Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 14849, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34290311

ABSTRACT

More than 70% of colorectal, prostate, ovarian, pancreatic and breast cancer specimens show expression of CD276 (B7-H3), a potential immune checkpoint family member. Several studies have shown that high CD276 expression in cancer cells correlates with a poor clinical prognosis. This has been associated with the presence of lower tumor infiltrating leukocytes. Among those, tumor-associated macrophages can comprise up to 50% of the tumor mass and are thought to support tumor growth through various mechanisms. However, a lack of information on CD276 function and interaction partner(s) impedes rigorous evaluation of CD276 as a therapeutic target in oncology. Therefore, we aimed to understand the relevance of CD276 in tumor-macrophage interaction by employing a 3D spheroid coculture system with human cells. Our data show a role for tumor-expressed CD276 on the macrophage recruitment into the tumor spheroid, and also in regulation of the extracellular matrix modulator PAI-1. Furthermore, our experiments focusing on macrophage-expressed CD276 suggest that the antibody-dependent CD276 engagement triggers predominantly inhibitory signaling networks in human macrophages.


Subject(s)
B7 Antigens/physiology , Gene Expression Regulation, Neoplastic/genetics , Gene Expression/genetics , Macrophages/pathology , Neoplasms/pathology , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , B7 Antigens/genetics , B7 Antigens/metabolism , Cell Line, Tumor , Humans , Leukocytes/pathology , Molecular Targeted Therapy , Neoplasms/immunology , Neoplasms/therapy , Prognosis , Signal Transduction/genetics , Signal Transduction/physiology
2.
Front Immunol ; 12: 676181, 2021.
Article in English | MEDLINE | ID: mdl-34093577

ABSTRACT

Immune checkpoints (ICs) have pivotal roles in regulating immune responses. The inhibitory ICs in the tumor microenvironment (TME) have been implicated in the immune evasion of tumoral cells. Therefore, identifying and targeting these inhibitory ICs might be critical for eliminating tumoral cells. V-domain immunoglobulin suppressor of T cell activation (VISTA) is a novel inhibitory IC that is expressed on myeloid cells, lymphoid cells, and tumoral cells; therefore, VISTA can substantially regulate innate and adaptive anti-tumoral immune responses. Besides, growing evidence indicates that VISTA blockade can enhance the sensitivity of tumoral cells to conventional IC-based immunotherapy, e.g., cytotoxic T lymphocyte antigen 4 (CTLA-4) inhibitors. In this regard, the current study aimed to review the current evidence about the structure and expression pattern of VISTA, its role in TME, the clinicopathological significance of VISTA, and its prognostic values in various cancers. Besides, this review intended to collect the lessons from the recent pre-clinical and clinical studies and propose a strategy to overcome tumor immune-resistance states.


Subject(s)
B7 Antigens/physiology , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/drug therapy , B7 Antigens/antagonists & inhibitors , B7 Antigens/chemistry , Clinical Trials as Topic , Humans , Tumor Microenvironment
3.
Int J Mol Sci ; 22(5)2021 Mar 06.
Article in English | MEDLINE | ID: mdl-33800752

ABSTRACT

The members of the B7 family, as immune checkpoint molecules, can substantially regulate immune responses. Since microRNAs (miRs) can regulate gene expression post-transcriptionally, we conducted a scoping review to summarize and discuss the regulatory cross-talk between miRs and new B7 family immune checkpoint molecules, i.e., B7-H3, B7-H4, B7-H5, butyrophilin like 2 (BTNL2), B7-H6, B7-H7, and immunoglobulin like domain containing receptor 2 (ILDR2). The current study was performed using a six-stage methodology structure and Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guideline. PubMed, Embase, Scopus, Cochrane, ProQuest, and Google Scholar were systematically searched to obtain the relevant records to 5 November 2020. Two authors independently reviewed the obtained records and extracted the desired data. After quantitative and qualitative analyses, we used bioinformatics approaches to extend our knowledge about the regulatory cross-talk between miRs and the abovementioned B7 family members. Twenty-seven articles were identified that fulfilled the inclusion criteria. Studies with different designs reported gene-miR regulatory axes in various cancer and non-cancer diseases. The regulatory cross-talk between the aforementioned B7 family molecules and miRs might provide valuable insights into the pathogenesis of various human diseases.


