EGCG targeting STAT3 transcriptionally represses PLXNC1 to inhibit M2 polarization mediated by gastric cancer cell-derived exosomal miR-92b-5p.

IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL
Jianfeng Yi, Zhenzhen Ye, Hao Xu, Hui Zhang, Hongtai Cao, Xin Li, Tianming Wang, Chunlu Dong, Yan Du, Shi Dong, Wence Zhou
{"title":"EGCG targeting STAT3 transcriptionally represses PLXNC1 to inhibit M2 polarization mediated by gastric cancer cell-derived exosomal miR-92b-5p.","authors":"Jianfeng Yi, Zhenzhen Ye, Hao Xu, Hui Zhang, Hongtai Cao, Xin Li, Tianming Wang, Chunlu Dong, Yan Du, Shi Dong, Wence Zhou","doi":"10.1016/j.phymed.2024.156137","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>M2-polarized tumor-associated macrophages (TAMs) predominate in tumor microenvironment (TME) and serve primary functions in tumor progression, including growth, angiogenesis, metastasis, immunosuppression, chemoresistance, and poor prognosis. The reversal of M2 polarization provides a new treatment strategy for cancer. Presently, the molecular mechanisms of M2 polarization have yet to be fully characterized, and there is a lack of effective therapeutic targets and drugs. Cancer cells initiate an immunosuppressive TME by recruiting macrophages and promoting M2 polarization through the secretion of inflammatory factors. Accordingly, blocking cancer cell-induced TAM M2 polarization may present a more effective strategy from the perspective of cancer cells. Hedyotis diffusa Willd (HDW) possesses immunomodulatory and antitumor properties, and is a precious and direct source of small molecule natural products with a dual function of inhibition of tumor growth and tumor cell-mediated M2 polarization.</p><p><strong>Objective: </strong>To identify a new target promoting gastric cancer (GC) cell growth and GC cell-mediated M2 polarization from mRNA profiles of GC cells treated with HDW injection (HDI) and to excavate a natural product from HDI that can regulate related mRNA and inhibit the aforementioned effects.</p><p><strong>Methods: </strong>RNA sequencing (RNA-seq) was used to analyze HDI-regulated differentially expressed mRNAs (HRmRNAs) in MKN45 cells. Weighted gene co-expression network analysis (WGCNA), univariate and multivariate Cox regression analysis, KM survival curves, and association analysis between HRmRNA and clinical characteristics/tumor infiltrating immune cells (TIICs) individually were utilized to screen out the target HRmRNA associated with prognosis and M2 macrophage infiltration in GC. shRNA lentiviral vectors were used for stably silencing, and transient overexpressing plasmids were constructed for overexpression. CCK8, EdU, colony formation, migration and invasion assays were used to validate the function of drugs and molecules in GC. HDI constituent analysis was performed using UHPLC-QE-MS. A network of HDI constituent-hub transcription factor (TF)-HRmRNA was constructed based on RNA-Seq, network pharmacology and TFs prediction. Exosome isolation and identification were performed using ultracentrifugation, NTA, TEM and western blot. Apoptosis and macrophage phenotypes were determined by flow cytometric analysis. Small RNA-Seq made exosomal miRNA identification. Small molecule interaction with targets were analyzed using molecular docking, SPR and CETSA. The direct relationship between transcription factors and promoters was verified using ChIP-QPCR and dual-luciferase reporter gene assay. A nude mice xenograft tumor model was established for vivo validation.</p><p><strong>Results: </strong>HDI inhibited MKN45 cell proliferation, migration, invasion and promoted apoptosis. RNA-Seq identified 2583 HRmRNAs. PLXNC1 was screened out as the target HRmRNA associated with prognosis and M2 macrophage infiltration in GC. PLXNC1 promoted GC cell proliferation and facilitated TAMs M2 polarization by transferring GC cell-derived exosomal miR-92b-5p, inhibiting SOCS7-STAT3 interactions and subsequently activating STAT3 in macrophages. M2 TAMs induced by PLXNC1-mediated GC cell-derived exosomes promoted GC cell migration and invasion. PLXNC1 regulated exosomal miR-92b-5p through the MEK1/MSK1/CREB1 pathway. STAT3 could transcriptionally regulate PLXNC1 expression in GC cells. The network of HDI constituent-hub TF-HRmRNA showed epigallocatechin gallate (EGCG) from HDI targeted STAT3 to transcriptionally regulate PLXNC1 expression. EGCG as a natural product directly bound to STAT3 to diminish its nuclear localization, resulting in the transcriptional repression of PLXNC1 and the reversal of M2 polarization induced by PLXNC1-mediated GC cell-derived exosomes.</p><p><strong>Conclusion: </strong>PLXNC1 is a novel target exerting dual effects on GC cell proliferation and GC cell-mediated M2 polarization. EGCG derived from HDI inhibits GC cell proliferation and targets STAT3 to inhibit M2 polarization induced by PLXNC1-mediated exosomes derived from GC cells, which may be a multi-target therapeutic agent for GC cell proliferation and immune microenvironment.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"135 ","pages":"156137"},"PeriodicalIF":6.7000,"publicationDate":"2024-10-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Phytomedicine","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1016/j.phymed.2024.156137","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"CHEMISTRY, MEDICINAL","Score":null,"Total":0}
引用次数: 0

