1
|
Yan H, Ji X, Li B. Advancing personalized, predictive, and preventive medicine in bladder cancer: a multi-omics and machine learning approach for novel prognostic modeling, immune profiling, and therapeutic target discovery. Front Immunol 2025; 16:1572034. [PMID: 40330458 PMCID: PMC12053186 DOI: 10.3389/fimmu.2025.1572034] [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: 02/06/2025] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
Objective This study aimed to identify and analyze immunogenic cell death (ICD)-related multi-omics features in bladder cancer (BLCA) using single-cell RNA sequencing (scRNA-seq) and bulk RNA-seq data. By integrating these datasets, we sought to construct a prognostic signature (ICDRS) and explore its clinical and biological implications, including its association with immune cell infiltration, tumor microenvironment (TME), and drug sensitivity. Methods Publicly available datasets from TCGA and GEO, including scRNA-seq (GSE222315, 9 samples) and bulk RNA-seq (TCGA-BLCA, 403 samples; GSE13507, 160 samples), were analyzed. Single-cell data were processed using Seurat, and ICD scores were calculated using single-sample gene set enrichment analysis (ssGSEA). Weighted gene co-expression network analysis (WGCNA) identified ICD-related modules, and machine learning algorithms (Lasso, Ridge, CoxBoost) were employed to construct the ICDRS. Survival analysis, immune infiltration, pathway enrichment, and drug sensitivity were evaluated to validate the model. Results The ICDRS, based on eight key genes (IL32, AHNAK, ANXA5, FN1, GSN, CNN3, FXYD3, CTSS), effectively stratified BLCA patients into high- and low-risk groups with significant differences in overall survival (OS, P < 0.001). High ICDRS scores were associated with immune-suppressive TME, including increased infiltration of T cells CD4 memory resting (P = 0.02) and macrophages M0/M1/M2 (P = 0.01). Pathway enrichment revealed correlations with cholesterol homeostasis, epithelial-mesenchymal transition (EMT), and KRAS signaling. Drug sensitivity analysis showed high-risk groups were resistant to Cisplatin (P = 0.003), Mitomycin C (P = 0.01), and Paclitaxel (P = 0.004), with IC50 values significantly higher than low-risk groups. Conclusion The ICDRS serves as a robust prognostic biomarker for BLCA, offering insights into tumor immune evasion mechanisms and potential therapeutic targets. Its integration with clinical features enhances personalized treatment strategies, highlighting the importance of ICD in BLCA immunotherapy and precision medicine. The model's predictive accuracy and biological relevance were validated across multiple datasets, underscoring its potential for clinical application.
Collapse
Affiliation(s)
- Han Yan
- Department of Pain Medicine, The First Hospital of China Medical University, Liaoning, Shenyang, China
| | - Xinyu Ji
- Department of Thoracic Surgery, The First Hospital of China Medical University, Liaoning, Shenyang, China
| | - Bohan Li
- Department of Urinary Surgery, The First Hospital of China Medical University, Liaoning, Shenyang, China
| |
Collapse
|
2
|
Li M, Cui Y, Wu X, Yang X, Huang C, Yu L, Yi P, Chen C. Integrating network pharmacology to investigate the mechanism of quercetin's action through AKT inhibition in co-expressed genes associated with polycystic ovary syndrome and endometrial cancer. Int J Biol Macromol 2025; 297:139468. [PMID: 39765297 DOI: 10.1016/j.ijbiomac.2025.139468] [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: 10/14/2024] [Revised: 12/30/2024] [Accepted: 01/01/2025] [Indexed: 01/30/2025]
Abstract
Endometrial cancer (EC) is a common gynecological malignancy for which polycystic ovarian syndrome (PCOS) has been identified as a significant risk factor. Quercetin, a widely distributed natural flavonoid, has demonstrated potential therapeutic effects in managing both PCOS and EC. However, the specific molecular targets of quercetin in the context of PCOS comorbid with EC (PCOS-EC) remain poorly defined. This study aims to elucidate the therapeutic potential of quercetin for treating PCOS-EC using network pharmacology, molecular dynamics simulations, and in vitro assays. The intersection of 379 PCOS-EC-associated targets with 361 quercetin targets identified 47 potential therapeutic targets of quercetin for PCOS-EC. Gene Ontology enrichment analysis revealed the biological functions, while Kyoto Encyclopedia of Genes and Genomes identified the pathways potentially involved in quercetin's effects against PCOS-EC. Protein-protein interaction network analysis highlighted six overlapping targets, namely, ACTB, AKT1, EGFR, ESR1, PTGS2, and TP53. Molecular docking and molecular dynamics simulations indicated that quercetin bound with high affinity to the hub genes, with AKT1 emerging as a central target. In vitro experiments confirmed that quercetin treatment significantly downregulated AKT expression in EC cells. These findings elucidate potential targets and molecular mechanisms through which quercetin exerts its therapeutic effects.
Collapse
Affiliation(s)
- Mengyuan Li
- Department of Obstetrics and Gynecology, Chongqing General Hospital, Chongqing University, Chongqing 401147, China; Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Yewei Cui
- School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xingfan Wu
- School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xunmei Yang
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Chenglong Huang
- Department of Clinical Laboratory, Medical Sciences Research Center, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Lili Yu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Ping Yi
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China.
| | - Cheng Chen
- Department of Obstetrics and Gynecology, Chongqing General Hospital, Chongqing University, Chongqing 401147, China.
| |
Collapse
|
3
|
Tang K, Tian J, Xu Y, Shang G, Peng X, Yue P, Wang Y, Chen S, Hu Z. Aflatoxin B1 Exposure Suppresses the Migration of Dendritic Cells by Reshaping the Cytoskeleton. Int J Mol Sci 2025; 26:1725. [PMID: 40004187 PMCID: PMC11854954 DOI: 10.3390/ijms26041725] [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: 01/28/2025] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Exposure to Aflatoxin B1 (AFB1) is considered a significant risk factor for human diseases, including the immune function impairment of immune cells. Dendritic cells (DCs), as essential antigen-presenting cells, play a pivotal role in bridging innate and adaptive immunity. However, the impact of AFB1 exposure on DCs has not been fully elucidated. In this study, we investigated the effects of AFB1 exposure on the migration ability of DCs and its underlying action model. Initially, we observed that AFB1 exposure inhibited the survival of DCs and altered their cellular morphology. Further investigation revealed that AFB1 promotes cell adhesion and inhibits DC migration by modulating the expression of cell adhesion molecules. Additionally, our findings indicated that cytoskeletal remodeling plays a crucial role in these processes. Experimental techniques such as immunofluorescence and RNA sequencing confirmed that AFB1 exposure regulates the expression of cytoskeleton-related genes. Moreover, we found that the perturbation of the gene expression profile through AFB1 exposure is associated with cell communication. Collectively, our study findings demonstrate that AFB1 can disrupt the expression of cytoskeleton- and adhesion-related molecules in DCs, thereby altering cell morphology and migration. These insights could provide new perspectives for further understanding the immunosuppressive effects of AFB1 and developing therapeutic strategies for diseases associated with AFB1 exposure.
Collapse
Affiliation(s)
- Kaiyi Tang
- Immune Cells and Antibody Engineering Research Center in University of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (School of Modern Industry for Health and Medicine)/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; (K.T.); (J.T.); (Y.X.); (G.S.); (P.Y.); (Y.W.)
| | - Jiaxiong Tian
- Immune Cells and Antibody Engineering Research Center in University of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (School of Modern Industry for Health and Medicine)/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; (K.T.); (J.T.); (Y.X.); (G.S.); (P.Y.); (Y.W.)
| | - Yujun Xu
- Immune Cells and Antibody Engineering Research Center in University of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (School of Modern Industry for Health and Medicine)/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; (K.T.); (J.T.); (Y.X.); (G.S.); (P.Y.); (Y.W.)
- Key Laboratory of Infectious Immune and Antibody Engineering in University of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Guizhou Medical University, Guiyang 550025, China;
| | - Guofu Shang
- Immune Cells and Antibody Engineering Research Center in University of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (School of Modern Industry for Health and Medicine)/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; (K.T.); (J.T.); (Y.X.); (G.S.); (P.Y.); (Y.W.)
- Key Laboratory of Infectious Immune and Antibody Engineering in University of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Guizhou Medical University, Guiyang 550025, China;
| | - Xiaoyan Peng
- Key Laboratory of Infectious Immune and Antibody Engineering in University of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Guizhou Medical University, Guiyang 550025, China;
| | - Ping Yue
- Immune Cells and Antibody Engineering Research Center in University of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (School of Modern Industry for Health and Medicine)/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; (K.T.); (J.T.); (Y.X.); (G.S.); (P.Y.); (Y.W.)
| | - Yun Wang
- Immune Cells and Antibody Engineering Research Center in University of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (School of Modern Industry for Health and Medicine)/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; (K.T.); (J.T.); (Y.X.); (G.S.); (P.Y.); (Y.W.)
| | - Sen Chen
- Immune Cells and Antibody Engineering Research Center in University of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (School of Modern Industry for Health and Medicine)/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; (K.T.); (J.T.); (Y.X.); (G.S.); (P.Y.); (Y.W.)
| | - Zuquan Hu
- Immune Cells and Antibody Engineering Research Center in University of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (School of Modern Industry for Health and Medicine)/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; (K.T.); (J.T.); (Y.X.); (G.S.); (P.Y.); (Y.W.)
- Key Laboratory of Infectious Immune and Antibody Engineering in University of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Guizhou Medical University, Guiyang 550025, China;
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guizhou Medical University, Guiyang 550025, China
| |
Collapse
|
4
|
Seo Y, Jang J, Ko KP, Zou G, Huang Y, Zhang S, Zhang J, Jun S, Chu W, Venkatesan V, Dhakshinamoorthy S, Park JI. Actin dysregulation induces immune evasion via oxidative stress-activated PD-L1 in gastric cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.18.629227. [PMID: 39763993 PMCID: PMC11702617 DOI: 10.1101/2024.12.18.629227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Diffuse gastric adenocarcinoma (DGAC) is an aggressive malignancy with limited therapeutic options, poor prognosis, and poorly understood biology. CRACD, an actin polymerization regulator, is often inactivated in gastric cancer, including DGAC. We found that genetic engineering of murine gastric organoids with Cracd ablation combined with Kras mutation and Trp53 loss induced aberrant cell plasticity, hyperproliferation, and hypermucinosis, the features that recapitulate DGAC transcriptional signatures. Notably, CRACD inactivation remodeled the immune landscape for immune evasion through PD-L1 enrichment in tumor cells. Mechanistically, CRACD loss disrupted actin dynamics, generating reactive oxygen species that activated HIF1α, which transactivated PD-L1 . Pharmacologic inhibition of HIF1α or PD-L1 restored immune surveillance and suppressed tumorigenesis. These findings reveal a novel role of actin homeostasis in limiting cell plasticity and immune evasion, position CRACD as a potential biomarker for stratifying patients with DGAC, and highlight HIF1α and PD-L1 as actionable therapeutic targets.
Collapse
|
5
|
Izquierdo M, Ruiz-Navarro J, Baldari CT, Roda-Navarro P. Editorial: Structure and function of the immunological and redirecting artificial synapses and their clinical implications. Front Immunol 2024; 15:1497118. [PMID: 39421740 PMCID: PMC11484066 DOI: 10.3389/fimmu.2024.1497118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Affiliation(s)
- Manuel Izquierdo
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Translational Research in Pediatric Oncology, Hematopoietic Transplantation and Cell Therapy, Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Javier Ruiz-Navarro
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Translational Research in Pediatric Oncology, Hematopoietic Transplantation and Cell Therapy, Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | | | - Pedro Roda-Navarro
- Department of Immunology, Ophthalmology and ear, nose and throat (ENT), School of Medicine, Universidad Complutense, Madrid, Spain
- Lymphocyte Immunobiology Group, Instituto de Investigación Sanitaria12 de Octubre (imas12), Madrid, Spain
| |
Collapse
|
6
|
Abou Kors T, Meier M, Mühlenbruch L, Betzler AC, Oliveri F, Bens M, Thomas J, Kraus JM, Doescher J, von Witzleben A, Hofmann L, Ezic J, Huber D, Benckendorff J, Barth TFE, Greve J, Schuler PJ, Brunner C, Blackburn JM, Hoffmann TK, Ottensmeier C, Kestler HA, Rammensee HG, Walz JS, Laban S. Multi-omics analysis of overexpressed tumor-associated proteins: gene expression, immunopeptide presentation, and antibody response in oropharyngeal squamous cell carcinoma, with a focus on cancer-testis antigens. Front Immunol 2024; 15:1408173. [PMID: 39136024 PMCID: PMC11317303 DOI: 10.3389/fimmu.2024.1408173] [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: 03/27/2024] [Accepted: 07/15/2024] [Indexed: 08/15/2024] Open
Abstract
Introduction The human leukocyte antigen complex (HLA) is essential for inducing specific immune responses to cancer by presenting tumor-associated peptides (TAP) to T cells. Overexpressed tumor associated antigens, mainly cancer-testis antigens (CTA), are outlined as essential targets for immunotherapy in oropharyngeal squamous cell carcinoma (OPSCC). This study assessed the degree to which presentation, gene expression, and antibody response (AR) of TAP, mainly CTA, are correlated in OPSCC patients to evaluate their potential as immunotherapy targets. Materials and methods Snap-frozen tumor (NLigand/RNA=40), healthy mucosa (NRNA=6), and healthy tonsils (NLigand=5) samples were obtained. RNA-Seq was performed using Illumina HiSeq 2500/NovaSeq 6000 and whole exome sequencing (WES) utilizing NextSeq500. HLA ligands were isolated from tumor tissue using immunoaffinity purification, UHPLC, and analyzed by tandem MS. Antibodies were measured in serum (NAb=27) utilizing the KREX™ CT262 protein array. Data analysis focused on 312 proteins (KREX™ CT262 panel + overexpressed self-proteins). Results 183 and 94 of HLA class I and II TAP were identified by comparative profiling with healthy tonsils. Genes from 26 TAP were overexpressed in tumors compared to healthy mucosa (LFC>1; FDR<0.05). Low concordance (r=0.25; p<0.0001) was found between upregulated mRNA and class I TAP. The specific mode of correlation of TAP was found to be dependent on clinical parameters. A lack of correlation was observed both between mRNA and class II TAP, as well as between class II tumor-unique TAP (TAP-U) presentation and antibody response (AR) levels. Discussion This study demonstrates that focusing exclusively on gene transcript levels fails to capture the full extent of TAP presentation in OPSCC. Furthermore, our findings reveal that although CTA are presented at relatively low levels, a few CTA TAP-U show potential as targets for immunotherapy.
Collapse
Affiliation(s)
- Tsima Abou Kors
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Matthias Meier
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Lena Mühlenbruch
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Eberhard Karls University and University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), Partner Site Tübingen, Tübingen, Germany
| | - Annika C. Betzler
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
- Core Facility Immune Monitoring, Ulm University Medical Center, Ulm, Germany
| | - Franziska Oliveri
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Martin Bens
- Core Facility Next Generation Sequencing, Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
| | - Jaya Thomas
- Cancer Sciences Unit, University of Southampton, Faculty of Medicine, Southampton, United Kingdom
| | - Johann M. Kraus
- Institute of Medical Systems Biology, Faculty of Medicine, Ulm University, Ulm, Germany
| | - Johannes Doescher
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
- Department of Otolaryngology, Augsburg University Hospital, Augsburg, Germany
| | - Adrian von Witzleben
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Linda Hofmann
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Jasmin Ezic
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Diana Huber
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | | | | | - Jens Greve
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Patrick J. Schuler
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
- Surgical Oncology Ulm, i2SOUL Consortium, Ulm, Germany
| | - Cornelia Brunner
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
- Core Facility Immune Monitoring, Ulm University Medical Center, Ulm, Germany
| | - Jonathan M. Blackburn
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Thomas K. Hoffmann
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
- Surgical Oncology Ulm, i2SOUL Consortium, Ulm, Germany
| | - Christian Ottensmeier
- Institute of Systems, Molecular and Integrative Biology, Liverpool Head and Neck Center, University of Liverpool, Faculty of Medicine, Liverpool, United Kingdom
| | - Hans A. Kestler
- Institute of Medical Systems Biology, Faculty of Medicine, Ulm University, Ulm, Germany
- Surgical Oncology Ulm, i2SOUL Consortium, Ulm, Germany
| | - Hans-Georg Rammensee
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), Partner Site Tübingen, Tübingen, Germany
| | - Juliane S. Walz
- Department of Peptide-based Immunotherapy, Eberhard Karls University and University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), Partner Site Tübingen, Tübingen, Germany
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Simon Laban
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
- Surgical Oncology Ulm, i2SOUL Consortium, Ulm, Germany
| |
Collapse
|
7
|
Martinez-Morga M, Garrigos D, Rodriguez-Montero E, Pombero A, Garcia-Lopez R, Martinez S. Pericytes Are Immunoregulatory Cells in Glioma Genesis and Progression. Int J Mol Sci 2024; 25:5072. [PMID: 38791110 PMCID: PMC11120873 DOI: 10.3390/ijms25105072] [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: 03/13/2024] [Revised: 05/03/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
Vascular co-option is a consequence of the direct interaction between perivascular cells, known as pericytes (PCs), and glioblastoma multiforme (GBM) cells (GBMcs). This process is essential for inducing changes in the pericytes' anti-tumoral and immunoreactive phenotypes. Starting from the initial stages of carcinogenesis in GBM, PCs conditioned by GBMcs undergo proliferation, acquire a pro-tumoral and immunosuppressive phenotype by expressing and secreting immunosuppressive molecules, and significantly hinder the activation of T cells, thereby facilitating tumor growth. Inhibiting the pericyte (PC) conditioning mechanisms in the GBM tumor microenvironment (TME) results in immunological activation and tumor disappearance. This underscores the pivotal role of PCs as a key cell in the TME, responsible for tumor-induced immunosuppression and enabling GBM cells to evade the immune system. Other cells within the TME, such as tumor-associated macrophages (TAMs) and microglia, have also been identified as contributors to this immunomodulation. In this paper, we will review the role of these three cell types in the immunosuppressive properties of the TME. Our conclusion is that the cellular heterogeneity of immunocompetent cells within the TME may lead to the misinterpretation of cellular lineage identification due to different reactive stages and the identification of PCs as TAMs. Consequently, novel therapies could be developed to disrupt GBM-PC interactions and/or PC conditioning through vascular co-option, thereby exposing GBMcs to the immune system.
Collapse
Affiliation(s)
- Marta Martinez-Morga
- Instituto de Neurociencias, Universidad Miguel Hernández–CSIC, Excellence Center Severo Ochoa, Campus de San Juan, Avda. Ramón y Cajal sn, 03550 Alicante, Spain; (M.M.-M.); (D.G.); (E.R.-M.); (A.P.); (R.G.-L.)
| | - Daniel Garrigos
- Instituto de Neurociencias, Universidad Miguel Hernández–CSIC, Excellence Center Severo Ochoa, Campus de San Juan, Avda. Ramón y Cajal sn, 03550 Alicante, Spain; (M.M.-M.); (D.G.); (E.R.-M.); (A.P.); (R.G.-L.)
| | - Elena Rodriguez-Montero
- Instituto de Neurociencias, Universidad Miguel Hernández–CSIC, Excellence Center Severo Ochoa, Campus de San Juan, Avda. Ramón y Cajal sn, 03550 Alicante, Spain; (M.M.-M.); (D.G.); (E.R.-M.); (A.P.); (R.G.-L.)
| | - Ana Pombero
- Instituto de Neurociencias, Universidad Miguel Hernández–CSIC, Excellence Center Severo Ochoa, Campus de San Juan, Avda. Ramón y Cajal sn, 03550 Alicante, Spain; (M.M.-M.); (D.G.); (E.R.-M.); (A.P.); (R.G.-L.)
| | - Raquel Garcia-Lopez
- Instituto de Neurociencias, Universidad Miguel Hernández–CSIC, Excellence Center Severo Ochoa, Campus de San Juan, Avda. Ramón y Cajal sn, 03550 Alicante, Spain; (M.M.-M.); (D.G.); (E.R.-M.); (A.P.); (R.G.-L.)
| | - Salvador Martinez
- Instituto de Neurociencias, Universidad Miguel Hernández–CSIC, Excellence Center Severo Ochoa, Campus de San Juan, Avda. Ramón y Cajal sn, 03550 Alicante, Spain; (M.M.-M.); (D.G.); (E.R.-M.); (A.P.); (R.G.-L.)
- Centro de Investigación Biomédica en Red en Salud Mental, CIBERSAM-ISCIII, 46010 Valencia, Spain
| |
Collapse
|
8
|
Biolato AM, Filali L, Krecke M, Thomas C, Hoffmann C. A comprehensive guide to study the immunological synapse using imaging flow cytometry. Methods Cell Biol 2024; 193:69-97. [PMID: 39919848 DOI: 10.1016/bs.mcb.2024.03.001] [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] [Indexed: 02/09/2025]
Abstract
Cytotoxic lymphocytes, such as cytotoxic T cells and natural killer (NK) cells, are instrumental in the recognition and eradication of pathogenic cells, notably those undergoing malignant transformation. Cytotoxic lymphocytes establish direct contact with cancer cells via the formation of a specialized cell-cell junction known as the lytic immunological synapse. This structure serves as a critical platform for lymphocytes to integrate surface signals from potential cancer cells and to direct their cytolytic apparatus toward the confirmed targets. Conversely, cancer cells evolve synaptic defense strategies to evade lymphocyte cytotoxicity. This chapter delineates protocols using imaging flow cytometry to examine and quantify important subcellular processes occurring within cytotoxic lymphocytes and cancer cells engaged into an immunological synapse. These processes encompass the spatial redistribution of cytoskeletal components, vesicles, organelles and cell surface molecules. We specifically describe methods to generate and select conjugates between MDA-MB-231 breast cancer cells or K-562 leukemic cells and either the NK-92MI cell line or primary human NK cells. In addition, we detail procedures to evaluate the synaptic polarization of the actin cytoskeleton, CD63-positive vesicular compartments, MHC class I molecules, as well as the microtubule-organizing center in effector cells.
Collapse
Affiliation(s)
- Andrea Michela Biolato
- Cytoskeleton and Cancer Progression, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Liza Filali
- Cytoskeleton and Cancer Progression, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Max Krecke
- Cytoskeleton and Cancer Progression, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Clément Thomas
- Cytoskeleton and Cancer Progression, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Céline Hoffmann
- Cytoskeleton and Cancer Progression, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg; National Cytometry Platform, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
9
|
Ruiz-Navarro J, Calvo V, Izquierdo M. Extracellular vesicles and microvilli in the immune synapse. Front Immunol 2024; 14:1324557. [PMID: 38268920 PMCID: PMC10806406 DOI: 10.3389/fimmu.2023.1324557] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/05/2023] [Indexed: 01/26/2024] Open
Abstract
T cell receptor (TCR) binding to cognate antigen on the plasma membrane of an antigen-presenting cell (APC) triggers the immune synapse (IS) formation. The IS constitutes a dedicated contact region between different cells that comprises a signaling platform where several cues evoked by TCR and accessory molecules are integrated, ultimately leading to an effective TCR signal transmission that guarantees intercellular message communication. This eventually leads to T lymphocyte activation and the efficient execution of different T lymphocyte effector tasks, including cytotoxicity and subsequent target cell death. Recent evidence demonstrates that the transmission of information between immune cells forming synapses is produced, to a significant extent, by the generation and secretion of distinct extracellular vesicles (EV) from both the effector T lymphocyte and the APC. These EV carry biologically active molecules that transfer cues among immune cells leading to a broad range of biological responses in the recipient cells. Included among these bioactive molecules are regulatory miRNAs, pro-apoptotic molecules implicated in target cell apoptosis, or molecules triggering cell activation. In this study we deal with the different EV classes detected at the IS, placing emphasis on the most recent findings on microvilli/lamellipodium-produced EV. The signals leading to polarized secretion of EV at the synaptic cleft will be discussed, showing that the IS architecture fulfills a fundamental task during this route.
Collapse
Affiliation(s)
- Javier Ruiz-Navarro
- Department of Metabolism and Cell Signaling, Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Víctor Calvo
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Manuel Izquierdo
- Department of Metabolism and Cell Signaling, Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
| |
Collapse
|