Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 13(9): 832-42, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22842346

ABSTRACT

The mechanisms by which tumor microenvironments modulate nucleic acid-mediated innate immunity remain unknown. Here we identify the receptor TIM-3 as key in circumventing the stimulatory effects of nucleic acids in tumor immunity. Tumor-associated dendritic cells (DCs) in mouse tumors and patients with cancer had high expression of TIM-3. DC-derived TIM-3 suppressed innate immune responses through the recognition of nucleic acids by Toll-like receptors and cytosolic sensors via a galectin-9-independent mechanism. In contrast, TIM-3 interacted with the alarmin HMGB1 to interfere with the recruitment of nucleic acids into DC endosomes and attenuated the therapeutic efficacy of DNA vaccination and chemotherapy by diminishing the immunogenicity of nucleic acids released from dying tumor cells. Our findings define a mechanism whereby tumor microenvironments suppress antitumor immunity mediated by nucleic acids.


Subject(s)
Dendritic Cells/immunology , HMGB1 Protein/immunology , Immunity, Innate , Neoplasms/immunology , Nucleic Acids/immunology , Receptors, Virus/immunology , Tumor Microenvironment/immunology , Animals , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , HMGB1 Protein/metabolism , Hepatitis A Virus Cellular Receptor 2 , Humans , Immunoblotting , Immunologic Surveillance/immunology , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Fluorescence , Neoplasms/metabolism , Receptors, Pattern Recognition/immunology , Receptors, Virus/metabolism , Reverse Transcriptase Polymerase Chain Reaction
2.
Allergol Int ; 73(1): 94-106, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37336695

ABSTRACT

BACKGROUND: Mepolizumab treatment improves symptom control and quality of life and reduces exacerbations in patients with severe eosinophilic asthma. However, biomarkers that predict therapeutic effectiveness must be determined for use in precision medicine. Herein, we elucidated the dynamics of various parameters before and after treatment as well as patient characteristics predictive of clinical responsiveness to mepolizumab after 1-year treatment. METHODS: Twenty-seven patients with severe asthma were treated with mepolizumab for one year. Asthma control test scores, pulmonary function tests, fractional exhaled nitric oxide levels, and blood samples were evaluated. Additionally, we explored the role of CD69-positive mucosal-associated invariant T (MAIT) cells as a candidate biomarker for predicting treatment effectiveness by evaluating an OVA-induced asthma murine model using MR1 knockout mice, where MAIT cells were absent. RESULTS: The frequencies of CD69-positive group 1 innate lymphoid cells, group 3 innate lymphoid cells, natural killer cells, and MAIT cells decreased after mepolizumab treatment. The frequency of CD69-positive MAIT cells and neutrophils was lower and serum periostin levels were higher in responders than in non-responders. In the OVA-induced asthma murine model, CD69-positive MAIT cell count in the whole mouse lung was significantly higher than that in the control mice. Moreover, OVA-induced eosinophilic airway inflammation was exacerbated in the MAIT cell-deficient MR1 knockout mice. CONCLUSIONS: This study shows that circulating CD69-positive MAIT cells, neutrophils, and serum periostin might predict the real-world response after 1-year mepolizumab treatment. Furthermore, MAIT cells potentially have a protective role against type 2 airway inflammation.


Subject(s)
Asthma , Mucosal-Associated Invariant T Cells , Humans , Animals , Mice , Neutrophils , Periostin , Immunity, Innate , Disease Models, Animal , Ovalbumin/therapeutic use , Quality of Life , Lymphocytes , Inflammation , Biomarkers , Mice, Knockout
3.
Curr Issues Mol Biol ; 45(11): 8907-8924, 2023 Nov 08.
Article in English | MEDLINE | ID: mdl-37998736

ABSTRACT

The mitogen-activated protein kinase (MAPK) signaling pathway is involved in the epithelial-mesenchymal transition (EMT) and asthma; however, the role of mitogen-activated protein kinase kinase kinase 19 (MAP3K19) remains uncertain. Therefore, we investigated the involvement of MAP3K19 in in vitro EMT and ovalbumin (OVA)-induced asthma murine models. The involvement of MAP3K19 in the EMT and the production of cytokines and chemokines were analyzed using a cultured bronchial epithelial cell line, BEAS-2B, in which MAP3K19 was knocked down using small interfering RNA. We also evaluated the involvement of MAP3K19 in the OVA-induced asthma murine model using Map3k19-deficient (MAP3K19-/-) mice. Transforming growth factor beta 1 (TGF-ß1) and tumor necrosis factor-like weak inducer of apoptosis (TWEAK) induced the MAP3K19 messenger RNA (mRNA) expression in the BEAS-2B cells. The knockdown of MAP3K19 enhanced the reduction in E-cadherin mRNA and the production of regulated upon activation normal T cell express sequence (RANTES) via stimulation with TWEAK alone or with the combination of TGF-ß1 and TWEAK. Furthermore, the expression of MAP3K19 mRNA was upregulated in both the lungs and tracheas of the mice in the OVA-induced asthma murine model. The MAP3K19-/- mice exhibited worsened eosinophilic inflammation and an increased production of RANTES in the airway epithelium compared with the wild-type mice. These findings indicate that MAP3K19 suppressed the TWEAK-stimulated airway epithelial response, including adhesion factor attenuation and RANTES production, and suppressed allergic airway inflammation in an asthma mouse model, suggesting that MAP3K19 regulates allergic airway inflammation in patients with asthma.

