rutacarpine通过抑制TNFR-MAPK/NF-κB和NLRP3炎症小体信号通路减轻尿酸钠晶体诱导的痛风炎症。

IF 2.2 3区 医学 Q2 INTEGRATIVE & COMPLEMENTARY MEDICINE
Min Li, Zhu-Jun Yin, Li Li, Yun-Yun Quan, Ting Wang, Xin Zhu, Rui-Rong Tan, Jin Zeng, Hua Hua, Qin-Xuan Wu, Jun-Ning Zhao
{"title":"rutacarpine通过抑制TNFR-MAPK/NF-κB和NLRP3炎症小体信号通路减轻尿酸钠晶体诱导的痛风炎症。","authors":"Min Li, Zhu-Jun Yin, Li Li, Yun-Yun Quan, Ting Wang, Xin Zhu, Rui-Rong Tan, Jin Zeng, Hua Hua, Qin-Xuan Wu, Jun-Ning Zhao","doi":"10.1007/s11655-025-4204-3","DOIUrl":null,"url":null,"abstract":"<p><strong>Objective: </strong>To investigate the anti-inflammatory effect of rutaecarpine (RUT) on monosodium urate crystal (MSU)-induced murine peritonitis in mice and further explored the underlying mechanism of RUT in lipopolysaccharide (LPS)/MSU-induced gout model in vitro.</p><p><strong>Methods: </strong>In MSU-induced mice, 36 male C57BL/6 mice were randomly divided into 6 groups of 8 mice each group, including the control group, model group, RUT low-, medium-, and high-doses groups, and prednisone acetate group. The mice in each group were orally administered the corresponding drugs or vehicle once a day for 7 consecutive days. The gout inflammation model was established by intraperitoneal injection of MSU to evaluate the anti-gout inflammatory effects of RUT. Then the proinflammatory cytokines were measured by enzyme-linked immunosorbent assay (ELISA) and the proportions of infiltrating neutrophils cytokines were detected by flow cytometry. In LPS/MSU-treated or untreated THP-1 macrophages, cell viability was observed by cell counting kit 8 and proinflammatory cytokines were measured by ELISA. The percentage of pyroptotic cells were detected by flow cytometry. Respectively, the mRNA and protein levels were measured by real-time quantitative polymerase chain reaction (qRT-PCR) and Western blot, the nuclear translocation of nuclear factor κB (NF-κB) p65 was observed by laser confocal imaging. Additionally, surface plasmon resonance (SPR) and molecular docking were applied to validate the binding ability of RUT components to tumor necrosis factor α (TNF-α) targets.</p><p><strong>Results: </strong>RUT reduced the levels of infiltrating neutrophils and monocytes and decreased the levels of the proinflammatory cytokines interleukin 1β (IL-1β) and interleukin 6 (IL-6, all P<0.01). In vitro, RUT reduced the production of IL-1β, IL-6 and TNF-α. In addition, RT-PCR revealed the inhibitory effects of RUT on the mRNA levels of IL-1β, IL-6, cyclooxygenase-2 and TNF-α (P<0.05 or P<0.01). Mechanistically, RUT markedly reduced protein expressions of tumor necrosis factor receptor (TNFR), phospho-mitogen-activated protein kinase (p-MAPK), phospho-extracellular signal-regulated kinase, phospho-c-Jun N-terminal kinase, phospho-NF-κB, phospho-kinase α/β, NOD-like receptor thermal protein domain associated protein 3 (NLRPS), cleaved-cysteinyl aspartate specific proteinase-1 and cleaved-gasdermin D in macrophages (P<0.05 or P<0.01). Molecularly, SPR revealed that RUT bound to TNF-α with a calculated equilibrium dissociation constant of 31.7 µmol/L. Molecular docking further confirmed that RUT could interact directly with the TNF-α protein via hydrogen bonding, van der Waals interactions, and carbon-hydrogen bonding.</p><p><strong>Conclusion: </strong>RUT alleviated MSU-induced peritonitis and inhibited the TNFR1-MAPK/NF-κB and NLRP3 inflammasome signaling pathway to attenuate gouty inflammation induced by LPS/MSU in THP-1 macrophages, suggesting that RUT could be a potential therapeutic candidate for gout.</p>","PeriodicalId":10005,"journal":{"name":"Chinese Journal of Integrative Medicine","volume":" ","pages":""},"PeriodicalIF":2.2000,"publicationDate":"2025-05-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Rutaecarpine Attenuates Monosodium Urate Crystal-Induced Gouty Inflammation via Inhibition of TNFR-MAPK/NF-κB and NLRP3 Inflammasome Signaling Pathways.