Tumor necroptosis-mediated shedding of cell surface proteins promotes metastasis of breast cancer by suppressing anti-tumor immunity.

Zhaoshan Liu, Swati Choksi, Hyung-Joon Kwon, Delong Jiao, Chengyu Liu, Zheng-Gang Liu
{"title":"Tumor necroptosis-mediated shedding of cell surface proteins promotes metastasis of breast cancer by suppressing anti-tumor immunity.","authors":"Zhaoshan Liu,&nbsp;Swati Choksi,&nbsp;Hyung-Joon Kwon,&nbsp;Delong Jiao,&nbsp;Chengyu Liu,&nbsp;Zheng-Gang Liu","doi":"10.1186/s13058-023-01604-9","DOIUrl":null,"url":null,"abstract":"<p><p>Necroptosis is a form of regulated necrosis and is executed by MLKL when MLKL is engaged in triggering the rupture of cell plasma membrane. MLKL activation also leads to the protease, ADAMs-mediated ectodomain shedding of cell surface proteins of necroptotic cells. Tumor necroptosis often happens in advanced solid tumors, and blocking necroptosis by MLKL deletion in breast cancer dramatically reduces tumor metastasis. It has been suggested that tumor necroptosis affects tumor progression through modulating the tumor microenvironment. However, the exact mechanism by which tumor necroptosis promotes tumor metastasis remains elusive. Here, we report that the ectodomain shedding of cell surface proteins of necroptotic cells is critical for the promoting effect of tumor necroptosis in tumor metastasis through inhibiting the anti-tumor activity of T cells. We found that blocking tumor necroptosis by MLKL deletion led to the dramatic reduction of tumor metastasis and significantly elevated anti-tumor activity of tumor-infiltrating and peripheral blood T cells. Importantly, the increased anti-tumor activity of T cells is a key cause for the reduced metastasis as the depletion of CD8+ T cells completely restored the level of metastasis in the Mlkl KO mice. Interestingly, the levels of some soluble cell surface proteins including sE-cadherin that are known to promote metastasis are also dramatically reduced in MLKL null tumors/mice. Administration of ADAMs pan inhibitor reduces the levels of soluble cell surface proteins in WT tumors/mice and leads to the dramatic decrease in metastasis. Finally, we showed the sE-cadherin/KLRG1 inhibitory receptor is the major pathway for necroptosis-mediated suppression of the anti-tumor activity of T cells and the promotion of metastasis. Hence, our study reveals a novel mechanism of tumor necroptosis-mediated promotion of metastasis and suggests that tumor necroptosis and necroptosis-activated ADAMs are potential targets for controlling metastasis.</p>","PeriodicalId":9283,"journal":{"name":"Breast Cancer Research : BCR","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2023-01-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9881343/pdf/","citationCount":"4","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Breast Cancer Research : BCR","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1186/s13058-023-01604-9","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 4

Abstract

Necroptosis is a form of regulated necrosis and is executed by MLKL when MLKL is engaged in triggering the rupture of cell plasma membrane. MLKL activation also leads to the protease, ADAMs-mediated ectodomain shedding of cell surface proteins of necroptotic cells. Tumor necroptosis often happens in advanced solid tumors, and blocking necroptosis by MLKL deletion in breast cancer dramatically reduces tumor metastasis. It has been suggested that tumor necroptosis affects tumor progression through modulating the tumor microenvironment. However, the exact mechanism by which tumor necroptosis promotes tumor metastasis remains elusive. Here, we report that the ectodomain shedding of cell surface proteins of necroptotic cells is critical for the promoting effect of tumor necroptosis in tumor metastasis through inhibiting the anti-tumor activity of T cells. We found that blocking tumor necroptosis by MLKL deletion led to the dramatic reduction of tumor metastasis and significantly elevated anti-tumor activity of tumor-infiltrating and peripheral blood T cells. Importantly, the increased anti-tumor activity of T cells is a key cause for the reduced metastasis as the depletion of CD8+ T cells completely restored the level of metastasis in the Mlkl KO mice. Interestingly, the levels of some soluble cell surface proteins including sE-cadherin that are known to promote metastasis are also dramatically reduced in MLKL null tumors/mice. Administration of ADAMs pan inhibitor reduces the levels of soluble cell surface proteins in WT tumors/mice and leads to the dramatic decrease in metastasis. Finally, we showed the sE-cadherin/KLRG1 inhibitory receptor is the major pathway for necroptosis-mediated suppression of the anti-tumor activity of T cells and the promotion of metastasis. Hence, our study reveals a novel mechanism of tumor necroptosis-mediated promotion of metastasis and suggests that tumor necroptosis and necroptosis-activated ADAMs are potential targets for controlling metastasis.

Abstract Image

Abstract Image

Abstract Image

肿瘤坏死介导的细胞表面蛋白的脱落通过抑制抗肿瘤免疫促进乳腺癌的转移。
坏死下垂是一种受调控的坏死形式,当MLKL参与触发细胞膜破裂时,由MLKL执行。MLKL的激活也会导致坏死细胞的蛋白酶、adams介导的细胞表面蛋白的外畴脱落。肿瘤坏死下垂常发生在晚期实体瘤中,在乳腺癌中通过MLKL缺失阻断坏死下垂可显著减少肿瘤转移。有研究认为,肿瘤坏死下垂通过调节肿瘤微环境影响肿瘤进展。然而,肿瘤坏死下垂促进肿瘤转移的确切机制尚不清楚。在这里,我们报道了坏死细胞表面蛋白的外畴脱落是通过抑制T细胞的抗肿瘤活性来促进肿瘤坏死在肿瘤转移中的作用的关键。我们发现,通过缺失MLKL阻断肿瘤坏死下垂可显著减少肿瘤转移,并显著提高肿瘤浸润细胞和外周血T细胞的抗肿瘤活性。重要的是,T细胞抗肿瘤活性的增加是减少转移的关键原因,因为CD8+ T细胞的消耗完全恢复了Mlkl KO小鼠的转移水平。有趣的是,一些可溶性细胞表面蛋白(包括已知促进转移的sE-cadherin)的水平在MLKL阴性肿瘤/小鼠中也显著降低。施用ADAMs pan抑制剂可降低WT肿瘤/小鼠中可溶性细胞表面蛋白的水平,并导致转移的显著减少。最后,我们发现sE-cadherin/KLRG1抑制受体是坏死介导的T细胞抗肿瘤活性抑制和促进转移的主要途径。因此,我们的研究揭示了肿瘤坏死坏死介导的促进转移的新机制,并提示肿瘤坏死下垂和坏死坏死激活的ADAMs是控制转移的潜在靶点。
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
自引率
0.00%
发文量
0
×
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