Your browser doesn't support javascript.
loading
Interference with the HNF4-dependent gene regulatory network diminishes ER stress in hepatocytes.
Shah, Anit; Huck, Ian; Duncan, Kaylia; Gansemer, Erica R; Apte, Udayan; Stamnes, Mark A; Rutkowski, D Thomas.
Afiliação
  • Shah A; Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA.
  • Huck I; Department of Pharmacology, Toxicology, and Therapeutics, Kansas University Medical Center, Kansas City, KS.
  • Duncan K; Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA.
  • Gansemer ER; Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA.
  • Apte U; Department of Pharmacology, Toxicology, and Therapeutics, Kansas University Medical Center, Kansas City, KS.
  • Stamnes MA; Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA.
  • Rutkowski DT; Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA.
bioRxiv ; 2023 Feb 09.
Article em En | MEDLINE | ID: mdl-36798396
ABSTRACT
In all eukaryotic cell types, the unfolded protein response (UPR) upregulates factors that promote protein folding and misfolded protein clearance to help alleviate endoplasmic reticulum (ER) stress. Yet ER stress in the liver is uniquely accompanied by the suppression of metabolic genes, the coordination and purpose of which is largely unknown. Here, we used unsupervised machine learning to identify a cluster of correlated genes that were profoundly suppressed by persistent ER stress in the liver. These genes, which encode diverse functions including metabolism, coagulation, drug detoxification, and bile synthesis, are likely targets of the master regulator of hepatocyte differentiation HNF4α. The response of these genes to ER stress was phenocopied by liver-specific deletion of HNF4 α. Strikingly, while deletion of HNF4α exacerbated liver injury in response to an ER stress challenge, it also diminished UPR activation and partially preserved ER ultrastructure, suggesting attenuated ER stress. Conversely, pharmacological maintenance of hepatocyte identity in vitro enhanced sensitivity to stress. Several pathways potentially link HNF4α to ER stress sensitivity, including control of expression of the tunicamycin transporter MFSD2A; modulation of IRE1/XBP1 signaling; and regulation of Pyruvate Dehydrogenase. Together, these findings suggest that HNF4α activity is linked to hepatic ER homeostasis through multiple mechanisms.

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Prognostic_studies Idioma: En Revista: BioRxiv Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Prognostic_studies Idioma: En Revista: BioRxiv Ano de publicação: 2023 Tipo de documento: Article