Abstract

Non-alcoholic fatty liver disease (NAFLD) is a major growing worldwide health problem. We previously reported that interruption of the enterohepatic circulation of bile acids using a non-absorbable apical sodium-dependent bile acid transporter inhibitor (ASBTi; SC-435) reduced the development of NAFLD in high fat diet fed mice. However, the ability of ASBTi treatment to impact the progression of NAFLD to non-alcoholic steatohepatitis (NASH) and fibrosis in a diet-induced mouse model remains untested. In the current study, we assessed whether ASBTi treatment is hepatoprotective in the choline-deficient, L-amino acid-defined (CDAA) diet model of NASH-induced fibrosis.Methods: Male C57Bl/6 mice were fed with: (A) choline-sufficient L-amino acid-defined diet (CSAA) (31 kcal% fat), (B) CSAA diet plus ASBTi (SC-435; 60 ppm), (C) CDAA diet, or (D) CDAA diet plus ASBTi. Body weight and food intake were monitored. After 22 weeks on diet, liver histology, cholesterol and triglyceride levels, and gene expression were measured. Fecal bile acid and fat excretion were measured, and intestinal fat absorption was determined using the sucrose polybehenate method.Results: ASBTi treatment reduced bodyweight gain in mice fed either the CSAA or CDAA diet, and prevented the increase in liver to body weight ratio observed in CDAA-fed mice. ASBTi significantly reduced hepatic total cholesterol levels in both CSAA and CDAA-fed mice. ASBTi-associated significant reductions in hepatic triglyceride levels and histological scoring for NAFLD activity were observed in CSAA but not CDAA-fed mice. These changes correlated with measurements of intestinal fat absorption, which was significantly reduced in ASBTi-treated mice fed the CSAA (85 vs. 94%, P < 0.001) but not CDAA diet (93 vs. 93%). As scored by Ishak staging of Sirius red stained liver sections, no hepatic fibrosis was evident in the CSAA diet mice. The CDAA diet-fed mice developed hepatic fibrosis, which was increased by the ASBTi.Conclusions: ASBT inhibition reduced intestinal fat absorption, bodyweight gain and hepatic steatosis in CSAA diet-fed mice. The effects of the ASBTi on steatosis and fat absorption were attenuated in the context of dietary choline-deficiency. Inhibition of intestinal absorption of fatty acids may be involved in the therapeutic effects of ASBTi treatment.

Highlights

  • Parallel to the global rise in obesity, the disease burden related to non-alcoholic fatty liver disease (NAFLD) is emerging as a major worldwide problem

  • Interruption of the enterohepatic circulation of bile acids was confirmed by gene expression measurements, with increases in hepatic cholesterol 7alpha-hydroxylase (Cyp7a1) mRNA as a marker of bile acid synthesis, and colonic intestinal bile acidbinding protein (Ibabp) mRNA as a marker of colon bile acid exposure (Figures 1C,D) in both the choline-sufficient L-amino acid-defined (CSAA) and choline-deficient L-amino aciddefined (CDAA) dietfed mice

  • Inhibiting the Apical sodium-dependent bile acid transporter (ASBT) in mice has been shown to increase the proportion of cholic acid (CA) plus its bacterial dehydroxylation product deoxycholic acid (DCA) and to reduce the proportion of 6-hydroxylated bile acid species, including alpha-muricholic acid, beta-muricholic acid, and the bacterial product omega-muricholic acid [10, 11, 29]

Read more

Summary

Introduction

Parallel to the global rise in obesity, the disease burden related to non-alcoholic fatty liver disease (NAFLD) is emerging as a major worldwide problem. Bile acid synthesis and enterohepatic cycling are tightly regulated to maintain a relatively constant whole-body bile acid pool size and restrict the systemic distribution of bile acids After their secretion along with bile into the duodenum, about 95% of the bile acids are reabsorbed by the apical sodium-dependent bile acid transporter (ASBT; called the ileal bile acid transporter, IBAT) in the distal small intestine, thereby limiting their flux into the colon. The intestinal microbiota harbor enzymes to deconjugate and convert primary bile acids into secondary bile acids, changing the bile acid pool size, composition, and physicochemical properties [6] These changes alter signaling through FXR and other bile acidactivated receptors, and modulates the metabolic response to bile acids [7]

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