1
|
Zhang S, Miao L, Tian X, Yang B, Luo B. Opportunities and challenges of immuno-oncology: A bibliometric analysis from 2014 to 2023. Hum Vaccin Immunother 2025; 21:2440203. [PMID: 39885669 PMCID: PMC11792843 DOI: 10.1080/21645515.2024.2440203] [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: 09/11/2024] [Revised: 11/22/2024] [Accepted: 12/06/2024] [Indexed: 02/01/2025] Open
Abstract
The emergence of immuno-oncology (IO) has led to revolutionary changes in the field of cancer treatment. Despite notable advancements in this field, a thorough exploration of its full depth and extent has yet to be performed. This study provides a comprehensive overview of publications pertaining to IO. Publications on IO from 2014 to 2023 were retrieved by searching the Web of Science Core Collection database (WoSCC). VOSviewer software and Citespace software were used for the visualized analysis. A total of 1,874 articles have been published in the IO domain. The number of publications and citations has been increasing annually. This study also examines the primary research directions within the field of IO. In conclusion, this study offers a comprehensive overview of the opportunities and challenges associated with IO, illuminating the current status of research and indicating potential future trajectories in this rapidly progressing field. This study provides a comprehensive survey of the current research status and hot spots within the field of IO. It will assist researchers in comprehending the current research emphasis and development trends in this field and offers guidance for future research directions.
Collapse
Affiliation(s)
- Siqi Zhang
- School of Clinical Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
- Department of Oncology, Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, China
- Department of Oncology, Hubei Province Academy of Traditional Chinese Medicine, Wuhan, China
| | - Lina Miao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoxia Tian
- School of Clinical Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Bingxu Yang
- School of Clinical Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Baoping Luo
- School of Clinical Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
- Department of Oncology, Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, China
- Department of Oncology, Hubei Province Academy of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
2
|
Liu Y, Dong K, Yao Y, Lu B, Wang L, Ji G, Zhang H, Zhao Z, Yang X, Huang R, Zhou W, Pan X, Cui X. Construction and validation of renal cell carcinoma tumor cell differentiation-related prognostic classification (RCC-TCDC): an integrated bioinformatic analysis and clinical study. Ann Med 2025; 57:2490830. [PMID: 40248945 PMCID: PMC12010653 DOI: 10.1080/07853890.2025.2490830] [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: 12/16/2024] [Revised: 02/28/2025] [Accepted: 03/08/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Renal cell carcinoma (RCC) is a heterogeneous malignancy with diverse gene expression patterns, molecular landscapes, and differentiation characteristics of tumor cells. It is imperative to develop molecular RCC classification based on tumor cell differentiation for precise risk stratification and personalized therapy. METHODS We obtained scRNA-seq profiles from GSE159115 and bulk RNA-seq profiles from TCGA-KIRC cohort. We then performed scRNA-seq cluster analysis, monocle2 pseudotime analysis, and prognostic analysis to obtain tumor cell differentiation-related prognostic genes (TCDGs). Subsequently, we conducted consensus clustering to construct the RCC tumor cell differentiation-related prognostic classification (RCC-TCDC) and implemented prognostic and multi-omics analyses. Moreover, we utilized Lasso regression to help develop a multivariable prognostic model. In addition, we performed correlation analysis and Cmap algorithm for regulatory network establishment and candidate inhibitor prediction. We eventually included 370 kidney neoplasm patients in Xinhua cohort to undergo immunohistochemical staining and scoring for classification and comprehensive statistical analyses, including Chi-square tests, Kaplan-Meier survival analyses, and multivariable Cox regression analysis . RESULTS 32 TCDGs were identifiedand RCC-TCDC was constructed to classify TCGA-KIRC patients into RCC-low differentiation (RCC-LD) (S100A11+ SH3BGRL3+, high risk), RCC-moderate differentiation (TSPAN7+, medium risk), and RCC-high differentiation (RCC-HD) (AQP1+ NPR3+, low risk). Notably, RCC-LD was validated as anindependent risk factor for both OS (p = 0.015, HR = 14.0, 95%CI = 1.67-117.8) and PFS (p = 0.010, HR = 4.0, 95%CI = 1.39-11.7) of RCC patients in Xinhua cohort, taking RCC-HD as reference. CONCLUSIONS We constructed and validated a robust molecular classification system, RCC-TCDC, elucidating three distinct RCC subtypes.
Collapse
Affiliation(s)
- Yifan Liu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Keqin Dong
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuntao Yao
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bingnan Lu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Wang
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guo Ji
- Department of Pathology, Shanghai Tenth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Haoyu Zhang
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zihui Zhao
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyue Yang
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Runzhi Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Wang Zhou
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiuwu Pan
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingang Cui
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Chen Y, Zhou C, Zhang X, Chen M, Wang M, Zhang L, Chen Y, Huang L, Sun J, Wang D, Chen Y. Construction of a novel radioresistance-related signature for prediction of prognosis, immune microenvironment and anti-tumour drug sensitivity in non-small cell lung cancer. Ann Med 2025; 57:2447930. [PMID: 39797413 PMCID: PMC11727174 DOI: 10.1080/07853890.2024.2447930] [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: 06/30/2024] [Revised: 11/26/2024] [Accepted: 12/12/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is a fatal disease, and radioresistance is an important factor leading to treatment failure and disease progression. The objective of this research was to detect radioresistance-related genes (RRRGs) with prognostic value in NSCLC. METHODS The weighted gene coexpression network analysis (WGCNA) and differentially expressed genes (DEGs) analysis were performed to identify RRRGs using expression profiles from TCGA and GEO databases. The least absolute shrinkage and selection operator (LASSO) regression and random survival forest (RSF) were used to screen for prognostically relevant RRRGs. Multivariate Cox regression was used to construct a risk score model. Then, Immune landscape and drug sensitivity were evaluated. The biological functions exerted by the key gene LBH were verified by in vitro experiments. RESULTS Ninety-nine RRRGs were screened by intersecting the results of DEGs and WGCNA, then 11 hub RRRGs associated with survival were identified using machine learning algorithms (LASSO and RSF). Subsequently, an eight-gene (APOBEC3B, DOCK4, IER5L, LBH, LY6K, RERG, RMDN2 and TSPAN2) risk score model was established and demonstrated to be an independent prognostic factor in NSCLC on the basis of Cox regression analysis. The immune landscape and sensitivity to anti-tumour drugs showed significant disparities between patients categorized into different risk score subgroups. In vitro experiments indicated that overexpression of LBH enhanced the radiosensitivity of A549 cells, and knockdown LBH reversed the cytotoxicity induced by X-rays. CONCLUSION Our study developed an eight-gene risk score model with potential clinical value that can be adopted for choice of drug treatment and prognostic prediction. Its clinical routine use may assist clinicians in selecting more rational practices for individuals, which is important for improving the prognosis of NSCLC patients. These findings also provide references for the development of potential therapeutic targets.
Collapse
Affiliation(s)
- Yanliang Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
| | - Chan Zhou
- Department of Geriatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xiaoqiao Zhang
- Department of Geriatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Min Chen
- Department of Geriatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Meifang Wang
- Department of Pulmonary and Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Lisha Zhang
- Department of Obstetrics, Tangshan Caofeidian District Hospital, Tangshan, Hebei, China
| | - Yanhui Chen
- Department of Neuroscience and Endocrinology, Tangshan Caofeidian District Hospital, Tangshan, Hebei, China
| | - Litao Huang
- Department of Clinical Research Management, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Junjun Sun
- Department of Emergency Surgery, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei, , China
| | - Dandan Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
| | - Yong Chen
- Department of Radio-Chemotherapy, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
4
|
Xu W, Li W, Kuai D, Li Y, Sun W, Liu X, Xu B. Identification of endoplasmic reticulum stress-related genes as prognostic markers in colon cancer. Cancer Biol Ther 2025; 26:2458820. [PMID: 40169935 PMCID: PMC11970746 DOI: 10.1080/15384047.2025.2458820] [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: 03/26/2024] [Revised: 12/20/2024] [Accepted: 01/22/2025] [Indexed: 04/03/2025] Open
Abstract
Endoplasmic reticulum stress (ERS) has been implicated in the pathogenesis of various cancers, including colon cancer, by regulating tumor cell survival, growth, and immune response. However, the specific genes involved in ERS that could serve as prognostic markers in colon cancer remain underexplored. This study aims to identify and validate endoplasmic reticulum stress related genes (ERSRGs) in colon cancer that correlate with patient prognosis, thereby enhancing the understanding of ERS in oncological outcomes and potential therapeutic targeting. We utilized bioinformatics analyses to identify ERSRGs from publicly available colon cancer datasets. Differential expression analysis and survival analysis were performed to assess the prognostic significance of these genes. Validation was conducted through quantitative real-time PCR (RT-qPCR) on selected colon cancer cell lines. Our study identified nine ERS related genes (ASNS, ATF4, ATF6B, BOK, CLU, DDIT3, MANF, SLC39A14, TRAF2) involved in critical pathways including IL-12, PI3K-AKT, IL-7, and IL-23 signaling, and linked to 1-, 3-, and 5-year survival of patients with colon cancer. A multivariate Cox model based on these ERS related genes demonstrated significant prognostic power. Further, TRAF2 strong correlated with immune cells infiltration, suggesting its potential roles in modulating immune responses in the tumor microenvironment. The RT-qPCR validation confirmed the differential expression of these genes in human colon cancer cell lines versus human normal colonic epithelial cell line. The identified ERSRGs could serve as valuable prognostic markers and may offer new insights into the therapeutic targeting of ERS in colon cancer.
Collapse
Affiliation(s)
- Wenjing Xu
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Wei Li
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Dayu Kuai
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yaqiang Li
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Wei Sun
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Xian Liu
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Baohong Xu
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Zhou M, Zhao W, Zhang X, Cheng Y, Wang M, Chen Y, Zhao L. Nicotinamide metabolism affects the prognosis of hepatocellular carcinoma by influencing the tumor microenvironment. Cytokine 2025; 191:156939. [PMID: 40228405 DOI: 10.1016/j.cyto.2025.156939] [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: 01/24/2025] [Revised: 03/15/2025] [Accepted: 04/05/2025] [Indexed: 04/16/2025]
Abstract
In this study, we utilized the public database along with single-cell genomics techniques to systematically analyze the expression patterns and clinical significance of key genes in the nicotinamide metabolism pathway in liver cancer samples. The findings indicate that differential nicotinamide metabolism-related key genes are expressed in liver cancer samples. The liver cancer samples were put into separate subgroups using consistency clustering analysis based on differential gene expression levels observed. Additionally, immune infiltration and drug sensitivity analysis also revealed differences between the two subgroups. Survival analysis suggested that the key genes were associated with prognosis. Finally, a prognostic model was established using the key genes, offering a fresh viewpoint on the molecular mechanism investigating liver cancer. This study demonstrated the significant correlation between key genes in the nicotinamide metabolism pathway and the occurrence and progression of liver cancer and indicated that these key genes could serve as prognostic markers and tailored treatment targets for liver cancer.
Collapse
Affiliation(s)
- Min Zhou
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210000, China
| | - Wenhui Zhao
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210000, China
| | - Xiaobo Zhang
- School of Life Sciences, Westlake University, Hangzhou, 310024, China
| | - Ye Cheng
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210000, China
| | - Mengxiang Wang
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210000, China
| | - Yan Chen
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210000, China.
| | - Lingrui Zhao
- School of Life Sciences, Westlake University, Hangzhou, 310024, China.
| |
Collapse
|
6
|
Dai S, Li B, Wu Q, Han S, Zhao Q, Wang Y, Zhang Y, Gao Y. Pan-cancer analysis reveals BAF complexes as immune-related biomarkers and validation in triple-negative breast cancer. Life Sci 2025; 372:123607. [PMID: 40194763 DOI: 10.1016/j.lfs.2025.123607] [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: 01/10/2025] [Revised: 03/11/2025] [Accepted: 04/01/2025] [Indexed: 04/09/2025]
Abstract
AIMS BAF complexes (BAFs), ATP-dependent regulators of chromatin structure, play a significant role in cancer progression. This pan-cancer study aimed to decode the potential of specific BAFs in the pathology, immunity, and therapy of targeted cancers. MATERIALS AND METHODS Data were retrieved from The Cancer Genome Atlas, Gene Expression Omnibus, and IMvigor210 databases and were analyzed for expression patterns, prognostic value, mutational signatures, biological pathways, tumor immune microenvironment (TIME) remodeling, and therapeutic resistance of BAFs. Experimental validation was also conducted. KEY FINDINGS BAFs exhibit abnormal expression in various human cancers. The BAFs model and nomogram (based on multiple variables) were developed as prognostic tools. BAFs regulate the TIME and influence the response to anti-PD-L1 therapy, particularly through ACTL6A, as observed in RNA sequencing and single-cell RNA sequencing datasets (high-resolution gene expression data at the single-cell level). ACTLA6 is a major adverse gene in the prognostic model. Patients with high ACTL6A expression showed significantly worse overall survival (hazard ratio = 1.32, 95 % CI: 1.26-1.39, p < 0.001). ACTL6A expression escalates with breast cancer (BRCA) malignancy, particularly in triple-negative BRCA (TNBC), and correlates with immune checkpoint expression while playing a crucial role in promoting cancer metastasis in TNBC. SIGNIFICANCE Our findings first emphasize the significance of a novel BAFs model for patient prognosis and corroborate the considerable role of BAFs as immune-related biomarkers in pan-cancer progression. ACTL6A has a dual role as an immune-related biomarker and potential therapeutic target in TNBC, deepening our comprehension of its function as an oncogene.
Collapse
Affiliation(s)
- Shuying Dai
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Department of Geriatrics, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Zhejiang 310006, China
| | - Bei Li
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Department of Geriatrics, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Zhejiang 310006, China
| | - Qingqian Wu
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Department of Geriatrics, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Zhejiang 310006, China
| | - Shuang Han
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Department of Geriatrics, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Zhejiang 310006, China; School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Qingwen Zhao
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Zhejiang 310006, China.
| | - Yule Wang
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Department of Geriatrics, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Zhejiang 310006, China
| | - Yingjuan Zhang
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Department of Geriatrics, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Zhejiang 310006, China
| | - Yue Gao
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Department of Geriatrics, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Zhejiang 310006, China.
| |
Collapse
|
7
|
Zou W, Zhang Z, Cao T, Li M. Mesenchymal stem cell transplantation ameliorates inflammation in spinal cord injury by inhibiting lactylation-related genes. Cytokine 2025; 191:156960. [PMID: 40345018 DOI: 10.1016/j.cyto.2025.156960] [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: 03/29/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND The immune microenvironment significantly influences neural regeneration in spinal cord injury (SCI). Lactate activates central nervous system (CNS) glial cells, prompting the secretion of proinflammatory cytokines and triggering an inflammatory response. Mesenchymal stem cells (MSCs) make a promising future for SCI therapy due to their immune regulation and anti-inflammatory properties. However, it is unclear whether MSCs inhibit inflammatory responses in the SCI microenvironment through lactylation regulation. This study aimed to identify lactylation-related genes (LRGs) in SCI and investigate their role in immune cell infiltration and MSC-mediated inflammation reduction. METHODS Transcription datasets of SCI patients were acquired from Gene Expression Omnibus (GEO). Differentially expressed genes (DEGs) underwent functional enrichment analysis, and CIBERSORT assessed immune cell infiltration in SCI. Crucial lactylation-related differentially expressed genes (LRDEGs) associated with SCI were identified via machine learning. The association between LRDEGs and inflammatory response in SCI mediated by immune cell infiltration was confirmed using Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG). Rats with subacute thoracic SCI were transplanted with hUC-MSCs, and transcriptome analyses were conducted on their spinal cords and retrieved hUC-MSCs, respectively. RESULTS The study identified 808 DEGs and 13 differentially infiltrated immune cell types in SCI patients compared to healthy controls. Multiple inflammatory response-related signaling pathways were activated in SCI. Seven LRDEGs, including LSP1, XRCC4, HSDL2, HNRNPH1, RPL14, IKZF1, and TP53, were recognized as key regulators. These genes are linked to immune cell infiltration and inflammatory responses in SCI. In SCI rats, the increased expression of LRDEGs and inflammatory cytokines were observed, which were significantly reduced after hUC-MSC transplantation. Differences in LRDEG expression patterns, enriched functions, and pathways between two SCI subtypes were statistically significant. CONCLUSIONS LRDEGs are involved in immune cell-mediated inflammatory response in SCI, and hUC-MSC transplantation reduces LRDEGs expression and inflammation response in the SCI microenvironment.
