Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 284
Filter
1.
Front Immunol ; 15: 1464480, 2024.
Article in English | MEDLINE | ID: mdl-39376569

ABSTRACT

Introduction: Over the past four years, the COVID-19 pandemic has posed serious global health challenges. The severe form of disease and death resulted from the failure of immune regulatory mechanisms, closely highlighted by the dual proinflammatory cytokine and soluble immune checkpoint (sICP) storm. Identifying the individual factors impacting on disease severity, evolution and outcome, as well as any additional interconnections, have become of high scientific interest. Methods: In this study, we evaluated a novel panel composed of ten sICPs for the predictive values of COVID-19 disease severity, mortality and Delta vs. Omicron variant infections in relation to hyperinflammatory biomarkers. The serum levels of sICPs from confirmed SARS-CoV-2 infected patients at hospital admission were determined by Luminex, and artificial neural network analysis was applied for defining the distinct patterns of molecular associations with each form of disease: mild, moderate, and severe. Results: Notably, distinct sICP profiles characterized various stages of disease and Delta infections: while sCD40 played a central role in all defined diagrams, the differences emerged from the distribution levels of four molecules recently found and relatively less investigated (sCD30, s4-1BB, sTIM-1, sB7-H3), and their associations with various hematological and biochemical inflammatory biomarkers. The artificial neural network analysis revealed the prominent role of serum sTIM-1 and Galectin-9 levels at hospital admission in discriminating between survivors and non-survivors, as well as the role of specific anti-interleukin therapy (Tocilizumab, Anakinra) in improving survival for patients with initially high sTIM-1 levels. Furthermore, strong associations between sCD40 and Galectin-9 with suPAR defined the Omicron variant infections, while the positive match of sCD40 with sTREM-1 serum levels characterized the Delta-infected patients. Conclusions: Of importance, this study provides a comprehensive analysis of circulatory immune factors governing the COVID-19 pathology, and identifies key roles of sCD40, sTIM-1, and Galectin-9 in predicting mortality.


Subject(s)
Biomarkers , COVID-19 , SARS-CoV-2 , Severity of Illness Index , Humans , COVID-19/immunology , COVID-19/mortality , COVID-19/blood , SARS-CoV-2/immunology , Male , Middle Aged , Female , Biomarkers/blood , Aged , Immune Checkpoint Proteins/metabolism , Immune Checkpoint Proteins/blood , Adult , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/blood , Cytokine Release Syndrome/mortality
2.
Front Immunol ; 15: 1448653, 2024.
Article in English | MEDLINE | ID: mdl-39355257

ABSTRACT

Background: Broad T cell phenotypic alterations and potential dysfunctions were prominent in COVID-19. There are few and inconclusive data about the role of immune checkpoints for T cell exhaustion/activation during SARS-CoV-2 infection in multiple myeloma (MM) patients. Methods: We tested T cell subsets and immune checkpoints in 177 MM patients with COVID-19, as well as in 32 healthy infected controls and 42 uninfected MM patients. The percentage of CD4+ and CD8+ subpopulation and immune checkpoints (PD-1, TIGIT, TIM-3, LAG-3, CTLA-4, OX40, and 4-1BB) were evaluated by flow cytometry. Results: We have found that pronounced lymphopenia and inverted CD4/CD8 ratio in severe COVID-19 patients were especially developed within the first month after infection. And T cell subset dysregulation was persistent in severe patients recovering from SARS-CoV-2 infection. Immune checkpoints on CD4+ T cells were variable and uncorrelated with the level of adaptive immunity, while the proportion of CD4+ T cells was positively correlated with humoral immune response. PD-1 and TIGIT on CD8+ T cells were significantly elevated in severe patients and sustained for more than 2 months, which was associated with impaired cellular immune function. Moreover, exhausted molecules PD-1 and TIGIT on T cells were reduced in immunotherapy patients. Conclusion: The prolonged T cell dysregulation after severe SARS-CoV-2 infection highlights the close surveillance from reinfection in MM patients even during convalescence. PD-1 and TIGIT on CD8+ T cells could be important prognostic factors to stratify prognosis in MM patients with COVID-19. Moreover, immunotherapy may downregulate the expression of exhausted checkpoints PD-1 and TIGIT, leading to T cell overactivation and severe COVID-19.


