Abstract

Transforming growth factor-β (TGF-β) is a potent cytokine that promotes the development of fibrogenic cells, stimulates the expression of fibrosis-related genes, and consequently results in hepatic fibrogenesis. The involvement of miRNAs in this process remains largely unknown. We showed that miR-122 was substantially expressed in hepatic stellate cells (HSCs) and fibroblasts, the major sources of fibrogenic cells in liver tissues. Notably, exposure to TGF-β led to significant downregulation of miR-122. Furthermore, reintroduction of miR-122 suppressed TGF-β-induced expression of fibrosis-related genes, including alpha smooth muscle actin (α-SMA), fibronectin 1 (FN1) and α1 type I collagen (COL1A1), in HSCs and fibroblasts. Subsequent mechanism investigations revealed that miR-122 directly inhibited FN1 expression by binding to its 3'-untranslated region and indirectly reduced the transcription of α-SMA and COL1A1 by inhibiting the expression of serum response factor (SRF), a key transcription factor that mediated the activation of fibrogenic cells. Further in vivo studies disclosed that intravenous injection of miR-122-expressing lentivirus successfully increased miR-122 level and reduced the amount of collagen fibrils, FN1 and SRF in the livers of CCl4-treated mice. These findings disclose a novel TGF-β-miR-122-FN1/SRF signaling cascade and its implication in hepatic fibrogenesis, and suggest miR-122 as a promising molecular target for anti-fibrosis therapy.

Highlights

  • Hepatic fibrosis is an outcome of the pathological response to chronic liver injuries and is characterized by the increased deposition and altered composition of extracellular matrix (ECM) [1]

  • Subsequent mechanism investigations revealed that miR-122 directly inhibited fibronectin 1 (FN1) expression by binding to its 3’-untranslated region and indirectly reduced the transcription of α-SMA and COL1A1 by inhibiting the expression of serum response factor (SRF), a key transcription factor that mediated the activation of fibrogenic cells

  • ECM is changed from a type IV collagen-rich composition to a type I collagen- and fibronectin 1 (FN1)rich composition, which distorts the architecture of the liver, and leads to hepatic cirrhosis and hepatocellular cancer (HCC) [1]

Read more

Summary

Introduction

Hepatic fibrosis is an outcome of the pathological response to chronic liver injuries and is characterized by the increased deposition and altered composition of extracellular matrix (ECM) [1]. The activation of fibrogenic cells is critical for hepatic fibrosis. In response to chronic liver injuries, the expression and secretion of transforming growth factor-β (TGF-β) significantly increases. TGF-β is the most potent fibrogenic cytokine that activates the HSCs and fibroblasts to express fibrosis-related genes, including alpha smooth muscle actin (α-SMA), FN1 and α1 type I collagen (COL1A1), and promotes www.impactjournals.com/oncotarget hepatic fibrogenesis [1, 2]. Α-SMA, besides as a key molecular marker for the activated fibrogenic cells, promotes contractile force and ECM stiffness [1, 2]. Great efforts have been dedicated to unravel the molecular mechanisms underlying liver fibrogenesis, whether microRNAs (miRNAs) may regulate liver fibrogenesis is still poorly understood

Methods
Results
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.