardiomyocytes in post-MI heart and restored cardiac function. Methods: c-Kit-CreERT2/mTeG mice were used where the c-Kit promoter drives expression of a tamoxifen (TX)-inducible Cre. Upon TX treatment Cre-mediated recombination drives expression of eGFP exclusively in c-Kit cells. Mice were crossed with a Diphtheria toxin receptor (DT) strain to selectively ablate heart c-Kit cells. Mice were subjected to myocardial infarction. 2 days later, mice were injected with Sfrp2 (0.5 μg) or saline at the infarct border zone. Heart tissue was analyzed 2 months post-MI for eGFP expression and various markers by microscopy. Results: c-Kit-CreERT2/mTeG model was tested. eGFP expression had no effect on in vitro c-Kit CPC differentiation or proliferation. In TX treated mice, >90% of c-Kit+ cells were eGFP+. No eGFP+ c-Kit+ cells were seen in vehicle-only mice (N=6, p<0.001). c-Kit negative cells, such as cardiomyocytes, endothelial cells, and smooth muscle cells, did not express eGFP following TX treatment. In sham animals, eGFP was localized to small c-Kit+ cells. In the vehicle MI group eGFP+ cardiomyocytes were rare (0.08% in infarct border zone. N=8, P=0.005). In contrast, the number of eGFP+ cardiomyocytes was significantly higher in the Sfrp2 treated group (15% in infarct border zone, N=10, P=0.0002). Co-localization of eGFP with blood vessels (0.77%, N=10) was infrequent. Induction of c-Kit CPC differentiation into cardiomyocytes by Sfrp2 was associated with lower fibrosis (15%, N=10, P=0.01), higher LV mass (58mg, N=10, P=0.01) and increased ejection fraction (16%, N=10, P=0.02). DT treatment ablated c-Kit cells (>95% N=4, P=0.01). Sfrp2 induction of c-Kit differentiation to cardiomyocytes and functional improvements were completely ablated by DT. Conclusion: Using c-Kit lineage-tracing and genetic ablation we have identified that these cells are important in cardiomyocyte generation in vivo in response to Sfrp2 and improves cardiac function following MI.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call