Subject(s)
COVID-19 , Multiple Myeloma , SARS-CoV-2 , Humans , COVID-19/immunology , Multiple Myeloma/immunology , Male , Middle Aged , Female , Aged , SARS-CoV-2/immunology , SARS-CoV-2/physiology , Prognosis , CD8-Positive T-Lymphocytes/immunology , Immune Checkpoint Proteins/metabolism , CD4-Positive T-Lymphocytes/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Adult , Biomarkers
3.
Mol Biol Rep ; 51(1): 1036, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39361074

ABSTRACT

BACKGROUND: Lung cancer is recognized as one of the leading causes of cancer-related deaths globally, with a significant increase in incidence and intricate pathogenic mechanisms. This study examines the expression profiles of Programmed Cell Death Protein 1 (PD-1), PD-1 ligand (PDL-1), ß-catenin, CD44, interleukin 6 (IL-6), and interleukin 10 (IL-10), as well as their correlations with the clinic-pathological features and diagnostic significance in lung cancer patients. METHODS AND RESULTS: The research involved lung cancer patients exhibiting various pathological characteristics, alongside demographically matched healthy controls. The expression levels of PD-1, PDL-1, ß-catenin, and CD44 were analyzed using Real-Time PCR, while circulating levels of IL-6 and IL-10 were assessed through ELISA assays. This investigation focused on peripheral blood mononuclear cells (PBMC) to evaluate these factors non-invasively. Findings indicated that levels of PD-1, PDL-1, and CD44 were significantly elevated in patients compared to controls, which coincided with a decrease in ß-catenin levels. Additionally, a concurrent rise in IL-6 and IL-10, both pro-inflammatory cytokines, was observed in patients, suggesting a potential regulatory role for these cytokines on the PD-1/PDL-1 axis, which may help tumors evade immune system checkpoints. The predictive value of these factors concerning lung tumors and metastasis was significant (Regression analysis). Furthermore, these markers demonstrated diagnostic potential in differentiating between patients and healthy controls, as well as between individuals with metastatic and non-metastatic tumors (ROC curve analysis). CONCLUSIONS: This study provides insights into the expression profiles of PD-1/PDL-1 immune system checkpoints and their regulatory factors in lung cancer, potentially paving the way for new therapeutic and diagnostic approaches.


Subject(s)
B7-H1 Antigen , Biomarkers, Tumor , Hyaluronan Receptors , Interleukin-10 , Interleukin-6 , Lung Neoplasms , Programmed Cell Death 1 Receptor , beta Catenin , Humans , Lung Neoplasms/blood , Lung Neoplasms/immunology , Lung Neoplasms/genetics , Lung Neoplasms/diagnosis , Male , Female , Biomarkers, Tumor/blood , Middle Aged , B7-H1 Antigen/blood , B7-H1 Antigen/genetics , beta Catenin/genetics , beta Catenin/blood , Hyaluronan Receptors/blood , Hyaluronan Receptors/genetics , Aged , Interleukin-6/blood , Programmed Cell Death 1 Receptor/blood , Interleukin-10/blood , Leukocytes, Mononuclear/metabolism , Adult , Case-Control Studies , Immune Checkpoint Proteins/genetics , Immune Checkpoint Proteins/metabolism , Immune Checkpoint Proteins/blood
4.
Front Immunol ; 15: 1463847, 2024.
Article in English | MEDLINE | ID: mdl-39372415

ABSTRACT

RNA modifications are epigenetic changes that alter the structure and function of RNA molecules, playing a crucial role in the onset, progression, and treatment of cancer. Immune checkpoint inhibitor (ICI) therapies, particularly PD-1 blockade and anti-CTLA-4 treatments, have changed the treatment landscape of virous cancers, showing great potential in the treatment of different cancer patients, but sensitivity to these therapies is limited to certain individuals. This review offers a comprehensive survey of the functions and therapeutic implications of the four principal RNA modifications, particularly highlighting the significance of m6A in the realms of immune cells in tumor and immunotherapy. This review starts by providing a foundational summary of the roles RNA modifications assume within the immune cell community, focusing on T cells, NK cells, macrophages, and dendritic cells. We then discuss how RNA modifications influence the intricate regulatory mechanisms governing immune checkpoint expression, modulation of ICI efficacy, and prediction of ICI treatment outcomes, and review drug therapies targeting genes regulated by RNA modifications. Finally, we explore the role of RNA modifications in gene editing, cancer vaccines, and adoptive T cell therapies, offering valuable insights into the use of RNA modifications in cancer immunotherapy.


Subject(s)
Immune Checkpoint Inhibitors , Immunotherapy , Neoplasms , Humans , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/genetics , Immunotherapy/methods , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Animals , RNA Processing, Post-Transcriptional , Immune Checkpoint Proteins/genetics , Immune Checkpoint Proteins/metabolism
5.
Front Immunol ; 15: 1424396, 2024.
Article in English | MEDLINE | ID: mdl-39346924

ABSTRACT

Immune checkpoint (IC) inhibition in glioblastoma (GBM) has not shown promising results in the last decade compared to other solid tumors. Several factors contributing to the lack of immunotherapy response include the profound immunosuppressive nature of GBM, highly redundant signaling pathways underlying immune checkpoints, and the negative immunogenic impact of current standard of care on the tumor microenvironment. In this review, we will discuss various ICs in the context of GBM, their interplay with the tumor immune microenvironment, relevant pre-clinical and clinical studies, and the impact of current treatment modalities on GBM IC blockade therapy. Understanding the molecular mechanisms that drive ICs, and how they contribute to an immunosuppressive tumor microenvironment is critical in advancing IC inhibition therapy in GBM. Furthermore, revisiting current treatment modalities and their impact on the immune landscape is instrumental in designing future combinatorial therapies that may overcome treatment resistance.


Subject(s)
Brain Neoplasms , Glioblastoma , Immune Checkpoint Inhibitors , Tumor Microenvironment , Humans , Glioblastoma/immunology , Glioblastoma/therapy , Tumor Microenvironment/immunology , Immune Checkpoint Inhibitors/therapeutic use , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Animals , Signal Transduction , Immunotherapy/methods , Immune Checkpoint Proteins/metabolism , Immune Checkpoint Proteins/genetics
6.
Int J Mol Sci ; 25(17)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39273224

ABSTRACT

Immune checkpoints are crucial molecules for the maintenance of antitumor immune responses. The activation or inhibition of these molecules is dependent on the interactions between receptors and ligands; such interactions can provide inhibitory or stimulatory signals to the various components of the immune system. Over the last 10 years, the inhibition of immune checkpoints, such as cytotoxic T lymphocyte antigen-4, programmed cell death-1, and programmed cell death ligand-1, has taken a leading role in immune therapy. This relatively recent therapy regime is based on the use of checkpoint inhibitors, which enhance the immune response towards various forms of cancer. For a subset of patients with specific forms of cancer, these inhibitors can induce a durable response to therapy; however, the medium response rate to such therapy remains relatively poor. Recent research activities have demonstrated that the disease response to this highly promising therapy resembles the response of many forms of cancer to chemotherapy, where an encouraging initial response is followed by acquired resistance to treatment and progress of the disease. That said, these inhibitors are now used as single agents or in combination with chemotherapies as first or second lines of treatment for about 50 types of cancer. The prevailing opinion regarding immune therapy suggests that for this approach of therapy to deliver on its promise, a number of challenges have to be circumvented. These challenges include understanding the resistance mechanisms to immune checkpoint blockade, the identification of more efficient inhibitors, extending their therapeutic benefits to a wider audience of cancer patients, better management of immune-related adverse side effects, and, more urgently the identification of biomarkers, which would help treating oncologists in the identification of patients who are likely to respond positively to the immune therapies and, last but not least, the prices of therapy which can be afforded by the highest number of patients. Numerous studies have demonstrated that understanding the interaction between these checkpoints and the immune system is essential for the development of efficient checkpoint inhibitors and improved immune therapies. In the present text, we discuss some of these checkpoints, their inhibitors, and some works in which mass spectrometry-based proteomic analyses were applied.


Subject(s)
Immune Checkpoint Inhibitors , Neoplasms , Proteomics , Humans , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/immunology , Proteomics/methods , Animals , Immunotherapy/methods , Immune Checkpoint Proteins/metabolism
7.
Front Immunol ; 15: 1434171, 2024.
Article in English | MEDLINE | ID: mdl-39238640

ABSTRACT

In recent years, there has been significant research interest in the field of immunotherapy for non-small cell lung cancer (NSCLC) within the academic community. Given the observed variations in individual responses, despite similarities in histopathologic type, immunohistochemical index, TNM stage, or mutation status, the identification of a reliable biomarker for early prediction of therapeutic responses is of utmost importance. Conventional medical imaging techniques primarily focus on macroscopic tumor monitoring, which may no longer adequately fulfill the requirements of clinical diagnosis and treatment. CT (computerized tomography) or PEF/CT-based radiomics has the potential to investigate the molecular-level biological attributes of tumors, such as PD-1/PD-L1 expression and tumor mutation burden, which offers a novel approach to assess the effectiveness of immunotherapy and forecast patient prognosis. The utilization of cutting-edge radiological imaging techniques, including radiomics, PET/CT, machine learning, and artificial intelligence, demonstrates significant potential in predicting diagnosis, treatment response, immunosuppressive characteristics, and immune-related adverse events. The current review highlights that CT scan-based radiomics is a reliable and feasible way to predict the benefits of immunotherapy in patients with advanced NSCLC.


Subject(s)
Biomarkers, Tumor , Carcinoma, Non-Small-Cell Lung , Immunotherapy , Lung Neoplasms , Tomography, X-Ray Computed , Humans , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/diagnosis , Lung Neoplasms/therapy , Lung Neoplasms/immunology , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/diagnosis , Tomography, X-Ray Computed/methods , Immunotherapy/methods , Treatment Outcome , Prognosis , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Proteins/metabolism , Immune Checkpoint Proteins/genetics , Radiomics
8.
Technol Cancer Res Treat ; 23: 15330338241281285, 2024.
Article in English | MEDLINE | ID: mdl-39248214

