IRE1α inhibition decreased TXNIP/NLRP3 inflammasome activation through miR-17-5p after neonatal hypoxic–ischemic brain injury in rats

Abstract Background The endoplasmic reticulum (ER) is responsible for the control of correct protein folding and protein function which is crucial for cell survival. However, under pathological conditions, such as hypoxia–ischemia (HI), there is an accumulation of unfolded proteins thereby triggerin...

Full description

Bibliographic Details
Main Authors: Di Chen, Brandon J. Dixon, Desislava M. Doycheva, Bo Li, Yang Zhang, Qin Hu, Yue He, Zongduo Guo, Derek Nowrangi, Jerry Flores, Valery Filippov, John H. Zhang, Jiping Tang
Format: Article
Language:English
Published: BMC 2018-02-01
Series:Journal of Neuroinflammation
Subjects:
Online Access:http://link.springer.com/article/10.1186/s12974-018-1077-9
id doaj-35cc4eb655054ed0a3bdcded8756876c
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Di Chen
Brandon J. Dixon
Desislava M. Doycheva
Bo Li
Yang Zhang
Qin Hu
Yue He
Zongduo Guo
Derek Nowrangi
Jerry Flores
Valery Filippov
John H. Zhang
Jiping Tang
spellingShingle Di Chen
Brandon J. Dixon
Desislava M. Doycheva
Bo Li
Yang Zhang
Qin Hu
Yue He
Zongduo Guo
Derek Nowrangi
Jerry Flores
Valery Filippov
John H. Zhang
Jiping Tang
IRE1α inhibition decreased TXNIP/NLRP3 inflammasome activation through miR-17-5p after neonatal hypoxic–ischemic brain injury in rats
Journal of Neuroinflammation
Hypoxia–ischemia
IRE1α
MicroRNA-17 (miR-17)
Neonatal
Nod-like receptor protein 3 (NLRP3)
Thioredoxin-interacting protein (TXNIP)
author_facet Di Chen
Brandon J. Dixon
Desislava M. Doycheva
Bo Li
Yang Zhang
Qin Hu
Yue He
Zongduo Guo
Derek Nowrangi
Jerry Flores
Valery Filippov
John H. Zhang
Jiping Tang
author_sort Di Chen
title IRE1α inhibition decreased TXNIP/NLRP3 inflammasome activation through miR-17-5p after neonatal hypoxic–ischemic brain injury in rats
title_short IRE1α inhibition decreased TXNIP/NLRP3 inflammasome activation through miR-17-5p after neonatal hypoxic–ischemic brain injury in rats
title_full IRE1α inhibition decreased TXNIP/NLRP3 inflammasome activation through miR-17-5p after neonatal hypoxic–ischemic brain injury in rats
title_fullStr IRE1α inhibition decreased TXNIP/NLRP3 inflammasome activation through miR-17-5p after neonatal hypoxic–ischemic brain injury in rats
title_full_unstemmed IRE1α inhibition decreased TXNIP/NLRP3 inflammasome activation through miR-17-5p after neonatal hypoxic–ischemic brain injury in rats
title_sort ire1α inhibition decreased txnip/nlrp3 inflammasome activation through mir-17-5p after neonatal hypoxic–ischemic brain injury in rats
publisher BMC
series Journal of Neuroinflammation
issn 1742-2094
publishDate 2018-02-01
description Abstract Background The endoplasmic reticulum (ER) is responsible for the control of correct protein folding and protein function which is crucial for cell survival. However, under pathological conditions, such as hypoxia–ischemia (HI), there is an accumulation of unfolded proteins thereby triggering the unfolded protein response (UPR) and causing ER stress which is associated with activation of several stress sensor signaling pathways, one of them being the inositol requiring enzyme-1 alpha (IRE1α) signaling pathway. The UPR is regarded as a potential contributor to neuronal cell death and inflammation after HI. In the present study, we sought to investigate whether microRNA-17 (miR-17), a potential IRE1α ribonuclease (RNase) substrate, arbitrates downregulation of thioredoxin-interacting protein (TXNIP) and consequent NLRP3 inflammasome activation in the immature brain after HI injury and whether inhibition of IRE1α may attenuate inflammation via miR-17/TXNIP regulation. Methods Postnatal day 10 rat pups (n = 287) were subjected to unilateral carotid artery ligation followed by 2.5 