Collapse
Affiliation(s)
- Weiwei Zou
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zelin Zhang
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Tingting Cao
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Mangmang Li
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
8
|
Chen C, Zou P, Wu X. Development and Validation of an Immune Prognostic Index Related to Infiltration of CD4+ and CD8+ T Cells in Colorectal Cancer. Mol Biotechnol 2025; 67:2758-2773. [PMID: 39026041 DOI: 10.1007/s12033-024-01237-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: 04/05/2024] [Accepted: 06/25/2024] [Indexed: 07/20/2024]
Abstract
Colorectal cancer (CRC) is a highly prevalent cancer worldwide, but treatment outcomes can vary significantly among patients with similar clinical or historical stages. This study aimed to investigate the differences in immune cell abundance associated with malignant progression in CRC patients. We utilized data from patients with CRC obtained from The Cancer Genome Atlas as our training set. To assess immune cell infiltration levels, an immune cell risk score (ICRS) was calculated. Furthermore, we performed network analysis to identify effective T cell-related genes (ETRGs) and subsequently constructed an effective T cell prognostic index (ETPI). The performance of the ETPI was evaluated through external validation using four Gene Expression Omnibus datasets. Additionally, a nomogram analysis and drug sensitivity analysis were conducted to explore the clinical utility of the ETRGs. We also examined the expression of ETRGs in clinical samples. Based on the ICRS, we identified activated CD4+ and CD8+ T cells as protective factors in terms of prognosis. Six ETRGs were identified to develop the ETPI, which exhibited remarkable prognostic performance. In the external validation of immunotherapy, the low ETPI group demonstrated a significantly lower recurrence rate. To optimize therapeutic strategies, we developed a nomogram. Notably, patients with different ETPI values exhibited varying responses to tumor pathway inhibitors. Finally, we observed higher protein expression of certain ETRGs in normal tissues compared to tumors. Our findings suggest that the ETPI may contribute to the precise selection of patients based on tumor microenvironment and key genomic landscape interactions, thereby optimizing drug benefits and informing clinical strategies in future.
Collapse
Affiliation(s)
- Chengru Chen
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, 518033, Guangdong Province, China
| | - Peng Zou
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, 518033, Guangdong Province, China
| | - Xiaobin Wu
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, 518033, Guangdong Province, China.
| |
Collapse
|
9
|
Li G, Zhou Q, Xie M, Zhao B, Zhang K, Luo Y, Kong L, Gao D, Guo Y. Identification of ageing-associated gene signatures in heart failure with preserved ejection fraction by integrated bioinformatics analysis and machine learning. Genes Dis 2025; 12:101478. [PMID: 40330147 PMCID: PMC12053710 DOI: 10.1016/j.gendis.2024.101478] [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: 03/09/2024] [Revised: 10/10/2024] [Accepted: 11/21/2024] [Indexed: 05/08/2025] Open
Abstract
The incidence of heart failure with preserved ejection fraction (HFpEF) increases with the ageing of populations. This study aimed to explore ageing-associated gene signatures in HFpEF to develop new diagnostic biomarkers and provide new insights into the underlying mechanisms of HFpEF. Mice were subjected to a high-fat diet combined with L-NG-nitroarginine methyl ester (l-NAME) to induce HFpEF, and next-generation sequencing was performed with HFpEF hearts. Additionally, separate datasets were acquired from the Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) were used to identify ageing-related DEGs. Support vector machine, random forest, and least absolute shrinkage and selection operator algorithms were employed to identify potential diagnostic genes from ageing-related DEGs. The diagnostic value was assessed using a nomogram and receiver operating characteristic curve. The gene and related protein expression were verified by reverse transcription PCR and western blotting. The immune cell infiltration in hearts was analysed using the single-sample gene-set enrichment analysis algorithm. The results showed that the merged HFpEF datasets comprised 103 genes, of which 15 ageing-related DEGs were further screened in. The ageing-related DEGs were primarily associated with immune and metabolism regulation. AGTR1a, NR3C1, and PRKAB1 were selected for nomogram construction and machine learning-based diagnostic value, displaying strong diagnostic potential. Additionally, ageing scores were established based on nine key DEGs, revealing noteworthy differences in immune cell infiltration across HFpEF subtypes. In summary, those results highlight the significance of immune dysfunction in HFpEF. Furthermore, ageing-related DEGs might serve as promising prognostic and predictive biomarkers for HFpEF.
Collapse
Affiliation(s)
- Guoxing Li
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Cardiovascular Disease Laboratory of Chongqing Medical University, Chongqing 400016, China
| | - Qingju Zhou
- Department of Health Management Center, Chongqing General Hospital, Chongqing University, Chongqing 400010, China
| | - Ming Xie
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing 400010, China
| | - Boying Zhao
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing 400010, China
| | - Keyu Zhang
- Cardiovascular Disease Laboratory of Chongqing Medical University, Chongqing 400016, China
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yuan Luo
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing 400010, China
| | - Lingwen Kong
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing 400010, China
| | - Diansa Gao
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Cardiovascular Disease Laboratory of Chongqing Medical University, Chongqing 400016, China
| | - Yongzheng Guo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Cardiovascular Disease Laboratory of Chongqing Medical University, Chongqing 400016, China
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing 400010, China
| |
Collapse
|
10
|
Huang J, Yu H, Yuan X, Zhong Y, Li X, Chen Y. TCN1 as an inflammatory regulator in psoriasis: Activation of the NF-κB pathway and potential therapeutic target. Int Immunopharmacol 2025; 157:114784. [PMID: 40318273 DOI: 10.1016/j.intimp.2025.114784] [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: 12/11/2024] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
OBJECTIVE This study investigates how TCN1 regulates inflammation and the cell cycle in psoriasis, focusing on the NF-κB pathway through in vitro experiments and bioinformatics analyses. METHODS DEGs were identified by analyzing transcriptome data from four datasets comparing psoriatic lesions and normal skin (GSE34248, GSE30999, GSE14905, and GSE13355). Validation of TCN1 expression following biologic treatment was conducted using GSE201827, GSE51440, and GSE117239. GO and GSEA were performed to explore biological pathways. The expression levels of TCN1 in psoriatic lesions and healthy skin were assessed by qPCR and immunohistochemistry (IHC). In vitro, HaCaT keratinocytes were stimulated with TNF-α and IL-17 A, and TCN1 expression was modulated through siRNA-mediated knockdown and plasmid-mediated overexpression. Subsequent changes in TCN1 and key inflammatory cytokines were evaluated by qPCR and Western blotting (WB). Furthermore, immunofluorescence assays were performed to visualize the subcellular localization of TCN1 and the nuclear translocation of phosphorylated p65 (p-p65) in HaCaT cells. Cell cycle progression was assessed using BrdU-PI flow cytometry. RESULTS TCN1 was upregulated in psoriatic lesions, and its expression levels were positively correlated with the PASI score. Following biologic treatment, TCN1 expression was reduced. TCN1 overexpression was associated with activation of the NF-κB signaling pathway, accompanied by increased synthesis of psoriasis-related inflammatory mediators, as well as an elevated proportion of cells in the S phase of the cell cycle. CONCLUSIONS TCN1 is essential in modulating inflammation and the cell cycle in psoriasis, implying its value as both a biomarker for diagnosis and a candidate for therapeutic intervention.
Collapse
Affiliation(s)
- Jian Huang
- Department of Dermatology, Guangdong College of Clinical Dermatology, Anhui Medical University, Hefei, Anhui Province, People's Republic of China; The Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui Province, People's Republic of China; Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Huanhuan Yu
- Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Xiuqing Yuan
- Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China; Shenzhen Children's Hospital, Shenzhen, Guangdong Province, People's Republic of China
| | - Yuanqiu Zhong
- Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Xinhui Li
- Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Yongfeng Chen
- Department of Dermatology, Guangdong College of Clinical Dermatology, Anhui Medical University, Hefei, Anhui Province, People's Republic of China; The Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui Province, People's Republic of China; Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.
| |
Collapse
|
11
|
Sriramadasu K, Ravichandran S, Li YH, Lai MT, Chiang AJ, Li CJ, Tsui KH, Chen CM, Chuang HH, Hwang T, Ding WY, Chung C, Chang CYY, Sheu JJC. Molecular evolution of driver mutations in cancer with microsatellite instability and their impact on tumor progression: Implications for precision medicine in patients with UCEC. Comput Biol Med 2025; 192:110275. [PMID: 40311467 DOI: 10.1016/j.compbiomed.2025.110275] [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: 11/11/2024] [Revised: 04/07/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025]
Abstract
Cancer development is driven by genetic alterations, particularly cancer driver mutations (CDMs), which are associated with aggressive phenotypes and shorter survival. In contrast, higher mutation loads caused by microsatellite instability (MSI) or mismatch repair deficiency (MMRd) can induce anti-cancer immunity, leading to tumor shrinkage and improved responses to immune checkpoint inhibitor (ICI) therapies. However, understanding how CDMs and MSI/MMRd influence cancer evolution remains limited. We opted uterine corpus endometrial carcinoma (UCEC) as a model in this study due to its MSI-high/MMRd characteristics. Somatic mutation screening revealed that UCEC has a significantly higher mutation rate in cancer driver genes compared to ovarian cancer (OVCA) and cervical squamous cell carcinoma (CSCC), despite these cancers arising from histologically connected organs in the reproductive tract. Interestingly, these CDMs did not necessarily drive tumor progression. Using a cutoff of 7.0 (mutations/Mb) for tumor mutation burden (TMB), we classified UCEC patients into two groups with distinct clinical features, genetic profiles, and drug sensitivities. Among the known CDMs, TP53 mutations and their functional networks emerged as key drivers in UCEC progression, while mutations in CTNNB1, PTEN, and ARID1A may enhance anti-tumor immunity, correlating with longer overall survivals. Drug screening using GDSC and CTRPv2 databases suggested that GSK-3 inhibitor IX may be effective for treating aggressive UCEC patients with a non-MSI phenotype. Curcumin showed efficacy for UCEC patients with MSI, especially with ICI therapy. Our study highlights the importance of immune regulation and tolerance over CDMs in cancer development, particularly in those with an MSI-high/MMRd phenotype. We propose that TMB could serve as a valuable screening method alongside molecular and histopathological classifications to guide treatment strategies for UCEC patients.
Collapse
Affiliation(s)
- Kalpana Sriramadasu
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804201, Taiwan
| | - Senthilkumar Ravichandran
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804201, Taiwan; Department of Dermatology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Yau-Hong Li
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804201, Taiwan; Department of Obstetrics and Gynecology, Pingtung Veterans General Hospital, Pingtung, 900053, Taiwan
| | - Ming-Tsung Lai
- Department of Pathology, Taichung Hospital, Ministry of Health and Welfare, Taichung, 403301, Taiwan
| | - An-Jen Chiang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804201, Taiwan; Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, 813414, Taiwan
| | - Chia-Jung Li
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, 813414, Taiwan; Institute of Biopharmaceutical Sciences, National Sun Yat-Sen University, Kaohsiung, 804201, Taiwan
| | - Kuan-Hao Tsui
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, 813414, Taiwan; Institute of Biopharmaceutical Sciences, National Sun Yat-Sen University, Kaohsiung, 804201, Taiwan
| | - Chih-Mei Chen
- Genetics Center, China Medical University Hospital, Taichung, 404332, Taiwan
| | - Hsiang-Hao Chuang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804201, Taiwan
| | - Tritium Hwang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804201, Taiwan
| | - Wendy Yarou Ding
- Genetics Center, China Medical University Hospital, Taichung, 404332, Taiwan
| | - Ching Chung
- Genetics Center, China Medical University Hospital, Taichung, 404332, Taiwan
| | - Cherry Yin-Yi Chang
- Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, 404332, Taiwan; Department of Medicine, School of Medicine, China Medical University Hospital, Taichung, 404333, Taiwan.
| | - Jim Jinn-Chyuan Sheu
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804201, Taiwan; Institute of Biopharmaceutical Sciences, National Sun Yat-Sen University, Kaohsiung, 804201, Taiwan; School of Chinese Medicine, China Medical University, Taichung, 404333, Taiwan; Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan.
| |
Collapse
|
12
|
Lyu G, Sun R, Liu X, Xu Z. A Novel Hypoxia-Featured Genes Prognostic Model for Identification of Hypoxia Subtypes in Diffuse Large B-Cell Lymphoma. Cell Biochem Biophys 2025; 83:2265-2279. [PMID: 39663278 DOI: 10.1007/s12013-024-01637-7] [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] [Accepted: 11/25/2024] [Indexed: 12/13/2024]
Abstract
Diffuse large B-cell lymphoma (DLBCL), known as the predominant type of aggressive B-cell lymphoma, is biologically and clinically heterogeneous. The prognosis of DLBCL is quite different among subtypes. Hypoxia is one of the key elements in tumor microenvironment, promoting tumor progression by means of various mechanisms, such as increased proliferation, altered metabolism, enhanced angiogenesis, and greater migratory capability, among others. The primary purpose of this research is to investigate the connection between hypoxia-featured genes (HFGs), prognosis in DLBCL, and their capacity association with the immune microenvironment. Various hypoxia-associated patterns for DLBCL patients from GEO and TCGA databases were identified by means of an unsupervised consensus clustering algorithm. CIBERSORT and IOBR package is used to identify different immune infiltration status. To develop a predictive model using hypoxia-related genes, we conducted univariate Cox regression, multivariate Cox regression, and LASSO regression assessment. Subsequently, we confirmed the predictive importance of these hypoxia-associated genes, highlighting hypoxia-associated characteristics, and explored the connection between the hypoxia model and the immune environment. Three hypoxia clusters were identified. We also observed that each pattern of hypoxia response was significantly related to different prognoses. It was found that the immune status among hypoxia clusters is different. After developing a prognostic risk model using 5 hypoxia-related genes, we discovered that the risk score is related to immune factors and how effective drugs are in treating DLBCL. In DLBCL patients, varying hypoxia patterns correlate with both prognostic outcomes and the immune microenvironment. Hypoxia-featured genes (HFGs) function as a standalone predictive element in these patients. It is also potentially a reliable indicator for predicting clinical responses to ICI therapy and traditional drugs.
Collapse
Affiliation(s)
- Geng Lyu
- Department of Laboratory Medicine, College of Health Science and Technology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ruixin Sun
- Department of Laboratory Medicine, College of Health Science and Technology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaxin Liu
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital affiliated to School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zizhen Xu
- Department of Laboratory Medicine, College of Health Science and Technology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
13
|
Shu F, Wang Y, Li L, Shi L, Zhang F, Ma Z, Mao D. Multi-omics integration and machine learning identify and validate neutrophil extracellular trap-associated gene signatures in chronic rhinosinusitis with nasal polyps. Clin Immunol 2025; 275:110473. [PMID: 40089249 DOI: 10.1016/j.clim.2025.110473] [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: 01/21/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
This study aimed to explore the molecular characteristics of neutrophil extracellular traps (NETs) in chronic rhinosinusitis with nasal polyps (CRSwNP). Differentially expressed gene analysis, weighted gene co-expression network analysis, and machine learning algorithms identified three core NETs-associated genes: CXCR4, CYBB, and PTAFR, which were significantly upregulated in CRSwNP patients. The diagnostic performance of these genes was evaluated using receiver operating characteristic (ROC) curves, and their clinical relevance was validated using multicenter data. Immune infiltration analysis showed strong correlations between these genes and neutrophil and immune cell infiltration. Single-cell RNA sequencing demonstrated that these genes were predominantly expressed in myeloid and immune cells and exhibited dynamic changes during disease progression. These genes may contribute to CRSwNP pathogenesis through IL-17 signaling and metabolism-related pathways. This study identifies novel biomarkers and therapeutic targets for precise diagnosis and personalized treatment of CRSwNP.
