[HTML][HTML] Loss of c-Met accelerates development of liver fibrosis in response to CCl4 exposure through deregulation of multiple molecular pathways

JU Marquardt, D Seo, LE Gómez-Quiroz… - … et Biophysica Acta (BBA …, 2012 - Elsevier
JU Marquardt, D Seo, LE Gómez-Quiroz, K Uchida, MC Gillen, M Kitade, P Kaposi-Novak…
Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease, 2012Elsevier
HGF/c-Met signaling plays a pivotal role in hepatocyte survival and tissue remodeling during
liver regeneration. HGF treatment accelerates resolution of fibrosis in experimental animal
models. Here, we utilized Metfl/fl; Alb-Cre+/− conditional knockout mice and a carbon
tetrachloride (CCl4)-induced liver fibrosis model to formally address the role of c-Met
signaling in hepatocytes in the context of chronic tissue injury. Histological changes during
injury (4weeks) and healing phase (4weeks) were monitored by immunohistochemistry; …
HGF/c-Met signaling plays a pivotal role in hepatocyte survival and tissue remodeling during liver regeneration. HGF treatment accelerates resolution of fibrosis in experimental animal models. Here, we utilized Metfl/fl;Alb-Cre+/− conditional knockout mice and a carbon tetrachloride(CCl4)-induced liver fibrosis model to formally address the role of c-Met signaling in hepatocytes in the context of chronic tissue injury. Histological changes during injury (4weeks) and healing phase (4weeks) were monitored by immunohistochemistry; expression levels of selected key fibrotic molecules were evaluated by western blotting, and time-dependent global transcriptomic changes were examined using a microarray platform. Loss of hepatocyte c-Met signaling altered hepatic microenvironment and aggravated hepatic fibrogenesis. Greater liver damage was associated with decreased hepatocyte proliferation, excessive stellate cell activation and rapid dystrophic calcification of necrotic areas. Global transcriptome analysis revealed a broad impact of c-Met on critical signaling pathways associated with fibrosis. Loss of hepatocyte c-Met caused a strong deregulation of chemotactic and inflammatory signaling (MCP-1, RANTES, Cxcl10) in addition to modulation of genes involved in reorganization of the cytoskeletal network (Actb, Tuba1a, Tuba8), intercellular communications and adhesion (Adam8, Icam1, Itgb2), control of cell proliferation (Ccng2, Csnk2a, Cdc6, cdk10), DNA damage and stress response (Rad9, Rad52, Ercc4, Gsta1 and 2, Jun). Our study demonstrates that deletion of c-Met receptor in hepatocytes results in pronounced changes in hepatic metabolism and microenvironment, and establishes an essential role for c-Met in maintaining the structural integrity and adaptive plasticity of the liver under adverse conditions.
Elsevier