1
|
Yang Z, Zhou P, Wang L, Yang X, Yang M, Xia J. High expression of HECW1 is associated with the poor prognosis and cancer progression of gastric cancer. World J Surg Oncol 2025; 23:204. [PMID: 40442732 PMCID: PMC12121130 DOI: 10.1186/s12957-025-03866-3] [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: 02/06/2025] [Accepted: 05/20/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND The E3 ubiquitin ligase HECW1 was found to be involved in ubiquitination modifications during malignant progression of multiple tumors. However, the prognostic role of HECW1 expression in gastric cancer (GC) remains unclear. METHODS The Tumor Immunoassay Resource (TIMER2.0) system evaluated the association of HECW1 with tumor-infiltrating lymphocytes in carcinomas. The UALCAN assessed HECW1 mRNA expression levels in GC tissues and examined their associations with clinicopathological characteristics. The Kaplan Meier-plotter analyzed the effect of HECW1 on the survival of GC patients. The cBioPortal retrieved information about genetic variants in HECW1 gene. Protein‒protein interaction (PPI) networks associated with HECW1 were explored using the STRING database. The functional effects of HECW1 on GC cells were evaluated through proliferation (Cell Counting Kit-8), apoptosis (Flow cytometry), and migration (Transwell and wound healing assays). The RNA-Seq was applied to explore the underlying mechanisms. RESULTS HECW1 demonstrated significant overexpression in GC tumor tissues, correlating with adverse clinical outcomes. Clinically, elevated HECW1 expression exhibited an inverse association with tumor-infiltrating CD8+ T lymphocytes while demonstrating a positive correlation with macrophages, DCs, and neutrophils infiltration, suggesting its potential involvement in tumor immune evasion mechanisms. Functional validation revealed that HECW1 knockdown markedly suppressed GC cell proliferation and migratory capacity, concurrently promoting apoptotic cell death. Mechanistic investigations identified that HECW1 exerts its oncogenic effects through dysregulation of the Hippo signaling pathway, with its silencing effectively attenuating tumor progression via pathway modulation. CONCLUSIONS HECW1 upregulation is significantly associated with poor prognosis and immune infiltration in GC patients, emphasizing its potential as a prognostic biomarker.
Collapse
Affiliation(s)
- Zhihui Yang
- Department of General Surgery, Institute of General Surgical Research, Jiangnan University Medical Center (Wuxi No.2 People's Hospital), School of Medicine, Wuxi, China
| | - Peng Zhou
- Department of General Surgery, People's Hospital of Nanjing Medical University, Wuxi, China
| | - Liping Wang
- Department of General Surgery, Institute of General Surgical Research, Jiangnan University Medical Center (Wuxi No.2 People's Hospital), School of Medicine, Wuxi, China
| | - Xiao Yang
- Department of General Surgery, Institute of General Surgical Research, Jiangnan University Medical Center (Wuxi No.2 People's Hospital), School of Medicine, Wuxi, China
| | - Mengqi Yang
- Department of General Surgery, People's Hospital of Nanjing Medical University, Wuxi, China
| | - Jiazeng Xia
- Department of General Surgery, Institute of General Surgical Research, Jiangnan University Medical Center (Wuxi No.2 People's Hospital), School of Medicine, Wuxi, China.
- Department of General Surgery, People's Hospital of Nanjing Medical University, Wuxi, China.
- Department of General Surgery, Institute of General Surgical Research, Jiangnan University Medical Center, 68 Zhongshan Lu, Wuxi, 214002, P.R. China.
| |
Collapse
|
2
|
Chen J, Zhai X, Kuang H, Zhang A. Prediction of neuroblastoma prognosis with a novel T-cell exhaustion-related gene signature. Sci Rep 2025; 15:17885. [PMID: 40404747 PMCID: PMC12098910 DOI: 10.1038/s41598-025-02661-0] [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: 08/06/2024] [Accepted: 05/15/2025] [Indexed: 05/24/2025] Open
Abstract
Neuroblastoma (NB) is the most common type of pediatric extra-cranial tumor that arises in the sympathetic nervous system. The heterogeneity of T-cell exhaustion (TEX) has been linked to the determination of distinct clinical outcomes and the effectiveness of immunotherapy in numerous adult malignancies. Therefore, studying the heterogeneous TEX landscape in NB as well as its impact on clinical outcomes is meaningful. The gene expression and clinical datasets of the Sequencing Quality Control (SEQC), E-MTAB-8248, and Therapeutically Applicable Research to Generate Effective Treatments (TARGET) cohorts were downloaded from publicly available databases. Two TEX-related clusters for neuroblastoma were identified in the SEQC cohort. Patients in TEX-C1 exhibited superior overall survival (OS) and event-free survival (EFS) rates compared with those in TEX-C2. And TEX-C1 had more immune cells infiltrating, as well as higher expression of immune checkpoint genes. A total of 1984 genes were differentially expressed between these two clusters, of which 1712 were associated with OS. A gene signature consisting of ten TEX-related genes was developed, and a risk score was computed for each patient. Based on the risk score, SEQC patients were split into high- and low-risk groups with significantly different survival rates. The risk score was an independent risk factor predicting survival and showed superior prediction power for 3, 5, and 10-year survival compared to individual clinical parameters. The signature was further confirmed in the TARGET and E-MTAB-8248 cohorts. This study has successfully constructed a risk score model for NB prognosis, utilizing TEX as its foundation. The model provides risk classification and survival evaluation, which can further guide treatment.
Collapse
Affiliation(s)
- Jiangtao Chen
- Department of Pediatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao Zhai
- Department of Pediatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Haiyang Kuang
- Department of Pediatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ai Zhang
- Department of Pediatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
3
|
Rojas-Diaz JM, Solorzano-Ibarra F, Garcia-Barrientos NT, Klimov-Kravtchenko K, Cruz-Ramos JA, Guitron-Aviña MS, Urciaga-Gutierrez PI, Ortiz-Lazareno PC, Tellez-Bañuelos MC, Bueno-Topete MR, Haramati J, Del Toro-Arreola S. Beyond Canonical Immune Checkpoints: Overexpression of TNFRSF Members 4-1BB and OX-40 Marks T Cells Exhibiting Phenotypic Features of Exhaustion in Cervical Carcinoma. Immunology 2025. [PMID: 40387515 DOI: 10.1111/imm.13945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 04/21/2025] [Accepted: 05/06/2025] [Indexed: 05/20/2025] Open
Abstract
T cells are pivotal in combating cancer; however, they can become exhausted during tumour progression, losing their cytotoxic capacity and upregulating inhibitory receptors including PD-1 and TIGIT. While checkpoint blockade has emerged as a potent treatment option for numerous cancers, patient selection, long-term efficacy, and adverse effects still remain an issue. For these reasons, it is important to investigate other pathways that might lead to selective reactivation of the immune system. Co-stimulatory TNFRSF receptors, including 4-1BB and OX-40, have emerged as promising targets for reactivating exhausted T cells. However, their expression on exhausted peripheral and tumour-infiltrating lymphocytes (TILs) is not well characterised, particularly in cervical cancer (CC), which remains the leading cause of gynaecological cancer mortality in low- and middle-income countries. To investigate the expression of these receptors, PBMCs were collected from CC patients and healthy donors, along with TILs from tumour biopsies, and analysed using multiparametric flow cytometry. Our findings revealed an increased population of phenotypically exhausted (PD-1+TIGIT+) CD4+ and CD8+ T cells in TILs, and, to a lesser extent, in peripheral blood and from CC patients. These exhausted T cell subsets exhibited selective overexpression of 4-1BB and OX-40 compared to phenotypically non-exhausted cells (PD-1-TIGIT-). In TILs, 4-1BB was overexpressed 12.7-fold in CD8 cells with the exhausted phenotype, OX-40 was overexpressed 3.3-fold; in CD4 cells with the exhausted phenotype, the overexpression was 7.8× and 3.8× for 4-1BB and OX-40, respectively. CD8 and CD4 T cells that were PD-1 + TIGIT+ 4-1BB+ were 7.3× and 16× more likely to be found in the tumour versus peripheral blood. Additionally, subpopulations of PD-1high T cells were significantly elevated in the tumour-infiltrating T cells and TIGIT expression was positively associated with PD-1 levels in peripheral patient CD8+ and CD4+ T cells, potentially indicating an advanced state of exhaustion. These findings suggest that TNFRSF members, especially 4-1BB, may serve as potential immunotherapeutic targets for reinvigorating exhausted T cells in CC.
Collapse
Affiliation(s)
- Jose Manuel Rojas-Diaz
- Instituto de Investigación en Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Fabiola Solorzano-Ibarra
- Instituto de Investigación en Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Nadia Tatiana Garcia-Barrientos
- Instituto de Investigación en Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Ksenia Klimov-Kravtchenko
- Instituto de Investigación en Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Jose Alfonso Cruz-Ramos
- Coordinación de Investigación, Subdirección de Desarrollo Institucional, Instituto Jalisciense de Cancerología, Guadalajara, Jalisco, Mexico
| | - Marcela Sofia Guitron-Aviña
- Instituto de Investigación en Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
- Laboratorio de Inmunología Traslacional, Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - Pedro Ivan Urciaga-Gutierrez
- Instituto de Investigación en Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Pablo Cesar Ortiz-Lazareno
- Centro de Investigación Biomédica de Occidente, División de Inmunología, Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, Mexico
| | - Martha Cecilia Tellez-Bañuelos
- Laboratorio de Inmunología Traslacional, Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - Miriam Ruth Bueno-Topete
- Instituto de Investigación en Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Jesse Haramati
- Laboratorio de Inmunología Traslacional, Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - Susana Del Toro-Arreola
- Instituto de Investigación en Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
- Laboratorio de Inmunología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| |
Collapse
|
4
|
Cao S, Lun S, Duan L, Gao Z, Wang X, Li Y, Zhang Y. Harnessing Calmodulin-Related Genes to Build a Prognostic Model in Esophageal Squamous Cell Carcinoma for a Comprehensive Analysis of Single-Cell Immune Characteristics and Drug Efficacy. J Immunother 2025:00002371-990000000-00140. [PMID: 40375794 DOI: 10.1097/cji.0000000000000561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/27/2025] [Indexed: 05/18/2025]
Abstract
SUMMARY Calmodulin (CALM) has a bearing on the prognosis of various cancers. However, the prognostic value of CALM in esophageal squamous cell carcinoma (ESCC) remains unelucidated. Differentially expressed genes (DEGs) were screened between normal and tumor groups of TCGA-ESCC sets. The intersection of DEGs with calmodulin-related genes (CRGs) yielded differentially expressed CRGs (DE-CRGs). A prognostic model was established using LASSO Cox regression analysis and multivariate Cox regression analysis. qPCR validated the expression of prognostic feature genes. Analysis of gene expression patterns of different cellular clusters was based on single-cell sequencing data. Lastly, GSEA enrichment, immune infiltration, mutational profiling, drug sensitivity, and molecular docking as well as cellular thermal shift assay (CETSA) were conducted for ESCC patients. A prognosis model with excellent predictive capability was created based on 4 feature genes (ATP2B3, CALB1, KCNQ1, and MYO1G). The qPCR results demonstrated that ATP2B3 and KCNQ1 were significantly downregulated in human ESCC cells, whereas CALB1 and MYO1G were upregulated (P<0.05). Single-cell analysis uncovered that MYO1G and KCNQ1 were mainly expressed in different cell clusters. Furthermore, this risk model was strongly associated with functional pathway enrichment, immune cell infiltration, and somatic mutations. We also identified AZD-8055 may be potential therapy for ESCC patients. The CETSA experiment demonstrated the existence of a favorable binding thermal stability between AZD-8055 and MYO1G. This research may identify potential biomarkers for predicting the prognosis of ESCC patients.
Collapse
Affiliation(s)
- Shasha Cao
- Henan Medical Key Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang, China
| | | | | | | | | | | | | |
Collapse
|
5
|
Dang B, Liang L, Li Z, Luo J, Zhong S. Bolstering CD8 + T Cells' Antitumor Immunity: A Promising Strategy to Improve the Response to Advanced Prostate Cancer Treatment. BIOLOGY 2025; 14:544. [PMID: 40427733 PMCID: PMC12108615 DOI: 10.3390/biology14050544] [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: 04/11/2025] [Revised: 05/07/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025]
Abstract
Prostate cancer is among the most frequently diagnosed and deadly cancers among men in the Western world. It is typically classified as an immune "cold" tumor due to its sparse immune cell presence and limited immunogenic response. Recent research has revealed the significant role of immune cells, especially CD8+ T cells, in both prostate cancer progression and treatment efficacy. This review integrates recent findings to provide a comprehensive overview of the current understanding of CD8+ T cell dynamics in prostate cancer and discusses emerging strategies to improve treatment outcomes. The ongoing exploration of new molecular targets and the development of innovative immunotherapeutic approaches hold promise for more effective management of prostate cancer, particularly in the context of advanced and resistant forms of the disease.
Collapse
Affiliation(s)
| | | | | | | | - Shangwei Zhong
- The Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang 421009, China; (B.D.); (L.L.); (Z.L.); (J.L.)
| |
Collapse
|
6
|
Wang J, Jia Y, Liu T, Liu X, Yin S, Chen J, Xu X, Zhang Y, Liu L. Tumor cell-intrinsic BIN1 deficiency promotes the immunosuppression and impedes ferroptosis of non-small cell lung cancer via G3BP1-mediated degradation of STAT1. J Exp Clin Cancer Res 2025; 44:141. [PMID: 40346580 PMCID: PMC12063428 DOI: 10.1186/s13046-025-03404-9] [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: 01/26/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Tumors often evade immune surveillance by limiting T cell infiltration. In non-small cell lung cancer (NSCLC), increased infiltration of CD8+ T cells is associated with a favorable response to immunotherapy. While BIN1 is recognized as a tumor suppressor gene, its role in shaping the tumor microenvironment in NSCLC has yet to be fully clarified. METHODS To investigate the relationship between BIN1 expression and CD8+T cell infiltration in NSCLC, we performed a comprehensive data analysis utilizing clinical information from NSCLC patients. BIN1 expression levels in NSCLC tissues were evaluated, and their correlation with CD8+T cells infiltration and patient survival outcomes was examined. Loss-of-function strategies targeting BIN1 were applied in syngeneic NSCLC mouse models to assess its functional significance. Tumor growth was monitored, and immune cell populations were analyzed in terms of frequency and functionality through mass cytometry and flow cytometry techniques. Cytokine secretion was profiled using multiplex assays. Additionally, RNA sequencing, immunoprecipitation-mass spectrometry, and molecular docking were employed to confirm direct interactions between BIN1 and cytokine-encoding genes. Finally, the regulatory role of BIN1 in ferroptosis in NSCLC cells were explored using metabolomics analysis, ROS measurement, and MDA detection. RESULTS We observed that BIN1 expression is downregulated in NSCLC tumor tissues, with its reduced expression strongly associated with advanced disease progression and poor prognosis. Bioinformatics analysis of immune infiltration in human NSCLC samples revealed a positive correlation between BIN1 expression in NSCLC tissues and CD8+ T cell infiltration. Furthermore, the prognostic impact of BIN1 on NSCLC patients is strongly linked to the level of CD8+ T cell infiltration. In syngeneic mouse models, the knockout of BIN1 in NSCLC cells significantly inhibited CD8+ T cell infiltration and impaired their cytotoxic function, facilitating tumor immune evasion. Mechanistically, we demonstrated that BIN1 directly interacts with G3BP1, and its knockout stabilizes G3BP1. This, in turn, promotes STAT1 degradation and reduces the secretion of T cell-recruiting chemokines such as CXCL10 and CCL5. Finally, our findings reveal that BIN1 influences ferroptosis in NSCLC cells through the G3BP1/STAT1/GSH pathway, thereby regulating NSCLC cell proliferation, migration, and invasion. CONCLUSION This study highlights the crucial role of the BIN1/G3BP1/STAT1/CD8+ tumor-infiltrating lymphocyte signaling pathway in the progression of NSCLC and its mechanisms of immune evasion. This fundings lay a foundation for the development of BIN1-targeted therapies aimed at improving tumor immunogenicity and transforming immunologically "cold" NSCLC into a more responsive disease.
