1
|
Chen Y, Tong Y, Ye X, Yang Y, Li H, Wu H, Zhai W, Li Y, Zhang Q, Zhou L, Sun J, Fan Y. A four gene risk score model for prognosis and immune microenvironment insights in small cell lung cancer based on CAF functional-related genes. Discov Oncol 2025; 16:923. [PMID: 40415048 DOI: 10.1007/s12672-025-02781-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025] Open
Abstract
Small cell lung cancer (SCLC) is still one of the most formidable challenges in oncology. In this study, we introduce an innovative risk scoring model rooted in cancer-associated fibroblast (CAF)-related functional genes, designed to predict patient prognosis and illuminate the microenvironment of SCLC. Through Kaplan-Meier survival analysis and receiver operating characteristic (ROC) curves, our model could effectively classify patients into high- and low-risk groups, with distinct survival outcomes and remarkable predictive accuracy, which has been evidenced by the AUC values. The low-risk patients showed a more active immune environment, characterized by more infiltration of dendritic cells, natural killer cells, and higher expression of immune co-stimulation molecules. On the contrary, high-risk patients displayed an enrichment of DNA repair and glycolysis pathways associated with tumor aggressiveness and treatment resistance. These results suggest that the risk model offers a nuanced view of response to immunotherapy that may guide the identification of patients who may benefit from immunotherapy. Moreover, we also verified the function of the key gene UBE2E2 by SCLC cell line experiments. Silencing UBE2E2 results in decreased cell proliferation and migration as well as increased apoptosis, which enhances its important role in SCLC biology. In summary, our study highlights the prognostic potential of the CAF-related functional gene risk model and its implications for predicting immune microenvironment status and guiding SCLC treatment strategies.
Collapse
Affiliation(s)
- Yunfei Chen
- Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yunfeng Tong
- Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xinyuan Ye
- Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yehao Yang
- Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hui Li
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
| | - Haicheng Wu
- Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou, 325035, China
| | - Wanchen Zhai
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
| | - Yuwei Li
- Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou, 325035, China
| | - Qian Zhang
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
| | - Linjing Zhou
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
| | - Jing Sun
- Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yun Fan
- Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
2
|
Ma K, Mao Q, Fei B, Ni T, Zhang Z, Ni H. Metabolic reprogramming and immune microenvironment characteristics in laryngeal carcinoma: advances in immunotherapy. Front Immunol 2025; 16:1589243. [PMID: 40370437 PMCID: PMC12075248 DOI: 10.3389/fimmu.2025.1589243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 04/08/2025] [Indexed: 05/16/2025] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is a prevalent malignancy with high mortality and recurrence rates, necessitating novel therapeutic strategies. Recent research highlights the pivotal role of metabolic reprogramming and immune microenvironment alterations in LSCC pathogenesis, providing promising avenues for targeted therapy. This review summarizes the metabolic characteristics of LSCC, including glycolysis, lipid metabolism, and amino acid biosynthesis, and their implications for tumor progression and therapeutic resistance. Additionally, this review further describes the tumor microenvironment's immunosuppressive landscape, including immune checkpoint regulation, tumor-associated macrophages, and T-cell dysfunction. The integration of metabolic and immune-targeted strategies represents a promising frontier in LSCC treatment, warranting further investigation.
Collapse
Affiliation(s)
- Kexin Ma
- Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Qingjie Mao
- Department of Otolaryngology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Bing Fei
- Department of Otolaryngology Head and Neck Surgery, Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, China
| | - Tingting Ni
- Department of Oncology, Nantong Tumor Hospital, Nantong, China
| | - Zhenxin Zhang
- Department of Otolaryngology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Haosheng Ni
- Department of Otolaryngology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| |
Collapse
|
3
|
Wang Y, He M, Li J, Li L. Immunotherapeutic strategies for invasive bladder cancer: a comprehensive review. Front Immunol 2025; 16:1591379. [PMID: 40370454 PMCID: PMC12075300 DOI: 10.3389/fimmu.2025.1591379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 04/10/2025] [Indexed: 05/16/2025] Open
Abstract
Bladder cancer is a prevalent malignancy, with muscle-invasive bladder cancer (MIBC) presenting a significant therapeutic challenge. Standard treatments, including radical cystectomy (RC) and neoadjuvant chemotherapy, pose substantial risks and impact quality of life, leading to increasing interest in bladder-preserving therapies (BPT). Immunotherapy has revolutionized bladder cancer management, with strategies ranging from intravesical Bacillus Calmette-Guérin (BCG) to immune checkpoint inhibitors targeting programmed cell death protein 1 (PD-1) and its ligand (PD-L1). In BCG-unresponsive non-muscle-invasive bladder cancer (NMIBC), PD-1 inhibitors such as pembrolizumab offer promising response rates. In MIBC, neoadjuvant immunotherapy with agents like atezolizumab and pembrolizumab improves pathological complete response (pCR) and facilitates bladder preservation. Combination regimens integrating radiotherapy, chemotherapy, and immunotherapy not only enhance treatment efficacy but also exploit mechanisms such as immunogenic cell death and antigen release that further augment antitumor immune responses. This review provides a comprehensive analysis of current immunotherapeutic strategies for invasive bladder cancer, highlighting their clinical applications and future potential.
Collapse
Affiliation(s)
- Yingying Wang
- Department of Pharmacy, The First People’s Hospital of Xiaoshan District, Hangzhou, China
| | - Min He
- Department of Pharmacy, The First People’s Hospital of Xiaoshan District, Hangzhou, China
| | - Jian Li
- Department of Pharmacy, The First People’s Hospital of Xiaoshan District, Hangzhou, China
| | - Li Li
- Department of Emergency, The First People’s Hospital of Xiaoshan District, Hangzhou, China
| |
Collapse
|
4
|
Yu H, Yang R, Li M, Li D, Xu Y. The role of Treg cells in colorectal cancer and the immunotherapy targeting Treg cells. Front Immunol 2025; 16:1574327. [PMID: 40308582 PMCID: PMC12040624 DOI: 10.3389/fimmu.2025.1574327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Colorectal cancer (CRC) is among the most prevalent and lethal cancers globally, accounting for approximately 10% of all cancer cases and deaths. Regulatory T (Treg) cells, which accumulate in CRC tissue, suppress anti-tumor immune responses and facilitate tumor progression. This review discusses Treg cell origins and functions, along with the mechanisms by which Tregs influence CRC development. In addition, we highlight therapeutic strategies targeting Tregs-such as immune checkpoint inhibitors and combinatorial approaches-to enhance effector T cell responses. A deeper understanding of Treg-mediated immunosuppression in CRC may inform the design of more effective immunotherapies and precision medicine strategies.
Collapse
Affiliation(s)
- Hanqing Yu
- Department of General Surgery, The Sixth People’s Hospital of Huizhou, Huizhou, China
| | - Ruiliang Yang
- Department of General Surgery, The Sixth People’s Hospital of Huizhou, Huizhou, China
| | - Meixiang Li
- Department of General Surgery, The Sixth People’s Hospital of Huizhou, Huizhou, China
| | - Dan Li
- Department of Internal Medicine, The Sixth People’s Hospital of Huizhou, Huizhou, China
| | - Yuanqing Xu
- Department of General Surgery, The Sixth People’s Hospital of Huizhou, Huizhou, China
| |
Collapse
|
5
|
Xu J, Zhou H, Liu Z, Huang Y, Zhang Z, Zou H, Wang Y. PDT-regulated immune gene prognostic model reveals tumor microenvironment in colorectal cancer liver metastases. Sci Rep 2025; 15:13129. [PMID: 40240471 PMCID: PMC12003684 DOI: 10.1038/s41598-025-97667-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 04/07/2025] [Indexed: 04/18/2025] Open
Abstract
Liver metastasis is the most common site of metastasis in colorectal cancer, and the prognosis of colorectal cancer patients with liver metastasis is extremely poor. Revealing the key genes of CLM and implementing targeted interventions is of great significance for colorectal cancer patients. By using the weighted gene co-expression network analysis (WGCNA) algorithm, key gene modules related to metastasis in colorectal cancer were identified. Subsequently, immune-regulating and prognostic-influencing key gene sets were identified from these modules to construct a prognostic model related to colorectal cancer metastasis. Genetic background differences underlying this model were analyzed using colorectal cancer methylation and mutation data, followed by Gene Ontology (GO) analysis and Gene Set Enrichment Analysis (GSEA) analysis of the relevant biological processes associated with the model. The value of predicting tumor drug response through the model was assessed using drug half maximal inhibitory concentration (IC50) data from colorectal cancer cell lines. Subsequently, utilizing single-cell sequencing data about liver metastasis, the colorectal cancer immune microenvironment reflected in the predictive model was analyzed, and a key gene set of the model was identified. Lastly, experimental validation was conducted to investigate the regulatory effects of photodynamic therapy (PDT) on the key genes of the model, and the cytotoxic effect of PDT on colorectal cancer was confirmed. An immune-related gene prognostic model regulating CLM was constructed, consisting of HSPA1A, ULBP2, RBP7, OXT, SLC11A1, INHBB, and ICOS. This model can predict the clinical response of colorectal cancer patients to Oxaliplatin, Cisplatin, Irinotecan, and 5-Fluorouracil. Single-cell sequencing results demonstrate that the model is associated with an immunosuppressive microenvironment in CLM. The higher the model's riskscore, the weaker the MHC-I, MHC-II, and various tumor immune signaling pathway networks in the colorectal cancer microenvironment. Causal analysis reveals that SLC11A1, ICOS, and HSPA1A play key roles in this model. PDT can kill colorectal cancer cells, inhibit colorectal cancer cell metastasis, significantly influence the expression of genes such as SLC11A1, ICOS, and HSPA1A in these processes, and suppress the infiltration of macrophages in the colorectal microenvironment, inhibiting the immune escape process of PD-1/PD-L1. A prognostic model based on immunity regulated by PDT has been established for assessing the prognosis of CLM patients, as well as clinical responses to chemotherapy drugs and immunotherapy.
Collapse
Affiliation(s)
- Jiachi Xu
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Hui Zhou
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Zhongtao Liu
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Yunpeng Huang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Zijian Zhang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Heng Zou
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
| | - Yongxiang Wang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
| |
Collapse
|
6
|
Li Z, Miao H, Bao W, Zhang L. Development and validation of a nomogram model of lung metastasis in breast cancer based on machine learning algorithm and cytokines. BMC Cancer 2025; 25:692. [PMID: 40229760 PMCID: PMC11998148 DOI: 10.1186/s12885-025-14101-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND The relationship between cytokines and lung metastasis (LM) in breast cancer (BC) remains unclear and current clinical methods for identifying breast cancer lung metastasis (BCLM) lack precision, thus underscoring the need for an accurate risk prediction model. This study aimed to apply machine learning algorithms for identifying the key risk factors for BCLM before developing a reliable prediction model centered on cytokines. METHODS This population-based retrospective study included 326 BC patients admitted to the Second Affiliated Hospital of Xuzhou Medical University between September 2018 and September 2023. After randomly assigning the patients to a training cohort (70%; n = 228) or a validation cohort (30%; n = 98) the risk factors for BCLM were identified using Least Absolute Shrinkage and Selection Operator (LASSO), Extreme Gradient Boosting (XGBoost) and Random Forest (RF) models. Significant risk factors were visualized with a Venn diagram and incorporated into a nomogram model, the performance of which was then evaluated according to three criteria, namely discrimination, calibration and clinical utility using calibration plots, receiver operating characteristic (ROC) curves and decision curve analysis (DCA). RESULTS Among the cohort, 70 patients developed LM. A nomogram was then developed to predict the 5-year and 10-year BCLM risk by incorporating five key variables, namely endocrine therapy, hsCRP, IL6, IFN-ɑ and TNF-ɑ. For the 5-year prediction model, the training and validation cohorts had AUC values of 0.786 (95% CI: 0.691-0.881) and 0.627 (95% CI: 0.441-0.813), respectively, while for the 10-year prediction model, the corresponding AUC values were 0.687 (95% CI: 0.528-0.847) and 0.797 (95% CI: 0.605-0.988), respectively. ROC analysis further confirmed the model's strong discriminative ability, while calibration plots indicated that the predicted and observed outcomes were in good agreement in both cohorts. Finally, DCA demonstrated the model's effectiveness in clinical practice. CONCLUSION Using machine learning algorithms, this study developed aa nomogram that could effectively identify BC patients who were at a higher risk of developing LM, thus providing a valuable tool for decision-making in clinical settings.
Collapse
Affiliation(s)
- Zhaoyi Li
- Department of Radiotherapy, The Second Affiliated Hospital of Xuzhou Medical University, Meijian Road 32, Xuzhou, 221000, China
| | - Hao Miao
- Xuzhou Medical University, Tongshan Road 209, Xuzhou, 221000, China
| | - Wei Bao
- Tongji University, Yangpu District, Siping Road 1239, Shanghai, 310000, China
| | - Lansheng Zhang
- Department of Radiotherapy, The Second Affiliated Hospital of Xuzhou Medical University, Meijian Road 32, Xuzhou, 221000, China.
| |
Collapse
|
7
|
Lin Z, Lv J, Dai C, Zhai Y, Jiang J, Gao Y, Li R, Fan J, Yu Y, Wu L, Yang Y. Identification of the disulfidptosis-related key gene CD2AP as a potential biomarker and new therapeutic target for LUAD patients by comprehensive multi-omics analysis. Discov Oncol 2025; 16:515. [PMID: 40214854 PMCID: PMC11992263 DOI: 10.1007/s12672-025-02308-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/03/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Lung Adenocarcinoma (LUAD) is a major subtype of Non-Small Cell Lung Cancer (NSCLC) with poor prognosis. Despite advances in molecular targeted therapy and immunotherapy, the five-year survival rate remains low. Disulfidptosis, a novel cell death mechanism, may play a role in tumor progression. CD2AP (CD2-associated protein), a key gene related to Disulfidptosis, is involved in cytoskeleton reorganization and signaling. This study aimed to explore CD2AP's function in LUAD and its potential as a biomarker and therapeutic target through multi-omics analysis. METHODS We analyzed CD2AP expression and clinical significance in LUAD using data from TCGA, GEO, and other public databases. We employed transcriptomics, methylation analysis, immune infiltration assays, and spatial transcriptomics. Kaplan-Meier survival analysis was used to assess the relationship between CD2AP expression and prognosis. Enrichment analysis identified biological processes and pathways related to CD2AP, while its association with the immune microenvironment and drug sensitivity was also evaluated. RESULTS CD2AP was significantly overexpressed in LUAD, and high expression correlated with poorer prognosis, including overall survival and progression-free survival. Enrichment analysis showed CD2AP is involved in cell adhesion, PI3K-Akt signaling, and immune escape, suggesting it promotes LUAD progression through these pathways. High CD2AP expression was associated with alterations in the tumor immune microenvironment and drug sensitivity, particularly to chemotherapeutics like Cisplatin, Etoposide, and Paclitaxel, and resistance to targeted therapies like Gefitinib. Spatial transcriptomics revealed higher CD2AP expression in tumor regions, especially in malignant cell-enriched areas. CONCLUSION This study highlights CD2AP's critical role in LUAD, particularly in immune microenvironment modulation, metabolic reprogramming, and drug response. CD2AP's high expression is linked to poor prognosis and may serve as a potential target for immunotherapy and drug response prediction.
