Shuang Li, Zheng Tao, Nan Wang, Yazhou Liu, Kai Xie, Haitao Ma
{"title":"综合单细胞和大量转录组分析揭示了食管鳞状细胞癌中T细胞有丝分裂突变相关基因的景观。","authors":"Shuang Li, Zheng Tao, Nan Wang, Yazhou Liu, Kai Xie, Haitao Ma","doi":"10.1186/s40246-025-00815-9","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Mitotic catastrophe (MC) is a well-recognized endogenous mechanism of tumor cell death, characterized as a delayed cell death process associated with aberrant mitosis. However, its prognostic significance in the context of intratumoral heterogeneity in esophageal squamous cell carcinoma (ESCC) remains largely unexplored.</p><p><strong>Methods: </strong>We performed an in-depth analysis of single-cell RNA sequencing (scRNA-seq) data from ESCC obtained from the Gene Expression Omnibus (GEO) database. MC scores for individual cells were calculated using the AddModuleScore function, and T cell specific gene modules were identified via the high-dimensional weighted gene co-expression network analysis (hdWGCNA) framework. To further elucidate the developmental trajectories and intercellular interactions of T cells, pseudotime analysis and cell-cell communication inference were conducted. A prognostic risk model was then constructed using three machine learning algorithms combined with multivariate Cox regression analysis. Following risk stratification, we performed immune infiltration profiling, drug sensitivity analysis, and molecular docking to comprehensively assess the functional implications of the risk model in ESCC. Based on preliminary results from quantitative Real-time PCR (qRT-PCR) and Western blotting (WB), we selected the hub gene SLF2 for functional validation using wound healing, Cell Counting Kit-8 (CCK-8) assay, Transwell, and colony formation assays.</p><p><strong>Results: </strong>Based on T cell mitotic catastrophe associated genes (MCAGs) and utilizing machine learning algorithms, we established a robust prognostic risk model for ESCC. The model demonstrated excellent stratification capability in predicting patient outcomes and effectively revealed the heterogeneity of the tumor immune microenvironment (TIME) and drug sensitivity. Furthermore, functional experiments confirmed that knockdown of the hub gene SLF2 significantly inhibited the migration, invasion, and proliferation of ESCC cells.</p><p><strong>Conclusion: </strong>The prognostic model based on MCAGs we developed serves as an effective tool for predicting outcomes in ESCC.T cell-specific MCAGs drive intratumoral heterogeneity in ESCC, serving as potential prognostic biomarkers and therapeutic targets.</p>","PeriodicalId":13183,"journal":{"name":"Human Genomics","volume":"19 1","pages":"99"},"PeriodicalIF":4.3000,"publicationDate":"2025-08-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12398072/pdf/","citationCount":"0","resultStr":"{\"title\":\"Integrative single-cell and bulk transcriptomic analysis reveals the landscape of T cell mitotic catastrophe associated genes in esophageal squamous cell carcinoma.\",\"authors\":\"Shuang Li, Zheng Tao, Nan Wang, Yazhou Liu, Kai Xie, Haitao Ma\",\"doi\":\"10.1186/s40246-025-00815-9\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background: </strong>Mitotic catastrophe (MC) is a well-recognized endogenous mechanism of tumor cell death, characterized as a delayed cell death process associated with aberrant mitosis. However, its prognostic significance in the context of intratumoral heterogeneity in esophageal squamous cell carcinoma (ESCC) remains largely unexplored.</p><p><strong>Methods: </strong>We performed an in-depth analysis of single-cell RNA sequencing (scRNA-seq) data from ESCC obtained from the Gene Expression Omnibus (GEO) database. MC scores for individual cells were calculated using the AddModuleScore function, and T cell specific gene modules were identified via the high-dimensional weighted gene co-expression network analysis (hdWGCNA) framework. To further elucidate the developmental trajectories and intercellular interactions of T cells, pseudotime analysis and cell-cell communication inference were conducted. A prognostic risk model was then constructed using three machine learning algorithms combined with multivariate Cox regression analysis. Following risk stratification, we performed immune infiltration profiling, drug sensitivity analysis, and molecular docking to comprehensively assess the functional implications of the risk model in ESCC. Based on preliminary results from quantitative Real-time PCR (qRT-PCR) and Western blotting (WB), we selected the hub gene SLF2 for functional validation using wound healing, Cell Counting Kit-8 (CCK-8) assay, Transwell, and colony formation assays.</p><p><strong>Results: </strong>Based on T cell mitotic catastrophe associated genes (MCAGs) and utilizing machine learning algorithms, we established a robust prognostic risk model for ESCC. The model demonstrated excellent stratification capability in predicting patient outcomes and effectively revealed the heterogeneity of the tumor immune microenvironment (TIME) and drug sensitivity. Furthermore, functional experiments confirmed that knockdown of the hub gene SLF2 significantly inhibited the migration, invasion, and proliferation of ESCC cells.