ABSTRACT

Objectives: Heparanase (HPSE), an endoglycosidase that cleaves heparan sulfate, regulates various biological processes related to tumor progression. We explore the prognostic value of HPSE and its relationship with immunotherapy response in patients with breast cancer, to improve the effectiveness of immunotherapy and increase the survival outcomes. Methods: In the study, we explored the prognostic value of HPSE through the The Cancer Genome Atlas (TCGA) database. By using the single-sample gene set enrichment analysis (ssGSEA) method, we measured the infiltration levels of 24 immune cell types in the tumor microenvironment. Cancer Therapeutics Response Portal (CTRP) and PRISM datasets provide the area under the dose-response curve (AUC) to measure drug sensitivity. Using nomograms, we predicted overall survival ability. In vivo studies, we investigated the relationship between HPSE and immune checkpoint proteins and pro-inflammatory cytokines by immunohistochemistry of Triple-Negative Breast Cancer tumors in mice. Results: Our model demonstrated that the integrating of HPSE with the clinical stage effectively predicts patients' survival time, highlighting high HPSE expression as a prognostic risk factor for breast cancer. Then the Receiver Operating Characteristic (ROC) curve [AUC of 1 year = 0.747, AUC of 3 years = 0.731] and Decision Curve Analysis (DCA) curve illustrated the satisfactory discriminative capacity of our model, emphasizing its valuable clinical applicability. Immune-related results showed that HPSE correlates strongly with immune infiltrating cells, immune-related genes, and the anti-cancer immunity cycle. In vivo studies have demonstrated that HPSE in breast cancer is associated with increased expression of immune checkpoint proteins CD274 and cytotoxic T lymphocyte-associated protein 4 (CTLA-4) and is positively correlated with the pro-inflammatory cytokine TNF-α. Meanwhile, we analyzed the 11 types of drugs that are sensitive to the HPSE gene. Conclusion: Our results show that HPSE can serve as an effective biomarker to predict the prognosis of breast cancer patients and reflect the impact of immunotherapy.


Subject(s)
B7-H1 Antigen , Biomarkers, Tumor , Breast Neoplasms , CTLA-4 Antigen , Gene Expression Regulation, Neoplastic , Glucuronidase , Tumor Microenvironment , Humans , Female , Prognosis , Glucuronidase/metabolism , Glucuronidase/genetics , Animals , Mice , Biomarkers, Tumor/metabolism , CTLA-4 Antigen/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Tumor Microenvironment/immunology , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Immune Checkpoint Proteins/metabolism , Immune Checkpoint Proteins/genetics , ROC Curve , Nomograms , Computational Biology/methods , Gene Expression Profiling , Databases, Genetic
9.
Int J Mol Sci ; 25(17)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39273326

ABSTRACT

Due to the genetic diversity between the mother and the fetus, heightened control over the immune system during pregnancy is crucial. Immunological parameters determined by clinicians in women with idiopathic recurrent spontaneous abortion (RSA) include the quantity and activity of Natural Killer (NK) and Natural Killer T (NKT) cells, the quantity of regulatory T lymphocytes, and the ratio of pro-inflammatory cytokines, which indicate imbalances in Th1 and Th2 cell response. The processes are controlled by immune checkpoint proteins (ICPs) expressed on the surface of immune cells. We aim to investigate differences in the expression of ICPs on T cells, T regulatory lymphocytes, NK cells, and NKT cells in peripheral blood samples collected from RSA women, pregnant women, and healthy multiparous women. We aim to discover new insights into the role of ICPs involved in recurrent pregnancy loss. Peripheral blood mononuclear cells (PBMCs) were isolated by gradient centrifugation from blood samples obtained from 10 multiparous women, 20 pregnant women (11-14th week of pregnancy), and 20 RSA women, at maximum of 72 h after miscarriage. The PBMCs were stained for flow cytometry analysis. Standard flow cytometry immunophenotyping of PBMCs was performed using antibodies against classical lymphocyte markers, including CD3, CD4, CD8, CD56, CD25, and CD127. Additionally, ICPs were investigated using antibodies against Programmed Death Protein-1 (PD-1, CD279), T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3, CD366), V-domain Ig suppressor of T cell activation (VISTA), T cell immunoglobulin and ITIM domain (TIGIT), and Lymphocyte activation gene 3 (LAG-3). We observed differences in the surface expression of ICPs in the analyzed subpopulations of lymphocytes between early pregnancy and RSA, after miscarriage, and in women. We noted diminished expression of PD-1 on T lymphocytes (p = 0.0046), T helper cells (CD3CD4 positive cells, p = 0.0165), T cytotoxic cells (CD3CD8 positive cells, p = 0.0046), T regulatory lymphocytes (CD3CD4CD25CD127 low positive cells, p = 0.0106), and NKT cells (CD3CD56/CD16 positive cells, p = 0.0438), as well as LAG-3 on lymphocytes T (p = 0.0225) T helper, p = 0.0426), T cytotoxic cells (p = 0.0458) and Treg (p = 0.0293), and cells from RSA women. Impaired expression of TIM-3 (p = 0.0226) and VISTA (p = 0.0039) on CD8 cytotoxic T and NK (TIM3 p = 0.0482; VISTA p = 0.0118) cells was shown, with an accompanying increased expression of TIGIT (p = 0.0211) on NKT cells. The changes in the expression of surface immune checkpoints indicate their involvement in the regulation of pregnancy. The data might be utilized to develop specific therapies for RSA women based on the modulation of ICP expression.