Collapse
Affiliation(s)
- Zhiwu Lin
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Jing Lv
- Department of Orthopedics, Ziyang Central Hospital, Ziyang, 641300, China
| | - Chuanqiang Dai
- Department of Orthopedics, Ziyang Central Hospital, Ziyang, 641300, China
| | - Yuanwei Zhai
- Department of Medical Imaging, Ziyang Central Hospital, Ziyang, 641300, China
| | - Jiudong Jiang
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Yang Gao
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Rulin Li
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Jiangang Fan
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Yang Yu
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Liang Wu
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China
| | - Yanchun Yang
- Department of Thoracic Surgery, Ziyang Central Hospital, Ziyang, 641300, China.
| |
Collapse
|
8
|
Xiang J, Wang J, Xiao H, Huang C, Wu C, Zhang L, Qian C, Xiang D. Targeting tumor-associated macrophages in colon cancer: mechanisms and therapeutic strategies. Front Immunol 2025; 16:1573917. [PMID: 40191202 PMCID: PMC11968422 DOI: 10.3389/fimmu.2025.1573917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/10/2025] [Indexed: 04/09/2025] Open
Abstract
Colon cancer (CC) remains a primary contributor to cancer-related fatalities worldwide, driven by difficulties in early diagnosis and constrained therapeutic options. Recent studies underscore the importance of the tumor microenvironment (TME), notably tumor-associated macrophages (TAMs), in fostering malignancy progression and therapy resistance. Through their inherent plasticity, TAMs facilitate immunosuppression, angiogenic processes, metastatic spread, and drug tolerance. In contrast to M1 macrophages, which promote inflammatory and tumoricidal responses, M2 macrophages support tumor expansion and dissemination by exerting immunosuppressive and pro-angiogenic influences. Consequently, manipulating TAMs has emerged as a potential avenue to enhance treatment effectiveness. This review outlines the origins, polarization states, and functions of TAMs in CC, highlights their role in driving tumor advancement, and surveys ongoing efforts to target these cells for better patient outcomes. Emerging therapeutic strategies aimed at modulating TAM functions - including depletion strategies, reprogramming approaches that shift M2-polarized TAMs toward an M1 phenotype, and inhibition of key signaling pathways sustaining TAM-mediated immunosuppression-are currently under active investigation. These approaches hold promise in overcoming TAM - induced resistance and improving immunotherapeutic efficacy in CC.
Collapse
Affiliation(s)
- Jianqin Xiang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing, China
| | - Jian Wang
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing, China
| | - Huihui Xiao
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing, China
| | - Chengchen Huang
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing, China
| | - Chunrong Wu
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing, China
| | - Lin Zhang
- Department of Gastroenterology, Chongqing University Jiangjin Hospital, Chongqing, China
| | - Chenyuan Qian
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing, China
| | - Debing Xiang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing, China
| |
Collapse
|
9
|
He J, Liu N, Zhao L. New progress in imaging diagnosis and immunotherapy of breast cancer. Front Immunol 2025; 16:1560257. [PMID: 40165974 PMCID: PMC11955504 DOI: 10.3389/fimmu.2025.1560257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Breast cancer (BC) is a predominant malignancy among women globally, with its etiology remaining largely elusive. Diagnosis primarily relies on invasive histopathological methods, which are often limited by sample representation and processing time. Consequently, non-invasive imaging techniques such as mammography, ultrasound, and Magnetic Resonance Imaging (MRI) are indispensable for BC screening, diagnosis, staging, and treatment monitoring. Recent advancements in imaging technologies and artificial intelligence-driven radiomics have enhanced precision medicine by enabling early detection, accurate molecular subtyping, and personalized therapeutic strategies. Despite reductions in mortality through traditional treatments, challenges like tumor heterogeneity and therapeutic resistance persist. Immunotherapies, particularly PD-1/PD-L1 inhibitors, have emerged as promising alternatives. This review explores recent developments in BC imaging diagnostics and immunotherapeutic approaches, aiming to inform clinical practices and optimize therapeutic outcomes.
Collapse
Affiliation(s)
- Jie He
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Nan Liu
- Department of Translational Medicine and Clinical Research, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Li Zhao
- Department of Radiology, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| |
Collapse
|
10
|
Liu Z, Huang S, Luo R, Shi X, Xiu M, Wang Y, Wang R, Zhang W, Lv M, Tang X. EXO1's pan-cancer roles: diagnostic, prognostic, and immunological analyses through bioinformatics. Discov Oncol 2025; 16:310. [PMID: 40074873 PMCID: PMC11903978 DOI: 10.1007/s12672-025-02045-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a leading cause of mortality worldwide, with human exonuclease 1 (EXO1) emerging as a key player in DNA repair and damage response pathways, critical for genomic stability and tumor evolution. The aim of this study was to conduct a comprehensive pan-cancer analysis to elucidate the multifaceted roles of EXO1 in various malignancies. Leveraging public databases including TCGA, GTEx, HPA, cBioPortal, UALCAN, STRING, CancerSEA and TISIDB database, we examined EXO1's expression, diagnostic potential, prognostic significance, mutational characteristics, functional roles, and immunological effects across different cancer types. EXO1 was found to be upregulated in multiple cancers, with significant diagnostic potential as indicated by high AUC values in ROC analyses. Elevated EXO1 expression correlated with adverse prognosis in several cancer types, including breast, lung, and pancreatic cancers. Epigenetic alterations, including DNA methylation and mRNA modifications, were also associated with EXO1 expression. Enrichment analyses identified EXO1-related genes involved in DNA recombination, replication, and repair, with GSEA implicating EXO1 in cell cycle regulation and DNA processing pathways. Importantly, immunogenomic analyses revealed EXO1's significant role in modulating the tumor microenvironment, as it is associated with immune cell infiltration and cytokine expression, suggesting its involvement in tumor immunology and immune response regulation. These results implied that EXO1 as a significant biomarker with prognostic and diagnostic potential across various malignancies, suggesting its potential as a therapeutic target and its involvement in immunomodulatory processes within the tumor microenvironment.
Collapse
Affiliation(s)
- Zheng Liu
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Taiping No.25, Jiangyang, Luzhou, Sichuan, China
| | - Shu Huang
- Department of Gastroenterology, Lianshui County People' Hospital, Huaian, China
- Department of Gastroenterology, Lianshui People' Hospital of Kangda College Affiliated to Nanjing Medical University, Huaian, China
| | - Rui Luo
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Taiping No.25, Jiangyang, Luzhou, Sichuan, China
| | - Xiaomin Shi
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Taiping No.25, Jiangyang, Luzhou, Sichuan, China
| | - Mingzhu Xiu
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Taiping No.25, Jiangyang, Luzhou, Sichuan, China
| | - Yizhou Wang
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Taiping No.25, Jiangyang, Luzhou, Sichuan, China
| | - Ruiyu Wang
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Taiping No.25, Jiangyang, Luzhou, Sichuan, China
| | - Wei Zhang
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Taiping No.25, Jiangyang, Luzhou, Sichuan, China
| | - Muhan Lv
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Taiping No.25, Jiangyang, Luzhou, Sichuan, China.
| | - Xiaowei Tang
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Taiping No.25, Jiangyang, Luzhou, Sichuan, China.
| |
Collapse
|
11
|
Hu W, Zhao X, Luo N, Xiao M, Feng F, An Y, Chen J, Rong L, Yang Y, Peng J. Circulating cell-free DNA methylation analysis of pancreatic cancer patients for early noninvasive diagnosis. Front Oncol 2025; 15:1552426. [PMID: 40129923 PMCID: PMC11930829 DOI: 10.3389/fonc.2025.1552426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 02/13/2025] [Indexed: 03/26/2025] Open
Abstract
Background Aberrant hypermethylation of genomic DNA CpG islands (CGIs) is frequently observed in human pancreatic cancer (PAC). A plasma cell-free DNA (cfDNA) methylation analysis method can be utilized for the early and noninvasive detection of PAC. This study also aimed to differentiate PAC from other cancer types. Methods We employed the methylated CpG tandem amplification and sequencing (MCTA-Seq) method, which targets approximately one-third of CGIs, on plasma samples from PAC patients (n = 50) and healthy controls (n = 52), as well as from cancerous and adjacent noncancerous tissue samples (n = 66). The method's efficacy in detecting PAC and distinguishing it from hepatocellular carcinoma (HCC), colorectal cancer (CRC), and gastric cancer (GC) was evaluated. Additionally, a methylation score and typing system for PAC was also established. Results We identified a total of 120 cfDNA methylation biomarkers, including IRX4, KCNS2, and RIMS4, for the detection of PAC in blood. A panel comprising these biomarkers achieved a sensitivity of 97% and 86% for patients in the discovery and validation cohorts, respectively, with a specificity of 100% in both cohorts. The methylation scoring and typing systems were clinically applicable. Furthermore, we identified hundreds of differentially methylated cfDNA biomarkers between PAC and HCC, CRC, and GC. Certain combinations of these markers can be used in a highly specific (approximately 100%) algorithm to differentiate PAC from HCC, CRC, and GC in blood. Conclusions Our study identified cfDNA methylation markers for PAC, offering a novel approach for the early, noninvasive diagnosis of PAC.
Collapse
Affiliation(s)
- Wenzhe Hu
- Department of Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- School of Oncology, Capital Medical University, Beijing, China
| | - Xudong Zhao
- Department of Endoscopy Center, Peking University First Hospital, Peking University, Beijing, China
| | - Nan Luo
- Department of Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- School of Oncology, Capital Medical University, Beijing, China
- Ninth School of Clinical Medicine, Peking University, Beijing, China
| | - Mengmeng Xiao
- Department of General Surgery, Peking University People’s Hospital, Peking University, Beijing, China
| | - Feng Feng
- Department of Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- School of Oncology, Capital Medical University, Beijing, China
| | - Yuan An
- Department of Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- School of Oncology, Capital Medical University, Beijing, China
| | - Jianfei Chen
- Department of Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- School of Oncology, Capital Medical University, Beijing, China
| | - Long Rong
- Department of Endoscopy Center, Peking University First Hospital, Peking University, Beijing, China
| | - Yinmo Yang
- Department of General Surgery, Peking University First Hospital, Peking University, Beijing, China
| | - Jirun Peng
- Department of Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- School of Oncology, Capital Medical University, Beijing, China
- Ninth School of Clinical Medicine, Peking University, Beijing, China
| |
Collapse
|
12
|
FANG ZIYI, SHAO YONGFU, HU MENG, YAN JIANING, YE GUOLIANG. Biological roles and molecular mechanism of circular RNAs in epithelial-mesenchymal transition of gastrointestinal malignancies. Oncol Res 2025; 33:549-566. [PMID: 40109856 PMCID: PMC11915071 DOI: 10.32604/or.2024.051589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 06/13/2024] [Indexed: 03/22/2025] Open
Abstract
Circular RNAs (circRNAs) are formed by splicing of precursor RNAs and covalently linked at the 5' and 3' ends. Dysregulated circRNAs are closely related to the epithelial-mesenchymal transition (EMT) of gastrointestinal malignancies. CircRNAs, including circRNA_0008717, circGOT1, circ-DOCK5, circVPS33B, circPVT1, circMET, circ-OXCT1, circ_67835, circRTN4, circ_0087502, circFNDC38, circ_PTEN1, circPGPEP1, and circ-E-Cad are involved in the EMT process of gastrointestinal malignancies through a variety of mechanisms, such as regulating EMT-inducing transcription factors, signaling pathways, and tumor microenvironments. Gastrointestinal (GI) malignancies are common malignant tumors worldwide, and the heterogeneity and easy metastasis of gastrointestinal malignancies limit the effectiveness of medical treatments. Therefore, investigating the molecular mechanisms involved in the pathogenesis of gastrointestinal malignancies is essential for clinical treatment. This article summarizes the biological roles and molecular mechanism of circRNAs in EMT of gastrointestinal malignancies, providing a theoretical basis for applying EMT-related circRNAs in targeted therapy.
Collapse
Affiliation(s)
- ZIYI FANG
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, 315020, China
| | - YONGFU SHAO
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, 315020, China
| | - MENG HU
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - JIANING YAN
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, 315020, China
| | - GUOLIANG YE
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, 315020, China
| |
Collapse
|
13
|
Zhao F, Chen M, Wu T, Ji M, Li F. Integration of single-cell and bulk RNA sequencing to identify a distinct tumor stem cells and construct a novel prognostic signature for evaluating prognosis and immunotherapy in LUAD. J Transl Med 2025; 23:222. [PMID: 39987127 PMCID: PMC11847374 DOI: 10.1186/s12967-025-06243-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/11/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are crucial for lung adenocarcinoma (LUAD). This study investigates tumor stem cell gene signatures in LUAD using single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing (RNA-seq), aiming to develop a prognostic tumor stem cell marker signature (TSCMS) model. METHODS LUAD scRNA-seq and RNA-seq data were analyzed. CytoTRACE software quantified the stemness score of tumor-derived epithelial cell clusters. Gene Set Variation Analysis (GSVA) identified potential biological functions in different clusters. The TSCMS model was constructed using Lasso-Cox regression, and its prognostic value was assessed through Kaplan-Meier, Cox regression, and receiver-operating characteristic (ROC) curve analyses. Immune infiltration was evaluated using the Cibersortx algorithm, and drug response prediction was performed using the pRRophetic package. TAF10 functional investigations in LUAD cells involved bioinformatics analysis, qRT-PCR, Western blot, immunohistochemistry, and assays for cell proliferation. RESULTS Seven distinct cell clusters were identified by CytoTRACE, with epithelial cell cluster 1 (Epi_C1) showing the highest stemness potential. The TSCMS model included 49 tumor stemness-related genes; high-risk patients exhibited lower immune and ESTIMATE scores and increased tumor purity. Significant differences in immune landscapes and chemotherapy sensitivity were observed between risk groups. TAF10 positively correlated with RNA expression-based stemness scores in various tumors, including LUAD. It was over-expressed in LUAD cell lines and clinical tumor tissues, with high expression linked to poor prognosis. Silencing TAF10 inhibited LUAD cell proliferation and tumor sphere formation. CONCLUSIONS This study demonstrates the TSCMS model's prognostic value in LUAD, reveals insights into immune infiltration and therapeutic response, and identifies TAF10 as a potential therapeutic target.