Abstract

Background: M2-polarized tumor-associated macrophages (TAMs) predominate in tumor microenvironment (TME) and serve primary functions in tumor progression, including growth, angiogenesis, metastasis, immunosuppression, chemoresistance, and poor prognosis. The reversal of M2 polarization provides a new treatment strategy for cancer. Presently, the molecular mechanisms of M2 polarization have yet to be fully characterized, and there is a lack of effective therapeutic targets and drugs. Cancer cells initiate an immunosuppressive TME by recruiting macrophages and promoting M2 polarization through the secretion of inflammatory factors. Accordingly, blocking cancer cell-induced TAM M2 polarization may present a more effective strategy from the perspective of cancer cells. Hedyotis diffusa Willd (HDW) possesses immunomodulatory and antitumor properties, and is a precious and direct source of small molecule natural products with a dual function of inhibition of tumor growth and tumor cell-mediated M2 polarization.

Objective: To identify a new target promoting gastric cancer (GC) cell growth and GC cell-mediated M2 polarization from mRNA profiles of GC cells treated with HDW injection (HDI) and to excavate a natural product from HDI that can regulate related mRNA and inhibit the aforementioned effects.

Methods: RNA sequencing (RNA-seq) was used to analyze HDI-regulated differentially expressed mRNAs (HRmRNAs) in MKN45 cells. Weighted gene co-expression network analysis (WGCNA), univariate and multivariate Cox regression analysis, KM survival curves, and association analysis between HRmRNA and clinical characteristics/tumor infiltrating immune cells (TIICs) individually were utilized to screen out the target HRmRNA associated with prognosis and M2 macrophage infiltration in GC. shRNA lentiviral vectors were used for stably silencing, and transient overexpressing plasmids were constructed for overexpression. CCK8, EdU, colony formation, migration and invasion assays were used to validate the function of drugs and molecules in GC. HDI constituent analysis was performed using UHPLC-QE-MS. A network of HDI constituent-hub transcription factor (TF)-HRmRNA was constructed based on RNA-Seq, network pharmacology and TFs prediction. Exosome isolation and identification were performed using ultracentrifugation, NTA, TEM and western blot. Apoptosis and macrophage phenotypes were determined by flow cytometric analysis. Small RNA-Seq made exosomal miRNA identification. Small molecule interaction with targets were analyzed using molecular docking, SPR and CETSA. The direct relationship between transcription factors and promoters was verified using ChIP-QPCR and dual-luciferase reporter gene assay. A nude mice xenograft tumor model was established for vivo validation.

Results: HDI inhibited MKN45 cell proliferation, migration, invasion and promoted apoptosis. RNA-Seq identified 2583 HRmRNAs. PLXNC1 was screened out as the target HRmRNA associated with prognosis and M2 macrophage infiltration in GC. PLXNC1 promoted GC cell proliferation and facilitated TAMs M2 polarization by transferring GC cell-derived exosomal miR-92b-5p, inhibiting SOCS7-STAT3 interactions and subsequently activating STAT3 in macrophages. M2 TAMs induced by PLXNC1-mediated GC cell-derived exosomes promoted GC cell migration and invasion. PLXNC1 regulated exosomal miR-92b-5p through the MEK1/MSK1/CREB1 pathway. STAT3 could transcriptionally regulate PLXNC1 expression in GC cells. The network of HDI constituent-hub TF-HRmRNA showed epigallocatechin gallate (EGCG) from HDI targeted STAT3 to transcriptionally regulate PLXNC1 expression. EGCG as a natural product directly bound to STAT3 to diminish its nuclear localization, resulting in the transcriptional repression of PLXNC1 and the reversal of M2 polarization induced by PLXNC1-mediated GC cell-derived exosomes.

Conclusion: PLXNC1 is a novel target exerting dual effects on GC cell proliferation and GC cell-mediated M2 polarization. EGCG derived from HDI inhibits GC cell proliferation and targets STAT3 to inhibit M2 polarization induced by PLXNC1-mediated exosomes derived from GC cells, which may be a multi-target therapeutic agent for GC cell proliferation and immune microenvironment.

