Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Arthritis Res Ther ; 21(1): 216, 2019 10 23.
Article in English | MEDLINE | ID: mdl-31647025

ABSTRACT

BACKGROUND: The goal of this study is to use comprehensive molecular profiling to characterize clinical response to anti-TNF therapy in a real-world setting and identify reproducible markers differentiating good responders and non-responders in rheumatoid arthritis (RA). METHODS: Whole-blood mRNA, plasma proteins, and glycopeptides were measured in two cohorts of biologic-naïve RA patients (n = 40 and n = 36) from the Corrona CERTAIN (Comparative Effectiveness Registry to study Therapies for Arthritis and Inflammatory coNditions) registry at baseline and after 3 months of anti-TNF treatment. Response to treatment was categorized by EULAR criteria. A cell type-specific data analysis was conducted to evaluate the involvement of the most common immune cell sub-populations. Findings concordant between the two cohorts were further assessed for reproducibility using selected NCBI-GEO datasets and clinical laboratory measurements available in the CERTAIN database. RESULTS: A treatment-related signature suggesting a reduction in neutrophils, independent of the status of response, was indicated by a high level of correlation (ρ = 0.62; p < 0.01) between the two cohorts. A baseline, response signature of increased innate cell types in responders compared to increased adaptive cell types in non-responders was identified in both cohorts. This result was further assessed by applying the cell type-specific analysis to five other publicly available RA datasets. Evaluation of the neutrophil-to-lymphocyte ratio at baseline in the remaining patients (n = 1962) from the CERTAIN database confirmed the observation (odds ratio of good/moderate response = 1.20 [95% CI = 1.03-1.41, p = 0.02]). CONCLUSION: Differences in innate/adaptive immune cell type composition at baseline may be a major contributor to response to anti-TNF treatment within the first 3 months of therapy.


Subject(s)
Adaptive Immunity/physiology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/genetics , Gene Expression Profiling/methods , Immunity, Innate/physiology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adaptive Immunity/drug effects , Adult , Aged , Antirheumatic Agents/pharmacology , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/immunology , Cohort Studies , Female , Humans , Immunity, Innate/drug effects , Male , Middle Aged , Prospective Studies , Treatment Outcome , Tumor Necrosis Factor-alpha/immunology
2.
Article in English | MEDLINE | ID: mdl-29440323

ABSTRACT

Glatiramer acetate (GA) is a synthetic amino acid copolymer that is approved for treatment of relapsing remitting multiple sclerosis (RRMS) and clinically isolated syndrome (CIS). GA reduces multiple sclerosis (MS) disease activity and has shown comparable efficacy with high-dose interferon-ß. The mechanism of action (MOA) of GA has long been an enigma. Originally, it was recognized that GA treatment promoted expansion of GA-reactive T-helper 2 and regulatory T cells, and induced the release of neurotrophic factors. However, GA treatment influences both innate and adaptive immune compartments, and it is now recognized that antigen-presenting cells (APCs) are the initial cellular targets for GA. The anti-inflammatory (M2) APCs induced following treatment with GA are responsible for the induction of anti-inflammatory T cells that contribute to its therapeutic benefit. Here, we review studies that have shaped our current understanding of the MOA of GA.


Subject(s)
Glatiramer Acetate/therapeutic use , Immunosuppressive Agents/therapeutic use , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Animals , Glatiramer Acetate/immunology , Humans , Immunosuppressive Agents/immunology , Mice , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Multiple Sclerosis, Relapsing-Remitting/immunology , Randomized Controlled Trials as Topic
3.
Mol Cell Proteomics ; 18(3): 534-545, 2019 03.
Article in English | MEDLINE | ID: mdl-30559323