h of hypoxia (8% O2). STF-083010, an IRE1α RNase inhibitor, was intranasally delivered at 1 h post-HI or followed by an additional one administration per day for 2 days. MiR-17-5p mimic or anti-miR-17-5p inhibitor was injected intracerebroventricularly at 48 h before HI. Infarct volume and body weight were used to evaluate the short-term effects while brain weight, gross and microscopic brain tissue morphologies, and neurobehavioral tests were conducted for the long-term evaluation. Western blots, immunofluorescence staining, reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR), and co-immunoprecipitation (Co-IP) were used for mechanism studies. Results Endogenous phosphorylated IRE1α expression was significantly increased after HI. Intranasal administration of STF-083010 alleviated brain injury and improved neurological behavior. MiR-17-5p expression was reduced after HI, and this decrease was attenuated by STF-083010 treatment. MiR-17-5p mimic administration ameliorated TXNIP expression, NLRP3 inflammasome activation, caspase-1 cleavage, and IL-1β production, as well as brain infarct volume. Conversely, anti-miR-17-5p inhibitor reversed IRE1α inhibition-induced decrease in TXNIP expression and inflammasome activation, as well as exacerbated brain injury after HI. Conclusions IRE1a-induced UPR pathway may contribute to inflammatory activation and brain injury following neonatal HI. IRE1a activation, through decay of miR-17-5p, elevated TXNIP expression to activate NLRP3 inflammasome and aggravated brain damage.
topic Hypoxia–ischemia
IRE1α
MicroRNA-17 (miR-17)
Neonatal
Nod-like receptor protein 3 (NLRP3)
Thioredoxin-interacting protein (TXNIP)
url http://link.springer.com/article/10.1186/s12974-018-1077-9
work_keys_str_mv AT dichen ire1ainhibitiondecreasedtxnipnlrp3inflammasomeactivationthroughmir175pafterneonatalhypoxicischemicbraininjuryinrats
AT brandonjdixon ire1ainhibitiondecreasedtxnipnlrp3inflammasomeactivationthroughmir175pafterneonatalhypoxicischemicbraininjuryinrats
AT desislavamdoycheva ire1ainhibitiondecreasedtxnipnlrp3inflammasomeactivationthroughmir175pafterneonatalhypoxicischemicbraininjuryinrats
AT boli ire1ainhibitiondecreasedtxnipnlrp3inflammasomeactivationthroughmir175pafterneonatalhypoxicischemicbraininjuryinrats
AT yangzhang ire1ainhibitiondecreasedtxnipnlrp3inflammasomeactivationthroughmir175pafterneonatalhypoxicischemicbraininjuryinrats
AT qinhu ire1ainhibitiondecreasedtxnipnlrp3inflammasomeactivationthroughmir175pafterneonatalhypoxicischemicbraininjuryinrats
AT yuehe ire1ainhibitiondecreasedtxnipnlrp3inflammasomeactivationthroughmir175pafterneonatalhypoxicischemicbraininjuryinrats
AT zongduoguo ire1ainhibitiondecreasedtxnipnlrp3inflammasomeactivationthroughmir175pafterneonatalhypoxicischemicbraininjuryinrats
AT dereknowrangi ire1ainhibitiondecreasedtxnipnlrp3inflammasomeactivationthroughmir175pafterneonatalhypoxicischemicbraininjuryinrats
AT jerryflores ire1ainhibitiondecreasedtxnipnlrp3inflammasomeactivationthroughmir175pafterneonatalhypoxicischemicbraininjuryinrats
AT valeryfilippov ire1ainhibitiondecreasedtxnipnlrp3inflammasomeactivationthroughmir175pafterneonatalhypoxicischemicbraininjuryinrats
AT johnhzhang ire1ainhibitiondecreasedtxnipnlrp3inflammasomeactivationthroughmir175pafterneonatalhypoxicischemicbraininjuryinrats
AT jipingtang ire1ainhibitiondecreasedtxnipnlrp3inflammasomeactivationthroughmir175pafterneonatalhypoxicischemicbraininjuryinrats
_version_ 1725052853028913152