\",\"authors\":\"Min Li, Zhu-Jun Yin, Li Li, Yun-Yun Quan, Ting Wang, Xin Zhu, Rui-Rong Tan, Jin Zeng, Hua Hua, Qin-Xuan Wu, Jun-Ning Zhao\",\"doi\":\"10.1007/s11655-025-4204-3\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Objective: </strong>To investigate the anti-inflammatory effect of rutaecarpine (RUT) on monosodium urate crystal (MSU)-induced murine peritonitis in mice and further explored the underlying mechanism of RUT in lipopolysaccharide (LPS)/MSU-induced gout model in vitro.</p><p><strong>Methods: </strong>In MSU-induced mice, 36 male C57BL/6 mice were randomly divided into 6 groups of 8 mice each group, including the control group, model group, RUT low-, medium-, and high-doses groups, and prednisone acetate group. The mice in each group were orally administered the corresponding drugs or vehicle once a day for 7 consecutive days. The gout inflammation model was established by intraperitoneal injection of MSU to evaluate the anti-gout inflammatory effects of RUT. Then the proinflammatory cytokines were measured by enzyme-linked immunosorbent assay (ELISA) and the proportions of infiltrating neutrophils cytokines were detected by flow cytometry. In LPS/MSU-treated or untreated THP-1 macrophages, cell viability was observed by cell counting kit 8 and proinflammatory cytokines were measured by ELISA. The percentage of pyroptotic cells were detected by flow cytometry. Respectively, the mRNA and protein levels were measured by real-time quantitative polymerase chain reaction (qRT-PCR) and Western blot, the nuclear translocation of nuclear factor κB (NF-κB) p65 was observed by laser confocal imaging. Additionally, surface plasmon resonance (SPR) and molecular docking were applied to validate the binding ability of RUT components to tumor necrosis factor α (TNF-α) targets.</p><p><strong>Results: </strong>RUT reduced the levels of infiltrating neutrophils and monocytes and decreased the levels of the proinflammatory cytokines interleukin 1β (IL-1β) and interleukin 6 (IL-6, all P<0.01). In vitro, RUT reduced the production of IL-1β, IL-6 and TNF-α. In addition, RT-PCR revealed the inhibitory effects of RUT on the mRNA levels of IL-1β, IL-6, cyclooxygenase-2 and TNF-α (P<0.05 or P<0.01). Mechanistically, RUT markedly reduced protein expressions of tumor necrosis factor receptor (TNFR), phospho-mitogen-activated protein kinase (p-MAPK), phospho-extracellular signal-regulated kinase, phospho-c-Jun N-terminal kinase, phospho-NF-κB, phospho-kinase α/β, NOD-like receptor thermal protein domain associated protein 3 (NLRPS), cleaved-cysteinyl aspartate specific proteinase-1 and cleaved-gasdermin D in macrophages (P<0.05 or P<0.01). Molecularly, SPR revealed that RUT bound to TNF-α with a calculated equilibrium dissociation constant of 31.7 µmol/L. Molecular docking further confirmed that RUT could interact directly with the TNF-α protein via hydrogen bonding, van der Waals interactions, and carbon-hydrogen bonding.</p><p><strong>Conclusion: </strong>RUT alleviated MSU-induced peritonitis and inhibited the TNFR1-MAPK/NF-κB and NLRP3 inflammasome signaling pathway to attenuate gouty inflammation induced by LPS/MSU in THP-1 macrophages, suggesting that RUT could be a potential therapeutic candidate for gout.</p>\",\"PeriodicalId\":10005,\"journal\":{\"name\":\"Chinese Journal of Integrative Medicine\",\"volume\":\" \",\"pages\":\"\"},\"PeriodicalIF\":2.2000,\"publicationDate\":\"2025-05-08\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Chinese Journal of Integrative Medicine\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1007/s11655-025-4204-3\",\"RegionNum\":3,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q2\",\"JCRName\":\"INTEGRATIVE & COMPLEMENTARY MEDICINE\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Chinese Journal of Integrative Medicine","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1007/s11655-025-4204-3","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"INTEGRATIVE & COMPLEMENTARY MEDICINE","Score":null,"Total":0}
引用次数: 0

