Abstract

Liraglutide, a glucagon-like peptide-1 receptor agonist (GLP-1RA), has been demonstrated to alleviate non-alcoholic fatty liver disease (NAFLD). However, the underlying mechanism has not been fully elucidated. Increasing evidence suggests that autophagy is involved in the pathogenesis of hepatic steatosis. In this study, we examined whether liraglutide could alleviate hepatic steatosis through autophagy-dependent lipid degradation and investigated the underlying mechanisms. Herein, the effects of liraglutide on NAFLD were evaluated in a high-fat diet (HFD)-induced mouse model of NAFLD as well as in mouse primary and HepG2 hepatocytes exposed to palmitic acid (PA). The expression of the GLP-1 receptor (GLP-1R) was measured in vivo and in vitro. Oil red O staining was performed to detect lipid accumulation in hepatocytes. Electron microscopy was used to observe the morphology of autophagic vesicles and autolysosomes. Autophagic flux activity was measured by infecting HepG2 cells with mRFP-GFP-LC3 adenovirus. The roles of GLP-1R and transcription factor EB (TFEB) in autophagy-lysosomal activation were explored using small interfering RNA. Liraglutide treatment alleviated hepatic steatosis in vivo and in vitro. In models of hepatic steatosis, microtubule-associated protein 1B light chain-3-II (LC3-II) and SQSTM1/P62 levels were elevated in parallel to blockade of autophagic flux. Liraglutide treatment restored autophagic activity by improving lysosomal function. Furthermore, treatment with autophagy inhibitor chloroquine weakened liraglutide-induced autophagy activation and lipid degradation. TFEB has been identified as a key regulator of lysosome biogenesis and autophagy. The protein levels of nuclear TFEB and its downstream targets CTSB and LAMP1 were decreased in hepatocytes treated with PA, and these decreases were reversed by liraglutide treatment. Knockdown of TFEB expression compromised the effects of liraglutide on lysosome biogenesis and hepatic lipid accumulation. Mechanistically, GLP-1R expression was decreased in HFD mouse livers as well as PA-stimulated hepatocytes, and liraglutide treatment reversed the downregulation of GLP-1R expression in vivo and in vitro. Moreover, GLP-1R inhibition could mimic the effect of the TFEB downregulation-mediated decrease in lysosome biogenesis. Thus, our findings suggest that liraglutide attenuated hepatic steatosis via restoring autophagic flux, specifically the GLP-1R-TFEB-mediated autophagy-lysosomal pathway.

Highlights

  • Non-alcoholic fatty liver disease (NAFLD), characterized by excessive triglyceride (TG) accumulation in the liver, is commonly associated with insulin resistance, obesity, diabetes, and cardiovascular disease (Arab et al, 2018)

  • Liraglutide treatment induced a transient reduction of food intake during the first 2 weeks, no significant differences were found between the high-fat diet (HFD) groups treated with or without liraglutide during 3– 4 weeks (Supplementary Figure S1)

  • Given that Transcription factor EB (TFEB) plays a critical role in the regulation of autophagy and lysosome biogenesis, we evaluated whether liraglutide alleviated steatosis by promoting

Read more

Summary

Introduction

Non-alcoholic fatty liver disease (NAFLD), characterized by excessive triglyceride (TG) accumulation in the liver, is commonly associated with insulin resistance, obesity, diabetes, and cardiovascular disease (Arab et al, 2018). The prevalence of NAFLD is estimated to be approximately 10–35% in the general population, making it the most common liver disease worldwide (Vernon et al, 2011). There is an urgent need to develop new preventive and therapeutic strategies to alleviate NAFLD. Liraglutide, a glucagon-like peptide-1 receptor agonist (GLP1RA), exhibits beneficial effects on weight loss, cardiovascular function, and NAFLD in addition to its glucose-lowering effect, especially in patients with obesity (Drucker, 2018; Muller et al, 2019). Previous studies suggested that liraglutide could alleviate high-fat diet (HFD)−induced hepatic lipid accumulation in a weight loss-independent manner (Lyu et al, 2020). The mechanism underlying the effects of liraglutide on NAFLD remains unclear

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