Abstract

Growth differentiation factor 11 (GDF11), a member of the transforming growth factor-β family, has been shown to act as a negative regulator in cardiac hypertrophy. Ca2+ signaling modulates cardiomyocyte growth; however, the role of Ca2+-dependent mechanisms in mediating the effects of GDF11 remains elusive. Here, we found that GDF11 induced intracellular Ca2+ increases in neonatal rat cardiomyocytes and that this response was blocked by chelating the intracellular Ca2+ with BAPTA-AM or by pretreatment with inhibitors of the inositol 1,4,5-trisphosphate (IP3) pathway. Moreover, GDF11 increased the phosphorylation levels and luciferase activity of Smad2/3 in a concentration-dependent manner, and the inhibition of IP3-dependent Ca2+ release abolished GDF11-induced Smad2/3 activity. To assess whether GDF11 exerted antihypertrophic effects by modulating Ca2+ signaling, cardiomyocytes were exposed to hypertrophic agents (100 nM testosterone or 50 μM phenylephrine) for 24 h. Both treatments increased cardiomyocyte size and [3H]-leucine incorporation, and these responses were significantly blunted by pretreatment with GDF11 over 24 h. Moreover, downregulation of Smad2 and Smad3 with siRNA was accompanied by inhibition of the antihypertrophic effects of GDF11. These results suggest that GDF11 modulates Ca2+ signaling and the Smad2/3 pathway to prevent cardiomyocyte hypertrophy.

Highlights

  • Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor-β (TGF-β) superfamily and has various roles in the cardiovascular system [1,2]

  • Our findings suggest that pretreatment of cardiomyocytes with GDF11 prevented hypertrophy induced by testosterone and phenylephrine by modulating the Smad2/3 pathway

  • We found that GDF11 increased Smad2/3 activity and prevented cardiomyocyte hypertrophy with the involvement of Smad2/3 proteins through intracellular Ca2+-dependent pathways

Read more

Summary

Introduction

Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor-β (TGF-β) superfamily and has various roles in the cardiovascular system [1,2]. Circulating GDF11 levels increase after induction of cardiac infarction in mice [3]. The degree of plasma GDF11 concentration increase is dependent on the severity of heart failure, suggesting that GDF11 is an adjustable mediator in cardiac remodeling [3]. Exogenous administration of GDF11, through controlled peptide delivery from secretory cells, decreases infarct size in the heart, which contributes to preservation of cardiac function [4]. Circulating GDF11 levels represent an important negative regulator of hypertrophic and remodeling processes, which contribute to heart failure. Exogenous administration of GDF11 blocks cardiac hypertrophy induced by various stimuli or prohypertrophic conditions [6]. Diverse effects of GDF11 have been reported in cardiac cells, the mechanisms underlying these effects are poorly understood

Methods
Results
Discussion
Conclusion
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