Collapse
Affiliation(s)
- Fengyun Zhao
- Cancer Research Institute of Zhongshan City, Zhongshan City People's Hospital, Zhongshan, 528403, Guangdong, China.
| | - Mengting Chen
- South China Normal University, Guangzhou, 510630, Guangdong, China
| | - Tianjiao Wu
- Guangdong Medical University, Zhanjiang, 523000, Guangdong, China
| | - Mingfang Ji
- Cancer Research Institute of Zhongshan City, Zhongshan City People's Hospital, Zhongshan, 528403, Guangdong, China
| | - Fugui Li
- Cancer Research Institute of Zhongshan City, Zhongshan City People's Hospital, Zhongshan, 528403, Guangdong, China.
| |
Collapse
|
14
|
Li Z, Lin X, Yang Y, Tian M, Zhang L, Huang F, Wen X, Wei Z, Tian Y. EXO1 is a key gene for lung-resident memory T cells and has diagnostic and predictive values for lung adenocarcinoma. Sci Rep 2025; 15:4002. [PMID: 39893221 PMCID: PMC11787328 DOI: 10.1038/s41598-025-88126-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/24/2025] [Indexed: 02/04/2025] Open
Abstract
Lung adenocarcinoma (LUAD) is a very common and lethal kind of lung malignancy. An increasing number of studies indicated that tissue-resident memory T (TRM) cells played significant roles in anti-cancer immunity. In our previous study, EXO1 was found to be a core gene for TRM cells in the prognosis of LUAD. However, the roles of EXO1 in the tumor microenvironment, and its application in the diagnosis and prognosis prediction of LUAD are still inadequately explored. In this study, the RNA expression, DNA methylation, CNV, somatic mutation data of EXO1, and the corresponding patients' clinical information from publicly available databases were analyzed using bioinformatic methods. The results were validated through immunohistochemical staining of EXO1 in LUAD samples. The results showed EXO1 was aberrantly highly expressed in LUAD tissues. High expression of EXO1 was a risky factor for LUAD patients. The expression level of EXO1 was associated with many clinical features such as TNM stages. It can also distinguish normal tissues and LUAD tumor tissues accurately. EXO1 expression was correlated with the infiltration of immune cells, and high expression of EXO1 was an adverse effect on LUAD patients receiving anti-PD-1/PD-L1 immunotherapy. Moreover, patients with EXO1 mutation had worse DSS, DFI and PFI.
Collapse
Affiliation(s)
- Zhuoqi Li
- Department of Radiotherapy Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250011, Jinan, P.R. China
| | - Xiaoyan Lin
- Department of Pathology, Shandong Provincial Hospital, Shandong University, 250021, Jinan, P.R. China
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021, Jinan, P.R. China
| | - Yuanhui Yang
- Department of Pathology, Shandong Provincial Hospital, Shandong University, 250021, Jinan, P.R. China
| | - Mei Tian
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250014, Jinan, P.R. China
| | - Lu Zhang
- Department of Radiotherapy Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250011, Jinan, P.R. China
| | - Fujing Huang
- Department of Radiotherapy Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250011, Jinan, P.R. China
| | - Xiao Wen
- Department of Radiotherapy Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250011, Jinan, P.R. China
| | - Zhigang Wei
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, 250014, Jinan, P.R. China.
| | - Yuan Tian
- Department of Radiotherapy Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250011, Jinan, P.R. China.
| |
Collapse
|
15
|
Shu Y, Li J. Disulfidptosis as a key regulator of glioblastoma progression and immune cell impairment. Front Immunol 2025; 16:1526296. [PMID: 39949776 PMCID: PMC11821639 DOI: 10.3389/fimmu.2025.1526296] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 01/13/2025] [Indexed: 02/16/2025] Open
Abstract
Background Glioblastoma, associated with poor prognosis and impaired immune function, shows potential interactions between newly identified disulfidptosis mechanisms and T cell exhaustion, yet these remain understudied. Methods Key genes were identified using Lasso regression, followed by multivariate analysis to develop a prognostic model. Single-cell pseudotemporal analysis explored disulfidptosis T-cell exhaustion (Tex) signaling in cell differentiation. Immune infiltration was assessed via ssGSEA, while transwell assays and immunofluorescence examined the effects of disulfidptosis-Tex genes on glioma cell behavior and immune response. Results Eleven disulfidptosis-Tex genes were found critical for glioblastoma survival outcomes. This gene set underpinned a model predicting patient prognosis. Single-cell analysis showed high disulfidptosis-Tex activity in endothelial cells. Memory T cell populations were linked to these genes. SMC4 inhibition reduced LN299 cell migration and increased chemotherapy sensitivity, decreasing CD4 and CD8 T cell activation. Conclusions Disulfidptosis-Tex genes are pivotal in glioblastoma progression and immune interactions, offering new avenues for improving anti-glioblastoma therapies through modulation of T cell exhaustion.
Collapse
|
16
|
Xie D, Liu Y, Xu FB, Zhang JS. The role of SASP in ischemic stroke: a deep dive into cellular mechanisms. Front Neurol 2025; 15:1513357. [PMID: 39931101 PMCID: PMC11809037 DOI: 10.3389/fneur.2024.1513357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/17/2024] [Indexed: 02/13/2025] Open
Abstract
Background The escalating incidence of ischemic stroke (IS) exerts a heavy toll on global health. Aging, a prominent risk factor, implicates the senescence-associated secretory phenotype (SASP) in IS pathogenesis. We postulated that alterations in SASP-related factor expression during IS correlate with remodeling of intercellular interaction networks and disease advancement. The present study endeavored to preliminarily dissect the SASP-IS nexus via combined bulk and single-cell transcriptome analysis. Methods Aggregated expression profiles from human peripheral blood bulk chips and MCAO mouse single-cell sequencing data, followed by SASP gene analysis. Executed protein interaction network and enrichment assays. Investigated immune infiltration in stroke patients, managed quality control and annotation of single-cell data, cherry-picked central cells based on SASP scores, unearthed essential genes via enrichment analysis, conducted pseudo-time and intercellular communication studies, and prognosticated drugs for hub genes. Finally, authenticated core gene expression in serum of MCAO and Sham rats using real-time fluorescent polymerase chain reaction (RT-qPCR). Results Fourteen hub genes were discerned. Seven cell types were annotated in MCAO mouse peripheral blood single-cell data. Basophils exhibited the highest SASP scores, with Lcp1 upregulated and Ccl3 downregulated in basophils of the MCAO group. Enrichment analysis divulged a significant association of Ccl3 with the cell apoptosis pathway and Lcp1 with immune responses. The Ccl3 gene is pivotal in basophils and basophil-neutrophil crosstalk. Additionally, we forecasted nagrestipen's regulatory function on Ccl3. RT-qPCR demonstrated a marked elevation in Lcp1 mRNA and a pronounced reduction in Ccl3 in the MCAO group relative to the Sham group. Conclusion The Ccl3 gene in basophils and its immune cell interaction is a linchpin in the IS immune microenvironment. Ccl3 and Lcp1 might potentially modulate IS progression by influencing SASP, proffering novel prospects for IS clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Dong Xie
- The Third Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yang Liu
- The Third Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Fang-Biao Xu
- The Third Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jin-Sheng Zhang
- Department of Encephalopathy, The Third Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
17
|
Li Y, Zeng M, Qin Y, Feng F, Wei H. The role of KRT18 in lung adenocarcinoma development: integrative bioinformatics and experimental validation. Discov Oncol 2024; 15:841. [PMID: 39729139 DOI: 10.1007/s12672-024-01728-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
Lung adenocarcinoma (LUAD) represents one of the most common subtypes of lung cancer with high rates of incidence and mortality, which contributes to substantial health and economic demand across the globe. Treatment today mainly consists of surgery, radiotherapy, and chemotherapy, but their efficacy in advanced stages is often suboptimal and emphasizes the clear need for new biomarkers and therapeutic targets. Using comprehensive bioinformatics analyses consisting of the Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), Human Protein Atlas (HPA) and Clinical Proteomic Tumor Analysis Consortium (CPTAC), immune infiltration analysis and functional enrichment analysis, and single-cell analysis, we examined the potential of keratin 18 (KRT18) as a candidate biomarker in advanced LUAD. KRT18 was significantly elevated in LUAD tissue relative to normal adjacent tissue (p < 0.05), and its expression was correlated with poor clinical-pathological features and inferior prognostic outcome. Furthermore, KRT18 expression was associated with several populations of immune cells, suggesting KRT18 may contribute to the local tumor microenvironment and potentially pathways of immune evasion. Survival analysis indicated that elevated KRT18 expression correlated with poor overall survival (OS), disease-specific survival (DSS), and progression-free interval (PFI), reinforcing its legitimacy as a prognostic tool (AUC = 0.846). Importantly, gene enrichment analysis found KRT18-associated genes enriched for pathways associated with lymphocyte differentiation and immune response pathways, which provides mechanistic insight into biological effects attributed to KRT18. Notably, NU.1025 has demonstrated the capability of reversing KRT18-modulated oncogenic features, and targeted therapeutic strategies can be developed moving forward. In conclusion, our data demonstrate that KRT18 has utility as a potential biomarker but may also serve as a therapeutic target in LUAD and merit further investigation into underlying mechanistic functions and potential therapeutic roles in the clinic.
Collapse
Affiliation(s)
- Yongjie Li
- School of Pharmacy, Shaoyang University, Shaoyang, 422000, Hunan, China
- Southwest Hunan Research Center of Engineering for Development and Utilization of Traditional Chinese Medicine, Shaoyang, 422000, Hunan, China
| | - Min Zeng
- Department of Respiratory and Critical Care Medicine, The Affiliated Shaoyang Hospital, Hengyang Medical School, University of South China, Shaoyang, 422000, Hunan, China.
| | - Yinan Qin
- Department of Pharmacy, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, Zhejiang, China
| | - Fen Feng
- School of Pharmacy, Shaoyang University, Shaoyang, 422000, Hunan, China
| | - Hailiang Wei
- School of Pharmacy, Shaoyang University, Shaoyang, 422000, Hunan, China
| |
Collapse
|
18
|
Liu M, Li Q, Meng X, Cui Y, Sun W, Wang H, Gao Q. Identification of gene signatures relevant to the efficacy of immune checkpoint inhibitors in non-small cell lung cancer. Medicine (Baltimore) 2024; 103:e40569. [PMID: 39654181 PMCID: PMC11630944 DOI: 10.1097/md.0000000000040569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/27/2024] [Accepted: 10/30/2024] [Indexed: 12/12/2024] Open
Abstract
Despite significant advancements in the treatment of non-small cell lung cancer (NSCLC) through immunotherapy, many patients still exhibit resistance to this approach. This study aims to identify the characteristics of individuals who can benefit from immunotherapy, especially immune checkpoint inhibitors (ICIs), and to investigate optimal strategies for patients who experience resistance to it. Data on gene expression patterns and clinical information from NSCLC patients who underwent immunotherapy were obtained from the Gene Expression Omnibus databases. A predictive signature for immunotherapy prognosis was developed using a training dataset and validated with validation datasets. Immune landscape and immunotherapy responsiveness analyses were conducted to assess the risk signature. Additionally, data from a study on immunotherapy were used to evaluate the correlation between MNX1 mutation and the effectiveness of ICIs, including clinical data and whole exome sequencing data. We identified 7 genes in NSCLC using RNA-seq data that were significantly associated with the efficacy of immunotherapy. Based on these genes, a risk signature was created to predict the efficacy of ICIs. Patients in the low-risk group had better outcomes compared to those in the high-risk group after receiving ICIs. Additionally, our analysis of the immune landscape revealed a significant association between the high-risk signature and an immunosuppressive state. We also discovered an unexpected role of tumor-specific MNX1 and HOXD1 in suppressing the immune response against cancer. Notably, NSCLC patients with MNX1 mutations experienced prolonged progression-free survival. Furthermore, we identified several medications that exhibited increased sensitivity in patients with high MNX1 expression, with topoisomerase inhibitors showing the highest level of sensitivity. This could be a potential strategy to improve the efficacy of ICIs. The risk signature has demonstrated its effectiveness in forecasting the prognosis of NSCLC treated with ICIs, enabling better patient stratification and more accurate prediction of immunotherapy response. Moreover, MNX1 and HOXD1 have been identified as key molecules related to immunotherapy resistance. Inhibition of these molecules, combined with current ICIs, offers novel strategies for the management of NSCLC patients.
Collapse
Affiliation(s)
- Min Liu
- Department of General Medicine, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Qiao Li
- The Eighth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaohong Meng
- The Eighth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yanan Cui
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Weirong Sun
- Radiological Department, The Eighth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hongmei Wang
- Department of General Medicine, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Qingjun Gao
- Department of Thoracic Surgery, The People’s Hospital of Rushan City, Weihai, PR China
| |
Collapse
|
19
|
Zhang B, Zhang B, Wang T, Huang B, Cen L, Wang Z. Integrated bulk and single-cell profiling characterize sphingolipid metabolism in pancreatic cancer. BMC Cancer 2024; 24:1347. [PMID: 39487387 PMCID: PMC11531184 DOI: 10.1186/s12885-024-13114-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Abnormal sphingolipid metabolism (SM) is closely linked to the incidence of cancers. However, the role of SM in pancreatic cancer (PC) remains unclear. This study aims to explore the significance of SM in the prognosis, immune microenvironment, and treatment of PC. METHODS Single-cell and bulk transcriptome data of PC were acquired via TCGA and GEO databases. SM-related genes (SMRGs) were obtained via MSigDB database. Consensus clustering was utilized to construct SM-related molecular subtypes. LASSO and Cox regression were utilized to build SM-related prognostic signature. ESTIMATE and CIBERSORT algorithms were employed to assess the tumour immune microenvironment. OncoPredict package was used to predict drug sensitivity. CCK-8, scratch, and transwell experiments were performed to analyze the function of ANKRD22 in PC cell line PANC-1 and BxPC-3. RESULTS A total of 153 SMRGs were acquired, of which 48 were linked to PC patients' prognosis. Two SM-related subtypes (SMRGcluster A and B) were identified in PC. SMRGcluster A had a poorer outcome and more active SM process compared to SMRGcluster B. Immune analysis revealed that SMRGcluster B had higher immune and stromal scores and CD8 + T cell abundance, while SMRGcluster A had a higher tumour purity score and M0 macrophages and activated dendritic cell abundance. PC with SMRGcluster B was more susceptible to gemcitabine, paclitaxel, and oxaliplatin. Then SM-related prognostic model (including ANLN, ANKRD22, and DKK1) was built, which had a very good predictive performance. Single-cell analysis revealed that in PC microenvironment, macrophages, epithelial cells, and endothelial cells had relatively higher SM activity. ANKRD22, DKK1, and ANLN have relatively higher expression levels in epithelial cells. Cell subpopulations with high expression of ANKRD22, DKK1, and ANLN had more active SM activity. In vitro experiments showed that ANKRD22 knockdown can inhibit the proliferation, migration, and invasion of PC cells. CONCLUSION This study revealed the important significance of SM in PC and identified SM-associated molecular subtypes and prognostic model, which provided novel perspectives on the stratification, prognostic prediction, and precision treatment of PC patients.
Collapse
Affiliation(s)
- Biao Zhang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Bolin Zhang
- Department of Visceral, Vascular and Endocrine Surgery, Martin-Luther-University Halle- Wittenberg, University Medical Center Halle, Halle, Germany
| | - Tingxin Wang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Bingqian Huang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Lijun Cen
- Department of Transfusion Medicine, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China.
