c-Met confers protection against chronic liver tissue damage and fibrosis progression after bile duct ligation in mice

Gastroenterology. 2009 Jul;137(1):297-308, 308.e1-4. doi: 10.1053/j.gastro.2009.01.068. Epub 2009 Feb 5.

Abstract

Background & aims: The hepatocyte growth factor (HGF)/mesenchymal-epithelial transition factor (c-Met) system is an essential inducer of hepatocyte growth and proliferation. Although a fundamental role for the HGF receptor c-Met has been shown in acute liver regeneration, its cell-specific role in hepatocytes during chronic liver injury and fibrosis progression has not been determined.

Methods: Hepatocyte-specific c-Met knockout mice (c-Met(Delta hepa)) using the Cre-loxP system were studied in a bile duct ligation (BDL) model. Microarray analyses were performed to define HGF/c-Met-dependent gene expression.

Results: Two strategies for c-Met deletion in hepatocytes to generate hepatocyte-specific c-Met knockout mice were tested. Early deletion during embryonic development was lethal, whereas post-natal Cre expression was successful, leading to the generation of viable c-Met(Delta hepa) mice. BDL in these mice resulted in extensive necrosis and lower proliferation rates of hepatocytes. Gene array analysis of c-Met(Delta hepa) mice revealed a significant reduction of anti-apoptotic genes in c-Met-deleted hepatocytes. These findings could be tested functionally because c-Met(Delta hepa) mice showed a stronger apoptotic response after BDL and Jo-2 stimulation. The phenotype was associated with increased expression of proinflammatory cytokines (tumor necrosis factor-alpha and interleukin-6) and an enhanced recruitment of neutrophils. Activation of these mechanisms triggered a stronger profibrogenic response as evidenced by increased transforming growth factor-beta(1), alpha-smooth muscle actin, collagen-1alpha messenger RNA expression, and enhanced collagen-fiber staining in c-Met(Delta hepa) mice.

Conclusions: Our results show that deletion of c-Met in hepatocytes leads to more liver cell damage and fibrosis in a chronic cholestatic liver injury model because c-Met triggers survival signals important for hepatocyte recovery.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Apoptosis* / genetics
  • Cell Proliferation
  • Cholestasis, Extrahepatic / complications*
  • Cholestasis, Extrahepatic / genetics
  • Cholestasis, Extrahepatic / metabolism
  • Cholestasis, Extrahepatic / pathology
  • Chronic Disease
  • Common Bile Duct / surgery
  • Cytokines / metabolism
  • Disease Models, Animal
  • Disease Progression
  • Extracellular Matrix Proteins / metabolism
  • Gene Expression Profiling / methods
  • Hepatic Stellate Cells / metabolism
  • Hepatic Stellate Cells / pathology
  • Hepatitis / metabolism
  • Hepatitis / pathology
  • Hepatocyte Growth Factor / metabolism
  • Hepatocytes / metabolism
  • Hepatocytes / pathology
  • Inflammation Mediators / metabolism
  • Ligation
  • Liver / metabolism*
  • Liver / pathology
  • Liver Cirrhosis / genetics
  • Liver Cirrhosis / metabolism
  • Liver Cirrhosis / pathology
  • Liver Cirrhosis / prevention & control*
  • Mice
  • Mice, Knockout
  • Necrosis
  • Neutrophil Infiltration
  • Oligonucleotide Array Sequence Analysis
  • Proto-Oncogene Proteins c-met / deficiency
  • Proto-Oncogene Proteins c-met / genetics
  • Proto-Oncogene Proteins c-met / metabolism*
  • Time Factors

Substances

  • Cytokines
  • Extracellular Matrix Proteins
  • Inflammation Mediators
  • Hepatocyte Growth Factor
  • Proto-Oncogene Proteins c-met