Collapse
Affiliation(s)
- Jiali Wang
- Department of Tumor Immunotherapy, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, 050035, P.R. China
| | - Yunlong Jia
- Department of Tumor Immunotherapy, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, 050035, P.R. China
| | - Tianxu Liu
- Department of Tumor Immunotherapy, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, 050035, P.R. China
| | - Xinyan Liu
- Department of Tumor Immunotherapy, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, 050035, P.R. China
| | - Shuxian Yin
- Department of Tumor Immunotherapy, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, 050035, P.R. China
| | - Jiaqi Chen
- Department of Tumor Immunotherapy, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, 050035, P.R. China
| | - Xiaoqing Xu
- Department of General Surgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Yi Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450002, China
| | - Lihua Liu
- Department of Tumor Immunotherapy, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, 050035, P.R. China.
- China International Cooperation Laboratory of Stem Cell Research, Hebei Medical University, Shijiazhuang, 050011, China.
| |
Collapse
|
7
|
Lu Y, Wang FY, Levine MS, Shi HR, Wang Y, Xiong X, Yang LM, Shi YQ, Zou T, Sessler JL, Liang H, Huang KB. Oxoisoaporphine Alkaloid Iridium(III) Derivative: An Immunogenic Cell Death Inducer That Engages the Autophagy-Dependent Regulator Cathepsin D. J Am Chem Soc 2025; 147:15216-15228. [PMID: 40279467 DOI: 10.1021/jacs.5c00255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
Autophagy has been recognized as one of the pathways for eliciting immunogenic cell death (ICD). However, the specific molecular target responsible for autophagy-mediated ICD has not yet been elucidated. Here, we report that an oxoisoaporphine alkaloid-modified iridium(III) complex (2a) displays autophagy-inducing ICD activity. Through unbiased thermal proteome profiling (TPP), this new complex was found to interact with the lysosomal protease cathepsin D (Cat D). Subsequent cellular and biochemical assays─including the cellular thermal shift assay, isothermal dose-response assay, enzymatic assays, and molecular docking─confirmed that 2a binds to and inhibits Cat D. Further pathway analysis demonstrated that 2a triggers autophagy-dependent ICD via the LKB1-AMPK-ULK1 signaling pathway by inhibiting Cat D. Several other autophagy-dependent ICD inducers were tested and likewise found to inhibit Cat D. In contrast, an earlier reported analogue of 2a, complex 1a, was found to bind and destabilize binding immunoglobulin protein (BiP) and promote its ICD activity through an endoplasmic reticulum stress response. We believe that the findings reported here will enhance the understanding of the novel mechanisms of ICD agents and pave the way for the design of new ICD inducers with high specificity and efficacy.
Collapse
Affiliation(s)
- Yuan Lu
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
- Guangxi College Key Laboratory of Innovation Research on Medical and Engineering Integration & Liuzhou Key Laboratory of Guizhong Characteristic Medicinal Resources Development, School of Medicine, Guangxi University of Science and Technology, Liuzhou 545005, China
| | - Feng-Yang Wang
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
- Key Laboratory of Medical Biotechnology and Translational Medicine, Education Department of Guangxi Zhuang Autonomous Region, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541004, China
| | - Matthew S Levine
- Department of Chemistry, The University of Texas at Austin, 105 E. 24th Street-A5300, Austin, Texas 78712-1224, United States
| | - Hai-Rong Shi
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Yuan Wang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou 510006, China
| | - Xiaolin Xiong
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou 510006, China
| | - Liang-Mei Yang
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Ya-Qian Shi
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Taotao Zou
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou 510006, China
| | - Jonathan L Sessler
- Department of Chemistry, The University of Texas at Austin, 105 E. 24th Street-A5300, Austin, Texas 78712-1224, United States
| | - Hong Liang
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Ke-Bin Huang
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| |
Collapse
|
8
|
Wang H, Qiu B, Li X, Ying Y, Wang Y, Chen H, Zeng F, Shi J, Huang J, Wu Z, Chen Z, Che X, Li Q, Fan Y, Li B, Wang Q, Huang C, Chen Y, Li T, Mo K, Wang Q, Cui C. Single cell analysis reveals that SPP1 + macrophages enhance tumor progression by triggering fibroblast extracellular vesicles. Transl Oncol 2025; 55:102347. [PMID: 40086324 PMCID: PMC11954126 DOI: 10.1016/j.tranon.2025.102347] [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: 11/21/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
Patients with liver metastatic colorectal cancer (mCRC) have a poor prognosis and are the leading cause of death in colorectal cancer (CRC) patients, but the mechanisms associated with CRC metastasis have not been fully elucidated. In this study, we obtained data from the Gene Expression Omnibus database and characterized the single-cell profiles of CRC, mCRC and healthy samples at single-cell resolution, and explored the cells that influence CRC metastasis. We find that AQP1+ CRC identified as highly malignant tumor cells exhibited proliferative and metastatic characteristics. Immunosuppressive properties are present in the tumor microenvironment (TME), while NOTCH3+ Fib is identified to play a facilitating role in the metastatic colonization of CRC. Importantly, we reveal that tumor-associated macrophages (TAM) characterized by SPP1-specific high expression may be involved in TME remodeling through intercellular communication. Specifically, SPP1+ TAM mediates the generation of Fib-derived extracellular vesicle through the APOE-LRP1 axis, which in turn delivers tumor growth-promoting factors in the TME. This study deepens the understanding of the mechanism of TME in mCRC and lays the scientific foundation for the development of therapeutic regimens for mCRC patients.
Collapse
Affiliation(s)
- Haocheng Wang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Bowen Qiu
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xinyu Li
- Experimental Center of BIOQGene, YuanDong International Academy Of Life Sciences, Hong Kong 999077, China; Systems Biology Research Center, Biology Institute, Guangxi Academy of Sciences, Nanning 530007, Guangxi, China
| | - Yao Ying
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yue Wang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Hungchen Chen
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Fanan Zeng
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Junyao Shi
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Junpeng Huang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Ziying Wu
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Zequn Chen
- Department of Gastrointestinal Surgery, First Ward of Maoming People's Hospital, Maoming 525000, China
| | - Xiao Che
- Department of Abdominal Hernia Surgery, Maoming People's Hospital, Southern Medical University, Maoming 525000, China
| | - Qingzhong Li
- Guangzhou University of Traditional Chinese Medicine, Maoming 525000, China
| | - Yingming Fan
- Department of General Surgery, Guangning County People's Hospital, Guangdong Medical University, Zhaoqing 526300, China
| | - Bingyao Li
- Department of General Surgery, Guangning People's Hospital, Zhaoqing 526300, China
| | - Qun Wang
- Department of Emergency, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou 510282, China
| | - Chengyu Huang
- Experimental Center of BIOQGene, YuanDong International Academy Of Life Sciences, Hong Kong 999077, China; Systems Biology Research Center, Biology Institute, Guangxi Academy of Sciences, Nanning 530007, Guangxi, China
| | - Yixuan Chen
- Experimental Center of BIOQGene, YuanDong International Academy Of Life Sciences, Hong Kong 999077, China; Systems Biology Research Center, Biology Institute, Guangxi Academy of Sciences, Nanning 530007, Guangxi, China
| | - Ting Li
- Experimental Center of BIOQGene, YuanDong International Academy Of Life Sciences, Hong Kong 999077, China; Systems Biology Research Center, Biology Institute, Guangxi Academy of Sciences, Nanning 530007, Guangxi, China
| | - Ke Mo
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Experimental Center of BIOQGene, YuanDong International Academy Of Life Sciences, Hong Kong 999077, China; Systems Biology Research Center, Biology Institute, Guangxi Academy of Sciences, Nanning 530007, Guangxi, China.
| | - Qian Wang
- Department of Gastrointestinal surgery, The Seventh Affiliated Hospital, Southern Medical University, Foshan 528000, China.
| | - Chunhui Cui
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| |
Collapse
|
9
|
Lu S, Zhou Q, Zhao R, Xie L, Cao WM, Feng YX. Unraveling UPR-mediated intercellular crosstalk: Implications for immunotherapy resistance mechanisms. Cancer Lett 2025; 617:217613. [PMID: 40054654 DOI: 10.1016/j.canlet.2025.217613] [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: 11/20/2024] [Revised: 02/10/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025]
Abstract
Endoplasmic reticulum (ER) is the critical organelle that regulates essential cellular processes, including protein synthesis, folding, and post-translational modification, as well as lipid metabolism and calcium homeostasis. Disruption in ER homeostasis leads to a condition known as ER stress, characterized by the accumulation of misfolded or unfolded proteins. This triggers the unfolded protein response (UPR), an adaptive pathway mediated by three ER-resident sensors: inositol-requiring enzyme 1α (IRE1α), protein kinase R-like ER kinase (PERK), and activating transcription factor 6 (ATF6). Increasing evidence highlights sustained UPR activation in malignant and immune cells within the tumor microenvironment (TME), which promotes tumor progression and metastasis while simultaneously impairing antitumor immunity. This review explores how UPR-driven intercellular signaling influences immunotherapy resistance, focusing on the alterations occurring in tumor cells as well as in the surrounding immune environment. By providing insights into these mechanisms, we aim to highlight the therapeutic potential of targeting the UPR pathways in modulating cancer immunity.
Collapse
Affiliation(s)
- Si Lu
- Department of Head and Neck Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China; Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou, China
| | - Qimin Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rongjie Zhao
- Department of Gynecological Radiotherapy, Zhejiang Cancer Hospital, Hangzhou, China
| | - Lei Xie
- Department of Head and Neck Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Wen-Ming Cao
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China.
| | - Yu-Xiong Feng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China; Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou, China.
| |
Collapse
|
10
|
Liu L, Hao Z, Yang X, Li Y, Wang S, Li L. Metabolic reprogramming in T cell senescence: a novel strategy for cancer immunotherapy. Cell Death Discov 2025; 11:161. [PMID: 40204707 PMCID: PMC11982223 DOI: 10.1038/s41420-025-02468-y] [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: 08/18/2024] [Revised: 03/25/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025] Open
Abstract
The complex interplay between cancer progression and immune senescence is critically influenced by metabolic reprogramming in T cells. As T cells age, especially within the tumor microenvironment, they undergo significant metabolic shifts that may hinder their proliferation and functionality. This manuscript reviews how metabolic alterations contribute to T cell senescence in cancer and discusses potential therapeutic strategies aimed at reversing these metabolic changes. We explore interventions such as mitochondrial enhancement, glycolytic inhibition, and lipid metabolism adjustments that could rejuvenate senescent T cells, potentially restoring their efficacy in tumor suppression. This review also focuses on the significance of metabolic interventions in T cells with aging and further explores the future direction of the metabolism-based cancer immunotherapy in senescent T cells.
Collapse
Affiliation(s)
- Li Liu
- The Operation Room, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhanying Hao
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Xi Yang
- Department of General Surgery, Sanya People's Hospital, Sanya, China
| | - Yan Li
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.
| | - Siyang Wang
- Department of Anesthesiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.
| | - Linze Li
- The Operation Room, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
11
|
Cui G, Shao Y, Wang J, Xu C, Zhang J, Zhong Z. Polymersome-mediated Cbl-b silencing activates T cells against solid tumors. Biomater Sci 2025; 13:2036-2046. [PMID: 40017436 DOI: 10.1039/d5bm00001g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Unleashing T cell function is critical for efficacious cancer immunotherapy. Here, we present an in vivo T cell activation strategy by silencing Casitas B-lineage lymphoma proto-oncogene b (Cbl-b), an intracellular checkpoint, to effectively combat solid tumors. The polymersomes are able to efficiently load and deliver siRNA against cblb to T cells both in vitro and in vivo, successfully silencing the cblb gene expression in primary T cells and enhancing the IL-2 receptor CD25 expression, which in turn enhances T cell function and prevents T cell exhaustion. In vitro and in vivo studies showed that siRNA against cblb caused an effective inhibition of tumor progression in subcutaneous B16-F10 and LLC models, in which a significant increase of effector T cells in peripheral blood mononuclear cells and an increase of effector T cells and a significant decrease of Treg cells in the tumor were clearly observed. This polymersome-mediated down-regulation of the cblb gene in T cells provides a promising approach for activating T cells and enhancing their anti-tumor capacity.
Collapse
Affiliation(s)
- Guanhong Cui
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China.
| | - Yu Shao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P.R. China.
| | - Junyao Wang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P.R. China.
| | - Congcong Xu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China.
- International College of Pharmaceutical Innovation, Soochow University, Suzhou, 215222, P.R. China
| | - Jinping Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P.R. China.
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China.
- International College of Pharmaceutical Innovation, Soochow University, Suzhou, 215222, P.R. China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
12
|
Pang Y, Liang J, Deng Y, Chen W, Shen Y, Li J, Wang X, Ren Z. Identification and validation of HOXC6 as a diagnostic biomarker for Ewing sarcoma: insights from machine learning algorithms and in vitro experiments. Front Immunol 2025; 16:1449355. [PMID: 40255403 PMCID: PMC12006176 DOI: 10.3389/fimmu.2025.1449355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 03/18/2025] [Indexed: 04/22/2025] Open
Abstract
Introduction Early diagnosis of Ewing sarcoma (ES) is critical for improving patient prognosis. However, the accurate diagnosis of ES remains challenging, underscoring the need for novel diagnostic biomarkers to enhance diagnostic precision and reliability. This study aimed to identify potential gene expression-based biomarkers for the diagnosis of ES. Methods We selected the GSE17679, GSE45544, and GSE68776 datasets from the Gene Expression Omnibus (GEO) database. After correcting for batch effects, we combined ES and normal tissue samples from the GSE17679 and GSE45544 datasets to create a combined cohort. Two-thirds of both the tumor and normal samples from the combined cohort were randomly selected for the training cohort, while the remaining one-third served as the internal validation cohort. Additionally, the GSE68776 dataset was used for external validation. To identify key diagnostic genes, we applied three machine learning algorithms: least absolute shrinkage and selection operator (LASSO), support vector machine recursive feature elimination (SVM-RFE), and random forest (RF). Results HOXC6 was identified as a key diagnostic biomarker for ES. It demonstrated strong diagnostic performance across all cohorts, with area under the curve (AUC) values of 0.956 (95% CI: 0.909-0.990) in the training cohort, 0.995 (95% CI: 0.977-1.000) in the internal validation cohort, and 0.966 (95% CI: 0.910-0.999) in the external validation cohort. Functional validation through HOXC6 knockdown in the RD-ES cell line revealed that its suppression significantly inhibited cell proliferation and migration. Furthermore, transcriptome sequencing suggested potential oncogenic mechanisms underlying HOXC6 function. Discussion These findings highlight HOXC6 as a promising diagnostic biomarker for ES, demonstrating robust performance across multiple datasets. Additionally, its functional role suggests potential as a therapeutic target.