摘要

目的:研究芦丁卡尔平(RUT)对尿酸单钠晶体(MSU)所致小鼠腹膜炎的抗炎作用,并进一步探讨RUT在脂多糖(LPS)/MSU诱导的痛风模型中的作用机制。方法:选取msu诱导小鼠36只雄性C57BL/6小鼠,随机分为6组,每组8只,分别为对照组、模型组、RUT低、中、高剂量组和醋酸泼尼松组。各组小鼠分别口服相应药物或载药,每天1次,连续7天。通过腹腔注射MSU建立痛风炎症模型,评价RUT抗痛风炎症作用。采用酶联免疫吸附法(ELISA)检测促炎细胞因子,流式细胞术检测浸润中性粒细胞细胞因子比例。LPS/ msu处理和未处理的THP-1巨噬细胞,用细胞计数试剂盒8观察细胞活力,ELISA检测促炎因子。流式细胞术检测热噬细胞百分率。分别采用实时定量聚合酶链反应(qRT-PCR)和Western blot检测mRNA和蛋白水平,激光共聚焦成像观察核因子κB (NF-κB) p65的核易位。此外,利用表面等离子体共振(SPR)和分子对接技术验证了RUT组分与肿瘤坏死因子α (TNF-α)靶点的结合能力。结果:RUT降低了浸润性中性粒细胞和单核细胞的水平,降低了促炎细胞因子白细胞介素1β (IL-1β)和白细胞介素6 (IL-6)的水平。结论:RUT减轻了MSU诱导的腹膜炎,抑制了TNFR1-MAPK/NF-κB和NLRP3炎性小体信号通路,减轻了LPS/MSU诱导的THP-1巨噬细胞的痛风炎症,提示RUT可能是痛风的潜在治疗候选药物。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
Rutaecarpine Attenuates Monosodium Urate Crystal-Induced Gouty Inflammation via Inhibition of TNFR-MAPK/NF-κB and NLRP3 Inflammasome Signaling Pathways.

Objective: To investigate the anti-inflammatory effect of rutaecarpine (RUT) on monosodium urate crystal (MSU)-induced murine peritonitis in mice and further explored the underlying mechanism of RUT in lipopolysaccharide (LPS)/MSU-induced gout model in vitro.

Methods: In MSU-induced mice, 36 male C57BL/6 mice were randomly divided into 6 groups of 8 mice each group, including the control group, model group, RUT low-, medium-, and high-doses groups, and prednisone acetate group. The mice in each group were orally administered the corresponding drugs or vehicle once a day for 7 consecutive days. The gout inflammation model was established by intraperitoneal injection of MSU to evaluate the anti-gout inflammatory effects of RUT. Then the proinflammatory cytokines were measured by enzyme-linked immunosorbent assay (ELISA) and the proportions of infiltrating neutrophils cytokines were detected by flow cytometry. In LPS/MSU-treated or untreated THP-1 macrophages, cell viability was observed by cell counting kit 8 and proinflammatory cytokines were measured by ELISA. The percentage of pyroptotic cells were detected by flow cytometry. Respectively, the mRNA and protein levels were measured by real-time quantitative polymerase chain reaction (qRT-PCR) and Western blot, the nuclear translocation of nuclear factor κB (NF-κB) p65 was observed by laser confocal imaging. Additionally, surface plasmon resonance (SPR) and molecular docking were applied to validate the binding ability of RUT components to tumor necrosis factor α (TNF-α) targets.

Results: RUT reduced the levels of infiltrating neutrophils and monocytes and decreased the levels of the proinflammatory cytokines interleukin 1β (IL-1β) and interleukin 6 (IL-6, all P<0.01). In vitro, RUT reduced the production of IL-1β, IL-6 and TNF-α. In addition, RT-PCR revealed the inhibitory effects of RUT on the mRNA levels of IL-1β, IL-6, cyclooxygenase-2 and TNF-α (P<0.05 or P<0.01). Mechanistically, RUT markedly reduced protein expressions of tumor necrosis factor receptor (TNFR), phospho-mitogen-activated protein kinase (p-MAPK), phospho-extracellular signal-regulated kinase, phospho-c-Jun N-terminal kinase, phospho-NF-κB, phospho-kinase α/β, NOD-like receptor thermal protein domain associated protein 3 (NLRPS), cleaved-cysteinyl aspartate specific proteinase-1 and cleaved-gasdermin D in macrophages (P<0.05 or P<0.01). Molecularly, SPR revealed that RUT bound to TNF-α with a calculated equilibrium dissociation constant of 31.7 µmol/L. Molecular docking further confirmed that RUT could interact directly with the TNF-α protein via hydrogen bonding, van der Waals interactions, and carbon-hydrogen bonding.

Conclusion: RUT alleviated MSU-induced peritonitis and inhibited the TNFR1-MAPK/NF-κB and NLRP3 inflammasome signaling pathway to attenuate gouty inflammation induced by LPS/MSU in THP-1 macrophages, suggesting that RUT could be a potential therapeutic candidate for gout.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
Chinese Journal of Integrative Medicine
Chinese Journal of Integrative Medicine 医学-全科医学与补充医学
CiteScore
5.90
自引率
3.40%
发文量
2413
审稿时长
3 months
期刊介绍: Chinese Journal of Integrative Medicine seeks to promote international communication and exchange on integrative medicine as well as complementary and alternative medicine (CAM) and provide a rapid forum for the dissemination of scientific articles focusing on the latest developments and trends as well as experiences and achievements on integrative medicine or CAM in clinical practice, scientific research, education and healthcare.
×
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