4.
Biochem Biophys Res Commun ; 675: 71-77, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37453260

ABSTRACT

Fibroblast growth factor 18 (FGF18) is elevated in several human cancers, such as gastrointestinal and ovarian cancers, and stimulates the proliferation of tumor cells. This suggests that FGF18 may be a promising candidate biomarker in cancer patients. However, the lack of a high-sensitivity enzyme-linked immunosorbent assay (ELISA) does not permit testing of this possibility. In this study, we generated monoclonal antibodies against human FGF18 and developed a high-sensitivity ELISA to measure human FGF18 at concentrations as low as 10 pg/mL. Of the eight tumor cell lines investigated, we detected human FGF18 in culture supernatants from four tumor cell lines, including HeLa, OVCAR-3, BxPC-3, and SW620 cells, albeit the production levels were relatively low in the latter two cell lines. Moreover, the in-house ELISA could detect murine FGF18 in sera from mice overexpressing murine Fgf18 in hepatocytes, although the sensitivity in detecting murine FGF18 was relatively low. This FGF18 ELISA could be a valuable tool to validate FGF18 as a potential biomarker for cancer patients and to test the contribution of FGF18 for various disease models invivo and in vitro.


Subject(s)
Apoptosis , Ovarian Neoplasms , Humans , Mice , Animals , Female , Cell Line, Tumor , Ovarian Neoplasms/pathology , Fibroblast Growth Factors/metabolism , Enzyme-Linked Immunosorbent Assay
5.
Immunity ; 39(6): 1070-81, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24315994

ABSTRACT

Phagocytosis of apoptotic cells by myeloid cells has been implicated in the maintenance of immune homeostasis. In this study, we found that T cell immunoglobulin- and mucin domain-containing molecule-4 (TIM-4) repressed tumor-specific immunity triggered by chemotherapy-induced tumor cell death. TIM-4 was found to be highly expressed on tumor-associated myeloid cells such as macrophages (TAMs) and dendritic cells (TADCs) and danger-associated molecular patterns (DAMPs) released from chemotherapy-damaged tumor cells induced TIM-4 on tumor-associated myeloid cells recruited from bone marrow-derived precursors. TIM-4 directly interacted with AMPKα1 and activated autophagy-mediated degradation of ingested tumors, leading to reduced antigen presentation and impaired CTL responses. Consistently, blockade of the TIM-4-AMPKα1-autophagy pathway augmented the antitumor effect of chemotherapeutics by enhancing tumor-specific CTL responses. Our finding provides insight into the immune tolerance mediated by phagocytosis of dying cells, and targeting of the TIM-4-AMPKα1 interaction constitutes a unique strategy for augmenting antitumor immunity and improving cancer chemotherapy.


Subject(s)
Antigen Presentation/immunology , Autophagy/immunology , Immune Tolerance/immunology , Macrophages/immunology , Membrane Proteins/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Dendritic Cells/immunology , Flow Cytometry , Gene Expression Regulation, Neoplastic , Mice , Neoplasms/physiopathology , Tumor Cells, Cultured
6.
Immunity ; 39(3): 584-98, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-24035363

ABSTRACT

Because immune responses simultaneously defend and injure the host, the immune system must be finely regulated to ensure the host's survival. Here, we have shown that when injected with high Toll-like receptor ligand doses or infected with lymphocytic choriomeningitis virus (LCMV) clone 13, which has a high viral turnover, inflammatory monocyte-derived dendritic cells (Mo-DCs) engulfed apoptotic erythroid cells. In this process, called hemophagocytosis, phosphatidylserine (PS) served as an "eat-me" signal. Type I interferons were necessary for both PS exposure on erythroid cells and the expression of PS receptors in the Mo-DCs. Importantly, hemophagocytosis was required for interleukin-10 (IL-10) production from Mo-DCs. Blocking hemophagocytosis or Mo-DC-derived IL-10 significantly increased cytotoxic T cell lymphocyte activity, tissue damage, and mortality in virus-infected hosts, suggesting that hemophagocytosis moderates immune responses to ensure the host's survival in vivo. This sheds light on the physiological relevance of hemophagocytosis in severe inflammatory and infectious diseases.


