CD8T

  • 文章类型: Journal Article
    被认为是转移相关基因,然而,在肿瘤微环境的背景下很少描述NME/NM23核苷二磷酸激酶4(NME4)。为了了解NME4在食管鳞状细胞癌(ESCC)中的免疫学意义,我们使用多重免疫组织化学分析NME4表达的临床病理和预后重要性。然后,在用C57BL/6小鼠品系建立了可以概括人类肿瘤微环境的同基因肿瘤模型后,我们检查了NME4表达的免疫学参与。探索潜在的分子机制,通过定量蛋白质组学和蛋白质微阵列筛选,我们调查了潜在的信号通路.在ESCC患者中,NME4表达的临床病理和预后重要性有限。在体内,单细胞RNA测序显示,NME4显著阻止CD8+T细胞浸润小鼠ESCC的肿瘤微环境.机械上,我们在鼠ESCC细胞系AKR中绘制了由NME4负控制的NFκB2-CCL5轴。总的来说,这些数据表明,NME4对NFκB2-CCL5轴的调节可防止ESCC中CD8+T细胞浸润.
    Thought of as a metastasis-associated gene, however, NME/NM23 nucleoside diphosphate kinase 4 (NME4) has rarely been described in the context of the tumour microenvironment. To understand the immunological implications of NME4 in oesophageal squamous cell carcinoma (ESCC), we used multiplex immunohistochemistry to analyse the clinicopathological and prognostic importance of NME4 expression. Then, after establishing a syngeneic tumour model with a C57BL/6 mouse strain that can recapitulate the tumour microenvironment of humans, we examined the immunological involvement of NME4 expression. To explore the underlying molecular mechanism, via quantitative proteomics and protein microarray screening, we investigated the potential signalling pathways involved. The clinicopathological and prognostic importance of NME4 expression is limited in ESCC patients. In vivo, single-cell RNA sequencing showed that NME4 strikingly prevented CD8+ T cells from infiltrating the tumour microenvironment in murine ESCC. Mechanistically, we mapped out the NFκB2-CCL5 axis that was negatively controlled by NME4 in the murine ESCC cell line AKR. Collectively, these data demonstrated that regulation of NFκB2-CCL5 axis by NME4 prevents CD8+ T cells infiltration in ESCC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    波形蛋白被认为是上皮-间质转化(EMT)的典型标志物,并且与以异常PD-L1表达为特征的肿瘤逃逸有关。然而,在食管鳞状细胞癌(ESCC)中,波形蛋白和PD-L1之间是否存在相关关系尚不清楚.首先通过ESCC组织微阵列中的多重免疫荧光染色,然后是异种移植小鼠模型,分析了波形蛋白在ESCC中的免疫学参与。在体内,在波形蛋白稳定沉默后,C57BL/6小鼠皮下移植AKR细胞。体内结果显示,除了PD-L1和PD-L2的表达,波形蛋白表达与CD8+T细胞浸润呈负相关。机械上,波形蛋白可直接与PD-L1相互作用,促进PD-L1在AKR细胞中的核转位。此外,SEMA6C,STC-2和TRAILR2被鉴定为由波形蛋白调节的细胞因子。与对照相比,STC-2和TRAILR2在与它们自己的初级抗体共培养中的阻断显示募集更多的CD8+T细胞。一起,这些数据强烈表明靶向Vimenin可以克服ESCC中的免疫循环.
    Vimentin has been considered a canonical marker of epithelial-mesenchymal transition (EMT) and is associated with tumor escape characterized by aberrant PD-L1 expression. However, whether there is a relationship between vimentin and PD-L1 in esophageal squamous cell carcinoma (ESCC) remains poorly understood. The immunological involvement of vimentin in ESCC was first analyzed by multiplex immunofluorescence staining in ESCC tissue microarray followed by a xenografted mouse model. In vivo, C57BL/6 mice were subcutaneously transplanted with AKR cells after stable silencing of vimentin. In vivo results showed that in addition to PD-L1 and PD-L2 expression, vimentin expression was inversely correlated with CD8+ T-cell infiltration. Mechanistically, vimentin can directly interact with PD-L1 and promote nuclear translocation of PD-L1 in AKR cells. In addition, SEMA6C, STC-2 and TRAILR2 were identified as cytokines modulated by vimentin. Blockade of STC-2 and TRAILR2 in co-culture with their own primary antibodies was shown to recruit more CD8+ T cells than controls. Together, these data strongly suggest targeting Vimenin to overcome the immune cycle in ESCC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号