UNI-MB - logo
UMNIK - logo
 
E-viri
Celotno besedilo
Recenzirano Odprti dostop
  • Histone acetylation of bile...
    Garrido, Amanda; Kim, Eunjeong; Teijeiro, Ana; Sánchez Sánchez, Paula; Gallo, Rosa; Nair, Ajay; Matamala Montoya, María; Perna, Cristian; Vicent, Guillermo P.; Muñoz, Javier; Campos-Olivas, Ramón; Melms, Johannes C.; Izar, Benjamin; Schwabe, Robert F.; Djouder, Nabil

    Journal of hepatology, 04/2022, Letnik: 76, Številka: 4
    Journal Article

    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. 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. 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. 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. 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. Display omitted •MCRS1 loss in hepatocytes deregulates the expression of BA transporters, causing perturbation of BA flow and cirrhosis.•Re-expression of the BA transporter NTCP in MCRS1-depleted hepatocytes restores the BA flow and prevents cirrhosis.•Accumulation of BAs in liver sinusoids activates liver fibroblasts via FXR leading to fibrosis.•MCRS1 loss increases histone acetylation of BA transporter genes, altering their expression levels.•MCRS1 and NTCP expression correlates with fibrotic marks in human cirrhosis samples.