</p><p><strong>Conclusion: </strong>The prognostic model based on MCAGs we developed serves as an effective tool for predicting outcomes in ESCC.T cell-specific MCAGs drive intratumoral heterogeneity in ESCC, serving as potential prognostic biomarkers and therapeutic targets.</p>\",\"PeriodicalId\":13183,\"journal\":{\"name\":\"Human Genomics\",\"volume\":\"19 1\",\"pages\":\"99\"},\"PeriodicalIF\":4.3000,\"publicationDate\":\"2025-08-29\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12398072/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Human Genomics\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1186/s40246-025-00815-9\",\"RegionNum\":3,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q2\",\"JCRName\":\"GENETICS & HEREDITY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Human Genomics","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1186/s40246-025-00815-9","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"GENETICS & HEREDITY","Score":null,"Total":0}
引用次数: 0
摘要
背景:有丝分裂突变(Mitotic catastrophe, MC)是一种公认的肿瘤细胞死亡的内源性机制,其特征是与异常有丝分裂相关的延迟细胞死亡过程。然而,其在食管鳞状细胞癌(ESCC)肿瘤内异质性背景下的预后意义仍未得到充分探讨。方法:我们对从Gene Expression Omnibus (GEO)数据库中获得的ESCC单细胞RNA测序(scRNA-seq)数据进行了深入分析。使用AddModuleScore函数计算单个细胞的MC评分,并通过高维加权基因共表达网络分析(hdWGCNA)框架鉴定T细胞特异性基因模块。为了进一步阐明T细胞的发育轨迹和细胞间相互作用,我们进行了伪时间分析和细胞间通讯推断。然后使用三种机器学习算法结合多变量Cox回归分析构建预后风险模型。在风险分层之后,我们进行了免疫浸润分析、药物敏感性分析和分子对接,以全面评估风险模型在ESCC中的功能意义。基于定量实时荧光定量PCR (qRT-PCR)和Western blotting (WB)的初步结果,我们选择了中心基因SLF2进行伤口愈合、细胞计数试剂盒-8 (CCK-8)试验、Transwell和菌落形成试验的功能验证。结果:基于T细胞有丝分裂灾难相关基因(MCAGs)并利用机器学习算法,我们建立了ESCC的稳健预后风险模型。该模型在预测患者预后方面表现出出色的分层能力,并有效揭示了肿瘤免疫微环境(TIME)和药物敏感性的异质性。此外,功能实验证实,敲低枢纽基因SLF2可显著抑制ESCC细胞的迁移、侵袭和增殖。结论:我们建立的基于MCAGs的预后模型是预测ESCC预后的有效工具。T细胞特异性MCAGs驱动ESCC肿瘤内异质性,作为潜在的预后生物标志物和治疗靶点。
Integrative single-cell and bulk transcriptomic analysis reveals the landscape of T cell mitotic catastrophe associated genes in esophageal squamous cell carcinoma.
Background: Mitotic catastrophe (MC) is a well-recognized endogenous mechanism of tumor cell death, characterized as a delayed cell death process associated with aberrant mitosis. However, its prognostic significance in the context of intratumoral heterogeneity in esophageal squamous cell carcinoma (ESCC) remains largely unexplored.
Methods: We performed an in-depth analysis of single-cell RNA sequencing (scRNA-seq) data from ESCC obtained from the Gene Expression Omnibus (GEO) database. MC scores for individual cells were calculated using the AddModuleScore function, and T cell specific gene modules were identified via the high-dimensional weighted gene co-expression network analysis (hdWGCNA) framework. To further elucidate the developmental trajectories and intercellular interactions of T cells, pseudotime analysis and cell-cell communication inference were conducted. A prognostic risk model was then constructed using three machine learning algorithms combined with multivariate Cox regression analysis. Following risk stratification, we performed immune infiltration profiling, drug sensitivity analysis, and molecular docking to comprehensively assess the functional implications of the risk model in ESCC. Based on preliminary results from quantitative Real-time PCR (qRT-PCR) and Western blotting (WB), we selected the hub gene SLF2 for functional validation using wound healing, Cell Counting Kit-8 (CCK-8) assay, Transwell, and colony formation assays.
Results: Based on T cell mitotic catastrophe associated genes (MCAGs) and utilizing machine learning algorithms, we established a robust prognostic risk model for ESCC. The model demonstrated excellent stratification capability in predicting patient outcomes and effectively revealed the heterogeneity of the tumor immune microenvironment (TIME) and drug sensitivity. Furthermore, functional experiments confirmed that knockdown of the hub gene SLF2 significantly inhibited the migration, invasion, and proliferation of ESCC cells.
Conclusion: The prognostic model based on MCAGs we developed serves as an effective tool for predicting outcomes in ESCC.T cell-specific MCAGs drive intratumoral heterogeneity in ESCC, serving as potential prognostic biomarkers and therapeutic targets.
期刊介绍:
Human Genomics is a peer-reviewed, open access, online journal that focuses on the application of genomic analysis in all aspects of human health and disease, as well as genomic analysis of drug efficacy and safety, and comparative genomics.
Topics covered by the journal include, but are not limited to: pharmacogenomics, genome-wide association studies, genome-wide sequencing, exome sequencing, next-generation deep-sequencing, functional genomics, epigenomics, translational genomics, expression profiling, proteomics, bioinformatics, animal models, statistical genetics, genetic epidemiology, human population genetics and comparative genomics.