Subject(s)
Abortion, Habitual , Biomarkers , Immune Checkpoint Proteins , Killer Cells, Natural , Humans , Female , Pregnancy , Abortion, Habitual/immunology , Abortion, Habitual/metabolism , Abortion, Habitual/blood , Adult , Biomarkers/blood , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Immune Checkpoint Proteins/metabolism , Immune Checkpoint Proteins/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Immunophenotyping , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/immunology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Antigens, CD/metabolism , Hepatitis A Virus Cellular Receptor 2/metabolism , Programmed Cell Death 1 Receptor/metabolism
11.
BMC Cancer ; 24(1): 1024, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160525

ABSTRACT

BACKGROUND: In the past few decades, researchers have made promising progress, including the development of immune checkpoint inhibitors (ICIs) in the therapy of bladder cancer (BLCA). Existing studies mainly focus on single immune checkpoint inhibitors but lack relevant studies on the gene expression profiles of multiple immune checkpoints. METHODS: RNA-sequencing profiling data and clinical information of BLCA patients and normal human bladder samples were acquired from the Cancer Genome Atlas and Gene Expression Omnibus databases and analyzed to identify different expression profiles of immune checkpoint genes (ICGs) after consensus clustering analysis. Based on the 526 intersecting differentially expressed genes, the LASSO Cox regression analysis was utilized to construct the ICG signature. RESULTS: According to the expression of ICGs, BLCA patients were divided into three subtypes with different phenotypic and mechanistic characteristics. Furthermore, the developed ICG signature were independent predictors of outcome in BLCA patients, and was correlated with the immune infiltration, the expression of ICGs and chemotherapeutic effect. CONCLUSIONS: This study systematically and comprehensively analyzed the expression profile of immune checkpoint genes, and established the ICG signature to investigate the differences in ICGs expression and tumor immune microenvironment, which will help risk stratification and accelerate precision medicine. Finally, we identified KRT23 as the most critical model gene, and highlighted KRT23 as a potential target to enhance immunotherapy against BLCA.


Subject(s)
Tumor Microenvironment , Urinary Bladder Neoplasms , Aged , Female , Humans , Male , Middle Aged , Biomarkers, Tumor/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Proteins/genetics , Immune Checkpoint Proteins/metabolism , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Prognosis , Transcriptome , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/therapy , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Keratins, Type I/genetics , Keratins, Type I/metabolism
12.
J Transl Med ; 22(1): 718, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39097734

ABSTRACT

BACKGROUND: An increasing number of clinical studies have begun to explore combination strategies with immune checkpoint inhibitors, aiming to present new opportunities for overcoming anti-PD-1 treatment resistance in gastric cancer. Unfortunately, the exploration of certain immune checkpoint inhibitor combination strategies has yielded suboptimal results. Therefore, it is necessary to comprehensively analyze the expression patterns of immune checkpoints and identify optimal combination regimens of anti-PD-1 inhibitors with other immune checkpoint inhibitors. METHODS: Leveraging single-cell RNA sequencing (scRNA-seq) and multivariate linear regression interaction models, we dissected the immune checkpoint expression characteristics of CD8+ T cells in gastric cancer and the immune checkpoint expression pattern (ICEP) mediating anti-PD-1 treatment resistance. Furthermore, we employed transcription factor analysis and CellOracle to explore the transcriptional regulatory mechanisms governing CD8+ T cell differentiation fates. Finally, we utilized Nichenet and spatial transcriptomic analysis to investigate the spatial expression patterns of immune checkpoints. RESULTS: Interaction analysis indicated that, among the known immune checkpoints, co-expression of NKG2A and PD-1 might exert a more profound inhibitory effect on the proliferative capacity of CD8+ T cells. The co-expression analysis revealed differential co-expression pattern of PD-1 and NKG2A, defined as ICEP1 (CD8+ T cells co-expressing PD-1, CTLA-4, TIGIT, LAG-3 or CD38) and ICEP2 (CD8+ T cells solely expressing NKG2A or co-expressing with other immune checkpoints), reflecting the co-occurrence pattern of PD-1 and the mutual exclusivity of NKG2A. Further, these two ICEP CD8+ T cell subsets represented distinct CD8+ T cell differentiation fates governed by MSC and RUNX3. Notably, ICEP2 CD8+ T cells were associated with anti-PD-1 therapy resistance in gastric cancer. This phenomenon may be attributed to the recruitment of LGMN+ macrophages mediated by the CXCL16-CXCR6 signaling pathway. CONCLUSION: This study unveiled two distinct ICEPs and the mutually exclusivity and co-occurrence characteristics of CD8+ T cells in gastric cancer. The ICEP2 CD8+ T cell subset, highly expressed in gastric cancer patients resistant to anti-PD-1 therapy, may be recruited by LGMN+ macrophages through CXCL16-CXCR6 axis. These findings provide evidence for NKG2A as a novel immunotherapeutic target in gastric cancer and offer new insights into combination strategies for immune checkpoint inhibitors in gastric cancer.


