Abstract

A hallmark of chronic liver injury is fibrosis, with accumulation of extracellular matrix orchestrated by activated hepatic stellate cells (HSCs). Glucocorticoids limit HSC activation in vitro, and tissue glucocorticoid levels are amplified by 11beta‐hydroxysteroid dehydrogenase‐1 (11βHSD1). Although 11βHSD1 inhibitors have been developed for type 2 diabetes mellitus and improve diet‐induced fatty liver in various mouse models, effects on the progression and/or resolution of liver injury and consequent fibrosis have not been characterized. We have used the reversible carbon tetrachloride‐induced model of hepatocyte injury and liver fibrosis to show that in two models of genetic 11βHSD1 deficiency (global, Hsd11b1 –/–, and hepatic myofibroblast‐specific, Hsd11b1 fl/fl/Pdgfrb‐cre) 11βHSD1 pharmacological inhibition in vivo exacerbates hepatic myofibroblast activation and liver fibrosis. In contrast, liver injury and fibrosis in hepatocyte‐specific Hsd11b1 fl/fl/albumin‐cre mice did not differ from that of controls, ruling out 11βHSD1 deficiency in hepatocytes as the cause of the increased fibrosis. In primary HSC culture, glucocorticoids inhibited expression of the key profibrotic genes Acta2 and Col1α1, an effect attenuated by the 11βHSD1 inhibitor [4‐(2‐chlorophenyl‐4‐fluoro‐1‐piperidinyl][5‐(1H‐pyrazol‐4‐yl)‐3‐thienyl]‐methanone. HSCs from Hsd11b1 –/– and Hsd11b1 fl/fl/Pdgfrb‐cre mice expressed higher levels of Acta2 and Col1α1 and were correspondingly more potently activated. In vivo [4‐(2‐chlorophenyl‐4‐fluoro‐1‐piperidinyl][5‐(1H‐pyrazol‐4‐yl)‐3‐thienyl]‐methanone administration prior to chemical injury recapitulated findings in Hsd11b1 –/– mice, including greater fibrosis. Conclusion: 11βHSD1 deficiency enhances myofibroblast activation and promotes initial fibrosis following chemical liver injury; hence, the effects of 11βHSD1 inhibitors on liver injury and repair are likely to be context‐dependent and deserve careful scrutiny as these compounds are developed for chronic diseases including metabolic syndrome and dementia. (Hepatology 2018;67:2167‐2181).

Highlights

  • A Hepatology specific small molecule 11βHSD1 inhibitor, to study the functional role of 11βHSD1 in murine models of toxin-induced liver fibrosis

  • For the first time, that 11βHSD1 deficiency or inhibition promotes MFB activation and liver fibrogenesis in the CCl4 model

  • Macrophage numbers (Fig. 5A-C) and cytokine mRNA levels of Mcp1and Il1 (Fig. 5D-E) were similar in GKO and control mice. Taken together these data suggest that enhanced pro-fibrotic response in GKO mice is not due to exacerbated inflammation or macrophageinduced fibrosis but is more likely to reflect a direct effect on hepatic stellate cell activation pathways

Read more

Summary

Materials and Methods

To induce steatosis and NASH in a model of NAFLD, GKO male mice and age-matched C57Bl/6J mice aged 610 months were fed commercially available diets (all from Dyets, Bethlehem, PA, US): control (Con, 518574), choline deficient (CDD, 518753) or methionine-choline deficient (MCDD, 518810), for 2 weeks as previously described [22]. Blinded hematoxylin and eosin (H&E) stained sections from the acute single dose CCl4 and the chronic CCl4 injury models (at 24h peak fibrosis) were evaluated by a pathologist using 2 separate ordinal scales.; Inflammation was scored using the inflammation component of the NAS scoring system for NASH [24] (0 – no inflammation; 1 - 4 necroinflammatory foci/20x field). Statistical analysis was performed using Graphpad prism or Statistica 7. 2-way ANOVA was used to test for the interaction of genotype/treatment analysis. 2-tailed unpaired Student’s t test or one-way ANOVA was used for the comparison of 2 (ie control versus KO mice) or 3 groups respectively (ie .control, CD, MCD diet comparisons)

Results
Findings
Discussion
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