Integrating multi-modal transcriptomics identifies cellular subtypes with distinct roles in PDAC progression.

IF 4.8 2区 医学 Q1 Medicine
Jun Wu, Tenghui Dai, Ziyue Li, Meng Pan, Wei Zhang, Hao Chen, Guansheng Zheng, Li Qiao, Qizhou Lian, Yang Liu, Jierong Chen
{"title":"Integrating multi-modal transcriptomics identifies cellular subtypes with distinct roles in PDAC progression.","authors":"Jun Wu, Tenghui Dai, Ziyue Li, Meng Pan, Wei Zhang, Hao Chen, Guansheng Zheng, Li Qiao, Qizhou Lian, Yang Liu, Jierong Chen","doi":"10.1007/s13402-025-01100-6","DOIUrl":null,"url":null,"abstract":"<p><strong>Purpose: </strong>Pancreatic ductal adenocarcinoma (PDAC) remains one of the deadliest malignancies, largely due to its highly immunosuppressive and fibrotic tumor microenvironment (TME). However, the spatial and functional organization of its cellular components remains poorly understood.</p><p><strong>Methods: </strong>We present an integrated transcriptomic atlas of the PDAC TME by combining single-cell RNA sequencing (n = 88; 187,520 cells), Visium spatial transcriptomics (n = 20; 67,933 spots), bulk RNA sequencing (n = 1,383), and high-resolution Xenium spatial transcriptomics (n = 2; 307,679 cells). Key findings were validated using scRNA-seq, bulk datasets, multiplex immunohistochemistry, and spatial imaging.</p><p><strong>Results: </strong>POSTN⁺ fibroblasts and SPP1⁺ macrophages consistently co-infiltrated across 12 independent bulk RNA-seq cohorts, and showed spatial correlation in both Visium and Xenium platforms. Those tumor-promoting cell states were enriched in hypoxic, angiogenesis, and epithelial-mesenchymal transition, and were linked to poor prognosis. In contrast, CCL4⁺ CD8⁺ effector T cells and IGHG1⁺ plasma cells co-occurred within immune-active niches, were enriched for cytotoxic and activation-related pathways, and were associated with improved patient survival. Notably, these protective immune subsets remained detectable despite the immunosuppressive nature of the PDAC TME. Expression specificity of POSTN, SPP1, CCL4, and IGHG1 was validated at the transcriptomic and protein levels.</p><p><strong>Conclusions: </strong>We delineate two opposing cellular programs in the PDAC TME-tumor-promoting stromal remodeling and anti-tumor immune activation-spatially organized in distinct niches. Those findings suggest that targeting POSTN⁺ fibroblasts and SPP1⁺ macrophages-mediated stromal interactions while promoting CCL4⁺ T cell and IGHG1⁺ plasma cell immunity, may offer new therapeutic strategies for PDAC.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":""},"PeriodicalIF":4.8000,"publicationDate":"2025-08-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cellular Oncology","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1007/s13402-025-01100-6","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"Medicine","Score":null,"Total":0}
引用次数: 0

Abstract

Purpose: Pancreatic ductal adenocarcinoma (PDAC) remains one of the deadliest malignancies, largely due to its highly immunosuppressive and fibrotic tumor microenvironment (TME). However, the spatial and functional organization of its cellular components remains poorly understood.

Methods: We present an integrated transcriptomic atlas of the PDAC TME by combining single-cell RNA sequencing (n = 88; 187,520 cells), Visium spatial transcriptomics (n = 20; 67,933 spots), bulk RNA sequencing (n = 1,383), and high-resolution Xenium spatial transcriptomics (n = 2; 307,679 cells). Key findings were validated using scRNA-seq, bulk datasets, multiplex immunohistochemistry, and spatial imaging.