Subject(s)
Apoptosis/immunology , Dendritic Cells/immunology , Lymphocytic choriomeningitis virus/immunology , Phagocytosis , Animals , Cell Differentiation , Dendritic Cells/metabolism , Erythroid Cells/immunology , Interferon Type I/metabolism , Interleukin-10/biosynthesis , Lymphocyte Activation , Lymphocytic Choriomeningitis/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Phosphatidylserines/metabolism , Receptors, Cell Surface/metabolism , T-Lymphocytes, Cytotoxic/immunology
7.
Allergol Int ; 70(3): 343-350, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33640239

ABSTRACT

BACKGROUND: Previous reports have shown that pathogen-associated patterns (PAMPs) induce the production of interleukin (IL)-1ß in macrophages. Moreover, studies using mouse models also suggest that chitin, which acts as a PAMP, induces adjuvant effects and eosinophilic infiltration in the lung. Thus, we investigated the effects of inhaled chitin in mouse models. METHODS: We developed mouse models of inhaled chitin particle-induced airway inflammation and steroid-resistant ovalbumin (OVA)-induced airway inflammation. Some experimental groups of mice were treated additionally with dexamethasone (DEX). Murine alveolar macrophages (AMs), which were purified from bronchoalveolar lavage (BAL) fluids, were incubated with chitin, and treated with or without DEX. RESULTS: The numbers of total cells, AMs, lymphocytes, eosinophils, and neutrophils among BAL-derived cells, as well as the IL-1ß levels in BAL fluids and the numbers of IL-1ß-positive cells in lung, were significantly increased by chitin stimulation. Airway hyperresponsiveness (AHR) was aggravated in mice of the chitin inflammation model compared to control animals. The production of IL-1ß was significantly increased in murine AMs by chitin treatment, but DEX administration did not inhibit this chitin-induced IL-1ß production. Furthermore, in mouse models, DEX treatment inhibited the OVA-induced airway inflammation and AHR but not the airway inflammation and AHR induced by chitin or the combination of OVA and chitin. CONCLUSIONS: These results suggest that inhaled chitin induces airway inflammation, AHR, and the production of IL-1ß. Furthermore, our findings demonstrate for the first time that inhaled chitin induces steroid-resistant airway inflammation and AHR. Inhaled chitin may contribute to features of steroid-resistant asthma.


Subject(s)
Chitin/immunology , Glucocorticoids/pharmacology , Inflammation/immunology , Lung/drug effects , Macrophages, Alveolar/drug effects , Respiratory Hypersensitivity/immunology , Administration, Inhalation , Animals , Asthma/chemically induced , Asthma/immunology , Asthma/physiopathology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Chitin/pharmacology , Dexamethasone/pharmacology , Disease Models, Animal , Drug Resistance , Inflammation/chemically induced , Inflammation/physiopathology , Interleukin-1beta/drug effects , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Lung/immunology , Lung/physiopathology , Macrophages, Alveolar/immunology , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Ovalbumin/immunology , Ovalbumin/pharmacology , Pathogen-Associated Molecular Pattern Molecules , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/physiopathology
8.
Exp Eye Res ; 199: 108190, 2020 10.
Article in English | MEDLINE | ID: mdl-32798537

ABSTRACT

PURPOSE: To determine whether the CD27/CD70 pathway plays a significant role in corneal allograft rejection by investigating the effect of blocking the CD27/CD70 pathway by anti-CD70 antibody on corneal allograft survival. METHODS: Orthotopic penetrating keratoplasty was performed using C57BL/6 donor grafts and BALB/c recipients. Expression of CD27 and CD70 on rejected cornea was examined by immunohistochemistry. Corneal transplant recipients received intraperitoneal injection of anti-CD70 antibody (FR70) or control rat IgG. Alloreactivity was measured by mixed lymphoid reaction (MLR) in recipients administered control rat IgG and those administered anti-CD70 antibody. Corneal expression of IFN-γ and IL-12 was also examined in both groups. Graft opacity was assessed over an 8-week period and graft survival was evaluated using Kaplan-Meier survival curves. Proportion of CD4+CD44+ memory T cells in lymph nodes was measured by flow cytometry. RESULTS: CD4+CD27+ cells and CD11c+CD70+ cells were present in rejected cornea. Anti-CD70 antibody administration suppressed alloreactivity in corneal allograft recipients, and inhibited IFN-γ expression in recipient cornea (p < 0.05). Anti-CD70 antibody suppressed opacity score of recipient cornea and prolonged corneal allograft survival (p < 0.05). Proportion of CD4+CD44+ memory T cells in recipient lymph nodes was reduced by anti-CD70 antibody treatment. CONCLUSION: The CD27/CD70 pathway plays a significant role in corneal allograft rejection by initiating alloreactive Th1 cells and preserving memory T cells. Anti-CD70 antibody administration prolongs corneal allograft survival indicating the potential therapeutic effect of CD27/CD70 pathway blockade on corneal allograft rejection.