Collapse
Affiliation(s)
- Fu Shu
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400000, People's Republic of China
| | - Yaping Wang
- Department of Otorhinolaryngology, Yongchuan Chinese Medicine Hospital Affiliated to Chongqing Medical University, Chongqing 400000, People's Republic of China
| | - Linglong Li
- Department of Otorhinolaryngology, Yongchuan Chinese Medicine Hospital Affiliated to Chongqing Medical University, Chongqing 400000, People's Republic of China
| | - Lei Shi
- Department of Otorhinolaryngology, The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 110847, People's Republic of China
| | - Feng Zhang
- Department of Otorhinolaryngology, Yongchuan Chinese Medicine Hospital Affiliated to Chongqing Medical University, Chongqing 400000, People's Republic of China
| | - Zhixuan Ma
- Department of Otorhinolaryngology, The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 110847, People's Republic of China
| | - Dehong Mao
- Department of Otorhinolaryngology, Yongchuan Chinese Medicine Hospital Affiliated to Chongqing Medical University, Chongqing 400000, People's Republic of China.
| |
Collapse
|
14
|
Zhang B, Zhu S, Zheng D, Zhang X, Xie W, Zhou S, Zheng S, Wang Q, Lin Z, Zheng Z, Chen Z, Lan E, Cui L, Ying H, Zhang Y, Lin X, Zhuang Q, Qian H, Hu X, Zhuang Y, Zhang Q, Jin Z, Jiang S, Ma Y. Development of a cuproptosis-related prognostic signature to reveal heterogeneity of the immune microenvironment and drug sensitivity in acute lymphoblastic leukemia. Eur J Med Res 2025; 30:435. [PMID: 40450339 DOI: 10.1186/s40001-025-02572-w] [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: 01/08/2025] [Accepted: 04/09/2025] [Indexed: 06/03/2025] Open
Abstract
BACKGROUND Cuproptosis is a brand-new copper-dependent type of cell death that has been linked to various tumors. However, the relationship between cuproptosis and acute lymphoblastic leukemia (ALL) remains to be further elaborated. METHODS In ALL, 12 cuproptosis-related genes (CRGs) were analyzed at genetic and single-cell levels. Two molecular clusters were identified using "ConsensusClusterPlus". With the least absolute shrinkage and selection operator, a prognostic signature was built based on cuproptosis. The prognosis, clinical parameters, biological function, immune cell infiltration, therapy sensitivities, and transcription factor regulation of the clusters and risk subsets were further compared. Kaplan Meier curves, time-ROC curves, and nomogram were employed to evaluate the accuracy of the signature. Lastly, qRT-PCR was used to detect prognostic genes in cell lines and clinical samples. RESULTS CRGs exhibited extensive genetic variations and heterogeneous expression profiles in ALL. Single-cell analysis demonstrated that CRGs were strongly correlated with the biological characteristics of cancer cells. Two clusters and risk subgroups with distinct clinicopathological features, prognoses, biological functions, and drug sensitivities were identified. The cuproptosis signature was crucial in characterizing tumor immune landscape and cancer cell self-renewal ability. Furthermore, we explored that subtype A and high-scoring groups were more sensitive to immunotherapy. Multiple drugs with higher sensitivity among high-risk subgroups have been predicted. Nomograms demonstrated the clinical applicability of cuproptosis in risk assessment. The model was further validated in the verification cohort, our clinical specimens, and cell lines. CONCLUSIONS The cuproptosis-based model can characterize the tumor microenvironment, forecast survival results, and aid in improving risk assessment and personalized therapy options in ALL.
Collapse
Affiliation(s)
- Bingxin Zhang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Shuxia Zhu
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Dong Zheng
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xinyi Zhang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Wenxia Xie
- Department of Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Shujuan Zhou
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Sisi Zheng
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Quanqiang Wang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Zhili Lin
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Ziwei Zheng
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Zixing Chen
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Enqing Lan
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Luning Cui
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Hansen Ying
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yu Zhang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xuanru Lin
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Qiang Zhuang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Honglan Qian
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xudong Hu
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yan Zhuang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Qianying Zhang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Zhouxiang Jin
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Songfu Jiang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Yongyong Ma
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
- Zhejiang Engineering Research Center for Hospital Emergency and Process Digitization, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
15
|
Liu B, He S, Li C, Xiong Z, Li Z, Feng C, Wang H, Tu C, Li Z. Leveraging multiple cell-death patterns based on machine learning to decipher the prognosis, immune, and immune therapeutic response of soft tissue sarcoma. Discov Oncol 2025; 16:917. [PMID: 40413669 DOI: 10.1007/s12672-025-02587-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: 01/04/2025] [Accepted: 05/06/2025] [Indexed: 05/27/2025] Open
Abstract
Soft tissue sarcomas (STS) imposes a substantial healthcare burden on society. The progression of these tumors is significantly influenced by diverse modes of programmed cell death (PCD), which can serve as valuable indicators for assessing prognosis and immune therapeutic response in STS. Nonetheless, the precise role of multiple cell death patterns in STS is yet to be clarified. We employed 96 machine-learning algorithm combination frameworks to identify novel cell death-related signatures (CDSigs) with the highest mean c-index, indicating their excellence. The independence test and comparison with previously published models further confirmed the stability and quality of these signatures for survival prediction in STS. The nomogram, comprising the cell death score (CDS) and clinical features, exhibited excellent predictive performance. Additionally, the CDSigs revealed associations with immune checkpoint genes and the immune microenvironment in STS. Furthermore, the results demonstrated that patients with lower CDS had the potential for greater benefit from immune therapeutic responses compared to those with higher CDS. Moreover, STS patients with low-risk scores exhibited heightened sensitivity to doxorubicin, axitinib, cisplatin, and camptothecin. Finally, the RT-qPCR results underscored significant differences in expression levels of several CDSigs genes between STS and normal cells. Overall, we comprehensively analyzed the multiple PCD in STS and established a novel CDSig for STS patients. This novel CDSig holds great promise in deciphering the prognosis, immune, and immune therapeutic response of STS.
Collapse
Affiliation(s)
- Binfeng Liu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Shasha He
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Chenbei Li
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Zijian Xiong
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Zhaoqi Li
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Chengyao Feng
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Hua Wang
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
- Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
- Shenzhen Research Institute of Central South University, Guangdong, 518063, China.
- National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China.
| | - Zhihong Li
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
- Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
- Shenzhen Research Institute of Central South University, Guangdong, 518063, China.
- National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
16
|
Tumuluru S, Godfrey JK, Cooper A, Yu J, Chen X, MacNabb BW, Venkataraman G, Zha Y, Pelzer B, Song J, Duns G, Sworder BJ, Raj S, Bolen C, Penuel E, Postovalova E, Kotlov N, Bagaev A, Fowler N, Shouval R, Smith SM, Alizadeh AA, Steidl C, Kline J. Integrative genomic analysis of DLBCL identifies immune environments associated with bispecific antibody response. Blood 2025; 145:2460-2472. [PMID: 39869833 DOI: 10.1182/blood.2024025355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/29/2025] Open
Abstract
ABSTRACT Most patients with diffuse large B-cell lymphoma (DLBCL) treated with immunotherapies such as bispecific antibodies (BsAbs) or chimeric antigen receptor (CAR) T cells fail to achieve durable treatment responses, underscoring the need for a deeper understanding of mechanisms that regulate the immune environment and response to treatment. Here, an integrative multiomics approach was applied to multiple large independent data sets to characterize DLBCL immune environments and to define their association with tumor cell-intrinsic genomic alterations and outcomes to CD19-directed CAR T-cell and CD20 × CD3 BsAb therapies. This approach effectively segregated DLBCLs into 4 immune quadrants (IQs) defined by cell-of-origin and immune-related gene set expression scores. These quadrants consisted of activated B cell-like (ABC) hot, ABC cold, germinal center B cell-like (GCB) hot, and GCB cold DLBCLs. Recurrent genomic alterations were enriched in each IQ, suggesting that lymphoma cell-intrinsic alterations contribute significantly to orchestrating unique DLBCL immune environments. For instance, SOCS1 loss-of-function mutations were significantly enriched among GCB hot DLBCLs, identifying a putative subset of inflamed DLBCLs that may be inherently susceptible to immunotherapy. In patients with relapsed/refractory DLBCL, DLBCL-IQ assignment correlated significantly with clinical benefit with a CD20 × CD3 BsAb (N = 74), but not with CD19-directed CAR T cells (Stanford, N = 51; Memorial Sloan Kettering Cancer Center, N = 69). Thus, DLBCL-IQ provides a new framework to conceptualize the DLBCL immune landscape and suggests the endogenous immune environment has a more significant impact on outcomes to BsAb than CAR T-cell treatment.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Antibodies, Bispecific/therapeutic use
- Antibodies, Bispecific/immunology
- Tumor Microenvironment/immunology
- Tumor Microenvironment/genetics
- Genomics/methods
- Antigens, CD19/immunology
- Immunotherapy, Adoptive
Collapse
Affiliation(s)
- Sravya Tumuluru
- Biological Sciences Division, Committee on Cancer Biology, The University of Chicago, Chicago, IL
| | - James K Godfrey
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope, Duarte, CA
| | - Alan Cooper
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL
| | - Jovian Yu
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL
| | - Xiufen Chen
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL
| | - Brendan W MacNabb
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA
| | | | - Yuanyuan Zha
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL
| | - Benedikt Pelzer
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Joo Song
- Department of Pathology, City of Hope, Duarte, CA
| | - Gerben Duns
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BC, Canada
| | - Brian J Sworder
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA
| | - Sandeep Raj
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | | | | | | | | | - Roni Shouval
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sonali M Smith
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL
| | - Ash A Alizadeh
- Division of Oncology, Department of Medicine, Stanford University, Palo Alto, CA
| | - Christian Steidl
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Justin Kline
- Biological Sciences Division, Committee on Cancer Biology, The University of Chicago, Chicago, IL
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL
| |
Collapse
|
17
|
Shu Z, Liu X, Li X, Fu S, Li S, Liu G, Tuo Z, Lan W, Lan B, Zhang Y. RAP1GAP is a prognostic biomarker and correlates with immune infiltrates in bladder cancer. Discov Oncol 2025; 16:863. [PMID: 40405009 PMCID: PMC12098257 DOI: 10.1007/s12672-025-02634-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 05/09/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND The role of RAP1GAP in tumor progression has garnered increasing attention; however, its prognostic value and immunological influence across various cancers remain uncertain. Our study presents a pan-cancer analysis to investigate its involvement in oncogenesis and immune regulation. METHODS Public databases were utilized to assess RAP1GAP expression across cancers. Cox regression analysis evaluated its prognostic value, while Pearson correlation examined associations with genomic heterogeneity, tumor stemness, immune cell infiltration, and immune checkpoints. Immunohistochemical staining of bladder cancer and adjacent tissues assessed RAP1GAP expression and clinical correlations. RESULTS RAP1GAP expression is differentially expressed in a variety of tumor types and predicts a better or worse prognosis for tumor patients. It was strongly linked to genomic heterogeneity and tumor stemness in multiple cancers. Immunohistochemistry showed increased RAP1GAP expression in bladder cancer. Immune cell analysis revealed high RAP1GAP expression was associated with greater infiltration of plasma cells, naive CD4 + T cells, Tregs, and eosinophils, while low expression correlated with increased CD8 + T cells, activated memory CD4 + T cells, and M1 macrophages. CONCLUSION RAP1GAP is a potential prognostic biomarker and immune regulator, with promising implications as an immunotherapeutic target for bladder cancer.
Collapse
Affiliation(s)
- Zehua Shu
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China
| | - Xinyi Liu
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiaoyan Li
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China
| | - Siming Fu
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China
| | - Sheng Li
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China
| | - Gaolei Liu
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhouting Tuo
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China
| | - Weihua Lan
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China
| | - Baohua Lan
- Department of Oncology, Chongqing Jiulongpo Science City People's Hospital, Chongqing, China.
| | - Yao Zhang
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
18
|
Zhao G, Ding J, Ma J, Jiang Y, Wang Y, Wang S, Li N. Integrative Analysis of Immune- and Metabolism-Related Genes Identifies Robust Prognostic Signature and PYCR1 as a Carcinogenic Regulator in Clear Cell Renal Cell Carcinoma. Int J Mol Sci 2025; 26:4953. [PMID: 40430095 PMCID: PMC12112471 DOI: 10.3390/ijms26104953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 05/04/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is distinguished by metabolic irregularities and unique immunological profiles. Nevertheless, the comprehensive examination of immune and metabolic attributes within the tumor microenvironment of ccRCC remains inadequately elucidated. In this study, we identified two distinct molecular subtypes (C1 and C2) of ccRCC using the non-negative matrix factorization (NMF) algorithm. Utilizing univariate and least absolute shrinkage and selection operator (LASSO) Cox regression analyses, we developed a prognostic signature comprising eight immune- and metabolism-related genes (IMRGs) associated with the tumor microenvironment. The validation of this signature was performed using both testing and entire datasets. A nomogram was developed using IMRGs prognostic signature and various clinical parameters, including age and TNM stage. We also performed the in vitro experiments to validate the carcinogenic role of PYCR1 in ccRCC cells. Subtype C1 exhibited a more favorable prognosis and higher levels of immune cell infiltration compared to subtype C2. The AUCs of the nomogram at 1-, 3-, and 5-year intervals (AUC = 0.874, 0.820, and 0.794) were slightly higher than those of the IMRGs signature alone (AUC = 0.773, 0.755, and 0.764). The association between risk score and immune checkpoint expressions, immunophenoscore (IPS), and microsatellite instability (MSI) collectively predicted treatment efficacy accurately. Additionally, in vitro experiments confirmed the involvement of PYCR1 in promoting the aggressive behaviors of ccRCC cells, as evidenced by reduced proliferation, invasion, and enhanced apoptosis upon PYCR1 knockdown. In conclusion, the IMRGs signature shows promise in predicting prognostic risk, assessing the effectiveness of immunotherapy, and tailoring treatment for ccRCC patients.
Collapse
Affiliation(s)
- Guo Zhao
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (G.Z.); (J.D.); (Y.J.); (Y.W.)
| | - Jiatong Ding
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (G.Z.); (J.D.); (Y.J.); (Y.W.)
| | - Jiaxiu Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300052, China;
| | - Yale Jiang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (G.Z.); (J.D.); (Y.J.); (Y.W.)
| | - Yuning Wang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (G.Z.); (J.D.); (Y.J.); (Y.W.)
| | - Shuhang Wang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (G.Z.); (J.D.); (Y.J.); (Y.W.)
| | - Ning Li
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (G.Z.); (J.D.); (Y.J.); (Y.W.)
| |
Collapse
|
19
|
Yan N, Liu J, Li G, Zhao L, Yang J, Guo Q, Zhou W, Luo Y, Gao Y. The ferroptosis-related gene GGTLC2 is identified as a novel biomarker for gastric cancer within the GGT family, with associations to immune infiltration and liver metastasis. Funct Integr Genomics 2025; 25:106. [PMID: 40397220 DOI: 10.1007/s10142-025-01614-0] [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: 09/14/2024] [Revised: 05/10/2025] [Accepted: 05/13/2025] [Indexed: 05/22/2025]
Abstract
Gastric cancer (GC) has a high incidence and poor prognosis, often metastasizing to the liver. Gamma-glutamyl transferase (GGT) is a key indicator of liver damage, and its family members are associated with various cancers. However, their expression and prognostic significance in GC remain unclear. This study utilized R to analyze the expression and prognosis of GGT family members using RNA-seq and clinical data from the TCGA database, applying Lasso regression for key gene identification. We identified GGTLC2 as a significant gene related to GC prognosis. We examined the clinical relevance, methylation levels, and copy number variations of GGTLC2 using the MEXPRESS database and performed GSEA analysis to identify enriched pathways. Additionally, we explored the relationship between GGTLC2 and immune cell infiltration, as well as immune-related genes, and established GGTLC2 knockdown and overexpression cell lines. The results indicate that GGTLC2 is highly expressed in GC and is associated with DNA methylation, copy number variation, and liver metastasis. Functionally, GGTLC2 is primarily enriched in oxidative stress and immune-related pathways, affecting immune infiltration and the expression of inflammatory factors in the tumor microenvironment. In vivo and in vitro studies demonstrate that knocking down GGTLC2 inhibits GC proliferation, invasion, and migration while promoting apoptosis and ferroptosis. Conversely, overexpressing GGTLC2 significantly increases the number of metastatic tumors in the liver. Overall, GGTLC2 may influence the occurrence, development, and liver metastasis of GC by inhibiting ferroptosis, making it a promising novel biomarker for the diagnosis and treatment of GC and its metastasis.
Collapse
Affiliation(s)
- Nan Yan
- Center for High Altitude Medicine, Laboratory of High Altitude Medicine in Qinghai Province, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, Qinghai, China
| | - Jianhong Liu
- College of Humanities and Technology, QingHai Open University, Xining City, China
| | - Gaofu Li
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Linglin Zhao
- Center for High Altitude Medicine, Laboratory of High Altitude Medicine in Qinghai Province, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, Qinghai, China
| | - Jie Yang
- Affiliated Hospital of Qinghai University, Xining, 810001, Qinghai, China
| | - Qijing Guo
- Department of Oncology, Air Force Medical Center. PLA, Beijing, China
| | - Wei Zhou
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Yushuang Luo
- Center for High Altitude Medicine, Laboratory of High Altitude Medicine in Qinghai Province, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, Qinghai, China.
