Wang Jiali, Jia Yunlong, Liu Tianxu, Liu Xinyan, Yin Shuxian, Chen Jiaqi, Xu Xiaoqing, Zhang Yi, Liu Lihua
Department of Tumor Immunotherapy, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, 050035, P.R. China.
Department of General Surgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
J Exp Clin Cancer Res. 2025 May 9;44(1):141. doi: 10.1186/s13046-025-03404-9.
Tumors often evade immune surveillance by limiting T cell infiltration. In non-small cell lung cancer (NSCLC), increased infiltration of CD8 T cells is associated with a favorable response to immunotherapy. While BIN1 is recognized as a tumor suppressor gene, its role in shaping the tumor microenvironment in NSCLC has yet to be fully clarified.
To investigate the relationship between BIN1 expression and CD8T cell infiltration in NSCLC, we performed a comprehensive data analysis utilizing clinical information from NSCLC patients. BIN1 expression levels in NSCLC tissues were evaluated, and their correlation with CD8T cells infiltration and patient survival outcomes was examined. Loss-of-function strategies targeting BIN1 were applied in syngeneic NSCLC mouse models to assess its functional significance. Tumor growth was monitored, and immune cell populations were analyzed in terms of frequency and functionality through mass cytometry and flow cytometry techniques. Cytokine secretion was profiled using multiplex assays. Additionally, RNA sequencing, immunoprecipitation-mass spectrometry, and molecular docking were employed to confirm direct interactions between BIN1 and cytokine-encoding genes. Finally, the regulatory role of BIN1 in ferroptosis in NSCLC cells were explored using metabolomics analysis, ROS measurement, and MDA detection.
We observed that BIN1 expression is downregulated in NSCLC tumor tissues, with its reduced expression strongly associated with advanced disease progression and poor prognosis. Bioinformatics analysis of immune infiltration in human NSCLC samples revealed a positive correlation between BIN1 expression in NSCLC tissues and CD8 T cell infiltration. Furthermore, the prognostic impact of BIN1 on NSCLC patients is strongly linked to the level of CD8 T cell infiltration. In syngeneic mouse models, the knockout of BIN1 in NSCLC cells significantly inhibited CD8 T cell infiltration and impaired their cytotoxic function, facilitating tumor immune evasion. Mechanistically, we demonstrated that BIN1 directly interacts with G3BP1, and its knockout stabilizes G3BP1. This, in turn, promotes STAT1 degradation and reduces the secretion of T cell-recruiting chemokines such as CXCL10 and CCL5. Finally, our findings reveal that BIN1 influences ferroptosis in NSCLC cells through the G3BP1/STAT1/GSH pathway, thereby regulating NSCLC cell proliferation, migration, and invasion.
This study highlights the crucial role of the BIN1/G3BP1/STAT1/CD8 tumor-infiltrating lymphocyte signaling pathway in the progression of NSCLC and its mechanisms of immune evasion. This fundings lay a foundation for the development of BIN1-targeted therapies aimed at improving tumor immunogenicity and transforming immunologically "cold" NSCLC into a more responsive disease.
肿瘤常常通过限制T细胞浸润来逃避免疫监视。在非小细胞肺癌(NSCLC)中,CD8 T细胞浸润增加与免疫治疗的良好反应相关。虽然BIN1被认为是一种肿瘤抑制基因,但其在NSCLC肿瘤微环境形成中的作用尚未完全阐明。
为了研究BIN1表达与NSCLC中CD8 T细胞浸润之间的关系,我们利用NSCLC患者的临床信息进行了全面的数据分析。评估了NSCLC组织中BIN1的表达水平,并检查了其与CD8 T细胞浸润及患者生存结果的相关性。在同基因NSCLC小鼠模型中应用针对BIN1的功能缺失策略,以评估其功能意义。监测肿瘤生长,并通过质谱流式细胞术和流式细胞术技术分析免疫细胞群体的频率和功能。使用多重检测分析细胞因子分泌情况。此外,采用RNA测序、免疫沉淀-质谱分析和分子对接来确认BIN1与细胞因子编码基因之间的直接相互作用。最后,利用代谢组学分析、活性氧测量和丙二醛检测,探索BIN1在NSCLC细胞铁死亡中的调节作用。
我们观察到NSCLC肿瘤组织中BIN1表达下调,其表达降低与疾病进展和预后不良密切相关。对人类NSCLC样本的免疫浸润进行生物信息学分析发现,NSCLC组织中BIN1表达与CD8 T细胞浸润呈正相关。此外,BIN1对NSCLC患者的预后影响与CD8 T细胞浸润水平密切相关。在同基因小鼠模型中,敲除NSCLC细胞中的BIN1可显著抑制CD8 T细胞浸润并损害其细胞毒性功能,促进肿瘤免疫逃逸。机制上,我们证明BIN1直接与G3BP1相互作用,其敲除使G3BP1稳定。这反过来又促进STAT1降解,并减少CXCL10和CCL5等T细胞招募趋化因子的分泌。最后,我们的研究结果表明,BIN1通过G3BP1/STAT1/谷胱甘肽途径影响NSCLC细胞的铁死亡,从而调节NSCLC细胞的增殖、迁移和侵袭。
本研究强调了BIN1/G3BP1/STAT1/CD8肿瘤浸润淋巴细胞信号通路在NSCLC进展及其免疫逃逸机制中的关键作用。这些发现为开发旨在提高肿瘤免疫原性并将免疫“冷”的NSCLC转变为更易反应疾病的BIN1靶向治疗奠定了基础。