Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
J Immunother Cancer ; 11(6)2023 06.
Article in English | MEDLINE | ID: mdl-37364935

ABSTRACT

BACKGROUND: Claudin18.2 (CLDN18.2) is a tight junction protein that has been identified as a clinically proven target in gastric cancer. Stimulation of 4-1BB with agonistic antibodies is also a promising strategy for immunotherapy and 4-1BB+ T cells were reported to be present within the tumor microenvironment of patients with gastric cancer. However, hepatotoxicity-mediated by 4-1BB activation was observed in clinical trials of agonistic anti-4-1BB monoclonal antibodies. METHODS: To specifically activate the 4-1BB+ T cells in tumor and avoid the on-target liver toxicity, we developed a novel CLDN18.2×4-1BB bispecific antibody (termed 'givastomig' or 'ABL111'; also known as TJ-CD4B or TJ033721) that was designed to activate 4-1BB signaling in a CLDN18.2 engagement-dependent manner. RESULTS: 4-1BB+ T cells were observed to be coexisted with CLDN18.2+ tumor cells in proximity by multiplex immunohistochemical staining of tumor tissues from patients with gastric cancer (n=60). Givastomig/ABL111 could bind to cell lines expressing various levels of CLDN18.2 with a high affinity and induce 4-1BB activation in vitro only in the context of CLDN18.2 binding. The magnitude of T-cell activation by givastomig/ABL111 treatment was closely correlated with the CLDN18.2 expression level of tumor cells from gastric cancer patient-derived xenograft model. Mechanistically, givastomig/ABL111 treatment could upregulate the expression of a panel of pro-inflammatory and interferon-γ-responsive genes in human peripheral blood mononuclear cells when co-cultured with CLDN18.2+ tumor cells. Furthermore, in humanized 4-1BB transgenic mice inoculated with human CLDN18.2-expressing tumor cells, givastomig/ABL111 induced a localized immune activation in tumor as evident by the increased ratio of CD8+/regulatory T cell, leading to the superior antitumor activity and long-lasting memory response against tumor rechallenge. Givastomig/ABL111 was well tolerated, with no systemic immune response and hepatotoxicity in monkeys. CONCLUSIONS: Givastomig/ABL111 is a novel CLDN18.2×4-1BB bispecific antibody which has the potential to treat patients with gastric cancer with a wide range of CLDN18.2 expression level through the restricted activation of 4-1BB+ T cells in tumor microenvironment to avoid the risk of liver toxicity and systemic immune response.


Subject(s)
Antibodies, Bispecific , Chemical and Drug Induced Liver Injury , Stomach Neoplasms , Mice , Animals , Humans , Stomach Neoplasms/drug therapy , Leukocytes, Mononuclear , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Lymphocyte Activation , Mice, Transgenic , Tumor Microenvironment , Claudins
2.
Nat Med ; 19(3): 322-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23396208

ABSTRACT

Regulatory T (Treg) cells suppress autoimmune disease, and impaired Treg cell function is associated with rheumatoid arthritis. Here we demonstrate that forkhead box P3 (FOXP3) transcriptional activity and, consequently, Treg cell suppressive function are regulated by phosphorylation at Ser418 in the C-terminal DNA-binding domain. In rheumatoid arthritis-derived Treg cells, the Ser418 site was specifically dephosphorylated by protein phosphatase 1 (PP1), whose expression and enzymatic activity were induced in the inflamed synovium by tumor necrosis factor α (TNF-α), leading to impaired Treg cell function. Moreover, TNF-α-induced Treg cell dysfunction correlated with increased numbers of interleukin-17 (IL-17)(+) and interferon-γ (IFN-γ)(+)CD4(+) T cells within the inflamed synovium in rheumatoid arthritis. Treatment with a TNF-α-specific antibody restored Treg cell function in subjects with rheumatoid arthritis, which was associated with decreased PP1 expression and increased FOXP3 phosphorylation in Treg cells. Thus, TNF-α controls the balance between Treg cells and pathogenic TH17 and TH1 cells in the synovium of individuals with rheumatoid arthritis through FOXP3 dephosphorylation.


