FOXA3 induction under endoplasmic reticulum stress contributes to non-alcoholic fatty liver disease

J Hepatol. 2021 Jul;75(1):150-162. doi: 10.1016/j.jhep.2021.01.042. Epub 2021 Feb 4.

Abstract

Background & aims: Chronic endoplasmic reticulum (ER) stress in the liver has been shown to play a causative role in non-alcoholic fatty liver disease (NAFLD) progression, yet the underlying molecular mechanisms remain to be elucidated. Forkhead box A3 (FOXA3), a member of the FOX family, plays critical roles in metabolic homeostasis, although its possible functions in ER stress and fatty liver progression are unknown.

Methods: Adenoviral delivery, siRNA delivery, and genetic knockout mice were used to crease FOXA3 gain- or loss-of-function models. Tunicamycin (TM) and a high-fat diet (HFD) were used to induce acute or chronic ER stress in mice. Chromatin immunoprecipiation (ChIP)-seq, luciferase assay, and adenoviral-mediated downstream gene manipulations were performed to reveal the transcriptional axis involved. Key axis protein levels in livers from healthy donors and patients with NAFLD were assessed via immunohistochemical staining.

Results: FOXA3 transcription is specifically induced by XBP1s upon ER stress. FOXA3 exacerbates the excessive lipid accumulation caused by the acute ER-inducer TM, whereas FOXA3 deficiency in hepatocytes and mice alleviates it. Importantly, FOXA3 deficiency in mice reduced diet-induced chronic ER stress, fatty liver, and insulin resistance. In addition, FOXA3 suppression via siRNA or adeno-associated virus delivery ameliorated the fatty liver phenotype in HFD-fed and db/db mice. Mechanistically, ChIP-Seq analysis revealed that FOXA3 directly regulates Period1 (Per1) transcription, which in turn promotes the expression of lipogenic genes, including Srebp1c, thus enhancing lipid synthesis. Of pathophysiological significance, FOXA3, PER1, and SREBP1c levels were increased in livers of obese mice and patients with NAFLD.

Conclusion: The present study identified FOXA3 as the bridging molecule that links ER stress and NAFLD progression. Our results highlighted the role of the XBP1s-FOXA3-PER1/Srebp1c transcriptional axis in the development of NAFLD and identified FOXA3 as a potential therapeutic target for fatty liver disease.

Lay summary: The molecular mechanisms linking endoplasmic reticulum stress to non-alcoholic fatty liver disease (NAFLD) progression remain undefined. Herein, via in vitro and in vivo analysis, we identified Forkhead box A3 (FOXA3) as a key bridging molecule. Of pathophysiological significance, FOXA3 protein levels were increased in livers of obese mice and patients with NAFLD, indicating that FOXA3 could be a potential therapeutic target in fatty liver disease.

Keywords: Endoplasmic reticulum stress; Hepatic steatosis; Lipogenesis; Non-alcoholic fatty liver disease; Triglyceride.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Drug Discovery
  • Endoplasmic Reticulum Stress*
  • Hepatocyte Nuclear Factor 3-gamma / metabolism*
  • Hepatocytes / metabolism
  • Humans
  • Lipogenesis / genetics
  • Mice
  • Mice, Knockout
  • Non-alcoholic Fatty Liver Disease / metabolism
  • Period Circadian Proteins / metabolism
  • Signal Transduction
  • Sterol Regulatory Element Binding Protein 1 / metabolism
  • X-Box Binding Protein 1 / metabolism

Substances

  • FOXA3 protein, human
  • Foxa3 protein, mouse
  • PER1 protein, human
  • Per1 protein, mouse
  • Period Circadian Proteins
  • SREBF1 protein, human
  • Srebf1 protein, mouse
  • Sterol Regulatory Element Binding Protein 1
  • X-Box Binding Protein 1
  • XBP1 protein, human
  • Xbp1 protein, mouse
  • Hepatocyte Nuclear Factor 3-gamma