EGCG靶向STAT3转录抑制PLXNC1,从而抑制胃癌细胞外泌体miR-92b-5p介导的M2极化。
背景:M2 极化的肿瘤相关巨噬细胞(TAMs)在肿瘤微环境(TME)中占主导地位,在肿瘤进展过程中发挥主要功能,包括生长、血管生成、转移、免疫抑制、化疗抵抗和不良预后。逆转 M2 极化为癌症提供了一种新的治疗策略。目前,M2 极化的分子机制尚未完全定性,也缺乏有效的治疗靶点和药物。癌细胞通过招募巨噬细胞并通过分泌炎症因子促进 M2 极化,从而启动免疫抑制 TME。因此,从癌细胞的角度来看,阻断癌细胞诱导的 TAM M2 极化可能是一种更有效的策略。Hedyotis diffusa Willd(HDW)具有免疫调节和抗肿瘤特性,是一种珍贵而直接的小分子天然产物来源,具有抑制肿瘤生长和肿瘤细胞介导的 M2 极化的双重功能:从注射用高密度脂蛋白(HDI)治疗的胃癌细胞的mRNA图谱中发现促进胃癌细胞生长和胃癌细胞介导的M2极化的新靶点,并从HDI中挖掘一种能调节相关mRNA并抑制上述作用的天然产物:方法:利用RNA测序(RNA-seq)分析MKN45细胞中受HDI调控的差异表达mRNAs(HRmRNAs)。利用加权基因共表达网络分析(WGCNA)、单变量和多变量Cox回归分析、KM生存曲线以及HRmRNA与临床特征/肿瘤浸润免疫细胞(TIICs)的关联分析,筛选出与GC预后和M2巨噬细胞浸润相关的目标HRmRNA。使用 CCK8、EdU、集落形成、迁移和侵袭试验来验证药物和分子在 GC 中的功能。利用超高效液相色谱-质谱联用仪(UHPLC-QE-MS)进行了 HDI 成分分析。基于RNA-Seq、网络药理学和TFs预测,构建了HDI成分-中枢转录因子(TF)-HRmRNA网络。利用超速离心、NTA、TEM和Western blot技术分离和鉴定了外泌体。通过流式细胞分析确定了细胞凋亡和巨噬细胞的表型。小 RNA-Seq 对外泌体 miRNA 进行了鉴定。利用分子对接、SPR和CETSA分析了小分子与靶标的相互作用。利用 ChIP-QPCR 和双荧光素酶报告基因试验验证了转录因子与启动子之间的直接关系。建立了裸鼠异种移植肿瘤模型进行体内验证:结果:HDI抑制了MKN45细胞的增殖、迁移和侵袭,并促进了细胞凋亡。RNA-Seq鉴定出2583个HRmRNA。结果:HDI抑制了MKN45细胞的增殖、迁移和侵袭,并促进了细胞凋亡。PLXNC1通过转移GC细胞衍生的外泌体miR-92b-5p、抑制SOCS7-STAT3相互作用并随后激活巨噬细胞中的STAT3,促进了GC细胞增殖并促进了TAMs M2极化。PLXNC1 介导的 GC 细胞衍生外泌体诱导的 M2 TAMs 促进了 GC 细胞的迁移和侵袭。PLXNC1通过MEK1/MSK1/CREB1途径调节外泌体miR-92b-5p。STAT3可转录调控PLXNC1在GC细胞中的表达。HDI成分-枢纽TF-HRmRNA网络显示,HDI中的表没食子儿茶素没食子酸酯(EGCG)可靶向STAT3转录调控PLXNC1的表达。EGCG作为一种天然产物直接与STAT3结合,减少了STAT3的核定位,从而抑制了PLXNC1的转录,并逆转了PLXNC1介导的GC细胞外泌体诱导的M2极化:结论:PLXNC1是一个新靶点,对GC细胞增殖和GC细胞介导的M2极化具有双重作用。从HDI中提取的EGCG可抑制GC细胞增殖,并靶向STAT3抑制PLXNC1介导的GC细胞外泌体诱导的M2极化,这可能是一种针对GC细胞增殖和免疫微环境的多靶点治疗药物。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
Phytomedicine
Phytomedicine 医学-药学
CiteScore
10.30
自引率
5.10%
发文量
670
审稿时长
91 days
期刊介绍: Phytomedicine is a therapy-oriented journal that publishes innovative studies on the efficacy, safety, quality, and mechanisms of action of specified plant extracts, phytopharmaceuticals, and their isolated constituents. This includes clinical, pharmacological, pharmacokinetic, and toxicological studies of herbal medicinal products, preparations, and purified compounds with defined and consistent quality, ensuring reproducible pharmacological activity. Founded in 1994, Phytomedicine aims to focus and stimulate research in this field and establish internationally accepted scientific standards for pharmacological studies, proof of clinical efficacy, and safety of phytomedicines.
×
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