Subject(s)
Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor-alpha/metabolism , Cells, Cultured , DNA-Binding Proteins/metabolism , Forkhead Transcription Factors/chemistry , Humans , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-7 Receptor alpha Subunit/biosynthesis , Phosphorylation , Protein Phosphatase 1/metabolism , RNA Interference , RNA, Small Interfering , Synovial Membrane/immunology , Synovial Membrane/metabolism , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Tumor Necrosis Factor-alpha/immunology
3.
J Mol Cell Biol ; 5(2): 85-98, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23243069

ABSTRACT

Deficiency in the TGF-ß-induced regulatory T (iTreg) cell differentiation is associated with compromised immune homeostasis and plays a key role in many autoimmune diseases. Therapeutic intervention to enhance in situ iTreg differentiation has become a promising treatment modality for autoimmune diseases. Here we describe that the development of autoimmune inflammation in experimental autoimmune encephalomyelitis (EAE) is associated with selective impairment of iTreg differentiation largely due to the increased production of TNF-α. The neutralization of TNF-α markedly increases iTreg differentiation, leading to the amelioration of EAE, whereas the depletion of iTreg cells abolishes the therapeutic effect of an anti-TNF-α antibody. The inhibition of iTreg differentiation by TNF-α is mediated through a signaling cascade involving the induction of TNF receptor II (TNFR2) expression and the activation of Akt. The activated Akt in turn interacts with Smad3, resulting in the inhibition of TGF-ß-induced Smad3 phosphorylation and consequently the reduction of p-Smad3 results in the decreased binding to the specific binding site of the foxp3 promoter, and finally foxp3 transcription itself. Interestingly, this regulatory pathway is iTreg cell specific as TNF-α does not activate Akt in naturally occurring regulatory T cells, therefore conferring a selective effect of TNF-α and its antagonism on iTreg cells. The study sheds new light on the critical role and underlying mechanism of TNF-α in the regulation of iTreg differentiation and provides a novel rationale for TNF-α antagonistic therapy for autoimmune diseases.


Subject(s)
Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Smad3 Protein/metabolism , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Enzyme Activation/genetics , Gene Expression Regulation/genetics , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Knockout , Proto-Oncogene Proteins c-akt/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics , Receptors, Tumor Necrosis Factor, Type II/metabolism , Response Elements/genetics , Signal Transduction/genetics , Smad3 Protein/genetics , T-Lymphocytes, Regulatory/pathology , Transforming Growth Factor beta/genetics , Tumor Necrosis Factor-alpha/genetics
4.
Trends Immunol ; 34(3): 120-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23116550

ABSTRACT

In autoimmune disease, a network of diverse cytokines is produced in association with disease susceptibility to constitute the 'cytokine milieu' that drives chronic inflammation. It remains elusive how cytokines interact in such a complex network to sustain inflammation in autoimmune disease. This has presented huge challenges for successful drug discovery because it has been difficult to predict how individual cytokine-targeted therapy would work. Here, we combine the principles of Chinese Taoism philosophy and modern bioinformatics tools to dissect multiple layers of arbitrary cytokine interactions into discernible interfaces and connectivity maps to predict movements in the cytokine network. The key principles presented here have important implications in our understanding of cytokine interactions and development of effective cytokine-targeted therapies for autoimmune disorders.


Subject(s)
Autoimmune Diseases/drug therapy , Computational Biology/methods , Cytokines/antagonists & inhibitors , Religious Philosophies , Antibodies, Monoclonal/therapeutic use , Asian People , Autoimmune Diseases/immunology , Autoimmune Diseases/physiopathology , Cytokines/immunology , Cytokines/metabolism , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/physiopathology
5.
Mol Immunol ; 49(1-2): 290-6, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21943782

ABSTRACT

Anakinra, a human recombinant IL-1 receptor antagonist, is approved for the treatment of RA. In this study, 12 patients received the placebo plus MTX treatment, 38 patients received Anakinra combined with MTX treatment. Compared with the placebo plus MTX group, serum levels of IL-17, IFN-γ, IL-21 and IL-1ß significantly decreased, the percentages of Th17 cells and Th1 cells were lower and the percentage of Treg cells was higher after receiving Anakinra combined with MTX treatment. The observed regulatory immune responses collectively correlated with clinical improvement in treated patients. A substantial response, ACR 20 at 24 w were consistent with those at 12 w, 16 w and 20 w, and was accompanied by a marked improvement in RA related laboratory parameters. The study reveals that the combination of Anakinra and MTX is safe and well tolerated, which induces regulatory immune responses and significantly provides greater clinical benefit than the placebo plus MTX group.


