Cargando…

Targeting macrophagic PIM-1 alleviates osteoarthritis by inhibiting NLRP3 inflammasome activation via suppressing mitochondrial ROS/Cl(−) efflux signaling pathway

BACKGROUND: Osteoarthritis (OA), in which macrophage-driven synovitis is considered closely related to cartilage destruction and could occur at any stage, is an inflammatory arthritis. However, there are no effective targets to cure the progression of OA. The NOD-, LRR-,and pyrin domain-containing p...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Zhen, Xie, Shujun, Qian, Jin, Gao, Fengqiang, Jin, Wenjian, Wang, Lingqiao, Yan, Lili, Chen, Hao, Yao, Wangxiang, Li, Maoqiang, Wang, Xuepeng, Zhu, Liulong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10329339/
https://www.ncbi.nlm.nih.gov/pubmed/37422640
http://dx.doi.org/10.1186/s12967-023-04313-1
_version_ 1785069996987121664
author Zhang, Zhen
Xie, Shujun
Qian, Jin
Gao, Fengqiang
Jin, Wenjian
Wang, Lingqiao
Yan, Lili
Chen, Hao
Yao, Wangxiang
Li, Maoqiang
Wang, Xuepeng
Zhu, Liulong
author_facet Zhang, Zhen
Xie, Shujun
Qian, Jin
Gao, Fengqiang
Jin, Wenjian
Wang, Lingqiao
Yan, Lili
Chen, Hao
Yao, Wangxiang
Li, Maoqiang
Wang, Xuepeng
Zhu, Liulong
author_sort Zhang, Zhen
collection PubMed
description BACKGROUND: Osteoarthritis (OA), in which macrophage-driven synovitis is considered closely related to cartilage destruction and could occur at any stage, is an inflammatory arthritis. However, there are no effective targets to cure the progression of OA. The NOD-, LRR-,and pyrin domain-containing protein 3 (NLRP3) inflammasome in synovial macrophages participates in the pathological inflammatory process and treatment strategies targeting it are considered to be an effective approach for OA. PIM-1 kinase, as a downstream effector of many cytokine signaling pathways, plays a pro-inflammatory role in inflammatory disease. METHODS: In this study, we evaluated the expression of the PIM-1 and the infiltration of synovial macrophages in the human OA synovium. The effects and mechanism of PIM-1 were investigated in mice and human macrophages stimulated by lipopolysaccharide (LPS) and different agonists such as nigericin, ATP, Monosodium urate (MSU), and Aluminum salt (Alum). The protective effects on chondrocytes were assessed by a modified co-culture system induced by macrophage condition medium (CM). The therapeutic effect in vivo was confirmed by the medial meniscus (DMM)-induced OA in mice. RESULTS: The expression of PIM-1 was increased in the human OA synovium which was accompanied by the infiltration of synovial macrophages. In vitro experiments, suppression of PIM-1 by SMI-4a, a specific inhibitor, rapidly inhibited the NLRP3 inflammasome activation in mice and human macrophages and gasdermin-D (GSDME)-mediated pyroptosis. Furthermore, PIM-1 inhibition specifically blocked the apoptosis-associated speck-like protein containing a CARD (ASC) oligomerization in the assembly stage. Mechanistically, PIM-1 inhibition alleviated the mitochondrial reactive oxygen species (ROS)/chloride intracellular channel proteins (CLICs)-dependent Cl(−) efflux signaling pathway, which eventually resulted in the blockade of the ASC oligomerization and NLRP3 inflammasome activation. Furthermore, PIM-1 suppression showed chondroprotective effects in the modified co-culture system. Finally, SMI-4a significantly suppressed the expression of PIM-1 in the synovium and reduced the synovitis scores and the Osteoarthritis Research Society International (OARSI) score in the DMM-induced OA model. CONCLUSIONS: Therefore, PIM-1 represented a new class of promising targets as a treatment of OA to target these mechanisms in macrophages and widened the road to therapeutic strategies for OA. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12967-023-04313-1.
