학술논문

Sphingosine 1-phosphate elicits RhoA-dependent proliferation and MRTF-A mediated gene induction in CPCs
Document Type
article
Source
Cellular Signalling. 28(8)
Subject
Stem Cell Research
Cardiovascular
Heart Disease
Underpinning research
1.1 Normal biological development and functioning
Animals
Biomarkers
Cell Lineage
Cell Nucleus
Cell Proliferation
Lysophospholipids
Male
Mice
Knockout
Myocardium
RNA
Messenger
Receptors
G-Protein-Coupled
Receptors
Lysosphingolipid
Serum Response Factor
Sphingosine
Stem Cells
Trans-Activators
Transcription
Genetic
Transcriptional Activation
rhoA GTP-Binding Protein
Sphingosine-1-phosphate
GPCR
Cardiac progenitor cells
MRTF-A
RhoA
Biochemistry and Cell Biology
Medical Physiology
Biochemistry & Molecular Biology
Language
Abstract
Although c-kit(+) cardiac progenitor cells (CPCs) are currently used in clinical trials there remain considerable gaps in our understanding of the molecular mechanisms underlying their proliferation and differentiation. G-protein coupled receptors (GPCRs) play an important role in regulating these processes in mammalian cell types thus we assessed GPCR mRNA expression in c-kit(+) cells isolated from adult mouse hearts. Our data provide the first comprehensive overview of the distribution of this fundamental class of cardiac receptors in CPCs and reveal notable distinctions from that of adult cardiomyocytes. We focused on GPCRs that couple to RhoA activation in particular those for sphingosine-1-phosphate (S1P). The S1P2 and S1P3 receptors are the most abundant S1P receptor subtypes in mouse and human CPCs while cardiomyocytes express predominantly S1P1 receptors. Treatment of CPCs with S1P, as with thrombin and serum, increased proliferation through a pathway requiring RhoA signaling, as evidenced by significant attenuation when Rho was inhibited by treatment with C3 toxin. Further analysis demonstrated that both S1P- and serum-induced proliferation are regulated through the S1P2 and S1P3 receptor subtypes which couple to Gα12/13 to elicit RhoA activation. The transcriptional co-activator MRTF-A was activated by S1P as assessed by its nuclear accumulation and induction of a RhoA/MRTF-A luciferase reporter. In addition S1P treatment increased expression of cardiac lineage markers Mef2C and GATA4 and the smooth muscle marker GATA6 through activation of MRTF-A. In conclusion, we delineate an S1P-regulated signaling pathway in CPCs that introduces the possibility of targeting S1P2/3 receptors, Gα12/13 or RhoA to influence the proliferation and commitment of c-kit(+) CPCs and improve the response of the myocardium following injury.