Subject(s)
CD8-Positive T-Lymphocytes , Drug Resistance, Neoplasm , Immune Checkpoint Inhibitors , NK Cell Lectin-Like Receptor Subfamily C , Stomach Neoplasms , Humans , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Gene Expression Regulation, Neoplastic , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Proteins/metabolism , Immune Checkpoint Proteins/genetics , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Programmed Cell Death 1 Receptor/metabolism , Stomach Neoplasms/immunology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
13.
Aging (Albany NY) ; 16(15): 11683-11728, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39120585

ABSTRACT

Drugs that target immune checkpoint have become the most popular weapon in cancer immunotherapy, yet only have practical benefits for a small percentage of patients. Tumor cells constantly interact with their microenvironment, which is made up of a variety of immune cells as well as endothelial cells and fibroblasts. Immune checkpoint expression and blocked signaling of immune cells in the tumor microenvironment (TME) are key to tumor progression. In this study, we perform deliberation convolution on the TCGA database for human lung, breast, and colorectal cancer to infer crosstalk between immune checkpoint receptors (ICRs) and ligands (ICLs) in TME of pan-carcinogenic solid tumor types, validated by flow cytometry. Analysis of immune checkpoints showed that there was little variation between different tumor types. It showed that CD160, LAG3, TIGIT were found to be highly expressed in CD8+ T cells instead of CD4+ T cells, PD-L1, PD-L2, CD86, LGALS9, TNFRSF14, LILRB4 and other ligands were highly expressed on macrophages, FVR, NECTIN2, FGL1 were highly expressed on Epithelial cells, CD200 was highly expressed in Endothelial cells, and CD80 was highly expressed in CD8 High expression on T cells. Overall, our study provides a new resource for the expression of immune checkpoints in TME on various types of cells. Significance: This study provides immune checkpoint expression of immune cells of multiple cancer types to infer immune mechanisms in the tumor microenvironment and provide ideas for the development of new immune checkpoint-blocking drugs.


Subject(s)
Immune Checkpoint Proteins , Tumor Microenvironment , Humans , Tumor Microenvironment/immunology , Immune Checkpoint Proteins/metabolism , Immune Checkpoint Proteins/genetics , Ligands , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Antigens, CD/metabolism , Antigens, CD/genetics , Nectins/metabolism , Nectins/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Lymphocyte Activation Gene 3 Protein , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Programmed Cell Death 1 Ligand 2 Protein/genetics , Macrophages/immunology , Macrophages/metabolism , Endothelial Cells/immunology , Endothelial Cells/metabolism , Female
14.
Sci Adv ; 10(35): eadn0164, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39196934

ABSTRACT

Natural killer (NK) cells often become dysfunctional during tumor progression, but the molecular mechanisms underlying this phenotype remain unclear. To explore this phenomenon, we set up mouse lymphoma models activating or not activating NK cells. Both tumor types elicited type I interferon production, leading to the expression of a T cell exhaustion-like signature in NK cells, which included immune checkpoint proteins (ICPs). However, NK cell dysfunction occurred exclusively in the tumor model that triggered NK cell activation. Moreover, ICP-positive NK cells demonstrated heightened reactivity compared to negative ones. Furthermore, the onset of NK cell dysfunction was swift and temporally dissociated from ICPs induction, which occurred as a later event during tumor growth. Last, NK cell responsiveness was restored when stimulation was discontinued, and interleukin-15 had a positive impact on this reversion. Therefore, our data demonstrate that the reactivity of NK cells is dynamically controlled and that NK cell dysfunction is a reversible process uncoupled from the expression of ICPs.


Subject(s)
Killer Cells, Natural , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Animals , Mice , Immune Checkpoint Proteins/metabolism , Immune Checkpoint Proteins/genetics , Lymphocyte Activation/immunology , Cell Line, Tumor , Interleukin-15/metabolism , Lymphoma/immunology , Lymphoma/pathology , Mice, Inbred C57BL , Disease Models, Animal , Humans
15.
AIDS ; 38(11): 1725-1727, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39088830

ABSTRACT

We explored the impact of immune dysregulation on pancreatic beta cell injury in HIV patients. Analyzing 105 participant samples, we observed lower IL-21 levels and elevated immune checkpoint levels (e.g. PD-1, CD27+, CD40+) in untreated HIV patients. Notably, soluble TIM-3 correlated positively with improved beta cell function and inversely with beta cell stress, suggesting its potential role in beta cell protection in untreated HIV.