- Affiliated Hospital of Qinghai University, Xining, 810001, Qinghai, China.
| | - Yue Gao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| |
Collapse
|
20
|
Li H, Chen H, Zhao T, Zhang W, Deng J, Xie W, Fan J, Lou H, Dong P, Han Z, Xing D, Mao S, Shen X, Xue X, Lu M. CD2AP shapes a stromal reduced tumor microenvironment and contributes to immunotherapy in gastric cancer. BMC Cancer 2025; 25:910. [PMID: 40399857 PMCID: PMC12096758 DOI: 10.1186/s12885-025-14248-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/29/2025] [Indexed: 05/23/2025] Open
Abstract
Gastric cancer (GC) ranks as the fifth most prevalent malignant tumor and stands as the fourth leading contributor to cancer-related fatalities on a global scale. The specific link between CD2 Associated Protein (CD2AP) expression and the tumor microenvironment (TME) remains unclear, and further exploration is needed to understand its potential role in immune response and as a target for immunotherapy in GC. Utilizing RNA sequencing data acquired from The Cancer Genome Atlas (TCGA) for a pan-cancer analysis, a comprehensive evaluation was carried out to determine the expression pattern and immunological involvement of CD2AP. Systematic association of CD2AP with immunological features within the stomach adenocarcinoma (STAD) TME was subsequently performed, encompassing factors like cancer immunity cycles, immune checkpoints, immunomodulators, tumor-infiltrating immune cells (TIICs). We found that CD2AP was enhanced expression in the TME of a variety of malignancies. CD2AP contributes to forming a stromal reduced TME in GC and improve the efficacy of immunotherapy. It was observed that patients with elevated levels of CD2AP, along with high scores on their CD4, CD20, and CD57 immune markers, tended to experience the most favorable prognosis. Furthermore, an IRS was constructed to accurately assess the prognosis of STAD patients. Since CD2AP was associated with the formation of stromal reduced TME in STAD, the expression of CD2AP can improve the effect of immunotherapy of STAD. CD2AP could emerge as a novel prognostic biomarker for STAD, offering a fresh avenue for molecular targeted therapy.
Collapse
Affiliation(s)
- Haoliang Li
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hua Chen
- Department of Radiation and Medical Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ting Zhao
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wenqi Zhang
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jing Deng
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wangkai Xie
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jianing Fan
- School of Second Clinical Medical, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Han Lou
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Pingping Dong
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zheng Han
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Dong Xing
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Sunzhong Mao
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Xian Shen
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Xiangyang Xue
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Mingdong Lu
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
21
|
Wang S, Zheng Q, Chen L. Integration of Bulk and Single-Cell RNA Sequencing to Identify RNA Modifications-Related Prognostic Signature in Ovarian Cancer. Int J Gen Med 2025; 18:2629-2647. [PMID: 40417417 PMCID: PMC12103173 DOI: 10.2147/ijgm.s523878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Accepted: 05/15/2025] [Indexed: 05/27/2025] Open
Abstract
Background Ovarian cancer (OC), a common fatal malignancy in women, has a poor prognosis. RNA modifications are associated with the development of OC. In this study, we aimed to identify and verify RNA modifications-related prognostic genes in OC by integrating bulk and single-cell RNA sequencing (scRNA-seq) data. Methods Transcriptome data came from public databases and RNA modifications-related genes (RMRGs) were obtained from literature. Candidate genes were identified by intersecting RMRGs with differentially expressed genes (DEGs) in OC patients. Prognostic genes were gained via machine learning techniques, particularly LASSO regression. A risk model was built to predict the prognosis. OC patients were divided into high-risk and low-risk groups according to risk score. Subsequent analyses covered enrichment analysis, immune microenvironment, mutation analysis, and chemotherapeutic drug sensitivity. In addition, scRNA-seq data was assessed for key cells and gene expression in them. Finally, RT-qPCR was applied to identify the expression of prognostic genes. Results LSM4, SNRPC, ZC3H13, LSM2, WTAP, DCP2, PUS7, and TUT1 were selected as prognostic genes. The risk model exhibited excellent predictive abilities. Seventeen pathways were enriched like calcium signaling pathway, 7 differential immune cells were identified like regulatory T cells and plasmacytoid dendritic cells, and TP53 had highest mutation rate. Half-maximal inhibitory concentrations (IC50) values of 47 drugs like paclitaxel differed between two risk groups. The prognostic genes were distributed mainly in fibroblast cells, epithelial cells and endothelial cells. During fibroblast cells differentiation, expression of prognostic genes fluctuated to varying degrees. The RT-qPCR demonstrated that the expression of LSM2, LSM4, PUS7, SNRPC, and TUT1 were upregulated in OC, while DCP2, WTAP, and ZC3H13 were downregulated. Conclusion We constructed an RNA modifications-related prognostic signature that can effectively predict clinical outcomes and therapeutic responses in patients with OC.
Collapse
Affiliation(s)
- Shaoyu Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
- Department of Obstetrics and Gynecology, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, People’s Republic of China
- Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
| | - Qiaomei Zheng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
- Department of Obstetrics and Gynecology, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, People’s Republic of China
- Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
| | - Lihong Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
- Department of Obstetrics and Gynecology, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, People’s Republic of China
- Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
| |
Collapse
|
22
|
Mauriello A, Cavalluzzo B, Ragone C, Tagliamonte M, Buonaguro L. Shared neoantigens' atlas for off-the-shelf cancer vaccine development. J Transl Med 2025; 23:558. [PMID: 40390041 PMCID: PMC12087128 DOI: 10.1186/s12967-025-06478-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 04/10/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND We have recently described that the most prevalent 100 mutations identified in human cancers, both single nucleotide variations (SNVs) and InDels, generate a handful number of shared mutated neoantigens (SNV and InDel-NeoAgs) in association with 5 HLA-A and 7 B haplotypes. METHODS In the present study, we expanded such analysis to 50 haplotypes in the three MHC class I loci (10 HLA-A, 27 HLA-B and 13 HLA-C), including all the mutated proteins identified in at least 5% of cancer patients. RESULTS Overall, the extended analysis identified 15 SNV-NeoAgs and 55 InDel-NeoAgs with a significant affinity improvement over the corresponding wt (DAI > 10). These targetable shared NeoAgs are prevalently derived from PIK3CAH1047R (6/15 SNV-NeoAgs) and LARP4BT163Hfs (30/55 InDel-NeoAgs). From the HLA perspective, the HLA-A*33:03 is associated with the largest number of SNV-NeoAgs (4/15 NeoAgs) and the HLA-B*58:01 is associated with the largest number of InDel-NeoAgs (16/55 NeoAgs). According to the distribution of each HLA haplotype in at least 10% of the regional populations, therapeutic cancer vaccines based on mutated shared SNV and InDel-NeoAgs, might be developed for COAD, STAD and UCEC cancers, with a global coverage, and for PAAD and UVM, with a regional coverage. CONCLUSIONS This represents the first in-depth analysis for the identification of a specific repertoire of shared mutated NeoAgs, most of which never reported before. Such shared SNV and InDel-NeoAgs are indispensable for the development of "off-the-shelf" cancer vaccines targeting a relevant percentage of cancers in a significant percentage of cancer patients worldwide.
Collapse
Affiliation(s)
- Angela Mauriello
- Innovative Immunological Models Unit, Istituto Nazionale Tumori IRCCS - "Fond G. Pascale", Via Mariano Semmola, 52, Naples, Italy
| | - Beatrice Cavalluzzo
- Innovative Immunological Models Unit, Istituto Nazionale Tumori IRCCS - "Fond G. Pascale", Via Mariano Semmola, 52, Naples, Italy
| | - Concetta Ragone
- Innovative Immunological Models Unit, Istituto Nazionale Tumori IRCCS - "Fond G. Pascale", Via Mariano Semmola, 52, Naples, Italy
| | - Maria Tagliamonte
- Innovative Immunological Models Unit, Istituto Nazionale Tumori IRCCS - "Fond G. Pascale", Via Mariano Semmola, 52, Naples, Italy.
| | - Luigi Buonaguro
- Innovative Immunological Models Unit, Istituto Nazionale Tumori IRCCS - "Fond G. Pascale", Via Mariano Semmola, 52, Naples, Italy.
| |
Collapse
|
23
|
He W, Wei M, Huang Y, Qin J, Liu M, Liu N, He Y, Chen C, Huang Y, Yin H, Zhang R. Integrated Bioinformatics Analysis and Cellular Experimental Validation Identify Lipoprotein Lipase Gene as a Novel Biomarker for Tumorigenesis and Prognosis in Lung Adenocarcinoma. BIOLOGY 2025; 14:566. [PMID: 40427755 PMCID: PMC12108960 DOI: 10.3390/biology14050566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 05/06/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025]
Abstract
Lung adenocarcinoma (LUAD) is one of the leading causes of death worldwide, and thus, more biomarker and therapeutic targets need to be explored. Herein, we aimed to explore new biomarkers of LUAD by integrating bioinformatics analysis with cell experiments. We firstly identified 266 druggable genes that were significantly differentially expressed between LUAD tissues and adjacent normal lung tissues. Among these genes, SMR analysis with p-value correction suggested that declining lipoprotein lipase (LPL) levels may be causally associated with an elevated risk of LUAD, which was corroborated by co-localization analysis. Analyses of clinical data showed that LPL in lung cancer tissues has considerable diagnostic value for LUAD, and elevated LPL levels were positively associated with improved patient survival outcomes. Cell experiments with an LPL activator proved these findings; the activator inhibited the proliferation and migration of lung cancer cells. Next, we found that LPL promoted the infiltration of immune cells such as DCs, IDCs, and macrophages in LUAD by mononuclear sequencing analysis and TIMER2.0. Meanwhile, patients with low levels of LPL expression demonstrated superior immunotherapeutic responses to anti-PD-1 therapy. We conclude that LPL acts as a diagnostic and prognostic marker for LUAD.
Collapse
Affiliation(s)
- Wanwan He
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; (W.H.); (M.W.); (Y.H.); (J.Q.); (M.L.); (N.L.); (Y.H.); (Y.H.)
| | - Meilian Wei
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; (W.H.); (M.W.); (Y.H.); (J.Q.); (M.L.); (N.L.); (Y.H.); (Y.H.)
| | - Yan Huang
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; (W.H.); (M.W.); (Y.H.); (J.Q.); (M.L.); (N.L.); (Y.H.); (Y.H.)
| | - Junsen Qin
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; (W.H.); (M.W.); (Y.H.); (J.Q.); (M.L.); (N.L.); (Y.H.); (Y.H.)
| | - Meng Liu
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; (W.H.); (M.W.); (Y.H.); (J.Q.); (M.L.); (N.L.); (Y.H.); (Y.H.)
| | - Na Liu
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; (W.H.); (M.W.); (Y.H.); (J.Q.); (M.L.); (N.L.); (Y.H.); (Y.H.)
| | - Yanli He
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; (W.H.); (M.W.); (Y.H.); (J.Q.); (M.L.); (N.L.); (Y.H.); (Y.H.)
| | - Chuanbing Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China;
| | - Yali Huang
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; (W.H.); (M.W.); (Y.H.); (J.Q.); (M.L.); (N.L.); (Y.H.); (Y.H.)
| | - Heng Yin
- Institute of Infectious Diseases, Guangzhou Medical University, Guangzhou 510182, China
| | - Ren Zhang
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; (W.H.); (M.W.); (Y.H.); (J.Q.); (M.L.); (N.L.); (Y.H.); (Y.H.)
| |
Collapse
|
24
|
Liang MZ, Huang XF, Zhu JC, Bao JX, Chen CL, Wang XW, Lou YW, Pan YT, Dai YW. A machine learning-based glycolysis and fatty acid metabolism-related prognostic signature is constructed and identified ACSL5 as a novel marker inhibiting the proliferation of breast cancer. Comput Biol Chem 2025; 119:108507. [PMID: 40403353 DOI: 10.1016/j.compbiolchem.2025.108507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 04/27/2025] [Accepted: 05/09/2025] [Indexed: 05/24/2025]
Abstract
INTRODUCTION A new perspective on cancer metabolism suggests that it varies by context and is diverse. Cancer metabolism reprogramming can create a heterogeneous microenvironment that affects immune cell infiltration and function, complicating the selection of treatment methods. However, the specifics of this relationship remain unclear in breast cancer. This research aims to explore how glycolysis and fatty acid metabolism (GF) influence the immune microenvironment and their predictive capabilities for immunotherapy responses and overall survival. METHODS We at first time identified 602 GF-related genes. Utilizing multiple datasets from various centers and employing 10 different machine learning algorithms, we developed a GF-related signature called GFSscore, driven by artificial intelligence. RESULTS The GFSscore served as an independent prognostic indicator and demonstrated greater robustness than other models. Its validity was validated through multiple databases. Our study found that breast cancer patients with a high GFSscore, indicative of a greater tendency towards glycolytic activity, experienced poorer prognosis due to immunosuppression from distinct immune evasion mechanisms. Conversely, those with a low GFSscore, more inclined towards fatty acid metabolism, had better outcomes. Additionally, the GFSscore has the potential to forecast how well a patient might respond to immunotherapy and their susceptibility to chemotherapy medications. Moreover, we found that the overexpressed ACSL5 gene inhibits the proliferation of BRCA through experiments. CONCLUSIONS The GFSscore may offer patients personalized therapy by identifying new therapeutic targets for tumors. By understanding the relationship between cancer metabolism and the immune microenvironment, we can better tailor treatments to individual patients.
Collapse
Affiliation(s)
- Mei-Zhen Liang
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Xian-Feng Huang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Jun-Chang Zhu
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Jing-Xia Bao
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Cheng-Liang Chen
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Xiao-Wu Wang
- Department of Burns and Skin Repair Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Yun-Wei Lou
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Ya-Ting Pan
- Yongkang First People's Hospital Medical Group, Jinhua, Zhejiang, China.
| | - Yin-Wei Dai
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
25
|
Zhang Y, Zhu H, Fan J, Zhao J, Xia Y, Zhang N, Xu H. A glutamine metabolism gene signature with prognostic and predictive value for colorectal cancer survival and immunotherapy response. Front Mol Biosci 2025; 12:1599141. [PMID: 40443528 PMCID: PMC12119274 DOI: 10.3389/fmolb.2025.1599141] [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: 04/02/2025] [Accepted: 04/23/2025] [Indexed: 06/02/2025] Open
Abstract
Background Colorectal cancer (CRC) remains a major cause of cancer mortality, and dysregulated glutamine metabolism has emerged as a potential therapeutic target. However, the precise role of glutamine in CRC progression and treatment response remains debated. Methods The authors collected transcriptome and microbiome information, from multiple sources to construct the GLMscore, a prognostic signature in CRC. To comprehensively characterize the biological features of GLMscore groups, the integration of transcriptomic profiling, KEGG pathway enrichment analysis, immune infiltration analysis, tumor immune microenvironment characterization, microbiome analysis, and tissue imaging were applied. Furthermore, CRC patients were stratified into GLMscore high and GLMscore low groups. The robustness of GLMscore was validated in both training and validation cohorts, and the predictive value for immunotherapy response was assessed. Finally, single-cell RNA sequencing (scRNA-seq) analysis was conducted to delineate the differences between GLMscore high and GLMscore low groups. Results High GLMscore was associated with elevated expression of pathways related to tumorigenesis, epithelial-mesenchymal transition (EMT), and angiogenesis. Furthermore, high GLMscore patients exhibited an immunosuppressive TME characterized by increased infiltration of M0 and M2 macrophages, reduced overall immune infiltration (supported by ESTIMATE and TIDE scores), and increased expression of immune exclusion and suppression pathways. Analysis of pathological whole-slide images (WSIs) revealed a lack of intratumoral tertiary lymphoid structures (TLSs) in high GLMscore patients. The GLMscore also predicted resistance to common chemotherapeutic agents (using GDSC data) and, importantly, predicted poor response to immunotherapy in the IMvigor210 cohort. Analysis of 16S rRNA gene sequencing data revealed an enrichment of potentially oncogenic microbiota, including Hungatella and Selenomonas, in high GLMscore group. Single-cell analysis further confirmed the immunosuppressive TME and identified increased cell-cell communication between inflammatory macrophages and tumor cells in high GLMscore group. Conclusion The authors innovatively constructed GLMscore, a robust scoring system in quantifying CRC patients, exploring the distinct biological features, tumor immune microenvironment and microbiome ecology, exhibiting high validity in predicting survival prognosis and clinical treatment efficacy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hong Xu
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
26
|
Wen H, Dai F, Wang H, Lin Y, Xu Z, Lyu Z. Identification and validation of SLC16A8 as a prognostic biomarker in clear cell renal cell carcinoma: a six-gene solute carrier signature. Exp Cell Res 2025; 448:114567. [PMID: 40268265 DOI: 10.1016/j.yexcr.2025.114567] [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: 01/15/2025] [Revised: 04/10/2025] [Accepted: 04/20/2025] [Indexed: 04/25/2025]
Abstract
Solute carrier (SLC) proteins are essential for nutrient transport, influencing tumor metabolism and growth while preserving cellular homeostasis. Despite the critical biological functions of these transporters, their applicability as therapeutic targets in clear cell renal cell carcinoma (ccRCC) remains largely unexplored. In the current study, we analyzed transcriptomic data and discovered 77 differentially expressed SLC genes in ccRCC, with 24 demonstrating predictive potential. Using Lasso regression, we developed a prognostic signature comprising six key genes: SLC2A3, SLC11A1, SLC14A1, SLC16A8, SLC22A6, and SLC28A1. This signature demonstrated strong diagnostic performance and served as an independent predictor of patient survival. Further analysis integrating clinical variables and risk scores enabled the construction of nomograms, which exhibited high predictive accuracy for patient outcomes. Immune profiling revealed distinct infiltration patterns between risk groups: high-risk patients showed elevated levels of memory B cells, activated CD4+ T cells, regulatory T cells (Tregs), M0 macrophages, and neutrophils. In contrast, their low-risk counterparts showed M1 macrophages, resting dendritic cells, and resting mast cells. Validation experiments confirmed that SLC16A8 was significantly overexpressed in ccRCC tissues compared to normal samples, correlating with poor prognosis. Functional studies demonstrated that SLC16A8 knockdown impaired tumor progression in vitro. Consistent with these findings, in vivo experiments demonstrated reduced tumor growth upon SLC16A8 knockdown. Mechanistically, decreased SLC16A8 attenuated PI3K/AKT signaling, suggesting a potential regulatory pathway in ccRCC progression. In summary, we established a six-gene SLC signature with significant prognostic value in ccRCC. Among these genes, SLC16A8 emerged as a promising biomarker and therapeutic target, warranting further investigation.
