Your browser doesn't support javascript.
loading
Rapid Recapitulation of Nonalcoholic Steatohepatitis upon Loss of Host Cell Factor 1 Function in Mouse Hepatocytes
Minocha, Shilpi; Villeneuve, Dominic; Praz, Viviane; Moret, Catherine; Lopes, Maykel; Pinatel, Danièle; Rib, Leonor; Guex, Nicolas; Herr, Winship.
Affiliation
  • Minocha S; Center for Integrative Genomics, Génopode, University of Lausanne, Lausanne, Switzerland.
  • Villeneuve D; Center for Integrative Genomics, Génopode, University of Lausanne, Lausanne, Switzerland.
  • Praz V; Center for Integrative Genomics, Génopode, University of Lausanne, Lausanne, Switzerland.
  • Moret C; Swiss Institute of Bioinformatics, Génopode, University of Lausanne, Lausanne, Switzerland.
  • Lopes M; Center for Integrative Genomics, Génopode, University of Lausanne, Lausanne, Switzerland.
  • Pinatel D; Center for Integrative Genomics, Génopode, University of Lausanne, Lausanne, Switzerland.
  • Rib L; Center for Integrative Genomics, Génopode, University of Lausanne, Lausanne, Switzerland.
  • Guex N; Center for Integrative Genomics, Génopode, University of Lausanne, Lausanne, Switzerland.
  • Herr W; Swiss Institute of Bioinformatics, Génopode, University of Lausanne, Lausanne, Switzerland.
Mol Cell Biol ; 39(5)2019 02 15.
Article in En | MEDLINE | ID: mdl-30559308
ABSTRACT
Host-cell factor 1 (HCF-1), encoded by the ubiquitously expressed X-linked gene Hcfc1, is an epigenetic coregulator important for mouse development and cell proliferation, including during liver regeneration. We used a hepatocyte-specific inducible Hcfc1 knock-out allele (called Hcfc1hepKO), to induce HCF-1 loss in hepatocytes of hemizygous Hcfc1hepKO/Y males by four days. In heterozygous Hcfc1hepKO/+ females, owing to random X-chromosome inactivation, upon Hcfc1hepKO allele induction, a 50/50 mix of HCF-1 positive and negative hepatocyte clusters is engineered. The livers with Hcfc1hepKO/Y hepatocytes displayed a 21-24-day terminal non-alcoholic fatty liver (NAFL) followed by non-alcoholic steatohepatitis (NASH) disease progression typical of severe NAFL disease (NAFLD). In contrast, in livers with heterozygous Hcfc1hepKO/+ hepatocytes, HCF-1-positive hepatocytes replaced HCF-1-negative hepatocytes and revealed only mild-NAFL development. Loss of HCF-1 led to loss of PGC1α protein, probably owing to its destabilization, and deregulation of gene expression particularly of genes involved in mitochondrial structure and function, likely explaining the severe Hcfc1 hepKO/Y liver pathology. Thus, HCF-1 is essential for hepatocyte function, likely playing both transcriptional and non-transcriptional roles. These genetically-engineered loss-of-HCF-1 mice can be used to study NASH as well as NAFLD resolution.
Subject(s)

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Host Factor 1 Protein / Host Cell Factor C1 / Non-alcoholic Fatty Liver Disease Type of study: Prognostic_studies Limits: Animals Language: En Journal: Mol Cell Biol Year: 2019 Document type: Article Affiliation country: Switzerland

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Host Factor 1 Protein / Host Cell Factor C1 / Non-alcoholic Fatty Liver Disease Type of study: Prognostic_studies Limits: Animals Language: En Journal: Mol Cell Biol Year: 2019 Document type: Article Affiliation country: Switzerland