IL-33/ST2 induces macrophage-dependent ROS production and TRPA1 activation that mediate pain-like responses by skin incision in mice

IF 12.4 1区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL
Theranostics Pub Date : 2024-08-19 DOI:10.7150/thno.97856
Ruoyao Xu, Yushuang Pan, Kaige Zheng, Muyan Chen, Chengyu Yin, Qimiao Hu, Jie Wang, Qing Yu, Peiyi Li, Yan Tai, Junfan Fang, Boyu Liu, Jianqiao Fang, Guihua Tian, Boyi Liu
{"title":"IL-33/ST2 induces macrophage-dependent ROS production and TRPA1 activation that mediate pain-like responses by skin incision in mice","authors":"Ruoyao Xu, Yushuang Pan, Kaige Zheng, Muyan Chen, Chengyu Yin, Qimiao Hu, Jie Wang, Qing Yu, Peiyi Li, Yan Tai, Junfan Fang, Boyu Liu, Jianqiao Fang, Guihua Tian, Boyi Liu","doi":"10.7150/thno.97856","DOIUrl":null,"url":null,"abstract":"<b>Background:</b> Insufficiently managed incisional (INC) pain severely affects patients' life quality and rehabilitation after a major operation. However, mechanisms underlying INC pain still remain poorly understood./n<b>Methods:</b> A mouse model of INC pain was established by skin plus deep muscle incision. Biochemistry assay, <i>in vivo</i> reactive oxygen species (ROS) imaging, Ca<sup>2+</sup> imaging combined with retrograde labelling, neuron tracing and nocifensive behavior test, etc. were utilized for mechanism investigation./n<b>Results:</b> We found pro-nociceptive cytokine interleukin -33 (IL-33) ranked among top up-regulated cytokines in incised tissues of INC pain model mice. IL-33 was predominantly expressed in keratinocytes around the incisional area. Neutralization of IL-33 or its receptor suppression of tumorigenicity 2 protein (ST2) or genetic deletion of <i>St2</i> gene (<i>St2</i><sup>-/-</sup>) remarkably ameliorated mechanical allodynia and improved gait impairments of model mice. IL-33 contributes to INC pain by recruiting macrophages, which subsequently release ROS in incised tissues via ST2-dependent mechanism. Transfer of excessive macrophages enhanced oxidative injury and reproduced mechanical allodynia in <i>St2</i><sup>-/-</sup> mice upon tissue incision. Overproduced ROS subsequently activated functionally up-regulated transient receptor potential ankyrin subtype-1 (TRPA1) channel innervating the incisional site to produce mechanical allodynia. Neither deleting <i>St2</i> nor attenuating ROS affected wound healing of model mice./n<b>Conclusions:</b> Our work uncovered a previously unrecognized contribution of IL-33/ST2 signaling in mediating mechanical allodynia and gait impairment of a mouse model of INC pain. Targeting IL-33/ST2 signaling could be a novel therapeutic approach for INC pain management.","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":null,"pages":null},"PeriodicalIF":12.4000,"publicationDate":"2024-08-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Theranostics","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.7150/thno.97856","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"MEDICINE, RESEARCH & EXPERIMENTAL","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Insufficiently managed incisional (INC) pain severely affects patients' life quality and rehabilitation after a major operation. However, mechanisms underlying INC pain still remain poorly understood./nMethods: A mouse model of INC pain was established by skin plus deep muscle incision. Biochemistry assay, in vivo reactive oxygen species (ROS) imaging, Ca2+ imaging combined with retrograde labelling, neuron tracing and nocifensive behavior test, etc. were utilized for mechanism investigation./nResults: We found pro-nociceptive cytokine interleukin -33 (IL-33) ranked among top up-regulated cytokines in incised tissues of INC pain model mice. IL-33 was predominantly expressed in keratinocytes around the incisional area. Neutralization of IL-33 or its receptor suppression of tumorigenicity 2 protein (ST2) or genetic deletion of St2 gene (St2-/-) remarkably ameliorated mechanical allodynia and improved gait impairments of model mice. IL-33 contributes to INC pain by recruiting macrophages, which subsequently release ROS in incised tissues via ST2-dependent mechanism. Transfer of excessive macrophages enhanced oxidative injury and reproduced mechanical allodynia in St2-/- mice upon tissue incision. Overproduced ROS subsequently activated functionally up-regulated transient receptor potential ankyrin subtype-1 (TRPA1) channel innervating the incisional site to produce mechanical allodynia. Neither deleting St2 nor attenuating ROS affected wound healing of model mice./nConclusions: Our work uncovered a previously unrecognized contribution of IL-33/ST2 signaling in mediating mechanical allodynia and gait impairment of a mouse model of INC pain. Targeting IL-33/ST2 signaling could be a novel therapeutic approach for INC pain management.
IL-33/ST2 可诱导巨噬细胞依赖性 ROS 生成和 TRPA1 激活,从而介导小鼠皮肤切口疼痛样反应
背景:切口(INC)疼痛处理不当会严重影响患者的生活质量和大手术后的康复。然而,人们对 INC 疼痛的机制仍然知之甚少:通过皮肤加深肌肉切口建立了 INC 疼痛小鼠模型。采用生化分析、体内活性氧成像、Ca2+成像结合逆行标记、神经元追踪和痛觉行为测试等方法进行机制研究:我们发现在 INC 疼痛模型小鼠的切口组织中,促痛觉细胞因子白细胞介素 33(IL-33)的上调率位居前列。IL-33 主要在切口周围的角质细胞中表达。中和 IL-33 或其受体抑制肿瘤生成 2 蛋白(ST2)或遗传性删除 St2 基因(St2-/-)可显著改善机械异感,并改善模型小鼠的步态障碍。IL-33通过招募巨噬细胞导致INC疼痛,巨噬细胞随后通过ST2依赖性机制在切口组织中释放ROS。转移过量的巨噬细胞会增强氧化损伤,并在 St2-/-小鼠身上重现组织切口时的机械痛觉。过量产生的 ROS 随后激活了支配切口部位的功能上调的瞬时受体电位淀粉样蛋白亚型-1(TRPA1)通道,从而产生机械异感。删除 St2 或减少 ROS 都不会影响模型小鼠的伤口愈合:我们的研究发现,IL-33/ST2 信号在介导 INC 疼痛小鼠模型的机械异感和步态障碍方面的作用以前从未被认识到。以 IL-33/ST2 信号为靶点可能是一种治疗 INC 疼痛的新方法。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
Theranostics
Theranostics MEDICINE, RESEARCH & EXPERIMENTAL-
CiteScore
25.40
自引率
1.60%
发文量
433
审稿时长
1 months
期刊介绍: Theranostics serves as a pivotal platform for the exchange of clinical and scientific insights within the diagnostic and therapeutic molecular and nanomedicine community, along with allied professions engaged in integrating molecular imaging and therapy. As a multidisciplinary journal, Theranostics showcases innovative research articles spanning fields such as in vitro diagnostics and prognostics, in vivo molecular imaging, molecular therapeutics, image-guided therapy, biosensor technology, nanobiosensors, bioelectronics, system biology, translational medicine, point-of-care applications, and personalized medicine. Encouraging a broad spectrum of biomedical research with potential theranostic applications, the journal rigorously peer-reviews primary research, alongside publishing reviews, news, and commentary that aim to bridge the gap between the laboratory, clinic, and biotechnology industries.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信