Collapse
Affiliation(s)
- Hantao Wen
- Institute of Precision Medicine, Peking University Shenzhen Hospital, PKU-Shenzhen Clinical Institute of Shantou University Medical College, Shenzhen, China, 518036
| | - Fang Dai
- Department of Urology, PKU-Shenzhen Clinical Institute of Anhui Medical University, Shenzhen, China, 518036
| | - Huming Wang
- Department of Urology, PKU-Shenzhen Clinical Institute of Anhui Medical University, Shenzhen, China, 518036
| | - Yu Lin
- Institute of Precision Medicine, Peking University Shenzhen Hospital, PKU-Shenzhen Clinical Institute of Shantou University Medical College, Shenzhen, China, 518036
| | - Zihan Xu
- Institute of Precision Medicine, Peking University Shenzhen Hospital, PKU-Shenzhen Clinical Institute of Shantou University Medical College, Shenzhen, China, 518036
| | - Zhaojie Lyu
- Institute of Precision Medicine, Peking University Shenzhen Hospital, PKU-Shenzhen Clinical Institute of Shantou University Medical College, Shenzhen, China, 518036; Department of Urology, PKU-Shenzhen Clinical Institute of Anhui Medical University, Shenzhen, China, 518036.
| |
Collapse
|
27
|
Lv Y, Cai X, Zheng H, Dai H. Identification of different lung adenocarcinoma subtypes in combination with antidiuretic hormone-related genes and creation of an associated index to predict prognosis and guide immunotherapy. Comput Biol Chem 2025; 119:108506. [PMID: 40412340 DOI: 10.1016/j.compbiolchem.2025.108506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 04/25/2025] [Accepted: 05/09/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is one of the most aggressive and rapidly lethal tumor types. Previous studies have demonstrated the involvement of antidiuretic hormone (ADH)-related genes in cancer. However, the role of ADH-related genes in LUAD remains unclear. Therefore, investigating the characteristics of these genes in LUAD is essential. METHODS Differentially expressed genes (DEGs) associated with ADH in LUAD were identified using the STRING database. Consensus clustering was performed, and a protein-protein interaction network was constructed for the DEGs between subtypes. Genes extracted from the PPI network underwent univariate, LASSO, and multivariate Cox regression analyses to develop a predictive model for LUAD. A nomogram integrating clinical data and risk scores was created, and its prognostic power for overall survival (OS) in LUAD patients was evaluated. Additionally, LUAD patients were analyzed for targeted therapies, immune landscape, functional enrichment, and mutation profiles. Finally, qRT-PCR was used to examine the expression of signature genes in LUAD cells. RESULTS Based on ADH-related DEGs, LUAD patients were stratified into two clusters (Cluster 1 and Cluster 2) with distinct survival outcomes. A predictive model incorporating nine feature genes was subsequently constructed using DEGs from these two subtypes. The receiver operating characteristic curve demonstrated the model's prognostic accuracy in predicting OS in LUAD patients. Compared to the high-risk group, patients in the low-risk group exhibited higher immune infiltration levels and immunophenoscore, along with lower tumor immune dysfunction and exclusion scores. Enrichment analysis revealed that immune response pathways and ligand-receptor interactions were the primary functional categories distinguishing the high- and low-risk groups. The low-risk group showed a significantly lower gene mutation burden. Drug sensitivity analysis identified several potential targeted therapies, including Dabrafenib, ARQ-680, Vemurafenib, BGB-283, MLN-2480, and GDC-0994, which might act on hub genes. qRT-PCR validation confirmed that DNAH12 was significantly downregulated in tumor tissues, while DKK1, DLX2, IGFBP1, NTSR1, RPE65, and VGF were markedly upregulated. CONCLUSION This study provided potential prognostic biomarkers for LUAD and might facilitate the development of effective immunotherapy strategies for LUAD patients.
Collapse
Affiliation(s)
- Yuankai Lv
- Department of Respiratory, Lishui People's Hospital, Lishui 323000, China
| | - Xiaoping Cai
- Department of Respiratory, Lishui People's Hospital, Lishui 323000, China
| | - Hao Zheng
- Department of Respiratory, Lishui People's Hospital, Lishui 323000, China
| | - Hong Dai
- Department of Anesthesiology, Lishui People's Hospital, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China.
| |
Collapse
|
28
|
Luo X, Xue D. Potential mechanisms of epigenetic regulation in diabetic retinopathy from the perspectives of multi-omics. Diabetol Metab Syndr 2025; 17:155. [PMID: 40369608 PMCID: PMC12076923 DOI: 10.1186/s13098-025-01723-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 05/02/2025] [Indexed: 05/16/2025] Open
Abstract
PURPOSE Diabetic retinopathy (DR) is a significant complication of diabetes, with complex pathogenesis involving epigenetic modifications. This study aimed to explore the epigenetic regulatory mechanisms contributing to DR. METHODS DR-related data, including DNA methylation, mRNA, and miRNA expression datasets, were obtained from the Gene Expression Omnibus database. Differential gene expression analysis was performed to identify differentially methylated genes and expressed mRNAs and miRNAs. Cross-analysis established the methylation-expression and miRNA-mRNA regulatory networks. A comprehensive DR-related epigenetic regulatory network was constructed, identifying hub genes. The expression characteristics of these hub genes in various immune cells were examined using single-cell RNA sequencing. RESULTS We identified 10,716 differentially methylated genes, 1,181 differentially expressed mRNAs, and 615 differentially expressed miRNAs in DR. The methylation-expression regulatory network was associated with pathways such as TGF-beta and ErbB signaling. The miRNA regulatory network was linked to pathways related to cellular senescence, adherents junctions, and endocytosis. Five hub genes were identified: TFRC, AP2M1, AP2A1, DAB2, and PPP1CB. Single-cell RNA sequencing revealed specific expression of these genes in particular immune cells, highlighting their potential roles in DR pathogenesis. CONCLUSION This study constructed a comprehensive epigenetic regulatory network for DR and identified key regulatory genes, offering new insights into the molecular mechanisms underlying DR and potential therapeutic targets for diagnosis and treatment.
Collapse
Affiliation(s)
- Xin Luo
- Department of Ophthalmic, Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital Northwest University, Xi'an, 710004, China
| | - Daxi Xue
- Department of Ophthalmic, Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital Northwest University, Xi'an, 710004, China.
| |
Collapse
|
29
|
Rao W, Zhang Q, Dai X, Yang Y, Lei Z, Kuang X, Xiao H, Zhu J, Xiong Y, Wang D, Yang L. A three-subtype prognostic classification based on base excision repair and oxidative stress genes in lung adenocarcinoma and its relationship with tumor microenvironment. Sci Rep 2025; 15:16647. [PMID: 40360689 PMCID: PMC12075871 DOI: 10.1038/s41598-025-98088-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 04/09/2025] [Indexed: 05/15/2025] Open
Abstract
Unrepaired DNA damage is the initiation of mutation and tumor-specific biological characteristics. Oxidative stress and base excision repair (BER) are the two main pathways to cope with oxidative DNA damage, which is closely related to the heterogeneity of Lung adenocarcinoma (LUAD), but their relationship with tumor biological characteristics is unclear, and a molecular subtyping based on comprehensive BER and oxidative stress gene expression is lacking. 501 samples from The Cancer Genome Atlas (TCGA) were classified into three subtypes based on genes related to BER and oxidative stress through hierarchical agglomerative cluster analysis. By integrating the nearest template prediction (NTP), four GEO datasets and 52 samples from our institution were analyzed for validation. Bioinformatic analysis was performed to define the diverse molecular characteristics, mutation background, tumor microenvironment, and prognosis. Three subtypes with distinct gene signatures were identified: relatively high BER and low oxidative stress gene expression (C1), low BER gene and high oxidative stress gene expression (C2), and high expression of both BER and oxidative stress genes (C3). C2 was characterized by a low mutation frequency in TP53 (29%) and a high mutation frequency in EGFR (20%), whereas a high frequency of mutation was seen in C3 in STK11 and KEAP1 genes. Additionally, differentially expressed genes among the three subtypes were particularly enriched in immune-related pathways, and the abundance of immune cells and Immunophenoscore were significantly higher in C2, while the Tumor Immune Dysfunction and Exclusion (TIDE) score was lower in C2, indicating a better response to immunotherapy. C2 was also associated with an improved survival outcome compared with C1 and C3, and this finding was validated in 978 samples from four independent GEO datasets and 52 samples at our institution by the NTP algorithm. The three-subtype classifications based on BER and oxidative stress gene expression offers potential for predicting the survival and response to immunotherapy of LUAD patients.
Collapse
Affiliation(s)
- Wen Rao
- Cancer Center, Daping Hospital and Army Medical Center of PLA, Army Medical University, No.10 Changjiangzhi Rd, Yuzhong District, Chongqing, People's Republic of China
- The 75th Group Army Hospital, Dali, Yunnan, People's Republic of China
| | - Qin Zhang
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Cancer Hospital Affiliate to School of Medicine,, University of Electronic Science and Technology of China, Sichuan, People's Republic of China
| | - Xiaoyan Dai
- Cancer Center, Daping Hospital and Army Medical Center of PLA, Army Medical University, No.10 Changjiangzhi Rd, Yuzhong District, Chongqing, People's Republic of China
| | - Yuxin Yang
- Cancer Center, Daping Hospital and Army Medical Center of PLA, Army Medical University, No.10 Changjiangzhi Rd, Yuzhong District, Chongqing, People's Republic of China
| | - Zhang Lei
- Cancer Center, Daping Hospital and Army Medical Center of PLA, Army Medical University, No.10 Changjiangzhi Rd, Yuzhong District, Chongqing, People's Republic of China
| | - Xunjie Kuang
- Cancer Center, Daping Hospital and Army Medical Center of PLA, Army Medical University, No.10 Changjiangzhi Rd, Yuzhong District, Chongqing, People's Republic of China
| | - He Xiao
- Cancer Center, Daping Hospital and Army Medical Center of PLA, Army Medical University, No.10 Changjiangzhi Rd, Yuzhong District, Chongqing, People's Republic of China
| | - Jianwu Zhu
- Cancer Center, Daping Hospital and Army Medical Center of PLA, Army Medical University, No.10 Changjiangzhi Rd, Yuzhong District, Chongqing, People's Republic of China
| | - Yanli Xiong
- Cancer Center, Daping Hospital and Army Medical Center of PLA, Army Medical University, No.10 Changjiangzhi Rd, Yuzhong District, Chongqing, People's Republic of China
| | - Dong Wang
- Chongqing University Qianjiang Hospital, Chongqing, People's Republic of China.
| | - Lujie Yang
- Cancer Center, Daping Hospital and Army Medical Center of PLA, Army Medical University, No.10 Changjiangzhi Rd, Yuzhong District, Chongqing, People's Republic of China.
| |
Collapse
|
30
|
Xiong Y, Tong Y. Development of macrophage M2 relate signature for predicting prognosis and immunotherapy response in ovarian cancer. Discov Oncol 2025; 16:750. [PMID: 40358821 PMCID: PMC12075037 DOI: 10.1007/s12672-025-02559-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 05/05/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Ovarian cancer ranks as the fifth most common cause of cancer-related deaths in women worldwide. Macrophages M2 is believed to support tumor growth by suppressing immune responses and promoting angiogenesis. METHODS A macrophage M2-related signature (MRS) was developed by applying machine learning methods that included 10 algorithms and utilized data from the TCGA, GSE14764 and GSE140082 datasets. The predictive capacity of the MRS for immunotherapy response was evaluated through various methods, including immunophenoscore, TIDE score, TMB score, immune escape score, as well as two immunotherapy cohorts (IMvigor210 and GSE91061). RESULTS The optimal MRS, developed using the lasso algorithm, served as an independent prognostic factor and demonstrated stable performance in predicting overall survival rates in ovarian cancer. In the TCGA dataset, the AUC values for the 1-, 3-, and 5-year ROC curves were 0.874, 0.808, and 0.813, respectively. The C-index of our MRS was superior to that of clinical stage, tumor grade, and several other established prognostic signatures. Ovarian cancer patients with low risk score exhibited higher ESTIMATE score, increased levels of immune cells, elevated PDI&CTLA4 immunophenoscore, higher TMB score, reduced TIDE score, and lower immune escape score. Additionally, the survival prediction nomogram displayed significant potential for clinical application in estimating the 1-, 3-, and 5-year overall survival rates of ovarian cancer patients. CONCLUSION Our study developed a novel MRS for ovarian cancer, which could act as an indicator for predicting the prognosis and immunotherapy response in ovarian cancer.
Collapse
Affiliation(s)
- Yifei Xiong
- Reproductive Medicine Center, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, China
| | - Yan Tong
- Reproductive Medicine Center, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, China.
| |
Collapse
|
31
|
Yang Y, Luo W, Feng Z, Chen X, Li J, Zuo L, Duan M, He X, Wang W, He F, Liu F. An integrative analysis combining bioinformatics, network pharmacology and experimental methods identified key genes of EGCG targets in Nasopharyngeal Carcinoma. Discov Oncol 2025; 16:742. [PMID: 40355769 PMCID: PMC12069167 DOI: 10.1007/s12672-025-02365-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 04/10/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Epigallocatechin gallate (EGCG), a frequently studied catechin in green tea, has been shown to be involved in the antiproliferation and apoptosis of human Nasopharyngeal carcinoma (NPC) cells. However, the pharmacological targets and mechanism by which EGCG can combat NPC patients remain to be studied in detail. METHODS Network pharmacology and bioinformatics were employed to investigate the molecular mechanisms underlying EGCG's therapeutic effects on NPC, with an emphasis on developing a prognostic risk model and identifying potential therapeutic targets. RESULTS A novel prognostic risk model was developed using univariate Cox regression, LASSO regression and multivariable Cox regression analyses, incorporating six genes to stratify patients into low- and highrisk groups. Kaplan-Meier analysis demonstrated significantly shorter progression-free survival in the high-risk group. The model's accuracy was further validated using time-dependent Receiver Operating Characteristic (ROC) curves. ESTIMATE analysis revealed significantly higher immune, stromal and overall ESTIMATE scores in the low-risk group compared to the high-risk group. Immune profiling indicated significant differences in five immune cell subtypes (memory B cells, regulatory T cells (Tregs), gamma delta T cells, activated NK cells and activated dendritic cells) between the two risk groups. Additionally, the low-risk group showed greater sensitivity to conventional chemotherapeutic agents. Immunohistochemistry and molecular docking analyses identified CYCS and MYL12B as promising targets for EGCG treatment. CONCLUSION This study utilised network pharmacology and bioinformatics to identify shared genes between EGCG and NPC, aiming to elucidate the molecular mechanisms through which EGCG inhibits NPC and to develop a prognostic model for assessing patient outcomes. The findings provide potential insights for the development of anti-NPC therapies and their clinical applications.
