Abstract

This study identifies signaling pathways that play key roles in the formation and maintenance of epicardial cells, a source of progenitors for coronary smooth muscle cells (SMCs). After epithelial to mesenchymal transition (EMT), mesenchymal cells invade the myocardium to form coronary SMCs. RhoA/Rho kinase activity is required for EMT and for differentiation into coronary SMCs, whereas cAMP activity is known to inhibit EMT in epithelial cells by an unknown mechanism. We use outgrowth of epicardial cells from E9.5 isolated mouse proepicardium (PE) explants, wild type and Epac1 null E12.5 mouse heart explants, adult rat epicardial cells, and immortalized mouse embryonic epicardial cells as model systems to identify signaling pathways that regulate RhoA activity to maintain the epicardial progenitor state. We demonstrate that RhoA activity is suppressed in the epicardial progenitor state, that the cAMP-dependent Rap1 GTP exchange factor (GEF), Epac, known to down-regulate RhoA activity through activation of Rap1 GTPase activity increased, that Rap1 activity increased, and that expression of the RhoA antagonistic Rnd proteins known to activate p190RhoGAP increased and associated with p190RhoGAP. Finally, EMT is associated with increased p63RhoGEF and RhoGEF-H1 protein expression, increased GEF-H1 activity, with a trend in increased p63RhoGEF activity. EMT is suppressed by partial silencing of p63RhoGEF and GEF-H1. In conclusion, we have identified new signaling molecules that act together to control RhoA activity and play critical roles in the maintenance of coronary smooth muscle progenitor cells in the embryonic epicardium. We suggest that their eventual manipulation could promote revascularization after myocardial injury.

Highlights

  • Epicardial cells are a potential source of progenitor cells for revascularization of the injured heart

  • Activation of Epac Suppresses and Silencing of Epac Enhances epithelial to mesenchymal transition (EMT) in Cultured embryonic epicardial cells (EECs) and Cell Migration of Epicardial Cells— We previously demonstrated that activation of the cAMP-dependent, protein kinase A (PKA)-independent Rap1 GTP exchange factor, Epac, relaxed smooth muscle (SM) tissues through increased Rap1 activity that resulted in decreased RhoA activity [19]

  • Using GSTRhoAG17A and Western blotting analysis to sediment active GTP exchange factor (GEF), we showed that the activity of GEF-H1 increased significantly with a consistent trend for an increase in p63RhoGEF but not VAV2, consistent with the changes in the expression level of GEF proteins with TGF-␤1-induced EMT

Read more

Summary

Background

Epicardial cells are a potential source of progenitor cells for revascularization of the injured heart. We have identified new signaling molecules that act together to control RhoA activity and play critical roles in the maintenance of coronary smooth muscle progenitor cells in the embryonic epicardium. We suggest that their eventual manipulation could promote revascularization after myocardial injury. We show up-regulation of the RhoA-specific GEFs p63RhoGEF and GEF-H1, expression, and activity upon growth factor-induced EMT, which is suppressed by partial silencing of these GEFs. Identification of responsible signaling molecules could provide new molecular targets for eventual manipulation of these SM progenitor cells and should be examined in the future for their roles during revascularization after myocardial injury or hypoxia

EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
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