Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 635: 283-290, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36308908

ABSTRACT

The V-set and transmembrane domain-containing protein (VSTM) family is a newly discovered immunoglobulin (Ig) superfamily that shares structural similarities with the B7-like transmembrane proteins. Although most VSTM5 members have been reported to exert immune-related functions, VSTM5 has been described as a regulator of neuronal morphogenesis and migration in the brain. Based on its close phylogenetic relationship with two immune checkpoints, VISTA and TIGIT, we investigated the potential role of VSTM5 in T-cell immune responses. VSTM5.Ig inhibits T-cell proliferation and cytokine production, induces T-cell apoptosis, and promotes the generation of regulatory T cells (Tregs) in in vitro T-cell assays. VSTM5 also contributes to the maintenance of T-cell anergy in vitro. Similarly, serum VSTM5.Ig produced using a recombinant plasmid in ovalbumin (OVA)-immunized mice inhibits both naive and effector T-cell immune responses. In addition, VSTM5.Ig enhances oral tolerance of cell-mediated and antibody responses in OVA-fed mice by inducing Tregs and T-cell clonal deletion. Consequently, our findings suggest that VSTM5 is a novel immune checkpoint that could be used to improve the therapeutic efficacy of tolerance-based therapies for autoimmune diseases.


Subject(s)
Antibody Formation , Immune Tolerance , Membrane Proteins , Animals , Mice , Immunoglobulins , Membrane Proteins/metabolism , Ovalbumin , Phylogeny , T-Lymphocytes, Regulatory
2.
Biochem Biophys Res Commun ; 628: 18-24, 2022 11 05.
Article in English | MEDLINE | ID: mdl-36063598

ABSTRACT

VSIG4, a newly identified co-inhibitory molecule belonging to the B7-related family, is exclusively expressed on tissue-resident macrophages and is involved in the suppression of T cell proliferation and cytokine production. We sought to characterize the role of VSIG4 in anti-tumor immunity in the tumor microenvironment, focusing on VSIG4-expressing tumor-associated macrophages (TAMs). We found that VSIG4-expressing TAMs negatively regulated antigen-specific T cell proliferation and cytokine production through direct inhibition via cell cycle arrest, but not apoptosis, as well as through their arginase 1 activity. Furthermore, VSIG4-expressing TAMs suppress tumor-specific CD8+ T cell cytotoxicity. Therefore, our results suggest that VSIG4-expressing TAMs could be a negative cellular regulator of anti-tumor immunity in the tumor microenvironment.


Subject(s)
Receptors, Complement , Tumor Microenvironment , Tumor-Associated Macrophages , Animals , Arginase/genetics , Arginase/metabolism , Cytokines/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Receptors, Complement/metabolism , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism
3.
Int J Immunopathol Pharmacol ; 36: 3946320221125588, 2022.
Article in English | MEDLINE | ID: mdl-36083857

ABSTRACT

OBJECTIVES: Lymphocyte-activation gene 3 (LAG-3) represents a potential immune checkpoint target for cancer treatment. We investigated LAG-3 expression and its prognostic value in patients with surgically treated clear cell renal cell carcinoma (RCC) and correlated LAG-3 expression with programmed cell death ligand 1(PD-L1). METHODS: We evaluated LAG-3 and PD-L1 expression using immunohistochemistry on tissue microarrays incorporating 134 primary excision specimens of clear cell RCC (ccRCC). The patients were analyzed as two groups: the whole cohort and those with metastatic RCC (mRCC). The cancer genome atlas (TCGA) data analysis of LAG-3 was done through UALCAN web servers. RESULTS: Using the UALCAN cancer transcriptional data analysis, we found that LAG-3 was overexpressed in ccRCC. LAG-3 expression was significantly correlated with PD-L1 expression in the whole cohort and in the mRCC group (all, p < 0.05). Both LAG-3⁺ RCC and PD-L1⁺ RCC presented with a higher TNM stage and higher Fuhrman nuclear grade (all, p < 0.05). PD-L1⁺/LAG-3⁺ RCC and PD-L1⁻/LAG-3⁺ RCC showed poorer cancer-specific survival (CSS) than PD-L1⁻/LAG-3⁻ RCC (all, p = 0.01). Similarly, PD-L1⁺/LAG-3⁺ mRCC and PD-L1⁻/LAG-3⁺ mRCC showed poorer CSS than PD-L1⁻/LAG-3⁻ mRCC (all, p < 0.05). Multivariate analysis showed that PD-L1⁺/LAG-3⁺ mRCC (hazard ratio: 3.19; 95% CI: 0.77-13.67; p = 0.033) was a predictor of poor CSS. CONCLUSION: Both LAG-3⁺ and PD-L1⁺ RCC have adverse pathological features, and their coexpression predicts worse clinical outcomes. Our findings suggest LAG-3 blockade in combination with programmed cell death 1/PD-L1 blockade as a potential therapeutic approach for RCC.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , B7-H1 Antigen/genetics , Carcinoma, Renal Cell/drug therapy , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/surgery , Lymphocytes/metabolism
4.
Cell Death Dis ; 12(6): 526, 2021 05 22.
Article in English | MEDLINE | ID: mdl-34023853

ABSTRACT

Thymic atrophy in sepsis is a critical disadvantage because it induces immunosuppression and increases the mortality rate as the disease progresses. However, the exact mechanism of thymic atrophy has not been fully elucidated. In this study, we discovered a novel role for VSIG4-positive peritoneal macrophages (V4(+) cells) as the principal cells that induce thymic atrophy and thymocyte apoptosis. In CLP-induced mice, V4(+) cells were activated after ingestion of invading microbes, and the majority of these cells migrated into the thymus. Furthermore, these cells underwent a phenotypic shift from V4(+) to V4(-) and from MHC II(low) to MHC II(+). In coculture with thymocytes, V4(+) cells mainly induced apoptosis in DP thymocytes via the secretion of TNF-α. However, there was little effect on CD4 or CD8 SP and DN thymocytes. V4(-) cells showed low levels of activity compared to V4(+) cells. Thymic atrophy in CLP-induced V4(KO) mice was much less severe than that in CLP-induced wild-type mice. In addition, V4(KO) peritoneal macrophages also showed similar activity to V4(-) cells. Taken together, the current study demonstrates that V4(+) cells play important roles in inducing immunosuppression via thymic atrophy in the context of severe infection. These data also suggest that controlling the function of V4(+) cells may play a crucial role in the development of new therapies to prevent thymocyte apoptosis in sepsis.


Subject(s)
Macrophages, Peritoneal/physiology , Receptors, Complement/metabolism , Sepsis/pathology , Thymocytes/physiology , Animals , Apoptosis/genetics , Cecum/pathology , Cecum/surgery , Disease Models, Animal , Female , Ligation , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Punctures , Receptors, Complement/genetics , Sepsis/genetics , Sepsis/metabolism , Thymocytes/metabolism , Thymocytes/pathology , Tumor Necrosis Factor-alpha/metabolism
5.
Cancer Immunol Immunother ; 70(11): 3113-3122, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33770210

ABSTRACT

V-domain immunoglobulin suppressor of T cell activation (VISTA) is an immune checkpoint molecule expressed in hematopoietic cells, granulocytes, macrophages, and monocytes. However, few studies to date have investigated VISTA expression, especially its clinical utility, in bladder cancer. The present retrospective study aimed to examine VISTA, programmed death ligand-1 (PD-L1), and CD45 expression by immunohistochemical and immunofluorescence staining of archived pathological tissue samples from 159 patients with primary bladder cancer. The correlation between VISTA expression in immune cells (ICs) and clinicopathologic variables including PD-L1 expression in ICs was examined. Briefly, the rates of VISTA-positive ICs and VISTA-positive tumor cells were 67.9% (108/159) and 30.8% (49/159), respectively. The VISTA expression in ICs of patients with bladder cancer, including those with non-muscle-invasive bladder cancer (NMIBC), was positively correlated with tumor stage, grade, size, and multiplicity. The VISTA expression in ICs was stronger in bladder cancer cases with PD-L1-positive ICs than in those with PD-L1-negative ICs (p < 0.001). The mean intravesical recurrence-free survival was shorter in NMIBC cases with VISTA-positive ICs than in those with VISTA-negative ICs (34.0 vs 39.9 months, p = 0.03, log-rank test). In this first study to investigate VISTA expression in bladder cancer, these results implicate VISTA as a potential immunotherapeutic target and immunologic biomarker in bladder cancer.


Subject(s)
B7 Antigens/metabolism , Biomarkers, Tumor/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasm Recurrence, Local/metabolism , Urinary Bladder Neoplasms/pathology , Aged , Biomarkers, Tumor/metabolism , Female , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Male , Middle Aged , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Retrospective Studies , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/metabolism
6.
BMB Rep ; 54(1): 12-20, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33298250

ABSTRACT

In the last decade, we have witnessed an unprecedented clinical success in cancer immunotherapies targeting the programmed cell-death ligand 1 (PD-L1) and programmed cell-death 1 (PD-1) pathway. Besides the fact that PD-L1 plays a key role in immune regulation in tumor microenvironment, recently a plethora of reports has suggested a new perspective of non-immunological functions of PD-L1 in the regulation of cancer intrinsic activities including mesenchymal transition, glucose and lipid metabolism, stemness, and autophagy. Here we review the current understanding on the regulation of expression and intrinsic protumoral activity of cancer-intrinsic PD-L1. [BMB Reports 2021; 54(1): 12-20].


Subject(s)
B7-H1 Antigen/immunology , Neoplasms/immunology , B7-H1 Antigen/genetics , Humans , Immunotherapy , Neoplasms/therapy , Tumor Microenvironment/immunology
7.
Clin Cancer Res ; 26(7): 1644-1655, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31941832

ABSTRACT

PURPOSE: Immune-checkpoint inhibitors have shown therapeutic efficacy in various malignant diseases. However, anti-programmed death (PD)-1 therapy has not shown clinical efficacy in multiple myeloma. EXPERIMENTAL DESIGN: Bone marrow (BM) mononuclear cells were obtained from 77 newly diagnosed multiple myeloma patients. We examined the expression of immune-checkpoint receptors in BM CD8+ T cells and their functional restoration by ex vivo treatment with anti-PD-1 and TGFß inhibitors. RESULTS: We confirmed the upregulation of PD-1 and PD-L1 expression in CD8+ T cells and myeloma cells, respectively, from the BM of multiple myeloma patients. PD-1-expressing CD8+ T cells from the BM of multiple myeloma patients coexpressed other checkpoint inhibitory receptors and exhibited a terminally differentiated phenotype. These results were also observed in BM CD8+ T cells specific to myeloma antigens NY-ESO-1 and HM1.24. BM CD8+ T cells from multiple myeloma patients exhibited reduced proliferation and cytokine production upon T-cell receptor stimulation. However, anti-PD-1 did not increase the proliferation of BM CD8+ T cells from multiple myeloma patients, indicating that T-cell exhaustion in multiple myeloma is hardly reversed by PD-1 blockade alone. Intriguingly, anti-PD-1 significantly increased the proliferation of BM CD8+ T cells from multiple myeloma patients in the presence of inhibitors of TGFß, which was overexpressed by myeloma cells. CONCLUSIONS: Our findings indicate that combined blockade of PD-1 and TGFß may be useful for the treatment of multiple myeloma.


Subject(s)
Antibodies, Monoclonal/pharmacology , Bone Marrow/immunology , CD8-Positive T-Lymphocytes/immunology , Multiple Myeloma/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, Antigen, T-Cell/immunology , Transforming Growth Factor beta/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Bone Marrow/drug effects , Bone Marrow/pathology , Cells, Cultured , Female , Humans , Male , Middle Aged , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Programmed Cell Death 1 Receptor/immunology , Transforming Growth Factor beta/metabolism
8.
Biol Pharm Bull ; 42(6): 915-922, 2019 Jun 01.
Article in English | MEDLINE | ID: mdl-30918133

ABSTRACT

The goal of the present study focused on the adverse reaction of contrast medium (CM) via the induction of inflammatory molecules in human umbilical vein endothelial cells (HUVECs). Ultravist-induced monocyte chemoattractant protein-1 (MCP-1) and vascular cell adhesion molecule-1 (VCAM-1) gene expression was markedly increased in interleukin-4 (IL-4)-pretreated HUVECs in a time- and dose-dependent manner and was paralleled by concomitant production of MCP-1 and VCAM-1 proteins. MCP-1 and VCAM-1 gene expression by Ultravist in combination with IL-4 was mediated by the c-Jun N-terminal kinases (JNK1/2) signaling pathway. IL-4-pretreated Ultravist-stimulated HUVECs showed greatly increased migration and adhesion of THP-1 cells. Cell migration was decreased by treatment of CCR2 antagonist, and cell adhesion was also decreased by VCAM-1 blocking antibody. Furthermore, when tested in vivo under similar conditions, MCP-1 protein was significantly increased in Ultravist combined with IL-4-injected mice. Taken together, our findings suggest that MCP-1 blocking may be crucial in preventing the endothelial dysfunction induced by contrast medium in patients with inflammatory disease and atherosclerosis.


Subject(s)
Chemokine CCL2/biosynthesis , Human Umbilical Vein Endothelial Cells/drug effects , Interleukin-4/pharmacology , Iohexol/analogs & derivatives , Vascular Cell Adhesion Molecule-1/biosynthesis , Animals , Cell Adhesion/drug effects , Cell Movement/drug effects , Cells, Cultured , Chemokine CCL2/metabolism , Contrast Media , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Iohexol/pharmacology , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction , THP-1 Cells , Vascular Cell Adhesion Molecule-1/metabolism
9.
Eur J Immunol ; 48(4): 632-643, 2018 04.
Article in English | MEDLINE | ID: mdl-29235119

ABSTRACT

Oral tolerance can prevent unnecessary immune responses against dietary antigens. Members of the B7 protein family play critical roles in the positive and/or negative regulation of T cell responses to interactions between APCs and T cells. V-set and Ig domain-containing 4 (VSIG4), a B7-related co-signaling molecule, has been known to act as a co-inhibitory ligand and may be critical in establishing immune tolerance. Therefore, we investigated the regulation of VSIG4 signaling in a food allergy and experimental oral tolerance murine models. We analyzed the contributions of the two main sites involved in oral tolerance, the mesenteric lymph node (MLN) and the liver, in VSIG4-mediated oral tolerance induction. Through the comparative analysis of major APCs, dendritic cells (DCs) and macrophages, we found that Kupffer cells play a critical role in inducing regulatory T cells (Tregs) and establishing immune tolerance against oral antigens via VSIG4 signaling. Taken together, these results suggest the possibility of VSIG4 signaling-based regulation of orally administered antigens.


Subject(s)
Dendritic Cells/immunology , Immune Tolerance/immunology , Kupffer Cells/immunology , Liver/immunology , Receptors, Complement/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, Differentiation/metabolism , B7 Antigens/immunology , Cells, Cultured , Female , Immunoglobulin G/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Liver/cytology , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/administration & dosage , Ovalbumin/immunology , Receptors, CCR7/genetics , Receptors, Complement/genetics , Signal Transduction
10.
Oncotarget ; 8(35): 58122-58132, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28938542

ABSTRACT

Multiple myeloma (MM) remains as an incurable disease, despite recent substantial improvements in treatment. Therefore, development of novel biomarkers for risk stratification and new therapeutic targets are imperative. One of the emerging treatments for MM is the immune checkpoint blockades. V-set Ig domain-containing 4 (VSIG4) is a lately studied B7-related immune checkpoint modulator. We assessed the VSIG4 expression in patients with MM and its prognostic impact. We analyzed 81 bone marrow and 66 extramedullary biopsy samples of MM patients using immunohistochemistry. VSIG4 mRNA expression data from the Multiple Myeloma Genomics Portal (MMGP) were analyzed to validate our results. The overall survival (OS) of the high VSIG4 expression group was significantly poorer than that of the low VSIG4 expression group (p = 0.046). VSIG4 expression was remained statistically significant after adjustment for revised international staging system (rISS) and Mayo stratification algorithm (mSMART) risk classification, respectively (p = 0.019 and 0.017). Corroborating results were also observed on analyses of VSIG4 expression in patients with extramedullary MM and external data from the MMGP. Our results suggest that VSIG4 expression in MM is an independent indicator of poor prognosis, implying a possible therapeutic target for immunotherapy for MM.

11.
Proc Natl Acad Sci U S A ; 114(29): E5881-E5890, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28673995

ABSTRACT

The lung is a prototypic organ that was evolved to reduce immunopathology during the immune response to potentially hazardous endogenous and exogenous antigens. In this study, we show that donor CD4+ T cells transiently induced expression of indoleamine 2,3-dioxygenase (IDO) in lung parenchyma in an IFN-γ-dependent manner early after allogeneic hematopoietic stem cell transplantation (HSCT). Abrogation of host IDO expression by deletion of the IDO gene or the IFN-γ gene in donor T cells or by FK506 treatment resulted in acute lethal pulmonary inflammation known as idiopathic pneumonia syndrome (IPS). Interestingly, IL-6 strongly induced IDO expression in an IFN-γ-independent manner when deacetylation of STAT3 was inhibited. Accordingly, a histone deacetylase inhibitor (HDACi) could reduce IPS in the state where IFN-γ expression was suppressed by FK506. Finally, l-kynurenine produced by lung epithelial cells and alveolar macrophages during IPS progression suppresses the inflammatory activities of lung epithelial cells and CD4+ T cells through the aryl hydrocarbon receptor pathway. Taken together, our results reveal that IDO is a critical regulator of acute pulmonary inflammation and that regulation of IDO expression by HDACi may be a therapeutic approach for IPS after HSCT.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hematopoietic Stem Cell Transplantation , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Pneumonia/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/immunology , Female , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation/mortality , Histone Deacetylase Inhibitors/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Kynurenine/metabolism , Lung/immunology , Lung/metabolism , Lung/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Pneumonia/drug therapy , Receptors, Aryl Hydrocarbon/immunology , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/immunology , Tacrolimus/pharmacology , Interferon gamma Receptor
12.
Cancer Immunol Res ; 4(9): 779-88, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27440711

ABSTRACT

B7 homolog 1 (B7-H1)-expressing myeloma cells not only inhibit myeloma-specific cytotoxic T lymphocytes (CTL), but also confer a proliferative advantage: resistance to antimyeloma chemotherapy. However, it remains unknown whether B7-H1 expressed on myeloma cells induces cellular responses associated with aggressive myeloma behaviors. To address this question, we analyzed the proliferation and drug sensitivity of B7-H1-expressing myeloma cells transfected with B7-H1-specific short-hairpin RNA or treated with programmed cell death (PD)-1-Fc-coupled beads. Knockdown of B7-H1 expression in myeloma cells significantly inhibited cell proliferation and increased apoptosis induced by the chemotherapeutic alkylating agent melphalan, with downregulation of the expression of cell cycle-related genes (CCND3 and CDK6) and antiapoptotic genes (BCL2 and MCL1). B7-H1 molecules thus contributed to myeloma cell-cycle progression and suppression of drug-induced apoptosis. B7-H1-expressing myeloma cells had a higher affinity for PD-1 than for CD80. PD-1-Fc bead-treated myeloma cells also became resistant to apoptosis that was induced by melphalan and the proteasome inhibitor bortezomib. Apoptosis resistance was associated with the PI3K/AKT pathway. Both myeloma cell drug resistance and antiapoptotic responses occurred through the PI3K/AKT signaling pathway, initiated from "reverse" stimulation of B7-H1 by PD-1. Therefore, B7-H1 itself may function as an oncogenic protein in myeloma cells. The interaction between B7-H1 on myeloma cells and PD-1 molecules not only inhibits tumor-specific CTLs but also induces drug resistance in myeloma cells through the PI3K/AKT signaling pathway. These observations provide mechanistic insights into potential immunotherapeutic benefits of blocking the B7-H1-PD-1 pathway. Cancer Immunol Res; 4(9); 779-88. ©2016 AACR.


Subject(s)
B7-H1 Antigen/metabolism , Drug Resistance, Neoplasm , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Programmed Cell Death 1 Receptor/metabolism , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , B7-1 Antigen/metabolism , B7-H1 Antigen/genetics , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm/genetics , Gene Knockdown Techniques , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Phosphatidylinositol 3-Kinases/metabolism , Programmed Cell Death 1 Receptor/genetics , Protein Binding , Proto-Oncogene Proteins c-akt , Signal Transduction
14.
Int J Mol Med ; 36(6): 1464-78, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26498453

ABSTRACT

Hepatitis C virus (HCV) E2 protein binds to CD81, which is a component of the B cell co-stimulatory complex. The E2-CD81 interaction leads to B cell proliferation, protein tyrosine phosphorylation and to the hypermutation of immunoglobulin genes. Epidemiological studies have reported a high prevalence of B cell non-Hodgkin lymphoma (NHL) in HCV-positive patients, suggesting a potential association between HCV and Epstein-Barr virus (EBV) in the genesis of B lymphocyte proliferative disorders. In the present study, in order to investigate the association between EBV and HCV in B cells, we created an in vitro EBV-induced B cell transformation model. CD81 was gradually overexpressed during transformation by EBV. B cells isolated from HCV-positive patients grew more rapidly and clumped together earlier than B cells isolated from healthy donors following EBV infection. Pre-stimulation of CD81 expressed by resting B cells with anti-CD81 monoclonal antibody (mAb) or HCV E2 accelerated the generation of lymphoblastoid cell lines (LCLs) by EBV infection. These cells proliferated prominently through the early expression of interleukin-10 and intracellular latent membrane protein (LMP)-l. By contrast, the overexpression of CD81 on EBV-transformed B cells by anti-CD81 mAb or HCV E2 protein induced apoptosis through reactive oxygen species (ROS)-mediated mitochondrial dysfunction. These results suggest that the engagement of CD81 expressed by B cells has differential effects on B cell fate (proliferation or apoptosis) according to EBV infection and the expression level of CD81.


Subject(s)
Apoptosis/immunology , B-Lymphocytes/immunology , Cell Proliferation , Cell Transformation, Viral/immunology , Herpesvirus 4, Human/immunology , Tetraspanin 28/immunology , Adult , Antibodies/immunology , Antibodies/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Blotting, Western , Cells, Cultured , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/virology , Female , Hepacivirus/immunology , Hepacivirus/physiology , Hepatitis C/immunology , Hepatitis C/metabolism , Hepatitis C/virology , Herpesvirus 4, Human/physiology , Host-Pathogen Interactions/immunology , Humans , Male , Membrane Potential, Mitochondrial/immunology , Microscopy, Confocal , Middle Aged , Protein Binding , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tetraspanin 28/metabolism , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism
15.
Int Immunopharmacol ; 29(2): 488-493, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26462591

ABSTRACT

Our previous study demonstrated that G-CSF treatment increased the expression of TLR2 in donor grafts; this contributed to rapid engraftment after allogeneic hematopoietic stem cell transplantation (HSCT) in mice. In the current study, we investigated the effects of upregulated TLR2 expression in G-CSF-mobilized donor grafts on acute graft-versus-host disease (GVHD). We found that TLR2 was highly expressed on myeloid cell populations but not T and B cells from the spleens of G-CSF-treated donor mice. After transplantation, the mortality and disease severity in recipients were not significantly different between G-CSF-treated TLR2-/- and wt donor grafts. Although endogenous TLR2 ligand was detected in the serum of both recipients, T cells from TLR2-/- and wt donors have the same ability regarding alloreactivity. Moreover, the blockade of TLR2 signaling in recipients by administering anti-TLR2 blocking antibody after BMT did not lead to a significant difference in acute GVHD compared with control IgG treatment. However, the hematopoietic ability of G-CSF-mobilized lin−c-kit+ HSCs from TLR2-/- donor grafts was lower than that from wt donor grafts. Our results demonstrate that upregulated TLR2 expression in G-CSF-mobilized donor grafts has no effect on acute GVHD, suggesting that TLR2 is a valuable target for increasing HSCT efficiency in order to enhance engraftment without exacerbating acute GVHD.


Subject(s)
Graft vs Host Disease/immunology , Granulocyte Colony-Stimulating Factor/pharmacology , Toll-Like Receptor 4/biosynthesis , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Hematopoietic Stem Cell Transplantation/methods , Mice , Mice, Knockout , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Stem Cell Transplantation , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Toll-Like Receptor 4/genetics
16.
Immunol Lett ; 165(2): 78-83, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25929804

ABSTRACT

VSIG4 acts as a co-inhibitory ligand to negatively regulate T cell proliferation and cytokine production, and its expression is restricted to macrophages. We hypothesized that endogenous VSIG4 impairs helper T cell functions and then inhibits the subsequent antibody response. Isotype switching of ovalbumin (OVA)-specific antibody subclasses to IgG1, IgG2a, IgG2b, and IgG3 was enhanced in OVA-immunized VSIG4 knockout (KO) mice. 2,4,6-Trinitrophenyl hapten (TNP) - Keyhole Limpet Hemocyanin (KLH)-primed B cells cocultured with OVA-primed CD4(+) T cells from OVA-immunized VSIG4 KO mice in the presence of TNP-OVA showed enhanced isotype switching to IgG subclasses compared to those cocultured with cells isolated from OVA-immunized wild-type (WT) mice. Furthermore, the levels of CD40L expression, the frequency of memory CD4(+) T cells, and the production of isotype switching-inducing cytokines increased significantly in OVA-primed CD4(+) T cells from VSIG4 KO mice. T cells from OVA-specific T cell receptor (TCR) transgenic mice produced more IFN-γ when cocultured with macrophages from VSIG4 KO mice compared to WT mice. Thus, our results demonstrate that macrophage-associated VSIG4 plays a negative role in helper T cell-dependent isotype switching by inhibiting helper T cell activation and differentiation, and suppressing the isotype switching-inducing cytokine production in antigen-primed CD4(+) helper T cells.


Subject(s)
Macrophages/physiology , Receptors, Complement/metabolism , T-Lymphocytes, Helper-Inducer/physiology , Animals , Antibody Formation/genetics , CD40 Ligand/metabolism , Cell Differentiation/genetics , Coculture Techniques , Cytokines/metabolism , HEK293 Cells , Humans , Immunoglobulin Class Switching/genetics , Immunologic Memory/genetics , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Complement/genetics
17.
J Immunol ; 194(12): 5801-11, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25934860

ABSTRACT

Regulatory T (Treg) cells act as terminators of T cell immuniy during acute phase of viral infection; however, their role and suppressive mechanism in chronic viral infection are not completely understood. In this study, we compared the phenotype and function of Treg cells during acute or chronic infection with lymphocytic choriomeningitis virus. Chronic infection, unlike acute infection, led to a large expansion of Treg cells and their upregulation of programmed death-1 (PD-1). Treg cells from chronically infected mice (chronic Treg cells) displayed greater suppressive capacity for inhibiting both CD8(+) and CD4(+) T cell proliferation and subsequent cytokine production than those from naive or acutely infected mice. A contact between Treg and CD8(+) T cells was necessary for the potent suppression of CD8(+) T cell immune response. More importantly, the suppression required cell-specific expression and interaction of PD-1 on chronic Treg cells and PD-1 ligand on CD8(+) T cells. Our study defines PD-1 upregulated on Treg cells and its interaction with PD-1 ligand on effector T cells as one cause for the potent T cell suppression and proposes the role of PD-1 on Treg cells, in addition to that on exhausted T cells, during chronic viral infection.


Subject(s)
B7-H1 Antigen/genetics , Immunomodulation , Programmed Cell Death 1 Receptor/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Virus Diseases/genetics , Virus Diseases/immunology , Animals , Antigens, CD/metabolism , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Communication/immunology , Chronic Disease , Disease Models, Animal , Female , Gene Expression , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Immunophenotyping , Integrin alpha Chains/metabolism , Lymphocyte Activation/immunology , Lymphocyte Count , Lymphocyte Depletion , Mice , Mice, Knockout , Phenotype , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
18.
Biochem Biophys Res Commun ; 459(2): 277-283, 2015 Apr 03.
Article in English | MEDLINE | ID: mdl-25725157

ABSTRACT

Aberrant B7-H4 expression in cancer tissues serves as a novel prognostic biomarker for poor survival in patients with cancer. However, the factor(s) that induce cancer cell-associated B7-H4 remain to be fully elucidated. We herein demonstrate that hypoxia upregulates B7-H4 transcription in primary CD138(+) multiple myeloma cells and cancer cell lines. In support of this finding, analysis of the Multiple Myeloma Genomics Portal (MMGP) data set revealed a positive correlation between the mRNA expression levels of B7-H4 and the endogenous hypoxia marker carbonic anhydrogenase 9. Hypoxia-induced B7-H4 expression was detected in the cytoplasm, but not in cancer cell membranes. Chromatin immunoprecipitation analysis demonstrated binding of hypoxia-inducible factor-1α (HIF-1α) to proximal hypoxia-response element (HRE) sites within the B7-H4 promoter. Knockdown of HIF-1α and pharmacological inhibition of HIF-1α diminished B7-H4 expression. Furthermore, knockdown of cytoplasmic B7-H4 in MCF-7 decreased the S-phase cell population under hypoxia. Finally, MMGP analysis revealed a positive correlation between the transcript levels of B7-H4 and proliferation-related genes including MKI67, CCNA1, and Myc in several patients with multiple myeloma. Our results provide insight into the mechanisms underlying B7-H4 upregulation and its role in cancer cell proliferation in a hypoxic tumor microenvironment.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Multiple Myeloma/metabolism , V-Set Domain-Containing T-Cell Activation Inhibitor 1/biosynthesis , Cell Hypoxia/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Chromatin Immunoprecipitation , Cytoplasm/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , MCF-7 Cells , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Promoter Regions, Genetic , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tumor Microenvironment/genetics , Tumor Microenvironment/physiology , V-Set Domain-Containing T-Cell Activation Inhibitor 1/antagonists & inhibitors , V-Set Domain-Containing T-Cell Activation Inhibitor 1/genetics
19.
Radiat Res ; 183(3): 271-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25706775

ABSTRACT

This study investigated the therapeutic effects of combined local irradiation and anti-HER2/neu antibody in a mixed tumor mouse model comprised of a nonmetastatic neu-positive tumor and a metastatic neu-negative tumor. While local irradiation alone could control the primary tumor in a dose-dependent manner, it did not improve mouse survival. Combined treatment comprised of local irradiation and anti-neu antibody of tumor-bearing BALB/c mice significantly improved mouse survival (P < 0.5), even though the tumor growth was similar to that of the irradiated-alone group. The combined treatment significantly reduced metastatic tumor masses in the lung and increased immune cell infiltration in primary tumor tissues. However, immune deficient nude mice with tumors did not exhibit prolonged survival in response to the combined treatment. Collectively, these results show that combined local irradiation and anti-neu antibody can elicit an immune-mediated abscopal effect to extend survival. Although the mechanism for abscopal effects induced by the combined treatment of radiation and anti-HER2/neu antibody was not elucidated, to our knowledge this is the first published study to describe the abscopal effect induced by the combination of local irradiation and the anti-HER2/neu antibody.


Subject(s)
Antibodies, Anti-Idiotypic/administration & dosage , Breast Neoplasms/radiotherapy , Immunotherapy , Receptor, ErbB-2/immunology , Animals , Antibodies, Anti-Idiotypic/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Female , Humans , Mice , Neoplasm Metastasis , Receptor, ErbB-2/genetics , Xenograft Model Antitumor Assays
20.
Life Sci ; 112(1-2): 82-9, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25072357

ABSTRACT

AIM: B7-H1, which belongs to the B7 family of costimulatory molecules, is implicated in the ability of tumors to evade the host immune response. The development of evasion mechanisms within the tumor microenvironment may be responsible for poor therapeutic responses. In this manuscript, we report that the 15-deoxy-δ(12,14)-prostaglandin J2 (15d-PGJ2), peroxisome proliferator-activated receptor gamma (PPARγ) activator leads to the downregulation of the cancer-associated expression of B7-H1 in response to interferon-gamma (IFN-γ) and the associated signaling cascades. MAIN METHODS: The expression of B7-H1 from IFN-γ-induced B16F10 melanoma cells was measured with flow cytometric analysis. The regulatory mechanisms of 15d-PGJ2 on cellular signaling pathways were examined using Western blot and electrophoretic mobility shift assays. KEY FINDINGS: The flow cytometric analysis revealed that the B7-H1 costimulatory molecule is significantly upregulated in B16F10 melanoma cells by stimulation with IFN-γ. However, 15d-PGJ2 strongly downregulates B7-H1 expression in IFN-γ-stimulated B16F10 melanoma cells. Furthermore, the significant damping effect of 15d-PGJ2 on B7-H1 expression involves the inhibition of the tyrosine phosphorylation of Janus kinase (Jak) and signal transducer(s) and activator(s) of transcription (STAT) and, thereby, the interferon regulatory factor-1 (IRF-1) trans-activation of STAT. These effects of 15d-PGJ2 were not abrogated by the PPARγ antagonist GW9662, indicating that they occur through a PPARγ-independent mechanism. SIGNIFICANCE: In this study, we demonstrate that 15d-PGJ2 suppresses the IFN-γ-elicited expression of B7-H1 by the inhibition of IRF-1 transcription via the Jak/STAT signaling pathway through a PPARγ-independent mechanism in mouse melanoma cells.


Subject(s)
B7-H1 Antigen/genetics , Gene Expression Regulation, Neoplastic , Interferon-gamma/pharmacology , Prostaglandin D2/analogs & derivatives , Signal Transduction/drug effects , Anilides/pharmacology , Animals , B7-H1 Antigen/metabolism , Cell Line, Tumor , Interferon Regulatory Factor-1/antagonists & inhibitors , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Janus Kinases/antagonists & inhibitors , Janus Kinases/genetics , Janus Kinases/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , PPAR gamma/metabolism , Prostaglandin D2/pharmacology , STAT Transcription Factors/antagonists & inhibitors , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...