Collapse
Affiliation(s)
- Yuhang Yang
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Wenqi Luo
- Department of Pathology, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Zhang Feng
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Xiaoyu Chen
- Department of Pathology, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Jinqing Li
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Long Zuo
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Meijiao Duan
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Xiaosong He
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Wenhua Wang
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Feng He
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China.
| | - Fangxian Liu
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China.
| |
Collapse
|
32
|
Liu C, Liao C, Sun B, Guo Z, Chen S, Liu S, Yuan X, Huang Z, Liu J, Deng M, Wang K, Wu R, Zhao J, Dong X. Tumour-infiltrating immune cells as a novel prognostic model for bladder cancer. Discov Oncol 2025; 16:725. [PMID: 40350535 PMCID: PMC12066389 DOI: 10.1007/s12672-025-02292-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 04/02/2025] [Indexed: 05/14/2025] Open
Abstract
Bladder cancer (BLCA) is the tenth most commonly diagnosed cancer and poses a significant challenge due to its complexity and associated high morbidity and mortality rates in the absence of optimal treatment. The tumor microenvironment (TME) is recognized as a critical factor in tumor initiation, progression and therapeutic response, and offers numerous potential targets for intervention. A comprehensive understanding of immune infiltration patterns in BLCA is essential for the development of effective prevention and treatment strategies. In this study, bioinformatics analysis was used to identify differentially expressed genes (DEGs) and tumor-infiltrating immune cells (TIICs) between BLCA tissues and adjacent normal tissues. Weighted gene co-expression network analysis (WGCNA) and protein-protein interaction (PPI) analysis were used to identify the top 10 hub genes with the most significant co-expression effects, and their potential relationship with patient prognosis was then predicted. The random survival forest (RSF) model was used to further identify six variables among the hub genes and establish a novel scoring system, defined as the tumor-infiltrating immune score (TIIS) to predict the prognosis of BLCA patients. In addition, the correlation analysis between TIIS and drug sensitivity was investigated using the Genomics of Drug Sensitivity in Cancer (GDSC) and Cancer Therapeutics Response Portal (CTRP) databases. Patients with high TIIS were found to have a poor prognosis but may be more sensitive to Cisplatin and certain novel agents. This study provided a systematic analysis of immune cell infiltration in BLCA and established TIIS to predict patient prognosis and the efficacy of specific drugs in the treatment of BLCA.
Collapse
Affiliation(s)
- Can Liu
- Department of Urology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Chaoyu Liao
- Department of Urology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Bishao Sun
- Department of Urology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Zhen Guo
- Urology Department, Chongqing Shapingba Hospital, School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Sihao Chen
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing, 400010, China
| | - Shixue Liu
- Urology Department, Chongqing Shapingba Hospital, School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Xiaoyu Yuan
- Urology Department, Chongqing Shapingba Hospital, School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Zeyu Huang
- Department of Urology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Jingui Liu
- Department of Urology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Min Deng
- Department of Urology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Kui Wang
- Department of Urology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Ruixin Wu
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China.
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing, 400010, China.
| | - Jiang Zhao
- Department of Urology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China.
| | - Xingyou Dong
- Urology Department, Chongqing Shapingba Hospital, School of Medicine, Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
33
|
Chen Z, Zhang Y. Development of an immune-related gene signature applying Ridge method for improving immunotherapy responses and clinical outcomes in lung adenocarcinoma. PeerJ 2025; 13:e19121. [PMID: 40352269 PMCID: PMC12066106 DOI: 10.7717/peerj.19121] [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: 11/13/2024] [Accepted: 02/17/2025] [Indexed: 05/14/2025] Open
Abstract
Background Lung adenocarcinoma (LUAD) is a major cause of cancer mortality. Considering the critical role of tumor infiltrating lymphocytes in effective immunotherapy, this study was designed to screen molecular markers related to tumor infiltrating cells in LUAD, aiming to improve immunotherapy response during LUAD therapy. Methods The ConsensusClusterPlus method was used for clustering immune molecular subtypes of LUAD. Immune cell infiltration and immunotherapeutic potential in each subtype was evaluated employing single-sample gene set enrichment analysis (ssGSEA), Tumor Immune Dysfunction and Exclusion (TIDE), and Immunophenoscore (IPS). Immune-related co-expression modules were classified by weighted gene co-expression network analysis (WGCNA) analysis. The sequencing data of immune-related genes were comprehensively analyzed by introducing a new computational framework and 10 machine learning algorithms (a total of 101 combinations) to determine the prognostic genes, which were further combined to develop an immune prognostic signature (IMMPS) using the stepCox and Ridge methods. The expression of the signature genes was validated by quantitative real-time PCR (qRT-PCR). Results Samples from The Cancer Genome Atlas dataset (TCGA-LUAD) were divided into two subtypes (immunosuppressive subgroup C1 and immune-activated subgroup C2); notably, the C2 subgroup was more likely to benefit from immunotherapy (p < 0.05). An IMMPS developed based on seven immune infiltrating cell-related genes (SEMA7A, EFHD2, CHST11, SLC24A4, MAL, JCHAIN, and SCARF1) could accurately predict the overall survival of LUAD in five LUAD cohorts, with an average C-index higher than 0.69. LUAD patients with a low IMMPS value had a higher immune cell infiltration (p < 0.05). In addition, the IMMPS exhibited better prediction performance in comparison to 154 published gene signatures, suggesting that the IMMPS was an independent prognostic risk factor for evaluating the overall survival of LUAD patients. Since BTNL9 was the most relevant immune checkpoint gene, in vitro experiment showed that the expression of the seven key genes (SEMA7A, EFHD2, CHST11, SLC24A4, MAL, JCHAIN, and SCARF1) in LUAD cell lines was consistent with that in normal lung epithelial cells after inhibiting BTNL9 expression (p < 0.05). Conclusions Our results contributed to a better understanding of immunological characteristics of LUAD. The IMMPS could serve as a promising tool for improving the clinical outcome of patients suffering from LUAD.
Collapse
Affiliation(s)
- Zhen Chen
- Department of Cardiothoracic Surgery, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
| | - Yongjun Zhang
- Department of Cardiothoracic Surgery, Xiangyang Central Hospital, Xiangyang, China
| |
Collapse
|
34
|
Cai L, Zhang S, Zheng F, Ji F, Wang J, Shi L, Chao L, Wang X, Zhang J, Chen Z. Identification of SCAMP2 as a regulator of NOTCH signaling in cisplatin resistance through a novel prognostic model for bladder cancer. Front Immunol 2025; 16:1573412. [PMID: 40406117 PMCID: PMC12095277 DOI: 10.3389/fimmu.2025.1573412] [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: 02/09/2025] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
Introduction Bladder cancer remains a major challenge in clinical oncology, particularly due to the development of platinum resistance, which severely impacts patient prognosis. Despite numerous attempts to create effective prognostic models, their clinical applicability has often been limited. Methods In this study, we utilized a robust statistical approach, LASSO-COX regression analysis, to develop a novel prognostic model for bladder cancer based on cisplatin sensitivity-related genes (CSRGs). The model was validated using both the TCGA-BLCA dataset and an independent validation set, GSE32894. Additionally, we employed various in vitro assays, including CCK-8 and EdU assays for cell proliferation, transwell assays for migration, and flow cytometry for apoptosis analysis, to investigate the biological function of the identified genes. Results Our prognostic model demonstrated superior predictive performance, with high AUC values. SCAMP2 was identified as a critical gene with elevated expression in bladder cancer, showing strong correlation with sensitivity to multiple anti-cancer drugs, including cisplatin. Further functional assays revealed that SCAMP2 mediates drug resistance in bladder cancer cells via the NOTCH signaling pathway. Additionally, in vivo experiments showed that SCAMP2 overexpression significantly enhanced cisplatin sensitivity in bladder cancer tissues. Discussion These findings underscore the potential of CSRGs, particularly SCAMP2, as critical biomarkers for bladder cancer prognosis. The identification of SCAMP2 as a regulator of NOTCH signaling in cisplatin resistance offers new insights into the molecular mechanisms of chemotherapy resistance and suggests potential therapeutic targets for overcoming drug resistance. Our model could guide personalized treatment strategies and improve bladder cancer patient outcomes.
Collapse
Affiliation(s)
- Longjun Cai
- Department of Urology, The Affiliated Suqian Hospital of Xuzhou Medical University, Nanjing Drum-Tower Hospital Group Suqian Hospital, Suqian, Jiangsu, China
| | - Shaoqi Zhang
- Department of Urology, The Affiliated Suqian Hospital of Xuzhou Medical University, Nanjing Drum-Tower Hospital Group Suqian Hospital, Suqian, Jiangsu, China
| | - Fangfang Zheng
- Department of Urology, The Affiliated Suqian Hospital of Xuzhou Medical University, Nanjing Drum-Tower Hospital Group Suqian Hospital, Suqian, Jiangsu, China
| | - Furong Ji
- Department of Urology, The Affiliated Suqian Hospital of Xuzhou Medical University, Nanjing Drum-Tower Hospital Group Suqian Hospital, Suqian, Jiangsu, China
| | - Jin Wang
- School of Public Health, Suzhou Medicine College of Soochow University, Suzhou, Jiangsu, China
| | - Long Shi
- Department of Urology, The Affiliated Suqian Hospital of Xuzhou Medical University, Nanjing Drum-Tower Hospital Group Suqian Hospital, Suqian, Jiangsu, China
| | - Liu Chao
- Department of Urology, The Affiliated Suqian Hospital of Xuzhou Medical University, Nanjing Drum-Tower Hospital Group Suqian Hospital, Suqian, Jiangsu, China
| | - Xiangyu Wang
- Department of Urology, The Affiliated Suqian Hospital of Xuzhou Medical University, Nanjing Drum-Tower Hospital Group Suqian Hospital, Suqian, Jiangsu, China
| | - Jianjun Zhang
- Department of Urology, The Affiliated Suqian Hospital of Xuzhou Medical University, Nanjing Drum-Tower Hospital Group Suqian Hospital, Suqian, Jiangsu, China
| | - Zhiyong Chen
- Department of Urology, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian, China
| |
Collapse
|
35
|
Guo X, Bai J, Wang X, Guo S, Shang Z, Shao Z. Evoking the Cancer-immunity cycle by targeting the tumor-specific antigens in Cancer immunotherapy. Int Immunopharmacol 2025; 154:114576. [PMID: 40168803 DOI: 10.1016/j.intimp.2025.114576] [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: 02/08/2025] [Revised: 03/17/2025] [Accepted: 03/27/2025] [Indexed: 04/03/2025]
Abstract
Cancer-related deaths continue to rise, largely due to the suboptimal efficacy of current treatments. Fortunately, immunotherapy has emerged as a promising alternative, offering new hope for cancer patients. Among various immunotherapy approaches, targeting tumor-specific antigens (TSAs) has gained particular attention due to its demonstrated success in clinical settings. Despite these advancements, there are still gaps in our understanding of TSAs. Therefore, this review explores the life cycle of TSAs in cancer, the methods used to identify them, and recent advances in TSAs-targeted cancer therapies. Enhancing medical professionals' understanding of TSAs will help facilitate the development of more effective TSAs-based cancer treatments.
Collapse
Affiliation(s)
- Xiaomeng Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Junqiang Bai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xinmiao Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Shutian Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhengjun Shang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial-Head and Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Zhe Shao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Day Surgery Center, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
36
|
Duan A, Zheng Y, Xiao G. Machine learning developed regulatory T cells-related signature for prognosis and immunotherapy benefit in oral squamous cell carcinoma. Am J Otolaryngol 2025; 46:104670. [PMID: 40398107 DOI: 10.1016/j.amjoto.2025.104670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 05/02/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is one of the most common malignancies with poor clinical outcome. Regulatory T cells (Tregs) have a dual role in maintaining immune homeostasis and suppressing anti-tumor immunity. The role of Tregs related genes (TRGs) in the prognosis of OSCC patients were rarely been reported. METHODS Integrative analysis procedure containing 10 machine learning methods was used to develop a Tregs related gene signature (TRS) using datasets from TCGA, GSE41613, GSE65858, and GSE117973 cohort. Several predicting scores were used to investigate the performance of TRS in predicting immunotherapy benefit. The biological function of TNFAIP3 was explored by in vitro assay. RESULTS The prognostic signature constructed using the LASSO algorithm was identified as the optimal TRS with the highest average C-index of 0.78. TRS served as a prognostic biomarker, with low TRS score correlating with favorable clinical outcome. Specifically, in TCGA dataset, the 1-, 3-, and 5-year AUC values were 0.796, 0.838 and 0.812, respectively. OSCC with low TRS score exhibited higher levels of immune-activated cells and immune-related functions, including CD8+ T, macrophage M1, and cytolytic activity. Low TRS indicated higher PD1&CTLA4 immunophenoscores, higher TMB, higher MSI, lower tumor immune dysfunction and exclusion score, decreased immune escape score, and lower intratumor heterogeneity scores. Additionally, OSCC cases with high TRS score showed elevated cancer-related hallmark score. Knock-down of TNFAIP3 inhibited OSCC cell proliferation. CONCLUSION This research established an optimal TRS for OSCC, which functions as a valuable indicator for forecasting clinical outcomes and assessing potential benefits from immunotherapy.
Collapse
Affiliation(s)
- Azhu Duan
- Department of stomatology, Shanghai Children's Hospital, Shanghai 200040, China
| | - Yicai Zheng
- Department of Stomatology, Shanghai Fifth People's Hospital, Fudan university, Shanghai 200240, China
| | - Guoxiu Xiao
- Department of Stomatology, Shanghai Fifth People's Hospital, Fudan university, Shanghai 200240, China.
| |
Collapse
|
37
|
Yin Y, Luo M. Lactylation-related risk model for prognostication and therapeutic responsiveness in uterine corpus endometrial carcinoma. Discov Oncol 2025; 16:677. [PMID: 40327181 PMCID: PMC12055729 DOI: 10.1007/s12672-025-02524-0] [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: 04/28/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Uterine corpus endometrial carcinoma (UCEC) is a prevalent gynecological cancer characterized by varied clinical outcomes and responses to treatment. Developing effective prognostic models is essential for guiding clinical decision-making. Recent research indicates that lactylation-a process impacting gene expression and immune responses-can affect tumor growth, metastasis, and immune evasion through histone modification. This study introduces a lactylation-related risk model aimed at predicting UCEC prognosis and providing insights into treatment efficacy. METHODS We analyzed transcriptomic data from The Cancer Genome Atlas (TCGA) for UCEC patients and identified two distinct lactylation-related patterns using consensus clustering. A risk model developed using Cox and Lasso regression has been studied for its ability to predict prognosis, immune cell infiltration, and treatment response. Additionally, we investigated the relationship between IGSF1 gene expression and clinical features. Gene Set Enrichment Analysis (GSEA) was performed to explore the function of the IGSF1 gene. RESULTS Two distinct lactylation-related clusters were identified, along with 156 differentially expressed genes between these clusters that are associated with the prognosis of UCEC. A risk model was developed based on three genes: IGSF1, ZFHX4, and SCGB2A1. This model effectively predicts clinical characteristics of UCEC patients, including immune cell infiltration, genetic variations, drug sensitivity, and response to immunotherapy. Notably, IGSF1 is linked to poor prognosis and is associated with immune activity, tumorigenesis, and cancer metabolism. CONCLUSIONS This study demonstrates that the lactylation-related risk model plays a crucial role in predicting prognosis and the efficacy of immunotherapy in UCEC, offering valuable insights for personalized treatment approaches.
Collapse
Affiliation(s)
- Yupeng Yin
- Department of Obstetrics and Gynecology, General Hospital of Southern Theatre Command, Guangzhou, 510010, China
| | - Min Luo
- Department of Obstetrics and Gynecology, General Hospital of Southern Theatre Command, Guangzhou, 510010, China.