Subject(s)
CD27 Ligand/antagonists & inhibitors , Cornea/metabolism , Corneal Transplantation , Graft Rejection/prevention & control , Graft Survival/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/antagonists & inhibitors , Allografts , Animals , CD27 Ligand/biosynthesis , Cornea/pathology , Disease Models, Animal , Graft Rejection/immunology , Graft Rejection/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tumor Necrosis Factor Receptor Superfamily, Member 7/biosynthesis
9.
J Immunol ; 199(11): 3733-3737, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29061768

ABSTRACT

Promising results of immune checkpoint inhibitors have indicated the use of immunotherapy against malignant tumors. However, they cause serious side effects, including autoimmune diseases and pneumonitis. T cell Ig and mucin domain (TIM)-3 is a new candidate immune checkpoint molecule; however, the potential toxicity associated with anti-TIM-3 treatment is unknown. In this study, we investigated the pathological contribution of anti-TIM-3 mAb in a bleomycin-induced lung inflammation and fibrosis model. Anti-TIM-3-treated mice showed more severe inflammation and peribronchiolar fibrosis compared with control IgG-treated mice. Anti-TIM-3 mAb was associated with increased numbers of myofibroblasts, collagen deposition, and TGF-ß1 production in lungs. TIM-3 expression was only detected on alveolar macrophages that protect against fibrosis by apoptotic cell clearance. Treatment with anti-TIM-3 mAb inhibited the phagocytic ability of alveolar macrophages in vivo, resulting in the defective clearance of apoptotic cells in lungs. In summary, anti-TIM-3 mAb treatment might cause pneumonitis and it should be used with caution in clinical settings.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Hepatitis A Virus Cellular Receptor 2/immunology , Immunotherapy/methods , Lung/pathology , Macrophages, Alveolar/immunology , Pneumonia/therapy , Pulmonary Fibrosis/therapy , Animals , Apoptosis , Bleomycin , Cells, Cultured , Collagen/metabolism , Humans , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Myofibroblasts/pathology , Pneumonia/chemically induced , Pneumonia/immunology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/immunology , Transforming Growth Factor beta1/metabolism
10.
Proc Natl Acad Sci U S A ; 113(27): E3901-10, 2016 07 05.
Article in English | MEDLINE | ID: mdl-27298374

ABSTRACT

Treatment for fibrosis represents a critical unmet need, because fibrosis is the leading cause of death in industrialized countries, and there is no effective therapy to counteract the fibrotic process. The development of fibrosis relates to the interplay between vessel injury, immune cell activation, and fibroblast stimulation, which can occur in various tissues. Immunotherapies have provided a breakthrough in the treatment of immune diseases. The glycoprotein OX40-OX40 ligand (OX40L) axis offers the advantage of a targeted approach to costimulatory signals with limited impact on the whole immune response. Using systemic sclerosis (SSc) as a prototypic disease, we report compelling evidence that blockade of OX40L is a promising strategy for the treatment of inflammation-driven fibrosis. OX40L is overexpressed in the fibrotic skin and serum of patients with SSc, particularly in patients with diffuse cutaneous forms. Soluble OX40L was identified as a promising serum biomarker to predict the worsening of lung and skin fibrosis, highlighting the role of this pathway in fibrosis. In vivo, OX40L blockade prevents inflammation-driven skin, lung, and vessel fibrosis and induces the regression of established dermal fibrosis in different complementary mouse models. OX40L exerts potent profibrotic effects by promoting the infiltration of inflammatory cells into lesional tissues and therefore the release of proinflammatory mediators, thereafter leading to fibroblast activation.


Subject(s)
OX40 Ligand/antagonists & inhibitors , OX40 Ligand/blood , Pulmonary Fibrosis/prevention & control , Scleroderma, Systemic/blood , Skin/pathology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Biomarkers/blood , Bleomycin , Case-Control Studies , Cells, Cultured , Drug Evaluation, Preclinical , Fibrosis , Fos-Related Antigen-2/genetics , Humans , Hypertension, Pulmonary/prevention & control , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Molecular Targeted Therapy , Pulmonary Fibrosis/genetics , Scleroderma, Systemic/drug therapy , Skin/metabolism , Transcription Factor AP-1/metabolism
11.
Exp Lung Res ; 44(7): 332-343, 2018 09.
Article in English | MEDLINE | ID: mdl-30676129

ABSTRACT

AIM OF THE STUDY: In patients with asthma, chronic inflammatory processes and the subsequent remodeling of the airways contribute to the symptoms and the pathophysiological changes. Epithelial-mesenchymal transition (EMT) is thought to play an important role in tissue remodeling. Previous reports show that tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) is a cytokine of the TNF superfamily, exerts pro-inflammatory effects, and enhances transforming growth factor (TGF)-ß-induced EMT in bronchial epithelial cells. In this study, we investigated the TWEAK-induced cytokine and chemokine production in the human bronchial epithelial cell line BEAS-2B during EMT. MATERIALS AND METHODS: Quantitative real-time RT-PCR, enzyme-linked immunosorbent assays, western blotting, and immunohistochemistry were used to define the production of cytokines and chemokines. RESULTS: We found that TWEAK increases mRNA and protein levels of thymic stromal lymphopoietin (TSLP), monocyte chemoattractant protein -1 (MCP-1), regulated upon activation normal T cell express sequence (RANTES), and IL-8 in BEAS-2B bronchial epithelial cells. Moreover, co-treatment with TWEAK and TGF-ß1 induces not only features of EMT but also enhances the production of TSLP and RANTES. Thymus- and activation-regulated chemokines (TARC) production is induced by the co-treatment of TWEAK and TGF-ß1 but not by TWEAK or TGF-ß1 stimulation alone. Furthermore, the increased mRNA expression of TSLP and RANTES after co-treatment with TWEAK and TGF-ß1 is prevented by inhibitors of Smad-independent signaling pathways. CONCLUSIONS: In the present study, we have revealed a novel mechanism for the production of asthma-related cytokines and chemokines in EMT driven by the co-stimulation with TWEAK and TGF-ß1. We conclude that cellular EMT processes caused by TWEAK and TGF-ß1 may contribute to chronic airway inflammation and remodeling.