Results: POSTN⁺ fibroblasts and SPP1⁺ macrophages consistently co-infiltrated across 12 independent bulk RNA-seq cohorts, and showed spatial correlation in both Visium and Xenium platforms. Those tumor-promoting cell states were enriched in hypoxic, angiogenesis, and epithelial-mesenchymal transition, and were linked to poor prognosis. In contrast, CCL4⁺ CD8⁺ effector T cells and IGHG1⁺ plasma cells co-occurred within immune-active niches, were enriched for cytotoxic and activation-related pathways, and were associated with improved patient survival. Notably, these protective immune subsets remained detectable despite the immunosuppressive nature of the PDAC TME. Expression specificity of POSTN, SPP1, CCL4, and IGHG1 was validated at the transcriptomic and protein levels.

Conclusions: We delineate two opposing cellular programs in the PDAC TME-tumor-promoting stromal remodeling and anti-tumor immune activation-spatially organized in distinct niches. Those findings suggest that targeting POSTN⁺ fibroblasts and SPP1⁺ macrophages-mediated stromal interactions while promoting CCL4⁺ T cell and IGHG1⁺ plasma cell immunity, may offer new therapeutic strategies for PDAC.

整合多模态转录组学鉴定在PDAC进展中具有不同作用的细胞亚型。
目的:胰腺导管腺癌(Pancreatic ductal adencarcinoma, PDAC)是最致命的恶性肿瘤之一,主要是由于其高度免疫抑制和纤维化的肿瘤微环境(tumor microenvironment, TME)。然而,其细胞成分的空间和功能组织仍然知之甚少。方法:我们通过结合单细胞RNA测序(n = 88, 187,520个细胞)、Visium空间转录组学(n = 20, 67,933个点)、大量RNA测序(n = 1,383)和高分辨率Xenium空间转录组学(n = 2, 307,679个细胞),构建了PDAC TME的综合转录组图谱。通过scRNA-seq、大量数据集、多重免疫组织化学和空间成像验证了关键发现。结果:POSTN +成纤维细胞和SPP1 +巨噬细胞在12个独立的bulk RNA-seq队列中一致共浸润,并且在Visium和Xenium平台中均表现出空间相关性。这些促肿瘤细胞状态富含缺氧、血管生成和上皮-间质转化,并与预后不良有关。相比之下,CCL4 + CD8 +效应T细胞和IGHG1 +浆细胞在免疫活性壁龛内共存,富集了细胞毒性和激活相关途径,并与患者生存率的提高有关。值得注意的是,尽管PDAC TME具有免疫抑制特性,但这些保护性免疫亚群仍可检测到。在转录组学和蛋白水平上验证了POSTN、SPP1、CCL4和IGHG1的表达特异性。结论:我们描述了PDAC中两个相反的细胞程序-促肿瘤间质重塑和抗肿瘤免疫激活-在空间上组织在不同的生态位。这些发现表明,靶向POSTN +成纤维细胞和SPP1 +巨噬细胞介导的基质相互作用,同时促进CCL4 + T细胞和IGHG1 +浆细胞免疫,可能为PDAC提供新的治疗策略。
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
Cellular Oncology
Cellular Oncology Biochemistry, Genetics and Molecular Biology-Cancer Research
CiteScore
10.40
自引率
1.50%
发文量
0
审稿时长
16 weeks
期刊介绍: The Official Journal of the International Society for Cellular Oncology Focuses on translational research Addresses the conversion of cell biology to clinical applications Cellular Oncology publishes scientific contributions from various biomedical and clinical disciplines involved in basic and translational cancer research on the cell and tissue level, technical and bioinformatics developments in this area, and clinical applications. This includes a variety of fields like genome technology, micro-arrays and other high-throughput techniques, genomic instability, SNP, DNA methylation, signaling pathways, DNA organization, (sub)microscopic imaging, proteomics, bioinformatics, functional effects of genomics, drug design and development, molecular diagnostics and targeted cancer therapies, genotype-phenotype interactions. A major goal is to translate the latest developments in these fields from the research laboratory into routine patient management. To this end Cellular Oncology forms a platform of scientific information exchange between molecular biologists and geneticists, technical developers, pathologists, (medical) oncologists and other clinicians involved in the management of cancer patients. In vitro studies are preferentially supported by validations in tumor tissue with clinicopathological associations.
×
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