Insulin-like growth factor-1 attenuates oxidative stress-induced hepatocyte premature senescence in liver fibrogenesis via regulating nuclear p53-progerin interaction

Cell Death Dis. 2019 Jun 6;10(6):451. doi: 10.1038/s41419-019-1670-6.

Abstract

Stress-induced premature senescence (SIPS), a state of cell growth arrest due to various stimuli, is implicated in the pathogeneses of hepatic fibrogenesis. Progerin, a permanently farnesylated mutant lamin A protein, likely leads to premature senescence to influent liver diseases. The previous reports showed that activation of insulin-like growth factor-1 (IGF-1) signaling could enhance cell longevity and attenuate liver fibrosis. However, the underlying mechanisms about hepatocyte premature senility in liver fibrosis, and how IGF-1 regulates cell premature aging and fibrogenesis, remain poorly understood. In the present study, we found the augment of hepatocyte oxidation and premature aging, along with the decrease of plasm IGF-1 level in patients with liver fibrosis and CCl4-induced liver injury rat models. Nevertheless, IGF-1 gene transfer to CCl4 rats to overexpress intrahepatic IGF-1 relieved hepatocyte oxidative stress and premature senescence, which was likely mediated by the p53/progerin pathway, to improve hepatic steatosis and fibrogenesis. In vitro, H2O2 caused abnormal accumulation of progerin in nuclear and activation of nuclear p53-progerin interaction to trigger primary rat hepatocyte premature senescence through the p21-independent pathway; while these effects were rescued by prolonged exogenous IGF-1 or the IGF-1 adenovirus vector. Furthermore, the IGF-1 adenovirus vector, transfected to H2O2-treated hepatocytes, reversed oxidative stress-induced premature senescence via enhancing cytoplasmic AKT1-p53 interaction and subsequently inhibiting nuclear p53-progerin interaction. Consequently, our data illuminate a novel role of IGF-1 in regulating stress-induced hepatocyte premature senescence in liver fibrosis: prolonged IGF-1 relieves oxidative stress-initiated hepatocyte premature senescence via inhibition of nuclear p53-progerin interaction to ameliorate hepatic steatosis and fibrogenesis.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Carbon Tetrachloride / toxicity
  • Cell Nucleus / genetics
  • Cell Nucleus / metabolism
  • Cellular Senescence / drug effects
  • Cellular Senescence / genetics*
  • Disease Models, Animal
  • Fatty Liver / chemically induced
  • Fatty Liver / genetics
  • Fatty Liver / pathology
  • Hepatocytes / cytology
  • Hepatocytes / drug effects
  • Hepatocytes / metabolism*
  • Hepatocytes / physiology
  • Humans
  • Hydrogen Peroxide / metabolism
  • Insulin-Like Growth Factor I / genetics
  • Insulin-Like Growth Factor I / metabolism*
  • Lamin Type A / chemistry
  • Lamin Type A / genetics
  • Lamin Type A / metabolism*
  • Liver / metabolism
  • Liver / pathology
  • Liver Cirrhosis / chemically induced
  • Liver Cirrhosis / genetics
  • Liver Cirrhosis / metabolism*
  • Liver Cirrhosis / pathology
  • Oxidative Stress
  • Protein Prenylation
  • Proto-Oncogene Proteins c-akt / metabolism
  • Rats
  • Signal Transduction / genetics
  • Tumor Suppressor Protein p53 / genetics
  • Tumor Suppressor Protein p53 / metabolism*
  • Up-Regulation / genetics

Substances

  • IGF1 protein, human
  • Lamin Type A
  • Tumor Suppressor Protein p53
  • insulin-like growth factor-1, rat
  • prelamin A
  • Insulin-Like Growth Factor I
  • Hydrogen Peroxide
  • Carbon Tetrachloride
  • Proto-Oncogene Proteins c-akt