Subject(s)
Antirheumatic Agents/administration & dosage , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Interleukin 1 Receptor Antagonist Protein/administration & dosage , Methotrexate/administration & dosage , Arthritis, Rheumatoid/blood , Cell Separation , Drug Therapy, Combination , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Interferon-gamma/blood , Interleukin-17/blood , Interleukin-1beta/blood , Interleukins/blood , Male , Middle Aged , Real-Time Polymerase Chain Reaction , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th1 Cells/drug effects , Th1 Cells/immunology , Th17 Cells/drug effects , Th17 Cells/immunology
6.
Immunity ; 35(2): 273-84, 2011 Aug 26.
Article in English | MEDLINE | ID: mdl-21835648

ABSTRACT

Neural progenitor cell (NPC) therapy is considered a promising treatment modality for multiple sclerosis (MS), potentially acting through neural repair. Here, we showed that intravenous administration of NPCs ameliorated experimental autoimmune encephalomyelitis (EAE) by selectively inhibiting pathogenic T helper 17 (Th17) cell differentiation. Leukemia inhibitory factor (LIF) produced by NPCs was responsible for the observed EAE suppression. Through the inducible LIF receptor expression, LIF inhibited the differentiation of Th17 cells in EAE mice and that from MS subjects. At the molecular level, LIF exerted an opposing effect on interleukin 6 (IL-6)-induced signal transducer and activator of transcription 3 (STAT3) phosphorylation required for Th17 cell differentiation by triggering a signaling cascade that activated extracellular signal-regulated MAP kinase (ERK) and upregulated suppressor of cytokine signaling 3 (SOCS3) expression. This study reveals a critical role for LIF in regulating Th17 cell differentiation and provides insights into the mechanisms of action of NPC therapy in MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Leukemia Inhibitory Factor/metabolism , Neurons/metabolism , Stem Cells/metabolism , Th17 Cells/metabolism , Animals , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , Neurons/immunology , Neurons/pathology , Neurons/transplantation , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Stem Cell Transplantation , Stem Cells/immunology , Stem Cells/pathology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/immunology , Suppressor of Cytokine Signaling Proteins/metabolism , Th17 Cells/immunology , Th17 Cells/pathology
7.
J Clin Invest ; 121(2): 658-70, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21266776

ABSTRACT

Th17 cells are a subset of CD4+ T cells with an important role in clearing certain bacterial and fungal pathogens. However, they have also been implicated in autoimmune diseases such as multiple sclerosis. Exposure of naive CD4+ T cells to IL-6 and TGF-ß leads to Th17 cell differentiation through a process in which many proteins have been implicated. We report here that ectopic expression of liver X receptor (LXR) inhibits Th17 polarization of mouse CD4+ T cells, while LXR deficiency promotes Th17 differentiation in vitro. LXR activation in mice ameliorated disease in the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis, whereas LXR deficiency exacerbated disease. Further analysis revealed that Srebp-1, which is encoded by an LXR target gene, mediated the suppression of Th17 differentiation by binding to the E-box element on the Il17 promoter, physically interacting with aryl hydrocarbon receptor (Ahr) and inhibiting Ahr-controlled Il17 transcription. The putative active site (PAS) domain of Ahr and the N-terminal acidic region of Srebp-1 were essential for this interaction. Additional analyses suggested that similar LXR-dependent mechanisms were operational during human Th17 differentiation in vitro. This study reports what we believe to be a novel signaling pathway underlying LXR-mediated regulation of Th17 cell differentiation and autoimmunity.


Subject(s)
Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-17/immunology , Orphan Nuclear Receptors/immunology , Th17 Cells/immunology , Animals , Autoimmunity/immunology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Liver X Receptors , Mice , Mice, Knockout , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/genetics , Signal Transduction/immunology , Sterol Regulatory Element Binding Protein 1/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Th17 Cells/cytology
8.
J Immunol ; 185(3): 1855-63, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20622114

ABSTRACT

Berberine (BBR), an isoquinoline alkaloid derived from plants, is widely used as an anti-inflammatory remedy in traditional Chinese medicine. In this study, we showed that BBR was efficacious in the amelioration of experimental autoimmune encephalomyelitis (EAE) through novel regulatory mechanisms involving pathogenic Th1 and Th17 cells. BBR inhibited differentiation of Th17 cells and, to a lesser degree, Th1 cells through direct actions on the JAK/STAT pathway, whereas it had no effect on the relative number of CD4(+)Foxp3(+) regulatory T cells. In addition, BBR indirectly influenced Th17 and Th1 cell functions through its effect on the expression and function of costimulatory molecules and the production of IL-6, which was attributable to the inhibition of NF-kappaB activity in CD11b(+) APCs. BBR treatment completely abolished the encephalitogenicity of MOG(35-55)-reactive Th17 cells in an adoptive transfer EAE model, and the same treatment significantly inhibited the ability of MOG(35-55)-reactive Th1 cells to induce EAE. This study provides new evidence that natural compounds, such as BBR, are of great value in the search for novel anti-inflammatory agents and therapeutic targets for autoimmune diseases.