- Key Laboratory of Molecular Pathology in Tumors of Guangxi, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China.
| | - Zhizhou Wang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
20
|
Si Y, Zhao Z, Meng X, Zhao K. RNA-seq and bulk RNA-seq data analysis of cancer-related fibroblasts (CAF) in LUAD to construct a CAF-based risk signature. Sci Rep 2024; 14:23243. [PMID: 39369095 PMCID: PMC11455853 DOI: 10.1038/s41598-024-74336-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 09/25/2024] [Indexed: 10/07/2024] Open
Abstract
Angiogenesis, metastasis, and resistance to therapy are all facilitated by cancer-associated fibroblasts (CAFs). A CAF-based risk signature can be used to predict patients' prognoses for Lung adenocarcinoma (LUAD) based on CAF characteristics. The Gene Expression Omnibus (GEO) database was used to gather signal-cell RNA sequencing (scRNA-seq) data for this investigation. The GEO and TCGA databases were used to gather bulk RNA-seq and microarray data for LUAD. The scRNA-seq data were analyzed using the Seurat R program based on the CAF markers. Our goal was to use differential expression analysis to discover differentially expressed genes (DEGs) across normal and tumor samples in the TCGA dataset. Pearson correlation analysis was utilized to discover prognostic genes related with CAF, followed by univariate Cox regression analysis. Using Lasso regression, a risk signature based on CAF-related prognostic genes was created. A nomogram model was created based on the clinical and pathological aspects. 5 CAF clusters were identified in LUAD, 4 of which were associated with prognosis. From 2811 DEGs, 1002 genes were found to be significantly correlated with CAF clusters, which led to the creation of a risk signature with 8 genes. These 8 genes were primarily connected with 41 pathways, such as antigen paocessing and presentation, apoptosis, and cell cycle. Meanwhile, the risk signature was significantly associated with stromal and immune scores, as well as some immune cells. Multivariate analysis revealed that risk signature was an independent prognostic factor for LUAD, and its value in predicting immunotherapeutic outcomes was confirmed. A novel nomogram integrating the stage and CAF-based risk signature was constructed, which exhibited favorable predictability and reliability in the prognosis prediction of LUAD. CAF-based risk signatures can be effective in predicting the prognosis of LUAD, and they may provide new strategies for cancer treatments by interpreting the response of LUAD to immunotherapy.
Collapse
Affiliation(s)
- Youjiao Si
- Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zhonghua Zhao
- Department of Oncology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Kaikai Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
21
|
Yan Y, Shen S, Li J, Su L, Wang B, Zhang J, Lu J, Luo H, Han P, Xu K, Shen X, Huang S. Cross-omics strategies and personalised options for lung cancer immunotherapy. Front Immunol 2024; 15:1471409. [PMID: 39391313 PMCID: PMC11465239 DOI: 10.3389/fimmu.2024.1471409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 08/30/2024] [Indexed: 10/12/2024] Open
Abstract
Lung cancer is one of the most common malignant tumours worldwide and its high mortality rate makes it a leading cause of cancer-related deaths. To address this daunting challenge, we need a comprehensive understanding of the pathogenesis and progression of lung cancer in order to adopt more effective therapeutic strategies. In this regard, integrating multi-omics data of the lung provides a highly promising avenue. Multi-omics approaches such as genomics, transcriptomics, proteomics, and metabolomics have become key tools in the study of lung cancer. The application of these methods not only helps to resolve the immunotherapeutic mechanisms of lung cancer, but also provides a theoretical basis for the development of personalised treatment plans. By integrating multi-omics, we have gained a more comprehensive understanding of the process of lung cancer development and progression, and discovered potential immunotherapy targets. This review summarises the studies on multi-omics and immunology in lung cancer, and explores the application of these studies in early diagnosis, treatment selection and prognostic assessment of lung cancer, with the aim of providing more personalised and effective treatment options for lung cancer patients.
Collapse
Affiliation(s)
- Yalan Yan
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Siyi Shen
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Jiamin Li
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Lanqian Su
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Binbin Wang
- Intensive Care Unit, Xichong People’s Hospital, Nanchong, China
| | - Jinghan Zhang
- Department of Anaesthesiology, Southwest Medical University, Luzhou, China
| | - Jiaan Lu
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Huiyan Luo
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Ping Han
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Ke Xu
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Xiang Shen
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Shangke Huang
- Department of Oncology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|
22
|
Yuan L, Shen Z, Shan Y, Zhu J, Wang Q, Lu Y, Shi H. Unveiling the landscape of pathomics in personalized immunotherapy for lung cancer: a bibliometric analysis. Front Oncol 2024; 14:1432212. [PMID: 39040448 PMCID: PMC11260632 DOI: 10.3389/fonc.2024.1432212] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 06/19/2024] [Indexed: 07/24/2024] Open
Abstract
Background Pathomics has emerged as a promising biomarker that could facilitate personalized immunotherapy in lung cancer. It is essential to elucidate the global research trends and emerging prospects in this domain. Methods The annual distribution, journals, authors, countries, institutions, and keywords of articles published between 2018 and 2023 were visualized and analyzed using CiteSpace and other bibliometric tools. Results A total of 109 relevant articles or reviews were included, demonstrating an overall upward trend; The terms "deep learning", "tumor microenvironment", "biomarkers", "image analysis", "immunotherapy", and "survival prediction", etc. are hot keywords in this field. Conclusion In future research endeavors, advanced methodologies involving artificial intelligence and pathomics will be deployed for the digital analysis of tumor tissues and the tumor microenvironment in lung cancer patients, leveraging histopathological tissue sections. Through the integration of comprehensive multi-omics data, this strategy aims to enhance the depth of assessment, characterization, and understanding of the tumor microenvironment, thereby elucidating a broader spectrum of tumor features. Consequently, the development of a multimodal fusion model will ensue, enabling precise evaluation of personalized immunotherapy efficacy and prognosis for lung cancer patients, potentially establishing a pivotal frontier in this domain of investigation.
Collapse
Affiliation(s)
- Lei Yuan
- Department of Thoracic Surgery, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Zhiming Shen
- Department of Thoracic Surgery, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Yibo Shan
- Department of Thoracic Surgery, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Jianwei Zhu
- Department of Thoracic Surgery, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Qi Wang
- Department of Thoracic Surgery, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Yi Lu
- Department of Thoracic Surgery, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Hongcan Shi
- Department of Thoracic Surgery, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| |
Collapse
|
23
|
Li Q, Fang J, Liu K, Luo P, Wang X. Multi-omic validation of the cuproptosis-sphingolipid metabolism network: modulating the immune landscape in osteosarcoma. Front Immunol 2024; 15:1424806. [PMID: 38983852 PMCID: PMC11231095 DOI: 10.3389/fimmu.2024.1424806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 06/06/2024] [Indexed: 07/11/2024] Open
Abstract
Background The current understanding of the mechanisms by which metal ion metabolism promotes the progression and drug resistance of osteosarcoma remains incomplete. This study aims to elucidate the key roles and mechanisms of genes involved in cuproptosis-related sphingolipid metabolism (cuproptosis-SPGs) in regulating the immune landscape, tumor metastasis, and drug resistance in osteosarcoma cells. Methods This study employed multi-omics approaches to assess the impact of cuproptosis-SPGs on the prognosis of osteosarcoma patients. Lasso regression analysis was utilized to construct a prognostic model, while multivariate regression analysis was applied to identify key core genes and generate risk coefficients for these genes, thereby calculating a risk score for each osteosarcoma patient. Patients were then stratified into high-risk and low-risk groups based on their risk scores. The ESTIMATE and CIBERSORT algorithms were used to analyze the level of immune cell infiltration within these risk groups to construct the immune landscape. Single-cell analysis was conducted to provide a more precise depiction of the expression patterns of cuproptosis-SPGs among immune cell subtypes. Finally, experiments on osteosarcoma cells were performed to validate the role of the cuproptosis-sphingolipid signaling network in regulating cell migration and apoptosis. Results In this study, seven cuproptosis-SPGs were identified and used to construct a prognostic model for osteosarcoma patients. In addition to predicting survival, the model also demonstrated reliability in forecasting the response to chemotherapy drugs. The results showed that a high cuproptosis-sphingolipid metabolism score was closely associated with reduced CD8 T cell infiltration and indicated poor prognosis in osteosarcoma patients. Cellular functional assays revealed that cuproptosis-SPGs regulated the LC3B/ERK signaling pathway, thereby triggering cell death and impairing migration capabilities in osteosarcoma cells. Conclusion The impact of cuproptosis-related sphingolipid metabolism on the survival and migration of osteosarcoma cells, as well as on CD8 T cell infiltration, highlights the potential of targeting copper ion metabolism as a promising strategy for osteosarcoma patients.
Collapse
Affiliation(s)
- Qingbiao Li
- Department of Orthopedics, Southern Medical University Pingshan Hospital (Pingshan District Peoples’ Hospital of Shenzhen), Shenzhen, Guangdong, China
| | - Jiarui Fang
- Department of Sport Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, China
| | - Kai Liu
- Department of Sport Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, China
| | - Peng Luo
- Department of Sport Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, China
| | - Xiuzhuo Wang
- Department of Orthopedics, Southern Medical University Pingshan Hospital (Pingshan District Peoples’ Hospital of Shenzhen), Shenzhen, Guangdong, China
| |
Collapse
|
24
|
Zhang C, Zhao X, Li F, Qin J, Yang L, Yin Q, Liu Y, Zhu Z, Zhang F, Wang Z, Liang H. Integrating single-cell and multi-omic approaches reveals Euphorbiae Humifusae Herba-dependent mitochondrial dysfunction in non-small-cell lung cancer. J Cell Mol Med 2024; 28:e18317. [PMID: 38801409 PMCID: PMC11129731 DOI: 10.1111/jcmm.18317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/25/2024] [Accepted: 04/03/2024] [Indexed: 05/29/2024] Open
Abstract
Euphorbiae Humifusae Herba (EHH) is a pivotal therapeutic agent with diverse pharmacological effects. However, a substantial gap exists in understanding its pharmacological properties and anti-tumour mechanisms. This study aimed to address this gap by exploring EHH's pharmacological properties, identifying NSCLC therapy-associated protein targets, and elucidating how EHH induces mitochondrial disruption in NSCLC cells, offering insights into novel NSCLC treatment strategies. String database was utilized to explore protein-protein interactions. Subsequently, single-cell analysis and multi-omics further unveiled the impact of EHH-targeted genes on the immune microenvironment of NSCLC, as well as their influence on immunotherapeutic responses. Finally, both in vivo and in vitro experiments elucidated the anti-tumour mechanisms of EHH, specifically through the assessment of mitochondrial ROS levels and alterations in mitochondrial membrane potential. EHH exerts its influence through engagement with a cluster of 10 genes, including the apoptotic gene CASP3. This regulatory impact on the immune milieu within NSCLC holds promise as an indicator for predicting responses to immunotherapy. Besides, EHH demonstrated the capability to induce mitochondrial ROS generation and perturbations in mitochondrial membrane potential in NSCLC cells, ultimately leading to mitochondrial dysfunction and consequent apoptosis of tumour cells. EHH induces mitochondrial disruption in NSCLC cells, leading to cell apoptosis to inhibit the progress of NSCLC.
Collapse
Affiliation(s)
- Chengcheng Zhang
- Department of Medical OncologyLonghua Hospital affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Xiaoxue Zhao
- Department of Medical OncologyLonghua Hospital affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Feng Li
- Department of RheumatologyLonghua Hospital affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Jingru Qin
- Department of Medical OncologyLonghua Hospital affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Lu Yang
- Department of Medical OncologyLonghua Hospital affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Qianqian Yin
- Department of Medical OncologyLonghua Hospital affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Yiyi Liu
- Department of Medical OncologyLonghua Hospital affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Zhiyao Zhu
- Department of Medical OncologyLonghua Hospital affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Fei Zhang
- Department of General SurgeryXinhua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhongqi Wang
- Department of Medical OncologyLonghua Hospital affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Haibin Liang
- Department of General SurgeryXinhua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
25
|
Wang Y, Zhu H, Zhang L, He J, Bo J, Wang J, Ding B, Ren M. Common immunological and prognostic features of lung and bladder cancer via smoking-related genes: PRR11 gene as potential immunotherapeutic target. J Cell Mol Med 2024; 28:e18384. [PMID: 38760964 PMCID: PMC11101993 DOI: 10.1111/jcmm.18384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/17/2024] [Accepted: 04/27/2024] [Indexed: 05/20/2024] Open
Abstract
Smoking is a well-known risk factor for non-small-cell lung cancer (NSCLC) and bladder urothelial carcinoma (BLCA). Despite this, there has been no investigation into a prognostic marker based on smoking-related genes that could universally predict prognosis in these cancers and correlate with immune checkpoint therapy. This study aimed to identify smoking-related differential genes in NSCLC and BLCA, analyse their roles in patient prognosis and immune checkpoint therapy through subgroup analyses, and shed light on PRR11 as a crucial prognostic gene in both cancers. By examining PRR11 co-expressed genes, a prognostic model was constructed and its impact on immunotherapy for NSCLC and BLCA was evaluated. Molecular docking and tissue microarray analyses were conducted to explore the correlation between PRR11 and its reciprocal gene SPDL1. Additionally, miRNAs associated with PRR11 were analysed. The study confirmed a strong link between smoking-related genes, prognosis, and immune checkpoint therapy in NSCLC and BLCA. PRR11 was identified as a key smoking-associated gene that influences the efficacy of immune checkpoint therapy by modulating the stemness of these cancers. A prognostic model based on PRR11 co-expressed genes in BLCA was established and its prognostic value was validated in NSCLC. Furthermore, it was found that PRR11 regulates PDL1 via SPDL1, impacting immunotherapeutic efficacy in both cancers. The involvement of hsa-miR-200b-3p in the regulation of SPDL1 expression by PRR11 was also highlighted. Overall, the study elucidates that PRR11 modulates patient immunotherapy by influencing PDL1 expression through its interaction with SPDL1, with potential upstream regulation by hsa-miR-200b-3p.
Collapse
Affiliation(s)
- YaXuan Wang
- Department of UrologyThe First Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - HaiXia Zhu
- Department of Central LaboratoryAffiliated Tumor Hospital of Nantong University & Nantong Tumor HospitalNantongChina
| | - Lu Zhang
- Department of UrologyThe First Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - JiaXing He
- Department of UrologyThe First Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Ji Bo
- Department of UrologyThe First Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - JianShe Wang
- Department of UrologyThe First Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - BeiChen Ding
- Department of UrologyThe First Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - MingHua Ren
- Department of UrologyThe First Affiliated Hospital of Harbin Medical UniversityHarbinChina
| |
Collapse
|
26
|
Guan H, Yuan Q, Lv K, Qi Y, Jiang Y, Zhang S, Miao D, Wang Z, Lin J. Dermoscopy-based Radiomics Help Distinguish Basal Cell Carcinoma and Actinic Keratosis: A Large-scale Real-world Study Based on a 207-combination Machine Learning Computational Framework. J Cancer 2024; 15:3350-3361. [PMID: 38817855 PMCID: PMC11134443 DOI: 10.7150/jca.94759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 03/13/2024] [Indexed: 06/01/2024] Open
Abstract
This study has used machine learning algorithms to develop a predictive model for differentiating between dermoscopic images of basal cell carcinoma (BCC) and actinic keratosis (AK). We compiled a total of 904 dermoscopic images from two sources - the public dataset (HAM10000) and our proprietary dataset from the First Affiliated Hospital of Dalian Medical University (DAYISET 1) - and subsequently categorised these images into four distinct cohorts. The study developed a deep learning model for quantitative analysis of image features and integrated 15 machine learning algorithms, generating 207 algorithmic combinations through random combinations and cross-validation. The final predictive model, formed by integrating XGBoost with Lasso regression, exhibited effective performance in the differential diagnosis of BCC and AK. The model demonstrated high sensitivity in the training set and maintained stable performance in three validation sets. The area under the curve (AUC) value reached 1.000 in the training set and an average of 0.695 in the validation sets. The study concludes that the constructed discriminative diagnostic model based on machine learning algorithms has excellent predictive capabilities that could enhance clinical decision-making efficiency, reduce unnecessary biopsies, and provide valuable guidance for further treatment.