- The First Clinical Medical College, Southern Medical University, Guangzhou, China.
| |
Collapse
|
38
|
Li Y, Song C, Wang H, Di W, Chen Y, Hu Y, Li P, Chen J, Ren Y, Gong J, Wang Q. Novel prognostic biomarkers in small cell lung cancer reveal mutational signatures, genomic mutations, and immune implications. Sci Rep 2025; 15:15592. [PMID: 40320401 PMCID: PMC12050310 DOI: 10.1038/s41598-025-00222-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 04/25/2025] [Indexed: 05/08/2025] Open
Abstract
Small cell lung cancer (SCLC) is a highly malignant lung cancer subtype with a dismal prognosis and limited treatment options. This study aimed to identify new prognostic molecular biomarkers for SCLC and explore their immune-related implications for treatment strategies. We analyzed 200 SCLC samples via whole-exome sequencing (WES) and 313 samples by targeted sequencing. A smoking-related SBS4 mutational signature was linked to poorer prognosis and lower tumor mutational burden (TMB), while the APOBEC-mediated SBS13 signature was associated with better prognosis and higher TMB. We identified a molecular subtype with the worst outcomes and lowest TMB in both cohorts. Among 38 high-frequency mutated genes associated with SCLC prognosis, only UNC13A mutations were beneficial. Patients with UNC13A mutations had favorable immune infiltration and tumor immunogenicity. Additionally, TP53 splice site mutations were related to the worst survival outcomes. In conclusion, we discovered new molecular biomarkers for SCLC prognosis. Our findings on their immunological characteristics offer insights for developing novel SCLC treatment strategies.
Collapse
Affiliation(s)
- Yuting Li
- Department of Radiation Oncology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453100, China
| | - Chen Song
- Department of Hematology Laboratory, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453100, China
| | - Haijun Wang
- Department of Pathology, Xinxiang Key Laboratory of Precision Medicine, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453100, China
| | - Wenyu Di
- Department of Pathology, Xinxiang Key Laboratory of Precision Medicine, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453100, China
| | - Yangyang Chen
- Department of Radiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453100, China
| | - Yuanyuan Hu
- Department of Radiation Oncology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453100, China
| | - Peiheng Li
- Department of Radiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453100, China
| | - Jie Chen
- Department of Radiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453100, China
| | - Yanfeng Ren
- Department of Health Statistics, Key Laboratory of Medicine and Health of Shandong Province, School of Public Health, Shandong Second Medical University, Baotong Xi Street, Weicheng District, Weifang, 261053, Shandong, China.
| | - Jing Gong
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Qinghua Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453100, China.
- Department of Health Statistics, Key Laboratory of Medicine and Health of Shandong Province, School of Public Health, Shandong Second Medical University, Baotong Xi Street, Weicheng District, Weifang, 261053, Shandong, China.
| |
Collapse
|
39
|
Tian T, Han H, Huang J, Ma J, Ran R. DBI as a Novel Immunotherapeutic Candidate in Colorectal Cancer: Dissecting Genetic Risk and the Immune Landscape via GWAS, eQTL, and pQTL. Biomedicines 2025; 13:1115. [PMID: 40426943 PMCID: PMC12109284 DOI: 10.3390/biomedicines13051115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 04/24/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Colorectal cancer (CRC) is a leading cause of cancer-related mortality worldwide. Identifying drug targets associated with CRC is crucial for developing targeted therapies. Methods: MR (IVW, Wald ratio, weighted median, and MR-Egger) and SMR analyses were used to screen candidate genes associated with CRC risk. Further validation was performed using The Cancer Genome Atlas (TCGA) to assess gene expression patterns and prognostic significance. Additionally, immune infiltration analysis was conducted to characterize the tumor immune microenvironment. Drug prediction was performed to explore potential therapeutic interventions. Results: Eight genes were identified associated with CRC. IGFBP3, CD72, SERPINH1, CHRDL2, LRP11, and SPARCL1 were linked to an increased risk of CRC, whereas DBI and HYAL1 were associated with a decreased risk of CRC. Notably, DBI exhibited a potentially favorable immune profile, negatively correlated with Tregs and MDSCs while positively associated with activated CD8+ and CD4+ T cells. Conclusions: Eight genes were identified as associated with CRC, among which DBI exhibited a potential protective role, correlating with improved patient survival, enhanced immune activation, and increased responsiveness to immunotherapy. The remaining proteins demonstrated diverse and complex functions within the tumor immune microenvironment, providing novel insights for the development of precision diagnostics and immunotherapeutic strategies.
Collapse
Affiliation(s)
- Ting Tian
- Department of Clinical Laboratory, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430070, China; (T.T.); (J.M.)
| | - Huan Han
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China; (H.H.); (J.H.)
| | - Jingtao Huang
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China; (H.H.); (J.H.)
| | - Jun’e Ma
- Department of Clinical Laboratory, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430070, China; (T.T.); (J.M.)
| | - Ruoxi Ran
- Department of Clinical Laboratory, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430070, China; (T.T.); (J.M.)
| |
Collapse
|
40
|
Xie Y, Wang F, Wei J, Shen Z, Song X, Wang Y, Chen H, Tao L, Zheng J, Lin L, Niu Z, Guan X, Zhou T, Xu Z, Liu Y, Du D, Pan H, Li S, Ji W, Zhou W, Yang Y, Tian J, Xu J, Hu H, Liang X. Noninvasive prognostic classification of ITH in HCC with multi-omics insights and therapeutic implications. SCIENCE ADVANCES 2025; 11:eads8323. [PMID: 40315307 PMCID: PMC12047409 DOI: 10.1126/sciadv.ads8323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 03/31/2025] [Indexed: 05/04/2025]
Abstract
Intratumoral heterogeneity (ITH) is a critical factor associated with treatment failure and disease relapse in hepatocellular carcinoma (HCC). However, decoding ITH in a noninvasive and comprehensive manner remains a notable challenge. In this study involving 851 patients from five centers, we developed a noninvasive prognostic classification for ITH using radiomics based on multisequence MRI, termed radiomics ITH (RITH) phenotypes. The RITH phenotypes highly correlated with prognosis and pathological ITH. In addition, through an integrated multi-omics analysis, we uncovered the molecular mechanisms underlying RITH, notably enhancing its biological interpretability. Specifically, high-RITH tumors demonstrated an enrichment of cancer-associated fibroblasts and activation of extracellular matrix remodeling. Our approach facilitates the noninvasive refined classification of ITH using radiomics and multi-omics, paving the way for tailored treatment strategies in HCC. Extracellular matrix-receptor interaction could be a potential therapeutic target in patients with high-RITH tumors. Given the routine use of radiologic imaging in oncology, our methodology ignites versatile framework for broader application to other solid tumors.
Collapse
Affiliation(s)
- Yangyang Xie
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Fang Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, 430022 Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, 430022 Wuhan, China
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
| | - Jingwei Wei
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, 100190 Beijing, China
- Beijing Key Laboratory of Molecular Imaging, 100190 Beijing, China
| | - Zefeng Shen
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Xue Song
- Department of Respiratory and Critical Care Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 310007 Hangzhou, China
| | - Yali Wang
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Hongjun Chen
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Liye Tao
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Junhao Zheng
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Lanfen Lin
- The College of Computer Science and Technology, Zhejiang University, 310027 Hangzhou, China
| | - Ziwei Niu
- The College of Computer Science and Technology, Zhejiang University, 310027 Hangzhou, China
| | - Xiaojun Guan
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009 Hangzhou, China
| | - Tianhan Zhou
- Department of General Surgery, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 310007 Hangzhou, China
| | - Zhengao Xu
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Yang Liu
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Danwei Du
- Department of Anorectal, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 310000 Hangzhou, China
| | - Haoyu Pan
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Shihao Li
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Wenbin Ji
- Department of Radiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, 317000 Taizhou, China
| | - Wei Zhou
- Department of Radiology, Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, 313000 Huzhou, China
| | - Yunjun Yang
- Department of Radiology, The First Affiliated Hospital, Wenzhou Medical University, 325006 Wenzhou, China
| | - Jie Tian
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, 100190 Beijing, China
- Beijing Key Laboratory of Molecular Imaging, 100190 Beijing, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 100191 Beijing, China
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, 710126 Xi’an, China
| | - Junjie Xu
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Hongjie Hu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
| | - Xiao Liang
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- School of Medicine, Shaoxing University, 312000 Shaoxing, China
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, 310000 Hangzhou, China
| |
Collapse
|
41
|
Liu Z, Jiang X, Ke Z, Wang W, Tang J, Dai Y. PAR2 deficiency impairs antitumor immunity and attenuates anti-PD1 efficacy in colorectal cancer. Pharmacol Res 2025; 215:107721. [PMID: 40174816 DOI: 10.1016/j.phrs.2025.107721] [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: 11/05/2024] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025]
Abstract
A T cell-inflamed tumor microenvironment is predictive of better prognosis and clinical response to immunotherapy. Proteinase-activated receptor 2 (PAR2), a member of G-protein coupled receptors is involved in inflammatory process and the progression of various cancers. However, the role of PAR2 in modulating the tumor microenvironment remains unclear. Here, we found that PAR2 high-expression was associated with a favorable prognosis in patients with colorectal cancer. Intriguingly, PAR2 expression in human colorectal cancer was mainly confined to tumor cells and was significantly associated with CD8+ T cell infiltration. Tumor-intrinsic PAR2 deficiency blunted antitumor immune responses to promote tumor growth and attenuated the therapeutic efficacy of anti-PD1 in a mouse model of colon cancer. Tumors with downregulated PAR2 showed decreased CD8+ T cell infiltration and impaired effector function. Mechanistically, PAR2 activation in tumor cells induced CXCL9 and CXCL10 production via PI3K/AKT/mTOR signaling, thereby enhancing CD8+ T cell recruitment in the tumor microenvironment. In addition, PAR2 was essential for dendritic cell activation and differentiation towards conventional type 1 subset. PAR2 deficiency in dendritic cells markedly impaired their ability to prime CD8+ T cells and control tumor growth in vivo. Thus, our findings identify new roles for PAR2 in promoting antitumor immunity and provide a promising target to improve immunotherapy efficacy in colorectal cancer.
Collapse
Affiliation(s)
- Zilin Liu
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Xuehui Jiang
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Ziliang Ke
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Weihong Wang
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Jianqiang Tang
- Department of General Surgery, Peking University First Hospital, Beijing, China.
| | - Yun Dai
- Department of Gastroenterology, Peking University First Hospital, Beijing, China.
| |
Collapse
|
42
|
Liu G, Pan LZ, Chen J, Ma J. Unveiling the role of PANoptosis-related genes in breast cancer: an integrated study by multi-omics analysis and machine learning algorithms. Breast Cancer Res Treat 2025; 211:35-50. [PMID: 39870964 DOI: 10.1007/s10549-025-07620-x] [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: 06/23/2024] [Accepted: 01/16/2025] [Indexed: 01/29/2025]
Abstract
BACKGROUND The heterogeneity of breast cancer (BC) necessitates the identification of novel subtypes and prognostic models to enhance patient stratification and treatment strategies. This study aims to identify novel BC subtypes based on PANoptosis-related genes (PRGs) and construct a robust prognostic model to guide individualized treatment strategies. METHODS The transcriptome data along with clinical data of BC patients were sourced from the TCGA and GEO databases. Consensus clustering was performed on 12 PRGs to ascertain potential BC subtypes, and variances in survival, infiltration of immune cells, and functional pathways among them were examined. A prognostic model was generated through 101 combinations of machine learning algorithms and validated across multiple cohorts. The response of patients towards immunotherapy were analyzed using multiple frameworks. RESULTS Consensus clustering of 12 PRGs identified two distinct BC subtypes, with subtype B exhibiting significantly lower overall survival (OS) rates compared to subtype A. Immune cell infiltration analysis revealed higher immune activity in subtype A. Functional pathway analysis revealed that subtype A exhibited a significant enrichment in immune-related pathways, while subtype B was associated with cell cycle and metabolic processes. An integrated machine learning framework integrating CoxBoost and Random Survival Forest (RSF) algorithms was developed, demonstrating high predictive performance across multiple cohorts. A nomogram combining age and risk score was constructed, showing excellent predictive performance. Immune landscape analysis revealed that the high-risk group exhibited a suppressive tumor immune microenvironment (TIME). Immunotherapy response prediction suggested that low-risk patients were more likely to benefit from PD-1 and CTLA-4 inhibitors. CONCLUSIONS Our study provides a comprehensive framework for BC subtype classification and prognostic prediction, offering valuable insights for personalized treatment strategies.
Collapse
Affiliation(s)
- Gang Liu
- Department of Thyroid and Breast Surgery, The People's Hospital of Suzhou New District, Suzhou, China
| | - Liang-Zhi Pan
- Party Committee Office, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
| | - Jie Chen
- Department of Internal Medicine, Huangshi Maternal and Child Health Hospital, Huangshi, China
| | - Jianying Ma
- Department of Breast Surgery, Thyroid Surgery, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, No.141, Tianjin Road, Huangshi, 435000, Hubei, China.
| |
Collapse
|
43
|
Liu B, Tao W, Zhou X, Xu LD, Luo Y, Yang X, Min Q, Huang M, Zhu Y, Cui X, Wang Y, Gong T, Zhang E, Huang YS, Chen W, Yan S, Wu N. Multi‑omics analysis identifies different molecular subtypes with unique outcomes in early-stage poorly differentiated lung adenocarcinoma. Mol Cancer 2025; 24:129. [PMID: 40312720 PMCID: PMC12044723 DOI: 10.1186/s12943-025-02333-7] [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: 02/22/2025] [Accepted: 04/12/2025] [Indexed: 05/03/2025] Open
Abstract
INTRODUCTION Early-stage poorly differentiated lung adenocarcinoma (LUAD) is plagued by a high risk of postoperative recurrence, and its prognostic heterogeneity complicates treatment and surveillance planning. We conducted this integrative multi-omics study to identify those patients with a truly high risk of adverse outcomes. METHODS Whole-exome, RNA and whole methylome sequencing were carried out on 101 treatment-naïve early-stage poorly differentiated LUADs. Integrated analyses were conducted to disclose molecular characteristics and explore molecular subtyping. Functional validation of key molecules was carried out through in vitro and in vivo experiments. RESULTS Recurrent tumors exhibited significantly higher ploidy (p = 0.024), the fraction of the genome altered (FGA, p = 0.042), and aneuploidy (p < 0.05) compared to non-recurrent tumors, as well as a higher frequency of CNVs. Additionally, recurrent tumors showed hypomethylation at both the global level and in CpG island regions. Integrative transcriptomic and methylation analyses identified three molecular subtypes (C1, C2, and C3), with the C1 subtype presenting the worst prognosis (p = 0.024). Although frequently mutated genes showed similar mutation frequencies across the three subtypes, the C1 subtype exhibited the highest tumor mutation burden (TMB), mutant-allele tumor heterogeneity (MATH), aneuploidy, and HLA loss of heterozygosity (HLA-LOH), along with relatively lower immune cell infiltration. Furthermore, GINS1 and CPT1C were found to promote LUAD progression, and their high expression correlated with a poor prognosis. CONCLUSIONS This multi-omics study identified three integrative subtypes with distinct prognostic implications, paving the way for more precise management and postoperative monitoring of early-stage poorly differentiated LUAD.
Collapse
Affiliation(s)
- Bing Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Wei Tao
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Xuantong Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Li-Di Xu
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Yanrui Luo
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Xin Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Qingjie Min
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Miao Huang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yuge Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xinrun Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yaqi Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Tongyang Gong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Enli Zhang
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Yu S Huang
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Weizhi Chen
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Shi Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Nan Wu
- State Key Laboratory of Molecular Oncology, Frontiers Science Center for Cancer Integrative Omics, Department of Thoracic Surgery II, Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
- Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Yunnan, China.
| |
Collapse
|
44
|
Chen G, Zheng Z, Ji Q, He R, Pan Z, Chen Y, Zhou Y, Wei Z, Sun H, Feng L. Tumor innervation in cervical cancer: Prognostic insights from myelin-associated risk signatures. FASEB Bioadv 2025; 7:e70004. [PMID: 40330434 PMCID: PMC12050960 DOI: 10.1096/fba.2024-00190] [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: 10/21/2024] [Revised: 12/15/2024] [Accepted: 02/10/2025] [Indexed: 05/08/2025] Open
Abstract
The reported frequencies of perineural invasion (PNI) in human cervical cancer, ranging from 7.0% to 35.1%, may underestimate the significant role that nerves play in cervical cancer progression. Neurosecretory factors can promote tumor migration and invasion, even in cases classified as "PNI-negative". This study aimed to clarify whether tumor innervation influences tumor progression and cervical cancer patient outcomes. We first evaluated the gene signatures of human myelinating Schwann cells (SCs) using the Inferring Pathway Activity and Suppression (IPAS) scoring system to predict the degree of tumor innervation in 304 cervical cancer patients from The Cancer Genome Atlas (TCGA) database. Subsequently, we constructed a myelin-associated risk prognostic signature using LASSO regression analysis. Finally, we obtained a risk score using a quantitative formula and categorized all samples into high- and low-risk score groups. Our results indicated that tumor innervation in cervical cancer is associated with poor patient survival. Higher levels of innervation were correlated with an impaired immune response and reduced expression of immune checkpoints, including PD-L1. The prognostic model demonstrated excellent consistency between predicted and actual survival outcomes. Overall, tumor innervation plays a crucial role in regulating cervical cancer prognosis. The identified prognostic risk signatures offer a valuable tool for risk stratification and prognostic prediction in clinical practice.
