investigator_user investigator user funding collaborators pending menu bell message arrow_up arrow_down filter layers globe marker add arrow close download edit facebook info linkedin minus plus save share search sort twitter remove user-plus user-minus
  • Project leads
  • Collaborators

Prohibitin 1 in Liver Injury and Cancer

Shelly Chi-Loo Lu

1 Collaborator(s)

Funding source

National Cancer Institute (NIH)
Prohibitin 1 (PHB1) is a highly conserved, ubiquitously expressed protein that participates in diverse processes including mitochondrial chaperone, growth and apoptosis. The role of PHB1 in vivo is unclear as embryonic deletion is lethal. We reported that mice lacking methionine adenosyltransferase 1A (MAT1A) have reduced PHB1 expression at the protein level, impaired mitochondrial function, and spontaneously develop steatohepatitis and hepatocellular carcinoma (HCC). To see if reduced PHB1 expression can contribute to the Mat1a knockout (KO) phenotype, we generated liver-specific Phb1 KO mice. At 3 weeks, liver-specific Phb1 KO mice exhibit biochemical and histologic liver injury. Immunohistochemistry revealed oxidative stress, fibrosis, hepatocyte dysplasia, and increased staining of preneoplastic markers. Mitochondrial function is impaired. Phb1 KO mice are sensitized to multiple forms of liver injury but the key mechanism appears to be independent of its role as a mitochondrial chaperone. Our preliminary data show PHB1 deficiency leads to higher total and nuclear histone deacetylase 4 (HDAC4) expression and activity. Interestingly, inhibiting HDAC activity prevented increased cell death in Phb1 KO hepatocytes induced by a variety of toxicants. We also found PHB1 protein level falls in cholestatic liver injury in mouse and humans, suggesting this could further perpetuate liver damage. Many Phb1 KO mice develop multifocal HCC by 35 weeks. Our preliminary data also support PHB1 can directly influence genes implicated in hepatocarcinogenesis. Two genes highly up-regulated in 4-week-old male KO mice livers are H19 and insulin-like growth factor 2 (IGF2). Acute knockdown of PHB1 in murine non-transformed AML12 cells raised cyclin D1, H19 and IGF2 expression, increased E2F binding to the cyclin D1 promoter, and proliferation. The opposite occurred with PHB1 overexpression. These results support PHB1 as a tumor suppressor in hepatocytes. The current application is to examine these highly novel areas and define the role of PHB1 in liver injury and HCC. Three specific aims are proposed: 1) examine the role of PHB1 in liver injury, 2) examine how PHB1 influences HCC development, and 3) determine if PHB1 down-regulation contributes to mitochondrial dysfunction, liver injury and HCC formation in the Mat1a KO mice. Our main hypotheses are 1) PHB1 protein stability falls during cholestatic liver injury and this is part of the mechanism of injury, 2) PHB1 regulates hepatocyte death via HDAC-mediated epigenetic changes, 3) PHB1 regulates HDAC4, H19 and IGF2 expression directly and their induction contribute to HCC formation in the liver-specific Phb1 KO mice, and 4) decreased PHB1 expression contributes to impaired mitochondrial function and HCC formation in the Mat1a KO mice. The application is hypothesis-driven and mechanistic and represents a new area of investigation as little is known of the biological functions of PHB in the liver. The ultimate goal is to translate results from the laboratory to bedside to optimize PHB1 function, which is essential to prevent liver injury and HCC, topics that are highly relevant to public health.

Related projects