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}
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.
Cell DiscoveryBiochemistry, 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.