Cargando…

Activation of the IRE1α Arm, but not the PERK Arm, of the Unfolded Protein Response Contributes to Fumonisin B1-Induced Hepatotoxicity

Previous studies by us or others have shown that endoplasmic reticulum (ER) stress was activated by fumonisin 1 (FB1) exposure, which is considered to be a critical event in the FB1-induced toxic effect. However, the detailed mechanisms underlying FB1-induced ER stress-mediated liver toxicity remain...

Descripción completa

Detalles Bibliográficos
Autores principales: Liu, Xiaoyi, Zhang, Enxiang, Yin, Shutao, Zhao, Chong, Fan, Lihong, Hu, Hongbo
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7020448/
https://www.ncbi.nlm.nih.gov/pubmed/31963346
http://dx.doi.org/10.3390/toxins12010055
_version_ 1783497747441123328
author Liu, Xiaoyi
Zhang, Enxiang
Yin, Shutao
Zhao, Chong
Fan, Lihong
Hu, Hongbo
author_facet Liu, Xiaoyi
Zhang, Enxiang
Yin, Shutao
Zhao, Chong
Fan, Lihong
Hu, Hongbo
author_sort Liu, Xiaoyi
collection PubMed
description Previous studies by us or others have shown that endoplasmic reticulum (ER) stress was activated by fumonisin 1 (FB1) exposure, which is considered to be a critical event in the FB1-induced toxic effect. However, the detailed mechanisms underlying FB1-induced ER stress-mediated liver toxicity remain elusive. The objectives of the present study were designed to address the following issues: (1) the contribution of each arm of the unfolded protein response (UPR); (2) the downstream targets of ER stress that mediated FB1-induced liver toxicity; and (3) the relationship between ER stress and oxidative stress triggered by FB1. We also investigated whether the inhibition of ER stress by its inhibitor could offer protection against FB1-induced hepatotoxicity in vivo, which has not been critically addressed previously. The results showed that the activation of the IRE1α axis, but not of the PERK axis, of UPR contributed to FB1-induced ER stress-mediated hepatocyte toxicity; the activation of the Bax/Bak-mediated mitochondrial pathway lay downstream of IRE1α to trigger mitochondrial-dependent apoptosis in response to FB1; FB1-induced oxidative stress and ER stress augmented each other through a positive feedback mechanism; tauroursodeoxycholic acid (TUDCA)-mediated ER stress inactivation is an effective approach to counteract FB1-induced hepatotoxicity in vivo. The data of the present study allow us to better understand the mechanisms of FB1-induced hepatotoxicity.
format Online
Article
Text
id pubmed-7020448
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-70204482020-03-09 Activation of the IRE1α Arm, but not the PERK Arm, of the Unfolded Protein Response Contributes to Fumonisin B1-Induced Hepatotoxicity Liu, Xiaoyi Zhang, Enxiang Yin, Shutao Zhao, Chong Fan, Lihong Hu, Hongbo Toxins (Basel) Article Previous studies by us or others have shown that endoplasmic reticulum (ER) stress was activated by fumonisin 1 (FB1) exposure, which is considered to be a critical event in the FB1-induced toxic effect. However, the detailed mechanisms underlying FB1-induced ER stress-mediated liver toxicity remain elusive. The objectives of the present study were designed to address the following issues: (1) the contribution of each arm of the unfolded protein response (UPR); (2) the downstream targets of ER stress that mediated FB1-induced liver toxicity; and (3) the relationship between ER stress and oxidative stress triggered by FB1. We also investigated whether the inhibition of ER stress by its inhibitor could offer protection against FB1-induced hepatotoxicity in vivo, which has not been critically addressed previously. The results showed that the activation of the IRE1α axis, but not of the PERK axis, of UPR contributed to FB1-induced ER stress-mediated hepatocyte toxicity; the activation of the Bax/Bak-mediated mitochondrial pathway lay downstream of IRE1α to trigger mitochondrial-dependent apoptosis in response to FB1; FB1-induced oxidative stress and ER stress augmented each other through a positive feedback mechanism; tauroursodeoxycholic acid (TUDCA)-mediated ER stress inactivation is an effective approach to counteract FB1-induced hepatotoxicity in vivo. The data of the present study allow us to better understand the mechanisms of FB1-induced hepatotoxicity. MDPI 2020-01-16 /pmc/articles/PMC7020448/ /pubmed/31963346 http://dx.doi.org/10.3390/toxins12010055 Text en © 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Liu, Xiaoyi
Zhang, Enxiang
Yin, Shutao
Zhao, Chong
Fan, Lihong
Hu, Hongbo
Activation of the IRE1α Arm, but not the PERK Arm, of the Unfolded Protein Response Contributes to Fumonisin B1-Induced Hepatotoxicity
title Activation of the IRE1α Arm, but not the PERK Arm, of the Unfolded Protein Response Contributes to Fumonisin B1-Induced Hepatotoxicity
title_full Activation of the IRE1α Arm, but not the PERK Arm, of the Unfolded Protein Response Contributes to Fumonisin B1-Induced Hepatotoxicity
title_fullStr Activation of the IRE1α Arm, but not the PERK Arm, of the Unfolded Protein Response Contributes to Fumonisin B1-Induced Hepatotoxicity
title_full_unstemmed Activation of the IRE1α Arm, but not the PERK Arm, of the Unfolded Protein Response Contributes to Fumonisin B1-Induced Hepatotoxicity
title_short Activation of the IRE1α Arm, but not the PERK Arm, of the Unfolded Protein Response Contributes to Fumonisin B1-Induced Hepatotoxicity
title_sort activation of the ire1α arm, but not the perk arm, of the unfolded protein response contributes to fumonisin b1-induced hepatotoxicity
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7020448/
https://www.ncbi.nlm.nih.gov/pubmed/31963346
http://dx.doi.org/10.3390/toxins12010055
work_keys_str_mv AT liuxiaoyi activationoftheire1aarmbutnottheperkarmoftheunfoldedproteinresponsecontributestofumonisinb1inducedhepatotoxicity
AT zhangenxiang activationoftheire1aarmbutnottheperkarmoftheunfoldedproteinresponsecontributestofumonisinb1inducedhepatotoxicity
AT yinshutao activationoftheire1aarmbutnottheperkarmoftheunfoldedproteinresponsecontributestofumonisinb1inducedhepatotoxicity
AT zhaochong activationoftheire1aarmbutnottheperkarmoftheunfoldedproteinresponsecontributestofumonisinb1inducedhepatotoxicity
AT fanlihong activationoftheire1aarmbutnottheperkarmoftheunfoldedproteinresponsecontributestofumonisinb1inducedhepatotoxicity
AT huhongbo activationoftheire1aarmbutnottheperkarmoftheunfoldedproteinresponsecontributestofumonisinb1inducedhepatotoxicity