Collapse
Affiliation(s)
- Yonghua Pang
- Department of Orthopedics, The 904th Hospital of the Joint Logistics Support Force, People's Liberation Army of China, Wuxi, Jiangsu, China
| | - Jiahui Liang
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yakai Deng
- Department of Orthopedics, The 904th Hospital of the Joint Logistics Support Force, People's Liberation Army of China, Wuxi, Jiangsu, China
| | - Weinan Chen
- Department of Orthopedics, The 904th Hospital of the Joint Logistics Support Force, People's Liberation Army of China, Wuxi, Jiangsu, China
| | - Yunyan Shen
- Department of Orthopedics, The 904th Hospital of the Joint Logistics Support Force, People's Liberation Army of China, Wuxi, Jiangsu, China
| | - Jing Li
- Department of Orthopedics, Linyi People's Hospital, Linyi, Shandong, China
| | - Xin Wang
- Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Zhiyao Ren
- Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
13
|
Lin H, Zhu S, Chen Y, Lu J, Xie C, Liao C, Huang X, Li G, Wu Y, Li Z, Hu J, Lin X, Tian Y, Li Q, Wang Z, Chen S. Targeting cTRIP12 counteracts ferroptosis resistance and augments sensitivity to immunotherapy in pancreatic cancer. Drug Resist Updat 2025; 81:101240. [PMID: 40154160 DOI: 10.1016/j.drup.2025.101240] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 04/01/2025]
Abstract
AIMS Current therapeutic strategies for pancreatic ductal adenocarcinoma (PDAC) have limited efficacy in increasing patient survival rates, largely due to ferroptosis resistance and immunosuppression. The aim of this study is to identify molecular mechanisms associated with ferroptosis resistance and immunosuppression in PDAC tumour cells. METHODS Circular RNA sequencing (circRNA-seq) was performed on clinical samples to identify potential circRNAs that mediate ferroptosis resistance. C11-BODIPY staining, FerroOrange staining, the glutathione ratio, malondialdehyde quantification, and transmission electron microscopy were employed to assess ferroptosis. RNA pulldown, mass spectrometry, RNA immunoprecipitation, and coimmunoprecipitation assays were conducted to investigate the molecular mechanisms involved. A HuNSG mouse xenograft tumour model was utilized to validate therapeutic agents. RESULTS A circRNA derived from TRIP12 (cTRIP12) was identified in PDAC samples resistant to ferroptosis. cTRIP12 knockdown increased the sensitivity of PDAC cells to ferroptosis and immunotherapy. Subsequent mechanistic studies revealed that cTRIP12 specifically binds to the O-linked N-acetylglucosamine transferase (OGT) protein and increases intracellular O-GlcNAcylation levels, leading to increased protein levels of ferritin heavy chain (FTH) and PD-L1 in tumour cells. Notably, high cTRIP12 expression suppressed ferroptosis sensitivity and increased immune resistance in PDAC cells by functioning as a protein scaffold through its interaction with OGT and protein kinase R-like endoplasmic reticulum kinase (PERK). cTRIP12 inhibition induced ferroptosis in PDAC cells by reducing FTH and PD-L1 expression and synergistically increased the immunotherapy efficacy. In vivo animal experiments confirmed that the triple therapy consisting of GSK2656157, erastin, and anti-CTLA-4 effectively suppressed the progression of PDAC in tumours with high cTRIP12 expression. CONCLUSION We elucidated the molecular mechanisms underlying the simultaneous occurrence of ferroptosis resistance and immune suppression in PDAC patients. Our study provides a novel therapeutic strategy that could promote ferroptosis in tumour cells and increase immunotherapy efficacy.
Collapse
Affiliation(s)
- Hongyi Lin
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Shuncang Zhu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Yinhao Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Jinpeng Lu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Chengke Xie
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Chengyu Liao
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China; Fuzhou University, Fuzhou 350001, China
| | - Xiaoxiao Huang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China; Fuzhou University, Fuzhou 350001, China
| | - Ge Li
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Yongding Wu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Zhiyuan Li
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Jianfei Hu
- Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | | | - Yifeng Tian
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China; Fuzhou University, Fuzhou 350001, China
| | - Qiaowei Li
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Fuzhou University, Fuzhou 350001, China; Fujian Provincial Institute of Clinical Geriatrics, Fuzhou 350001, China; Fujian Key Laboratory of Geriatrics, Fuzhou 350001, China; Fujian Provincial Center for Geriatrics, Fuzhou 350001, China.
| | - Zuwei Wang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China; Fuzhou University, Fuzhou 350001, China.
| | - Shi Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China; Fuzhou University, Fuzhou 350001, China; Fujian Provincial Institute of Clinical Geriatrics, Fuzhou 350001, China; Fujian Key Laboratory of Geriatrics, Fuzhou 350001, China; Fujian Provincial Center for Geriatrics, Fuzhou 350001, China.
| |
Collapse
|
14
|
Xu Q, Li L, Zhu R. T Cell Exhaustion in Allergic Diseases and Allergen Immunotherapy: A Novel Biomarker? Curr Allergy Asthma Rep 2025; 25:18. [PMID: 40091122 DOI: 10.1007/s11882-025-01199-5] [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] [Accepted: 03/06/2025] [Indexed: 03/19/2025]
Abstract
PURPOSE OF REVIEW This review explores the emerging role of T cell exhaustion in allergic diseases and allergen immunotherapy (AIT). It aims to synthesize current knowledge on the mechanisms of T cell exhaustion, evaluate its potential involvement in allergic inflammation, and assess its implications as a novel biomarker for predicting and monitoring AIT efficacy. RECENT FINDINGS Recent studies highlight that T cell exhaustion, characterized by co-expression of inhibitory receptors (e.g., PD-1, CTLA-4, TIM-3), diminished cytokine production, and altered transcriptional profiles, may suppress type 2 inflammation in allergic diseases. In allergic asthma, exhausted CD4 + T cells exhibit upregulated inhibitory receptors, correlating with reduced IgE levels and airway hyperreactivity. During AIT, prolonged high-dose allergen exposure drives allergen-specific Th2 and T follicular helper (Tfh) cell exhaustion, potentially contributing to immune tolerance. Notably, clinical improvements in AIT correlate with depletion of allergen-specific Th2 cells and persistent expression of exhaustion markers (e.g., PD-1, CTLA-4) during maintenance phases. Blockade of inhibitory receptors (e.g., PD-1) enhances T cell activation, underscoring their dual regulatory role in allergy. T cell exhaustion represents a double-edged sword in allergy: it may dampen pathological inflammation in allergic diseases while serving as a mechanism for AIT-induced tolerance. The co-expression of inhibitory receptors on allergen-specific T cells emerges as a promising biomarker for AIT efficacy. Future research should clarify the transcriptional and metabolic drivers of exhaustion in allergy, validate its role across diverse allergic conditions, and optimize strategies to harness T cell exhaustion for durable immune tolerance. These insights could revolutionize therapeutic approaches and biomarker development in allergy management.
Collapse
Affiliation(s)
- Qingxiu Xu
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Le Li
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Rongfei Zhu
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
15
|
Zhao J, Cheng L, Yang J, Xu F, Qi W, Liao K, Zhou L, Cao L, Chen J, Lin Y. Tumor-nerve interactions in cancer regulation and progression. Cancer Lett 2025; 612:217483. [PMID: 39842496 DOI: 10.1016/j.canlet.2025.217483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/15/2025] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
Tumor-nerve interactions play a critical role in tumor progression, metastasis, and treatment resistance, redefining our understanding of the tumor microenvironment. This review provides a comprehensive analysis of how the peripheral and central nervous systems contribute to cancer biology, focusing on mechanisms of neural invasion, immune evasion, and tumor adaptation. It has highlighted the emerging potential of repurposing nervous system-targeted drugs originally developed for neurodegenerative and autoimmune diseases as innovative cancer therapies. The review also addresses key challenges, including the limitations of current experimental models and the complexity of translating preclinical findings to clinical applications. By bridging the gap between neuroscience and oncology, this interdisciplinary study aims to discover novel therapeutic strategies to improve outcomes for cancer patients.
Collapse
Affiliation(s)
- Jianyi Zhao
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Proton-Therapy, Shanghai, 201801, China
| | - Lilin Cheng
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Jian Yang
- Department of Neurosurgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Feifei Xu
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Proton-Therapy, Shanghai, 201801, China
| | - Weixiang Qi
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Proton-Therapy, Shanghai, 201801, China
| | - Keman Liao
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Proton-Therapy, Shanghai, 201801, China
| | - Li Zhou
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Proton-Therapy, Shanghai, 201801, China
| | - Lu Cao
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Proton-Therapy, Shanghai, 201801, China.
| | - Jiayi Chen
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Proton-Therapy, Shanghai, 201801, China.
| | - Yingying Lin
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Proton-Therapy, Shanghai, 201801, China.
| |
Collapse
|
16
|
Arciga BM, Walters DM, Kimchi ET, Staveley-O'Carroll KF, Li G, Teixeiro E, Rachagani S, Kaifi JT. Pulsed electric field ablation as a candidate to enhance the anti-tumor immune response to immune checkpoint inhibitors. Cancer Lett 2025; 609:217361. [PMID: 39608443 PMCID: PMC11625606 DOI: 10.1016/j.canlet.2024.217361] [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: 10/30/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
Cancer ablation with pulsed electric fields (PEFs) involves the delivery of high-voltage, short-duration electrical pulses that destabilize tumor cells, leading to cellular death. Unlike most conventional ablation technologies, PEF ablation is non-thermal, allowing for safe and targeted energy delivery to the tumor without damaging surrounding tissue and critical structures. PEFs allow for specific dosing, predictable treatment zones, and preservation of the extracellular matrix and adjacent vascular tissues. Preclinical and preliminary clinical data suggest that PEF ablation may induce inflammatory changes in the tumor microenvironment (TME) that engage host innate and adaptive immune cells, stimulating an anti-tumor response. Specifically, PEF promotes local and systemic anti-tumor immune activation through immunogenic cell death and the release of damage-associated molecular patterns (DAMPs) and tumor antigens. This tumor-specific immune activation could potentially enhance response to immune checkpoint inhibitor (ICI) therapies. Furthermore, PEF ablation induces the formation of tertiary lymphoid structures (TLSs) in the TME, which are predictive biomarkers for responsiveness to ICI across several solid tumors. This combination of effects activates antigen-presenting cells and stimulates the effector T cell response, which is often inhibited in ICI-resistant cancer patients. In this review, the onco-immunological characteristics of PEF ablation are discussed, with special emphasis placed on the clinical potential of PEF ablation to induce anti-cancer immune responses and enhance responsiveness to ICI therapy in ablated and non-ablated (abscopal) tumors.
Collapse
Affiliation(s)
- Blake M Arciga
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA; Hugh E. Stephenson Jr., M.D., Department of Surgery, University of Missouri, Columbia, MO, USA
| | - Dustin M Walters
- Department of Surgery, University of Connecticut, Farmington, CT, USA
| | - Eric T Kimchi
- Department of Surgery, University of Connecticut, Farmington, CT, USA
| | | | - Guangfu Li
- Department of Surgery, University of Connecticut, Farmington, CT, USA
| | - Emma Teixeiro
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA; Hugh E. Stephenson Jr., M.D., Department of Surgery, University of Missouri, Columbia, MO, USA; Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA; Siteman Cancer Center, Washington University, St. Louis, MO, USA
| | - Satyanarayana Rachagani
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA; Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, USA; Siteman Cancer Center, Washington University, St. Louis, MO, USA; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA.
| | - Jussuf T Kaifi
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA; Hugh E. Stephenson Jr., M.D., Department of Surgery, University of Missouri, Columbia, MO, USA; Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, USA; Siteman Cancer Center, Washington University, St. Louis, MO, USA; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA.
| |
Collapse
|
17
|
Shen J, Gong X, Ren H, Tang X, Yu H, Tang Y, Chen S, Ji M. Identification and validation of CDK1 as a promising therapeutic target for Eriocitrin in colorectal cancer: a combined bioinformatics and experimental approach. BMC Cancer 2025; 25:76. [PMID: 39806333 PMCID: PMC11731355 DOI: 10.1186/s12885-025-13448-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: 11/12/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a prevalent malignancy worldwide, associated with significant morbidity and mortality. Cyclin-dependent kinase 1 (CDK1) plays a crucial role in cell cycle regulation and has been implicated in various cancers. This study aimed to evaluate the prognostic value of CDK1 in CRC and to identify traditional Chinese medicines (TCM) that can target CDK1 as potential treatments for CRC. METHODS The expression and prognostic value of CDK1 were analyzed through TCGA, GEO, GEPIA, UALCAN and HPA databases. An ESTIMATE analysis was applied to estimate the proportions of stromal and immune cells in tumor samples. GO and KEGG enrichment analyses were performed to clarify the functional roles of CDK1-related genes. CCK-8, colony formation, cell migration, cell invasion, and wound healing assays were employed to explore tumor-promoting role of CDK1. Molecular docking, cellular thermal shift, and isothermal dose-response assays were employed to identify potential inhibitors of CDK1. RESULTS CDK1 was highly expressed in CRC and associated with a poorer prognosis. The expression of CDK1 was also correlated with the levels of immune cells infiltration. CDK1-related genes were primarily involved in the cell cycle and the P53 signaling pathway. Knockdown of CDK1 inhibited the proliferation, migration, and invasion of CRC cells in vitro. Furthermore, Eriocitrin emerged as a potential inhibitor, exerting its anti-tumor effects by targeting and inhibiting CDK1 activity. CONCLUSION CDK1 plays a critical role in CRC prognosis. Eriocitrin, a potential CDK1 inhibitor derived from TCM, highlights a promising new therapeutic strategy for CRC treatment.
Collapse
Affiliation(s)
- Jiemiao Shen
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, P. R. China
| | - Xing Gong
- Department of Environment Health, Nanjing Medical University Affiliated Nanjing Municipal Center for Disease Control and Prevention, 2 Zizhulin, Nanjing, 210003, P. R. China
| | - Haili Ren
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, P. R. China
| | - Xia Tang
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, P. R. China
| | - Hairong Yu
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, P. R. China
| | - Yilu Tang
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, P. R. China
| | - Shen Chen
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, P. R. China.
| | - Minghui Ji
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, P. R. China.
| |
Collapse
|
18
|
Dai Y, Dong C, Wang Z, Zhou Y, Wang Y, Hao Y, Chen P, Liang C, Li G. Infiltrating T lymphocytes and tumor microenvironment within cholangiocarcinoma: immune heterogeneity, intercellular communication, immune checkpoints. Front Immunol 2025; 15:1482291. [PMID: 39845973 PMCID: PMC11750830 DOI: 10.3389/fimmu.2024.1482291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025] Open
Abstract
Cholangiocarcinoma is the second most common primary liver cancer, and its global incidence has increased in recent years. Radical surgical resection and systemic chemotherapy have traditionally been the standard treatment options. However, the complexity of cholangiocarcinoma subtypes often presents a challenge for early diagnosis. Additionally, high recurrence rates following radical treatment and resistance to late-stage chemotherapy limit the benefits for patients. Immunotherapy has emerged as an effective strategy for treating various types of cancer, and has shown efficacy when combined with chemotherapy for cholangiocarcinoma. Current immunotherapies targeting cholangiocarcinoma have predominantly focused on T lymphocytes within the tumor microenvironment, and new immunotherapies have yielded unsatisfactory results in clinical trials. Therefore, it is essential to achieve a comprehensive understanding of the unique tumor microenvironment of cholangiocarcinoma and the pivotal role of T lymphocytes within it. In this review, we describe the heterogeneous immune landscape and intercellular communication in cholangiocarcinoma and summarize the specific distribution of T lymphocytes. Finally, we review potential immune checkpoints in cholangiocarcinoma.
