Histone acetylation of bile acid transporter genes plays a critical role in cirrhosis

J Hepatol. 2022 Apr;76(4):850-861. doi: 10.1016/j.jhep.2021.12.019. Epub 2021 Dec 25.

Abstract

Background & aims: Owing to the lack of genetic animal models that adequately recreate key clinical characteristics of cirrhosis, the molecular pathogenesis of cirrhosis has been poorly characterized, and treatments remain limited. Hence, we aimed to better elucidate the pathological mechanisms of cirrhosis using a novel murine model.

Methods: We report on the first murine genetic model mimicking human cirrhosis induced by hepatocyte-specific elimination of microspherule protein 1 (MCRS1), a member of non-specific lethal (NSL) and INO80 chromatin-modifier complexes. Using this genetic tool with other mouse models, cell culture and human samples, combined with quantitative proteomics, single nuclei/cell RNA sequencing and chromatin immunoprecipitation assays, we investigated mechanisms of cirrhosis.

Results: MCRS1 loss in mouse hepatocytes modulates the expression of bile acid (BA) transporters - with a pronounced downregulation of Na+-taurocholate cotransporting polypeptide (NTCP) - concentrating BAs in sinusoids and thereby activating hepatic stellate cells (HSCs) via the farnesoid X receptor (FXR), which is predominantly expressed in human and mouse HSCs. Consistently, re-expression of NTCP in mice reduces cirrhosis, and genetic ablation of FXR in HSCs suppresses fibrotic marks in mice and in vitro cell culture. Mechanistically, deletion of a putative SANT domain from MCRS1 evicts histone deacetylase 1 from its histone H3 anchoring sites, increasing histone acetylation of BA transporter genes, modulating their expression and perturbing BA flow. Accordingly, human cirrhosis displays decreased nuclear MCRS1 and NTCP expression.

Conclusions: Our data reveal a previously unrecognized function of MCRS1 as a critical histone acetylation regulator, maintaining gene expression and liver homeostasis. MCRS1 loss induces acetylation of BA transporter genes, perturbation of BA flow, and consequently, FXR activation in HSCs. This axis represents a central and universal signaling event in cirrhosis, which has significant implications for cirrhosis treatment.

Lay summary: By genetic ablation of MCRS1 in mouse hepatocytes, we generate the first genetic mouse model of cirrhosis that recapitulates human features. Herein, we demonstrate that the activation of the bile acid/FXR axis in liver fibroblasts is key in cirrhosis development.

Keywords: Bile acid transporter; Bile acids; Cirrhosis; FXR; Fibroblasts; Hepatic stellate cells; Histone acetylation; Liver fibrosis; MCRS1; NTCP.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Acetylation
  • Animals
  • Bile Acids and Salts / metabolism
  • Carrier Proteins
  • Histones* / metabolism
  • Liver / pathology
  • Liver Cirrhosis / pathology
  • Membrane Glycoproteins
  • Mice
  • RNA-Binding Proteins* / genetics
  • RNA-Binding Proteins* / metabolism
  • Receptors, Cytoplasmic and Nuclear* / genetics
  • Receptors, Cytoplasmic and Nuclear* / metabolism

Substances

  • Bile Acids and Salts
  • Carrier Proteins
  • Histones
  • Mcrs1 protein, mouse
  • Membrane Glycoproteins
  • RNA-Binding Proteins
  • Receptors, Cytoplasmic and Nuclear
  • bile acid binding proteins