Subject(s)
Berberine/therapeutic use , Cell Differentiation/drug effects , Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-17/biosynthesis , Th1 Cells/drug effects , Th1 Cells/immunology , Amino Acid Sequence , Animals , Cells, Cultured , Drugs, Chinese Herbal/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , Th1 Cells/cytology
9.
Arthritis Rheum ; 62(10): 2900-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20533542

ABSTRACT

OBJECTIVE: Osteopontin (OPN) that is aberrantly produced in rheumatoid synovium is thought to play an important role in rheumatoid arthritis (RA). This study was undertaken to investigate the role of OPN in the differentiation and accumulation of Th17 cells in rheumatoid synovium. METHODS: Peripheral blood mononuclear cells and purified CD4+ T cells derived from patients with RA or healthy controls were used to test the effect of OPN in vitro. Cytokine expression was determined by enzyme-linked immunosorbent assay and quantitative polymerase chain reaction. Intracellular staining and flow cytometry were used to detect the percentages of Th17 cells and OPN receptors. Signaling and molecular events were analyzed by immunoblotting and chromatin immunoprecipitation. RESULTS: The levels of OPN correlated significantly with interleukin-17 (IL-17) production and the frequency of Th17 cells in the synovial fluid (SF) of RA patients. Endogenous OPN produced in RA SF was responsible for markedly increased production of IL-17 in T cells, which was blocked by OPN antibody. The effect of OPN in Th17 differentiation was mediated through a mechanism independent of the IL-6/STAT-3 pathway or other cytokines and specifically involved the OPN receptors CD44 and CD29 and the transcription factor retinoic acid-related orphan receptor (ROR). Furthermore, OPN was found to induce H3 acetylation of the IL17A gene promoter, mainly through the CD44 binding domain in CD4+ T cells, allowing the interaction of the IL17A gene locus with ROR. CONCLUSION: This study reveals new evidence of the critical role of OPN in Th17 differentiation in rheumatoid synovitis.


Subject(s)
Arthritis, Rheumatoid/physiopathology , Cell Differentiation/physiology , Interleukin-17/metabolism , Osteopontin/physiology , Synovial Fluid/physiology , Synovial Membrane/physiopathology , T-Lymphocyte Subsets/metabolism , Adult , CD4-Positive T-Lymphocytes/metabolism , Case-Control Studies , Cells, Cultured , Female , Humans , Male , Middle Aged
10.
Hum Immunol ; 71(4): 334-41, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20079789

ABSTRACT

IL-21 is a type I cytokine that like IL-2, IL-4, IL-7, IL-9, and IL-15 uses the common gamma chain of cytokine receptor. IL-21 has been shown to regulate the function of T cells, B cells, natural killer cells, and dendritic cells in immune responses. Although activated CD4(+) T cells produce IL-21, recent data suggest that novel subsets of effector T cells are the major producers in immune responses. In this study, we show that IL-21 expression correlates with the presence of Th17 cells in synovial fluid (SF) and peripheral blood in rheumatoid arthritis patients. Human CCR6+ CD4(+) T cells produce high levels of both IL-21 and IL-17. Similar to mouse T cells, IL-21 auto-regulates its own production in human CD4(+) T cells. IL-21 potently enhances Th17 proliferation and suppresses Foxp3 expression, leading to the expression of RORC. IL-21 is therefore an autocrine cytokine that regulates human Th17 cells in rheumatoid arthritis, and serves as a good target for treating this autoimmune disease.


Subject(s)
Arthritis, Rheumatoid/immunology , Interleukins/pharmacology , T-Lymphocyte Subsets/drug effects , T-Lymphocytes/drug effects , Adult , Aged , Animals , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/pathology , CD4 Antigens/biosynthesis , Cell Proliferation/drug effects , Cells, Cultured , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Interleukin-17/metabolism , Interleukins/immunology , Male , Mice , Middle Aged , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Synovial Membrane/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
11.
Nat Med ; 16(2): 191-7, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20062065

