Cargando…

Interference with the HNF4-dependent gene regulatory network diminishes endoplasmic reticulum stress in hepatocytes

BACKGROUND: 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 gene...

Descripción completa

Detalles Bibliográficos
Autores principales: Shah, Anit, Huck, Ian, Duncan, Kaylia, Gansemer, Erica R., Liu, Kaihua, Adajar, Reed C., Apte, Udayan, Stamnes, Mark A., Rutkowski, D. Thomas
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Lippincott Williams & Wilkins 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10578741/
https://www.ncbi.nlm.nih.gov/pubmed/37820274
http://dx.doi.org/10.1097/HC9.0000000000000278
_version_ 1785121574976749568
author Shah, Anit
Huck, Ian
Duncan, Kaylia
Gansemer, Erica R.
Liu, Kaihua
Adajar, Reed C.
Apte, Udayan
Stamnes, Mark A.
Rutkowski, D. Thomas
author_facet Shah, Anit
Huck, Ian
Duncan, Kaylia
Gansemer, Erica R.
Liu, Kaihua
Adajar, Reed C.
Apte, Udayan
Stamnes, Mark A.
Rutkowski, D. Thomas
author_sort Shah, Anit
collection PubMed
description BACKGROUND: 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 are largely unknown. METHODS: Here, we combined in silico machine learning, in vivo liver-specific deletion of the master regulator of hepatocyte differentiation HNF4α, and in vitro manipulation of hepatocyte differentiation state to determine how the UPR regulates hepatocyte identity and toward what end. RESULTS: Machine learning identified 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 HNF4α. 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. CONCLUSIONS: Together, our findings suggest that the UPR regulates hepatocyte identity through HNF4α to protect ER homeostasis even at the expense of liver function.
format Online
Article
Text
id pubmed-10578741
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Lippincott Williams & Wilkins
record_format MEDLINE/PubMed
spelling pubmed-105787412023-10-17 Interference with the HNF4-dependent gene regulatory network diminishes endoplasmic reticulum stress in hepatocytes Shah, Anit Huck, Ian Duncan, Kaylia Gansemer, Erica R. Liu, Kaihua Adajar, Reed C. Apte, Udayan Stamnes, Mark A. Rutkowski, D. Thomas Hepatol Commun Original Article BACKGROUND: 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 are largely unknown. METHODS: Here, we combined in silico machine learning, in vivo liver-specific deletion of the master regulator of hepatocyte differentiation HNF4α, and in vitro manipulation of hepatocyte differentiation state to determine how the UPR regulates hepatocyte identity and toward what end. RESULTS: Machine learning identified 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 HNF4α. 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. CONCLUSIONS: Together, our findings suggest that the UPR regulates hepatocyte identity through HNF4α to protect ER homeostasis even at the expense of liver function. Lippincott Williams & Wilkins 2023-10-12 /pmc/articles/PMC10578741/ /pubmed/37820274 http://dx.doi.org/10.1097/HC9.0000000000000278 Text en Copyright © 2023 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the American Association for the Study of Liver Diseases. https://creativecommons.org/licenses/by-nc-nd/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution-Non Commercial-No Derivatives License 4.0 (https://creativecommons.org/licenses/by-nc-nd/4.0/) (CCBY-NC-ND), where it is permissible to download and share the work provided it is properly cited. The work cannot be changed in any way or used commercially without permission from the journal. http://creativecommons.org/licenses/by-nc-nd/4.0/ (https://creativecommons.org/licenses/by-nc-nd/4.0/)
spellingShingle Original Article
Shah, Anit
Huck, Ian
Duncan, Kaylia
Gansemer, Erica R.
Liu, Kaihua
Adajar, Reed C.
Apte, Udayan
Stamnes, Mark A.
Rutkowski, D. Thomas
Interference with the HNF4-dependent gene regulatory network diminishes endoplasmic reticulum stress in hepatocytes
title Interference with the HNF4-dependent gene regulatory network diminishes endoplasmic reticulum stress in hepatocytes
title_full Interference with the HNF4-dependent gene regulatory network diminishes endoplasmic reticulum stress in hepatocytes
title_fullStr Interference with the HNF4-dependent gene regulatory network diminishes endoplasmic reticulum stress in hepatocytes
title_full_unstemmed Interference with the HNF4-dependent gene regulatory network diminishes endoplasmic reticulum stress in hepatocytes
title_short Interference with the HNF4-dependent gene regulatory network diminishes endoplasmic reticulum stress in hepatocytes
title_sort interference with the hnf4-dependent gene regulatory network diminishes endoplasmic reticulum stress in hepatocytes
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10578741/
https://www.ncbi.nlm.nih.gov/pubmed/37820274
http://dx.doi.org/10.1097/HC9.0000000000000278
work_keys_str_mv AT shahanit interferencewiththehnf4dependentgeneregulatorynetworkdiminishesendoplasmicreticulumstressinhepatocytes
AT huckian interferencewiththehnf4dependentgeneregulatorynetworkdiminishesendoplasmicreticulumstressinhepatocytes
AT duncankaylia interferencewiththehnf4dependentgeneregulatorynetworkdiminishesendoplasmicreticulumstressinhepatocytes
AT gansemerericar interferencewiththehnf4dependentgeneregulatorynetworkdiminishesendoplasmicreticulumstressinhepatocytes
AT liukaihua interferencewiththehnf4dependentgeneregulatorynetworkdiminishesendoplasmicreticulumstressinhepatocytes
AT adajarreedc interferencewiththehnf4dependentgeneregulatorynetworkdiminishesendoplasmicreticulumstressinhepatocytes
AT apteudayan interferencewiththehnf4dependentgeneregulatorynetworkdiminishesendoplasmicreticulumstressinhepatocytes
AT stamnesmarka interferencewiththehnf4dependentgeneregulatorynetworkdiminishesendoplasmicreticulumstressinhepatocytes
AT rutkowskidthomas interferencewiththehnf4dependentgeneregulatorynetworkdiminishesendoplasmicreticulumstressinhepatocytes