Collapse
Affiliation(s)
- Hewen Guan
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Qihang Yuan
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Kejia Lv
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yushuo Qi
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yuankuan Jiang
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Shumeng Zhang
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Dong Miao
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zhiyi Wang
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jingrong Lin
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
27
|
Petroni G, Pillozzi S, Antonuzzo L. Exploiting Tertiary Lymphoid Structures to Stimulate Antitumor Immunity and Improve Immunotherapy Efficacy. Cancer Res 2024; 84:1199-1209. [PMID: 38381540 PMCID: PMC11016894 DOI: 10.1158/0008-5472.can-23-3325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/04/2024] [Accepted: 02/19/2024] [Indexed: 02/23/2024]
Abstract
Tumor-associated tertiary lymphoid structures (TLS) have been associated with favorable clinical outcomes and response to immune checkpoint inhibitors in many cancer types, including non-small cell lung cancer. Although the detailed cellular and molecular mechanisms underlying these clinical associations have not been fully elucidated, growing preclinical and clinical studies are helping to elucidate the mechanisms at the basis of TLS formation, composition, and regulation of immune responses. However, a major challenge remains how to exploit TLS to enhance naïve and treatment-mediated antitumor immune responses. Here, we discuss the current understanding of tumor-associated TLS, preclinical models that can be used to study them, and potential therapeutic interventions to boost TLS formation, with a particular focus on lung cancer research.
Collapse
Affiliation(s)
- Giulia Petroni
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Firenze, Italy
| | - Lorenzo Antonuzzo
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy
- Clinical Oncology Unit, Careggi University Hospital, Firenze, Italy
| |
Collapse
|
28
|
Rong D, Su Y, Jia D, Zeng Z, Yang Y, Wei D, Lu H, Cao Y. Experimentally validated oxidative stress -associated prognostic signatures describe the immune landscape and predict the drug response and prognosis of SKCM. Front Immunol 2024; 15:1387316. [PMID: 38660305 PMCID: PMC11039952 DOI: 10.3389/fimmu.2024.1387316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
Background Skin Cutaneous Melanoma (SKCM) incidence is continually increasing, with chemotherapy and immunotherapy being among the most common cancer treatment modalities. This study aims to identify novel biomarkers for chemotherapy and immunotherapy response in SKCM and explore their association with oxidative stress. Methods Utilizing TCGA-SKCM RNA-seq data, we employed Weighted Gene Co-expression Network Analysis (WGCNA) and Protein-Protein Interaction (PPI) networks to identify six core genes. Gene co-expression analysis and immune-related analysis were conducted, and specific markers associated with oxidative stress were identified using Gene Set Variation Analysis (GSVA). Single-cell analysis revealed the expression patterns of Oxidative Stress-Associated Genes (OSAG) in the tumor microenvironment. TIDE analysis was employed to explore the association between immune therapy response and OSAG, while CIBERSORT was used to analyze the tumor immune microenvironment. The BEST database demonstrated the impact of the Oxidative Stress signaling pathway on chemotherapy drug resistance. Immunohistochemical staining and ROC curve evaluation were performed to assess the protein expression levels of core genes in SKCM and normal samples, with survival analysis utilized to determine their diagnostic value. Results We identified six central genes associated with SKCM metastasis, among which the expression of DSC2 and DSC3 involved in the oxidative stress pathway was closely related to immune cell infiltration. DSC2 influenced drug resistance in SKMC patients. Furthermore, downregulation of DSC2 and DSC3 expression enhanced the response of SKCM patients to immunotherapy. Conclusion This study identified two Oxidative Stress-Associated genes as novel biomarkers for SKCM. Additionally, targeting the oxidative stress pathway may serve as a new strategy in clinical practice to enhance SKCM chemotherapy and sensitivity.
Collapse
Affiliation(s)
- Dongyun Rong
- Clinical Medical School, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yushen Su
- Clinical Medical School, Guizhou Medical University, Guiyang, Guizhou, China
| | - Dechao Jia
- Clinical Medical School, Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhirui Zeng
- Department of anorectal surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yan Yang
- Department of Internal Medicine, The Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou, China
| | - Dalong Wei
- Department of Burns, Plastic Surgery and Wound Repair, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Key Laboratory of Tumor Molecular Pathology of Baise, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Honguan Lu
- Clinical Medical School, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yu Cao
- Clinical Medical School, Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
29
|
Ding H, Teng Y, Gao P, Zhang Q, Wang M, Yu Y, Fan Y, Zhu L. Construction of a prognostic model for lung adenocarcinoma based on m6A/m5C/m1A genes. Hum Mol Genet 2024; 33:563-582. [PMID: 38142284 DOI: 10.1093/hmg/ddad208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/15/2023] [Accepted: 12/07/2023] [Indexed: 12/25/2023] Open
Abstract
BACKGROUND Developing a prognostic model for lung adenocarcinoma (LUAD) that utilizes m6A/m5C/m1A genes holds immense importance in providing precise prognosis predictions for individuals. METHODS This study mined m6A/m5C/m1A-related differential genes in LUAD based on public databases, identified LUAD tumor subtypes based on these genes, and further built a risk prognostic model grounded in differential genes between subtypes. The immune status between high- and low-risk groups was investigated, and the distribution of feature genes in tumor immune cells was analyzed using single-cell analysis. Based on the expression levels of feature genes, a projection of chemotherapeutic and targeted drugs was made for individuals identified as high-risk. Ultimately, cell experiments were further verified. RESULTS The 6-gene risk prognosis model based on differential genes between tumor subtypes had good predictive performance. Individuals classified as low-risk exhibited a higher (P < 0.05) abundance of infiltrating immune cells. Feature genes were mainly distributed in tumor immune cells like CD4+T cells, CD8+T cells, and regulatory T cells. Four drugs with relatively low IC50 values were found in the high-risk group: Elesclomol, Pyrimethamine, Saracatinib, and Temsirolimus. In addition, four drugs with significant positive correlation (P < 0.001) between IC50 values and feature gene expression were found, including Alectinib, Estramustine, Brigatinib, and Elesclomol. The low expression of key gene NTSR1 reduced the IC50 value of irinotecan. CONCLUSION Based on the m6A/m5C/m1A-related genes in LUAD, LUAD patients were divided into 2 subtypes, and a m6A/m5C/m1A-related LUAD prognostic model was constructed to provide a reference for the prognosis prediction of LUAD.
Collapse
Affiliation(s)
- Hao Ding
- Department of Respiratory Disease, Affiliated People's Hospital of Jiangsu University, NO. 8 Dianli Road, Runzhou District, Zhenjiang City, Jiangsu Province 212002, China
| | - Yuanyuan Teng
- Department of Respiratory Disease, Affiliated People's Hospital of Jiangsu University, NO. 8 Dianli Road, Runzhou District, Zhenjiang City, Jiangsu Province 212002, China
| | - Ping Gao
- Department of Respiratory Disease, Affiliated People's Hospital of Jiangsu University, NO. 8 Dianli Road, Runzhou District, Zhenjiang City, Jiangsu Province 212002, China
| | - Qi Zhang
- Department of Respiratory Disease, Affiliated People's Hospital of Jiangsu University, NO. 8 Dianli Road, Runzhou District, Zhenjiang City, Jiangsu Province 212002, China
| | - Mengdi Wang
- Department of Respiratory Disease, Affiliated People's Hospital of Jiangsu University, NO. 8 Dianli Road, Runzhou District, Zhenjiang City, Jiangsu Province 212002, China
| | - Yi Yu
- Department of General Practice, Jiankang Road Community Health Service Center, NO. 239 Zhongshan East Road, Jingkou District, Zhenjiang City, Jiangsu Province 212008, China
| | - Yueping Fan
- Department of Respiratory, Jurong Branch Hospital, Affiliated Hospital of Jiangsu University, NO. 8 Huayang South Road, Jurong City, Zhenjiang City, Jiangsu Province 212400, China
| | - Li Zhu
- Department of Nephrology, Affiliated People's Hospital of Jiangsu University, NO. 8 Dianli Road, Runzhou District, Zhenjiang City, Jiangsu Province 212002, China
| |
Collapse
|
30
|
Qiao X, Sun J, Ren P, Guo H, Xu H, Bao C, Jiang C. Integrated single-cell sequencing, spatial transcriptome sequencing and bulk RNA sequencing highlights the molecular characteristics of parthanatos in gastric cancer. Aging (Albany NY) 2024; 16:5471-5500. [PMID: 38499384 PMCID: PMC11006479 DOI: 10.18632/aging.205658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 02/08/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND Parthanatos is a novel programmatic form of cell death based on DNA damage and PARP-1 dependency. Nevertheless, its specific role in the context of gastric cancer (GC) remains uncertain. METHODS In this study, we integrated multi-omics algorithms to investigate the molecular characteristics of parthanatos in GC. A series of bioinformatics algorithms were utilized to explore clinical heterogeneity of GC and further predict the clinical outcomes. RESULTS Firstly, we conducted a comprehensive analysis of the omics features of parthanatos in various human tumors, including genomic mutations, transcriptome expression, and prognostic relevance. We successfully identified 7 cell types within the GC microenvironment: myeloid cell, epithelial cell, T cell, stromal cell, proliferative cell, B cell, and NK cell. When compared to adjacent non-tumor tissues, single-cell sequencing results from GC tissues revealed elevated scores for the parthanatos pathway across multiple cell types. Spatial transcriptomics, for the first time, unveiled the spatial distribution characteristics of parthanatos signaling. GC patients with different parthanatos signals often exhibited distinct immune microenvironment and metabolic reprogramming features, leading to different clinical outcomes. The integration of parthanatos signaling and clinical indicators enabled the creation of novel survival curves that accurately assess patients' survival times and statuses. CONCLUSIONS In this study, the molecular characteristics of parthanatos' unicellular and spatial transcriptomics in GC were revealed for the first time. Our model based on parthanatos signals can be used to distinguish individual heterogeneity and predict clinical outcomes in patients with GC.
Collapse
Affiliation(s)
- Xiuli Qiao
- Department of Gastroenterology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiaao Sun
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Pingping Ren
- Department of Gastroenterology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hui Guo
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hua Xu
- Department of Gastroenterology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Chongchan Bao
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Key Laboratory of Molecular Pathology in Tumors of Guangxi, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Chunmeng Jiang
- Department of Gastroenterology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
31
|
Song D, Yang Q, Li L, Wei Y, Zhang C, Du H, Ren G, Li H. Novel prognostic biomarker TBC1D1 is associated with immunotherapy resistance in gliomas. Front Immunol 2024; 15:1372113. [PMID: 38529286 PMCID: PMC10961388 DOI: 10.3389/fimmu.2024.1372113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 02/28/2024] [Indexed: 03/27/2024] Open
Abstract
Background Glioma, an aggressive brain tumor, poses a challenge in understanding the mechanisms of treatment resistance, despite promising results from immunotherapy. Methods We identified genes associated with immunotherapy resistance through an analysis of The Cancer Genome Atlas (TCGA), Chinese Glioma Genome Atlas (CGGA), and Gene Expression Omnibus (GEO) databases. Subsequently, qRT-PCR and western blot analyses were conducted to measure the mRNA and protein levels of TBC1 Domain Family Member 1 (TBC1D1), respectively. Additionally, Gene Set Enrichment Analysis (GSEA) was employed to reveal relevant signaling pathways, and the expression of TBC1D1 in immune cells was analyzed using single-cell RNA sequencing (scRNA-seq) data from GEO database. Tumor Immune Dysfunction and Exclusion (TIDE) database was utilized to assess T-cell function, while Tumor Immunotherapy Gene Expression Resource (TIGER) database was employed to evaluate immunotherapy resistance in relation to TBC1D1. Furthermore, the predictive performance of molecules on prognosis was assessed using Kaplan-Meier plots, nomograms, and ROC curves. Results The levels of TBC1D1 were significantly elevated in tumor tissue from glioma patients. Furthermore, high TBC1D1 expression was observed in macrophages compared to other cells, which negatively impacted T cell function, impaired immunotherapy response, promoted treatment tolerance, and led to poor prognosis. Inhibition of TBC1D1 was found to potentially synergistically enhance the efficacy of immunotherapy and prolong the survival of cancer patients with gliomas. Conclusion Heightened expression of TBC1D1 may facilitate an immunosuppressive microenvironment and predict a poor prognosis. Blocking TBC1D1 could minimize immunotherapy resistance in cancer patients with gliomas.
Collapse
Affiliation(s)
- Daqiang Song
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Qian Yang
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liuying Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuxian Wei
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chong Zhang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huimin Du
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongzhong Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
32
|
Liu K, Wang Y, Shao W, Tang C, Zhao S, Xu J, Xu P, Cheng Q, Huang S, Ji P, Qiu S. Unveiling the oncogenic role of CLDN11-secreting fibroblasts in gastric cancer peritoneal metastasis through single-cell sequencing and experimental approaches. Int Immunopharmacol 2024; 129:111647. [PMID: 38335659 DOI: 10.1016/j.intimp.2024.111647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/14/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Fibroblasts are necessary to the progression of cancer. However, the role of fibroblasts in peritoneal metastasis (PM) of gastric cancer (GC) remains elusive. In this study, we would explore the role of fibroblasts mediated cell interaction in PM of GC. METHODS Single-cell sequencing data from public database GSE183904 was used to explore the specific fibroblast cluster. Fibroblasts were extracted from PM and GC tissues. The expression level of CXCR7 was verified by western blot, immunohistochemistry. The role of CLDN11 was investigate through in vitro and in vivo study. Multiple immunohistochemistry was used to characterize the tumor microenvironment. RESULTS CXCR7-positive fibroblasts were significantly enriched in PM of GC. CXCR7 could promote the expression of CLDN11 through activation of the AKT pathway in fibroblasts. Fibroblasts promote the GC proliferation and peritoneal metastasis by secreting CLDN11 in vitro and in vivo. Furthermore, it was revealed that CXCR7-positive fibroblasts were significantly associated with M2-type macrophages infiltration in tissues. CONCLUSION CXCR7-positive fibroblasts play an essential role in PM of GC via CLDN11. Therapy targeting CXCR7-positive fibroblasts or CLDN11 may be helpful in the treatment of GC with PM.
