Abstract

Large-conductance and Ca2+-activated K+ (BK) channels are expressed in human hepatic stellate cells (HSCs), where they have roles in normal hepatic microcirculation, as well as in portal hypertension in liver cirrhosis through the regulation of contractility in activated HSCs. Nevertheless, whether BK channel activity exerts protective effects against aberrant HSC activation and hepatic fibrosis is unknown. Here, we report that BK channels are expressed in activated primary rat HSCs as well as in a human HSC line. Moreover, whole-cell K+ currents recorded from activated HSCs were markedly increased by exposure to rottlerin, a BK channel-specific activator, but were inhibited by treatment with the BK channel-specific inhibitor, paxilline, suggesting that BK channels are functional in activated HSCs. Overexpression but not downregulation of the BK channel pore-forming alpha subunit, KCNMA1, led to reduced migration and collagen expression in activated HSCs. Consistently, rottlerin treatment suppressed the fibrogenic cell function both in vitro and in CCl4-induced liver fibrosis in vivo. Microarray and pathway analysis, combined with a luciferase reporter assay and western blotting, further showed that rottlerin treatment led to a significant downregulation of the profibrotic TGFβ1/SMAD3 and JAK/STAT3 signaling pathways, both in vitro and in vivo. Our findings not only link BK channel function to profibrotic signaling pathways, but also provide evidence that BK channel activation represents a promising therapeutic strategy for the treatment of liver fibrosis.

Highlights

  • Liver fibrosis is a frequent complication related to chronic liver diseases, and is associated with high morbidity and high mortality worldwide

  • hepatic stellate cells (HSCs) become activated in response to a variety of profibrotic factors, such as transforming growth factor beta 1 (TGFβ1), that are generated during chronic liver injury, and exhibit phenotypes characterized by enhanced aortic smooth muscle actin (ACTA2) expression (Hernandez-Gea and Friedman, 2011; Tsuchida and Friedman, 2017; Shang et al, 2018), collagen generation, contraction, migration, and proliferation (Reynaert et al, 2002; Friedman, 2010; Page et al, 2014; Seki and Schwabe, 2015)

  • We found that KCNMA1 was expressed at both the mRNA and protein level in TGFβ1-activated LX2 cells (Figures 1A,B) that showed robust expression of actin alpha 2 (ACTA2), a marker for activated HSCs (Tsuchida and Friedman, 2017; Shang et al, 2018)

Read more

Summary

Introduction

Liver fibrosis is a frequent complication related to chronic liver diseases, and is associated with high morbidity and high mortality worldwide. HSCs reside in Disse’s space, where they exhibit a quiescent phenotype. HSCs become activated in response to a variety of profibrotic factors, such as transforming growth factor beta 1 (TGFβ1), that are generated during chronic liver injury, and exhibit phenotypes characterized by enhanced aortic smooth muscle actin (ACTA2) expression (Hernandez-Gea and Friedman, 2011; Tsuchida and Friedman, 2017; Shang et al, 2018), collagen generation, contraction, migration, and proliferation (Reynaert et al, 2002; Friedman, 2010; Page et al, 2014; Seki and Schwabe, 2015)

Methods
Results
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