Targeting RAC1 reactivates pyroptosis to reverse paclitaxel resistance in ovarian cancer by suppressing P21-activated kinase 4

IF 10.7 Q1 MEDICINE, RESEARCH & EXPERIMENTAL MedComm Pub Date : 2024-09-02 DOI:10.1002/mco2.719
Jiangchun Wu, Yong Wu, Tianyi Zhao, Xiangwei Wang, Qinhao Guo, Simin Wang, Siyu Chen, Xingzhu Ju, Jin Li, Xiaohua Wu, Zhong Zheng
{"title":"Targeting RAC1 reactivates pyroptosis to reverse paclitaxel resistance in ovarian cancer by suppressing P21-activated kinase 4","authors":"Jiangchun Wu,&nbsp;Yong Wu,&nbsp;Tianyi Zhao,&nbsp;Xiangwei Wang,&nbsp;Qinhao Guo,&nbsp;Simin Wang,&nbsp;Siyu Chen,&nbsp;Xingzhu Ju,&nbsp;Jin Li,&nbsp;Xiaohua Wu,&nbsp;Zhong Zheng","doi":"10.1002/mco2.719","DOIUrl":null,"url":null,"abstract":"<p>Pyroptosis may play an important role in the resistance of ovarian cancer (OC) to chemotherapy. However, the mechanism by which pyroptosis modulation can attenuate chemotherapy resistance has not been comprehensively studied in OC. Here, we demonstrated that RAS-associated C3 botulinum toxin substrate 1 (RAC1) is highly expressed in OC and is negatively correlated with patient outcomes. Through cell function tests and in vivo tumor formation tests, we found that RAC1 can promote tumor growth by mediating paclitaxel (PTX) resistance. RAC1 can mediate OC progression by inhibiting pyroptosis, as evidenced by high-throughput automated confocal imaging, the release of lactate dehydrogenase (LDH), the expression of the inflammatory cytokines IL-1β/IL-18 and the nucleotide oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome. Mechanically, RNA-seq, gene set enrichment analysis (GSEA), coimmunoprecipitation (Co-IP), mass spectrometry (MS), and ubiquitination tests further confirmed that RAC1 inhibits caspase-1/gasdermin D (GSDMD)-mediated canonical pyroptosis through the P21-activated kinase 4 (PAK4)/mitogen-activated protein kinase (MAPK) pathway, thereby promoting PTX resistance in OC cells. Finally, the whole molecular pathway was verified by the results of in vivo drug combination tests, clinical specimen detection and the prognosis. In summary, our results suggest that the combination of RAC1 inhibitors with PTX can reverse PTX resistance by inducing pyroptosis through the PAK4/MAPK pathway.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":null,"pages":null},"PeriodicalIF":10.7000,"publicationDate":"2024-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11366825/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"MedComm","FirstCategoryId":"1085","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/mco2.719","RegionNum":0,"RegionCategory":null,"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

Pyroptosis may play an important role in the resistance of ovarian cancer (OC) to chemotherapy. However, the mechanism by which pyroptosis modulation can attenuate chemotherapy resistance has not been comprehensively studied in OC. Here, we demonstrated that RAS-associated C3 botulinum toxin substrate 1 (RAC1) is highly expressed in OC and is negatively correlated with patient outcomes. Through cell function tests and in vivo tumor formation tests, we found that RAC1 can promote tumor growth by mediating paclitaxel (PTX) resistance. RAC1 can mediate OC progression by inhibiting pyroptosis, as evidenced by high-throughput automated confocal imaging, the release of lactate dehydrogenase (LDH), the expression of the inflammatory cytokines IL-1β/IL-18 and the nucleotide oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome. Mechanically, RNA-seq, gene set enrichment analysis (GSEA), coimmunoprecipitation (Co-IP), mass spectrometry (MS), and ubiquitination tests further confirmed that RAC1 inhibits caspase-1/gasdermin D (GSDMD)-mediated canonical pyroptosis through the P21-activated kinase 4 (PAK4)/mitogen-activated protein kinase (MAPK) pathway, thereby promoting PTX resistance in OC cells. Finally, the whole molecular pathway was verified by the results of in vivo drug combination tests, clinical specimen detection and the prognosis. In summary, our results suggest that the combination of RAC1 inhibitors with PTX can reverse PTX resistance by inducing pyroptosis through the PAK4/MAPK pathway.

Abstract Image

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
通过抑制 P21 激活激酶 4,靶向 RAC1 可重新激活热蛋白沉积,从而逆转卵巢癌对紫杉醇的耐药性。
化脓过程可能在卵巢癌(OC)的化疗耐药性中扮演重要角色。然而,关于化疗耐药性的减弱机制尚未在卵巢癌中得到全面研究。在这里,我们证明了RAS相关的C3肉毒毒素底物1(RAC1)在OC中高表达,并与患者的预后呈负相关。通过细胞功能测试和体内肿瘤形成测试,我们发现 RAC1 可通过介导紫杉醇(PTX)抗性促进肿瘤生长。通过高通量自动共聚焦成像、乳酸脱氢酶(LDH)的释放、炎性细胞因子IL-1β/IL-18和含核苷酸寡聚化结构域样受体家族吡啉结构域3(NLRP3)炎性小体的表达,RAC1可通过抑制热变态反应介导OC的进展。从机制上看,RNA-seq、基因组富集分析(GSEA)、共免疫沉淀(Co-IP)、质谱(MS)和泛素化检测进一步证实,RAC1通过P21激活激酶4(PAK4)/缄默激活蛋白激酶(MAPK)通路抑制了caspase-1/gasdermin D(GSDMD)介导的典型热蛋白沉积,从而促进了OC细胞对PTX的抗性。最后,体内联合用药试验、临床标本检测和预后结果验证了整个分子通路。综上所述,我们的研究结果表明,RAC1抑制剂与PTX联用可通过PAK4/MAPK通路诱导热凋亡,从而逆转PTX耐药。
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 去求助
来源期刊
CiteScore
6.70
自引率
0.00%
发文量
0
审稿时长
10 weeks
期刊最新文献
Copper homeostasis and cuproptosis in health and disease Liver cirrhosis: molecular mechanisms and therapeutic interventions High serum magnesium level is associated with increased mortality in patients with sepsis: an international, multicenter retrospective study N1-methylpseudouridine modification level correlates with protein expression, immunogenicity, and stability of mRNA Issue Information
×
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
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1