结直肠癌微环境中空间分辨的C1QC+巨噬细胞- cd4 + T细胞生态位:对免疫治疗反应的影响

IF 13 1区 生物学 Q1 CELL BIOLOGY
Hangyu Zhang, Libing Hong, Zhen Dong, Shan Xin, Bo Lin, Jinlin Cheng, Weihong Tian, Bin Li, Jing Wang, Xiaoyan Liu, Chuan Liu, Yuzhi Jin, Yanzhi Feng, Ge Su, Xuqi Sun, Qiqi Liu, Xiaomeng Dai, Yang Gao, Zhou Tong, Lulu Liu, Xudong Zhu, Yi Zheng, Peng Zhao, Tiannan Guo, Weijia Fang, Xuanwen Bao
{"title":"结直肠癌微环境中空间分辨的C1QC+巨噬细胞- cd4 + T细胞生态位:对免疫治疗反应的影响","authors":"Hangyu Zhang, Libing Hong, Zhen Dong, Shan Xin, Bo Lin, Jinlin Cheng, Weihong Tian, Bin Li, Jing Wang, Xiaoyan Liu, Chuan Liu, Yuzhi Jin, Yanzhi Feng, Ge Su, Xuqi Sun, Qiqi Liu, Xiaomeng Dai, Yang Gao, Zhou Tong, Lulu Liu, Xudong Zhu, Yi Zheng, Peng Zhao, Tiannan Guo, Weijia Fang, Xuanwen Bao","doi":"10.1038/s41421-025-00811-2","DOIUrl":null,"url":null,"abstract":"<p><p>Colorectal cancer (CRC), including both microsatellite instability (MSI) and microsatellite stability (MSS) subtypes, frequently exhibits intrinsic resistance to immunotherapy. However, the spatial tumor microenvironment (TME) and its role in distinguishing immunotherapy responders from non-responders remain poorly understood. In this study, spatial multiomics, including imaging mass cytometry (n = 50 in-house), spatial proteomics (n = 50 in-house), and spatial transcriptomics (n = 9 in-house), were employed to elucidate the spatial TME of metastatic CRC (mCRC) patients receiving immunotherapy. These methodologies were integrated with single-cell RNA sequencing (scRNA-seq), bulk RNA-seq, and bulk proteomics for comprehensive analysis and validation. A spatial immune atlas containing 314,774 cells was constructed. We found that C1QC<sup>+</sup> resident tissue macrophages (RTMs) were more abundant in responders regardless of microsatellite status. Co-localization of C1QC<sup>+</sup> RTMs with CD4<sup>+</sup> T cells was observed in responders, and MHC-II expression facilitated their interaction. In contrast, cancer-associated fibroblasts inhibited this interaction in non-responders. Moreover, whole genome screening identified key genes involved in antigen presentation in C1QC<sup>+</sup> RTMs. Hence, our study highlights the importance of spatial immune mapping in revealing the complex spatial topology of CRC and corresponding immunotherapy response.</p>","PeriodicalId":9674,"journal":{"name":"Cell Discovery","volume":"11 1","pages":"60"},"PeriodicalIF":13.0000,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12219098/pdf/","citationCount":"0","resultStr":"{\"title\":\"Spatially resolved C1QC<sup>+</sup> macrophage-CD4<sup>+</sup> T cell niche in colorectal cancer microenvironment: implications for immunotherapy response.\",\"authors\":\"Hangyu Zhang, Libing Hong, Zhen Dong, Shan Xin, Bo Lin, Jinlin Cheng, Weihong Tian, Bin Li, Jing Wang, Xiaoyan Liu, Chuan Liu, Yuzhi Jin, Yanzhi Feng, Ge Su, Xuqi Sun, Qiqi Liu, Xiaomeng Dai, Yang Gao, Zhou Tong, Lulu Liu, Xudong Zhu, Yi Zheng, Peng Zhao, Tiannan Guo, Weijia Fang, Xuanwen Bao\",\"doi\":\"10.1038/s41421-025-00811-2\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><p>Colorectal cancer (CRC), including both microsatellite instability (MSI) and microsatellite stability (MSS) subtypes, frequently exhibits intrinsic resistance to immunotherapy. However, the spatial tumor microenvironment (TME) and its role in distinguishing immunotherapy responders from non-responders remain poorly understood. In this study, spatial multiomics, including imaging mass cytometry (n = 50 in-house), spatial proteomics (n = 50 in-house), and spatial transcriptomics (n = 9 in-house), were employed to elucidate the spatial TME of metastatic CRC (mCRC) patients receiving immunotherapy. These methodologies were integrated with single-cell RNA sequencing (scRNA-seq), bulk RNA-seq, and bulk proteomics for comprehensive analysis and validation. A spatial immune atlas containing 314,774 cells was constructed. We found that C1QC<sup>+</sup> resident tissue macrophages (RTMs) were more abundant in responders regardless of microsatellite status. Co-localization of C1QC<sup>+</sup> RTMs with CD4<sup>+</sup> T cells was observed in responders, and MHC-II expression facilitated their interaction. In contrast, cancer-associated fibroblasts inhibited this interaction in non-responders. Moreover, whole genome screening identified key genes involved in antigen presentation in C1QC<sup>+</sup> RTMs. Hence, our study highlights the importance of spatial immune mapping in revealing the complex spatial topology of CRC and corresponding immunotherapy response.</p>\",\"PeriodicalId\":9674,\"journal\":{\"name\":\"Cell Discovery\",\"volume\":\"11 1\",\"pages\":\"60\"},\"PeriodicalIF\":13.0000,\"publicationDate\":\"2025-07-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12219098/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Cell Discovery\",\"FirstCategoryId\":\"99\",\"ListUrlMain\":\"https://doi.org/10.1038/s41421-025-00811-2\",\"RegionNum\":1,\"RegionCategory\":\"生物学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"CELL BIOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cell Discovery","FirstCategoryId":"99","ListUrlMain":"https://doi.org/10.1038/s41421-025-00811-2","RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"CELL BIOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

