PARP1-mediated PPARα poly(ADP-ribosyl)ation suppresses fatty acid oxidation in non-alcoholic fatty liver disease

J Hepatol. 2017 May;66(5):962-977. doi: 10.1016/j.jhep.2016.11.020. Epub 2016 Dec 12.

Abstract

Background & aims: PARP1 is a key mediator of cellular stress responses and critical in multiple physiological and pathophysiological processes of cells. However, whether it is involved in the pathogenesis of non-alcoholic fatty liver disease (NAFLD) remains elusive.

Methods: We analysed PARP1 activity in the liver of mice on a high fat diet (HFD), and samples from NAFLD patients. Gain- or loss-of-function approaches were used to investigate the roles and mechanisms of hepatic PARP1 in the pathogenesis of NAFLD.

Results: PARP1 is activated in fatty liver of HFD-fed mice. Pharmacological or genetic manipulations of PARP1 are sufficient to alter the HFD-induced hepatic steatosis and inflammation. Mechanistically we identified peroxisome proliferator-activated receptor α (PPARα) as a substrate of PARP1-mediated poly(ADP-ribosyl)ation. This poly(ADP-ribosyl)ation of PPARα inhibits its recruitment to target gene promoters and its interaction with SIRT1, a key regulator of PPARα signaling, resulting in suppression of fatty acid oxidation upregulation induced by fatty acids. Moreover, we show that PARP1 is a transcriptional repressor of PPARα gene in human hepatocytes, and its activation suppresses the ligand (fenofibrate)-induced PPARα transactivation and target gene expression. Importantly we demonstrate that liver biopsies of NAFLD patients display robust increases in PARP activity and PPARα poly(ADP-ribosyl)ation levels.

Conclusions: Our data indicate that PARP1 is activated in fatty liver, which prevents maximal activation of fatty acid oxidation by suppressing PPARα signaling. Pharmacological inhibition of PARP1 may alleviate PPARα suppression and therefore have therapeutic potential for NAFLD.

Lay summary: PARP1 is activated in the non-alcoholic fatty liver of mice and patients. Inhibition of PARP1 activation alleviates lipid accumulation and inflammation in fatty liver of mice.

Keywords: High fat diet; Non-alcoholic fatty liver disease; PPARα; Poly(ADP-ribose) polymerase 1; Transcriptional regulation.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Diet, High-Fat
  • Fatty Acids / metabolism*
  • Hep G2 Cells
  • Hepatocytes / metabolism
  • Humans
  • Male
  • Mice
  • Mice, Inbred C57BL
  • Non-alcoholic Fatty Liver Disease / metabolism*
  • Oxidation-Reduction
  • PPAR alpha / metabolism*
  • Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha / physiology
  • Poly (ADP-Ribose) Polymerase-1 / physiology*
  • Poly Adenosine Diphosphate Ribose / metabolism*
  • Sirtuin 1 / physiology

Substances

  • Fatty Acids
  • PPAR alpha
  • PPARGC1A protein, human
  • Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha
  • Poly Adenosine Diphosphate Ribose
  • PARP1 protein, human
  • Poly (ADP-Ribose) Polymerase-1
  • SIRT1 protein, human
  • Sirtuin 1