format Online
Article
Text
id pubmed-10329339
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-103293392023-07-09 Targeting macrophagic PIM-1 alleviates osteoarthritis by inhibiting NLRP3 inflammasome activation via suppressing mitochondrial ROS/Cl(−) efflux signaling pathway Zhang, Zhen Xie, Shujun Qian, Jin Gao, Fengqiang Jin, Wenjian Wang, Lingqiao Yan, Lili Chen, Hao Yao, Wangxiang Li, Maoqiang Wang, Xuepeng Zhu, Liulong J Transl Med Research BACKGROUND: Osteoarthritis (OA), in which macrophage-driven synovitis is considered closely related to cartilage destruction and could occur at any stage, is an inflammatory arthritis. However, there are no effective targets to cure the progression of OA. The NOD-, LRR-,and pyrin domain-containing protein 3 (NLRP3) inflammasome in synovial macrophages participates in the pathological inflammatory process and treatment strategies targeting it are considered to be an effective approach for OA. PIM-1 kinase, as a downstream effector of many cytokine signaling pathways, plays a pro-inflammatory role in inflammatory disease. METHODS: In this study, we evaluated the expression of the PIM-1 and the infiltration of synovial macrophages in the human OA synovium. The effects and mechanism of PIM-1 were investigated in mice and human macrophages stimulated by lipopolysaccharide (LPS) and different agonists such as nigericin, ATP, Monosodium urate (MSU), and Aluminum salt (Alum). The protective effects on chondrocytes were assessed by a modified co-culture system induced by macrophage condition medium (CM). The therapeutic effect in vivo was confirmed by the medial meniscus (DMM)-induced OA in mice. RESULTS: The expression of PIM-1 was increased in the human OA synovium which was accompanied by the infiltration of synovial macrophages. In vitro experiments, suppression of PIM-1 by SMI-4a, a specific inhibitor, rapidly inhibited the NLRP3 inflammasome activation in mice and human macrophages and gasdermin-D (GSDME)-mediated pyroptosis. Furthermore, PIM-1 inhibition specifically blocked the apoptosis-associated speck-like protein containing a CARD (ASC) oligomerization in the assembly stage. Mechanistically, PIM-1 inhibition alleviated the mitochondrial reactive oxygen species (ROS)/chloride intracellular channel proteins (CLICs)-dependent Cl(−) efflux signaling pathway, which eventually resulted in the blockade of the ASC oligomerization and NLRP3 inflammasome activation. Furthermore, PIM-1 suppression showed chondroprotective effects in the modified co-culture system. Finally, SMI-4a significantly suppressed the expression of PIM-1 in the synovium and reduced the synovitis scores and the Osteoarthritis Research Society International (OARSI) score in the DMM-induced OA model. CONCLUSIONS: Therefore, PIM-1 represented a new class of promising targets as a treatment of OA to target these mechanisms in macrophages and widened the road to therapeutic strategies for OA. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12967-023-04313-1. BioMed Central 2023-07-08 /pmc/articles/PMC10329339/ /pubmed/37422640 http://dx.doi.org/10.1186/s12967-023-04313-1 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Zhang, Zhen
Xie, Shujun
Qian, Jin
Gao, Fengqiang
Jin, Wenjian
Wang, Lingqiao
Yan, Lili
Chen, Hao
Yao, Wangxiang
Li, Maoqiang
Wang, Xuepeng
Zhu, Liulong
Targeting macrophagic PIM-1 alleviates osteoarthritis by inhibiting NLRP3 inflammasome activation via suppressing mitochondrial ROS/Cl(−) efflux signaling pathway
title Targeting macrophagic PIM-1 alleviates osteoarthritis by inhibiting NLRP3 inflammasome activation via suppressing mitochondrial ROS/Cl(−) efflux signaling pathway
title_full Targeting macrophagic PIM-1 alleviates osteoarthritis by inhibiting NLRP3 inflammasome activation via suppressing mitochondrial ROS/Cl(−) efflux signaling pathway
title_fullStr Targeting macrophagic PIM-1 alleviates osteoarthritis by inhibiting NLRP3 inflammasome activation via suppressing mitochondrial ROS/Cl(−) efflux signaling pathway
title_full_unstemmed Targeting macrophagic PIM-1 alleviates osteoarthritis by inhibiting NLRP3 inflammasome activation via suppressing mitochondrial ROS/Cl(−) efflux signaling pathway
title_short Targeting macrophagic PIM-1 alleviates osteoarthritis by inhibiting NLRP3 inflammasome activation via suppressing mitochondrial ROS/Cl(−) efflux signaling pathway
title_sort targeting macrophagic pim-1 alleviates osteoarthritis by inhibiting nlrp3 inflammasome activation via suppressing mitochondrial ros/cl(−) efflux signaling pathway
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10329339/
https://www.ncbi.nlm.nih.gov/pubmed/37422640
http://dx.doi.org/10.1186/s12967-023-04313-1
work_keys_str_mv AT zhangzhen targetingmacrophagicpim1alleviatesosteoarthritisbyinhibitingnlrp3inflammasomeactivationviasuppressingmitochondrialroscleffluxsignalingpathway
AT xieshujun targetingmacrophagicpim1alleviatesosteoarthritisbyinhibitingnlrp3inflammasomeactivationviasuppressingmitochondrialroscleffluxsignalingpathway
AT qianjin targetingmacrophagicpim1alleviatesosteoarthritisbyinhibitingnlrp3inflammasomeactivationviasuppressingmitochondrialroscleffluxsignalingpathway
AT gaofengqiang targetingmacrophagicpim1alleviatesosteoarthritisbyinhibitingnlrp3inflammasomeactivationviasuppressingmitochondrialroscleffluxsignalingpathway
AT jinwenjian targetingmacrophagicpim1alleviatesosteoarthritisbyinhibitingnlrp3inflammasomeactivationviasuppressingmitochondrialroscleffluxsignalingpathway
AT wanglingqiao targetingmacrophagicpim1alleviatesosteoarthritisbyinhibitingnlrp3inflammasomeactivationviasuppressingmitochondrialroscleffluxsignalingpathway
AT yanlili targetingmacrophagicpim1alleviatesosteoarthritisbyinhibitingnlrp3inflammasomeactivationviasuppressingmitochondrialroscleffluxsignalingpathway
AT chenhao targetingmacrophagicpim1alleviatesosteoarthritisbyinhibitingnlrp3inflammasomeactivationviasuppressingmitochondrialroscleffluxsignalingpathway
AT yaowangxiang targetingmacrophagicpim1alleviatesosteoarthritisbyinhibitingnlrp3inflammasomeactivationviasuppressingmitochondrialroscleffluxsignalingpathway
AT limaoqiang targetingmacrophagicpim1alleviatesosteoarthritisbyinhibitingnlrp3inflammasomeactivationviasuppressingmitochondrialroscleffluxsignalingpathway
AT wangxuepeng targetingmacrophagicpim1alleviatesosteoarthritisbyinhibitingnlrp3inflammasomeactivationviasuppressingmitochondrialroscleffluxsignalingpathway
AT zhuliulong targetingmacrophagicpim1alleviatesosteoarthritisbyinhibitingnlrp3inflammasomeactivationviasuppressingmitochondrialroscleffluxsignalingpathway