Subject(s)
Chemokine CCL17/biosynthesis , Chemokine CCL5/biosynthesis , Cytokine TWEAK/pharmacology , Cytokines/biosynthesis , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Transforming Growth Factor beta1/pharmacology , Airway Remodeling , Asthma/metabolism , Bronchi/cytology , Cell Line , Humans , Thymic Stromal Lymphopoietin
12.
Biochem Biophys Res Commun ; 463(4): 739-45, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26049110

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) are newly identified, potent producers of type 2 cytokines, such as IL-5 and IL-13, and contribute to the development of allergic lung inflammation induced by cysteine proteases. Although it has been shown that inducible costimulator (ICOS), a costimulatory molecule, is expressed on ILC2s, the role of ICOS in ILC2 responses is largely unknown. In the present study, we investigated whether the interaction of ICOS with its ligand B7-related protein-1 (B7RP-1) can promote ILC2 activation. Cytokine production in ILC2s purified from mouse lungs was significantly increased by coculture with B7RP-1-transfected cells, and increased cytokine production was inhibited by monoclonal antibody-mediated blocking of the ICOS/B7RP-1 interaction. ILC2 expansion and eosinophil influx induced by papain, a cysteine protease antigen, in mouse lungs were significantly abrogated by blocking the ICOS/B7RP-1 interaction. Dendritic cells (DCs) in the lungs expressed B7RP-1 and the number of DCs markedly increased with papain administration. B7RP-1 expression on lung DCs was reduced after papain administration. This downregulation of B7RP-1 expression may be an indication of ICOS/B7RP-1 binding. These results indicate that ILC2s might interact with B7RP-1-expressing DCs in allergic inflammatory lung, and ICOS signaling can positively regulate the protease allergen-induced ILC2 activation followed by eosinophil infiltration into the lungs.


Subject(s)
Immunity, Innate/immunology , Inducible T-Cell Co-Stimulator Protein/physiology , Lung/immunology , Animals , Flow Cytometry , Lung/drug effects , Male , Mice , Mice, Inbred C57BL , Papain/pharmacology
13.
Int Immunol ; 26(7): 369-81, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24486565

ABSTRACT

Infection by Theiler's murine encephalomyelitis virus (TMEV) in the central nervous system (CNS) induces an immune-mediated demyelinating disease in susceptible mouse strains and serves as a relevant infection model for human multiple sclerosis. T-cell immunoglobulin and mucin domain-3 (TIM-3) has been demonstrated to play a crucial role in the maintenance of peripheral tolerance. In this study, we examined the regulatory role of the TIM-3 pathway in the development of TMEV-induced demyelinating disease (TMEV-IDD). The expression of TIM-3 was increased at both protein and mRNA levels in the spinal cords of mice with TMEV-IDD compared with naive controls. In addition, by utilizing a blocking mAb, we demonstrate that TIM-3 negatively regulates TMEV-specific ex vivo production of IFN-γ and IL-10 by CD4(+) T cells and IFN-γ by CD8(+) T cells from the CNS of mice with TMEV-IDD at 36 days post-infection (dpi). In vivo blockade of TIM-3 by using the anti-TIM-3 mAb resulted in significant exacerbation of the development of TMEV-IDD both clinically and histologically. The number of infiltrating mononuclear cells in the CNS was also increased in mice administered with anti-TIM-3 mAb both at the induction phase (10 dpi) and at the effector phase (36 dpi). Flow cytometric analysis of intracellular cytokines revealed that the number of CD4(+) T cells producing TNF, IL-4, IL-10 and IL-17 was significantly increased at the effector phase in the CNS of anti-TIM-3 mAb-treated mice. These results suggest that the TIM-3 pathway plays a critical role in the regulation of TMEV-IDD.