Collapse
Affiliation(s)
- Kanghui Liu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yanjuan Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Wenwen Shao
- Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong, China
| | - Chong Tang
- Department of General Surgery, Nantong First People's Hospital, Nantong, Jiangsu Province, China
| | - Siguo Zhao
- Department of Clinical Medicine, The First School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Jiafeng Xu
- Department of Clinical Medicine, The First School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Peng Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Quan Cheng
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Shansong Huang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Peicheng Ji
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Shengkui Qiu
- Department of General Surgery, Nantong First People's Hospital, Nantong, Jiangsu Province, China.
| |
Collapse
|
33
|
Yang Q, Li B, Luan T, Wang X, Duan B, Wei C, Chen S. Exploring blood lipids-immunity associations following HBV vaccination: evidence from a large cross-sectional study. Front Cell Infect Microbiol 2024; 14:1369661. [PMID: 38524185 PMCID: PMC10959126 DOI: 10.3389/fcimb.2024.1369661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 02/19/2024] [Indexed: 03/26/2024] Open
Abstract
Introduction Serological responses following hepatitis B vaccination are crucial for preventing hepatitis B (HBV). However, the potential relationship between serum lipid levels and immunity from HBV vaccination remains poorly understood. Methods In this study, we conducted an analysis of the National Health and Nutrition Examination Survey (NHANES) data spanning from 2003 to 2016. Multivariable weighted logistic regression models, generalized linear analysis, stratified models, smooth curve fitting, segmentation effect analysis and sensitivity analysis were utilized to assess the relationships. Results After adjusting for relevant covariates, we observed that low levels of high-density lipoprotein cholesterol (HDL) were independently linked to a significantly lower seroprotective rate. Compared to HDL levels of ≥ 60 mg/dL, the odds ratios (ORs) for individuals with borderline levels (40-59 mg/dL for men, 50-59 mg/dL for women) and low levels (< 40 mg/dL for men, < 50 mg/dL for women) were 0.83 (95% CI 0.69-0.99) and 0.65 (95% CI 0.56-0.78), respectively. This association was particularly pronounced in individuals aged 40 or older. Conversely, higher levels of the triglyceride to HDL (TG/HDL) ratio (OR, 0.90; 95% CI, 0.84-0.98), total cholesterol to HDL (Chol/HDL) ratio (OR, 0.77; 95% CI, 0.64-0.92), and low-density lipoprotein to HDL (LDL/HDL) ratio (OR, 0.85; 95% CI, 0.76-0.96) were associated with a decreased likelihood of seroprotection. Conclusion This study suggests that lipid levels may play a role in modulating the immune response following HBV vaccination.
Collapse
Affiliation(s)
- Qian Yang
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Benhua Li
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tiankuo Luan
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyu Wang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bixia Duan
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chengcheng Wei
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shi Chen
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
34
|
Lin Z, Li X, Shi H, Cao R, Zhu L, Dang C, Sheng Y, Fan W, Yang Z, Wu S. Decoding the tumor microenvironment and molecular mechanism: unraveling cervical cancer subpopulations and prognostic signatures through scRNA-Seq and bulk RNA-seq analyses. Front Immunol 2024; 15:1351287. [PMID: 38482016 PMCID: PMC10933018 DOI: 10.3389/fimmu.2024.1351287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/13/2024] [Indexed: 04/13/2024] Open
Abstract
Background Cervical carcinoma (CC) represents a prevalent gynecological neoplasm, with a discernible rise in prevalence among younger cohorts observed in recent years. Nonetheless, the intrinsic cellular heterogeneity of CC remains inadequately investigated. Methods We utilized single-cell RNA sequencing (scRNA-seq) transcriptomic analysis to scrutinize the tumor epithelial cells derived from four specimens of cervical carcinoma (CC) patients. This method enabled the identification of pivotal subpopulations of tumor epithelial cells and elucidation of their contributions to CC progression. Subsequently, we assessed the influence of associated molecules in bulk RNA sequencing (Bulk RNA-seq) cohorts and performed cellular experiments for validation purposes. Results Through our analysis, we have discerned C3 PLP2+ Tumor Epithelial Progenitor Cells as a noteworthy subpopulation in cervical carcinoma (CC), exerting a pivotal influence on the differentiation and progression of CC. We have established an independent prognostic indicator-the PLP2+ Tumor EPCs score. By stratifying patients into high and low score groups based on the median score, we have observed that the high-score group exhibits diminished survival rates compared to the low-score group. The correlations observed between these groups and immune infiltration, enriched pathways, single-nucleotide polymorphisms (SNPs), drug sensitivity, among other factors, further underscore their impact on CC prognosis. Cellular experiments have validated the significant impact of ATF6 on the proliferation and migration of CC cell lines. Conclusion This study enriches our comprehension of the determinants shaping the progression of CC, elevates cognizance of the tumor microenvironment in CC, and offers valuable insights for prospective CC therapies. These discoveries contribute to the refinement of CC diagnostics and the formulation of optimal therapeutic approaches.
Collapse
Affiliation(s)
- Zhiheng Lin
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xinhan Li
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Hengmei Shi
- Department of Obstetrics and Gynecology, Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Renshuang Cao
- Wangjing Hospital of Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Lijun Zhu
- Longhua Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chunxiao Dang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yawen Sheng
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Weisen Fan
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | | | - Siyu Wu
- Department of Gynecology and Obstetrics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Qingdao, China
| |
Collapse
|
35
|
Liu X, Xu F, Zhao K, Liu Y, Ye G, Zhang X, Qu Y. Comprehending the cuproptosis and cancer-immunity cycle network: delving into the immune landscape and its predictive role in breast cancer immunotherapy responses and clinical endpoints. Front Immunol 2024; 15:1344023. [PMID: 38312844 PMCID: PMC10834629 DOI: 10.3389/fimmu.2024.1344023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/02/2024] [Indexed: 02/06/2024] Open
Abstract
Background The role of cuproptosis, a phenomenon associated with tumor metabolism and immunological identification, remains underexplored, particularly in relation to the cancer-immunity cycle (CIC) network. This study aims to rigorously examine the impact of the cuproptosis-CIC nexus on immune reactions and prognostic outcomes in patients with breast cancer (BC), striving to establish a comprehensive prognostic model. Methods In the study, we segregated data obtained from TCGA, GEO, and ICGC using CICs retrieved from the TIP database. We constructed a genetic prognostic framework using the LASSO-Cox model, followed by its validation through Cox proportional hazards regression. This framework's validity was further confirmed with data from ICGC and GEO. Explorations of the tumor microenvironment were carried out through the application of ESTIMATE and CIBERSORT algorithms, as well as machine learning techniques, to identify potential treatment strategies. Single-cell sequencing methods were utilized to delineate the spatial distribution of key genes within the various cell types in the tumor milieu. To explore the critical role of the identified CICs, experiments were conducted focusing on cell survival and migration abilities. Results In our research, we identified a set of 4 crucial cuproptosis-CICs that have a profound impact on patient longevity and their response to immunotherapy. By leveraging these identified CICs, we constructed a predictive model that efficiently estimates patient prognoses. Detailed analyses at the single-cell level showed that the significance of CICs. Experimental approaches, including CCK-8, Transwell, and wound healing assays, revealed that the protein HSPA9 restricts the growth and movement of breast cancer cells. Furthermore, our studies using immunofluorescence techniques demonstrated that suppressing HSPA9 leads to a notable increase in ceramide levels. Conclusion This research outlines a network of cuproptosis-CICs and constructs a predictive nomogram. Our model holds great promise for healthcare professionals to personalize treatment approaches for individuals with breast cancer. The work provides insights into the complex relationship between the cuproptosis-CIC network and the cancer immune microenvironment, setting the stage for novel approaches to cancer immunotherapy. By focusing on the essential gene HSPA9 within the cancer-immunity cycle, this strategy has the potential to significantly improve the efficacy of treatments against breast cancer.
Collapse
Affiliation(s)
- Xiangwei Liu
- Department of Breast Surgery, The First People’s Hospital of Foshan, Foshan, China
| | - Feng Xu
- Department of Anesthesiology, The First People’s Hospital of Foshan, Foshan, China
| | - Kunkun Zhao
- Department of Breast Surgery, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, China
| | - Yunfei Liu
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Guolin Ye
- Department of Breast Surgery, The First People’s Hospital of Foshan, Foshan, China
| | - Xin Zhang
- Department of Pathology, the Second People’s Hospital of Foshan, Foshan, China
| | - Yanyu Qu
- Department of Pathology, the Second People’s Hospital of Foshan, Foshan, China
| |
Collapse
|
36
|
Zhao Y, Ren J. 18F-FAPI-04 PET/CT parameters predict PD-L1 expression in esophageal squamous cell carcinoma. Front Immunol 2023; 14:1266843. [PMID: 38035081 PMCID: PMC10684668 DOI: 10.3389/fimmu.2023.1266843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/25/2023] [Indexed: 12/02/2023] Open
Abstract
Purpose This prospective study examined whether metabolism parameters obtained using the tracer 18F-AlFNOTA-fibroblast activation protein inhibitor (FAPI)-04 (denoted as 18F-FAPI-04) in positron emission tomography/computed tomography (PET/CT) can predict programmed death ligand-1 (PD-L1) expression in patients with locally advanced esophageal squamous cell carcinoma (LA-ESCC). Patients and methods The 24 enrolled LA-ESCC patients underwent an 18F-FAPI-04 PET/CT scan. The maximum, mean, peak and standard deviation standard uptake values (SUVmax, SUVmean, SUVpeak and SUVsd), metabolic tumor volume (MTV), and total lesion FAP (TLF) expression of the primary tumor were collected. Additionally, we evaluated PD-L1 expression on cancer cells by immunohistochemistry and immunofluorescence methods. Patients were divided into negative and positive expressions according to the expression of PD-L1 (CPS < 10 and CPS ≥ 10), and the variables were compared between the two groups. Results The SUVmax, SUVmean, SUVpeak and SUVsd were significantly higher in patients with positive expression than in negative expression (all p < 0.05). Receiver operating characteristic curve analysis identified SUVmean (area under the curve [AUC] = 0.882, p = 0.004), SUVsd (AUC = 0.874, p = 0.005), SUVpeak (AUC = 0.840, p = 0.010) and SUVmax (AUC = 0.765, p = 0.045) as significant predictors of the PD-L1 positive expression, with cutoff values of 9.67, 1.90, 9.67 and 13.71, respectively. On univariate logistic regression analysis, SUVmean (p = 0.045), SUVsd (p = 0.024), and SUVpeak (p = 0.031) were significantly correlated with the PD-L1 positive expression. On multivariable logistic regression analysis, SUVsd (p = 0.035) was an optimum predictor factor for PD-L1 positive expression. Conclusion 18F-FAPI-04 PET/CT parameters, including SUVmean, SUVpeak, and SUVsd, correlated with PD-L1 expression in patients with LA-ESCC, and thus SUVsd was an optimum predictor for PD-L1 positive expression, which could help to explore the existence of immune checkpoints and select ESCC candidates for immunotherapy.
Collapse
Affiliation(s)
- Yaqing Zhao
- Department of General Affairs Section, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jiazhong Ren
- Department of Medical Imaging, PET-CT Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
37
|
Shi T, Li M, Yu Y. Machine learning-enhanced insights into sphingolipid-based prognostication: revealing the immunological landscape and predictive proficiency for immunomotherapy and chemotherapy responses in pancreatic carcinoma. Front Mol Biosci 2023; 10:1284623. [PMID: 38028544 PMCID: PMC10643633 DOI: 10.3389/fmolb.2023.1284623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Background: With a poor prognosis for affected individuals, pancreatic adenocarcinoma (PAAD) is known as a complicated and diverse illness. Immunocytes have become essential elements in the development of PAAD. Notably, sphingolipid metabolism has a dual function in the development of tumors and the invasion of the immune system. Despite these implications, research on the predictive ability of sphingolipid variables for PAAD prognosis is strikingly lacking, and it is yet unclear how they can affect PAAD immunotherapy and targeted pharmacotherapy. Methods: The investigation process included SPG detection while also being pertinent to the prognosis for PAAD. Both the analytical capability of CIBERSORT and the prognostic capability of the pRRophetic R package were used to evaluate the immunological environments of the various HCC subtypes. In addition, CCK-8 experiments on PAAD cell lines were carried out to confirm the accuracy of drug sensitivity estimates. The results of these trials, which also evaluated cell survival and migratory patterns, confirmed the usefulness of sphingolipid-associated genes (SPGs). Results: As a result of this thorough investigation, 32 SPGs were identified, each of which had a measurable influence on the dynamics of overall survival. This collection of genes served as the conceptual framework for the development of a prognostic model, which was carefully assembled from 10 chosen genes. It should be noted that this grouping of patients into cohorts with high and low risk was a sign of different immune profiles and therapy responses. The increased abundance of SPGs was identified as a possible sign of inadequate responses to immune-based treatment approaches. The careful CCK-8 testing carried out on PAAD cell lines was of the highest importance for providing clear confirmation of drug sensitivity estimates. Conclusion: The significance of Sphingolipid metabolism in the complex web of PAAD development is brought home by this study. The novel risk model, built on the complexity of sphingolipid-associated genes, advances our understanding of PAAD and offers doctors a powerful tool for developing personalised treatment plans that are specifically suited to the unique characteristics of each patient.
Collapse
Affiliation(s)
| | | | - Yabin Yu
- Department of Hepatobiliary Surgery, The Affiliated Huaian No 1 People’s Hospital of Nanjing Medical University, Huaian, China
| |
Collapse
|
38
|
Zhao X, Zhang J, Liu J, Luo S, Ding R, Miao X, Wu T, Jia J, Cheng X. Molecular characterization of cancer-intrinsic immune evasion genes indicates prognosis and tumour microenvironment infiltration in osteosarcoma. Aging (Albany NY) 2023; 15:10272-10290. [PMID: 37796192 PMCID: PMC10599718 DOI: 10.18632/aging.205074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/08/2023] [Indexed: 10/06/2023]
Abstract
Cancer-intrinsic immune evasion (IE) to cells is a critical factor in tumour growth and progression, yet the molecular characterization of IE genes (IEGs) in osteosarcoma remains underexplored. In this study, 85 osteosarcoma patients were comprehensively analyzed based on 182 IEGs, leading to the identification of two IE clusters linked to distinct biological processes and clinical outcomes. In addition, two IE clusters demonstrated diverse immune cell infiltration patterns, with IEGcluster A displaying increased levels compared to IEGcluster B. Moreover, an IE score was identified as an independent prognostic factor and nomogram may serve as a practical tool for the individual prognostic evaluation of patients with osteosarcoma. Finally, GBP1, a potential biomarker with high expression in osteosarcoma was identified. The findings of this study highlight the presence of two IE clusters, each associated with differing patient outcomes and immune infiltration properties. The IE score may serve to assess individual patient IE characteristics, enhance comprehension of immune features, and guide more efficacious treatment approaches.
