Abstract

Complement is known to play a role in alcoholic fatty liver disease (AFLD), but the underlying mechanisms are poorly understood, thereby constraining the development of a rational approach for therapeutic intervention in the complement system. C3 deficiency has been shown to impart protective effects against ethanol-induced hepatic steatosis and inflammation. Here we demonstrate a protection effect in wild-type mice by treatment with CR2-Crry, a specific inhibitor of C3 activation. The expression of glycine transfer (t) RNA-derived fragments (Gly-tRFs) is upregulated in ethanol-fed mice and inhibition of Gly-tRFs in vivo decreases chronic ethanol feeding-induced hepatosteatosis without affecting inflammation. The expression of Gly-tRF was downregulated in C3-deficient or CR2-Crry-treated mice, but not in C5-deficient mice; Gly-tRF expression was restored by the C3 activation products C3a or Asp (C3a-des-Arg) via the regulation of CYP2E1. Transcriptome profiling of hepatic tissues showed that Gly-tRF inhibitors upregulate the expression of sirtuin1 (Sirt1) and subsequently affect downstream lipogenesis and β-oxidation pathways. Mechanistically, Gly-tRF interacts with AGO3 to downregulate Sirt1 expression via sequence complementarity in the 3′ UTR. Notably, the expression levels of C3d, CYP2E1 and Gly-tRF are upregulated, whereas Sirt1 is decreased in AFLD patients compared to healthy controls. Collectively, our findings suggest that C3 activation products contribute to hepatosteatosis by regulating the expression of Gly-tRF. Complement inhibition at the C3 activation step and treatment with Gly-tRF inhibitors may be potential and precise therapeutic approaches for AFLD.

Highlights

  • Alcoholic liver disease is a leading cause of morbidity and mortality worldwide

  • We found that component 3 (C3) activation is involved in regulation of both hepatosteatosis and inflammatory responses and contributes to liver ischemia-reperfusion injury (IRI) in Alcoholic fatty liver disease (AFLD) mice

  • U6 was used as an internal control for RNA loading. d AML12 cells were transfected with ANG small interfering RNA (siRNA), and after 12 h treated with 100 mM ethanol

Read more

Summary

Introduction

Alcoholic liver disease is a leading cause of morbidity and mortality worldwide. Alcoholic fatty liver disease (AFLD) is a consequence of excessive alcohol consumption. When patients continue to drink, AFLD progresses to more severe forms of liver injury such as steatohepatitis, fibrosis, cirrhosis, and hepatocellular carcinoma.[1] Excessive ethanol intake is estimated to be the cause of 4.5% of these diseases and 3.8% of all deaths around the world.[2,3] AFLD is characterized by a number of symptoms, such as steatosis and inflammation, which contribute to liver injury. While AFLD has been extensively studied, current understanding of the pathogenesis of AFLD is still limited

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