结直肠癌(CRC),包括微卫星不稳定性(MSI)和微卫星稳定性(MSS)亚型,经常表现出对免疫治疗的内在抗性。然而,空间肿瘤微环境(TME)及其在区分免疫治疗应答者和无应答者中的作用仍然知之甚少。在这项研究中,空间多组学,包括成像细胞术(n = 50),空间蛋白质组学(n = 50)和空间转录组学(n = 9),被用于阐明接受免疫治疗的转移性结直肠癌(mCRC)患者的空间TME。这些方法与单细胞RNA测序(scRNA-seq)、大量RNA-seq和大量蛋白质组学相结合,进行全面的分析和验证。构建了包含314,774个细胞的空间免疫图谱。我们发现,无论微卫星状态如何,应答者中C1QC+常驻组织巨噬细胞(RTMs)都更丰富。在应答者中观察到C1QC+ RTMs与CD4+ T细胞共定位,MHC-II的表达促进了它们的相互作用。相反,癌症相关成纤维细胞在无应答者中抑制这种相互作用。此外,全基因组筛选鉴定了C1QC+ rtm中参与抗原呈递的关键基因。因此,我们的研究强调了空间免疫图谱在揭示结直肠癌复杂的空间拓扑结构和相应的免疫治疗反应方面的重要性。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
Spatially resolved C1QC+ macrophage-CD4+ T cell niche in colorectal cancer microenvironment: implications for immunotherapy response.

Colorectal cancer (CRC), including both microsatellite instability (MSI) and microsatellite stability (MSS) subtypes, frequently exhibits intrinsic resistance to immunotherapy. However, the spatial tumor microenvironment (TME) and its role in distinguishing immunotherapy responders from non-responders remain poorly understood. In this study, spatial multiomics, including imaging mass cytometry (n = 50 in-house), spatial proteomics (n = 50 in-house), and spatial transcriptomics (n = 9 in-house), were employed to elucidate the spatial TME of metastatic CRC (mCRC) patients receiving immunotherapy. These methodologies were integrated with single-cell RNA sequencing (scRNA-seq), bulk RNA-seq, and bulk proteomics for comprehensive analysis and validation. A spatial immune atlas containing 314,774 cells was constructed. We found that C1QC+ resident tissue macrophages (RTMs) were more abundant in responders regardless of microsatellite status. Co-localization of C1QC+ RTMs with CD4+ T cells was observed in responders, and MHC-II expression facilitated their interaction. In contrast, cancer-associated fibroblasts inhibited this interaction in non-responders. Moreover, whole genome screening identified key genes involved in antigen presentation in C1QC+ RTMs. Hence, our study highlights the importance of spatial immune mapping in revealing the complex spatial topology of CRC and corresponding immunotherapy response.

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
Cell Discovery
Cell Discovery Biochemistry, Genetics and Molecular Biology-Molecular Biology
CiteScore
24.20
自引率
0.60%
发文量
120
审稿时长
20 weeks
期刊介绍: Cell Discovery is a cutting-edge, open access journal published by Springer Nature in collaboration with the Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences (CAS). Our aim is to provide a dynamic and accessible platform for scientists to showcase their exceptional original research. Cell Discovery covers a wide range of topics within the fields of molecular and cell biology. We eagerly publish results of great significance and that are of broad interest to the scientific community. With an international authorship and a focus on basic life sciences, our journal is a valued member of Springer Nature's prestigious Molecular Cell Biology journals. In summary, Cell Discovery offers a fresh approach to scholarly publishing, enabling scientists from around the world to share their exceptional findings in molecular and cell biology.
×
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:604180095
Book学术官方微信