Wu Jun, Dai Tenghui, Li Ziyue, Pan Meng, Zhang Wei, Chen Hao, Zheng Guansheng, Qiao Li, Lian Qizhou, Liu Yang, Chen Jierong
Laboratory Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China.
School of Biomedical Science, Hunan University, Changsha, 410082, China.
Cell Oncol (Dordr). 2025 Aug 25. doi: 10.1007/s13402-025-01100-6.
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.
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.
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.
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.
胰腺导管腺癌(PDAC)仍然是最致命的恶性肿瘤之一,这主要归因于其高度免疫抑制和纤维化的肿瘤微环境(TME)。然而,其细胞成分的空间和功能组织仍知之甚少。
我们通过整合单细胞RNA测序(n = 88;187,520个细胞)、Visium空间转录组学(n = 20;67,933个斑点)、批量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),呈现了PDAC TME的综合转录组图谱。关键发现通过单细胞RNA测序、批量数据集、多重免疫组织化学和空间成像进行了验证。
POSTN⁺成纤维细胞和SPP1⁺巨噬细胞在12个独立的批量RNA测序队列中持续共同浸润,并在Visium和Xenium平台上显示出空间相关性。那些促肿瘤细胞状态在缺氧、血管生成和上皮-间质转化中富集,并与预后不良相关。相比之下,CCL4⁺ CD8⁺效应T细胞和IGHG1⁺浆细胞在免疫活性龛中共存,富含细胞毒性和激活相关途径,并与患者生存率提高相关。值得注意的是,尽管PDAC TME具有免疫抑制性质,但这些保护性免疫亚群仍可检测到。POSTN、SPP1、CCL4和IGHG1的表达特异性在转录组和蛋白质水平上得到了验证。
我们在PDAC TME中描绘了两个相反的细胞程序——促肿瘤基质重塑和抗肿瘤免疫激活——在不同的龛中进行空间组织。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。