ABSTRACT

The importance of IgG glycosylation, Fc-gamma receptor (FcγR) single nucleotide polymorphisms and FcγR copy number variations in fine tuning the immune response has been well established. There is a growing appreciation of the importance of glycosylation of FcγRs in modulating the FcγR-IgG interaction based on the association between the glycosylation of recombinant FcγRs and the kinetics and affinity of the FcγR-IgG interaction. Although glycosylation of recombinant FcγRs has been recently characterized, limited knowledge exists on the glycosylation of endogenous human FcγRs. In order to improve the structural understanding of FcγRs expressed on human cells we characterized the site specific glycosylation of native human FcγRIII from neutrophils of 50 healthy donors and from matched plasma for 43 of these individuals. Through this analysis we have confirmed site specific glycosylation patterns previously reported for soluble FcγRIII from a single donor, identified FcγRIIIb specific Asn45 glycosylation and an allelic effect on glycosylation at Asn162 of FcγRIIIb. Identification of FcγRIIIb specific glycosylation allows for assignment of FcγRIIIb alleles and relative copy number of the two alleles where DNA/RNA is not available. Intriguingly the types of structures found to be elevated at Asn162 in the NA2 allele have been shown to destabilize the Fc:FcγRIII interaction resulting in a faster dissociation rate. These differences in glycosylation may in part explain the differential activity reported for the two alleles which have similar in vitro affinity for IgG.


Subject(s)
Asparagine/chemistry , Receptors, IgG/chemistry , Receptors, IgG/metabolism , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Dosage , Genotype , Glycosylation , Healthy Volunteers , Humans , Immunoglobulin Fc Fragments/metabolism , Mannose/chemistry , Mass Spectrometry , Models, Molecular , Neutrophils/immunology , Plasma/immunology , Receptors, IgG/genetics
4.
Sci Transl Med ; 8(365): 365ra158, 2016 11 16.
Article in English | MEDLINE | ID: mdl-27856797

ABSTRACT

Autoantibody immune complex (IC) activation of Fcγ receptors (FcγRs) is a common pathogenic hallmark of multiple autoimmune diseases. Given that the IC structural features that elicit FcγR activation are poorly understood and the FcγR system is highly complex, few therapeutics can directly block these processes without inadvertently activating the FcγR system. To address these issues, the structure activity relationships of an engineered panel of multivalent Fc constructs were evaluated using sensitive FcγR binding and signaling cellular assays. These studies identified an Fc valency with avid binding to FcγRs but without activation of immune cell effector functions. These observations directed the design of a potent trivalent immunoglobulin G-Fc molecule that broadly inhibited IC-driven processes in a variety of immune cells expressing FcγRs. The Fc trimer, Fc3Y, was highly efficacious in three different animal models of autoimmune diseases. This recombinant molecule may represent an effective therapeutic candidate for FcγR-mediated autoimmune diseases.


Subject(s)
Antigen-Antibody Complex/immunology , Autoimmune Diseases/therapy , Immune Complex Diseases/therapy , Immunoglobulin Fc Fragments/immunology , Receptors, IgG/immunology , Animals , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity/drug effects , Arthritis/immunology , Arthritis/therapy , Arthritis, Experimental/immunology , Arthritis, Experimental/therapy , Autoantibodies/immunology , Autoimmune Diseases/immunology , Cell Line , Epidermolysis Bullosa Acquisita/immunology , Epidermolysis Bullosa Acquisita/therapy , Humans , Immune Complex Diseases/immunology , Immunoglobulin G/immunology , Male , Mice , Mice, Inbred C57BL , Monocytes/cytology , Phagocytes , Platelet Activation , Purpura, Thrombocytopenic, Idiopathic/immunology , Purpura, Thrombocytopenic, Idiopathic/therapy , Signal Transduction
5.
Neurol Neuroimmunol Neuroinflamm ; 2(6): e179, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26601118