Collapse
Affiliation(s)
- Guoqiang Chen
- Department of GynecologyThe People's Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen UniversityShenzhenChina
| | - Zhen Zheng
- Department of Obstetrics and GynecologyNational Clinical Research Centre for Obstetric and Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Qingqing Ji
- Department of AnesthesiologyShidong Hospital Affiliated to University of Shanghai for Science and TechnologyShanghaiChina
| | - Ruihua He
- Department of PharmacyShanghai Fourth People's Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Zhouyuan Pan
- Department of GynecologyThe People's Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen UniversityShenzhenChina
| | - Yunxia Chen
- Department of GynecologyThe People's Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen UniversityShenzhenChina
| | - Yuqing Zhou
- Department of GynecologyThe People's Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen UniversityShenzhenChina
| | - Zhihong Wei
- Department of GynecologyThe People's Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen UniversityShenzhenChina
| | - Hao Sun
- Department of Obstetrics and GynecologyShanghai Changzheng Hospital of Naval Medical UniversityShanghaiChina
| | - Lixia Feng
- Department of GynecologyThe People's Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen UniversityShenzhenChina
| |
Collapse
|
45
|
Shang J, Zhou C, He M, Huang XY, Qin CF, Wu A. Mutation S139N on Zika virus prM protein shifts immune response from Asian to contemporary strain. Brain Behav Immun 2025; 126:247-259. [PMID: 39986659 DOI: 10.1016/j.bbi.2025.02.012] [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: 08/11/2024] [Revised: 01/26/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025] Open
Abstract
Zika virus (ZIKV) has been associated with neurological diseases like microcephaly and Guillain-Barré syndrome. The S139N single mutation on the prM protein of the FSS13025 Asian strain increases the mortality rate in mice. Therefore, it is a valuable tool for studying the impact of immune responses on neural damage. Here, we used single-cell sequencing technology to systematically assess the immune response induced by three ZIKV strains: Asian ancestral strain FSS13025/2010, FSS13025 strain with S139N mutation (FSS13025-S139N), and contemporary strain GZ01/2016. By infecting 1-day-old mice, we observed that the immune spectrum elicited by FSS13025-S139N mutant resembled that induced by the contemporary strain. The FSS13025-S139N strain induces the proliferation of inflammatory microglial cells earlier than the FSS13025 strain, similar to GZ01. A specific cell cluster, Microglia_Ccr7, was induced by the S139N mutant strain and GZ01 strain, which suppresses T cell activation through the PDCD1LG2-PDCD1 signaling pathway. Furthermore, the proliferation of CD8+ T cells was weakened and prolonged in S139N strain-infected samples. Finally, we found that the S139N mutant strain causes more apoptosis of neurons compared to the FSS13025 strain. These results indicate that the S139N mutation plays an important role in the immune response pattern of ZIKV and prolongs the duration of neuroinflammation.
Collapse
Affiliation(s)
- Jingzhe Shang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123 Jiangsu, China; Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China
| | - Chao Zhou
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences (AMMS), Beijing 100071, China
| | - Mengjiao He
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences (AMMS), Beijing 100071, China
| | - Xing-Yao Huang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences (AMMS), Beijing 100071, China
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences (AMMS), Beijing 100071, China.
| | - Aiping Wu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123 Jiangsu, China; Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China.
| |
Collapse
|
46
|
Zeng J, Zhang R, Xu H, Zhang C, Lu L. Integrative single-cell RNA sequencing and mendelian randomization analysis reveal the potential role of synaptic vesicle cycling-related genes in Alzheimer's disease. J Prev Alzheimers Dis 2025; 12:100097. [PMID: 40021385 DOI: 10.1016/j.tjpad.2025.100097] [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: 12/25/2024] [Revised: 02/15/2025] [Accepted: 02/17/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) involves alterations in synaptic vesicle cycling (SVC), which significantly affect neuronal communication and function. Therefore, a thorough investigation into the potential roles of SVC-related genes (SVCRGs) in AD can enhance the identification of critical biomarkers that may influence disease progression and treatment responses. METHODS The datasets used in this study were sourced exclusively from public databases. By integrating differential expression analysis with Mendelian randomization (MR), we identified SVCRGs as biomarkers for AD. Functional characterization of these biomarkers was performed, followed by integration into a nomogram. Further investigation of immune infiltration in AD patients and healthy individuals was carried out. Ultimately, the potential cellular mechanisms of AD were explored through single-cell RNA sequencing (scRNA-seq) analysis. RESULTS ATP6V1D, ATP6V1G2, CLTB, and NSF were identified as biomarkers, exhibiting a positive correlation with each other and a downregulated expression in AD. These markers were pinpointed as protective factors for AD [odds ratio (OR) < 1, P < 0.05], with potential to reduce the risk of the disease. Integrated into a nomogram, they demonstrated satisfactory diagnostic performance and clinical utility, surpassing the use of single gene. They were collectively enriched in pathways related to "interferon gamma response", "inflammatory response", and "TNFα signaling via NFκB". Additionally, an increase in infiltration of 17 immune cell types in AD was noted, particularly cells associated with neuroinflammation such as activated CD8 T cells and various dendritic cells (DCs), suggesting an inflammatory milieu in AD while also displaying a negative correlation with the biomarkers. The cell types were further annotated, revealing specific expressions of biomarkers and uncovering the heterogeneity of excitatory neurons. A significant reduction in the overall number of excitatory neurons under AD conditions was observed, alongside consistent expression of biomarkers during the developmental stages of excitatory neurons. CONCLUSION By using MR, we firstly identified four SVCRGs as protective factors for AD, functioning through pathways associated with mitochondrial dysfunction, chronic inflammation, immune dysregulation, and neuronal damage. These genes had the potential to modulate immune cell infiltration activated in AD patients and exhibited cell-type-specific expression profiles within AD-related cellular contexts. Their findings provide novel insights and valuable references for future research on AD pathogenesis and therapeutic strategies.
Collapse
Affiliation(s)
- Junfeng Zeng
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Ruihua Zhang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Huihua Xu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Chengwu Zhang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, Shanxi, China.
| | - Li Lu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Key Laboratory of Cellular Physiology of Chinese Ministry of Education, Shanxi Medical University, Taiyuan 030001, Shanxi, China.
| |
Collapse
|
47
|
Lai G, Xie B, Zhang C, Zhong X, Deng J, Li K, Liu H, Zhang Y, Liu A, Liu Y, Fan J, Zhou T, Wang W, Huang A. Comprehensive analysis of immune subtype characterization on identification of potential cells and drugs to predict response to immune checkpoint inhibitors for hepatocellular carcinoma. Genes Dis 2025; 12:101471. [PMID: 40092490 PMCID: PMC11907441 DOI: 10.1016/j.gendis.2024.101471] [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: 10/17/2023] [Revised: 04/12/2024] [Accepted: 11/02/2024] [Indexed: 03/19/2025] Open
Abstract
Immunosubtyping enables the segregation of immune responders from non-responders. However, numerous studies failed to focus on the integration of cellular heterogeneity and immunophenotyping in the prediction of hepatocellular carcinoma (HCC) patients' response to immune checkpoint inhibitors (ICIs). We categorized HCC patients into various immune subtypes based on feature scores linked to ICI response. Single-cell sequencing technology was to investigate the cellular heterogeneity of different immune subtypes and acquire significant ICI response-associated cells. Candidate drugs were identified using a blend of various drug databases and network approaches. HCC patients were divided into two distinct immune subtypes based on characterization scores of 151 immune-related gene sets. Patients in both subtypes showed varying overall survival, immunity levels, biological activities, and TP53 mutation rates. Subtype 1-related natural killer cells showed a positive correlation with immune-promoting scores but a negative correlation with immune-suppressing scores. Notably, docetaxel sensitivity in HCC patients rose as the levels of subtype 1-related natural killer cells increased. Our study demonstrated that immune subtypes have cellular heterogeneity in predicting response to ICIs. A combination of subtype 1-associated natural killer cells and docetaxel may offer new hope for ICI treatment in HCC.
Collapse
Affiliation(s)
- Guichuan Lai
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Biao Xie
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Cong Zhang
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Xiaoni Zhong
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Jielian Deng
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Kangjie Li
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Hui Liu
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Yuan Zhang
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Anbin Liu
- Department of Applied Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Yi Liu
- Department of Applied Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Jie Fan
- Department of Epidemiology, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Tianyi Zhou
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Wei Wang
- Department of Applied Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Ailong Huang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
48
|
Tuo Z, Gao M, Jiang C, Zhang D, Chen X, Jiang Z, Wang J. Construction of M2 macrophage-related gene signature for predicting prognosis and revealing different immunotherapy response in bladder cancer patients. Clin Transl Oncol 2025; 27:2191-2206. [PMID: 39347941 DOI: 10.1007/s12094-024-03698-9] [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: 06/12/2024] [Accepted: 08/22/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Bladder cancer development is closely associated with the dynamic interaction and communication between M2 macrophages and tumor cells. However, specific biomarkers for targeting M2 macrophages in immunotherapy remain limited and require further investigation. METHODS In this study, we identified key co-expressed genes in M2 macrophages and developed gene signatures to predict prognosis and immunotherapy response in patients. Public database provided the bioinformatics data used in the analysis. We created and verified an M2 macrophage-related gene signature in these datasets using Lasso-Cox analysis. RESULTS The predictive value and immunological functions of our risk model were examined in bladder cancer patients, and 158 genes were found to be significantly positively correlated with M2 macrophages. Moreover, we identified two molecular subgroups of bladder cancer with markedly different immunological profiles and clinical prognoses. The five key risk genes identified in this model were validated, including CALU, ECM1, LRP1, CYTL1, and CCDC102B, demonstrating the model can accurately predict prognosis and identify unique responses to immunotherapy in patients with bladder cancer. CONCLUSIONS In summary, we constructed and validated a five-gene signature related to M2 macrophages, which shows strong potential for forecasting bladder cancer prognosis and immunotherapy response.
Collapse
Affiliation(s)
- Zhouting Tuo
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Mingzhu Gao
- Department of Oncology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Chao Jiang
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Duobing Zhang
- Department of Urology, Suzhou Hospital of Anhui Medical University, Suzhou, 234000, China
- Department of Urology, Suzhou Municipal Hospital of Anhui Province, Suzhou, 234000, China
| | - Xin Chen
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Zhiwei Jiang
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| | - Jinyou Wang
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| |
Collapse
|
49
|
Wang T, Bai Y, Dong Y, Qin J, Zhou X, Wang A, Liu D, Li X, Ma Z, Hu Y. A comprehensive analysis of deubiquitinase USP20 on prognosis and immunity in pan-cancer. FASEB J 2025; 39:e70499. [PMID: 40270318 DOI: 10.1096/fj.202402603r] [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: 10/27/2024] [Revised: 02/05/2025] [Accepted: 03/20/2025] [Indexed: 04/25/2025]
Abstract
USP20 is a deubiquitinase enzyme in the ubiquitin-proteasome system that plays a role in the development and progression of tumors. However, the relationships between USP20 expression and clinical prognosis and tumor immunity remain unclear. In this study, the USP20 expression and its relationships with potential prognostic value, the tumor microenvironment (TME), immune-related genes, the tumor mutational burden (TMB), microsatellite instability (MSI), homologous recombination deficiency, cancer stemness, and correlated signaling pathways were investigated via The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), Cancer Cell Line Encyclopedia (CCLE), STRING, Gene Expression Profiling Interactive Analysis (GEPIA2), and the Human Protein Atlas (HPA). Moreover, we explored the oncogenic capability of USP20 in breast cancer. Data analysis was performed via GraphPad Prism and the R package. The results indicated that the expression of USP20 was upregulated in most cancers and was associated with survival in 17 tumor types. Furthermore, USP20 expression was strongly correlated with immune infiltration and the expression of immunomodulatory genes. We also verified the correlations between USP20 expression and tumor heterogeneity, cancer stemness, and the corresponding signaling pathways. Moreover, our work revealed that USP20 was highly expressed and predicted a poor outcome in patients with breast cancer. Basic experiments verified that USP20 overexpression promoted both the proliferation and migration of breast cancer cells. This study comprehensively investigated the expression of USP20 and its correlation with clinical prognostic assessment and tumor immune modulation across cancers, indicating that USP20 might have utility as a biomarker associated with prognosis and cancer immunotherapy.
Collapse
Affiliation(s)
- Ting Wang
- School of Medicine, Nankai University, Tianjin, China
- Department of Oncology, The First Medical Center of PLA General Hospital, Beijing, China
| | - Yibing Bai
- Department of Oncology, The First Medical Center of PLA General Hospital, Beijing, China
- Graduate School, Medical School of Chinese PLA, Beijing, China
| | - Yi Dong
- Department of Oncology, The First Medical Center of PLA General Hospital, Beijing, China
- Graduate School, Medical School of Chinese PLA, Beijing, China
| | - Jiapei Qin
- Department of Oncology, The First Medical Center of PLA General Hospital, Beijing, China
- Graduate School, Medical School of Chinese PLA, Beijing, China
| | - Xin Zhou
- Department of Oncology, The First Medical Center of PLA General Hospital, Beijing, China
- Graduate School, Medical School of Chinese PLA, Beijing, China
| | - An Wang
- Department of Oncology, The First Medical Center of PLA General Hospital, Beijing, China
- Graduate School, Medical School of Chinese PLA, Beijing, China
| | - Dong Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiaoyan Li
- Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Zhiqiang Ma
- Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Yi Hu
- School of Medicine, Nankai University, Tianjin, China
- Department of Oncology, The First Medical Center of PLA General Hospital, Beijing, China
- Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
50
|
Qiu Y, Wang Y, Liu J, Sun K, Liu B, Hou Q. Single-cell sequencing unveils the transcriptomic landscape of castration-resistant prostate cancer-associated fibroblasts and their association with prognosis and immunotherapy response. BMC Cancer 2025; 25:813. [PMID: 40307786 PMCID: PMC12044937 DOI: 10.1186/s12885-025-14212-x] [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: 10/04/2024] [Accepted: 04/23/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND The tumor microenvironment (TME) is increasingly acknowledged as a determinant in the malignant transformation and progression of castration-resistant prostate cancer (CRPC). Cancer-associated fibroblasts (CAFs), as a pivotal stromal cellular component in TME, are implicated in tumor progression and immune escape. However, the molecular characteristics and biological functions of CRPC-CAFs in prostate cancer necessitate further investigation. METHODS We ascertained the differential transcriptomic profiles between CRPC-CAFs and PCa-CAFs through single-cell RNA-sequencing (scRNA-seq). Bulk RNA-seq data were employed to assess the prognostic implications of CRPC-CAFs in PCa. In addition, we examined the impact of CRPC-CAFs on the efficacy of immunotherapy and the composition of the tumor immune milieu. Furthermore, a subcutaneous PCa model was applied to determine the potential of TGF-β signaling blockade to augment the response to immunotherapeutic interventions. RESULTS We observed a pronounced increase in the proportion of CAFs in CRPC compared to those in primary PCa. The functional pathways implicated in TGF-β signaling and ECM remodeling were remarkably upregulated in CRPC-CAFs. Moreover, gene regulatory network analysis uncovered substantial differences in the transcription factor activity profiles between CRPC-CAFs and PCa-CAFs. The elevated CRPC-CAFs abundance was associated with diminished recurrence-free survival and immunotherapy insensitivity. Substantially elevated infiltration of inhibitory immune cells and upregulated expression levels of immunosuppressive molecules were observed in patients with high CRPC-CAFs abundance. Importantly, administration of anti-TGF-β therapy remarkably potentiated the efficacy of anti-PD-1 immunotherapy through upregulating the anti-tumor immune response in the PCa model. CONCLUSION Our results highlighted the impact of CRPC-CAFs on clinical prognosis and immunosuppressive tumor milieu, indicating that CRPC-CAFs may function as a promising therapeutic target for CRPC.
Collapse
Affiliation(s)
- Yifeng Qiu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China
- International Cancer Center, Shenzhen Key Laboratory, Hematology Institution of Shenzhen University, Shenzhen, China
| | - Yuhan Wang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China
| | - Jiahe Liu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China
| | - Kai Sun
- Department of Radiology, the Third People's Hospital of Longgang District, Shenzhen, China
- Shenzhen Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, 518116, China
| | - Baohua Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China.
| | - Qi Hou
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China.
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China.
- International Cancer Center, Shenzhen Key Laboratory, Hematology Institution of Shenzhen University, Shenzhen, China.
| |
Collapse
|