Collapse
Affiliation(s)
- Yunyan Dai
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Chenyang Dong
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Zhiming Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yunpeng Zhou
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yi Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yi Hao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Pinggui Chen
- Department of Nuclear Medicine, Nanyang First People’s Hospital, Nanyang, Henan, China
| | - Chaojie Liang
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
- Department of biliary and Pancreatic Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Gaopeng Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
19
|
Wan R, Fu B, Fu X, Liu Z, Simayi N, Fu Y, Liang H, Li C, Huang W. SMAD4 Regulates the Expression of LCK Affecting Chimeric Antigen Receptor-T Cells Proliferation Through PI3K/Akt Signaling Pathway. J Cell Physiol 2025; 240:e31520. [PMID: 39763264 PMCID: PMC11704455 DOI: 10.1002/jcp.31520] [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: 06/17/2024] [Revised: 12/06/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025]
Abstract
The proliferation of CAR-T cells was hindered and cannot play its killing function well in solid tumors. And yet the regulatory mechanism of CAR-T cell proliferation is not fully understood. Here, we showed that recombinant expression of CD19CAR in T cells significantly increased the basal activation level of CAR-T cells and LCK activation. Both LCK and SMAD4 were essential for CAR-T cells proliferation since over-express LCK or SMAD4 significantly promotes CAR-T cells proliferation, while knock-down LCK or SMAD4 expression inhibited the proliferation of CAR-T cells seriously. More cells go into apoptosis when knock-down LCK or SMAD4 expression, and the cell cycle was arrested in G2/M or S phase, respectively. Over-express LCK or SMAD4 significantly promotes phosphorylation of PI3K and Akt, while it was inhibited when cells were treated with PI3K and Akt inhibitors (LY294002 or MK2206). Further mechanism exploration experiments showed that SMAD4 bound on the promoter region of LCK regulating its expression. Taken together, we reported that the transcription factor SMAD4 regulated the expression of LCK and further involved in the PI3K/Akt signaling pathway to affect the proliferation of CAR-T cells.
Collapse
Affiliation(s)
- Rongxue Wan
- Department of Transfusion Medicine, School of BiotechnologySouthern Medical UniversityGuangzhouGuangdongChina
- Guangzhou Blood Center, Institute of Blood Transfusion and HematologyGuangzhou Medical UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Bowen Fu
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
- The Third Affiliated HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Xiaokang Fu
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Zengping Liu
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Nafeisha Simayi
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Yongshui Fu
- Department of Transfusion Medicine, School of BiotechnologySouthern Medical UniversityGuangzhouGuangdongChina
- Guangzhou Blood Center, Institute of Blood Transfusion and HematologyGuangzhou Medical UniversityGuangzhouGuangdongChina
- Institute of Blood Transfusion and HematologyGuangzhou First People's HospitalGuangzhouGuangdongChina
| | - Huaqin Liang
- Guangzhou Blood Center, Institute of Blood Transfusion and HematologyGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Chengyao Li
- Department of Transfusion Medicine, School of BiotechnologySouthern Medical UniversityGuangzhouGuangdongChina
| | - Wenhua Huang
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
20
|
Chowdhury D, Das A, Mishra M, Khutere T, Bodakhe SH. Physiological markers for immunotherapeutics: a review. J Chemother 2024:1-24. [PMID: 39711144 DOI: 10.1080/1120009x.2024.2443701] [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: 09/09/2024] [Revised: 11/22/2024] [Accepted: 12/10/2024] [Indexed: 12/24/2024]
Abstract
Immunotherapy has been advanced through multiple approaches, including immunogenic cytokines, monoclonal antibodies, therapeutic vaccinations, adoptive cell transfer, stem cell transplantation, and oncolytic viruses. This review analyses various strategies in genomics, transcriptomics, single-cell techniques, computational analysis, big data, and imaging technologies for the identification of tumour microbiota and microenvironments. Immunotherapy is becoming acknowledged as a feasible cancer treatment method, facilitating innovative cancer medicines and personalized medicine techniques.
Collapse
Affiliation(s)
- Durlav Chowdhury
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Ashmita Das
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Mrityunjay Mishra
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Trinkal Khutere
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Surendra H Bodakhe
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| |
Collapse
|
21
|
Zhang X, Zhang C, Lu S, Dong J, Tang N, Wang Y, Han W, Pan X, Zhang X, Liu D, Shyh-Chang N, Wang Y, Feng G, Wang H. Miltefosine reinvigorates exhausted T cells by targeting their bioenergetic state. Cell Rep Med 2024; 5:101869. [PMID: 39657666 PMCID: PMC11722131 DOI: 10.1016/j.xcrm.2024.101869] [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: 11/05/2023] [Revised: 09/05/2024] [Accepted: 11/15/2024] [Indexed: 12/12/2024]
Abstract
T cell exhaustion presents a major challenge for the efficacy of both immune checkpoint inhibitors (ICBs) and chimeric antigen receptor T (CAR-T) cell immunotherapies. To address this issue, we generate hypofunctional CAR-T cells that imitate the exhaustion state. By screening a Food and Drug Administration (FDA)-approved small molecule library using this model, we identify miltefosine as a potent molecule that restores the impaired function of CAR-T cells in a PD-1/PD-L1-independent manner. Impressively, in the terminally exhausted state where PD-1 antibody treatment is ineffective, miltefosine still enhances CAR-T cell activity. Single-cell sequencing analysis reveals that miltefosine treatment significantly increases the population of effector cells. Mechanistically, miltefosine improves impaired glycolysis and oxidative phosphorylation in hypofunctional CAR-T cells. In both allogeneic and syngeneic tumor models, miltefosine effectively enhances the solid tumor clearance ability of CAR-T cells and T cells, demonstrating its potential as an effective immunotherapeutic drug.
Collapse
Affiliation(s)
- Xingying Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Chenze Zhang
- National Key Laboratory of Efficacy and Mechanism on Chinese Medicine for Metabolic Diseases, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shan Lu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingxi Dong
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Na Tang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yao Wang
- Chinese People's Liberation Army General Hospital, Beijing 100176, China
| | - Weidong Han
- Chinese People's Liberation Army General Hospital, Beijing 100176, China
| | - Xi Pan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiang Zhang
- National Key Laboratory of Efficacy and Mechanism on Chinese Medicine for Metabolic Diseases, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Duan Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ng Shyh-Chang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Yu Wang
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Guihai Feng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Haoyi Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
22
|
Li W, He H, Wang H, Wen W. Dynamics of liver cancer cellular taxa revealed through single-cell RNA sequencing: Advances and challenges. Cancer Lett 2024; 611:217394. [PMID: 39689824 DOI: 10.1016/j.canlet.2024.217394] [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: 08/20/2024] [Revised: 10/13/2024] [Accepted: 12/14/2024] [Indexed: 12/19/2024]
Abstract
Liver cancer is a leading cause of death worldwide, representing a substantial public health challenge. The advent of single-cell RNA sequencing has significantly advanced our understanding of cellular dynamics from the onset of liver cancer to therapeutic intervention. This technology has unveiled profound insights into cancer heterogeneity and the tumor microenvironment (TME), enabling the identification of key molecular drivers and phenotypic landscapes of liver cancer at a single-cell resolution. This review highlights recent advancements in mapping functional cell subsets, phenotypic alterations, and the diversity of the TME. These insights are pivotal for advancing targeted therapies and developing prognostic tools. Moreover, this review covers the ongoing challenges and advances from tumor initiation to progression, offering a detailed perspective on advancing personalized treatment.
Collapse
Affiliation(s)
- Wenxin Li
- Third Affiliated Hospital of Naval Medical University (Second Military Medical University), National Center for Liver Cancer, Shanghai, 200438, China; Department of Clinical Laboratory Medicine, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, 200438, China
| | - Huisi He
- Third Affiliated Hospital of Naval Medical University (Second Military Medical University), National Center for Liver Cancer, Shanghai, 200438, China; Department of Oncology, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, 200438, China
| | - Hongyang Wang
- Third Affiliated Hospital of Naval Medical University (Second Military Medical University), National Center for Liver Cancer, Shanghai, 200438, China; The Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai, 200438, China.
| | - Wen Wen
- Third Affiliated Hospital of Naval Medical University (Second Military Medical University), National Center for Liver Cancer, Shanghai, 200438, China; Department of Clinical Laboratory Medicine, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, 200438, China; The Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai, 200438, China.
| |
Collapse
|
23
|
Yu H, Li J, Peng S, Liu Q, Chen D, He Z, Xiang J, Wang B. Tumor microenvironment: Nurturing cancer cells for immunoevasion and druggable vulnerabilities for cancer immunotherapy. Cancer Lett 2024; 611:217385. [PMID: 39645024 DOI: 10.1016/j.canlet.2024.217385] [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: 08/14/2024] [Revised: 09/29/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
The tumor microenvironment (TME) is an intricate ecosystem where cancer cells thrive, encompassing a wide array of cellular and non-cellular components. The TME co-evolves with tumor progression in a spatially and temporally dynamic manner, which endows cancer cells with the adaptive capability of evading immune surveillance. To this end, diverse cancer-intrinsic mechanisms were exploited to dampen host immune system, such as upregulating immune checkpoints, impairing antigens presentation and competing for nutrients. In this review, we discuss how cancer immunoevasion is tightly regulated by hypoxia, one of the hallmark biochemical features of the TME. Moreover, we comprehensively summarize how immune evasiveness of cancer cells is facilitated by the extracellular matrix, as well as soluble components of TME, including inflammatory factors, lactate, nutrients and extracellular vesicles. Given their important roles in dictating cancer immunoevasion, various strategies to target TME components are proposed, which holds promising translational potential in developing novel therapeutics to sensitize anti-cancer immunotherapy such as immune checkpoint blockade.
Collapse
Affiliation(s)
- Hongyang Yu
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Jinyang Li
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Shiyin Peng
- School of Medicine, Chongqing University, Chongqing, China
| | - Qin Liu
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Dongfeng Chen
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Zongsheng He
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China.
| | - Junyu Xiang
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China.
| | - Bin Wang
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China; Institute of Pathology and Southwest Cancer Center, And Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China; Jinfeng Laboratory, Chongqing, 401329, China.
| |
Collapse
|
24
|
Shi X, Cheng X, Jiang A, Shi W, Zhu L, Mou W, Glaviano A, Liu Z, Cheng Q, Lin A, Wang L, Luo P. Immune Checkpoints in B Cells: Unlocking New Potentials in Cancer Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403423. [PMID: 39509319 PMCID: PMC11653663 DOI: 10.1002/advs.202403423] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/26/2024] [Indexed: 11/15/2024]
Abstract
B cells are crucial component of humoral immunity, and their role in the tumor immune microenvironment (TME) has garnered significant attention in recent years. These cells hold great potential and application prospects in the field of tumor immunotherapy. Research has demonstrated that the TME can remodel various B cell functions, including proliferation, differentiation, antigen presentation, and antibody production, thereby invalidating the anti-tumor effects of B cells. Concurrently, numerous immune checkpoints (ICs) on the surface of B cells are upregulated. Aberrant B-cell IC signals not only impair the function of B cells themselves, but also modulate the tumor-killing effects of other immune cells, ultimately fostering an immunosuppressive TME and facilitating tumor immune escape. Blocking ICs on B cells is beneficial for reversing the immunosuppressive TME and restoring anti-tumor immune responses. In this paper, the intricate connection between B-cell ICs and the TME is delved into, emphasizing the critical role of targeting B-cell ICs in anti-tumor immunity, which may provide valuable insights for the future development of tumor immunotherapy based on B cells.
Collapse
Affiliation(s)
- Xiaoye Shi
- Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdong510282China
- The Second School of Clinical MedicineSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Xiangshu Cheng
- College of Bioinformatics Science and TechnologyHarbin Medical University157 Baojian Road. Nangang District, HarbinHeilongiiang150076China
| | - Aimin Jiang
- Department of UrologyChanghai HospitalNaval Medical University (Second Military Medical University)Shanghai200433China
| | - Wenjie Shi
- Molecular and Experimental SurgeryUniversity Clinic for General‐Visceral‐Vascular‐ and Trans‐Plantation SurgeryMedical Faculty University Hospital MagdeburgOtto‐von Guericke University39120MagdeburgGermany
| | - Lingxuan Zhu
- Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdong510282China
| | - Weiming Mou
- Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdong510282China
- Department of UrologyShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080China
| | - Antonino Glaviano
- Department of BiologicalChemical and Pharmaceutical Sciences and TechnologiesUniversity of PalermoPalermo90123Italy
| | - Zaoqu Liu
- Institute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100730China
| | - Quan Cheng
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Anqi Lin
- Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdong510282China
| | - Linhui Wang
- Department of UrologyChanghai HospitalNaval Medical University (Second Military Medical University)Shanghai200433China
| | - Peng Luo
- Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdong510282China
- Cancer Centre and Institute of Translational MedicineFaculty of Health SciencesUniversity of MacauMacau SAR999078China
| |
Collapse
|
25
|
Chen L, Wu GZ, Wu T, Shang HH, Wang WJ, Fisher D, Hiens NTT, Musabaev E, Zhao L. Cell Cycle-Related LncRNA-Based Prognostic Model for Hepatocellular Carcinoma: Integrating Immune Microenvironment and Treatment Response. Curr Med Sci 2024; 44:1217-1231. [PMID: 39681799 DOI: 10.1007/s11596-024-2924-9] [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: 04/08/2024] [Accepted: 08/04/2024] [Indexed: 12/18/2024]
Abstract
OBJECTIVE Hepatocellular carcinoma (HCC) presents substantial genetic and phenotypic diversity, making it challenging to predict patient outcomes. There is a clear need for novel biomarkers to better identify high-risk individuals. Long non-coding RNAs (lncRNAs) are known to play key roles in cell cycle regulation and genomic stability, and their dysregulation has been closely linked to HCC progression. Developing a prognostic model based on cell cycle-related lncRNAs could open up new possibilities for immunotherapy in HCC patients. METHODS Transcriptomic data and clinical samples were obtained from the TCGA-HCC dataset. Cell cycle-related gene sets were sourced from existing studies, and coexpression analysis identified relevant lncRNAs (correlation coefficient >0.4, P<0.001). Univariate analysis identified prognostic lncRNAs, which were then used in a LASSO regression model to create a risk score. This model was validated via cross-validation. HCC samples were classified on the basis of their risk scores. Correlations between the risk score and tumor mutational burden (TMB), tumor immune infiltration, immune checkpoint gene expression, and immunotherapy response were evaluated via R packages and various methods (TIMER, CIBERSORT, CIBERSORT-ABS, QUANTISEQ, MCP-COUNTER, XCELL, and EPIC). RESULTS Four cell cycle-related lncRNAs (AC009549.1, AC090018.2, PKD1P6-NPIPP1, and TMCC1-AS1) were significantly upregulated in HCC. These lncRNAs were used to create a risk score (risk score=0.492×AC009549.1+1.390×AC090018.2+1.622×PKD1P6-NPIPP1+0.858×TMCC1-AS1). This risk score had superior predictive value compared to traditional clinical factors (AUC=0.738). A nomogram was developed to illustrate the 1-year, 3-year, and 5-year overall survival (OS) rates for individual HCC patients. Significant differences in TMB, immune response, immune cell infiltration, immune checkpoint gene expression, and drug responsiveness were observed between the high-risk and low-risk groups. CONCLUSION The risk score model we developed enhances the prognostication of HCC patients by identifying those at high risk for poor outcomes. This model could lead to new immunotherapy strategies for HCC patients.
Collapse
Affiliation(s)
- Lin Chen
- Department of Infectious Diseases, Tsinghua University Affiliated Chuiyangliu Hospital, Beijing, 100021, China.
| | - Guo-Zhi Wu
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Tao Wu
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Hao-Hu Shang
- Jingchuan County People's Hospital, Jingliang, 744300, China
| | - Wei-Juan Wang
- Department of Infectious Diseases, Tsinghua University Affiliated Chuiyangliu Hospital, Beijing, 100021, China
| | - David Fisher
- Department of Medical Biosciences, Faculty of Natural Sciences, University of the Western Cape, Cape Town, 7100, South Africa
| | | | - Erkin Musabaev
- The Research Institute of Virology, Ministry of Health, Tashkent, 100133, Uzbekistan
| | - Lei Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
26
|
Kalim M, Jing R, Guo W, Xing H, Lu Y. Functional diversity and regulation of IL-9-producing T cells in cancer immunotherapy. Cancer Lett 2024; 606:217306. [PMID: 39426662 PMCID: PMC11675864 DOI: 10.1016/j.canlet.2024.217306] [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: 06/18/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/21/2024]
Abstract
IL-9-producing T cells (T9) regulate immunological responses that affect various cellular biological processes, though their precise function remains fully understood. Previous studies have linked T9 cells to conditions such as allergic disorders, parasitic infection clearance, and various types of cancers. While the functional heterogeneity of IL-9 and T9 cells in cancer development has been documented, these cells present promising therapeutic opportunities for treating solid tumors. This review highlights the roles of IL-9 and T9 cells in cancer progression and treatment responses, focusing on potential discrepancies in IL-9/IL-9R signaling between murine tumors and cancer patients. Additionally, we discuss the regulation of tumor-specific Th9/Tc9 cell differentiation, the therapeutic potential of these cells, and current strategies to enhance their anti-tumor activities.
