Celastrol通过靶向SHP2和上调MHC-I增强黑色素瘤中CD8+T细胞免疫

IF 6.7 1区 医学 Q1 CHEMISTRY, MEDICINAL
Qing Kong , Suqing Liu , Shan He , Zhuyu Luo , Rui Lei , Ruilong Wang , Xiao Liu , Jinfeng Wu
{"title":"Celastrol通过靶向SHP2和上调MHC-I增强黑色素瘤中CD8+T细胞免疫","authors":"Qing Kong ,&nbsp;Suqing Liu ,&nbsp;Shan He ,&nbsp;Zhuyu Luo ,&nbsp;Rui Lei ,&nbsp;Ruilong Wang ,&nbsp;Xiao Liu ,&nbsp;Jinfeng Wu","doi":"10.1016/j.phymed.2025.156731","DOIUrl":null,"url":null,"abstract":"<div><h3>Background</h3><div>Celastrol (CEL) has demonstrated promising anti-cancer properties, yet its specific mechanisms against melanoma remain insufficient. This study investigated the CEL's anti-tumor effects and determined its potential mechanisms in the regulation of MHC-I expression in melanoma. In addition, we also tested its efficacy in sensitizing immune checkpoint inhibitors (ICIs) to melanoma.</div></div><div><h3>Methods</h3><div>CEL's anti-tumor activity was evaluated in B16F10 melanoma-bearing C57BL/6 mice across five groups (control, CEL 0.5 mg/kg, CEL 1 mg/kg, CEL 2 mg/kg, and ICIs), the tumor volume, histopathology, and body weight were assessed. Mechanistic insights were obtained through network pharmacology and RNA sequencing in B16F10 cells. Differential gene and pathway analysis were validated using qRT-PCR, Western blotting, and flow cytometry. CD8+<em>T</em> cell activation and cytotoxicity were analyzed in co-culture with CEL-pretreated B16F10 cells using flow cytometry and ELISA. CEL's interaction with potential targets was determined by molecular docking, surface plasmon resonance (SPR), and siRNA. The synergistic effect of CEL combined with ICIs was confirmed in B16F10-bearing C57BL/6 mice, and tumor-infiltrating T cells were assessed by flow cytometry across four groups (control, CEL, ICIs, CEL+ICIs).</div></div><div><h3>Results</h3><div>CEL exhibited a significant anti-tumor effect in B16F10 melanoma-bearing mice. Mechanistically, CEL-pretreated B16F10 cells notably enhanced CD8+<em>T</em> cell activation and promoted IFNγ and TNFα secretion, leading to B16F10 cell death. CEL upregulated MHC-I expression through activation of the JAK/STAT1 pathway in B16F10 cells. The binding assay revealed that CEL interacted with SHP2, with an affinity of 37.93 μM. When SHP2 was silenced in B16F10 cells by siRNA, CEL failed to induce MHC-I upregulation. Moreover, CEL combined with ICIs produced superior antitumor efficacy compared to ICIs alone, which was accompanied by increased CD8+<em>T</em> cell infiltration in melanoma.</div></div><div><h3>Conclusion</h3><div>CEL enhanced CD8+<em>T</em> cell immunity by upregulating MHC-I expression in melanoma cells, these effects were at least partially through targeting SHP2 and activating JAK/STAT1 pathway. CEL might be a novel sensitizer for ICIs in melanoma.</div></div>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"142 ","pages":"Article 156731"},"PeriodicalIF":6.7000,"publicationDate":"2025-04-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Celastrol enhanced CD8+T cell immunity in melanoma by targeting SHP2 and upregulating MHC-I\",\"authors\":\"Qing Kong ,&nbsp;Suqing Liu ,&nbsp;Shan He ,&nbsp;Zhuyu Luo ,&nbsp;Rui Lei ,&nbsp;Ruilong Wang ,&nbsp;Xiao Liu ,&nbsp;Jinfeng Wu\",\"doi\":\"10.1016/j.phymed.2025.156731\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<div><h3>Background</h3><div>Celastrol (CEL) has demonstrated promising anti-cancer properties, yet its specific mechanisms against melanoma remain insufficient. This study investigated the CEL's anti-tumor effects and determined its potential mechanisms in the regulation of MHC-I expression in melanoma. In addition, we also tested its efficacy in sensitizing immune checkpoint inhibitors (ICIs) to melanoma.</div></div><div><h3>Methods</h3><div>CEL's anti-tumor activity was evaluated in B16F10 melanoma-bearing C57BL/6 mice across five groups (control, CEL 0.5 mg/kg, CEL 1 mg/kg, CEL 2 mg/kg, and ICIs), the tumor volume, histopathology, and body weight were assessed. Mechanistic insights were obtained through network pharmacology and RNA sequencing in B16F10 cells. Differential gene and pathway analysis were validated using qRT-PCR, Western blotting, and flow cytometry. CD8+<em>T</em> cell activation and cytotoxicity were analyzed in co-culture with CEL-pretreated B16F10 cells using flow cytometry and ELISA. CEL's interaction with potential targets was determined by molecular docking, surface plasmon resonance (SPR), and siRNA. The synergistic effect of CEL combined with ICIs was confirmed in B16F10-bearing C57BL/6 mice, and tumor-infiltrating T cells were assessed by flow cytometry across four groups (control, CEL, ICIs, CEL+ICIs).</div></div><div><h3>Results</h3><div>CEL exhibited a significant anti-tumor effect in B16F10 melanoma-bearing mice. Mechanistically, CEL-pretreated B16F10 cells notably enhanced CD8+<em>T</em> cell activation and promoted IFNγ and TNFα secretion, leading to B16F10 cell death. CEL upregulated MHC-I expression through activation of the JAK/STAT1 pathway in B16F10 cells. The binding assay revealed that CEL interacted with SHP2, with an affinity of 37.93 μM. When SHP2 was silenced in B16F10 cells by siRNA, CEL failed to induce MHC-I upregulation. Moreover, CEL combined with ICIs produced superior antitumor efficacy compared to ICIs alone, which was accompanied by increased CD8+<em>T</em> cell infiltration in melanoma.</div></div><div><h3>Conclusion</h3><div>CEL enhanced CD8+<em>T</em> cell immunity by upregulating MHC-I expression in melanoma cells, these effects were at least partially through targeting SHP2 and activating JAK/STAT1 pathway. CEL might be a novel sensitizer for ICIs in melanoma.</div></div>\",\"PeriodicalId\":20212,\"journal\":{\"name\":\"Phytomedicine\",\"volume\":\"142 \",\"pages\":\"Article 156731\"},\"PeriodicalIF\":6.7000,\"publicationDate\":\"2025-04-14\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Phytomedicine\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://www.sciencedirect.com/science/article/pii/S0944711325003708\",\"RegionNum\":1,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"CHEMISTRY, MEDICINAL\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Phytomedicine","FirstCategoryId":"3","ListUrlMain":"https://www.sciencedirect.com/science/article/pii/S0944711325003708","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

摘要

背景雌二醇(Celastrol,CEL)具有良好的抗癌特性,但其抗击黑色素瘤的具体机制仍不充分。本研究调查了 CEL 的抗肿瘤作用,并确定了其调节黑色素瘤中 MHC-I 表达的潜在机制。此外,我们还测试了它在使免疫检查点抑制剂(ICIs)对黑色素瘤敏感方面的功效。方法在B16F10黑色素瘤C57BL/6小鼠中评估了CEL的抗肿瘤活性,共分五组(对照组、CEL 0.5 mg/kg组、CEL 1 mg/kg组、CEL 2 mg/kg组和ICIs组),评估了肿瘤体积、组织病理学和体重。通过网络药理学和 B16F10 细胞中的 RNA 测序,获得了对机理的深入了解。利用 qRT-PCR、Western 印迹和流式细胞术验证了差异基因和通路分析。在与 CEL 预处理过的 B16F10 细胞共培养时,使用流式细胞仪和 ELISA 分析了 CD8+T 细胞的活化和细胞毒性。通过分子对接、表面等离子体共振(SPR)和 siRNA 确定了 CEL 与潜在靶点的相互作用。结果CEL在B16F10黑色素瘤小鼠中表现出显著的抗肿瘤效果。从机理上讲,CEL预处理的B16F10细胞显著增强了CD8+T细胞的活化,促进了IFNγ和TNFα的分泌,从而导致B16F10细胞死亡。CEL 通过激活 B16F10 细胞中的 JAK/STAT1 通路上调 MHC-I 的表达。结合试验显示,CEL与SHP2相互作用,亲和力为37.93 μM。当用 siRNA 沉默 B16F10 细胞中的 SHP2 时,CEL 无法诱导 MHC-I 上调。结论CEL通过上调黑色素瘤细胞中MHC-I的表达来增强CD8+T细胞免疫,这些作用至少部分是通过靶向SHP2和激活JAK/STAT1通路实现的。CEL 可能是黑色素瘤 ICIs 的新型增敏剂。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

Celastrol enhanced CD8+T cell immunity in melanoma by targeting SHP2 and upregulating MHC-I

Celastrol enhanced CD8+T cell immunity in melanoma by targeting SHP2 and upregulating MHC-I

Background

Celastrol (CEL) has demonstrated promising anti-cancer properties, yet its specific mechanisms against melanoma remain insufficient. This study investigated the CEL's anti-tumor effects and determined its potential mechanisms in the regulation of MHC-I expression in melanoma. In addition, we also tested its efficacy in sensitizing immune checkpoint inhibitors (ICIs) to melanoma.

Methods

CEL's anti-tumor activity was evaluated in B16F10 melanoma-bearing C57BL/6 mice across five groups (control, CEL 0.5 mg/kg, CEL 1 mg/kg, CEL 2 mg/kg, and ICIs), the tumor volume, histopathology, and body weight were assessed. Mechanistic insights were obtained through network pharmacology and RNA sequencing in B16F10 cells. Differential gene and pathway analysis were validated using qRT-PCR, Western blotting, and flow cytometry. CD8+T cell activation and cytotoxicity were analyzed in co-culture with CEL-pretreated B16F10 cells using flow cytometry and ELISA. CEL's interaction with potential targets was determined by molecular docking, surface plasmon resonance (SPR), and siRNA. The synergistic effect of CEL combined with ICIs was confirmed in B16F10-bearing C57BL/6 mice, and tumor-infiltrating T cells were assessed by flow cytometry across four groups (control, CEL, ICIs, CEL+ICIs).

Results

CEL exhibited a significant anti-tumor effect in B16F10 melanoma-bearing mice. Mechanistically, CEL-pretreated B16F10 cells notably enhanced CD8+T cell activation and promoted IFNγ and TNFα secretion, leading to B16F10 cell death. CEL upregulated MHC-I expression through activation of the JAK/STAT1 pathway in B16F10 cells. The binding assay revealed that CEL interacted with SHP2, with an affinity of 37.93 μM. When SHP2 was silenced in B16F10 cells by siRNA, CEL failed to induce MHC-I upregulation. Moreover, CEL combined with ICIs produced superior antitumor efficacy compared to ICIs alone, which was accompanied by increased CD8+T cell infiltration in melanoma.

Conclusion

CEL enhanced CD8+T cell immunity by upregulating MHC-I expression in melanoma cells, these effects were at least partially through targeting SHP2 and activating JAK/STAT1 pathway. CEL might be a novel sensitizer for ICIs in melanoma.
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
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