ABSTRACT

Interleukin-7 receptor (IL-7R) is genetically associated with susceptibility to multiple sclerosis. Here we describe that IL-7 is essential for survival and expansion of pathogenic T helper type 17 (T(H)17) cells in experimental autoimmune encephalomyelitis (EAE). IL-7 directly expanded effector T(H)17 cells in EAE and human T(H)17 cells from subjects with multiple sclerosis, whereas it was not required for T(H)17 differentiation. IL-7R antagonism rendered differentiated T(H)17 cells susceptible to apoptosis through the inhibition of Janus kinase-signal transducer and activator of transcription-5 (JAK-STAT5) pathway and altered expression of the prosurvival protein Bcl-2 and the proapoptotic protein Bax, leading to decreased severity of EAE. In contrast, T(H)1 and regulatory T (T(reg)) cells were less susceptible to or not affected by IL-7R antagonism in vivo. The selectivity was attributable to minimal expression of IL-7Ralpha in T(reg) cells and correlated with a high level of Socs1 (encoding suppressor of cytokine signaling-1) expression in T(H)1 cells. The study reveals a unique, previously undescribed role of IL-7-IL-7R in T(H)17 cell survival and expansion and has implications in the treatment of autoimmune disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-7/immunology , T-Lymphocytes, Helper-Inducer/cytology , Animals , Cell Differentiation , Janus Kinases/metabolism , Mice , Mice, Transgenic , STAT5 Transcription Factor/metabolism
12.
Cell Res ; 20(4): 480-91, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20084083

ABSTRACT

Alpha-galactosylceramide (alpha-GC) is widely known to activate invariant natural killer T (iNKT) cells to suppress myelin antigen-specific Th1 responses, protecting susceptible mice against experimental autoimmune encephalomyelitis (EAE). Here, we demonstrate an unexpected finding that high doses of alpha-GC exacerbated, rather than ameliorated, EAE. Similar results were observed when MOG(35-55)-specific T cells treated with high-dose alpha-GC were transferred into naïve syngeneic recipient mice. Further study showed that high doses of alpha-GC directly enhance the Th17 and Th1 response by activation of CD4(+)CD44(+) memory T cells through phosphorylation of STAT3 and activation of NF-kappaB. Unlike the activation of iNKT cells by low doses of alpha-GC, high doses of alpha-GC directly interacted with CD1d expressed on T cells and activated Th17 and Th1 cells. Furthermore, antigen-presenting cells (APCs) predominantly express CD1d1, whereas the majority of CD4(+) T cells express CD1d2. Knockdown of CD1d1 or CD1d2 gene expression by RNAi interfered with the activation of iNKT or Th17/Th1 cells, respectively. Therefore, alpha-GC treatment could improve or worsen EAE by engaging either APCs or Th17/Th1 cells depending on the dose used.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Galactosylceramides/toxicity , Interleukin-17/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antigen-Presenting Cells/immunology , Antigens, CD1/metabolism , Antigens, CD1d/genetics , Antigens, CD1d/metabolism , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/metabolism , Gene Knockdown Techniques , Mice , NF-kappa B/metabolism , Natural Killer T-Cells/immunology , RNA Interference , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Helper-Inducer/cytology , Th1 Cells/immunology
13.
Int Immunol ; 21(12): 1329-40, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19822525

ABSTRACT

MBP-specific autoreactive T cells are considered pro-inflammatory T cells and thought to play an important role in the pathogenesis of multiple sclerosis (MS). Here, we report that MBP(83-99)-specific T cells generated from MS patients (n = 7) were comprised of pro-inflammatory and regulatory subsets of distinct phenotypes. The pro-inflammatory phenotype was characterized by high production of IFN-gamma, IL-6, IL-21 and IL-17 and low expression of FOXP3, whereas the regulatory subset expressed high levels of FOXP3 and exhibited potent regulatory functions. The regulatory subset of MBP-specific T cells appeared to expand from the CD4(+)CD25(-) T-cell pool. Their FOXP3 expression was stable, independent of the activation state and it correlated with suppressive function and inversely with the production of IFN-gamma, IL-6, IL-21 and IL-17. In contrast, the phenotype and function of FOXP3(low) MBP-specific T cells were adaptive and dependent on IL-6. The higher frequency of FOXP3(high) MBP-specific T cells was observed when IL-6 was neutralized in the culture of PBMC with MBP. The study provides new evidence that MBP-specific T cells are susceptible to pro-inflammatory cytokine milieu and act as either pro-inflammatory or regulatory T cells.