Subject(s)
HIV Infections , Insulin-Secreting Cells , Humans , HIV Infections/complications , HIV Infections/immunology , Insulin-Secreting Cells/immunology , Male , Female , Middle Aged , Adult , Hepatitis A Virus Cellular Receptor 2 , Interleukins/blood , Immune Checkpoint Proteins/metabolism
16.
Gene ; 930: 148835, 2024 Dec 20.
Article in English | MEDLINE | ID: mdl-39127414

ABSTRACT

BACKGROUND: Gliomas are the most common primary malignant tumours of the central nervous system, and neddylation may be a potential target for the treatment of gliomas. Our study analysed neddylation's potential role in gliomas of different pathological types and its correlation with immunotherapy. METHODS: Genes required for model construction were sourced from existing literature, and their expression data were extracted from the TCGA and CGGA databases. LASSO regression was employed to identify genes associated with the prognosis of glioma patients in TCGA and to establish a clinical prognostic model. Biological changes in glioma cell lines following intervention with hub genes were evaluated using the CCK-8 assay and transwell assay. The genes implicated in the model construction were validated across various cell lines using Western blot. We conducted analyses to examine correlations between model scores and clinical data, tumor microenvironments, and immune checkpoints. Furthermore, we investigated potential differences in molecular functions and mechanisms among different groups. RESULTS: We identified 249 genes from the Reactome database and analysed their expression profiles in the TCGA and CGGA databases. After using LASSO-Cox, four genes (BRCA1, BIRC5, FBXL16 and KLHL25, p < 0.05) with significant correlations were identified. We selected FBXL16 for validation in in vitro experiments. Following FBXL16 overexpression, the proliferation, migration, and invasion abilities of glioma cell lines all showed a decrease. Then, we constructed the NEDD Index for gliomas. The nomogram indicated that this model could serve as an independent prognostic marker. Analysis of the tumour microenvironment and immune checkpoints revealed that the NEDD index was also correlated with immune cell infiltration and the expression levels of various immune checkpoints. CONCLUSION: The NEDD index can serve as a practical tool for predicting the prognosis of glioma patients, and it is correlated with immune cell infiltration and the expression levels of immune checkpoints.


Subject(s)
Biomarkers, Tumor , Brain Neoplasms , Gene Expression Regulation, Neoplastic , Glioma , Humans , Glioma/genetics , Glioma/immunology , Glioma/pathology , Prognosis , Cell Line, Tumor , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , NEDD8 Protein/genetics , NEDD8 Protein/metabolism , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Immune Checkpoint Proteins/genetics , Immune Checkpoint Proteins/metabolism , Cell Proliferation/genetics , F-Box Proteins/genetics , F-Box Proteins/metabolism , Female , Gene Expression Profiling/methods , Databases, Genetic , Cell Movement/genetics , Male
17.
Front Immunol ; 15: 1442749, 2024.
Article in English | MEDLINE | ID: mdl-39206195

ABSTRACT

Background: Immunoglobulin G subclass deficiencies (IgGsd) comprise a wide clinical spectrum from no symptoms to repeated respiratory infections and risk for the development of lung damage. Our aims were to investigate whether the immunological phenotype of IgGsd patients on and off immunoglobulin replacement therapy (IgRT) was reflected in the clinical features of IgGsd. Method: Thirty patients with IgGsd were included in this prospective study of 18 months of IgRT, followed by 7-18 months of IgRT discontinuation. Blood samples were collected when patients were on and off IgRT and compared with samples from 34 cross-sectional healthy controls. An in-depth lymphocyte phenotyping was performed by flow cytometry and plasma levels of immune checkpoints were assessed. Results: IgG3 subclass deficiency was most common. Patients with IgGsd had decreased levels of activated T cells and B cells and plasma levels of negative immune checkpoint molecules correlated negatively with T cell and B cell activation. The decreased T cell activation level was unaffected by IgRT, while the B cell activation was partly restored. Of note, decreased levels of activated regulatory T cells (Tregs) were found in IgGsd patients and was partly restored during IgRT. The profile of comorbidities did not associate with Treg levels. Discussion: IgGsd is associated with decreased B cell and T cell activation including Tregs, and increased plasma levels of negative immune checkpoint molecules. The consequence of reduced activated Tregs in IgGsd remains unclear. Decreased immune cell activation was partly restored during IgRT, demonstrating that IgRT may contribute to improved immune function in patients with IgGsd.


Subject(s)
IgG Deficiency , Immunoglobulin G , Lymphocyte Activation , T-Lymphocytes, Regulatory , Humans , Male , Female , T-Lymphocytes, Regulatory/immunology , Middle Aged , Adult , IgG Deficiency/immunology , Lymphocyte Activation/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Cross-Sectional Studies , Prospective Studies , Aged , B-Lymphocytes/immunology , Immune Checkpoint Proteins/genetics , Immune Checkpoint Proteins/metabolism , Immunophenotyping , Immunoglobulins, Intravenous/therapeutic use
18.
Int J Mol Sci ; 25(15)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39125946