spelling doaj-35cc4eb655054ed0a3bdcded8756876c2020-11-25T01:38:35ZengBMCJournal of Neuroinflammation1742-20942018-02-0115111810.1186/s12974-018-1077-9IRE1α inhibition decreased TXNIP/NLRP3 inflammasome activation through miR-17-5p after neonatal hypoxic–ischemic brain injury in ratsDi Chen0Brandon J. Dixon1Desislava M. Doycheva2Bo Li3Yang Zhang4Qin Hu5Yue He6Zongduo Guo7Derek Nowrangi8Jerry Flores9Valery Filippov10John H. Zhang11Jiping Tang12Cerebrovascular Diseases Laboratory, Institute of Neuroscience, Chongqing Medical UniversityDepartment of Basic Sciences, Loma Linda University School of MedicineDepartment of Basic Sciences, Loma Linda University School of MedicineDepartment of Basic Sciences, Loma Linda University School of MedicineDepartment of Basic Sciences, Loma Linda University School of MedicineDepartment of Basic Sciences, Loma Linda University School of MedicineDepartment of Basic Sciences, Loma Linda University School of MedicineDepartment of Basic Sciences, Loma Linda University School of MedicineDepartment of Basic Sciences, Loma Linda University School of MedicineDepartment of Basic Sciences, Loma Linda University School of MedicineDepartment of Basic Sciences, Loma Linda University School of MedicineDepartment of Basic Sciences, Loma Linda University School of MedicineDepartment of Basic Sciences, Loma Linda University School of MedicineAbstract Background The endoplasmic reticulum (ER) is responsible for the control of correct protein folding and protein function which is crucial for cell survival. However, under pathological conditions, such as hypoxia–ischemia (HI), there is an accumulation of unfolded proteins thereby triggering the unfolded protein response (UPR) and causing ER stress which is associated with activation of several stress sensor signaling pathways, one of them being the inositol requiring enzyme-1 alpha (IRE1α) signaling pathway. The UPR is regarded as a potential contributor to neuronal cell death and inflammation after HI. In the present study, we sought to investigate whether microRNA-17 (miR-17), a potential IRE1α ribonuclease (RNase) substrate, arbitrates downregulation of thioredoxin-interacting protein (TXNIP) and consequent NLRP3 inflammasome activation in the immature brain after HI injury and whether inhibition of IRE1α may attenuate inflammation via miR-17/TXNIP regulation. Methods Postnatal day 10 rat pups (n = 287) were subjected to unilateral carotid artery ligation followed by 2.5 h of hypoxia (8% O2). STF-083010, an IRE1α RNase inhibitor, was intranasally delivered at 1 h post-HI or followed by an additional one administration per day for 2 days. MiR-17-5p mimic or anti-miR-17-5p inhibitor was injected intracerebroventricularly at 48 h before HI. Infarct volume and body weight were used to evaluate the short-term effects while brain weight, gross and microscopic brain tissue morphologies, and neurobehavioral tests were conducted for the long-term evaluation. Western blots, immunofluorescence staining, reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR), and co-immunoprecipitation (Co-IP) were used for mechanism studies. Results Endogenous phosphorylated IRE1α expression was significantly increased after HI. Intranasal administration of STF-083010 alleviated brain injury and improved neurological behavior. MiR-17-5p expression was reduced after HI, and this decrease was attenuated by STF-083010 treatment. MiR-17-5p mimic administration ameliorated TXNIP expression, NLRP3 inflammasome activation, caspase-1 cleavage, and IL-1β production, as well as brain infarct volume. Conversely, anti-miR-17-5p inhibitor reversed IRE1α inhibition-induced decrease in TXNIP expression and inflammasome activation, as well as exacerbated brain injury after HI. Conclusions IRE1a-induced UPR pathway may contribute to inflammatory activation and brain injury following neonatal HI. IRE1a activation, through decay of miR-17-5p, elevated TXNIP expression to activate NLRP3 inflammasome and aggravated brain damage.http://link.springer.com/article/10.1186/s12974-018-1077-9Hypoxia–ischemiaIRE1αMicroRNA-17 (miR-17)NeonatalNod-like receptor protein 3 (NLRP3)Thioredoxin-interacting protein (TXNIP)