Subject(s)
Poliomyelitis/genetics , RNA, Messenger/immunology , Receptors, Virus/immunology , Spinal Cord/immunology , Theilovirus/immunology , Animals , Antibodies, Monoclonal/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Disease Models, Animal , Female , Gene Expression Regulation , Hepatitis A Virus Cellular Receptor 2 , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/metabolism , Interleukin-10/biosynthesis , Interleukin-10/metabolism , Interleukin-17/biosynthesis , Interleukin-17/metabolism , Interleukin-4/biosynthesis , Interleukin-4/metabolism , Mice , Multiple Sclerosis , Peripheral Tolerance , Poliomyelitis/immunology , Poliomyelitis/pathology , Poliomyelitis/virology , RNA, Messenger/genetics , Receptors, Virus/genetics , Signal Transduction , Spinal Cord/pathology , Theilovirus/pathogenicity
14.
J Immunol ; 190(1): 88-96, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23180822

ABSTRACT

TIM-3 is constitutively expressed on subsets of macrophages and dendritic cells. Its expression on other cells of the innate immune system and its role in fetomaternal tolerance has not yet been explored. In this study, we investigate the role of TIM-3-expressing innate immune cells in the regulation of tolerance at the fetomaternal interface (FMI) using an allogeneic mouse model of pregnancy. Blockade of TIM-3 results in accumulation of inflammatory granulocytes and macrophages at the uteroplacental interface and upregulation of proinflammatory cytokines. Furthermore, TIM-3 blockade inhibits the phagocytic potential of uterine macrophages resulting in a build up of apoptotic bodies at the uteroplacental interface that elicits a local immune response. In response to inflammatory cytokines, Ly-6C(hi)G(neg) monocytic myeloid-derived suppressor cells expressing inducible NO synthase and arginase 1 are induced. However, these suppressive cells fail to downregulate the inflammatory cascade induced by inflammatory granulocytes (Ly-6C(int)G(hi)) and apoptotic cells; the increased production of IFN-γ and TNF-α by inflammatory granulocytes leads to abrogation of tolerance at the FMI and fetal rejection. These data highlight the interplay between cells of the innate immune system at the FMI and their influence on successful pregnancy in mice.


Subject(s)
Immune Tolerance , Immunity, Cellular , Immunity, Innate , Receptors, Virus/physiology , Animals , Apoptosis/immunology , Cell Line , Coculture Techniques , Female , Hepatitis A Virus Cellular Receptor 2 , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Pregnancy , Receptors, Virus/biosynthesis , Uterus/cytology , Uterus/immunology , Uterus/metabolism
15.
J Immunol ; 191(8): 4447-55, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24038092

ABSTRACT

Dendritic cells (DCs) are the central architects of the immune response, inducing inflammatory or tolerogenic immunity, dependent on their activation status. As such, DCs are highly attractive therapeutic targets and may hold the potential to control detrimental immune responses. TIM-4, expressed on APCs, has complex functions in vivo, acting both as a costimulatory molecule and a phosphatidylserine receptor. The effect of TIM-4 costimulation on T cell activation remains unclear. In this study, we demonstrate that Ab blockade of DC-expressed TIM-4 leads to increased induction of induced regulatory T cells (iTregs) from naive CD4(+) T cells, both in vitro and in vivo. iTreg induction occurs through suppression of IL-4/STAT6/Gata3-induced Th2 differentiation. In addition, blockade of TIM-4 on previously activated DCs still leads to increased iTreg induction. iTregs induced under TIM-4 blockade have equivalent potency to control and, upon adoptive transfer, significantly prolong skin allograft survival in vivo. In RAG(-/-) recipients of skin allografts adoptively transferred with CD4(+) T cells, we show that TIM-4 blockade in vivo is associated with a 3-fold prolongation in allograft survival. Furthermore, in this mouse model of skin transplantation, increased induction of allospecific iTregs and a reduction in T effector responses were observed, with decreased Th1 and Th2 responses. This enhanced allograft survival and protolerogenic skewing of the alloresponse is critically dependent on conversion of naive CD4(+) to Tregs in vivo. Collectively, these studies identify blockade of DC-expressed TIM-4 as a novel strategy that holds the capacity to induce regulatory immunity in vivo.


Subject(s)
Dendritic Cells/immunology , Graft Survival , Membrane Proteins/metabolism , Skin Transplantation , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Allografts/immunology , Animals , Antibodies, Monoclonal/immunology , Cell Differentiation/immunology , Cells, Cultured , GATA3 Transcription Factor/immunology , Homeodomain Proteins/genetics , Interleukin-4/immunology , Lymphocyte Activation , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , STAT6 Transcription Factor/immunology , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Th2 Cells/immunology
16.
J Immunol ; 191(9): 4562-72, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24068667

ABSTRACT

T cell Ig and mucin domain (TIM)-4 is involved in immune regulation. However, the pathological function of TIM-4 has not been understood and remains to be clarified in various disease models. In this study, DBA/1 mice were treated with anti-TIM-4 mAb during the induction or effector phase of collagen-induced arthritis (CIA). Anti-TIM-4 treatment in the induction phase exacerbated the development of CIA. In vitro experiments suggest that CD4 T cells bind to TIM-4 on APCs, which induces inhibitory effect to CD4 T cells. In contrast, therapeutic treatment with anti-TIM-4 mAb just before or after the onset or even at later stage of CIA significantly suppressed the development and progression by reducing proinflammatory cytokines in the ankle joints without affecting T or B cell responses. Consistently, clinical arthritis scores of collagen Ab-induced arthritis, which is not mediated by T or B cells, were significantly reduced in anti-TIM-4-treated mice with a concomitant decrease of proinflammatory cytokines in the joints. In vitro, macrophages secreted proinflammatory cytokines in response to TIM-4-Ig protein and LPS, which were reduced by the anti-TIM-4 mAb. The anti-TIM-4 mAb also inhibited the differentiation and bone-resorbing activity of osteoclasts. These results indicate that TIM-4 has two distinct functions depending on the stage of arthritis. The therapeutic effect of anti-TIM-4 mAb on arthritis is mediated by the inhibition of proinflammatory cytokine production by inflammatory cells, osteoclast differentiation, and bone resorption, suggesting that TIM-4 might be an appropriate target for the therapeutic treatment of arthritis.


