胃癌腹膜转移时,肿瘤相关巨噬细胞的特异性细胞系转变引起腹腔肿瘤细胞的免疫逃避。

IF 6 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Gastric Cancer Pub Date : 2024-05-01 Epub Date: 2024-03-09 DOI:10.1007/s10120-024-01486-6
Yilin Li, Lei Jiang, Yang Chen, Yanyan Li, Jiajia Yuan, Jialin Lu, Zizhen Zhang, Shengde Liu, Xujiao Feng, Jiaxin Xiong, Yan Jiang, Xiaotian Zhang, Jian Li, Lin Shen
{"title":"胃癌腹膜转移时,肿瘤相关巨噬细胞的特异性细胞系转变引起腹腔肿瘤细胞的免疫逃避。","authors":"Yilin Li, Lei Jiang, Yang Chen, Yanyan Li, Jiajia Yuan, Jialin Lu, Zizhen Zhang, Shengde Liu, Xujiao Feng, Jiaxin Xiong, Yan Jiang, Xiaotian Zhang, Jian Li, Lin Shen","doi":"10.1007/s10120-024-01486-6","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Gastric cancer with peritoneal metastasis (PM-GC), recognized as one of the deadliest cancers. However, whether and how the tumor cell-extrinsic tumor microenvironment (TME) is involved in the therapeutic failure remains unknown. Thus, this study systematically assessed the immunosuppressive tumor microenvironment in ascites from patients with PM-GC, and its contribution to dissemination and immune evasion of ascites-disseminated tumor cells (aDTCs).</p><p><strong>Methods: </strong>Sixty-three ascites and 43 peripheral blood (PB) samples from 51 patients with PM-GC were included in this study. aDTCs in ascites and circulating tumor cells (CTCs) in paired PB were immunophenotypically profiled. Using single-cell RNA transcriptional sequencing (scRNA-seq), crosstalk between aDTCs and the TME features of ascites was inspected. Further studies on the mechanism underlying aDTCs-immune cells crosstalk were performed on in vitro cultured aDTCs.</p><p><strong>Results: </strong>Immune cells in ascites interact with aDTCs, prompting their immune evasion. Specifically, we found that the tumor-associated macrophages (TAMs) in ascites underwent a continuum lineage transition from cathepsin<sup>high</sup> (CTS<sup>high</sup>) to complement 1q<sup>high</sup> (C1Q<sup>high</sup>) TAM. CTS<sup>high</sup> TAM initially attracted the metastatic tumor cells to ascites, thereafter, transitioning terminally to C1Q<sup>high</sup> TAM to trigger overproliferation and immune escape of aDTCs. Mechanistically, we demonstrated that C1Q<sup>high</sup> TAMs significantly enhanced the expression of PD-L1 and NECTIN2 on aDTCs, which was driven by the activation of the C1q-mediated complement pathway.</p><p><strong>Conclusions: </strong>For the first time, we identified an immunosuppressive macrophage transition from CTS<sup>high</sup> to C1Q<sup>high</sup> TAM in ascites from patients with PM-GC. This may contribute to developing potential TAM-targeted immunotherapies for PM-GC.</p>","PeriodicalId":12684,"journal":{"name":"Gastric Cancer","volume":null,"pages":null},"PeriodicalIF":6.0000,"publicationDate":"2024-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11016508/pdf/","citationCount":"0","resultStr":"{\"title\":\"Specific lineage transition of tumor-associated macrophages elicits immune evasion of ascitic tumor cells in gastric cancer with peritoneal metastasis.\",\"authors\":\"Yilin Li, Lei Jiang, Yang Chen, Yanyan Li, Jiajia Yuan, Jialin Lu, Zizhen Zhang, Shengde Liu, Xujiao Feng, Jiaxin Xiong, Yan Jiang, Xiaotian Zhang, Jian Li, Lin Shen\",\"doi\":\"10.1007/s10120-024-01486-6\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background: </strong>Gastric cancer with peritoneal metastasis (PM-GC), recognized as one of the deadliest cancers. However, whether and how the tumor cell-extrinsic tumor microenvironment (TME) is involved in the therapeutic failure remains unknown. Thus, this study systematically assessed the immunosuppressive tumor microenvironment in ascites from patients with PM-GC, and its contribution to dissemination and immune evasion of ascites-disseminated tumor cells (aDTCs).</p><p><strong>Methods: </strong>Sixty-three ascites and 43 peripheral blood (PB) samples from 51 patients with PM-GC were included in this study. aDTCs in ascites and circulating tumor cells (CTCs) in paired PB were immunophenotypically profiled. Using single-cell RNA transcriptional sequencing (scRNA-seq), crosstalk between aDTCs and the TME features of ascites was inspected. Further studies on the mechanism underlying aDTCs-immune cells crosstalk were performed on in vitro cultured aDTCs.</p><p><strong>Results: </strong>Immune cells in ascites interact with aDTCs, prompting their immune evasion. Specifically, we found that the tumor-associated macrophages (TAMs) in ascites underwent a continuum lineage transition from cathepsin<sup>high</sup> (CTS<sup>high</sup>) to complement 1q<sup>high</sup> (C1Q<sup>high</sup>) TAM. CTS<sup>high</sup> TAM initially attracted the metastatic tumor cells to ascites, thereafter, transitioning terminally to C1Q<sup>high</sup> TAM to trigger overproliferation and immune escape of aDTCs. Mechanistically, we demonstrated that C1Q<sup>high</sup> TAMs significantly enhanced the expression of PD-L1 and NECTIN2 on aDTCs, which was driven by the activation of the C1q-mediated complement pathway.</p><p><strong>Conclusions: </strong>For the first time, we identified an immunosuppressive macrophage transition from CTS<sup>high</sup> to C1Q<sup>high</sup> TAM in ascites from patients with PM-GC. This may contribute to developing potential TAM-targeted immunotherapies for PM-GC.</p>\",\"PeriodicalId\":12684,\"journal\":{\"name\":\"Gastric Cancer\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":6.0000,\"publicationDate\":\"2024-05-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11016508/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Gastric Cancer\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1007/s10120-024-01486-6\",\"RegionNum\":1,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"2024/3/9 0:00:00\",\"PubModel\":\"Epub\",\"JCR\":\"Q1\",\"JCRName\":\"GASTROENTEROLOGY & HEPATOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Gastric Cancer","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1007/s10120-024-01486-6","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/3/9 0:00:00","PubModel":"Epub","JCR":"Q1","JCRName":"GASTROENTEROLOGY & HEPATOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