ABSTRACT

OBJECTIVE: Glatiramer acetate (GA; Copaxone), a disease-modifying therapy for multiple sclerosis (MS), promotes development of anti-inflammatory (M2, type II) monocytes that can direct differentiation of regulatory T cells. We investigated the innate immune signaling pathways that participate in GA-mediated M2 monocyte polarization. METHODS: Monocytes were isolated from myeloid differentiation primary response gene 88 (MyD88)-deficient, Toll-IL-1 receptor domain-containing adaptor inducing interferon (IFN)-ß (TRIF)-deficient, IFN-α/ß receptor subunit 1 (IFNAR1)-deficient, and wild-type (WT) mice and human peripheral blood. GA-treated monocytes were stimulated with Toll-like receptor ligands, then evaluated for activation of kinases and transcription factors involved in innate immunity, and secretion of proinflammatory cytokines. GA-treated mice were evaluated for cytokine secretion and susceptibility to experimental autoimmune encephalomyelitis. RESULTS: GA-mediated inhibition of proinflammatory cytokine production by monocytes occurred independently of MyD88 and nuclear factor-κB, but was blocked by TRIF deficiency. Furthermore, GA did not provide clinical benefit in TRIF-deficient mice. GA inhibited activation of p38 mitogen-activated protein kinase, an upstream regulator of activating transcription factor (ATF)-2, and c-Jun N-terminal kinase 1, which regulates IFN regulatory factor 3 (IRF3). Consequently, nuclear translocation of ATF-2 and IRF3, components of the IFN-ß enhanceosome, was impaired. Consistent with these observations, GA inhibited production of IFN-ß in vivo in WT mice, but did not modulate proinflammatory cytokine production by monocytes from IFNAR1-deficient mice. CONCLUSION: Our results demonstrate that GA inhibits the type I IFN pathway in M2 polarization of monocytes independently of MyD88, providing an important mechanism connecting innate and adaptive immune modulation in GA therapy and valuable insight regarding its potential use with other MS treatments.

6.
Nat Commun ; 6: 8164, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26353940

ABSTRACT

Autoimmunity and macrophage recruitment into the central nervous system (CNS) are critical determinants of neuroinflammatory diseases. However, the mechanisms that drive immunological responses targeted to the CNS remain largely unknown. Here we show that fibrinogen, a central blood coagulation protein deposited in the CNS after blood-brain barrier disruption, induces encephalitogenic adaptive immune responses and peripheral macrophage recruitment into the CNS leading to demyelination. Fibrinogen stimulates a unique transcriptional signature in CD11b(+) antigen-presenting cells inducing the recruitment and local CNS activation of myelin antigen-specific Th1 cells. Fibrinogen depletion reduces Th1 cells in the multiple sclerosis model, experimental autoimmune encephalomyelitis. Major histocompatibility complex (MHC) II-dependent antigen presentation, CXCL10- and CCL2-mediated recruitment of T cells and macrophages, respectively, are required for fibrinogen-induced encephalomyelitis. Inhibition of the fibrinogen receptor CD11b/CD18 protects from all immune and neuropathologic effects. Our results show that the final product of the coagulation cascade is a key determinant of CNS autoimmunity.


Subject(s)
Autoimmunity/immunology , Brain/immunology , Demyelinating Diseases/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Fibrinogen/immunology , Genes, MHC Class II/immunology , Macrophages/immunology , Spinal Cord/immunology , Th1 Cells/immunology , Adaptive Immunity/drug effects , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Animals , Antigen Presentation/drug effects , Antigen Presentation/genetics , Antigen Presentation/immunology , Autoimmunity/drug effects , Autoimmunity/genetics , Blood-Brain Barrier , Brain/drug effects , Brain/metabolism , Brain/pathology , CD11b Antigen/genetics , CD11b Antigen/immunology , CX3C Chemokine Receptor 1 , Cell Proliferation , Chemokine CCL2/immunology , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Chemokines , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Demyelinating Diseases/genetics , Fibrin , Fibrinogen/pharmacology , Flow Cytometry , Gene Expression Profiling , Genes, MHC Class II/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Immunohistochemistry , Mice , Mice, Knockout , Microglia , Myelin-Oligodendrocyte Glycoprotein/immunology , Rats , Receptors, Antigen, T-Cell/immunology , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology
7.
Neurol Neuroimmunol Neuroinflamm ; 1(2): e22, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25340074

ABSTRACT

OBJECTIVE: Studies evaluating T-cell recognition of myelin oligodendrocyte glycoprotein (MOG) in multiple sclerosis (MS) and its model, experimental autoimmune encephalomyelitis (EAE), have focused mostly on its 117 amino acid (aa) extracellular domain, especially peptide (p) 35-55. We characterized T-cell responses to the entire 218 aa MOG sequence, including its transmembrane and cytoplasmic domains. METHODS: T-cell recognition in mice was examined using overlapping peptides and intact full-length mouse MOG. EAE was evaluated by peptide immunization and by adoptive transfer of MOG epitope-specific T cells. Frequency of epitope-specific T cells was examined by ELISPOT. RESULTS: Three T-cell determinants of MOG were discovered in its transmembrane and cytoplasmic domains, p119-132, p181-195, and p186-200. Transmembrane MOG p119-132 induced clinical EAE, CNS inflammation, and demyelination as potently as p35-55 in C57BL/6 mice and other H-2(b) strains. p119-128 contained its minimal encephalitogenic epitope. p119-132 did not cause disease in EAE-susceptible non-H-2(b) strains, including Biozzi, NOD, and PL/J. MOG p119-132-specific T cells produced Th1 and Th17 cytokines and transferred EAE to wild-type recipient mice. After immunization with full-length MOG, a significantly higher frequency of MOG-reactive T cells responded to p119-132 than to p35-55, demonstrating that p119-132 is an immunodominant encephalitogenic epitope. MOG p181-195 did not cause EAE, and MOG p181-195-specific T cells could not transfer EAE into wild-type or highly susceptible T- and B-cell-deficient mice. CONCLUSIONS: Transmembrane and cytoplasmic domains of MOG contain immunodominant T-cell epitopes in EAE. A CNS autoantigen can also contain nonpathogenic stimulatory T-cell epitopes. Recognition that a myelin antigen contains multiple encephalitogenic and nonencephalitogenic determinants may have implications for therapeutic development in MS.

8.
J Neuroinflammation ; 11: 29, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24498870

ABSTRACT

Oral atorvastatin has prevented or reversed paralysis in the multiple sclerosis (MS) model experimental autoimmune encephalomyelitis (EAE), and reduced development of new MS lesions in clinical trials. Besides inhibiting development of encephalitogenic T cells, atorvastatin treatment of EAE has been associated with an induction of anti-inflammatory myelin-reactive T-helper type (Th)-2 cells. To investigate the clinical significance of atorvastatin-mediated Th2 differentiation, we first evaluated atorvastatin treatment in interleukin (IL)-4 green fluorescent protein-enhanced transcript (4-GET) reporter mice. Atorvastatin treatment failed to induce IL-4-producing Th2 cells in vivo; however, when T cells from atorvastatin-treated 4-GET mice were reactivated in vitro, T cells preferentially differentiated into Th2 cells, while antigen-specific T-cell proliferation and secretion of proinflammatory cytokines (interferon gamma, IL-17, tumor necrosis factor and IL-12) were reduced. Oral atorvastatin also prevented or reversed EAE in signal transducer and activator of transcription 6-deficient (STAT6-/-) mice, which cannot generate IL-4-producing Th2 cells. Further, atorvastatin treatment did not induce or expand Foxp3+ regulatory T cells in either wild-type or STAT6-/- mice. In vivo proliferation of T cells, as measured by incorporation of bromodeoxyuridine, was inhibited in atorvastatin-treated wild-type and STAT6-/- mice. These data imply that atorvastatin ameliorates central nervous system autoimmune disease primarily by inhibiting proliferation of proinflammatory encephalitogenic T cells, and not simply through induction of anti-inflammatory Th2 cells. This cytostatic effect may be a relevant mechanism of action when considering use of statins in MS and other inflammatory conditions.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Forkhead Transcription Factors/metabolism , Heptanoic Acids/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Pyrroles/therapeutic use , T-Lymphocytes, Regulatory/physiology , Th2 Cells/physiology , Adenylyl Cyclases/genetics , Animals , Atorvastatin , Cell Differentiation , Cell Proliferation/drug effects , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Heptanoic Acids/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Interleukin-4/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pyrroles/pharmacology , Rats , STAT6 Transcription Factor/deficiency
9.
J Exp Med ; 210(13): 2921-37, 2013 Dec 16.
Article in English | MEDLINE | ID: mdl-24323356

ABSTRACT

Whether B cells serve as antigen-presenting cells (APCs) for activation of pathogenic T cells in the multiple sclerosis model experimental autoimmune encephalomyelitis (EAE) is unclear. To evaluate their role as APCs, we engineered mice selectively deficient in MHC II on B cells (B-MHC II(-/-)), and to distinguish this function from antibody production, we created transgenic (Tg) mice that express the myelin oligodendrocyte glycoprotein (MOG)-specific B cell receptor (BCR; IgH(MOG-mem)) but cannot secrete antibodies. B-MHC II(-/-) mice were resistant to EAE induced by recombinant human MOG (rhMOG), a T cell- and B cell-dependent autoantigen, and exhibited diminished Th1 and Th17 responses, suggesting a role for B cell APC function. In comparison, selective B cell IL-6 deficiency reduced EAE susceptibility and Th17 responses alone. Administration of MOG-specific antibodies only partially restored EAE susceptibility in B-MHC II(-/-) mice. In the absence of antibodies, IgH(MOG-mem) mice, but not mice expressing a BCR of irrelevant specificity, were fully susceptible to acute rhMOG-induced EAE, also demonstrating the importance of BCR specificity. Spontaneous opticospinal EAE and meningeal follicle-like structures were observed in IgH(MOG-mem) mice crossed with MOG-specific TCR Tg mice. Thus, B cells provide a critical cellular function in pathogenesis of central nervous system autoimmunity independent of their humoral involvement, findings which may be relevant to B cell-targeted therapies.


Subject(s)
Antigen-Presenting Cells/immunology , B-Lymphocytes/immunology , Central Nervous System/immunology , Genes, MHC Class II , Myelin Sheath/immunology , Animals , Cell Proliferation , Cell Separation , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Flow Cytometry , Gene Expression Regulation , Genetic Predisposition to Disease , Immunoglobulins/immunology , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Th1 Cells/immunology , Th17 Cells/immunology
10.
Ann Neurol ; 68(3): 369-83, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20641064

ABSTRACT

OBJECTIVE: Clinical studies indicate that anti-CD20 B-cell depletion may be an effective multiple sclerosis (MS) therapy. We investigated mechanisms of anti-CD20-mediated immune modulation using 2 paradigms of experimental autoimmune encephalomyelitis (EAE). METHODS: Murine EAE was induced by recombinant myelin oligodendrocyte glycoprotein (rMOG), a model in which B cells are considered to contribute pathogenically, or MOG peptide (p)35-55, which does not require B cells. RESULTS: In EAE induced by rMOG, B cells became activated and, when serving as antigen-presenting cells (APCs), promoted differentiation of proinflammatory MOG-specific Th1 and Th17 cells. B-cell depletion prevented or reversed established rMOG-induced EAE, which was associated with less central nervous system (CNS) inflammation, elimination of meningeal B cells, and reduction of MOG-specific Th1 and Th17 cells. In contrast, in MOG p35-55-induced EAE, B cells did not become activated or efficiently polarize proinflammatory MOG-specific T cells, similar to naive B cells. In this setting, anti-CD20 treatment exacerbated EAE, and did not impede development of Th1 or Th17 cells. Irrespective of the EAE model used, B-cell depletion reduced the frequency of CD4(+)CD25(+)Foxp3(+) regulatory T cells (Treg), and increased the proinflammatory polarizing capacity of remaining myeloid APCs. INTERPRETATION: Our study highlights distinct roles for B cells in CNS autoimmunity. Clinical benefit from anti-CD20 treatment may relate to inhibition of proinflammatory B cell APC function. In certain clinical settings, however, elimination of unactivated B cells, which participate in regulation of T cells and other APC, may be undesirable. Differences in immune responses to MOG protein and peptide may be important considerations when choosing an EAE model for testing novel B cell-targeting agents for MS.


Subject(s)
Antibodies/therapeutic use , Antigens, CD20/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Lymphocyte Activation/immunology , Animals , Antigens, CD20/genetics , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Flow Cytometry/methods , Forkhead Transcription Factors/metabolism , Glycoproteins/adverse effects , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/adverse effects , Statistics, Nonparametric , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology
11.
J Immunol ; 183(11): 7259-67, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19915062

ABSTRACT

SWAP-70-like adapter of T cells (SLAT; also known as Def6) is a novel guanine nucleotide exchange factor for Rho GTPases that has been previously shown to play a role in CD4+ T cell activation and Th1/Th2 differentiation. However, the role of SLAT/Def6 in autoimmunity and its associated Th1- and Th17-specific responses has not yet been clearly elucidated. We used a prototypical and pathologically relevant Th1/Th17-mediated autoimmune model, that is, experimental autoimmune encephalomyelitis, to assess the role of SLAT/Def6 in autoantigen-specific T cell response. We found that T cell-expressed SLAT/Def6 was critical for experimental autoimmune encephalomyelitis development and pathogenesis, as evidenced by the resistance of Def6-deficient (Def6(-/-)) mice to clinical signs of the disease associated with a lack of CNS inflammation and demyelination in myelin oligodendrocyte glycoprotein-immunized Def6(-/-) mice. Moreover, Def6 deficiency resulted in a severely diminished myelin oligodendrocyte glycoprotein-specific CD4+ T cell proliferation as well as a defect in IFN-gamma and IL-17 production in secondary lymphoid organs and the CNS. Lastly, Def6(-/-) CD4+ T cells were grossly deficient in their ability to differentiate into Th17 cells both in vitro and in vivo in a T cell-intrinsic manner. Therefore, our study establishes T cell-expressed SLAT/Def6 as a pivotal positive regulator of Th17 inflammatory responses and, thus, essential in controlling autoimmune and inflammatory diseases.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-17/immunology , Nuclear Proteins/immunology , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Guanine Nucleotide Exchange Factors , Interleukin-17/biosynthesis , Mice , Mice, Knockout , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism
13.
Nat Med ; 13(8): 935-43, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17676050

ABSTRACT

Treatment with glatiramer acetate (GA, copolymer-1, Copaxone), a drug approved for multiple sclerosis (MS), in a mouse model promoted development of anti-inflammatory type II monocytes, characterized by increased secretion of interleukin (IL)-10 and transforming growth factor (TGF)-beta, and decreased production of IL-12 and tumor necrosis factor (TNF). This anti-inflammatory cytokine shift was associated with reduced STAT-1 signaling. Type II monocytes directed differentiation of T(H)2 cells and CD4+CD25+FoxP3+ regulatory T cells (T(reg)) independent of antigen specificity. Type II monocyte-induced regulatory T cells specific for a foreign antigen ameliorated experimental autoimmune encephalomyelitis (EAE), indicating that neither GA specificity nor recognition of self-antigen was required for their therapeutic effect. Adoptive transfer of type II monocytes reversed EAE, suppressed T(H)17 cell development and promoted both T(H)2 differentiation and expansion of T(reg) cells in recipient mice. This demonstration of adoptive immunotherapy by type II monocytes identifies a central role for these cells in T cell immune modulation of autoimmunity.


Subject(s)
Monocytes/cytology , Monocytes/immunology , Multiple Sclerosis/immunology , T-Lymphocytes/immunology , Animals , Cell Differentiation/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Glatiramer Acetate , Mice , Mice, Transgenic , Monocytes/classification , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology , Myelin Sheath/metabolism , Peptides/therapeutic use , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
15.
J Neuroimmunol ; 178(1-2): 140-8, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16860400

ABSTRACT

Statins, inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A reductase, are widely prescribed for their cholesterol-lowering properties to reduce atherogenesis and cardiovascular morbidity. Over recent years, statins have also been shown to exert pleiotropic immunomodulatory effects that might be of therapeutic benefit in autoimmune disorders. The primary mechanism by which statins alter immune function appears to be mediated through the inhibition of post-translational protein prenylation of small GTP-binding proteins and is largely independent of lipid-lowering. In experimental autoimmune encephalomyelitis (EAE), the mouse model for multiple sclerosis (MS), statins prevent or reverse paralysis and were recently shown to exert synergistic benefit when combined with agents approved for MS therapy. Based primarily upon the beneficial effects in EAE, statins are now being tested in patients in MS clinical trials.


Subject(s)
Autoimmune Diseases of the Nervous System/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Signal Transduction/immunology , Animals , Antigen Presentation/drug effects , Brain/drug effects , Brain/immunology , Clinical Trials as Topic , Humans , Spinal Cord/drug effects , Spinal Cord/immunology , T-Lymphocytes/drug effects
16.
J Clin Invest ; 116(4): 1037-44, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16543951

ABSTRACT

One approach to improving efficacy in MS therapy is to identify medications that provide additive or synergistic benefit in combination. Orally administered cholesterol-lowering HMG-CoA reductase inhibitors (known as statins), which exhibit immunomodulatory properties and are effective in treatment of the MS model EAE, are being tested in MS. As atorvastatin can enhance protective Th2 responses and has a different mechanism of action than glatiramer acetate (GA), a parenterally administered immunomodulatory agent approved for MS treatment, we tested whether the combination of these agents could be beneficial in EAE. Combination therapy using suboptimal doses of atorvastatin and GA prevented or reversed clinical and histologic EAE. Secretion of proinflammatory Th1 cytokines was reduced--and conversely Th2 cytokine secretion was increased--in these mice, but not in mice treated with each drug alone at the same doses. Monocytes treated with the combination of suboptimal doses of atorvastatin and GA secreted an antiinflammatory type II cytokine pattern and, when used as APCs, promoted Th2 differentiation of naive myelin-specific T cells. Our results demonstrate that agents with different mechanisms of immune modulation can combine in a synergistic manner for the treatment of CNS autoimmunity and provide rationale for testing the combination of atorvastatin and GA in MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Heptanoic Acids/therapeutic use , Immunologic Factors/pharmacology , Peptides/therapeutic use , Pyrroles/therapeutic use , Animals , Atorvastatin , Cells, Cultured , Cytokines/metabolism , Drug Therapy, Combination , Female , Glatiramer Acetate , Heptanoic Acids/pharmacology , Immunologic Factors/therapeutic use , Mice , Mice, Inbred Strains , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Peptides/pharmacology , Pyrroles/pharmacology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology
17.
Trends Immunol ; 27(4): 164-7, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16530483

ABSTRACT

Disturbances in crosstalk between the immune system and the sympathetic nervous system (SNS) can contribute to the pathogenesis of Th1-mediated autoimmunity. Recent studies indicate that neuropeptide Y (NPY) has a major role in the regulation of Th1 responses. The precise role of NPY has been an enigma, but a recent study provides a breakthrough, demonstrating that NPY has a bimodal role as a negative regulator of T cells and an activator of antigen-presenting cell function.


Subject(s)
Inflammation/immunology , Neuropeptide Y/immunology , Neuropeptide Y/metabolism , Animals , Antigen-Presenting Cells/immunology , Inflammation/metabolism , Neurons/immunology , Neurons/metabolism , Neuropeptide Y/deficiency , Neuropeptide Y/genetics , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th1 Cells/metabolism
18.
J Exp Med ; 203(2): 401-12, 2006 Feb 20.
Article in English | MEDLINE | ID: mdl-16476765

ABSTRACT

3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase is a critical enzyme in the mevalonate pathway that regulates the biosynthesis of cholesterol as well as isoprenoids that mediate the membrane association of certain GTPases. Blockade of this enzyme by atorvastatin (AT) inhibits the destructive proinflammatory T helper cell (Th)1 response during experimental autoimmune encephalomyelitis and may be beneficial in the treatment of multiple sclerosis and other Th1-mediated autoimmune diseases. Here we present evidence linking specific isoprenoid intermediates of the mevalonate pathway to signaling pathways that regulate T cell autoimmunity. We demonstrate that the isoprenoid geranylgeranyl-pyrophosphate (GGPP) mediates proliferation, whereas both GGPP and its precursor, farnesyl-PP, regulate the Th1 differentiation of myelin-reactive T cells. Depletion of these isoprenoid intermediates in vivo via oral AT administration hindered these T cell responses by decreasing geranylgeranylated RhoA and farnesylated Ras at the plasma membrane. This was associated with reduced extracellular signal-regulated kinase (ERK) and p38 phosphorylation and DNA binding of their cotarget c-fos in response to T cell receptor activation. Inhibition of ERK and p38 mimicked the effects of AT and induced a Th2 cytokine shift. Thus, by connecting isoprenoid availability to regulation of Th1/Th2 fate, we have elucidated a mechanism by which AT may suppress Th1-mediated central nervous system autoimmune disease.


Subject(s)
Cell Differentiation/drug effects , Encephalomyelitis, Autoimmune, Experimental/immunology , Heptanoic Acids/pharmacology , Pyrroles/pharmacology , Terpenes/pharmacology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Atorvastatin , Cell Differentiation/immunology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Cholesterol/blood , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Female , Heptanoic Acids/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mice , Mice, Inbred Strains , Mice, Transgenic , Protein Prenylation/drug effects , Pyrroles/administration & dosage , Th1 Cells/cytology , Th1 Cells/drug effects , Th2 Cells/cytology , Th2 Cells/drug effects , ras Proteins/antagonists & inhibitors , ras Proteins/metabolism , ras Proteins/physiology , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
19.
Nat Clin Pract Neurol ; 1(2): 106-12, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16932506

ABSTRACT

Statins, a family of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors, are used primarily to reduce atherogenesis and cardiovascular morbidity. Surprisingly, they have also been shown to have immunomodulatory properties that might be of benefit for the treatment of autoimmune disorders. Statins can prevent and even reverse ongoing paralysis in experimental autoimmune encephalomyelitis--the mouse model for multiple sclerosis--and on the basis of these findings, statins are now being tested in patients with multiple sclerosis in clinical trials.


Subject(s)
Central Nervous System/drug effects , Central Nervous System/immunology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , Antigens, Surface/immunology , Antigens, Surface/metabolism , Central Nervous System/physiopathology , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Clinical Trials as Topic/statistics & numerical data , Drug Therapy, Combination , Humans , Multiple Sclerosis/physiopathology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Treatment Outcome
20.
Melanoma Res ; 14(6): 453-61, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15577315

ABSTRACT

In contrast with melanocytes, melanomas display constitutive expression of HLA-DR (HLA-DR+). This abnormal expression has been associated with tumour progression and metastatic dissemination. We have previously reported that this deregulation of HLA-D genes is due to the abnormal constitutive expression of the lymphocyte-specific isoform of class II transactivator (B-CIITA), in addition to its fibroblast form (F-CIITA), which is usually expressed in major histocompatibility complex (MHC) class II-negative interferon-gamma-induced cell types, such as melanocytes. In this study, we investigated the abnormal expression of B-CIITA in a panel of melanoma cell lines displaying differential HLA-DR expression profiles, and analysed whether such a molecular event can participate in tumour progression. Our results showed that the abnormal expression of B-CIITA did not have any particular effect, in comparison with F-CIITA, on the classical activity of CIITA HLA-D gene regulation. As CIITA has also been shown to regulate genes other than HLA-D, we evaluated the modulation of those encoding cyclin D1, YARS (tyrosyl-tRNA synthetase) and TRIP1 (transforming growth factor (TGF)-beta receptor-interacting protein), proteins involved in cell cycle/apoptosis balance, angiogenesis and resistance to TGF-beta, respectively. In contrast with other cell types, neither B-CIITA nor F-CIITA was able to modulate these genes in melanoma cell lines. Thus, the activity of CIITA, whether lymphocyte-specific or fibroblast-specific, is restricted to HLA-D gene expression in these tumours. Accordingly, our data suggest that CIITA is not involved per se in tumour progression; rather, it is the MHC class II molecules themselves, through tumour antigen presentation and the induction of tumour antigen-specific CD4 lymphocyte anergy, that may participate in immune escape and melanoma progression.


Subject(s)
Gene Expression Regulation, Neoplastic , HLA-DR Antigens/metabolism , Melanoma/metabolism , Nuclear Proteins/metabolism , Skin Neoplasms/metabolism , Trans-Activators/metabolism , Animals , B-Lymphocytes/metabolism , COS Cells , Chlorocebus aethiops , Cyclin D1/metabolism , Disease Progression , Eukaryotic Initiation Factor-3 , Fibroblasts/metabolism , Genes, MHC Class II/physiology , Humans , Melanoma/immunology , Nuclear Proteins/genetics , Protein Isoforms , Proteins/metabolism , Skin Neoplasms/immunology , Trans-Activators/genetics , Tumor Cells, Cultured , Tyrosine-tRNA Ligase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...