Subject(s)
Antibodies, Monoclonal/immunology , Arthritis, Experimental/immunology , Membrane Proteins/immunology , Membrane Proteins/metabolism , Animals , Antibodies, Monoclonal/therapeutic use , Antigen-Presenting Cells/immunology , B-Lymphocytes/immunology , Bone Resorption/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Collagen , Cytokines/biosynthesis , Lipopolysaccharides , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Mice, Knockout , Osteoclasts/drug effects , Osteoclasts/immunology
17.
Exp Lung Res ; 41(7): 370-82, 2015.
Article in English | MEDLINE | ID: mdl-26151756

ABSTRACT

BACKGROUND:  Macrophages include the classically activated pro-inflammatory M1 macrophages (M1s) and alternatively activated anti-inflammatory M2 macrophages (M2s). The M1s are activated by both interferon-γ and Toll-like receptor ligands, including lipopolysaccharide (LPS), and have potent pro-inflammatory activity. In contrast, Th2 cytokines activate the M2s, which are involved in the immune response to parasites, promotion of tissue remodeling, and immune regulatory functions. Although alveolar macrophages (AMs) play an essential role in the pulmonary immune system, little is known about their phenotypes. METHODS:  Quantitative reverse transcription polymerase chain reaction and flow cytometry were used to define the characteristics of alveolar macrophages derived from untreated naïve mice and from murine models of both ovalbumin (OVA)-induced allergic airway inflammation and LPS-induced acute airway inflammation. AMs were co-cultured with CD4(+) T cells and were pulsed with tritiated thymidine to assess proliferative responses. RESULTS:  We characterized in detail murine AMs and found that these cells were not completely consistent with the current M1 versus M2-polarization model. OVA-induced allergic and LPS-induced acute airway inflammation promoted the polarization of AMs towards the current M2-skewed and M1-skewed phenotypes, respectively. Moreover, our data also show that CD11c(+) CD11b(+) AMs from the LPS-treated mice play a regulatory role in antigen-specific T-cell proliferation in vitro. CONCLUSIONS:  These characteristics of AMs depend on the incoming pathogens they encounter and on the phase of inflammation and do not correspond to the current M1 versus M2-polarization model. These findings may facilitate an understanding of their contributions to the pulmonary immune system in airway inflammation.


Subject(s)
Hypersensitivity/immunology , Inflammation/immunology , Lipopolysaccharides/immunology , Lung/immunology , Macrophages, Alveolar/immunology , Ovalbumin/immunology , Allergens/immunology , Animals , Asthma/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation/physiology , Cytokines/immunology , Female , Mice , Mice, Inbred BALB C
18.
Am J Physiol Renal Physiol ; 306(10): F1210-21, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24623145

ABSTRACT

The T-cell immunoglobulin mucin 1, also known as kidney injury molecule-1, modulates CD4+ T-cell responses and is also expressed by damaged proximal tubules within the kidney. Both Th subset imbalance (Th1/Th2/Th17) and regulatory T-cell and B-cell alterations contribute to the pathogenesis of autoimmune disease. This study investigated the effects of an inhibitory anti-T-cell immunoglobulin mucin 1 antibody (RMT1-10) in lupus-prone MRL-Fas(lpr) mice. MRL-Fas(lpr) mice were treated with RMT1-10 or a control antibody intraperitoneally twice weekly from 3 mo of age for 16 wk. RMT1-10 treatment significantly improved survival, limited the development of lymphadenopathy and skin lesions, preserved renal function and decreased proteinuria, reduced serum anti-DNA antibody levels, and attenuated renal leukocyte accumulation. Th1 and Th17 cellular responses systemically and intrarenally were reduced, but regulatory T and B cells were increased. RMT1-10 treatment also reduced glomerular immunoglobulin and C3 deposition and suppressed cellular proliferation and apoptosis. Urinary excretion and renal expression of kidney injury molecule-1 was reduced, reflecting diminished interstitial injury. As RMT1-10 attenuated established lupus nephritis, manipulating immune system T-cell immunoglobulin mucin 1 may represent a therapeutic strategy in autoimmune diseases affecting the kidney.