Collapse
Affiliation(s)
- Muhammad Kalim
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX, 77030, USA
| | - Rui Jing
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX, 77030, USA
| | - Wei Guo
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX, 77030, USA
| | - Hui Xing
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX, 77030, USA
| | - Yong Lu
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
27
|
You H, Geng S, Li S, Imani M, Brambilla D, Sun T, Jiang C. Recent advances in biomimetic strategies for the immunotherapy of glioblastoma. Biomaterials 2024; 311:122694. [PMID: 38959533 DOI: 10.1016/j.biomaterials.2024.122694] [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: 04/08/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
Immunotherapy is regarded as one of the most promising approaches for treating tumors, with a multitude of immunotherapeutic thoughts currently under consideration for the lethal glioblastoma (GBM). However, issues with immunotherapeutic agents, such as limited in vivo stability, poor blood-brain barrier (BBB) penetration, insufficient GBM targeting, and represented monotherapy, have hindered the success of immunotherapeutic interventions. Moreover, even with the aid of conventional drug delivery systems, outcomes remain suboptimal. Biomimetic strategies seek to overcome these formidable drug delivery challenges by emulating nature's intelligent structures and functions. Leveraging the variety of biological structures and functions, biomimetic drug delivery systems afford a versatile platform with enhanced biocompatibility for the co-delivery of diverse immunotherapeutic agents. Moreover, their inherent capacity to traverse the BBB and home in on GBM holds promise for augmenting the efficacy of GBM immunotherapy. Thus, this review begins by revisiting the various thoughts and agents on immunotherapy for GBM. Then, the barriers to successful GBM immunotherapy are analyzed, and the corresponding biomimetic strategies are explored from the perspective of function and structure. Finally, the clinical translation's current state and prospects of biomimetic strategy are addressed. This review aspires to provide fresh perspectives on the advancement of immunotherapy for GBM.
Collapse
Affiliation(s)
- Haoyu You
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shuo Geng
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shangkuo Li
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Mohammad Imani
- Department of Science, Iran Polymer and Petrochemical Institute, Tehran 14977-13115, Iran; Center for Nanoscience and Nanotechnology, Institute for Convergence Science & Technology, Tehran 14588-89694, Iran
| | - Davide Brambilla
- Faculty of Pharmacy, University of Montreal, Montreal Quebec H3T 1J4, Canada
| | - Tao Sun
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
28
|
Zhang X, Liu L. Senescent T Cells: The Silent Culprit in Acute Myeloid Leukemia Progression? Int J Mol Sci 2024; 25:12550. [PMID: 39684260 DOI: 10.3390/ijms252312550] [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: 10/31/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Malignant tumors can evade immune surveillance and elimination through multiple mechanisms, with the induction of immune cell dysfunction serving as a crucial strategy. Mounting evidence indicates that T cell senescence constitutes the primary mechanism underlying T cell dysfunction in acute myeloid leukemia (AML) and represents one of the potential causes of immunotherapy failure. AML usually progresses rapidly and is highly susceptible to drug resistance, thereby resulting in recurrence and patient mortality. Hence, disrupting the immune interface within the bone marrow microenvironment of AML has emerged as a critical objective for synergistically enhancing tumor immunotherapy. In this review, we summarize the general characteristics, distinctive phenotypes, and regulatory signaling networks of senescent T cells and highlight their potential clinical significance in the bone marrow microenvironment of AML. Additionally, we discuss potential therapeutic strategies for alleviating and reversing T cell senescence.
Collapse
Affiliation(s)
- Xiaolan Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lingbo Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
29
|
Liu K, Hoover AR, Wang L, Sun Y, Valerio TI, Furrer C, Adams J, Yang J, Li M, Chen WR. Localized ablative immunotherapy enhances antitumor immunity by modulating the transcriptome of tumor-infiltrating Gamma delta T cells. Cancer Lett 2024; 604:217267. [PMID: 39307410 PMCID: PMC11471373 DOI: 10.1016/j.canlet.2024.217267] [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: 06/28/2024] [Revised: 09/10/2024] [Accepted: 09/18/2024] [Indexed: 09/26/2024]
Abstract
Gamma delta T cells (γδT cells) play crucial roles in the immune response against tumors, yet their functional dynamics under different cancer therapies remain poorly understood. Laser Ablative Immunotherapy (LAIT) is a novel cancer treatment modality combining local photothermal therapy (PTT) and intratumoral injection of an immunostimulant, N-dihydrogalactochitosan (glycated chitosan, GC). LAIT has been shown to induce systemic antitumor immune responses in pre-clinical studies and clinical trials, eradicating both treated local tumors and untreated distant metastases. In this study, we used LAIT to treat breast tumors in a mouse model and investigated the effects of LAIT on tumor-infiltrating γδT cells using single-cell RNA sequencing (scRNAseq). We characterized the γδT cells from tumors in control, PTT, GC, and LAIT (PTT + GC) groups, by identifying six distinct subtypes: activated, cytotoxic, cycling cytotoxic, IFN-enriched, antigen-presenting, and IL17-producing γδT cells. Differential gene expression analysis revealed that LAIT significantly upregulated genes associated with T cell activation, leukocyte adhesion, and interferon signaling in treated tumor tissues while downregulating genes involved in protein folding and stress responses. LAIT also uniquely increased the proportion of IL17-producing γδT cells, which correlated with prolonged survival in breast cancer patients, as analyzed using TCGA data. Furthermore, the transcriptomic profiles of γδT cells in LAIT-treated tumors closely resembled those in immune checkpoint inhibitor (ICI)-treated patients, suggesting potential synergistic effects. Our findings indicate that LAIT modulates the γδT cell transcriptome, enhancing their antitumor capabilities and providing a basis for combining LAIT with ICI therapy to improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Kaili Liu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Ashley R Hoover
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA; Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Lin Wang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Yuanhong Sun
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Trisha I Valerio
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Coline Furrer
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Jacob Adams
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Jingxuan Yang
- Department of Medicine, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Min Li
- Department of Medicine, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Wei R Chen
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
30
|
Dong L, Zhu Y, Zhang H, Gao L, Zhang Z, Xu X, Ying L, Zhang L, Li Y, Yun Z, Zhu D, Han C, Xu T, Yang H, Ju S, Chen X, Zhang H, Xie J. Open-Source Throttling of CD8 + T Cells in Brain with Low-Intensity Focused Ultrasound-Guided Sequential Delivery of CXCL10, IL-2, and aPD-L1 for Glioblastoma Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407235. [PMID: 39264011 DOI: 10.1002/adma.202407235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/14/2024] [Indexed: 09/13/2024]
Abstract
Improving clinical immunotherapy for glioblastoma (GBM) relies on addressing the immunosuppressive tumor microenvironment (TME). Enhancing CD8+ T cell infiltration and preventing its exhaustion holds promise for effective GBM immunotherapy. Here, a low-intensity focused ultrasound (LIFU)-guided sequential delivery strategy is developed to enhance CD8+ T cells infiltration and activity in the GBM region. The sequential delivery of CXC chemokine ligand 10 (CXCL10) to recruit CD8+ T cells and interleukin-2 (IL-2) to reduce their exhaustion is termed an "open-source throttling" strategy. Consequently, up to 3.39-fold of CD8+ T cells are observed with LIFU-guided sequential delivery of CXCL10, IL-2, and anti-programmed cell death 1 ligand 1 (aPD-L1), compared to the free aPD-L1 group. The immune checkpoint inhibitors (ICIs) therapeutic efficacy is substantially enhanced by the reversed immunosuppressive TME due to the expansion of CD8+ T cells, resulting in the elimination of tumor, prolonged survival time, and long-term immune memory establishment in orthotopic GBM mice. Overall, LIFU-guided sequential cytokine and ICIs delivery offers an "open-source throttling" strategy of CD8+ T cells, which may present a promising strategy for brain-tumor immunotherapy.
Collapse
Affiliation(s)
- Lei Dong
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology; Department of Oncology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, China
| | - Yini Zhu
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu, 210009, China
| | - Haoge Zhang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Basic Medicine Research and Innovation Center of Ministry of Education, State Key Laboratory of Digital Medical Engineering, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Lin Gao
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Basic Medicine Research and Innovation Center of Ministry of Education, State Key Laboratory of Digital Medical Engineering, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Zhiqi Zhang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Basic Medicine Research and Innovation Center of Ministry of Education, State Key Laboratory of Digital Medical Engineering, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Xiaoxuan Xu
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Basic Medicine Research and Innovation Center of Ministry of Education, State Key Laboratory of Digital Medical Engineering, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Leqian Ying
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology; Department of Oncology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, China
| | - Lu Zhang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology; Department of Oncology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, China
| | - Yue Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, 999078, China
| | - Zhengcheng Yun
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology; Department of Oncology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, China
| | - Danqi Zhu
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Basic Medicine Research and Innovation Center of Ministry of Education, State Key Laboratory of Digital Medical Engineering, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Chang Han
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Basic Medicine Research and Innovation Center of Ministry of Education, State Key Laboratory of Digital Medical Engineering, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Tingting Xu
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology; Department of Oncology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, China
| | - Hui Yang
- Department of Biochemistry and Molecular Biology, Medical School of Southeast University, Nanjing, China
| | - Shenghong Ju
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Basic Medicine Research and Innovation Center of Ministry of Education, State Key Laboratory of Digital Medical Engineering, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Xiaoyuan Chen
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Basic Medicine Research and Innovation Center of Ministry of Education, State Key Laboratory of Digital Medical Engineering, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Haijun Zhang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology; Department of Oncology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, China
| | - Jinbing Xie
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Basic Medicine Research and Innovation Center of Ministry of Education, State Key Laboratory of Digital Medical Engineering, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| |
Collapse
|
31
|
Wang Y, Gao B, Jiao T, Zhang W, Shi H, Jiang H, Li X, Li J, Ge X, Pan K, Li C, Mao G, Lu S. CCL5/CCR5/CYP1A1 pathway prompts liver cancer cells to survive in the combination of targeted and immunological therapies. Cancer Sci 2024; 115:3552-3569. [PMID: 39183447 PMCID: PMC11531955 DOI: 10.1111/cas.16320] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024] Open
Abstract
Combination therapy of anti-programmed cell death protein-1 (PD-1) antibodies and tyrosine kinase inhibitors (TKIs) has significantly improved the prognosis for hepatocellular carcinoma (HCC), but many patients still have unsatisfactory outcomes. CD8 T cells are known to exert a pivotal function in the immune response against tumors. Nevertheless, most CD8 T cells in HCC tissues are in a state of exhaustion, losing the cytotoxic activity against malignant cells. Cytokines, mainly secreted by immune cells, play an important role in the occurrence and development of tumors. Here, we demonstrated the changes in exhausted CD8T cells during combination therapy by single-cell RNA sequencing (scRNA-seq) analysis on tumor samples before and after treatment. Combination therapy exerted a substantial impact on the exhausted CD8T cells, particularly in terms of cytokine expression. CCL5 was the most abundantly expressed cytokine in CD8T cells and exhausted CD8T cells, and its expression increased further after treatment. Subsequently, we discovered the CCL5/CCR5/CYP1A1 pathway through RNA sequencing (RNA-seq) on CCL5-stimulated Huh7 cells and verified through a series of experiments that this pathway can mediate the resistance of liver cancer cells to lenvatinib. Tissue experiments showed that after combination therapy, the CCL5/CCR5/CYP1A1 pathway was activated, which can benefit the residual tumor cells to survive treatment. Tumor-bearing mouse experiments demonstrated that bergamottin (BGM), a competitive inhibitor of CYP1A1, can enhance the efficacy of both lenvatinib and combination therapy. Our research revealed one mechanism by which hepatoma cells can survive the combination therapy, providing a theoretical basis for the refined treatment of HCC.
Collapse
Affiliation(s)
- Yafei Wang
- Nankai University School of Medicine, Nankai UniversityTianjinChina
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Biao Gao
- Nankai University School of Medicine, Nankai UniversityTianjinChina
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Tianyu Jiao
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Wenwen Zhang
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Huizhong Shi
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Hao Jiang
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Xuerui Li
- Nankai University School of Medicine, Nankai UniversityTianjinChina
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Junfeng Li
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Xinlan Ge
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Ke Pan
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Chonghui Li
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Guankun Mao
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| | - Shichun Lu
- Nankai University School of Medicine, Nankai UniversityTianjinChina
- Faculty of Hepato‐Pancreato‐Biliary SurgeryChinese PLA General HospitalBeijingChina
- Institute of Hepatobiliary Surgery of Chinese PLABeijingChina
- Key Laboratory of Digital Hepatobiliary Surgery of Chinese PLABeijingChina
| |
Collapse
|
32
|
Zhang C, Wu Q, Yang H, Zhang H, Liu C, Yang B, Hu Q. Ferroptosis-related gene signature for predicting prognosis and identifying potential therapeutic drug in EGFR wild-type lung adenocarcinoma. Commun Biol 2024; 7:1416. [PMID: 39478024 PMCID: PMC11525656 DOI: 10.1038/s42003-024-07117-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
Epidermal growth factor receptor wild type lung adenocarcinoma (EGFRWT LUAD) still has limited treatment options and unsatisfactory clinical outcomes. Ferroptosis, as a form of cell death, has been reported to play a dual role in regulating tumor cell survival. In this study, we constructed a 3-ferroptosis-gene signature, FeSig, and verified its accuracy and efficacy in predicting EGFRWT LUAD prognosis at both the RNA and protein levels. Patients with higher FeSig scores were found to have worse clinical outcomes. Additionally, we explored the relationship between FeSig and tumor microenvironment, revealing that enhanced interactions between fibroblasts and tumor cells in FeSighigh patients causing tumor resistance to ferroptosis. To address this challenge, we screened potential drugs from NCI-60 (The US National Cancer Institute 60 human tumour cell line anticancer drug screen) and Connectivity map database, ultimately identifying 6-mercatopurine (6-MP) as a promising candidate. Both in vitro and in vivo experiments demonstrated its efficacy in treating FeSighigh EGFRWT LUAD tumor models. In summary, we develop a novel FeSig for predicting prognosis and guiding drug application.
Collapse
Affiliation(s)
- Chuankai Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Qi Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Hongwei Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Hui Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Changqing Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Bo Yang
- The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Qingsong Hu
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China.
| |
Collapse
|
33
|
Chen S, Tang Z, Wan Q, Huang W, Li X, Huang X, Zheng S, Lu C, Wu J, Li Z, Liu X. Machine learning and single-cell RNA sequencing reveal relationship between intratumor CD8 + T cells and uveal melanoma metastasis. Cancer Cell Int 2024; 24:359. [PMID: 39478575 PMCID: PMC11523669 DOI: 10.1186/s12935-024-03539-3] [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: 07/19/2024] [Accepted: 10/17/2024] [Indexed: 11/03/2024] Open
Abstract
PURPOSE Uveal melanoma (UM) is adults' most common primary intraocular malignant tumor. It has been observed that 40% of patients experience distant metastasis during subsequent treatment. While there exist multigene models developed using machine learning methods to assess metastasis and prognosis, the immune microenvironment's specific mechanisms influencing the tumor microenvironment have not been clarified. Single-cell transcriptome sequencing can accurately identify different types of cells in a tissue for precise analysis. This study aims to develop a model with fewer genes to evaluate metastasis risk in UM patients and provide a theoretical basis for UM immunotherapy. METHODS RNA-seq data and clinical information from 79 μm patients from TCGA were used to construct prognostic models. Mechanisms were probed using two single-cell datasets derived from the GEO database. After screening for metastasis-related genes, enrichment analysis was performed using GO and KEGG. Prognostic genes were screened using log-rank test and one-way Cox regression, and prognostic models were established using LASSO regression analysis and multifactor Cox regression analysis. The TCGA-UVM dataset was used as internal validation and dataset GSE22138 as external validation data. A time-dependent subject work characteristic curve (time-ROC) was established to assess the predictive ability of the model. Subsequently, dimensionality reduction, clustering, pseudo-temporal analysis and cellular communication analysis were performed on GSE138665 and GSE139829 to explore the underlying mechanisms involved. Cellular experiments were also used to validate the relevant findings. RESULTS Based on clinical characteristics and RNA-seq transcriptomic data from 79 samples in the TCGA-UVM cohort, 247 metastasis-related genes were identified. Survival models for three genes (SLC25A38, EDNRB, and LURAP1) were then constructed using lasso regression and multifactorial cox regression. Kaplan-Meier survival analysis showed that the high-risk group was associated with poorer overall survival (OS) and metastasis-free survival (MFS) in UM patients. Time-dependent ROC curves demonstrated high predictive performance in 6 m, 18 m, and 30 m prognostic models. Cell scratch assay showed that the 24 h and 48 h migration rates of cells with reduced expression of the three genes were significantly higher than those of the si-NC group. CD8 + T cells may play an important role in tumour metastasis as revealed by immune infiltration analysis. An increase in the percentage of cytotoxic CD8 + T cells in the metastatic high-risk group was found in the exploration of single-cell transcriptome data. The communication intensity of cytotoxic CD8 was significantly enhanced. It was also found that the CD8 + T cells in the two groups were in different states, although the number of CD8 + T cells in the high-risk group increased, they were mostly in the exhausted and undifferentiated state, while in the low-risk group, the CD8 + T cells were mostly in the functional state. CONCLUSIONS We developed a precise and stable 3-gene model to predict the metastatic risk and prognosis of patients. CD8 + T cells exhaustion in the tumor microenvironment play a crucial role in UM metastasis.
Collapse
Affiliation(s)
- Shuming Chen
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Zichun Tang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Qiaoqian Wan
- Department of Anaesthesiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Weidi Huang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Xie Li
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Xixuan Huang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Shuyan Zheng
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Caiyang Lu
- Hunan Centre for Drug Review and Adverse Reaction Monitoring, Changsha, Hunan, 410013, China
| | - Jinzheng Wu
- Hunan Provincial Drug Administration, Changsha, Hunan, 410013, China
| | - Zhuo Li
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Hunan Provincial Key Laboratory of Critical Quality Attribute of Cell Therapy Products, Changsha, Hunan, 410013, China.
| | - Xiao Liu
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| |
Collapse
|
34
|
Zhao Z, Chen M, Sun K, Gu X. CD8+ T cell associated scoring model helps prognostic diagnosis and immunotherapy selection in patients with colon adenocarcinoma. Heliyon 2024; 10:e37998. [PMID: 39386801 PMCID: PMC11462492 DOI: 10.1016/j.heliyon.2024.e37998] [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: 06/03/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024] Open
Abstract
Objective T cell-mediated immunity plays a crucial role in the immune response against tumors, with CD 8+ T cells playing a leading role in the eradication of cancer cells. Material and methods A total of 5 datasets were included in this study. Single cell transcriptome data were used to discover CD8+ T cell marker genes, and Bulk transcriptome data from TCGA and GEO were jointly analyzed to screen candidate prognostic genes. lasso regression was performed to construct prognostic models. Immunotherapy cohort (IMvigor 210 and GSE78220) was applied to validate the diagnostic power of markers. Result Single-cell transcriptome data identified 65 CD8+ T cell marker genes, highlighting their importance in T cell-mediated immune responses. Among these, 11 genes were identified as CD8+ T-associated differential genes through analysis of bulk data from TCGA and GEO. A prognostic model for 5 genes was identified based on Lasso regression, dividing colon adenocarcinoma (COAD) patients into high- and low-risk groups. This model exhibited higher prognostic accuracy compared to traditional clinicopathological characteristics (age, pathological stage, histological grading). Moreover, the risk score derived from this model successfully differentiated patient responses to immunotherapy, as validated in the IMvigor 210 and GSE78220 cohorts. Conclusion Our research introduces a novel prognostic signature based on CD8+ T cell marker genes, demonstrating significant predictive power for prognosis and immunotherapy response in COAD patients. This model offers a potential tool for improving patient stratification and personalizing treatment strategies.
Collapse
Affiliation(s)
- Zheng Zhao
- Deparment of General Surgery, Gongli Hospital, Pudong New Area, Shanghai, China
| | - Mingkai Chen
- Deparment of Gastroenterlogy, Zhengzhou Yihe Hospital, Zhengzhou, China
| | - Kuanxue Sun
- Deparment of General Surgery, Gongli Hospital, Pudong New Area, Shanghai, China
| | - Xinqi Gu
- Department of Gastroenterlogy, Shanghai Pudong Hospital, Pudong Medical Center of Fudan University, Shanghai, China
| |
Collapse
|
35
|
Chen F, Qian WB, Chen ZH, Qian J, Luo C. T cell exhaustion methylation signature drives differential immune responses in glioblastoma. Discov Oncol 2024; 15:530. [PMID: 39377985 PMCID: PMC11461406 DOI: 10.1007/s12672-024-01412-3] [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: 07/24/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND Methylation-related signatures play crucial roles in tumorigenesis and progression. However, their roles in the immune response in primary glioblastoma (GBM) remains unclear. METHODS We analyzed the differential expression of specific members of T cell exhaustion-related pathways in GBM from the perspective of T cell exhaustion. We further screened for significantly negatively correlated methylation sites as candidate methylation markers for T cell exhaustion. Using consensus clustering, we divided the samples into two categories with significant differences in overall survival (OS). We then performed univariate and multivariate Cox regression analyses to construct the T Cell Exhaustion Methylation (TEXM) signature. Finally, we confirmed that this signature served as an independent prognostic factor, and further characterized it in terms of drug resistance and immunotherapy. RESULTS We identified 95 significantly differentially expressed T cell exhaustion-related genes and 51 methylation markers associated with T cell exhaustion. The cancer samples were classified according to methylation site markers, thus indicating two subtypes with significant differences in OS: subtype A and subtype B. Tumor scores, stromal scores, tumor purity, and ESTIMATE scores all showed significant differences between subtypes (P < 0.05). Univariate Cox regression analysis identified five methylation sites significantly associated with OS, and multivariate Cox regression analysis was used to construct the TEXM signature model by using these five methylation sites. Significant differences in OS were found between the groups with high and low TEXM signature scores, on the basis of calculation of the TEXM signature scores of tumor samples and using the median score to divide them into high and low score groups. Survival analysis revealed that the high score group had poorer OS and DFS than the low score group in the validation set. Notably, we observed a significant difference in drug sensitivity between the high and low TEXM signature score groups, with the high score group showing higher drug resistance and poorer prognosis. The tumor immune state, as predicted with Tracking Tumor Immunophenotype (TIP), revealed significant differences in antitumor immune scores between the high and low TEXM signature score groups. Finally, we identified 43 significantly differentially regulated metabolism-associated biological processes. CONCLUSION The epigenetic methylation-related TEXM signature plays a key role in driving differential immune responses in GBM.
Collapse
Affiliation(s)
- Feng Chen
- Department of Neurosurgery, Tongji Hospital, School of Medicine, Tongji University, No. 389, Xinchun Road, Shanghai, 200065, China
| | - Wen-Bo Qian
- Department of Neurosurgery, Tongji Hospital, School of Medicine, Tongji University, No. 389, Xinchun Road, Shanghai, 200065, China
| | - Zhen-Hua Chen
- Department of Neurosurgery, Affiliated Hospital 2 of Nantong University, Nantong, China
| | - Jun Qian
- Department of Neurosurgery, Tongji Hospital, School of Medicine, Tongji University, No. 389, Xinchun Road, Shanghai, 200065, China.
| | - Chun Luo
- Department of Neurosurgery, Tongji Hospital, School of Medicine, Tongji University, No. 389, Xinchun Road, Shanghai, 200065, China.
| |
Collapse
|
36
|
Liu D, Wang L, Guo Y. Advances in and prospects of immunotherapy for prostate cancer. Cancer Lett 2024; 601:217155. [PMID: 39127338 DOI: 10.1016/j.canlet.2024.217155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/07/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024]
Abstract
Immunotherapy has shown promising therapeutic effects in hematological malignancies and certain solid tumors and has emerged as a critical and highly potential treatment modality for cancer. However, prostate cancer falls under the category of immune-resistant cold tumors, for which immunotherapy exhibits limited efficacy in patients with solid tumors. Thus, it is important to gain a deeper understanding of the tumor microenvironment in prostate cancer to facilitate immune system activation and overcome immune suppression to advance immunotherapy for prostate cancer. In this review, we discuss the immunosuppressive microenvironment of prostate cancer, which is characterized by the presence of few tumor-infiltrating lymphocytes, abundant immunosuppressive cells, low immunogenicity, and a noninflammatory phenotype, which significantly influences the efficacy of immunotherapy for prostate cancer. Immunotherapy is mainly achieved by activating the host immune system and overcoming immunosuppression. In this regard, we summarize the therapeutic advances in immune checkpoint blockade, immunogenic cell death, reversal of the immunosuppressive tumor microenvironment, tumor vaccines, immune adjuvants, chimeric antigen receptor T-cell therapy, and overcoming penetration barriers in prostate cancer, with the aim of providing novel research insights and approaches to enhance the effectiveness of immunotherapy for prostate cancer.
Collapse
Affiliation(s)
- Deng Liu
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China; Bioengineering College, Chongqing University, Chongqing, 400044, China
| | - Luofu Wang
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Yanli Guo
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
37
|
Li Z, Wu Z, You X, Tang N. Pan-cancer analysis reveals that TK1 promotes tumor progression by mediating cell proliferation and Th2 cell polarization. Cancer Cell Int 2024; 24:329. [PMID: 39343871 PMCID: PMC11440694 DOI: 10.1186/s12935-024-03515-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND TK1 (Thymidine kinase 1) is a member of the thymidine kinase family and has been observed to be significantly upregulated in a variety of cancer types. However, the exact roles of TK1 in tumor progression and the tumor immune microenvironment are not fully understood. This study aims to investigate the comprehensive involvement of TK1 in pan-cancer through the utilization of bioinformatics analysis, validation of pathological tissue samples, and in vitro experimental investigations. METHODS The expression profiles together with diagnostic and prognostic role of TK1 in pan-cancer were investigated though TCGA, TARGET, GTEx, and CPTAC databases. The single-sample gene set enrichment analysis (ssGSEA) and single-cell sequencing datasets were used to examine the relationship between TK1 and immune infiltration. The expression of TK1 were verified in hepatocellular carcinoma (HCC) through qPCR, western blotting and immunohistochemical assays. The proliferative capacity of HCC cell lines was assessed through CCK-8 and colony formation assays, while cytokine levels were measured via ELISA. Furthermore, flow cytometry was utilized to analyze cell cycle distribution and the proportions of Th2 cells. RESULTS TK1 was overexpressed in most cancers and demonstrated significant diagnostic and prognostic value. Among the various immune cells in pan-cancer, Th2 cells exhibited the closest association with TK1. Furthermore, the single-cell atlas provided insights into the distribution and proportion of TK1 in immune cells of HCC. In vitro experiments revealed an elevated expression of TK1 in HCC tissue and cell lines, and its role in influencing HCC cell proliferation by regulating G0/G1 phase arrest. Additionally, TK1 in cancer cells was found to potentially modulate Th2 cell polarization through the chemokine CCL5. CONCLUSION TK1 holds immense potential as a biomarker for pan-cancer diagnosis and prognosis. Additionally, targeting the expression of TK1 represents a promising therapeutic approach that can enhance the efficacy of current anti-tumor immunotherapy by modulating Th2 cell polarization and multiple mechanisms.
Collapse
Affiliation(s)
- Zhecheng Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhaoyi Wu
- Department of Thyroid and Breast Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Human Normal University, Changsha, 410008, China
| | - Xing You
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Neng Tang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
38
|
Li Z, Zhao Y, Huang P, Wu Z, Ouyang D, Nyarko AO, Ai L, Zhang Z, Chang S. Pan-cancer analysis of the immunological and oncogenic roles of ATAD2 with verification in papillary thyroid carcinoma. Sci Rep 2024; 14:22263. [PMID: 39333272 PMCID: PMC11436736 DOI: 10.1038/s41598-024-73274-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024] Open
Abstract
ATAD2 (ATPase Family AAA Domain-Containing 2) is highly expressed across varies tumor types, yet its common roles in tumor progression and immune interaction remain unclear. We analyzed the expression and alteration profiles of ATAD2, along with its diagnostic and prognostic role in pan-cancer, utilizing TCGA, GTEx, CPTAC, HPA, and cBioPortal databases. Furthermore, we examined the relationship between ATAD2 and immune infiltration utilizing single-cell sequencing data and TCGA database. Additionally, the expression and oncogenic functions of ATAD2 were verified in papillary thyroid carcinoma (PTC) through MTT, wound-healing, transwell, and flow cytometry assays. Our results revealed significant overexpression of ATAD2 in most cancers, strongly associated with poor prognosis. Amplification was the most frequent alteration type of ATAD2, with its mutation correlating with improved overall survival. ATAD2 was positively correlated with multiple inhibitory immune checkpoints and closely associated with the immunosuppressive microenvironment, particularly in PTC. In vitro experiments demonstrated that ATAD2 promoted the proliferation, migration, and invasion of PTC cells by activating the PI3K-AKT pathway and modulating the G1/S cell cycle checkpoint. Collectively, ATAD2 holds promise as a biomarker for pan-cancer diagnosis and prognosis, as well as a predictor of immunotherapeutic responsiveness and a therapeutic target to enhance the efficacy of existing anti-tumor immune therapies.
Collapse
Affiliation(s)
- Zhecheng Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yaxin Zhao
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Peng Huang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhaoyi Wu
- Department of Thyroid and Breast Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Human Normal University, Changsha, 410008, China
| | - Dengjie Ouyang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Adolphus Osei Nyarko
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Lei Ai
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhejia Zhang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Shi Chang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
39
|
Xu W, Sang S, Wang J, Guo S, Zhang X, Zhou H, Chen Y. Identification of telomere-related lncRNAs and immunological analysis in ovarian cancer. Front Immunol 2024; 15:1452946. [PMID: 39355254 PMCID: PMC11442270 DOI: 10.3389/fimmu.2024.1452946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/26/2024] [Indexed: 10/03/2024] Open
Abstract
Background Ovarian cancer (OC) is a global malignancy characterized by metastatic invasiveness and recurrence. Long non-coding RNAs (lncRNAs) and Telomeres are closely connected with several cancers, but their potential as practical prognostic markers in OC is less well-defined. Methods Relevant mRNA and clinical data for OC were sourced from The Cancer Genome Atlas (TCGA) database. The telomere-related lncRNAs (TRLs) prognostic model was established by univariate/LASSO/multivariate regression analyses. The effectiveness of the TRLs model was evaluated and measured via the nomogram. Additionally, immune infiltration, tumor mutational load (TMB), and drug sensitivity were evaluated. We validated the expression levels of prognostic genes. Subsequently, PTPRD-AS1 knockdown was utilized to perform the CCK8 assay, colony formation assay, transwell assay, and wound healing assay of CAOV3 cells. Results A six-TRLs prognostic model (PTPRD-AS1, SPAG5-AS1, CHRM3-AS2, AC074286.1, FAM27E3, and AC018647.3) was established, which can effectively predict patient survival rates and was successfully validated using external datasets. According to the nomogram, the model could effectively predict prognosis. Furthermore, we detected the levels of regulatory T cells and M2 macrophages were comparatively higher in the high-risk TRLs group, but the levels of activated CD8 T cells and monocytes were the opposite. Finally, the low-risk group was more sensitive to anti-cancer drugs. The mRNA levels of PTPRD-AS1, SPAG5-AS1, FAM27E3, and AC018647.3 were significantly over-expressed in OC cell lines (SKOV3, A2780, CAOV3) in comparison to normal IOSE-80 cells. AC074286.1 were over-expressed in A2780 and CAOV3 cells and CHRM3-AS2 only in A2780 cells. PTPRD-AS1 knockdown decreased the proliferation, cloning, and migration of CAOV3 cells. Conclusion Our study identified potential biomarkers for the six-TRLs model related to the prognosis of OC.
Collapse
Affiliation(s)
- Weina Xu
- Department of TCM, Zhoujiadu Community Health Service of Shanghai Pudong New Area, Shanghai, China
| | - Shuliu Sang
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jun Wang
- Department of TCM, Zhoujiadu Community Health Service of Shanghai Pudong New Area, Shanghai, China
| | - Shanshan Guo
- Department of Gynecology, Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao Zhang
- Department of Gynecology, Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hailun Zhou
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yijia Chen
- Department of Gynecology, Longhua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
40
|
Wang B, Liu W, Zhang M, Li Y, Tang H, Wang Y, Song C, Song B, Tan B. Circ_0001947 encapsulated by small extracellular vesicles promotes gastric cancer progression and anti-PD-1 resistance by modulating CD8 + T cell exhaustion. J Nanobiotechnology 2024; 22:563. [PMID: 39272146 PMCID: PMC11401313 DOI: 10.1186/s12951-024-02826-5] [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: 05/27/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND While small extracellular vesicles (sEVs)-derived circular RNAs (circRNAs) have been emerged as significant players in cancer, the function and underlying mechanism of sEVs-derived circRNAs in anti-cancer immunity remain unclear. METHODS Gastric cancer (GC)-derived circRNAs were identified using RNA-seq data from GEO datasets and quantitative reverse transcription polymerase chain reaction (qRT-PCR), RNA immunoprecipitation, dual-luciferase assay, and bioinformatics analysis were performed to investigate the regulatory axis. Transwell assay, wound healing assay, cell counting kit-8 (CCK-8) assay, and xenograft models were used to evaluate its role in GC progression in vivo and in vitro. The delivery of specific circRNAs into sEVs were verified through electron microscopy, nanoparticle tracking analysis (NTA) and fuorescence in situ hybridization (FISH). Flow cytometric analysis and immunohistochemical staining were conducted to find out how specific circRNAs mediated CD8+ T cell exhaustion and resistant to anti-programmed cell death 1 (PD-1) therapy. RESULTS We identified that circ_0001947, packaged by GC-derived sEVs, was obviously elevated in GC and was associated with poor clinical outcome. High circ0001947 level augmented the proliferation, migration, and invasion of GC cells. Mechanistically, circ0001947 sponged miR-661 and miR-671-5p to promote the expression of CD39, which further facilitated CD8+ T cell exhaustion and immune resistance. Conversely, blocking circ_0001947 attenuated CD8+ T cell exhaustion and increased the response to anti-PD-1 therapy. CONCLUSIONS Our study manifested the therapeutic potential of targeting sEVs-transmitted circ_0001947 to prohibit CD8+ T cell exhaustion and immune resistance in GC.
Collapse
Affiliation(s)
- Bingyu Wang
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Wenbo Liu
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Mingming Zhang
- Hebei Key Laboratory of Metabolic Disease, Shijiazhuang, 050011, China
| | - Yong Li
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Hongyue Tang
- Hebei Key Laboratory of Metabolic Disease, Shijiazhuang, 050011, China
| | - Yingying Wang
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Chao Song
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Buyun Song
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Bibo Tan
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China.
| |
Collapse
|
41
|
Li X, Yang G, Ma L, Tang B, Tao T. N 6-methyladenosine (m 6A) writer METTL5 represses the ferroptosis and antitumor immunity of gastric cancer. Cell Death Discov 2024; 10:402. [PMID: 39261486 PMCID: PMC11390903 DOI: 10.1038/s41420-024-02166-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/20/2024] [Accepted: 08/27/2024] [Indexed: 09/13/2024] Open
Abstract
Emerging evidence has shown that ferroptosis and antitumor immunity response of T lymphocytes play critical roles in multiple malignancies, including gastric cancer (GC). Here, the present research aims to reveal the function of novel N6-methyladenosine (m6A) methyltransferase METTL5 on GC immune microenvironment. Clinically, elevated METTL5 was negatively correlated to the prognosis of GC patients. METTL5 high-expression repressed the Fe2+ accumulation and ferroptosis to promote the GC immune evasion escaping from activated PBMCs' killing effect. Mechanistically, upregulation of METTL5 promoted NRF2 mRNA stability, thereby inactivating the ferroptosis and repressing PBMCs' cells antitumor immunity. One valuable finding is that ferroptosis inhibitor (Ferrostatin-1, Fer-1) could reduce the antitumor immunity of cocultured PBMCs. In other words, the increase of ferroptosis might contribute to the anti-tumor efficacy of immunotherapy. Further study revealed that m6A reader IGF2BP1 mediated the stability of NRF2 mRNA via METTL5/m6A/NRF2 axis. Collectively, these results demonstrate that METTL5 functions as an oncogene in GC immune microenvironment, and highlights a critical role in T lymphocytes' antitumor immunity.
Collapse
Affiliation(s)
- Xinli Li
- Department of Clinical Laboratory, Zibo Central Hospital, Zibo, China
| | - Guoqiang Yang
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China
| | - Lihong Ma
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China
| | - Bingxi Tang
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China
| | - Tao Tao
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China.
| |
Collapse
|
42
|
Hu Y, Zhang Y, Shi F, Yang R, Yan J, Han T, Guan L. Reversal of T-cell exhaustion: Mechanisms and synergistic approaches. Int Immunopharmacol 2024; 138:112571. [PMID: 38941674 DOI: 10.1016/j.intimp.2024.112571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 06/30/2024]
Abstract
T cells suffer from long-term antigen stimulation and insufficient energy supply, leading to a decline in their effector functions, memory capabilities, and proliferative capacity, ultimately resulting in T cell exhaustion and an inability to perform normal immune functions in the tumor microenvironment. Therefore, exploring how to restore these exhausted T cells to a state with effector functions is of great significance. Exhausted T cells exhibit a spectrum of molecular alterations, such as heightened expression of inhibitory receptors, shifts in transcription factor profiles, and modifications across epigenetic, metabolic, and transcriptional landscapes. This review provides a comprehensive overview of various strategies to reverse T cell exhaustion, including immune checkpoint blockade, and explores the potential synergistic effects of combining multiple approaches to reverse T cell exhaustion. It offers new insights and methods for achieving more durable and effective reversal of T cell exhaustion.
Collapse
Affiliation(s)
- Yang Hu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yaqi Zhang
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang 453003, China
| | - Fenfen Shi
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Ruihan Yang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jiayu Yan
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Tao Han
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang 453003, China.
| | - Liping Guan
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
43
|
Wu J, Li L, Cheng Z. System analysis based on T-cell exhaustion-related genes identifies PTPRT as a promising diagnostic and prognostic biomarker for gastric cancer. Sci Rep 2024; 14:21049. [PMID: 39251810 PMCID: PMC11384728 DOI: 10.1038/s41598-024-72135-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 09/04/2024] [Indexed: 09/11/2024] Open
Abstract
Multiple investigations have demonstrated the crucial involvement of T-cell exhaustion (TEX) in anti-tumor immune response and their strong correlation with prognosis. This study aimed at creating a strong signature using TEX for gastric cancer through bioinformatics analysis and experimental validation. We utilized data from The Cancer Genome Atlas (TCGA) databases to retrieve RNA-seq data from patients with stomach adenocarcinoma (STAD). Genes related to TEX were discovered using gene set variance analysis (GSVA) and weighted gene correlation network analysis (WGCNA). Subsequently, prognostic signature based on TEX was developed using LASSO-Cox analysis. Relationship between key genes and immune cells were examined. Finally, biological function of a key TEX-related gene PTPRT in gastric cancer was verified by in vivo experiment. A total of 29 TEX-related biomarkers were screened by WGCNA and random forest. Among them, five core signatures (PTPRT, CAV2, PPIH, PRDM2, and FGF1), further identified by LASSO-Cox, were considered as strong predictors of prognosis for gastric cancer and associated with immune infiltration. PTPRT gene had the largest number of SNPs, with the most mutation types. In vivo experiments revealed that PTPRT overexpression significantly inhibited tumor malignant progression and accelerated apoptosis through stimulating the secretion of killer cytokines such as TNF-α and IFN-γ. In addition, flow cytometry revealed that PTPRT overexpression alleviated TEX by increasing the abundance of CD8+ T cells, with inhibition of cell surface PD-1 and Tim-3. The predictive prognostic value of TEX gene expression levels was evaluated in patients with gastric cancer, providing a new perspective for precision immuno-oncology studies.
Collapse
Affiliation(s)
- Jianli Wu
- Medical School, Huanghe S&T University, No. 666 Zijingshan South Road, Zhengzhou, 450015, Henan, People's Republic of China
| | - Le Li
- Medical School, Huanghe S&T University, No. 666 Zijingshan South Road, Zhengzhou, 450015, Henan, People's Republic of China
| | - Zhenyun Cheng
- Medical School, Huanghe S&T University, No. 666 Zijingshan South Road, Zhengzhou, 450015, Henan, People's Republic of China.
| |
Collapse
|
44
|
Zou S, Zhang L, Jiang C, Li F, Yang Y, Deng X, Zhang J, Chen H, Jiang L, Cheng X, Deng L, Lin L, Shen B, Wen C, Zhan Q. Driver mutation subtypes involve with differentiated immunophenotypes influencing pancreatic cancer outcomes. Cancer Lett 2024; 599:217134. [PMID: 39094824 DOI: 10.1016/j.canlet.2024.217134] [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: 04/10/2024] [Revised: 07/03/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024]
Abstract
Despite many studies focusing on the prognostic biomarkers in pancreatic adenocarcinomas (PAADs), there is ill-informed about the relationships between their genomic features and immune characteristics. Herein, we deeply investigated the involvement of major driver mutation subtypes with immunophenotypes impacting PAAD outcomes. Based on public data analyses of RNA expression-based immune subtypes in PAAD, in contrast to KRAS G12D & TP53 co-mutant patients with poor outcomes, the best immune subtype C3 (inflammatory) characterized by high Th1/Th2 ratio was relatively enriched in KRASnon-G12DTP53wt patients with better survival, whereas the inferior subtype C2 (IFN-γ dominant) with low Th1/Th2 ratio was more common in the former than in the latter. Moreover, contrary to the highly immunosuppressive microenvironment (high Treg, high ratio of Treg to tumor-specific CD4+ T cell) in KRASG12DTP53mut patients, KRASG12VTP53wt individuals exhibited an inflamed context profiled by multiplex immunohistochemistry. It could be responsible for their outstanding survival advantage over others in postsurgical PAAD patients receiving adjuvant chemotherapy as shown by our cohort. Together, KRASG12VTP53wt may be a promising biomarker for prognostic evaluation and screening certain candidates with PAAD to get desirable survival benefit from adjuvant chemotherapy.
Collapse
Affiliation(s)
- Siyi Zou
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Er Road, Shanghai, 200025, PR China; Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, PR China
| | - Lei Zhang
- Genecast Biotechnology Co., Ltd, 88 Danshan Road, Xidong Chuangrong Building, Suite C 1310-1318, Xishan District, Wuxi City, Jiangsu, 214104, PR China
| | - Cen Jiang
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Er Road, Shanghai, 200025, PR China
| | - Fanlu Li
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Er Road, Shanghai, 200025, PR China; Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, PR China
| | - Ying Yang
- Genecast Biotechnology Co., Ltd, 88 Danshan Road, Xidong Chuangrong Building, Suite C 1310-1318, Xishan District, Wuxi City, Jiangsu, 214104, PR China
| | - Xiaxing Deng
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Er Road, Shanghai, 200025, PR China; Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, PR China
| | - Jiao Zhang
- Genecast Biotechnology Co., Ltd, 88 Danshan Road, Xidong Chuangrong Building, Suite C 1310-1318, Xishan District, Wuxi City, Jiangsu, 214104, PR China
| | - Hao Chen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Er Road, Shanghai, 200025, PR China; Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, PR China
| | - Lingxi Jiang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Er Road, Shanghai, 200025, PR China; Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, PR China
| | - Xueyan Cheng
- Genecast Biotechnology Co., Ltd, 88 Danshan Road, Xidong Chuangrong Building, Suite C 1310-1318, Xishan District, Wuxi City, Jiangsu, 214104, PR China
| | - Lisha Deng
- Genecast Biotechnology Co., Ltd, 88 Danshan Road, Xidong Chuangrong Building, Suite C 1310-1318, Xishan District, Wuxi City, Jiangsu, 214104, PR China
| | - Lin Lin
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Er Road, Shanghai, 200025, PR China.
| | - Baiyong Shen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Er Road, Shanghai, 200025, PR China; Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, PR China.
| | - Chenlei Wen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Er Road, Shanghai, 200025, PR China; Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, PR China.
| | - Qian Zhan
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Er Road, Shanghai, 200025, PR China; Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, PR China.
| |
Collapse
|
45
|
Jiang S, Tang Y, Wang X, Guo H, Chen L, Hu G, Cui Y, Liang S, Zuo J, Luo Z, Chen X, Wang X. ARHGAP4 promotes colon cancer metastasis through the TGF-β signaling pathway and may be associated with T cell exhaustion. Biochem Biophys Res Commun 2024; 722:150172. [PMID: 38805788 DOI: 10.1016/j.bbrc.2024.150172] [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: 04/02/2024] [Revised: 05/13/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Colon cancer is a prevalent invasive neoplasm in the gastrointestinal system with a high degree of malignancy. Despite extensive research, the underlying mechanisms of its recurrence and metastasis remain elusive.Rho GTPase activating protein 4 (ARHGAP4), a member of the small GTPases protein family, may be closely related to tumor metastasis, and its expression is increased in colon cancer. However, the role of ARHGAP4 in colon cancer metastasis is uncertain. This study investigates the impact of ARHGAP4 on the metastasis of colon cancer cells. Our objective is to determine the role of ARHGAP4 in regulating the invasive behavior of colon cancer cells. METHODS We downloaded colon adenocarcinoma (COAD) data from the Cancer Genome Atlas (TCGA), and performed differential analysis and survival analysis. By using the CIBERSORT algorithm, we evaluated the proportion of infiltrating immune cells in colon cancer. We further analyzed whether ARHGAP4 is associated with T cell exhaustion. Finally, we investigated the impact of ARHGAP4 knockdown on the migration and invasion of colon cancer cells through in vitro cell experiments. Additionally, we utilized western blotting to assess the expression of protein related to the TGF-β signaling pathway and epithelial-mesenchymal transition (EMT). RESULTS We found that ARHGAP4 is upregulated in colon cancer. Subsequent survival analysis revealed that the high-expression group had significantly lower survival rates compared to the low-expression group. Immune infiltration analysis showed that ARHGAP4 was not only positively correlated with CD8+ T cells, but also positively correlated with T cell exhaustion markers programmed cell death 1 (PDCD-1), cytotoxic T-lymphocyte associated protein 4 (CTLA-4), and lymphocyte activating 3 (LAG-3). In vitro cell experiments, the knockdown of ARHGAP4 inhibited the migration and invasion of colon cancer cells. Among EMT-related proteins, when ARHGAP4 was knocked down, the expression of E-cadherin was increased, while the expression of N-cadherin and Vimentin was decreased. Meanwhile, the expression of TGF-β1, p-Smad2, and p-Smad3, which are associated with the TGF-β/Smad pathway, all decreased. CONCLUSION ARHGAP4 promotes colon cancer metastasis through the TGF-β/Smad signaling pathway and may be associated with T cell exhaustion. It plays an important role in the progression of colon cancer and may serve as a potential target for diagnosis and treatment of colon cancer.
Collapse
Affiliation(s)
- Shuanghong Jiang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Shunqing District, Nanchong City, 637000, Sichuan, China; Digestive Endoscopy Center, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Shunqing District, Nanchong City, 637000, Sichuan, China
| | - Yong Tang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Shunqing District, Nanchong City, 637000, Sichuan, China
| | - Xiaobo Wang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Shunqing District, Nanchong City, 637000, Sichuan, China
| | - Haiyang Guo
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Shunqing District, Nanchong City, 637000, Sichuan, China
| | - Lin Chen
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Shunqing District, Nanchong City, 637000, Sichuan, China
| | - Guangbing Hu
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Shunqing District, Nanchong City, 637000, Sichuan, China
| | - Yutong Cui
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Shunqing District, Nanchong City, 637000, Sichuan, China
| | - Shiqi Liang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Shunqing District, Nanchong City, 637000, Sichuan, China
| | - Ji Zuo
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Shunqing District, Nanchong City, 637000, Sichuan, China
| | - Zichen Luo
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Shunqing District, Nanchong City, 637000, Sichuan, China
| | - Xinrui Chen
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Shunqing District, Nanchong City, 637000, Sichuan, China
| | - Xianfei Wang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Shunqing District, Nanchong City, 637000, Sichuan, China; Digestive Endoscopy Center, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Shunqing District, Nanchong City, 637000, Sichuan, China.
| |
Collapse
|
46
|
Zhang J, Du B, Wang Y, Cui Y, Wang S, Zhao Y, Li Y, Li X. The role of CD8 PET imaging in guiding cancer immunotherapy. Front Immunol 2024; 15:1428541. [PMID: 39072335 PMCID: PMC11272484 DOI: 10.3389/fimmu.2024.1428541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024] Open
Abstract
Currently, immunotherapy is being widely used for treating cancers. However, the significant heterogeneity in patient responses is a major challenge for its successful application. CD8-positive T cells (CD8+ T cells) play a critical role in immunotherapy. Both their infiltration and functional status in tumors contribute to treatment outcomes. Therefore, accurate monitoring of CD8+ T cells, a potential biomarker, may improve therapeutic strategy. Positron emission tomography (PET) is an optimal option which can provide molecular imaging with enhanced specificity. This review summarizes the mechanism of action of CD8+ T cells in immunotherapy, and highlights the recent advancements in PET-based tracers that can visualize CD8+ T cells and discusses their clinical applications to elucidate their potential role in cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yaming Li
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xuena Li
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
47
|
Wu H, Wu Z, Li H, Wang Z, Chen Y, Bao J, Chen B, Xu S, Xia E, Ye D, Dai X. Glycosylphosphatidylinositol anchor biosynthesis pathway-based biomarker identification with machine learning for prognosis and T cell exhaustion status prediction in breast cancer. Front Immunol 2024; 15:1392940. [PMID: 39015576 PMCID: PMC11249538 DOI: 10.3389/fimmu.2024.1392940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/11/2024] [Indexed: 07/18/2024] Open
Abstract
As the primary component of anti-tumor immunity, T cells are prone to exhaustion and dysfunction in the tumor microenvironment (TME). A thorough understanding of T cell exhaustion (TEX) in the TME is crucial for effectively addressing TEX in clinical settings and promoting the efficacy of immune checkpoint blockade therapies. In eukaryotes, numerous cell surface proteins are tethered to the plasma membrane via Glycosylphosphatidylinositol (GPI) anchors, which play a crucial role in facilitating the proper translocation of membrane proteins. However, the available evidence is insufficient to support any additional functional involvement of GPI anchors. Here, we investigate the signature of GPI-anchor biosynthesis in the TME of breast cancer (BC)patients, particularly its correlation with TEX. GPI-anchor biosynthesis should be considered as a prognostic risk factor for BC. Patients with high GPI-anchor biosynthesis showed more severe TEX. And the levels of GPI-anchor biosynthesis in exhausted CD8 T cells was higher than normal CD8 T cells, which was not observed between malignant epithelial cells and normal mammary epithelial cells. In addition, we also found that GPI -anchor biosynthesis related genes can be used to diagnose TEX status and predict prognosis in BC patients, both the TEX diagnostic model and the prognostic model showed good AUC values. Finally, we confirmed our findings in cells and clinical samples. Knockdown of PIGU gene expression significantly reduced the proliferation rate of MDA-MB-231 and MCF-7 cell lines. Immunofluorescence results from clinical samples showed reduced aggregation of CD8 T cells in tissues with high expression of GPAA1 and PIGU.
Collapse
Affiliation(s)
- Haodong Wu
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhixuan Wu
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hongfeng Li
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ziqiong Wang
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yao Chen
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingxia Bao
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Buran Chen
- School of Molecular Science, University of Western Australia, Perth, WA, Australia
| | - Shuning Xu
- Department of Computer Information Systems, Georgia State University, Atlanta, GA, United States
| | - Erjie Xia
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Daijiao Ye
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xuanxuan Dai
- Department of Breast Surgery, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
48
|
Chen H, Wu Y, Wang J, Li Y, Chen Y, Wang X, Lv H, Liu X. Tilianin enhances the antitumor effect of sufentanil on non-small cell lung cancer. J Biochem Mol Toxicol 2024; 38:e23761. [PMID: 38952040 DOI: 10.1002/jbt.23761] [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: 01/02/2024] [Revised: 04/18/2024] [Accepted: 06/24/2024] [Indexed: 07/03/2024]
Abstract
Non-small cell cancer (NSCLC) is the most common cancer in the world, but its effective therapeutic methods are limited. Tilianin and sufentanil alleviate various human tumors. This research aimed to clarify the functions and mechanisms of Tilianin and sufentanil in NSCLC. The functions of Tilianin and sufentanil on NSCLC cell viability, apoptosis, mitochondrial dysfunction, and immunity in vitro were examined using Cell Counting Kit-8 assay, flow cytometry, reactive oxygen species level analysis, CD8+ T cell percentage analysis, Western blot, and enzyme-linked immunosorbent assay, respectively. The molecular mechanism regulated by Tilianin and sufentanil in NSCLC was assessed using Western blot, and immunofluorescence assays. Meanwhile, the roles of Tilianin and sufentanil in NSCLC tumor growth, apoptosis, and immunity in vivo were determined by establishing a tumor xenograft mouse model, immunohistochemistry, and Western blot assays. When sufentanil concentration was proximity 2 nM, the inhibition rate of NSCLC cell viability was 50%. The IC50 for A549 cells was 2.36 nM, and the IC50 for H1299 cells was 2.18 nM. The IC50 of Tilianin for A549 cells was 38.7 μM, and the IC50 of Tilianin for H1299 cells was 44.6 μM. Functionally, 0.5 nM sufentanil and 10 μM Tilianin reduced NSCLC cell (A549 and H1299) viability in a dose-dependent manner. Also, 0.5 nM sufentanil and 10 μM Tilianin enhanced NSCLC cell apoptosis, yet this impact was strengthened after a combination of Tilianin and Sufentanil. Furthermore, 0.5 nM sufentanil and 10 μM Tilianin repressed NSCLC cell mitochondrial dysfunction and immunity, and these impacts were enhanced after a combination of Tilianin and Sufentanil. Mechanistically, 0.5 nM sufentanil and 10 μM Tilianin repressed the NF-κB pathway in NSCLC cells, while this repression was strengthened after a combination of Tilianin and Sufentanil. In vivo experimental data further clarified that 1 µg/kg sufentanil and 10 mg/kg Tilianin reduced NSCLC growth, immunity, and NF-κB pathway-related protein levels, yet these trends were enhanced after a combination of Tilianin and Sufentanil. Tilianin strengthened the antitumor effect of sufentanil in NSCLC.
Collapse
Affiliation(s)
- Huixia Chen
- Department of Anesthesiology, Handan Central Hospital, Handan, China
| | - Yulin Wu
- Department of Anesthesia Operation Center, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Jiazheng Wang
- Department of Anesthesiology, Handan Central Hospital, Handan, China
| | - Yike Li
- Department of Anesthesiology, Handan Central Hospital, Handan, China
| | - Yongxue Chen
- Department of Anesthesiology, Handan Central Hospital, Handan, China
| | - Xinbo Wang
- Department of Anesthesiology, Handan Central Hospital, Handan, China
| | - Hangyu Lv
- Department of Anesthesiology, Handan Central Hospital, Handan, China
| | - Xinyan Liu
- Department of Anesthesiology, Handan Central Hospital, Handan, China
| |
Collapse
|
49
|
Yue B, Gao Y, Hu Y, Zhan M, Wu Y, Lu L. Harnessing CD8 + T cell dynamics in hepatitis B virus-associated liver diseases: Insights, therapies and future directions. Clin Transl Med 2024; 14:e1731. [PMID: 38935536 PMCID: PMC11210506 DOI: 10.1002/ctm2.1731] [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: 02/05/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/29/2024] Open
Abstract
Hepatitis B virus (HBV) infection playsa significant role in the etiology and progression of liver-relatedpathologies, encompassing chronic hepatitis, fibrosis, cirrhosis, and eventual hepatocellularcarcinoma (HCC). Notably, HBV infection stands as the primary etiologicalfactor driving the development of HCC. Given the significant contribution ofHBV infection to liver diseases, a comprehensive understanding of immunedynamics in the liver microenvironment, spanning chronic HBV infection,fibrosis, cirrhosis, and HCC, is essential. In this review, we focused on thefunctional alterations of CD8+ T cells within the pathogenic livermicroenvironment from HBV infection to HCC. We thoroughly reviewed the roles ofhypoxia, acidic pH, metabolic reprogramming, amino acid deficiency, inhibitory checkpointmolecules, immunosuppressive cytokines, and the gut-liver communication in shapingthe dysfunction of CD8+ T cells in the liver microenvironment. Thesefactors significantly impact the clinical prognosis. Furthermore, we comprehensivelyreviewed CD8+ T cell-based therapy strategies for liver diseases,encompassing HBV infection, fibrosis, cirrhosis, and HCC. Strategies includeimmune checkpoint blockades, metabolic T-cell targeting therapy, therapeuticT-cell vaccination, and adoptive transfer of genetically engineered CD8+ T cells, along with the combined usage of programmed cell death protein-1/programmeddeath ligand-1 (PD-1/PD-L1) inhibitors with mitochondria-targeted antioxidants.Given that targeting CD8+ T cells at various stages of hepatitis Bvirus-induced hepatocellular carcinoma (HBV + HCC) shows promise, we reviewedthe ongoing need for research to elucidate the complex interplay between CD8+ T cells and the liver microenvironment in the progression of HBV infection toHCC. We also discussed personalized treatment regimens, combining therapeuticstrategies and harnessing gut microbiota modulation, which holds potential forenhanced clinical benefits. In conclusion, this review delves into the immunedynamics of CD8+ T cells, microenvironment changes, and therapeuticstrategies within the liver during chronic HBV infection, HCC progression, andrelated liver diseases.
Collapse
Affiliation(s)
- Bing Yue
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and TreatmentZhuhai Institute of Translational MedicineZhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Jinan UniversityZhuhaiGuangdongChina
| | - Yuxia Gao
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and TreatmentZhuhai Institute of Translational MedicineZhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Jinan UniversityZhuhaiGuangdongChina
| | - Yi Hu
- Microbiology and Immunology DepartmentSchool of MedicineFaculty of Medical ScienceJinan UniversityGuangzhouGuangdongChina
| | - Meixiao Zhan
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and TreatmentZhuhai Institute of Translational MedicineZhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Jinan UniversityZhuhaiGuangdongChina
| | - Yangzhe Wu
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and TreatmentZhuhai Institute of Translational MedicineZhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Jinan UniversityZhuhaiGuangdongChina
| | - Ligong Lu
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and TreatmentZhuhai Institute of Translational MedicineZhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Jinan UniversityZhuhaiGuangdongChina
| |
Collapse
|
50
|
Zhang X, Cui X, Li P, Zhao Y, Ren Y, Zhang H, Zhang S, Li C, Wang X, Shi L, Sun T, Hao J, Yao Z, Chen J, Gao X, Yang J. EGC enhances tumor antigen presentation and CD8 + T cell-mediated antitumor immunity via targeting oncoprotein SND1. Cancer Lett 2024; 592:216934. [PMID: 38710299 DOI: 10.1016/j.canlet.2024.216934] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 05/08/2024]
Abstract
The Staphylococcal nuclease and Tudor domain containing 1 (SND1) has been identified as an oncoprotein. Our previous study demonstrated that SND1 impedes the major histocompatibility complex class I (MHC-I) assembly by hijacking the nascent heavy chain of MHC-I to endoplasmic reticulum-associated degradation. Herein, we aimed to identify inhibitors to block SND1-MHC-I binding, to facilitate the MHC-I presentation and tumor immunotherapy. Our findings validated the importance of the K490-containing sites in SND1-MHC-I complex. Through structure-based virtual screening and docking analysis, (-)-Epigallocatechin (EGC) exhibited the highest docking score to prevent the binding of MHC-I to SND1 by altering the spatial conformation of SND1. Additionally, EGC treatment resulted in increased expression levels of membrane-presented MHC-I in tumor cells. The C57BL/6J murine orthotopic melanoma model validated that EGC increases infiltration and activity of CD8+ T cells in both the tumor and spleen. Furthermore, the combination of EGC with programmed death-1 (PD-1) antibody demonstrated a superior antitumor effect. In summary, we identified EGC as a novel inhibitor of SND1-MHC-I interaction, prompting MHC-I presentation to improve CD8+ T cell response within the tumor microenvironment. This discovery presents a promising immunotherapeutic candidate for tumors.
Collapse
Affiliation(s)
- Xinxin Zhang
- Tianjin Key Laboratory of Cellular and Molecular Immunology, and Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin, China; The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China; State Key Laboratory of Experimental Hematology, Tianjin, China; Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Xiaoteng Cui
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Peiying Li
- Tianjin Key Laboratory of Cellular and Molecular Immunology, and Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin, China; The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China; State Key Laboratory of Experimental Hematology, Tianjin, China; Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Yan Zhao
- Tianjin Key Laboratory of Cellular and Molecular Immunology, and Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin, China; The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China; State Key Laboratory of Experimental Hematology, Tianjin, China; Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Yuanyuan Ren
- Tianjin Key Laboratory of Cellular and Molecular Immunology, and Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin, China; The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China; State Key Laboratory of Experimental Hematology, Tianjin, China; Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Heng Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Shijie Zhang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Chufeng Li
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xinting Wang
- Tianjin Key Laboratory of Cellular and Molecular Immunology, and Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin, China; The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China; State Key Laboratory of Experimental Hematology, Tianjin, China; Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Lei Shi
- Tianjin Key Laboratory of Cellular and Molecular Immunology, and Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin, China; The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China; State Key Laboratory of Experimental Hematology, Tianjin, China; Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Tao Sun
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Jihui Hao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhi Yao
- Tianjin Key Laboratory of Cellular and Molecular Immunology, and Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin, China; The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China; State Key Laboratory of Experimental Hematology, Tianjin, China; Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Jun Chen
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin, China.
| | - Xingjie Gao
- Tianjin Key Laboratory of Cellular and Molecular Immunology, and Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin, China; The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China; State Key Laboratory of Experimental Hematology, Tianjin, China; Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China.
| | - Jie Yang
- Tianjin Key Laboratory of Cellular and Molecular Immunology, and Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin, China; The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China; State Key Laboratory of Experimental Hematology, Tianjin, China; Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China.
| |
Collapse
|