背景:胃癌腹膜转移(PM-GC)是公认的最致命癌症之一。然而,肿瘤细胞外肿瘤微环境(TME)是否以及如何参与治疗失败仍是未知数。因此,本研究系统评估了PM-GC患者腹水中的免疫抑制性肿瘤微环境及其对腹水播散肿瘤细胞(aDTCs)扩散和免疫逃避的贡献:对腹水中的aDTCs和配对PB中的循环肿瘤细胞(CTCs)进行免疫表型分析。通过单细胞 RNA 转录测序(scRNA-seq),检测了腹水中 aDTCs 与 TME 特征之间的相互影响。在体外培养的aDTCs上进一步研究了aDTCs与免疫细胞串扰的机制:结果:腹水中的免疫细胞与 aDTCs 相互作用,促使它们逃避免疫。具体而言,我们发现腹水中的肿瘤相关巨噬细胞(TAMs)经历了从高猫蛋白酶(CTShigh)到高补体1q(C1Qhigh)TAM的连续系转变。CTShigh TAM最初吸引腹水中的转移性肿瘤细胞,随后最终过渡到C1Qhigh TAM,从而引发aDTCs的过度增殖和免疫逃逸。从机理上讲,我们发现高C1Q TAM能显著增强aDTCs上PD-L1和NECTIN2的表达,而这是由C1q介导的补体途径激活所驱动的:我们首次在 PM-GC 患者腹水中发现了巨噬细胞从 CTShigh 向 C1Qhigh TAM 的免疫抑制转变。这可能有助于开发针对 PM-GC 的潜在 TAM 靶向免疫疗法。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

摘要图片

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
Specific lineage transition of tumor-associated macrophages elicits immune evasion of ascitic tumor cells in gastric cancer with peritoneal metastasis.

Background: Gastric cancer with peritoneal metastasis (PM-GC), recognized as one of the deadliest cancers. However, whether and how the tumor cell-extrinsic tumor microenvironment (TME) is involved in the therapeutic failure remains unknown. Thus, this study systematically assessed the immunosuppressive tumor microenvironment in ascites from patients with PM-GC, and its contribution to dissemination and immune evasion of ascites-disseminated tumor cells (aDTCs).

Methods: Sixty-three ascites and 43 peripheral blood (PB) samples from 51 patients with PM-GC were included in this study. aDTCs in ascites and circulating tumor cells (CTCs) in paired PB were immunophenotypically profiled. Using single-cell RNA transcriptional sequencing (scRNA-seq), crosstalk between aDTCs and the TME features of ascites was inspected. Further studies on the mechanism underlying aDTCs-immune cells crosstalk were performed on in vitro cultured aDTCs.

Results: Immune cells in ascites interact with aDTCs, prompting their immune evasion. Specifically, we found that the tumor-associated macrophages (TAMs) in ascites underwent a continuum lineage transition from cathepsinhigh (CTShigh) to complement 1qhigh (C1Qhigh) TAM. CTShigh TAM initially attracted the metastatic tumor cells to ascites, thereafter, transitioning terminally to C1Qhigh TAM to trigger overproliferation and immune escape of aDTCs. Mechanistically, we demonstrated that C1Qhigh TAMs significantly enhanced the expression of PD-L1 and NECTIN2 on aDTCs, which was driven by the activation of the C1q-mediated complement pathway.

Conclusions: For the first time, we identified an immunosuppressive macrophage transition from CTShigh to C1Qhigh TAM in ascites from patients with PM-GC. This may contribute to developing potential TAM-targeted immunotherapies for PM-GC.

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
Gastric Cancer
Gastric Cancer 医学-胃肠肝病学
CiteScore
14.70
自引率
2.70%
发文量
80
审稿时长
6-12 weeks
期刊介绍: Gastric Cancer is an esteemed global forum that focuses on various aspects of gastric cancer research, treatment, and biology worldwide. The journal promotes a diverse range of content, including original articles, case reports, short communications, and technical notes. It also welcomes Letters to the Editor discussing published articles or sharing viewpoints on gastric cancer topics. Review articles are predominantly sought after by the Editor, ensuring comprehensive coverage of the field. With a dedicated and knowledgeable editorial team, the journal is committed to providing exceptional support and ensuring high levels of author satisfaction. In fact, over 90% of published authors have expressed their intent to publish again in our esteemed journal.
期刊最新文献
Failure of evaluation on ABC classification for gastric cancer screening. Correction: An artificial intelligence system for comprehensive pathologic outcome prediction in early gastric cancer through endoscopic image analysis (with video). Survival among patients cured from gastric adenocarcinoma compared to the background population. SMARCA4-deficient undifferentiated gastric carcinoma: a case series and literature review. A machine learning model for predicting the lymph node metastasis of early gastric cancer not meeting the endoscopic curability criteria.
×
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