Subject(s)
Inflammation/immunology , Myelin Basic Protein/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes , Cell Differentiation , Forkhead Transcription Factors/metabolism , Humans , Interleukin-6/immunology , Interleukin-6/metabolism , Lymphocyte Activation , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
14.
J Immunol ; 183(10): 6135-44, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19841165

ABSTRACT

The exact role of adult thymus in autoimmune disease state is poorly understood. We show here that thymus regulated experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis, as evidenced by loss of spontaneous recovery in thymectomized EAE mice. There was progressive enrichment for CD4 single-positive Foxp3(+) regulatory T cells in thymocytes during the course of EAE and they suppressed the disease when adoptively transferred. Thymus was shown to undergo an active process characterized by accelerated differentiation and proliferation of regulatory T (Treg) cells through a mechanism involving increased expression of IL-7 in stromal cells and dynamic expression of IL-7 receptor in thymic Treg cells. This process preceded EAE recovery and selectively affected Treg over non-Treg cells in the thymus, leading to increased output of thymic Treg cells and self-regulation of EAE. The study reveals a novel role of thymus in self-regulation of autoimmune condition.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-7/immunology , Receptors, Interleukin-7/immunology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Adoptive Transfer , Animals , Cell Differentiation/immunology , Cell Proliferation , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Forkhead Transcription Factors/immunology , Glycoproteins/immunology , Interleukin-7/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/immunology , Receptors, Interleukin-7/metabolism , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/metabolism
15.
Eur J Immunol ; 39(9): 2525-36, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19670379

ABSTRACT

IFN-beta currently serves as one of the major treatments for MS. Its anti-inflammatory mechanism has been reported as involving a shift in cytokine balance from Th1 to Th2 in the T-cell response against elements of the myelin sheath. In addition to the Th1 and Th2 groups, two other important pro-inflammatory cytokines, IL-17 and osteopontin (OPN), are believed to play important roles in CNS inflammation in the pathogenesis of MS. In this study, we examined the potential effects of IFN-beta on the regulation of OPN and IL-17 in MS patients. We found that IFN-beta used in vitro at 0.5-3 ng/mL significantly inhibited the production of OPN in primary T cells derived from PBMC. The inhibition of OPN was determined to occur at the CD4(+) T-cell level. In addition, IFN-beta inhibited the production of IL-17 and IL-21 in CD4(+) T cells. It has been described that IFN-beta suppresses IL-17 production through the inhibition of a monocytic cytokine, the intracellular translational isoform of OPN. Our further investigation demonstrated that IFN-beta also acted directly on the CD4(+) T cells to regulate OPN and IL-17 expression through the type I IFN receptor-mediated activation of STAT1 and suppression of STAT3 activity. Administration of IFN-beta to EAE mice ameliorated the disease severity. Furthermore, spinal cord infiltration of OPN(+) and IL-17(+) cells decreased in IFN-beta-treated EAE mice along with decreases in serum levels of OPN and IL-21. Importantly, decreased OPN production by IFN-beta treatment contributes to the reduced migratory activity of T cells. Taken together, the results from both in vitro and in vivo experiments indicate that IFN-beta treatment can down-regulate the OPN and IL-17 production in MS. This study provides new insights into the mechanism of action of IFN-beta in the treatment of MS.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Interferon-beta/pharmacology , Interleukin-17/antagonists & inhibitors , Multiple Sclerosis/immunology , Osteopontin/antagonists & inhibitors , Adult , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Movement/drug effects , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Glycoproteins/pharmacology , Humans , Interleukin-17/biosynthesis , Interleukins/antagonists & inhibitors , Interleukins/blood , Interleukins/metabolism , Male , Mice , Middle Aged , Myelin-Oligodendrocyte Glycoprotein , Osteopontin/biosynthesis , Osteopontin/blood , Peptide Fragments/pharmacology , STAT1 Transcription Factor/agonists , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism
16.
Arthritis Rheum ; 60(7): 1957-65, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19565503

ABSTRACT

OBJECTIVE: Osteopontin (OPN) is a proinflammatory protein with a critical role in leukocyte migration. Although OPN has been implicated in rheumatoid arthritis (RA), its underlying mechanism remains unknown. In this study, we investigated the role and molecular mechanism of OPN in the induction of 2 key chemokines, monocyte chemoattractant protein 1 (MCP-1) and macrophage inflammatory protein 1beta (MIP-1beta), in RA. METHODS: Enzyme-linked immunosorbent assay and quantitative polymerase chain reaction were used to determine chemokine expression. Leukocyte migration in the presence of OPN was measured by chemotaxis assay. Signaling and molecular events were analyzed by immunoblotting and chromatin immunoprecipitation. RESULTS: The effect of OPN on inflammatory cell migration was mediated through its unique property of inducing the expression of MCP-1 and MIP-1beta in CD14+ monocytes. The concentration of OPN was significantly elevated in RA patients and appeared to correlate with the serum levels of inflammation markers and increased expression of MCP-1 or MIP-1beta in monocytes in RA patients. Endogenous production of OPN in RA synovial fluid was attributable to increased production of MCP-1 or MIP-1beta, and this effect could be blocked by an anti-OPN antibody. Furthermore, the structural motif responsible for this property resided within residues 50-83 of human OPN, sparing the known RGD or SVVYGLR sequences. It was evident that the effect of OPN on chemokine expression was mediated through both the NF-kappaB and MAPK pathways, involving the activation of IKKbeta, p38, and JNK. CONCLUSION: These results support a unique role of OPN in leukocyte migration, in the context of perpetuation of rheumatoid synovitis through the induction of MCP-1 and MIP-1beta.


Subject(s)
Arthritis, Rheumatoid/metabolism , Chemokine CCL2/metabolism , Chemokine CCL4/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/metabolism , Osteopontin/metabolism , Signal Transduction/physiology , Adult , Aged , Arthritis, Rheumatoid/pathology , Case-Control Studies , Cell Movement/physiology , Cells, Cultured , Female , Humans , I-kappa B Kinase/metabolism , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Lipopolysaccharide Receptors/metabolism , MAP Kinase Kinase 4/metabolism , Male , Middle Aged , Synovial Fluid/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
17.
J Immunol ; 183(1): 246-53, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19542436

ABSTRACT

Th17 and Th1 play an important role in multiple sclerosis for which copolymer I (COP-I) is a treatment option. We described here that the treatment effect of COP-I correlated with its unique regulatory properties on differentiation and survival of Th17 in experimental autoimmune encephalomyelitis mice, which was mediated through down-regulation of STAT3 phosphorylation. The effect of COP-I on Th17 differentiation required CD14(+) monocytes through IL-6 signaling as a key mediator to regulate STAT3 phosphorylation and subsequent RORgammat expression in Th17 cells. The observed effect was markedly dampened when monocytes were genetically deficient for IL-6. Similar regulatory properties of COP-I were demonstrated in human Th17 differentiation. The study revealed the differential regulatory roles and the novel mechanism of action of COP-I chiefly responsible for its treatment efficacy in experimental autoimmune encephalomyelitis and multiple sclerosis.


Subject(s)
Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-17/physiology , Peptides/administration & dosage , Peptides/pharmacology , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Amino Acid Sequence , Animals , Cell Differentiation/drug effects , Cells, Cultured , Coculture Techniques , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Injections, Subcutaneous , Interleukin-17/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Nuclear Receptor Subfamily 1, Group F, Member 3 , Phosphorylation/drug effects , Phosphorylation/immunology , Receptors, Retinoic Acid/biosynthesis , Receptors, Retinoic Acid/genetics , Receptors, Thyroid Hormone/biosynthesis , Receptors, Thyroid Hormone/genetics , T-Lymphocytes, Helper-Inducer/drug effects
18.
Cell Res ; 19(5): 574-83, 2009 May.
Article in English | MEDLINE | ID: mdl-19188932

ABSTRACT

Glatiramer acetate (GA) is an immunomodulatory peptide drug used to treat multiple sclerosis. Its treatment effect has been expanded to other autoimmune conditions such as uveoretinitis, inflammatory bowel disease, graft rejection and hepatic fibrosis. Here, we report that GA was effective in altering the clinical course of diabetes in cyclophosphamide (CY)-potentiated non-obese diabetic (CY-NOD) mice. Treatment with GA significantly reduced the diabetic rate in the mice and ameliorated insulitis, which coincided with increased CD4+CD25+Foxp3+ T cell response in treated mice. GA treatment led to increased expression of transcription factor Foxp3 and elevated production of interleukin-4 (IL-4) both in vivo and in vitro. It was evident that the effect of GA on up-regulation of Foxp3 was mediated partially through IL-4. IL-4 was found to maintain Foxp3 expression and regulatory function of CD4+CD25+ regulatory T cells (Tregs). This study provides new evidence that GA has treatment potential for type 1 diabetes through the induction of Tregs and that increased IL-4 production is partially responsible for the enhanced Treg's function in GA treatment.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Hypoglycemic Agents/pharmacology , Peptides/pharmacology , T-Lymphocytes, Regulatory/immunology , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/drug therapy , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Glatiramer Acetate , Interleukin-4/metabolism , Mice , Mice, Inbred NOD , T-Lymphocytes, Regulatory/metabolism , Up-Regulation
19.
J Immunol ; 181(11): 7581-92, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19017947

ABSTRACT

Chemokines and chemokine receptors play critical roles in directing the migration of alloreactive donor T cells into graft-vs-host disease (GVHD) target organs. However, blockade of GVHD by antagonist Ab against chemokine receptors remains an elusive goal. Using a mouse model of human GVHD, we demonstrate that in vivo administration of anti-CXCR3 Ab for 21 days (long-term), but not for 7 days (short-term), inhibits alloreactive CD8(+) T cell-mediated GVHD. During a graft-vs-host reaction, infused donor CD8(+) T cells generate two subsets of potent inducers of GVHD: CXCR3(+)CD8(+) and CXCR3(-)CD8(+) T cells. Compared with CXCR3(+)CD8(+) T cells, CXCR3(-)CD8(+) T cells produce less granzyme B, Fas ligand, IFN-gamma, and TNF-alpha. Interestingly, stimulation with either dendritic cells or IL-2 induces a dynamic conversion between CXCR3(+)CD8(+) and CXCR3(-)CD8(+) T cells. Short-term anti-CXCR3 Ab treatment inhibits only CXCR3(+)CD8(+) T cell-mediated GVHD, but not the disease induced by CXCR3(-)CD8(+) T cells. Prolonged in vivo administration of anti-CXCR3 Ab significantly reduces the infiltration of alloreactive CD8(+) T cells into GVHD target organs and inhibits GVHD mediated by either CXCR3(+)CD8(+) or CXCR3(-)CD8(+) T cells. Thus, we have established a novel and effective approach with the potential to give rise to new clinical methods for preventing and treating GVHD after allogeneic hematopoietic stem cell transplantation.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD8-Positive T-Lymphocytes/immunology , Graft vs Host Disease/drug therapy , Receptors, CXCR3/antagonists & inhibitors , Animals , Antibodies, Monoclonal/immunology , Disease Models, Animal , Fas Ligand Protein/immunology , Graft vs Host Disease/immunology , Granzymes/immunology , Hematopoietic Stem Cell Transplantation , Humans , Interferon-gamma/immunology , Mice , Receptors, CXCR3/immunology , Time Factors , Transplantation, Homologous , Tumor Necrosis Factor-alpha/immunology
20.
J Immunol ; 181(2): 1491-8, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18606704

ABSTRACT

Berbamine (BM) is an herbal compound derived from Berberis vulgaris L commonly used in traditional Chinese medicine. In this study, we show that BM has potent anti-inflammatory properties through novel regulatory mechanisms, leading to reduced encephalitogenic T cell responses and amelioration of experimental autoimmune encephalomyelitis (EAE). The treatment effect of BM was attributable to its selective inhibitory effect on the production and action of IFN-gamma in CD4(+) T cells, which was mediated through altered STAT4 expression in T cells. BM was found to up-regulate SLIM, a ubiquitin E3 ligase for STAT4, and promote STAT4 degradation, resulting in markedly decreased IFN-gamma production in CD4(+) T cells in EAE mice. Regulation of IFN-gamma by BM had profound anti-inflammatory actions through its effect on both CD4(+) T cells and APCs. BM-treated APCs exhibited reduced stimulatory function as a result of altered expression of PD-L1, CD80, and CD86 in treated mice. The treatment effect of BM in EAE was directly related to its action on IFN-gamma, and was abolished in IFN-gamma knockout mice. The study also confirmed that BM was able to inhibit NFAT translocation through effecting calcium mobilization in lymphocytes. However, this effect was not directly responsible for the treatment efficacy of BM in EAE. The study has important implications in our approaches to evaluating the utility of natural compounds in drug discovery and to probing the role of cytokine network in the development of autoimmune conditions.


Subject(s)
Benzylisoquinolines/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Interferon-gamma/metabolism , STAT4 Transcription Factor/metabolism , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , B7-2 Antigen/immunology , B7-2 Antigen/metabolism , B7-H1 Antigen , Benzylisoquinolines/chemistry , Benzylisoquinolines/pharmacology , CD4-Positive T-Lymphocytes , Calcium/metabolism , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Down-Regulation , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Interferon-gamma/immunology , LIM Domain Proteins , Male , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Proteins , Myelin-Associated Glycoprotein/immunology , Myelin-Associated Glycoprotein/metabolism , Myelin-Oligodendrocyte Glycoprotein , Peptides/immunology , Peptides/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Ubiquitin-Protein Ligases/immunology , Ubiquitin-Protein Ligases/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...