Collapse
Affiliation(s)
- Xiaokun Zhao
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Jian Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
- Institute of Orthopedics of Jiangxi, Nanchang, Jiangxi 330006, China
| | - Jiahao Liu
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Shengzhong Luo
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Rui Ding
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xinxin Miao
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
- Institute of Minimally Invasive Orthopedics, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Tianlong Wu
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
- Institute of Minimally Invasive Orthopedics, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Jingyu Jia
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xigao Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
- Institute of Orthopedics of Jiangxi, Nanchang, Jiangxi 330006, China
- Institute of Minimally Invasive Orthopedics, Nanchang University, Nanchang, Jiangxi 330006, China
| |
Collapse
|
39
|
Zou L, Meng L, Xu Y, Wang K, Zhang J. Revealing the diagnostic value and immune infiltration of senescence-related genes in endometriosis: a combined single-cell and machine learning analysis. Front Pharmacol 2023; 14:1259467. [PMID: 37860112 PMCID: PMC10583561 DOI: 10.3389/fphar.2023.1259467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 09/05/2023] [Indexed: 10/21/2023] Open
Abstract
Introduction: Endometriosis is a prevalent and recurrent medical condition associated with symptoms such as pelvic discomfort, dysmenorrhea, and reproductive challenges. Furthermore, it has the potential to progress into a malignant state, significantly impacting the quality of life for affected individuals. Despite its significance, there is currently a lack of precise and non-invasive diagnostic techniques for this condition. Methods: In this study, we leveraged microarray datasets and employed a multifaceted approach. We conducted differential gene analysis, implemented weighted gene co-expression network analysis (WGCNA), and utilized machine learning algorithms, including random forest, support vector machine, and LASSO analysis, to comprehensively explore senescence-related genes (SRGs) associated with endometriosis. Discussion: Our comprehensive analysis, which also encompassed profiling of immune cell infiltration and single-cell analysis, highlights the therapeutic potential of this gene assemblage as promising targets for alleviating endometriosis. Furthermore, the integration of these biomarkers into diagnostic protocols promises to enhance diagnostic precision, offering a more effective diagnostic journey for future endometriosis patients in clinical settings. Results: Our meticulous investigation led to the identification of a cluster of genes, namely BAK1, LMNA, and FLT1, which emerged as potential discerning biomarkers for endometriosis. These biomarkers were subsequently utilized to construct an artificial neural network classifier model and were graphically represented in the form of a Nomogram.
Collapse
Affiliation(s)
- Lian Zou
- Chongqing Emergency Medical Center, Department of Obstetrics and Gynecology in Chongging University Central Hospital, Chongqing, China
| | - Lou Meng
- Chongqing Emergency Medical Center, Department of Obstetrics and Gynecology in Chongging University Central Hospital, Chongqing, China
| | - Yan Xu
- Chongqing Emergency Medical Center, Department of Obstetrics and Gynecology in Chongging University Central Hospital, Chongqing, China
| | - Kana Wang
- Department of Gynecology, West China Second Hospital of Sichuan University, Chengdu, China
| | - Jiawen Zhang
- Department of Gynecology, West China Second Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
40
|
Zhang P, Dong S, Sun W, Zhong W, Xiong J, Gong X, Li J, Lin H, Zhuang Y. Deciphering Treg cell roles in esophageal squamous cell carcinoma: a comprehensive prognostic and immunotherapeutic analysis. Front Mol Biosci 2023; 10:1277530. [PMID: 37842637 PMCID: PMC10568469 DOI: 10.3389/fmolb.2023.1277530] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023] Open
Abstract
Background: Esophageal squamous cell carcinoma (ESCC) is a prevalent and aggressive form of cancer that poses significant challenges in terms of prognosis and treatment. Regulatory T cells (Treg cells) have gained attention due to their influential role in immune modulation within the tumor microenvironment (TME). Understanding the intricate interactions between Treg cells and the tumor microenvironment is essential for unraveling the mechanisms underlying ESCC progression and for developing effective prognostic models and immunotherapeutic strategies. Methods: A combination of single-cell RNA sequencing (scRNA-seq) and bulk RNA-seq analysis was utilized to explore the role of Treg cells within the TME of ESCC. The accuracy and applicability of the prognostic model were assessed through multi-dimensional evaluations, encompassing an examination of the model's performance across various dimensions, such as the mutation landscape, clinical relevance, enrichment analysis, and potential implications for immunotherapy strategies. Results: The pivotal role of the macrophage migration inhibitory factor (MIF) signaling pathway within the ESCC TME was investigated, with a focus on its impact on Treg cells and other subpopulations. Through comprehensive integration of bulk sequencing data, a Treg-associated signature (TAS) was constructed, revealing that ESCC patients with elevated TAS (referred to as high-TAS individuals) experienced significantly improved prognoses. Heightened immune infiltration and increased expression of immune checkpoint markers were observed in high-TAS specimens. The model's validity was established through the IMvigor210 dataset, demonstrating its robustness in predicting prognosis and responsiveness to immunotherapy. Heightened therapeutic benefits were observed in immune-based interventions for high-TAS ESCC patients. Noteworthy differences in pathway enrichment patterns emerged between high and low-TAS cohorts, highlighting potential avenues for therapeutic exploration. Furthermore, the clinical relevance of key model genes was substantiated by analyzing clinical samples from ten paired tumor and adjacent tissues, revealing differential expression levels. Conclusion: The study established a TAS that enables accurate prediction of patient prognosis and responsiveness to immunotherapy. This achievement holds significant implications for the clinical management of ESCC, offering valuable insights for informed therapeutic interventions.
Collapse
Affiliation(s)
- Pengpeng Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shiyang Dong
- Department of General Surgery, Fuyang Tumour Hospital, Fuyang, China
| | - Wei Sun
- Department of Thoracic Surgery, The Second Hospital of Nanjing, Nanjing, China
| | - Wan Zhong
- Department of General Surgery, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingwen Xiong
- Department of Sports Rehabilitation, Southwest Medical University, Luzhou, China
| | - Xiangjin Gong
- Department of Sports Rehabilitation, Southwest Medical University, Luzhou, China
| | - Jun Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Haoran Lin
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu Zhuang
- Department of Thoracic Surgery, Nanjing Chest Hospital, Nanjing, China
- Afliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
41
|
Li Y, Cai H, Yang J, Xie X, Pei S, Wu Y, Zhang J, Song G, Zhang J, Zhang Q, Chi H, Yang G. Decoding tumor heterogeneity in uveal melanoma: basement membrane genes as novel biomarkers and therapeutic targets revealed by multi-omics approaches for cancer immunotherapy. Front Pharmacol 2023; 14:1264345. [PMID: 37822877 PMCID: PMC10562578 DOI: 10.3389/fphar.2023.1264345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/15/2023] [Indexed: 10/13/2023] Open
Abstract
Background: Uveal melanoma (UVM) is a primary intraocular malignancy that poses a significant threat to patients' visual function and life. The basement membrane (BM) is critical for establishing and maintaining cell polarity, adult function, embryonic and organ morphogenesis, and many other biological processes. Some basement membrane protein genes have been proven to be prognostic biomarkers for various cancers. This research aimed to develop a novel risk assessment system based on BMRGs that would serve as a theoretical foundation for tailored and accurate treatment. Methods: We used gene expression profiles and clinical data from the TCGA-UVM cohort of 80 UVM patients as a training set. 56 UVM patients from the combined cohort of GSE84976 and GSE22138 were employed as an external validation dataset. Prognostic characteristics of basement membrane protein-related genes (BMRGs) were characterized by Lasso, stepwise multifactorial Cox. Multivariate analysis revealed BMRGs to be independent predictors of UVM. The TISCH database probes the crosstalk of BMEGs in the tumor microenvironment at the single-cell level. Finally, we investigated the function of ITGA5 in UVM using multiple experimental techniques, including CCK8, transwell, wound healing assay, and colony formation assay. Results: There are three genes in the prognostic risk model (ADAMTS10, ADAMTS14, and ITGA5). After validation, we determined that the model is quite reliable and accurately forecasts the prognosis of UVM patients. Immunotherapy is more likely to be beneficial for UVM patients in the high-risk group, whereas the survival advantage may be greater for UVM patients in the low-risk group. Knockdown of ITGA5 expression was shown to inhibit the proliferation, migration, and invasive ability of UVM cells in vitro experiments. Conclusion: The 3-BMRGs feature model we constructed has excellent predictive performance which plays a key role in the prognosis, informing the individualized treatment of UVM patients. It also provides a new perspective for assessing pre-immune efficacy.
Collapse
Affiliation(s)
- Yunyue Li
- Queen Mary College, Medical School of Nanchang University, Nanchang, China
| | - Huabao Cai
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jinyan Yang
- School of Stomatology, Southwest Medical University, Luzhou, China
| | - Xixi Xie
- School of Stomatology, Southwest Medical University, Luzhou, China
| | - Shengbin Pei
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yifan Wu
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jinhao Zhang
- School of Stomatology, Southwest Medical University, Luzhou, China
| | - Guobin Song
- School of Stomatology, Southwest Medical University, Luzhou, China
| | - Jieying Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qinhong Zhang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH, United States
| |
Collapse
|
42
|
Zhang S, Jiang C, Jiang L, Chen H, Huang J, Zhang J, Wang R, Chi H, Yang G, Tian G. Uncovering the immune microenvironment and molecular subtypes of hepatitis B-related liver cirrhosis and developing stable a diagnostic differential model by machine learning and artificial neural networks. Front Mol Biosci 2023; 10:1275897. [PMID: 37808522 PMCID: PMC10556489 DOI: 10.3389/fmolb.2023.1275897] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 09/14/2023] [Indexed: 10/10/2023] Open
Abstract
Background: Hepatitis B-related liver cirrhosis (HBV-LC) is a common clinical disease that evolves from chronic hepatitis B (CHB). The development of cirrhosis can be suppressed by pharmacological treatment. When CHB progresses to HBV-LC, the patient's quality of life decreases dramatically and drug therapy is ineffective. Liver transplantation is the most effective treatment, but the lack of donor required for transplantation, the high cost of the procedure and post-transplant rejection make this method unsuitable for most patients. Methods: The aim of this study was to find potential diagnostic biomarkers associated with HBV-LC by bioinformatics analysis and to classify HBV-LC into specific subtypes by consensus clustering. This will provide a new perspective for early diagnosis, clinical treatment and prevention of HCC in HBV-LC patients. Two study-relevant datasets, GSE114783 and GSE84044, were retrieved from the GEO database. We screened HBV-LC for feature genes using differential analysis, weighted gene co-expression network analysis (WGCNA), and three machine learning algorithms including least absolute shrinkage and selection operator (LASSO), support vector machine recursive feature elimination (SVM-RFE), and random forest (RF) for a total of five methods. After that, we constructed an artificial neural network (ANN) model. A cohort consisting of GSE123932, GSE121248 and GSE119322 was used for external validation. To better predict the risk of HBV-LC development, we also built a nomogram model. And multiple enrichment analyses of genes and samples were performed to understand the biological processes in which they were significantly enriched. And the different subtypes of HBV-LC were analyzed using the Immune infiltration approach. Results: Using the data downloaded from GEO, we developed an ANN model and nomogram based on six feature genes. And consensus clustering of HBV-LC classified them into two subtypes, C1 and C2, and it was hypothesized that patients with subtype C2 might have milder clinical symptoms by immune infiltration analysis. Conclusion: The ANN model and column line graphs constructed with six feature genes showed excellent predictive power, providing a new perspective for early diagnosis and possible treatment of HBV-LC. The delineation of HBV-LC subtypes will facilitate the development of future clinical treatment of HBV-LC.
Collapse
Affiliation(s)
- Shengke Zhang
- Department of Clinical Medicine, School of Clinical Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chenglu Jiang
- Department of Clinical Medicine, School of Clinical Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lai Jiang
- Department of Clinical Medicine, School of Clinical Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Haiqing Chen
- Department of Clinical Medicine, School of Clinical Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jinbang Huang
- Department of Clinical Medicine, School of Clinical Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jieying Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Rui Wang
- Department of General Surgery (Hepatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, China
| | - Hao Chi
- Department of Clinical Medicine, School of Clinical Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, United States
| | - Gang Tian
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China
| |
Collapse
|
43
|
Guo Q, Li K, Jiang N, Zhou R, Rao XR, Wu CY. A novel risk model of three gefitinib-related genes FBP1, SBK1 and AURKA is related to the immune microenvironment and is predicting prognosis of lung adenocarcinoma patients. Aging (Albany NY) 2023; 15:9633-9660. [PMID: 37737707 PMCID: PMC10564433 DOI: 10.18632/aging.205040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/21/2023] [Indexed: 09/23/2023]
Abstract
PURPOSE Gefitinib, an anticancer drug, has been reported to potentially improve the prognosis of patients with lung adenocarcinoma (LUAD). This study aims to investigate the roles and mechanisms of Gefitinib. METHODS The effects of Gefitinib on the growth and migration of LUAD cells were assessed using various methods, including CCK-8, flow cytometry, wound healing, and Transwell assays. To analyze the function and mechanisms of the differentially expressed Gefitinib target genes (GTGs), data from the TCGA database were utilized. Kaplan-Meier survival and ROC analysis identified prognostic-related GTGs and constructed a prognostic nomogram in LUAD. Consensus clustering, COX analysis and survival analysis evaluated the relationship between GTGs and the prognosis of LUAD patients. The mechanisms of the risk model involved LUAD progression, and the relationship between the risk model and immune microenvironment were investigated. RESULTS Gefitinib could inhibit proliferation, migration and invasion and promote cell apoptosis. 84 DEGTGs were involved in RAS, MAPK, ERBB pathways. The DEGTGs (FBP1, SBK1, and AURKA) were the independent risk factors for dismal prognosis of LUAD patients and were used to establish risk model and nomogram. Gefitinib could promote the expression of FBP1 and inhibit the expression of SBK1 and AURKA. High-risk LUAD patients had the dismal prognosis, and the high-risk score group was significantly associated with the immune microenvironment. CONCLUSION FBP1, SBK1, and AURKA are prognostic risk factors, and the risk model and nomogram of FBP1, SBK1 and AURKA are associated with dismal prognosis and immune cell infiltration, and have huge prospects for application in evaluating the prognosis in LUAD.