Subject(s)
B7 Antigens/physiology , Immune Checkpoint Proteins/physiology , MicroRNAs/physiology , B7 Antigens/genetics , B7 Antigens/immunology , Blood Coagulation , Chemokines/physiology , Complement Activation , Computational Biology/methods , Disease/genetics , Gene Regulatory Networks , Humans , Immune Checkpoint Proteins/genetics , Immune Checkpoint Proteins/immunology , MicroRNAs/genetics , Signal Transduction
4.
Int J Radiat Biol ; 97(2): 149-155, 2021.
Article in English | MEDLINE | ID: mdl-33253600

ABSTRACT

PURPOSE: DNA damage is one of the main consequences of exposure to ionizing irradiation (IR). Recent studies indicate that IR can modulate the expression of immune system-related genes. However, the effects of IR on the expression of genes and pathways of the B7-CD28 superfamily remain poorly defined. The aim of this study was to evaluate the modulation of genes and pathways related to the B7-CD28 superfamily in response to IR. MATERIALS AND METHODS: In this study, we used transcriptome data available from the Gene Expression Omnibus (GEO) database to investigate the modulation of the response of genes and pathways of samples of human peripheral blood irradiated with doses of 150, 300, and 600 cGy. The data were obtained at 6 and 24 h after irradiation. The relationship between genes and pathways was established through the Reactome database. The behavior of these pathways was analyzed using mathematical methods based on relative activity and diversity. Analysis of variance (ANOVA) followed by multiple comparisons tests (Bonferroni and Tamhanes) was used to identify differentially expressed genes. Data on transcriptomes were analyzed through ViaComplex V.1.0 and IBM SPSS Statistics 22. RESULTS: For the pathways investigated in this study, we observed that the effects produced by these doses significantly modified the behavior of five pathways associated with the immune system. Also, the dose of 300 cGy might trigger signaling for the activation of T cells through the negative regulation (p < .05) of the co-inhibitory PDCD1LG2 gene. Positive regulation caused by 300 cGy (p < .05) of the CD80 receptor was observed by us, which might be related to a stimulatory signal. According to our findings, this dose induced the production of cytokines and genes that are associated with the activation and differentiation of T cells. CONCLUSIONS: Our findings indicate that the irradiation modulated the organization of the biological system, suggesting that 300 cGy is more efficient in activating the immune system.


Subject(s)
B7 Antigens/genetics , Blood Cells/radiation effects , CD28 Antigens/genetics , B7 Antigens/physiology , Blood Cells/immunology , CD28 Antigens/physiology , Female , Gene Expression/radiation effects , Humans , Male , Signal Transduction/radiation effects
5.
J Invest Dermatol ; 141(5): 1177-1187.e3, 2021 05.
Article in English | MEDLINE | ID: mdl-33091425

ABSTRACT

Acute graft-versus-host disease (aGVHD) induced by allogenic hematopoietic stem cell transplantation is an immunological disorder in which donor lymphocytes attack recipient organs. It has been proven that recipient nonhematopoietic tissue cells, such as keratinocytes, are sufficient as immunological targets for allogenic donor T cells, whereas Langerhans cells (LCs) are potent professional hematopoietic antigen-presenting cells existing in the target epidermis and eliminated during the early phase of mucocutaneous aGVHD. Moreover, LCs have been reported to negatively regulate various types of immune responses. Here, we present data showing that initial depletion of recipient LCs exacerbates mucocutaneous lesions in a murine model of allogenic bone marrow transplantation-induced aGVHD. Furthermore, another murine model of mucocutaneous aGVHD induced in mice with keratinocytes genetically expressing chicken ovalbumin by transfer of ovalbumin-specific CD8+ OT-I cells also showed that LC-depleted recipient mice develop aggravated mucocutaneous disease owing to decreased apoptosis of skin-infiltrating OT-I cells. Moreover, coexisting LCs directly induce apoptosis and inhibit the proliferation of OT-I cells in vitro partially via B7 family proteins. Collectively, our results indicate that LCs negatively regulate mucocutaneous aGVHD-like lesions in situ by inhibiting the number of infiltrating CD8+ T cells.


Subject(s)
Bone Marrow Transplantation/adverse effects , CD8-Positive T-Lymphocytes/immunology , Graft vs Host Disease/immunology , Langerhans Cells/physiology , Skin Diseases/immunology , Acute Disease , Animals , Apoptosis , B7 Antigens/physiology , Cells, Cultured , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , V-Set Domain-Containing T-Cell Activation Inhibitor 1/physiology
6.
BMC Endocr Disord ; 20(1): 123, 2020 Aug 12.
Article in English | MEDLINE | ID: mdl-32787907

ABSTRACT

BACKGROUND: Type 1 Diabetes (T1D) is a T cell-mediated autoimmune disorder caused by the destruction of insulin-secreting cells. B7-H3 (CD276) plays a vital role in T cell response. However, B7-H3 expression and its clinical significance in T1D remain unclear. The aim of this study was to investigate the correlations between the expression of B7-H3 and clinical parameters in T1D patients. The possible role of B7-H3 gene variants with T1D was also discussed. METHODS: Four B7-H3 single nucleotide polymorphisms (SNPs) were genotyped in 121 T1D patients and 120 healthy controls by polymerase chain reaction (PCR) direct sequencing. Expression of membrane B7-H3 (mB7-H3) in peripheral blood lymphocytes was determined by flow cytometry. Levels of soluble B7-H3 (sB7-H3) in serum were analyzed by enzyme linked immunosorbent assay (ELISA). RESULTS: The B7-H3 haplotype T-A-C-T was less frequently observed in T1D patients compared to the controls (OR: 0.31, 95% CI: 0.16-0.61). B7-H3 expression on monocytes showed significant upregulation in T1D patients and was positively correlated with several clinical features including ALT, fast C-peptide 120 min, HbAlc, IFN-γ, IL-6 and TNF-α (P < 0.05). The concentration of sB7-H3 in serum increased in T1D patients (P < 0.0001). We also observed that B7-H3-T-A-C-T was associated with the decreased release of sB7-H3 but not the membrane form. CONCLUSIONS: B7-H3 may act as a potential biomarker related to the pathogenesis of T1D. The B7-H3-T-A-C-T polymorphism variant is associated with the low risk of T1D as well as less release of sB7-H3.


Subject(s)
B7 Antigens/genetics , Diabetes Mellitus, Type 1/genetics , Adult , Asian People/genetics , Asian People/statistics & numerical data , B7 Antigens/physiology , Case-Control Studies , China/epidemiology , Diabetes Mellitus, Type 1/epidemiology , Female , Gene Expression , Genetic Association Studies , Genetic Predisposition to Disease , Haplotypes , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Young Adult
7.
Science ; 367(6475)2020 01 17.
Article in English | MEDLINE | ID: mdl-31949051

ABSTRACT

Negative checkpoint regulators (NCRs) temper the T cell immune response to self-antigens and limit the development of autoimmunity. Unlike all other NCRs that are expressed on activated T lymphocytes, V-type immunoglobulin domain-containing suppressor of T cell activation (VISTA) is expressed on naïve T cells. We report an unexpected heterogeneity within the naïve T cell compartment in mice, where loss of VISTA disrupted the major quiescent naïve T cell subset and enhanced self-reactivity. Agonistic VISTA engagement increased T cell tolerance by promoting antigen-induced peripheral T cell deletion. Although a critical player in naïve T cell homeostasis, the ability of VISTA to restrain naïve T cell responses was lost under inflammatory conditions. VISTA is therefore a distinctive NCR of naïve T cells that is critical for steady-state maintenance of quiescence and peripheral tolerance.


Subject(s)
B7 Antigens/physiology , Membrane Proteins/physiology , Peripheral Tolerance/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal , B7 Antigens/genetics , Lymphocyte Activation , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Peripheral Tolerance/genetics , Receptors, Antigen, T-Cell/physiology
8.
Clin Lab Med ; 39(1): 107-123, 2019 03.
Article in English | MEDLINE | ID: mdl-30709500

ABSTRACT

There is growing evidence supporting the genetic variability outside of HLA system that is contributing to the variation in transplant outcomes. Determining novel predictors could help to identify patients at risk and tailor their immunosuppressive regimens. This article discusses the various single nucleotide polymorphisms in costimulatory molecules and cytokines that have been evaluated for their effect on transplantation. An overview of how gene polymorphism studies are conducted and factors to consider in the experimental design to ensure meaningful data can be concluded are discussed.


Subject(s)
B7 Antigens/genetics , Cytokines/genetics , Graft Rejection/immunology , Polymorphism, Single Nucleotide , Transplantation , B7 Antigens/metabolism , B7 Antigens/physiology , Genome-Wide Association Study , Humans , Receptors, Tumor Necrosis Factor/genetics , T-Lymphocytes/immunology , T-Lymphocytes/physiology , Treatment Outcome , Tumor Necrosis Factor-alpha/genetics
9.
Oncogene ; 38(1): 88-102, 2019 01.
Article in English | MEDLINE | ID: mdl-30082909

ABSTRACT

B7-H3 is a tumor-promoting glycoprotein that is expressed at low levels in most normal tissues, but is overexpressed in various human cancers which is associated with disease progression and poor patient outcome. Although numerous publications have reported the correlation between B7-H3 and cancer progression in many types of cancers, mechanistic studies on how B7-H3 regulates cancer malignancy are rare, and the mechanisms underlying the role of B7-H3 in drug resistance are almost unknown. Here we report a novel finding that upregulation of B7-H3 increases the breast cancer stem cell population and promotes cancer development. Depletion of B7-H3 in breast cancer significantly inhibits the cancer stem cells. By immunoprecipitation and mass spectrometry, we found that B7-H3 is associated with the major vault protein (MVP) and activates MEK through MVP-enhancing B-RAF and MEK interaction. B7-H3 expression increases stem cell population by binding to MVP which regulates the activation of the MAPK kinase pathway. Depletion of MVP blocks the activation of MEK induced by B7-H3 and dramatically inhibits B7-H3 induced stem cells. This study reports novel functions of B7-H3 in regulating breast cancer stem cell enrichment. The novel mechanism for B7-H3-induced stem cell propagation by regulating MVP/MEK signaling axis independent of the classic Ras pathway may have important implications in the development of strategies for overcoming cancer cell resistance to chemotherapy.


Subject(s)
B7 Antigens/physiology , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/physiology , MAP Kinase Kinase Kinases/physiology , Neoplasm Proteins/physiology , Neoplastic Stem Cells/metabolism , Vault Ribonucleoprotein Particles/physiology , Animals , B7 Antigens/antagonists & inhibitors , B7 Antigens/chemistry , B7 Antigens/genetics , Breast Neoplasms/pathology , Butadienes/pharmacology , Butadienes/therapeutic use , CRISPR-Cas Systems , Cell Line, Tumor , Cell Polarity , Enzyme Activation , Female , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , Mice , Mice, Nude , Nanog Homeobox Protein/biosynthesis , Nanog Homeobox Protein/genetics , Neoplasm Invasiveness , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Nitriles/pharmacology , Nitriles/therapeutic use , Protein Domains , Protein Interaction Mapping , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/metabolism , RNA Interference , RNA, Guide, Kinetoplastida/genetics , RNA, Small Interfering/pharmacology , Recombinant Proteins/metabolism , SOXB1 Transcription Factors/biosynthesis , SOXB1 Transcription Factors/genetics , Sequence Deletion , Spheroids, Cellular , Transfection , Up-Regulation
10.
Biochim Biophys Acta Rev Cancer ; 1868(2): 571-583, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29056539

ABSTRACT

Inhibitory or stimulatory immune checkpoint molecules are expressed on a sizeable fraction of tumor cells in different tumor types. It was thought that the main function of tumor cell-associated immune checkpoint molecules would be the modulation (down- or upregulation) of antitumor immune responses. In recent years, however, it has become clear that the expression of immune checkpoint molecules on tumor cells has important consequences on the biology of the tumor cells themselves. In particular, a causal relationship between the expression of these molecules and the acquisition of malignant traits has been demonstrated. Thus, immune checkpoint molecules have been shown to promote the epithelial-mesenchymal transition of tumor cells, the acquisition of tumor-initiating potential and resistance to apoptosis and antitumor drugs, as well as the propensity to disseminate and metastasize. Herein, we review this evidence, with a main focus on PD-L1, the most intensively investigated tumor cell-associated immune checkpoint molecule and for which most information is available. Then, we discuss more concisely other tumor cell-associated immune checkpoint molecules that have also been shown to induce the acquisition of malignant traits, such as PD-1, B7-H3, B7-H4, Tim-3, CD70, CD28, CD137, CD40 and CD47. Open questions in this field as well as some therapeutic approaches that can be derived from this knowledge, are also addressed.


Subject(s)
B7-H1 Antigen/physiology , Neoplasms/etiology , Animals , B7 Antigens/physiology , CD47 Antigen/physiology , Epithelial-Mesenchymal Transition , Humans , Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/physiology , Programmed Cell Death 1 Receptor/physiology , Proto-Oncogene Proteins c-akt/physiology , TOR Serine-Threonine Kinases/physiology , Tumor Microenvironment , V-Set Domain-Containing T-Cell Activation Inhibitor 1/physiology
11.
J Exp Med ; 214(9): 2795-2810, 2017 Sep 04.
Article in English | MEDLINE | ID: mdl-28768709

ABSTRACT

T cell-dependent germinal center (GC) responses require coordinated interactions of T cells with two antigen-presenting cell (APC) populations, B cells and dendritic cells (DCs), in the presence of B7- and CD40-dependent co-stimulatory pathways. Contrary to the prevailing paradigm, we found unique cellular requirements for B7 and CD40 expression in primary GC responses to vaccine immunization with protein antigen and adjuvant: B7 was required on DCs but was not required on B cells, whereas CD40 was required on B cells but not on DCs in the generation of antigen-specific follicular helper T cells, antigen-specific GC B cells, and high-affinity class-switched antibody production. There was, in fact, no requirement for coexpression of B7 and CD40 on the same cell in these responses. Our findings support a substantially revised model for co-stimulatory function in the primary GC response, with crucial and distinct contributions of B7- and CD40-dependent pathways expressed by different APC populations and with important implications for understanding how to optimize vaccine responses or limit autoimmunity.


Subject(s)
Antigen-Presenting Cells/physiology , B7 Antigens/physiology , CD40 Antigens/physiology , Germinal Center/physiology , Animals , Antibody Formation/physiology , B-Lymphocytes/physiology , Dendritic Cells/physiology , Immunoglobulin G/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Cell Antigen Receptor Specificity/physiology , T-Lymphocytes/physiology
12.
Cancer Res ; 76(8): 2231-42, 2016 04 15.
Article in English | MEDLINE | ID: mdl-27197253

ABSTRACT

B7-H3 is a member of B7 family of immunoregulatory transmembrane glycoproteins expressed by T cells. While B7-H3 overexpression is associated with poor outcomes in multiple cancers, it also has immune-independent roles outside T cells and its precise mechanistic contributions to cancer are unclear. In this study, we investigated the role of B7-H3 in metabolic reprogramming of cancer cells in vitro and in vivo We found that B7-H3 promoted the Warburg effect, evidenced by increased glucose uptake and lactate production in B7-H3-expressing cells. B7-H3 also increased the protein levels of HIF1α and its downstream targets, LDHA and PDK1, key enzymes in the glycolytic pathway. Furthermore, B7-H3 promoted reactive oxygen species-dependent stabilization of HIF1α by suppressing the activity of the stress-activated transcription factor Nrf2 and its target genes, including the antioxidants SOD1, SOD2, and PRX3. Metabolic imaging of human breast cancer xenografts in mice confirmed that B7-H3 enhanced tumor glucose uptake and tumor growth. Together, our results illuminate the critical immune-independent contributions of B7-H3 to cancer metabolism, presenting a radically new perspective on B7 family immunoregulatory proteins in malignant progression. Cancer Res; 76(8); 2231-42. ©2016 AACR.


Subject(s)
B7 Antigens/physiology , Breast Neoplasms/metabolism , Glucose/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Reactive Oxygen Species/metabolism , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Female , Heterografts , Humans , Mice , Mice, Nude
13.
Tumour Biol ; 37(3): 2961-71, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26411671

ABSTRACT

The coinhibitory molecules, B7-H3 and B7-H4, have shown negative regulation in T cell activation and tumor-associated macrophage (TAM) polarization in tumor-specific immunity. Here, we investigated the expression of B7-H3 and B7-H4 in human and murine esophageal squamous cell carcinoma (ESCC) tissues to define their clinical significance and mechanism in a tumor microenvironment. In the present study, B7-H3 and B7-H4 were expressed in 90.6 and 92.7 % samples, respectively. High B7-H3 and B7-H4 expression was associated with advanced TNM stage and lymph node metastasis (p < 0.05, respectively). Patients with both B7-H3 and B7-H4 high-expressed tumors had the poorest prognosis (26.7 months), whereas those with both low-expressed tumors had the best survival (56.7 months). B7-H3 and B7-H4 expression were inclined to be positively related to the infiltration intensity of Treg cells and TAMs (p < 0.05, respectively), and B7-H3 expression is negatively associated with the intensity of CD8(+) T cells (p < 0.05). In 4-nitroquinoline 1-oxide (4-NQO)-induced murine models, high B7-H3 expression could only be detected at carcinoma stage, but abnormal B7-H4 expression appeared a little earlier at dysplasia stage. In vitro studies revealed that knockdown of B7-H3 on tumor cells suppressed ESCC cell migration and invasion, while knockdown of B7-H4 could inhibit ESCC cell growth. Overall, B7-H3 and B7-H4 are involved in ESCC progression and development and their coexpression could be valuable prognostic indicators. Interference of these negative regulatory molecules might be a new strategy for treating ESCC.


Subject(s)
B7 Antigens/physiology , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/pathology , V-Set Domain-Containing T-Cell Activation Inhibitor 1/physiology , 4-Nitroquinoline-1-oxide/toxicity , Adult , Aged , Animals , B7 Antigens/analysis , Carcinoma, Squamous Cell/chemically induced , Cell Line, Tumor , Esophageal Neoplasms/chemically induced , Esophageal Squamous Cell Carcinoma , Female , Humans , Lymphocytes, Tumor-Infiltrating/physiology , Mice , Mice, Inbred C57BL , Middle Aged , Quinolones/toxicity , V-Set Domain-Containing T-Cell Activation Inhibitor 1/analysis
14.
PLoS One ; 10(6): e0130126, 2015.
Article in English | MEDLINE | ID: mdl-26065426

ABSTRACT

B7-H3 is a cell surface molecule in the immunoglobulin superfamily that is frequently upregulated in response to autoantigens and pathogens during host T cell immune responses. However, B7-H3's role in the differential regulation of T cell subsets remains largely unknown. Therefore, we constructed a new B7-H3 deficient mouse strain (B7-H3 KO) and evaluated the functions of B7-H3 in the regulation of Th1, Th2, and Th17 subsets in experimental autoimmune encephalomyelitis (EAE), experimental asthma, and collagen-induced arthritis (CIA); these mouse models were used to predict human immune responses in multiple sclerosis, asthma, and rheumatoid arthritis, respectively. Here, we demonstrate that B7-H3 KO mice have significantly less inflammation, decreased pathogenesis, and limited disease progression in both EAE and CIA mouse models when compared with littermates; these results were accompanied by a decrease in IFN-γ and IL-17 production. In sharp contrast, B7-H3 KO mice developed severe ovalbumin (OVA)-induced asthma with characteristic infiltrations of eosinophils in the lung, increased IL-5 and IL-13 in lavage fluid, and elevated IgE anti-OVA antibodies in the blood. Our results suggest B7-H3 has a costimulatory function on Th1/Th17 but a coinhibitory function on Th2 responses. Our studies reveal that B7-H3 could affect different T cell subsets which have important implications for regulating pathogenesis and disease progression in human autoimmune disease.


Subject(s)
Arthritis, Experimental/pathology , Asthma/pathology , B7 Antigens/physiology , Encephalomyelitis, Autoimmune, Experimental/pathology , Inflammation/pathology , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Animals , Apoptosis , Arthritis, Experimental/chemically induced , Arthritis, Experimental/immunology , Asthma/chemically induced , Asthma/immunology , Blotting, Western , Cell Differentiation , Cell Proliferation , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Humans , Immunoenzyme Techniques , Inflammation/chemically induced , Inflammation/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
15.
Hepatology ; 60(4): 1196-210, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24845056

ABSTRACT

UNLABELLED: The low-grade inflammatory state present in obesity contributes to obesity-related metabolic dysregulation, including nonalcoholic steatohepatitis (NASH) and insulin resistance. Intercellular interactions between immune cells or between immune cells and hepatic parenchymal cells contribute to the exacerbation of liver inflammation and steatosis in obesity. The costimulatory molecules, B7.1 and B7.2, are important regulators of cell-cell interactions in several immune processes; however, the role of B7 costimulation in obesity-related liver inflammation is unknown. Here, diet-induced obesity (DIO) studies in mice with genetic inactivation of both B7.1 and B7.2 (double knockout; DKO) revealed aggravated obesity-related metabolic dysregulation, reduced insulin signalling in the liver and adipose tissue (AT), glucose intolerance, and enhanced progression to steatohepatitis resulting from B7.1/B7.2 double deficiency. The metabolic phenotype of B7.1/B7.2 double deficiency upon DIO was accompanied by increased hepatic and AT inflammation, associated with largely reduced numbers of regulatory T cells (Tregs) in these organs. In order to assess the role of B7 costimulation in DIO in a non-Treg-lacking environment, we performed antibody (Ab)-mediated inhibition of B7 molecules in wild-type mice in DIO. Antibody-blockade of both B7.1 and B7.2 improved the metabolic phenotype of DIO mice, which was linked to amelioration of hepatic steatosis and reduced inflammation in liver and AT. CONCLUSION: Our study demonstrates a dual role of B7 costimulation in the course of obesity-related sequelae, particularly NASH. The genetic inactivation of B7.1/B7.2 deteriorates obesity-related liver steatosis and metabolic dysregulation, likely a result of the intrinsic absence of Tregs in these mice, rendering DKO mice a novel murine model of NASH. In contrast, inhibition of B7 costimulation under conditions where Tregs are present may provide a novel therapeutic approach for obesity-related metabolic dysregulation and, especially, NASH.


Subject(s)
B7 Antigens/physiology , Metabolic Syndrome/physiopathology , Non-alcoholic Fatty Liver Disease/physiopathology , Obesity/physiopathology , Animals , B7 Antigens/deficiency , B7 Antigens/genetics , Cell Communication/physiology , Disease Models, Animal , Liver/pathology , Male , Mice , Mice, Knockout , Phenotype , T-Lymphocytes, Regulatory/pathology
16.
Cancer Res ; 74(7): 1924-32, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24691993

ABSTRACT

V-domain Ig suppressor of T cell activation (VISTA) is a potent negative regulator of T-cell function that is expressed on hematopoietic cells. VISTA levels are heightened within the tumor microenvironment, in which its blockade can enhance antitumor immune responses in mice. In humans, blockade of the related programmed cell death 1 (PD-1) pathway has shown great potential in clinical immunotherapy trials. Here, we report the structure of human VISTA and examine its function in lymphocyte negative regulation in cancer. VISTA is expressed predominantly within the hematopoietic compartment with highest expression within the myeloid lineage. VISTA-Ig suppressed proliferation of T cells but not B cells and blunted the production of T-cell cytokines and activation markers. Our results establish VISTA as a negative checkpoint regulator that suppresses T-cell activation, induces Foxp3 expression, and is highly expressed within the tumor microenvironment. By analogy to PD-1 and PD-L1 blockade, VISTA blockade may offer an immunotherapeutic strategy for human cancer.


Subject(s)
B7 Antigens/physiology , Immunoglobulins/physiology , Lymphocyte Activation , T-Lymphocytes/immunology , Animals , B7 Antigens/chemistry , B7-H1 Antigen , Female , Humans , Immunoglobulins/chemistry , Mice , Mice, Inbred C57BL
17.
Cancer Res ; 74(7): 1933-44, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24691994

ABSTRACT

V-domain Ig suppressor of T-cell activation (VISTA) is a novel negative checkpoint ligand that is homologous to PD-L1 and suppresses T-cell activation. This study demonstrates the multiple mechanisms whereby VISTA relieves negative regulation by hematopoietic cells and enhances protective antitumor immunity. VISTA is highly expressed on myeloid cells and Foxp3(+)CD4(+) regulatory cells, but not on tumor cells within the tumor microenvironment (TME). VISTA monoclonal antibody (mAb) treatment increased the number of tumor-specific T cells in the periphery and enhanced the infiltration, proliferation, and effector function of tumor-reactive T cells within the TME. VISTA blockade altered the suppressive feature of the TME by decreasing the presence of monocytic myeloid-derived suppressor cells and increasing the presence of activated dendritic cells within the tumor microenvironment. In addition, VISTA blockade impaired the suppressive function and reduced the emergence of tumor-specific Foxp3(+)CD4(+) regulatory T cells. Consequently, VISTA mAb administration as a monotherapy significantly suppressed the growth of both transplantable and inducible melanoma. Initial studies explored a combinatorial regimen using VISTA blockade and a peptide-based cancer vaccine with TLR agonists as adjuvants. VISTA blockade synergized with the vaccine to effectively impair the growth of established tumors. Our study therefore establishes a foundation for designing VISTA-targeted approaches either as a monotherapy or in combination with additional immune-targeted strategies for cancer immunotherapy.


Subject(s)
B7 Antigens/physiology , Immunoglobulins/physiology , Lymphocyte Activation , Neoplasms/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Cancer Vaccines/therapeutic use , Mice , Mice, Inbred C57BL , Neoplasms/drug therapy , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment
18.
Urology ; 83(2): 324-30, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24246324

ABSTRACT

OBJECTIVE: To determine whether seminal B7-H3 levels are correlated to semen parameters and affect human sperm functions. METHODS: A total of 83 healthy donors of proven fertility (aged 22-37 years) and 176 infertile men (aged 21-38 years) were recruited. Computer-assisted semen analysis and enzyme-linked immunosorbent assay were used to assess the correlations between seminal B7-H3 levels and semen parameters. Flow cytometry and fluorescence microscopy were used to detect the putative receptor for B7-H3. Computer-assisted semen analysis and FITC-conjugated pisum sativum agglutinin staining were performed for assessing sperm motility, capacitation, and acrosome reaction (AR) after incubation with various concentrations of B7-H3 for 0-4 hours in vitro. RESULTS: Seminal B7-H3 level was significantly higher in the healthy donors than that in the infertile men (P <.05), and closely associated with sperm concentrations and progressive motility (all P <.05), but not the other parameters examined (all P >.05). A putative receptor for B7-H3 was detected on the surface of sperm, with no significant differences in expression between the healthy donors and infertile men (P >.05). Seminal B7-H3 promoted sperm progressive motility in a time- and dose-dependent manner in vitro, although having no significant influence on sperm capacitation and AR. CONCLUSION: B7-H3 showed a favorable effect on human sperm motility, without affecting sperm capacitation and AR.


Subject(s)
B7 Antigens/physiology , Sperm Motility/physiology , Adult , B7 Antigens/analysis , Humans , Male , Semen/chemistry , Semen Analysis , Young Adult
19.
Monoclon Antib Immunodiagn Immunother ; 32(6): 395-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24328742

ABSTRACT

The B7 family member B7-H3 (CD276) plays a key role during an immune response but its function remains controversial. In this study, we found that murine B7-H3 up-regulated the proliferation and cytokine production of T cells. Our study suggested that there was no interaction of murine B7-H3 with a triggering receptor expressed on myeloid cells (TREM)-like transcript 2 (TLT-2). Further studies demonstrated that mouse B7-H3 specifically bound to T cells and its receptor was not murine TLT-2. Moreover, murine B7-H3 was a positive co-stimulatory molecule in the regulation of T cell-mediated immune responses.


Subject(s)
B7 Antigens/physiology , Lymphocyte Activation , T-Lymphocytes/immunology , Animals , CHO Cells , Cell Proliferation , Cricetinae , Cricetulus , Cytokines/metabolism , HEK293 Cells , Humans , Mice , Protein Binding , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism
20.
Exp Cell Res ; 319(1): 96-102, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-22999863

ABSTRACT

Macrophages are the prominent components of solid tumors and have complex dual functions in their interaction with cancer cells. Strong evidence suggests that TAM is a part of inflammatory circuits that promote tumor progression. B7-homologue 3 (B7-H3), a recently identified homologue of B7.1/2 (CD80/86), has been described to exert co-stimulatory and immune regulatory functions. Here, we showed that a fraction of macrophages in tumor stroma expressed surface B7-H3 molecule. Normal macrophages, which did not express B7-H3, would be induced expressing B7-H3 molecule when culturing with tumor cell. Although a lung cancer cell line constitutively expressed B7-H3 mRNA and protein in plasma, primary tumor cell isolated from the transplanted lung carcinoma model expressed B7-H3 on the surface. Interestingly, in transplanted lung carcinoma model, the expression of membrane-bound B7-H3 in tumor cells was increased as prolonging of tumor transformation. In support, IL-10 released from TAM could stimulate cancer cell expression of membrane bound B7-H3. Furthermore, Lung cancer and TAM-related B7-H3 was identified as a strong inhibitor of T-cell effect and influenced the outcome of T cell immune response. In conclusion, TAM-tumor cell interaction-induced membrane-bound B7-H3 represents a novel immune escape mechanism which links the pro-inflammatory response to immune tolerance in the tumor milieu.


Subject(s)
B7 Antigens/biosynthesis , B7 Antigens/genetics , Carcinoma, Lewis Lung/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , T-Lymphocytes/immunology , Animals , B7 Antigens/physiology , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Coculture Techniques , Female , Lung Neoplasms/pathology , Macrophages, Alveolar/pathology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation/methods , T-Lymphocytes/pathology , Tumor Escape/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...