ABSTRACT

Despite the numerous studies on the clinical aspects of early-onset preeclampsia, our understanding of the immunological consequences of inadequate placenta development remains incomplete. The Th1-predominance characteristic of early-onset preeclampsia significantly impacts maternal immunotolerance, and the role of immune checkpoint molecules in these mechanisms is yet to be fully elucidated. Our study aims to fill these crucial knowledge gaps. A total of 34 pregnant women diagnosed with early-onset preeclampsia and 34 healthy pregnant women were enrolled in this study. A mononuclear cell fragment from the venous blood was separated and frozen. The CD8+ and CD8- NK cell subpopulations were identified and compared to their immune checkpoint molecule expressions using multicolor flow cytometry. The serum CD226 levels were measured by ELISA. Based on our measures, the frequency of the CD8- subpopulation was significantly higher than that of the CD8+ counterpart in both the NKdim and NKbright subsets. Significantly lower CD226 surface expressions were detected in the preeclamptic group compared to healthy women in all the investigated subpopulations. However, while no difference was observed in the level of the soluble CD226 molecule between the two groups, the CD112 and CD155 surface expressions were significantly different. Our study's findings underscore the significant role of the CD8+ and CD8- NK subpopulations in the Th1-dominated immune environment. This deepens our understanding of early-onset preeclampsia and suggests that each subpopulation could contribute to the compensation mechanisms and the restoration of the immunological balance in this condition, a crucial step toward developing effective interventions.


Subject(s)
CD8-Positive T-Lymphocytes , Killer Cells, Natural , Pre-Eclampsia , Humans , Female , Pregnancy , Pre-Eclampsia/immunology , Pre-Eclampsia/blood , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Adult , Antigens, Differentiation, T-Lymphocyte/metabolism , Immune Checkpoint Proteins/metabolism , Case-Control Studies
19.
Int J Mol Sci ; 25(13)2024 Jul 04.
Article in English | MEDLINE | ID: mdl-39000441

ABSTRACT

Although inhibitors targeting the PD1/PD-L1 immune checkpoint are showing comparably good outcomes, a significant percentage of head and neck squamous cell carcinoma (HNSCC) patients do not respond to treatment. Apart from using different treatment strategies, another possibility would be to target other immune checkpoints operating in these non-responding tumors. To obtain an overview of which checkpoint ligands are expressed on HNSCC tumor cells and if these ligands are affected by HGF/MET signaling, we used mRNA sequencing and antibody-based techniques for identifying checkpoint ligands in six HNSCC tumor cell lines. Furthermore, we compared our results to mRNA sequencing data. From the checkpoint ligands we investigated, VISTA was expressed the highest at the RNA level and was also the most ubiquitously expressed. PD-L2 and B7-H3 were expressed comparably lower and were not present in all cell lines to the same extent. B7-H4, however, was only detectable in the Detroit 562 cell line. Concerning the effect of HGF on the ligand levels, PD-L2 expression was enhanced with HGF stimulation, whereas other checkpoint ligand levels decreased with stimulation. B7-H4 levels in the Detroit 562 cell line drastically decreased with HGF stimulation. This is of interest because both the checkpoint ligand and the growth factor are reported to be connected to epithelial-mesenchymal transition in the literature.


Subject(s)
Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms , Hepatocyte Growth Factor , Immune Checkpoint Proteins , Proto-Oncogene Proteins c-met , Signal Transduction , Squamous Cell Carcinoma of Head and Neck , Humans , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins c-met/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/immunology , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/genetics , Cell Line, Tumor , Immune Checkpoint Proteins/metabolism , Immune Checkpoint Proteins/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Programmed Cell Death 1 Ligand 2 Protein/genetics , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , B7 Antigens/metabolism , B7 Antigens/genetics
20.
Int Immunopharmacol ; 139: 112663, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39079196

ABSTRACT

Psoriasis and atopic dermatitis (AD) are both chronic inflammatory skin diseases, which are common and difficult to cure. Currently, the emerging biologics have demonstrated outstanding efficacy, but not all patients are able to benefit from them, and traditional systemic treatments come with many severe side effects. The emergence of immune checkpoints brings new hope to solve this problem. Immune checkpoints regulate T cell activation. Upon damage to the co-inhibitory molecules, the inhibition on T cells is removed, leading to the excessive activation of T cells. In this review, we delineate and highlight the expression and function of immune checkpoint molecules (CTLA-4, PD-1, TIM-3, TIGIT, VISTA, LAG-3, OX40, GITR) in psoriasis and AD. We provide preclinical and clinical studies supporting a potential therapeutic approach of targeting these checkpoints for inflammatory skin diseases. Moreover, the complexity of immune checkpoints and safety of clinical application are discussed.


Subject(s)
Dermatitis, Atopic , Psoriasis , Humans , Dermatitis, Atopic/immunology , Dermatitis, Atopic/drug therapy , Psoriasis/immunology , Psoriasis/drug therapy , Animals , Immune Checkpoint Proteins/metabolism , Immune Checkpoint Proteins/genetics , Immune Checkpoint Inhibitors/therapeutic use , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL