The role of FERMT2 in the tumor microenvironment and immunotherapy in pan-cancer using comprehensive single-cell and bulk sequencing

被引:1
|
作者
Wu, Guang-hao [1 ]
He, Chao [2 ]
Che, Gang [2 ]
Zhou, Zheng [2 ]
Chen, Bi-ying [2 ]
Wu, Hai-ming [3 ]
Chen, Jian-feng [3 ]
Zhu, Wei-pu [4 ]
Yang, Yan [2 ]
Zhou, Zhan [5 ,6 ]
Teng, Li-song [2 ]
Wang, Hai-yong [2 ]
机构
[1] Hangzhou Normal Univ, Sch Clin Med, Med Coll, Hangzhou, Peoples R China
[2] Zhejiang Univ, Affiliated Hosp 1, Sch Med, Dept Surg Oncol, 79 Qingchun Rd, Hangzhou, Peoples R China
[3] Yiwu Cent Hosp, Dept Gastrointestinal Surg, Jinhua, Peoples R China
[4] Zhejiang Univ, Dept Polymer Sci & Engn, MOE Key Lab Macromol Synth & Functionalizat, Hangzhou, Peoples R China
[5] Zhejiang Univ, Inst Drug Metab & Pharmaceut Anal, Hangzhou, Peoples R China
[6] Zhejiang Univ, Coll Pharmaceut Sci, Zhejiang Prov Key Lab Anticanc Drug Res, Hangzhou, Peoples R China
基金
中国国家自然科学基金;
关键词
Pan-cancer; Tumor microenvironment; Single-cell transcriptomics; Cancer-associated fibroblasts; TGF-BETA; FIBROBLASTS; KINDLIN-2; EXPRESSION; PROMOTES;
D O I
10.1016/j.heliyon.2024.e30505
中图分类号
O [数理科学和化学]; P [天文学、地球科学]; Q [生物科学]; N [自然科学总论];
学科分类号
07 ; 0710 ; 09 ;
摘要
FERMT2 has been identified as a participant in integrin-linked kinase signaling pathways, influencing epithelial-mesenchymal transition and thereby affecting tumor initiation, progression, and invasion. While the character of FERMT2 in the tumor microenvironment (TME) as well as its implications for immunotherapy remain unclear. Thus, we conducted a comprehensive analysis to assess the prognostic significance of FERMT2 using Kaplan-Meier analysis. In addition, we employed enrichment analysis to uncover potential underlying molecular mechanisms. Using "Immunedeconv" package, we evaluated the immune characteristics of FERMT2 within TME. Furthermore, we determined the expression levels of FERMT2 in various cell types within TME, based on single-cell sequencing data. To confirm the co-expression of FERMT2 and markers of cancer-associated fibroblasts (CAFs), we performed multiplex immunofluorescence staining on tissue paraffin sections across various cancer types. Our analysis disclosed a significant correlation between elevated FERMT2 expression and unfavorable prognosis in specific cancer types. Furthermore, we identified a strong correlation between FERMT2 expression and diverse immune-related factors, including immune checkpoint molecules, immune cell infiltration, microsatellite instability (MSI), and tumor mutational burden (TMB). Additionally, there was a significant correlation between FERMT2 expression and immune-related pathways, particularly those associated with activating, migrating, and promoting the growth of fibroblasts in diverse cancer types. Interestingly, we observed consistent co-expression of FERMT2 in both malignant tumor cells and stromal cells, particularly within CAFs. Notably, our findings also indicated that FERMT2 , in particular, exhibited elevated expression levels within tumor tissues and co -expressed with alpha-SMA in CAFs based on the multiplex immunofluorescence staining results.
引用
收藏
页数:16
相关论文
共 50 条
  • [21] Single-cell sequencing reveals the role of aggrephagy-related patterns in tumor microenvironment, prognosis and immunotherapy in endometrial cancer
    Yuan, Yuquan
    Ren, Chunyan
    Shu, Jin
    Zhu, Keyang
    Li, Ganghui
    Liu, Bao
    Huang, Jianrong
    Huang, Yinde
    Zhao, Chengzhi
    FRONTIERS IN ONCOLOGY, 2025, 15
  • [22] Comprehensive pan-cancer single-cell analysis reveals glycolysis-related signatures as predictive biomarkers for immunotherapy response and their role in bladder cancer
    Li, Yingjie
    Yang, Wenjie
    Chen, Hualin
    Jin, Zhaoheng
    Dong, Jie
    Ma, Lin
    Ji, Zhigang
    INTERNATIONAL IMMUNOPHARMACOLOGY, 2025, 152
  • [23] A pan-cancer single-cell transcriptional analysis of antigen-presenting cancer-associated fibroblasts in the tumor microenvironment
    Chen, Juntao
    Chen, Renhui
    Huang, Jingang
    FRONTIERS IN IMMUNOLOGY, 2024, 15
  • [24] Integration of Pan-Cancer Single-Cell and Spatial Transcriptomics Reveals Stromal Cell Features and Therapeutic Targets in Tumor Microenvironment
    Du, Yanhua
    Shi, Jintong
    Wang, Jiaxin
    Xun, Zhenzhen
    Yu, Zhuo
    Sun, Hongxiang
    Bao, Rujuan
    Zheng, Junke
    Li, Zhigang
    Ye, Youqiong
    CANCER RESEARCH, 2024, 84 (02) : 192 - 210
  • [25] Single-Cell Informatics for Tumor Microenvironment and Immunotherapy
    Tian, Jiabao
    Bai, Xinyu
    Quek, Camelia
    INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, 2024, 25 (08)
  • [26] Integrative analysis of pan-cancer single-cell data reveals a tumor ecosystem subtype predicting immunotherapy response
    Zeng, Shengjie
    Chen, Liuxun
    Tian, Jinyu
    Liu, Zhengxin
    Liu, Xudong
    Tang, Haibin
    Wu, Hao
    Liu, Chuan
    NPJ PRECISION ONCOLOGY, 2024, 8 (01)
  • [27] Pan-cancer analysis of IFN-? with possible immunotherapeutic significance: a verification of single-cell sequencing and bulk omics research
    Wei, Xiaoying
    Ruan, Hanyi
    Zhang, Yan
    Qin, Tianyu
    Zhang, Yujie
    Qin, Yan
    Li, Wei
    FRONTIERS IN IMMUNOLOGY, 2023, 14
  • [28] Comprehensive analysis of single cell and bulk RNA sequencing reveals the heterogeneity of melanoma tumor microenvironment and predicts the response of immunotherapy
    Zhang, Yuan
    Zhang, Cong
    He, Jing
    Lai, Guichuan
    Li, Wenlong
    Zeng, Haijiao
    Zhong, Xiaoni
    Xie, Biao
    INFLAMMATION RESEARCH, 2024, 73 (08) : 1393 - 1409
  • [29] Immunotherapy and pan-apoptotic characterization of the tumor microenvironment in gastric cancer (STAD): a single-cell multidimensional analysis
    Zhang, Sheng
    Wang, Jianhong
    Zhang, Huan
    Li, Benhua
    Gao, Shun
    DISCOVER ONCOLOGY, 2024, 15 (01)
  • [30] Pan-cancer spatially resolved single-cell analysis reveals the crosstalk between cancer-associated fibroblasts and tumor microenvironment
    Ma, Chenxi
    Yang, Chengzhe
    Peng, Ai
    Sun, Tianyong
    Ji, Xiaoli
    Mi, Jun
    Wei, Li
    Shen, Song
    Feng, Qiang
    MOLECULAR CANCER, 2023, 22 (01)