Collapse
Affiliation(s)
- Qiang Guo
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Kai Li
- Department of Hepatobiliary and Pancreatic Surgery, The People’s Hospital of Jianyang City, Jianyang, China
| | - Ni Jiang
- Department of Obstetrics and Gynecology, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Rui Zhou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin-Rui Rao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuang-Yan Wu
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
44
|
Xu W, Zhang W, Zhao D, Wang Q, Zhang M, Li Q, Zhu W, Xu C. Unveiling the role of regulatory T cells in the tumor microenvironment of pancreatic cancer through single-cell transcriptomics and in vitro experiments. Front Immunol 2023; 14:1242909. [PMID: 37753069 PMCID: PMC10518406 DOI: 10.3389/fimmu.2023.1242909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 08/28/2023] [Indexed: 09/28/2023] Open
Abstract
Background In order to investigate the impact of Treg cell infiltration on the immune response against pancreatic cancer within the tumor microenvironment (TME), and identify crucial mRNA markers associated with Treg cells in pancreatic cancer, our study aims to delve into the role of Treg cells in the anti-tumor immune response of pancreatic cancer. Methods The ordinary transcriptome data for this study was sourced from the GEO and TCGA databases. It was analyzed using single-cell sequencing analysis and machine learning. To assess the infiltration level of Treg cells in pancreatic cancer tissues, we employed the CIBERSORT method. The identification of genes most closely associated with Treg cells was accomplished through the implementation of weighted gene co-expression network analysis (WGCNA). Our analysis of single-cell sequencing data involved various quality control methods, followed by annotation and advanced analyses such as cell trajectory analysis and cell communication analysis to elucidate the role of Treg cells within the pancreatic cancer microenvironment. Additionally, we categorized the Treg cells into two subsets: Treg1 associated with favorable prognosis, and Treg2 associated with poor prognosis, based on the enrichment scores of the key genes. Employing the hdWGCNA method, we analyzed these two subsets to identify the critical signaling pathways governing their mutual transformation. Finally, we conducted PCR and immunofluorescence staining in vitro to validate the identified key genes. Results Based on the results of immune infiltration analysis, we observed significant infiltration of Treg cells in the pancreatic cancer microenvironment. Subsequently, utilizing the WGCNA and machine learning algorithms, we ultimately identified four Treg cell-related genes (TRGs), among which four genes exhibited significant correlations with the occurrence and progression of pancreatic cancer. Among them, CASP4, TOB1, and CLEC2B were associated with poorer prognosis in pancreatic cancer patients, while FYN showed a correlation with better prognosis. Notably, significant differences were found in the HIF-1 signaling pathway between Treg1 and Treg2 cells identified by the four genes. These conclusions were further validated through in vitro experiments. Conclusion Treg cells played a crucial role in the pancreatic cancer microenvironment, and their presence held a dual significance. Recognizing this characteristic was vital for understanding the limitations of Treg cell-targeted therapies. CASP4, FYN, TOB1, and CLEC2B exhibited close associations with infiltrating Treg cells in pancreatic cancer, suggesting their involvement in Treg cell functions. Further investigation was warranted to uncover the mechanisms underlying these associations. Notably, the HIF-1 signaling pathway emerged as a significant pathway contributing to the duality of Treg cells. Targeting this pathway could potentially revolutionize the existing treatment approaches for pancreatic cancer.
Collapse
Affiliation(s)
- Wei Xu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Wenjia Zhang
- Shanghai Clinical College, Anhui Medical University, Shanghai, China
- Department of Respiratory Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Dongxu Zhao
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Man Zhang
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- The Laboratory of Emergency Medicine, School of the Secondary Clinical Medicine, Xuzhou Medical University, Xuzhou, China
| | - Qiang Li
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Wenxin Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Gastroenterology, Kunshan Third People’s Hospital, Suzhou, Jiangsu, China
| | - Chunfang Xu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
45
|
Zhang P, Zhang X, Cui Y, Gong Z, Wang W, Lin S. Revealing the role of regulatory T cells in the tumor microenvironment of lung adenocarcinoma: a novel prognostic and immunotherapeutic signature. Front Immunol 2023; 14:1244144. [PMID: 37671160 PMCID: PMC10476870 DOI: 10.3389/fimmu.2023.1244144] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 07/31/2023] [Indexed: 09/07/2023] Open
Abstract
Background Regulatory T cells (Tregs), are a key class of cell types in the immune system. In the tumor microenvironment (TME), the presence of Tregs has important implications for immune response and tumor development. Relatively little is known about the role of Tregs in lung adenocarcinoma (LUAD). Methods Tregs were identified using but single-cell RNA sequencing (scRNA-seq) analysis and interactions between Tregs and other cells in the TME were investigated. Next, we used multiple bulk RNA-seq datasets to construct risk models based on marker genes of Tregs and explored differences in prognosis, mutational landscape, immune cell infiltration and immunotherapy between high- and low-risk groups, and finally, qRT-PCR and cell function experiments were performed to validate the model genes. Results The cellchat analysis showed that MIF-(CD74+CXCR4) pairs play a key role in the interaction of Tregs with other cell subpopulations, and the Tregs-associated signatures (TRAS) could well classify multiple LUAD cohorts into high- and low-risk groups. Immunotherapy may offer greater potential benefits to the low-risk group, as indicated by their superior survival, increased infiltration of immune cells, and heightened expression of immune checkpoints. Finally, the experiment verified that the model genes LTB and PTTG1 were relatively highly expressed in cancer tissues, while PTPRC was relatively highly expressed in paracancerous tissues. Colony Formation assay confirmed that knockdown of PTTG1 reduced the proliferation ability of LUAD cells. Conclusion TRAS were constructed using scRNA-seq and bulk RNA-seq to distinguish patient risk subgroups, which may provide assistance in the clinical management of LUAD patients.
Collapse
Affiliation(s)
- Pengpeng Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xiao Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yanan Cui
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zetian Gong
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shengrong Lin
- Department of Thoracic Surgery, Dongtai People’s Hospital, Dongtai, China
| |
Collapse
|
46
|
Han X, Yan Z, Fan K, Guan X, Hu B, Li X, Ou Y, Cui B, An L, Zhang Y, Gong J. The combined signatures of telomere and immune cell landscape provide a prognostic and therapeutic biomarker in glioma. Front Immunol 2023; 14:1220100. [PMID: 37662954 PMCID: PMC10470026 DOI: 10.3389/fimmu.2023.1220100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/20/2023] [Indexed: 09/05/2023] Open
Abstract
Background Gliomas, the most prevalent primary malignant tumors of the central nervous system in adults, exhibit slow growth in lower-grade gliomas (LGG). However, the majority of LGG cases progress to high-grade gliomas, posing challenges for prognostication. The tumor microenvironment (TME), characterized by telomere-related genes and immune cell infiltration, strongly influences glioma growth and therapeutic response. Therefore, our objective was to develop a Telomere-TME (TM-TME) classifier that integrates telomere-related genes and immune cell landscape to assess prognosis and therapeutic response in glioma. Methods This study encompassed LGG patients from the TCGA and CCGA databases. TM score and TME score were derived from the expression signatures of telomere-related genes and the presence of immune cells in LGG, respectively. The TM-TME classifier was established by combining TM and TME scores to effectively predict prognosis. Subsequently, we conducted Kaplan-Meier survival estimation, univariate Cox regression analysis, and receiver operating characteristic curves to validate the prognostic prediction capacity of the TM-TME classifier across multiple cohorts. Gene Ontology (GO) analysis, biological processes, and proteomaps were performed to annotate the functional aspects of each subgroup and visualize the cellular signaling pathways. Results The TM_low+TME_high subgroup exhibited superior prognosis and therapeutic response compared to other subgroups (P<0.001). This finding could be attributed to distinct tumor somatic mutations and cancer cellular signaling pathways. GO analysis indicated that the TM_low+TME_high subgroup is associated with the neuronal system and modulation of chemical synaptic transmission. Conversely, the TM_high+TME_low subgroup showed a strong association with cell cycle and DNA metabolic processes. Furthermore, the classifier significantly differentiated overall survival in the TCGA LGG cohort and served as an independent prognostic factor for LGG patients in both the TCGA cohort (P<0.001) and the CGGA cohort (P<0.001). Conclusion Overall, our findings underscore the significance of the TM-TME classifier in predicting prognosis and immune therapeutic response in glioma, shedding light on the complex immune landscape within each subgroup. Additionally, our results suggest the potential of integrating risk stratification with precision therapy for LGG.
Collapse
Affiliation(s)
- Xu Han
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zihan Yan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kaiyu Fan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xueyi Guan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bohan Hu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiang Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yunwei Ou
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bing Cui
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Lingxuan An
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Yaohua Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Jian Gong
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| |
Collapse
|
47
|
Xu W, Jiang T, Shen K, Zhao D, Zhang M, Zhu W, Liu Y, Xu C. GADD45B regulates the carcinogenesis process of chronic atrophic gastritis and the metabolic pathways of gastric cancer. Front Endocrinol (Lausanne) 2023; 14:1224832. [PMID: 37608794 PMCID: PMC10441793 DOI: 10.3389/fendo.2023.1224832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/18/2023] [Indexed: 08/24/2023] Open
Abstract
Background Gastric cancer continues to be a significant global healthcare challenge, and its burden remains substantial. The development of gastric cancer (GC) is closely linked to chronic atrophic gastritis (CAG), yet there is a scarcity of research exploring the underlying mechanisms of CAG-induced carcinogenesis. Methods In this study, we conducted a comprehensive investigation into the oncogenes involved in CAG using both bulk transcriptome and single-cell transcriptome data. Our approach employed hdWGCNA to identify pathogenic genes specific to CAG, with non-atrophic gastritis (NAG) serving as the control group. Additionally, we compared CAG with GC, using normal gastric tissue as the control group in the single-cell transcriptome analysis. By intersecting the identified pathogenic genes, we pinpointed key network molecules through protein interaction network analysis. To further refine the gene selection, we applied LASSO, SVM-RFE, and RF techniques, which resulted in a set of cancer-related genes (CRGs) associated with CAG. To identify CRGs potentially linked to gastric cancer progression, we performed a univariate COX regression analysis on the gene set. Subsequently, we explored the relationship between CRGs and immune infiltration, drug sensitivity, and clinical characteristics in gastric cancer patients. We employed GSVA to investigate how CRGs regulated signaling pathways in gastric cancer cells, while an analysis of cell communication shed light on the impact of CRGs on signal transmission within the gastric cancer tumor microenvironment. Lastly, we analyzed changes in metabolic pathways throughout the progression of gastric cancer. Results Using hdWGCNA, we have identified a total of 143 pathogenic genes that were shared by CAG and GC. To further investigate the underlying mechanisms, we conducted protein interaction network analysis and employed machine learning screening techniques. As a result, we have identified 15 oncogenes that are specifically associated with chronic atrophic gastritis. By performing ROC reanalysis and prognostic analysis, we have determined that GADD45B is the most significant gene involved in the carcinogenesis of CAG. Immunohistochemical staining and differential analysis have revealed that GADD45B expression was low in GC tissues while high in normal gastric tissues. Moreover, based on prognostic analysis, high expression of GADD45B has been correlated with poor prognosis in GC patients. Additionally, an analysis of immune infiltration has shown a relationship between GADD45B and the infiltration of various immune cells. By correlating GADD45B with clinical characteristics, we have found that it primarily affects the depth of invasion in GC. Through cell communication analysis, we have discovered that the CD99 signaling pathway network and the CDH signaling pathway network are the main communication pathways that significantly alter the microenvironment of gastric tissue during the development of chronic atrophic gastritis. Specifically, GADD45B-low GC cells were predominantly involved in the network communication of the CDH signaling pathway, while GADD45B-high GC cells played a crucial role in both signaling pathways. Furthermore, we have identified several metabolic pathways, including D-Glutamine and D-glutamate metabolism and N-Glycan biosynthesis, among others, that played important roles in the occurrence and progression of GC, in addition to the six other metabolic pathways. In summary, our study highlighted the discovery of 143 pathogenic genes shared by CAG and GC, with a specific focus on 15 oncogenes associated with CAG. We have identified GADD45B as the most important gene in the carcinogenesis of CAG, which exhibited differential expression in GC tissues compared to normal gastric tissues. Moreover, GADD45B expression was correlated with patient prognosis and is associated with immune cell infiltration. Our findings also emphasized the impact of the CD99 and CDH signaling pathway networks on the microenvironment of gastric tissue during the development of CAG. Additionally, we have identified key metabolic pathways involved in GC progression. Conclusion GADD45B, an oncogene implicated in chronic atrophic gastritis, played a critical role in GC development. Decreased expression of GADD45B was associated with the onset of GC. Moreover, GADD45B expression levels were closely tied to poor prognosis in GC patients, influencing the infiltration patterns of various cells within the tumor microenvironment, as well as impacting the metabolic pathways involved in GC progression.
Collapse
Affiliation(s)
- Wei Xu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Tianxiao Jiang
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Kanger Shen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Dongxu Zhao
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Man Zhang
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wenxin Zhu
- Department of Gastroenterology, Kunshan Third People’s Hospital, Suzhou, Jiangsu, China
| | - Yunfei Liu
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Chunfang Xu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
48
|
Xia Z, Chen S, He M, Li B, Deng Y, Yi L, Li X. Editorial: Targeting metabolism to activate T cells and enhance the efficacy of checkpoint blockade immunotherapy in solid tumors. Front Immunol 2023; 14:1247178. [PMID: 37575246 PMCID: PMC10415066 DOI: 10.3389/fimmu.2023.1247178] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 07/20/2023] [Indexed: 08/15/2023] Open
Affiliation(s)
- Zhijia Xia
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Shi Chen
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Miao He
- Laboratory Animal Center, Chongqing Medical University, Chongqing, China
| | - Benhua Li
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yayuan Deng
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin Yi
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaosong Li
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, China
| |
Collapse
|
49
|
Liu T, Li C, Zhang J, Hu H, Li C. Unveiling efferocytosis-related signatures through the integration of single-cell analysis and machine learning: a predictive framework for prognosis and immunotherapy response in hepatocellular carcinoma. Front Immunol 2023; 14:1237350. [PMID: 37575252 PMCID: PMC10414188 DOI: 10.3389/fimmu.2023.1237350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) represents a prominent gastrointestinal malignancy with a grim clinical outlook. In this regard, the discovery of novel early biomarkers holds substantial promise for ameliorating HCC-associated mortality. Efferocytosis, a vital immunological process, assumes a central position in the elimination of apoptotic cells. However, comprehensive investigations exploring the role of efferocytosis-related genes (EFRGs) in HCC are sparse, and their regulatory influence on HCC immunotherapy and targeted drug interventions remain poorly understood. Methods RNA sequencing data and clinical characteristics of HCC patients were acquired from the TCGA database. To identify prognostically significant genes in HCC, we performed the limma package and conducted univariate Cox regression analysis. Subsequently, machine learning algorithms were employed to identify hub genes. To assess the immunological landscape of different HCC subtypes, we employed the CIBERSORT algorithm. Furthermore, single-cell RNA sequencing (scRNA-seq) was utilized to investigate the expression levels of ERFGs in immune cells and to explore intercellular communication within HCC tissues. The migratory capacity of HCC cells was evaluated using CCK-8 assays, while drug sensitivity prediction reliability was determined through wound-healing assays. Results We have successfully identified a set of nine genes, termed EFRGs, that hold significant potential for the establishment of a hepatocellular carcinoma-specific prognostic model. Furthermore, leveraging the individual risk scores derived from this model, we were able to stratify patients into two distinct risk groups, unveiling notable disparities in terms of immune infiltration patterns and response to immunotherapy. Notably, the model's capacity to accurately predict drug responses was substantiated through comprehensive experimental investigations, encompassing wound-healing assay, and CCK8 experiments conducted on the HepG2 and Huh7 cell lines. Conclusions We constructed an EFRGs model that serves as valuable tools for prognostic assessment and decision-making support in the context of immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Tao Liu
- Colorectal and Anal Surgery Department, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Chao Li
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians University, Munich, Germany
| | - Jiantao Zhang
- Colorectal and Anal Surgery Department, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Han Hu
- Colorectal and Anal Surgery Department, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Chenyao Li
- Colorectal and Anal Surgery Department, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|