Cargando…

IL-1β receptor antagonist (IL-1Ra) combined with autophagy inducer (TAT-Beclin1) is an effective alternative for attenuating extracellular matrix degradation in rat and human osteoarthritis chondrocytes

BACKGROUND: Autophagy induction is an effective approach for OA therapy. IL-1β is one of the major inflammatory cytokines linked to OA pathological progression, and its receptor blockade interrupts OA cartilage destruction. The objective of this study was to decipher the link between autophagy and r...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Fen, Liu, Jijie, Chen, Xiaolei, Zheng, Xinpeng, Qu, Ning, Zhang, Bing, Xia, Chun
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6617669/
https://www.ncbi.nlm.nih.gov/pubmed/31291980
http://dx.doi.org/10.1186/s13075-019-1952-5
_version_ 1783433743712649216
author Wang, Fen
Liu, Jijie
Chen, Xiaolei
Zheng, Xinpeng
Qu, Ning
Zhang, Bing
Xia, Chun
author_facet Wang, Fen
Liu, Jijie
Chen, Xiaolei
Zheng, Xinpeng
Qu, Ning
Zhang, Bing
Xia, Chun
author_sort Wang, Fen
collection PubMed
description BACKGROUND: Autophagy induction is an effective approach for OA therapy. IL-1β is one of the major inflammatory cytokines linked to OA pathological progression, and its receptor blockade interrupts OA cartilage destruction. The objective of this study was to decipher the link between autophagy and regulatory mechanism of IL-1β and to investigate the effect of IL-1β receptor blockade by IL-1 receptor antagonist (IL-1Ra) combined with or without an autophagy inducer (TAT-Beclin1) on extracellular matrix (ECM) in OA chondrocytes in vitro and in vivo. METHODS: IL-1β-treated rat and human OA chondrocytes were cultured in response to IL-1Ra. The expression and distribution of signal molecules regulating ECM synthesis and autophagy were investigated via western blotting, immunoprecipitation, real-time PCR, immunofluorescence, and transmission electron microscope technique. Furthermore, after intra-articular injection of IL-1Ra, TAT-Beclin1, and a combination of both in a rat OA model established by anterior cruciate ligament transection and medial meniscus resection, the morphological changes of cartilage and related signal molecule expression levels were monitored using H.E., Safranin O-Fast green, and immunohistochemistry staining. RESULTS: Reduced autophagy by IL-1β contributed to ECM degradation, and blockade of IL-1β by IL-1Ra restored autophagy and attenuated ECM degradation in rat and human OA chondrocytes, as well as in a rat OA model. Akt/mTOR/ULK1, Akt/mTOR/NF-κB, and LC3B deacetylation were involved in autophagy regulated by IL-1β. Intra-articular injection of IL-1Ra combined with TAT-Beclin1 was more effective than IL-1Ra alone. CONCLUSIONS: IL-1Ra restored autophagy and attenuated ECM degradation, with an implication that blocking IL-1β combined with enhancing autophagy might be a potential therapeutic strategy for OA. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s13075-019-1952-5) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-6617669
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-66176692019-07-22 IL-1β receptor antagonist (IL-1Ra) combined with autophagy inducer (TAT-Beclin1) is an effective alternative for attenuating extracellular matrix degradation in rat and human osteoarthritis chondrocytes Wang, Fen Liu, Jijie Chen, Xiaolei Zheng, Xinpeng Qu, Ning Zhang, Bing Xia, Chun Arthritis Res Ther Research Article BACKGROUND: Autophagy induction is an effective approach for OA therapy. IL-1β is one of the major inflammatory cytokines linked to OA pathological progression, and its receptor blockade interrupts OA cartilage destruction. The objective of this study was to decipher the link between autophagy and regulatory mechanism of IL-1β and to investigate the effect of IL-1β receptor blockade by IL-1 receptor antagonist (IL-1Ra) combined with or without an autophagy inducer (TAT-Beclin1) on extracellular matrix (ECM) in OA chondrocytes in vitro and in vivo. METHODS: IL-1β-treated rat and human OA chondrocytes were cultured in response to IL-1Ra. The expression and distribution of signal molecules regulating ECM synthesis and autophagy were investigated via western blotting, immunoprecipitation, real-time PCR, immunofluorescence, and transmission electron microscope technique. Furthermore, after intra-articular injection of IL-1Ra, TAT-Beclin1, and a combination of both in a rat OA model established by anterior cruciate ligament transection and medial meniscus resection, the morphological changes of cartilage and related signal molecule expression levels were monitored using H.E., Safranin O-Fast green, and immunohistochemistry staining. RESULTS: Reduced autophagy by IL-1β contributed to ECM degradation, and blockade of IL-1β by IL-1Ra restored autophagy and attenuated ECM degradation in rat and human OA chondrocytes, as well as in a rat OA model. Akt/mTOR/ULK1, Akt/mTOR/NF-κB, and LC3B deacetylation were involved in autophagy regulated by IL-1β. Intra-articular injection of IL-1Ra combined with TAT-Beclin1 was more effective than IL-1Ra alone. CONCLUSIONS: IL-1Ra restored autophagy and attenuated ECM degradation, with an implication that blocking IL-1β combined with enhancing autophagy might be a potential therapeutic strategy for OA. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s13075-019-1952-5) contains supplementary material, which is available to authorized users. BioMed Central 2019-07-10 2019 /pmc/articles/PMC6617669/ /pubmed/31291980 http://dx.doi.org/10.1186/s13075-019-1952-5 Text en © The Author(s). 2019 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research Article
Wang, Fen
Liu, Jijie
Chen, Xiaolei
Zheng, Xinpeng
Qu, Ning
Zhang, Bing
Xia, Chun
IL-1β receptor antagonist (IL-1Ra) combined with autophagy inducer (TAT-Beclin1) is an effective alternative for attenuating extracellular matrix degradation in rat and human osteoarthritis chondrocytes
title IL-1β receptor antagonist (IL-1Ra) combined with autophagy inducer (TAT-Beclin1) is an effective alternative for attenuating extracellular matrix degradation in rat and human osteoarthritis chondrocytes
title_full IL-1β receptor antagonist (IL-1Ra) combined with autophagy inducer (TAT-Beclin1) is an effective alternative for attenuating extracellular matrix degradation in rat and human osteoarthritis chondrocytes
title_fullStr IL-1β receptor antagonist (IL-1Ra) combined with autophagy inducer (TAT-Beclin1) is an effective alternative for attenuating extracellular matrix degradation in rat and human osteoarthritis chondrocytes
title_full_unstemmed IL-1β receptor antagonist (IL-1Ra) combined with autophagy inducer (TAT-Beclin1) is an effective alternative for attenuating extracellular matrix degradation in rat and human osteoarthritis chondrocytes
title_short IL-1β receptor antagonist (IL-1Ra) combined with autophagy inducer (TAT-Beclin1) is an effective alternative for attenuating extracellular matrix degradation in rat and human osteoarthritis chondrocytes
title_sort il-1β receptor antagonist (il-1ra) combined with autophagy inducer (tat-beclin1) is an effective alternative for attenuating extracellular matrix degradation in rat and human osteoarthritis chondrocytes
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6617669/
https://www.ncbi.nlm.nih.gov/pubmed/31291980
http://dx.doi.org/10.1186/s13075-019-1952-5
work_keys_str_mv AT wangfen il1breceptorantagonistil1racombinedwithautophagyinducertatbeclin1isaneffectivealternativeforattenuatingextracellularmatrixdegradationinratandhumanosteoarthritischondrocytes
AT liujijie il1breceptorantagonistil1racombinedwithautophagyinducertatbeclin1isaneffectivealternativeforattenuatingextracellularmatrixdegradationinratandhumanosteoarthritischondrocytes
AT chenxiaolei il1breceptorantagonistil1racombinedwithautophagyinducertatbeclin1isaneffectivealternativeforattenuatingextracellularmatrixdegradationinratandhumanosteoarthritischondrocytes
AT zhengxinpeng il1breceptorantagonistil1racombinedwithautophagyinducertatbeclin1isaneffectivealternativeforattenuatingextracellularmatrixdegradationinratandhumanosteoarthritischondrocytes
AT quning il1breceptorantagonistil1racombinedwithautophagyinducertatbeclin1isaneffectivealternativeforattenuatingextracellularmatrixdegradationinratandhumanosteoarthritischondrocytes
AT zhangbing il1breceptorantagonistil1racombinedwithautophagyinducertatbeclin1isaneffectivealternativeforattenuatingextracellularmatrixdegradationinratandhumanosteoarthritischondrocytes
AT xiachun il1breceptorantagonistil1racombinedwithautophagyinducertatbeclin1isaneffectivealternativeforattenuatingextracellularmatrixdegradationinratandhumanosteoarthritischondrocytes