Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68.832
Filter
1.
Cancer Immunol Immunother ; 73(10): 192, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39105820

ABSTRACT

PURPOSE: Immune checkpoint inhibitors have revolutionized the treatment of renal cell carcinoma (RCC), but many patients do not respond to therapy and the majority develop resistant disease over time. Thus, there is increasing need for alternative immunomodulating agents. The co-inhibitory molecule T-cell immunoglobulin and ITIM domain (TIGIT) may play a role in resistance to approved immune checkpoint inhibitors and is being investigated as a potential therapeutic target. The purpose of this study was to quantify TIGIT positivity in tumor-infiltrating T cells in RCC. METHODS: We employed tissue microarrays containing specimens from primary RCC tumors, adjacent normal renal tissue, and RCC metastases to quantify TIGIT within tumor-infiltrating CD3+ T cells using quantitative immunofluorescent analysis. We also compared these results to TIGIT+ CD3+ levels in four other tumor types (melanoma, non-small cell lung, cervical, and head and neck cancers). RESULTS: We did not observe significant differences in TIGIT positivity between primary RCC tumors and patient-matched metastatic samples. We found that the degree of TIGIT positivity in RCC is comparable to that in lung cancer but lower than that in melanoma, cervical, and head and neck cancers. Correlation analysis comparing TIGIT positivity to previously published, patient-matched spatial proteomic data by our group revealed a negative association between TIGIT and the checkpoint proteins PD-1 and LAG3. CONCLUSION: Our findings support careful evaluation of TIGIT expression on T cells in primary or metastatic RCC specimens for patients who may be treated with TIGIT-targeting antibodies, as increased TIGIT positivity might be associated with a greater likelihood of response to therapy.


Subject(s)
Antigens, CD , Carcinoma, Renal Cell , Kidney Neoplasms , Lymphocyte Activation Gene 3 Protein , Lymphocytes, Tumor-Infiltrating , Programmed Cell Death 1 Receptor , Receptors, Immunologic , Humans , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/metabolism , Receptors, Immunologic/metabolism , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Kidney Neoplasms/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Programmed Cell Death 1 Receptor/metabolism , Antigens, CD/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Female , Male , Biomarkers, Tumor/metabolism
2.
Cell ; 187(16): 4336-4354.e19, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39121847

ABSTRACT

Exhausted CD8 T (Tex) cells in chronic viral infection and cancer have sustained co-expression of inhibitory receptors (IRs). Tex cells can be reinvigorated by blocking IRs, such as PD-1, but synergistic reinvigoration and enhanced disease control can be achieved by co-targeting multiple IRs including PD-1 and LAG-3. To dissect the molecular changes intrinsic when these IR pathways are disrupted, we investigated the impact of loss of PD-1 and/or LAG-3 on Tex cells during chronic infection. These analyses revealed distinct roles of PD-1 and LAG-3 in regulating Tex cell proliferation and effector functions, respectively. Moreover, these studies identified an essential role for LAG-3 in sustaining TOX and Tex cell durability as well as a LAG-3-dependent circuit that generated a CD94/NKG2+ subset of Tex cells with enhanced cytotoxicity mediated by recognition of the stress ligand Qa-1b, with similar observations in humans. These analyses disentangle the non-redundant mechanisms of PD-1 and LAG-3 and their synergy in regulating Tex cells.


Subject(s)
Antigens, CD , CD8-Positive T-Lymphocytes , Histocompatibility Antigens Class I , Lymphocyte Activation Gene 3 Protein , NK Cell Lectin-Like Receptor Subfamily D , Programmed Cell Death 1 Receptor , Animals , Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Mice , Programmed Cell Death 1 Receptor/metabolism , NK Cell Lectin-Like Receptor Subfamily D/metabolism , Histocompatibility Antigens Class I/metabolism , Humans , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Mice, Inbred C57BL , High Mobility Group Proteins/metabolism , High Mobility Group Proteins/genetics , Cytotoxicity, Immunologic , Cell Proliferation , Killer Cells, Natural/metabolism , Killer Cells, Natural/immunology
3.
Cell ; 187(16): 4373-4388.e15, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39121849

ABSTRACT

Relatlimab (rela; anti-LAG-3) plus nivolumab (nivo; anti-PD-1) is safe and effective for treatment of advanced melanoma. We designed a trial (NCT03743766) where advanced melanoma patients received rela, nivo, or rela+nivo to interrogate the immunologic mechanisms of rela+nivo. Analysis of biospecimens from this ongoing trial demonstrated that rela+nivo led to enhanced capacity for CD8+ T cell receptor signaling and altered CD8+ T cell differentiation, leading to heightened cytotoxicity despite the retention of an exhaustion profile. Co-expression of cytotoxic and exhaustion signatures was driven by PRDM1, BATF, ETV7, and TOX. Effector function was upregulated in clonally expanded CD8+ T cells that emerged after rela+nivo. A rela+nivo intratumoral CD8+ T cell signature was associated with a favorable prognosis. This intratumoral rela+nivo signature was validated in peripheral blood as an elevated frequency of CD38+TIM3+CD8+ T cells. Overall, we demonstrated that cytotoxicity can be enhanced despite the retention of exhaustion signatures, which will inform future therapeutic strategies.


Subject(s)
CD8-Positive T-Lymphocytes , Lymphocyte Activation Gene 3 Protein , Melanoma , Programmed Cell Death 1 Receptor , Humans , Antigens, CD/metabolism , Antigens, CD/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cytotoxicity, Immunologic , High Mobility Group Proteins , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Lymphocyte Activation Gene 3 Protein/antagonists & inhibitors , Melanoma/immunology , Melanoma/drug therapy , Melanoma/genetics , Nivolumab/therapeutic use , Nivolumab/pharmacology , Positive Regulatory Domain I-Binding Factor 1/metabolism , Positive Regulatory Domain I-Binding Factor 1/genetics , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Signal Transduction
4.
Int J Med Sci ; 21(10): 1890-1902, 2024.
Article in English | MEDLINE | ID: mdl-39113896

ABSTRACT

Objective: The immune response initiated by SARS-CoV-2 infection in pregnancy is poorly elucidated. We aimed to access and compare the antiviral cellular responses and lymphocytes activation between healthy pregnancies and pregnant women infected with SARS-CoV-2. Methods: We detected the immunological changes of lymphocytes in peripheral blood of healthy non-pregnant women, non-pregnant women with COVID-19, healthy pregnant women, pregnant women with COVID-19 and convalescent group by flow cytometry. In vitro blockade was used to identify NKT-like cell activation through ICOS-ICOSL pathway. Results: We found that CD3+CD56+ NKT-like cells decreased significantly in COVID-19 positive pregnant women compared to healthy pregnant women. NKT-like cells of pregnant women expressed higher level of activating receptors CD69 and NKp46 after SARS-CoV-2 infection. Particularly, they also increased the expression of the co-stimulatory molecule ICOS. NKT-like cells of pregnant women with COVID-19 up-regulated the expression of IFN-γ, CD107a and Ki67. Meanwhile, we found that ICOSL expression was significantly increased on pDCs in pregnant women with COVID-19. Blocking ICOS in vitro significantly decreased the antiviral activity of NKT-like cells in COVID-19 positive pregnant women, suggesting that ICOS-ICOSL may play an important role in the virus clearance by NKT-like cells. Conclusions: During SARS-CoV-2 infection, NKT-like cells of pregnant women activated through ICOS-ICOSL pathway and played an important role in the antiviral response.


Subject(s)
COVID-19 , Inducible T-Cell Co-Stimulator Ligand , Inducible T-Cell Co-Stimulator Protein , Natural Killer T-Cells , Pregnancy Complications, Infectious , SARS-CoV-2 , Humans , Female , Pregnancy , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , COVID-19/immunology , COVID-19/virology , Inducible T-Cell Co-Stimulator Protein/metabolism , Adult , Inducible T-Cell Co-Stimulator Ligand/metabolism , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/virology , SARS-CoV-2/immunology , Lymphocyte Activation/immunology , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Signal Transduction/immunology , Interferon-gamma/metabolism , Natural Cytotoxicity Triggering Receptor 1/metabolism , Lectins, C-Type/metabolism
5.
BMC Cardiovasc Disord ; 24(1): 431, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39148029

ABSTRACT

BACKGROUND: Hyperhomocysteinemia (HHcy) is an independent risk factor for atherosclerosis (AS). Endothelial mesenchymal transition (EndMT) refers to the process in which endothelial cells lose endothelial cell morphology and characteristic gene expression, and acquire phenotypic characteristics and gene expression related to mesenchymal cells. Numerous studies have confirmed that EndMT is involved in the formation of atherosclerosis. Catalpol is one of the active components of Rehmannia, which has antioxidant, anti-inflammatory, anti-tumor, neuroprotective and other biological activities. Studies have shown that catalpol can reduce atherosclerotic plaque induced by high sugar or fat. However, the effect of catalpol on HHCY-induced EndMT is unclear. METHODS AND RESULTS: In vitro HHcy-treated primary human umbilical vein endothelial cells (HUVECs) were used to construct a cell model, and the antioxidants N-acetylcysteine (NAC) and catalase alcohol were administered. In vivo C57BL/6N mice were given a diet fed with 4.4% high methionine chow to construct a HHcy mice model and were treated with catalpol. The results showed that hhcy could induce morphological transformation of endothelial cells into mesenchymal cells, increase intracellular ROS content, up-regulate α-SMA, N-cadherin, p-p65 protein expression, down-regulate VE-cadherin, CD31 protein expression, induce pathological changes of aortic root endothelium, and increase aortic endothelial ROS content. Catalpol reversed these hhcy induced outcomes. CONCLUSIONS: Catalpol inhibits HHcy-induced EndMT, and the underlying mechanism may be related to the ROS/NF-κB signaling pathway. Catalpol may be a potential drug for the treatment of HHcy-related AS.


Subject(s)
Disease Models, Animal , Human Umbilical Vein Endothelial Cells , Hyperhomocysteinemia , Iridoid Glucosides , Mice, Inbred C57BL , NF-kappa B , Reactive Oxygen Species , Signal Transduction , Animals , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/pathology , Humans , Signal Transduction/drug effects , Iridoid Glucosides/pharmacology , Reactive Oxygen Species/metabolism , Hyperhomocysteinemia/drug therapy , Hyperhomocysteinemia/metabolism , Hyperhomocysteinemia/complications , Cells, Cultured , NF-kappa B/metabolism , Antioxidants/pharmacology , Oxidative Stress/drug effects , Male , Cadherins/metabolism , Epithelial-Mesenchymal Transition/drug effects , Atherosclerosis/pathology , Atherosclerosis/metabolism , Atherosclerosis/drug therapy , Atherosclerosis/prevention & control , Atherosclerosis/etiology , Transcription Factor RelA/metabolism , Antigens, CD/metabolism , Endothelial-Mesenchymal Transition
6.
Pathol Oncol Res ; 30: 1611813, 2024.
Article in English | MEDLINE | ID: mdl-39148954

ABSTRACT

Hereditary breast and ovarian cancer is a well-known genetic condition, inherited mainly in an autosomal dominant way, which elevates the risk of developing malignancies at a young age in heterozygous carriers. Advances in new generation sequencing have enabled medical professionals to determine whether a patient is harbouring mutations in moderate- or high penetrance susceptibility genes. We conducted a retrospective analysis among 275 patients who underwent genetic counselling and multigene panel testing for hereditary breast and ovarian cancer syndrome in our department. From these patients 74.5% (205/275) were affected by some type of malignancy, while the remaining 25.5% (70/275) had a positive family history of different cancers, suggesting a genetic predisposition. These tests confirmed a genetic variant in 29.8% and 28.6% of these patient groups respectively. The results also mirrored our general knowledge concerning the genetic background of hereditary breast and ovarian cancer, as variants in either one of the BRCA1 and BRCA2 genes proved to be the most common cause among our patients with 41.5%. Our test also detected a novel mutation in the CDH1 gene and three patients with double heterozygosity in two different susceptibility genes. This study demonstrates the relevance of genetic counselling and non-BRCA gene sequencing among cancer patients and patients who fulfil the criteria for genetic testing, while also providing important details about the genetic profile of Hungarian patients.


Subject(s)
BRCA1 Protein , BRCA2 Protein , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Mutation , Ovarian Neoplasms , Humans , Female , Retrospective Studies , Adult , Middle Aged , BRCA2 Protein/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , BRCA1 Protein/genetics , High-Throughput Nucleotide Sequencing/methods , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Aged , Genetic Testing/methods , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Genetic Counseling , Young Adult , Biomarkers, Tumor/genetics , Antigens, CD , Cadherins
7.
PeerJ ; 12: e17842, 2024.
Article in English | MEDLINE | ID: mdl-39131609

ABSTRACT

Background: Ferroptosis is a non-apoptotic iron-dependent form of cell death implicated in various cancer pathologies. However, its precise role in tumor growth and progression of cervical cancer (CC) remains unclear. Transferrin receptor protein 1 (TFRC), a key molecule associated with ferroptosis, has been identified as influencing a broad range of pathological processes in different cancers. However, the prognostic significance of TFRC in CC remains unclear. The present study utilized bioinformatics to explore the significance of the ferroptosis-related gene TFRC in the progression and prognosis of CC. Methods: We obtained RNA sequencing data and corresponding clinical information on patients with CC from The Cancer Genome Atlas (TCGA), Genotype Tissue Expression (GTEx) and Gene Expression Omnibus (GEO) databases. Using least absolute shrinkage and selection operator (LASSO) Cox regression, we then generated a multigene signature of five ferroptosis-related genes (FRGs) for the prognostic prediction of CC. We investigated the relationship between TFRC gene expression and immune cell infiltration by employing single-sample GSEA (ssGSEA) analysis. The potential functional role of the TFRC gene was evaluated through gene set enrichment analysis (GSEA). Immunohistochemistry and qPCR was employed to assess TFRC mRNA and protein expression in 33 cases of cervical cancer. Furthermore, the relationship between TFRC mRNA expression and overall survival (OS) was investigated in patients. Results: CC samples had significantly higher TFRC gene expression levels than normal tissue samples. Higher TFRC gene expression levels were strongly associated with higher cancer T stages and OS events. The findings of multivariate analyses illustrated that the OS in CC patients with high TFRC expression is shorter than in patients with low TFRC expression. Significant increases were observed in the levels of TFRC mRNA and protein expression in patients diagnosed with CC. Conclusion: Increased TFRC expression in CC was associated with disease progression, an unfavorable prognosis, and dysregulated immune cell infiltration. In addition, it highlights ferroptosis as a promising therapeutic target for CC.


Subject(s)
Ferroptosis , Receptors, Transferrin , Tumor Microenvironment , Uterine Cervical Neoplasms , Humans , Female , Ferroptosis/genetics , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/mortality , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Prognosis , Gene Expression Regulation, Neoplastic , Antigens, CD/genetics , Middle Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism
8.
Cell ; 187(16): 4144-4146, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39121844

ABSTRACT

LAG-3 is the third immune checkpoint pathway successfully targeted for cancer therapy. Although ineffective as a monotherapy, combination of LAG-3 and PD-1 blockade improves survival from advanced melanoma. In this issue of Cell, two studies in mice and a human clinical trial provide insights on LAG-3 in immune regulation.


Subject(s)
Antigens, CD , CD8-Positive T-Lymphocytes , Lymphocyte Activation Gene 3 Protein , CD8-Positive T-Lymphocytes/immunology , Humans , Animals , Mice , Antigens, CD/metabolism , Antigens, CD/immunology , Melanoma/immunology , Melanoma/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/immunology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use
9.
Sci Transl Med ; 16(759): eadg1915, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39110779

ABSTRACT

Severe alcohol-associated hepatitis (AH) is a life-threatening form of alcohol-associated liver disease. Liver neutrophil infiltration is a hallmark of AH, yet the effects of alcohol on neutrophil functions remain elusive. Identifying therapeutic targets to reduce neutrophil-mediated liver damage is essential. Bruton's tyrosine kinase (BTK) plays an important role in neutrophil development and function; however, the role of BTK in AH is unknown. Using RNA sequencing of circulating neutrophils, we found an increase in Btk expression (P = 0.05) and phosphorylated BTK (pBTK) in patients with AH compared with healthy controls. In vitro, physiologically relevant doses of alcohol resulted in a rapid, TLR4-mediated induction of pBTK in neutrophils. In a preclinical model of AH, administration of a small-molecule BTK inhibitor (evobrutinib) or myeloid-specific Btk knockout decreased proinflammatory cytokines and attenuated neutrophil-mediated liver damage. We found that pBTK was essential for alcohol-induced bone marrow granulopoiesis and liver neutrophil infiltration. In vivo, BTK inhibition or myeloid-specific Btk knockout reduced granulopoiesis, circulating neutrophils, liver neutrophil infiltration, and liver damage in a mouse model of AH. Mechanistically, using liquid chromatography-tandem mass spectrometry, we identified CD84 as a kinase target of BTK, which is involved in granulopoiesis. In vitro, CD84 promoted alcohol-induced interleukin-1ß and tumor necrosis factor-α in primary human neutrophils, which was inhibited by CD84-blocking antibody treatment. Our findings define the role of BTK and CD84 in regulating neutrophil inflammation and granulopoiesis, with potential therapeutic implications in AH.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase , Liver Diseases, Alcoholic , Neutrophils , Agammaglobulinaemia Tyrosine Kinase/metabolism , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Animals , Humans , Neutrophils/metabolism , Neutrophils/drug effects , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Protein Kinase Inhibitors/pharmacology , Mice , Male , Liver/pathology , Liver/metabolism , Liver/drug effects , Granulocytes/metabolism , Granulocytes/drug effects , Mice, Inbred C57BL , Antigens, CD/metabolism , Mice, Knockout , Toll-Like Receptor 4/metabolism , Phosphorylation/drug effects
10.
Am J Reprod Immunol ; 92(2): e13912, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39113660

ABSTRACT

PROBLEM: There is a higher incidence of irritable bowel syndrome with miscarriages, and recurrent miscarriages of otherwise normal embryos have been linked to subnormal expression of the immune checkpoint inhibitor CD200L. We sought to determine if alterations in the expression of the CD200 immune checkpoint inhibitor occur in colonic tissue in IBS-D patients. METHOD OF STUDY: Quantitative immunohistochemical staining of biopsies from proximal and distal colon or rectum for the inhibitory CD200L and CD200S molecules was done. CD56 cells were also enumerated as they play a role in recurrent miscarriages and may express CD200S. RESULTS: CD200L was decreased and CD200S was unchanged in epithelium but not stroma of 3 IBS-D cases. One case had an increase in both CD200L and CD200S. CD56 cells were also stained for CD200S. Degranulation was assessed by the percentage of extracellular CD200S that was increased as epithelial CD200L decreased. CONCLUSIONS: This pilot study was promising and warrants a larger sample to determine if a correlation between uterine implantation site CD200L and CD200S expression in normal and failing implantation sites is needed. Colonic epithelial CD200L may then provide useful information about the pathogenesis of the spontaneous miscarriage in individual cases.


Subject(s)
Abortion, Habitual , Antigens, CD , Diarrhea , Irritable Bowel Syndrome , Humans , Female , Irritable Bowel Syndrome/immunology , Irritable Bowel Syndrome/metabolism , Abortion, Habitual/immunology , Abortion, Habitual/metabolism , Antigens, CD/metabolism , Adult , Diarrhea/immunology , Pregnancy , Pilot Projects , Immune Tolerance , Signal Transduction , CD56 Antigen/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Colon/pathology , Colon/immunology , Colon/metabolism
11.
Front Immunol ; 15: 1432015, 2024.
Article in English | MEDLINE | ID: mdl-39144149

ABSTRACT

Therapeutic strategies targeting non-adaptive immune cells are currently in clinical development. γδT cells are a small subtype of T cells (1-10% of total T cells) that mediate their effector function without the necessity of the antigen presenting machinery, and also share functional properties with innate cells. Among the different γδT subtypes, antibodies against Vγ9Vδ2T have reported signs of clinical efficacy in early clinical studies. In this review we describe the biology of this subtype of non-conventional T cells and provide insights into the mechanism of action of novel antibodies that activate these cells. We will focus on antibodies targeting the BTN3A ligand and bi-specific γδT cell engagers. We will review in detail the advantages of these strategies including the potential for overcoming mechanisms of resistance to check point inhibitors, or the much more adequate safety profile compared with agents activating classical T cells. Limitations identified during the first studies in humans and strategies to overcome them will be revised and discussed. Finally, clinical options for future clinical development will be suggested.


Subject(s)
Receptors, Antigen, T-Cell, gamma-delta , Humans , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Animals , Butyrophilins/immunology , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Immunotherapy/methods , Intraepithelial Lymphocytes/immunology , Antigens, CD
12.
Cell ; 187(16): 4355-4372.e22, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39121848

ABSTRACT

Overcoming immune-mediated resistance to PD-1 blockade remains a major clinical challenge. Enhanced efficacy has been demonstrated in melanoma patients with combined nivolumab (anti-PD-1) and relatlimab (anti-LAG-3) treatment, the first in its class to be FDA approved. However, how these two inhibitory receptors synergize to hinder anti-tumor immunity remains unknown. Here, we show that CD8+ T cells deficient in both PD-1 and LAG-3, in contrast to CD8+ T cells lacking either receptor, mediate enhanced tumor clearance and long-term survival in mouse models of melanoma. PD-1- and LAG-3-deficient CD8+ T cells were transcriptionally distinct, with broad TCR clonality and enrichment of effector-like and interferon-responsive genes, resulting in enhanced IFN-γ release indicative of functionality. LAG-3 and PD-1 combined to drive T cell exhaustion, playing a dominant role in modulating TOX expression. Mechanistically, autocrine, cell-intrinsic IFN-γ signaling was required for PD-1- and LAG-3-deficient CD8+ T cells to enhance anti-tumor immunity, providing insight into how combinatorial targeting of LAG-3 and PD-1 enhances efficacy.


Subject(s)
Antigens, CD , CD8-Positive T-Lymphocytes , Interferon-gamma , Lymphocyte Activation Gene 3 Protein , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor , Programmed Cell Death 1 Receptor/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Animals , Interferon-gamma/metabolism , Mice , Antigens, CD/metabolism , Autocrine Communication , Humans , Melanoma/immunology , Melanoma/drug therapy , Female , Cell Line, Tumor , Melanoma, Experimental/immunology , T-Cell Exhaustion
13.
Int J Mol Sci ; 25(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39125969

ABSTRACT

To date, not many studies have presented evidence of SARS-CoV-2 infecting the female reproductive system. Furthermore, so far, no effect of the administration of anti-COVID 19 vaccines has been reported to affect the quality of oocytes retrieved from women who resorted to assisted reproduction technology (ART). The FF metabolic profiles of women who had been infected by SARS-CoV-2 before IVF treatments or after COVID-19 vaccination were examined by 1H NMR. Immunochemical characterization of proteins and cytokines involved in the redox and inflammatory pathways was performed. The increased expression of SOD2 and NQO1, the lack of alteration of IL-6 and CXCL10 levels, as well as the increased expression of CD39, suggested that, both sharing similar molecular mechanisms or proceeding along different routes, the redox balance is controlled in the FF of both vaccinated and recovered women compared to controls. The lower amount of metabolites known to have proinflammatory activity, i.e., TMAO and lipids, further supported the biochemical results, suggesting that the FF microenvironment is controlled so as to guarantee oocyte quality and does not compromise the outcome of ART. In terms of the number of blastocysts obtained after ICSI and the pregnancy rate, the results are also comforting.


Subject(s)
COVID-19 Vaccines , COVID-19 , Follicular Fluid , Metabolomics , Oxidation-Reduction , SARS-CoV-2 , Humans , Female , COVID-19/prevention & control , COVID-19/immunology , COVID-19/metabolism , Follicular Fluid/metabolism , Adult , COVID-19 Vaccines/immunology , SARS-CoV-2/immunology , Pregnancy , Metabolomics/methods , Superoxide Dismutase/metabolism , Inflammation/metabolism , Cytokines/metabolism , Vaccination , Antigens, CD/metabolism , Metabolome , Apyrase
14.
Nat Commun ; 15(1): 6726, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39112502

ABSTRACT

Arteriviruses infect a variety of mammalian hosts, but the receptors used by these viruses to enter cells are poorly understood. We identified the neonatal Fc receptor (FcRn) as an important pro-viral host factor via comparative genome-wide CRISPR-knockout screens with multiple arteriviruses. Using a panel of cell lines and divergent arteriviruses, we demonstrate that FcRn is required for the entry step of arterivirus infection and serves as a molecular barrier to arterivirus cross-species infection. We also show that FcRn synergizes with another known arterivirus entry factor, CD163, to mediate arterivirus entry. Overexpression of FcRn and CD163 sensitizes non-permissive cells to infection and enables the culture of fastidious arteriviruses. Treatment of multiple cell lines with a pre-clinical anti-FcRn monoclonal antibody blocked infection and rescued cells from arterivirus-induced death. Altogether, this study identifies FcRn as a novel pan-arterivirus receptor, with implications for arterivirus emergence, cross-species infection, and host-directed pan-arterivirus countermeasure development.


Subject(s)
Histocompatibility Antigens Class I , Receptors, Fc , Receptors, Virus , Receptors, Fc/metabolism , Receptors, Fc/genetics , Humans , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/genetics , Animals , Receptors, Virus/metabolism , Receptors, Virus/genetics , Cell Line , Virus Internalization , Antigens, CD/metabolism , Antigens, CD/genetics , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/genetics , HEK293 Cells
15.
Front Immunol ; 15: 1397967, 2024.
Article in English | MEDLINE | ID: mdl-38947317

ABSTRACT

Introduction: CD39 plays an important role in the immunoregulation and inhibition of effector cells. It is expressed on immune cells, including Tregs, and on extracellular vesicles (EVs) budding from the plasma membrane. Platelet transfusion may induce alloimmunization against HLA-I antigens, leading to refractoriness to platelet transfusion with severe consequences for patients. Tregs may play a key role in determining whether alloimmunization occurs in patients with hematologic disorders. We hypothesized that CD39+ EVs might play an immunoregulatory role, particularly in the context of platelet transfusions in patients with hematologic disorders. Such alloimmunization leads to the production of alloantibodies and is sensitive to the regulatory action of CD39. Methods: We characterized CD39+ EVs in platelet concentrates by flow cytometry. The absolute numbers and cellular origins of CD39+ EVs were evaluated. We also performed functional tests to evaluate interactions with immune cells and their functions. Results: We found that CD39+ EVs from platelet concentrates had an inhibitory phenotype that could be transferred to the immune cells with which they interacted: CD4+ and CD8+ T lymphocytes (TLs), dendritic cells, monocytes, and B lymphocytes (BLs). Moreover, the concentration of CD39+ EVs in platelet concentrates varied and was very high in 10% of concentrates. The number of these EVs present was determinant for EV-cell interactions. Finally, functional interactions were observed with BLs, CD4+ TLs and CD39+ EVs for immunoglobulin production and lymphoproliferation, with potential implications for the immunological management of patients.


Subject(s)
Blood Platelets , Extracellular Vesicles , Tetraspanin 29 , Humans , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Blood Platelets/immunology , Blood Platelets/metabolism , Tetraspanin 29/metabolism , Cell Communication/immunology , Platelet Transfusion , Female , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Male , Apyrase/metabolism , Apyrase/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Antigens, CD
16.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(6): 1126-1134, 2024 Jun 20.
Article in Chinese | MEDLINE | ID: mdl-38977342

ABSTRACT

OBJECTIVE: To investigate the protective effect of recombinant Schistosoma japonicum cystatin (rSj-Cys) against acute liver injury induced by lipopolysaccharide (LPS) and D-GalN in mice. METHODS: Adult male C57BL/6J mice with or without LPS/D-GaIN-induced acute liver injury were given intraperitoneal injections of rSj-Cys or PBS 30 min after modeling (n=18), and serum and liver tissues samples were collected from 8 mice in each group 6 h after modeling. The survival of the remaining 10 mice in each group within 24 h was observed. Serum levels of ALT, AST, TNF-α and IL-6 of the mice were measured, and liver pathologies was observed with HE staining. The hepatic expressions of macrophage marker CD68, Bax, Bcl-2 and endoplasmic reticulum stress (ERS)-related proteins were detected using immunohistochemistry or immunoblotting, and TUNEL staining was used to detect hepatocyte apoptosis. RESULTS: The survival rates of PBS- and rSj-Cys-treated mouse models of acute liver injury were 30% and 80% at 12 h and were 10% and 60% at 24 h after modeling, respectively; no death occurred in the two control groups within 24 h. The mouse models showed significantly increased serum levels of AST, ALT, IL-6 and TNF-α and serious liver pathologies with increased hepatic expressions of CD68 and Bax, lowered expression of Bcl-2, increased hepatocyte apoptosis, and up-regulated expressions of ERS-related signaling pathway proteins GRP78, CHOP and NF-κB p-p65. Treatment of the mouse models significantly lowered the levels of AST, ALT, IL-6 and TNF-α, alleviated liver pathologies, reduced hepatic expressions of CD68, Bax, GRP78, CHOP and NF-κB p-p65, and enhanced the expression of Bcl-2. In the normal control mice, rSj-Cys injection did not produce any significant changes in these parameters compared with PBS. CONCLUSION: rSj-Cys alleviates LPS/D-GalN-induced acute liver injury in mice by suppressing ERS, attenuating inflammation and inhibiting hepatocyte apoptosis.


Subject(s)
Apoptosis , Cystatins , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Hepatocytes , Inflammation , Mice, Inbred C57BL , Schistosoma japonicum , Animals , Mice , Endoplasmic Reticulum Stress/drug effects , Apoptosis/drug effects , Male , Hepatocytes/metabolism , Hepatocytes/drug effects , Cystatins/pharmacology , Liver/pathology , Liver/metabolism , Lipopolysaccharides , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6/metabolism , Recombinant Proteins/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Galactosamine , Antigens, CD/metabolism , Chemical and Drug Induced Liver Injury/drug therapy , CD68 Molecule
17.
Hematology ; 29(1): 2372482, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38994874

ABSTRACT

BACKGROUND: CD83 are closely related to the pathogenesis of immune thrombocytopenia (ITP), but the exact mechanism remains unclear. AIM: To explore the relationship between CD83 and CD4+ T cell subsets and clarify the role of CD83 in the pathogenesis of ITP. METHODS: RT-qPCR and Flow cytometry were used to illustrate CD83 expression. The downregulation and overexpression of DC-CD83 were co-cultured with CD4+ T cells to detect cell proliferation, co-cultured supernatant cytokines and Tregs expression. RESULTS: The results indicate that the ITP patients showed higher expression of CD83 than the healthy controls. The proliferation of CD4+ T cells was inhibited by downregulation of DCs-CD83 but promoted by overexpression of DCs-CD83. siRNA-CD83 inhibited proinflammatory IFN-γ and IL-17 secretion while raising TGF-ß, IL-10 concentrations. Overexpression of DCs-CD83 promoted Tregs expression. CONCLUSION: The Th1/Th2 and Th17/Tregs polarization were reversed via interfering DCs with siRNA-CD83. CD83 plays an important role in ITP pathogenesis, suggesting novel treatment for ITP patients.


Subject(s)
Antigens, CD , CD83 Antigen , Immunoglobulins , Membrane Glycoproteins , Purpura, Thrombocytopenic, Idiopathic , Humans , Purpura, Thrombocytopenic, Idiopathic/immunology , Purpura, Thrombocytopenic, Idiopathic/pathology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Antigens, CD/metabolism , Immunoglobulins/genetics , Immunoglobulins/metabolism , Female , Male , Adult , Middle Aged , Cytokines/metabolism , T-Lymphocytes, Regulatory/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism
18.
Front Immunol ; 15: 1415102, 2024.
Article in English | MEDLINE | ID: mdl-39007132

ABSTRACT

Human regulatory T cells (Treg) suppress other immune cells. Their dysfunction contributes to the pathophysiology of autoimmune diseases, including type 1 diabetes (T1D). Infusion of Tregs is being clinically evaluated as a novel way to prevent or treat T1D. Genetic modification of Tregs, most notably through the introduction of a chimeric antigen receptor (CAR) targeting Tregs to pancreatic islets, may improve their efficacy. We evaluated CAR targeting of human Tregs to monocytes, a human ß cell line and human islet ß cells in vitro. Targeting of HLA-A2-CAR (A2-CAR) bulk Tregs to HLA-A2+ cells resulted in dichotomous cytotoxic killing of human monocytes and islet ß cells. In exploring subsets and mechanisms that may explain this pattern, we found that CD39 expression segregated CAR Treg cytotoxicity. CAR Tregs from individuals with more CD39low/- Tregs and from individuals with genetic polymorphism associated with lower CD39 expression (rs10748643) had more cytotoxicity. Isolated CD39- CAR Tregs had elevated granzyme B expression and cytotoxicity compared to the CD39+ CAR Treg subset. Genetic overexpression of CD39 in CD39low CAR Tregs reduced their cytotoxicity. Importantly, ß cells upregulated protein surface expression of PD-L1 and PD-L2 in response to A2-CAR Tregs. Blockade of PD-L1/PD-L2 increased ß cell death in A2-CAR Treg co-cultures suggesting that the PD-1/PD-L1 pathway is important in protecting islet ß cells in the setting of CAR immunotherapy. In summary, introduction of CAR can enhance biological differences in subsets of Tregs. CD39+ Tregs represent a safer choice for CAR Treg therapies targeting tissues for tolerance induction.


Subject(s)
Apyrase , Receptors, Chimeric Antigen , T-Lymphocytes, Regulatory , Humans , Apyrase/immunology , Apyrase/metabolism , T-Lymphocytes, Regulatory/immunology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Cytotoxicity, Immunologic , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , HLA-A2 Antigen/immunology , HLA-A2 Antigen/genetics , HLA-A2 Antigen/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Antigens, CD
19.
Bull Exp Biol Med ; 177(1): 115-123, 2024 May.
Article in English | MEDLINE | ID: mdl-38963596

ABSTRACT

The cardiac perivascular niche is a cellular microenvironment of a blood vessel. The principles of niche regulation are still poorly understood. We studied the effect of TGFß1 on cells forming the cardiac perivascular niche using 3D cell culture (cardiospheres). Cardiospheres contained progenitor (c-Kit), endothelial (CD31), and mural (αSMA) cells, basement membrane proteins (laminin) and extracellular matrix proteins (collagen I, fibronectin). TGFß1 treatment decreased the length of CD31+ microvasculature, VE cadherin protein level, and proportion of NG2+ cells, and increased proportion of αSMA+ cells and transgelin/SM22α protein level. We supposed that this effect is related to the stabilizing function of TGFß1 on vascular cells: decreased endothelial cell proliferation, as shown for HUVEC, and activation of mural cell differentiation.


Subject(s)
Cell Differentiation , Cell Proliferation , Transforming Growth Factor beta1 , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factor beta1/metabolism , Cell Differentiation/drug effects , Humans , Cell Proliferation/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Animals , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Cadherins/metabolism , Laminin/metabolism , Laminin/pharmacology , Muscle Proteins/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Endothelial Cells/cytology , Fibronectins/metabolism , Fibronectins/pharmacology , Antigens, CD/metabolism , Myocardium/metabolism , Myocardium/cytology , Stem Cell Niche/drug effects , Stem Cell Niche/physiology , Collagen Type I/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/cytology , Cell Culture Techniques, Three Dimensional/methods
20.
Int J Mol Sci ; 25(13)2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38999968

ABSTRACT

Renal cell carcinoma (RCC) accounts for approximately 90-95% of all kidney cancers in adults, with clear cell RCC (ccRCC) being the most frequently identified subtype. RCC is known for its responsiveness to immunotherapy, making it an area of significant research interest. Immune checkpoint (IC) molecules, which regulate immune surveillance, are established therapeutic targets in RCC. The aim of this study was to analyze the influence of HVEM and CD160 gene polymorphisms on ccRCC susceptibility and patient overall survival (OS) over a ten-year period of observation. We genotyped three HVEM single nucleotide polymorphisms (SNPs): rs1886730, rs2234167, and rs8725, as well as two CD160 SNPs: rs744877 and rs2231375, in 238 ccRCC patients and 521 controls. Our findings indicated that heterozygosity within rs2231375 and/or rs2234167 increases ccRCC risk. Furthermore, in women, heterozygosity within HVEM SNPs rs8725 and rs1886730 is also associated with an increased ccRCC risk. The presence of a minor allele for rs1886730, rs2234167, rs8725, and rs2231375 was also correlated with certain clinical features of ccRCC. Moreover, rs1886730 was found to be associated with OS. In conclusion, our study highlights an association between HVEM and CD160 polymorphisms and the risk of developing ccRCC as well as OS.


Subject(s)
Antigens, CD , Carcinoma, Renal Cell , GPI-Linked Proteins , Genetic Predisposition to Disease , Kidney Neoplasms , Polymorphism, Single Nucleotide , Receptors, Tumor Necrosis Factor, Member 14 , Humans , Female , Male , Receptors, Tumor Necrosis Factor, Member 14/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/pathology , Middle Aged , Antigens, CD/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/mortality , Kidney Neoplasms/pathology , Aged , GPI-Linked Proteins/genetics , Receptors, Immunologic/genetics , Adult , Case-Control Studies , Genotype
SELECTION OF CITATIONS
SEARCH DETAIL