Subject(s)
Autoimmune Diseases/physiopathology , Kidney Diseases/physiopathology , Lupus Erythematosus, Systemic/physiopathology , Membrane Proteins/physiology , Severity of Illness Index , Animals , Antibodies, Anti-Idiotypic/immunology , Antibodies, Anti-Idiotypic/pharmacology , Apoptosis/drug effects , Autoimmune Diseases/epidemiology , Cell Proliferation/drug effects , Disease Models, Animal , Female , Hepatitis A Virus Cellular Receptor 1 , Kidney/pathology , Kidney Diseases/epidemiology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/immunology , Mice , Mice, Inbred MRL lpr , Proteinuria/prevention & control
19.
Biochem Biophys Res Commun ; 444(2): 235-40, 2014 Feb 07.
Article in English | MEDLINE | ID: mdl-24462862

ABSTRACT

In mice, splenic conventional dendritic cells (cDCs) can be separated, based on their expression of CD8α into CD8(-) and CD8(+) cDCs. Although previous experiments demonstrated that injection of antigen (Ag)-pulsed CD8(-) cDCs into mice induced CD4 T cell differentiation toward Th2 cells, the mechanism involved is unclear. In the current study, we investigated whether OX40 ligand (OX40L) on CD8(-) cDCs contributes to the induction of Th2 responses by Ag-pulsed CD8(-) cDCs in vivo, because OX40-OX40L interactions may play a preferential role in Th2 cell development. When unseparated Ag-pulsed OX40L-deficient cDCs were injected into syngeneic BALB/c mice, Th2 cytokine (IL-4, IL-5, and IL-10) production in lymph node cells was significantly reduced. Splenic cDCs were separated to CD8(-) and CD8(+) cDCs. OX40L expression was not observed on freshly isolated CD8(-) cDCs, but was induced by anti-CD40 mAb stimulation for 24 h. Administration of neutralizing anti-OX40L mAb significantly inhibited IL-4, IL-5, and IL-10 production induced by Ag-pulsed CD8(-) cDC injection. Moreover, administration of anti-OX40L mAb with Ag-pulsed CD8(-) cDCs during a secondary response also significantly inhibited Th2 cytokine production. Thus, OX40L on CD8(-) cDCs physiologically contributes to the development of Th2 cells and secondary Th2 responses induced by Ag-pulsed CD8(-) cDCs in vivo.


Subject(s)
CD8 Antigens/immunology , Dendritic Cells/immunology , OX40 Ligand/immunology , Th2 Cells/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , CD40 Antigens/immunology , CD8 Antigens/metabolism , Cells, Cultured , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Female , Flow Cytometry , Hemocyanins/immunology , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-4/immunology , Interleukin-4/metabolism , Interleukin-5/immunology , Interleukin-5/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , OX40 Ligand/genetics , OX40 Ligand/metabolism , Spleen/immunology , Spleen/metabolism , Th2 Cells/drug effects , Th2 Cells/metabolism
20.
Arterioscler Thromb Vasc Biol ; 33(11): 2558-65, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23990206

ABSTRACT

OBJECTIVE: Atherosclerosis is a chronic autoimmune-like disease in which lipids and fibrous elements accumulate in the arterial blood vessels. T cells are present within atherosclerotic plaques, and their activation is partially dependent on costimulatory signals, which can either provide positive or negative signals that promote T-cell activation or limit T-cell responses, respectively. T-cell immunoglobulin and mucin domain 3 (Tim-3) is a coinhibitory type 1 transmembrane protein that affects the function of several immune cells involved in atherosclerosis, such as monocytes, macrophages, effector T cells, and regulatory T cells. In the present study, we determined the role of Tim-3 in the development of atherosclerosis. APPROACH AND RESULTS: Western-type diet-fed low-density lipoprotein receptor-deficient (LDLr(-/-)) mice were treated with an anti-Tim-3 antibody for 3 and 8 weeks. Anti-Tim-3 administration increased fatty streak formation with 66% and increased atherosclerotic plaque formation after 8 weeks with 35% in the aortic root and with 50% in the aortic arch. Furthermore, blockade of Tim-3 signaling increased percentages of circulating monocytes with 33% and lesional macrophages with 20%. In addition, anti-Tim-3 administration increased CD4(+) T cells with 17%, enhanced their activation status, and reduced percentages of regulatory T cells with 18% and regulatory B cells with 37%. CONCLUSIONS: It is known that Tim-3 acts as a negative regulator of both innate and adaptive immune responses, and in the present study, we show that anti-Tim-3 treatment augments lesion development, accompanied by an increase in the number of monocytes/macrophages and CD4(+) T cells and by decreased regulatory T cells and regulatory B cells.


Subject(s)
Atherosclerosis/immunology , Atherosclerosis/metabolism , Receptors, Virus/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Blocking/immunology , Antibodies, Blocking/pharmacology , Aorta/immunology , Aorta/metabolism , Apoptosis/immunology , B-Lymphocytes, Regulatory/immunology , Hepatitis A Virus Cellular Receptor 2 , Macrophages/metabolism , Mice , Mice, Knockout , Monocytes/immunology , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/metabolism , Receptors, Virus/genetics , Receptors, Virus/metabolism , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL