Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Immunity ; 53(2): 384-397.e5, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32673565

ABSTRACT

Dysregulated Th17 cell responses underlie multiple inflammatory and autoimmune diseases, including autoimmune uveitis and its animal model, EAU. However, clinical trials targeting IL-17A in uveitis were not successful. Here, we report that Th17 cells were regulated by their own signature cytokine, IL-17A. Loss of IL-17A in autopathogenic Th17 cells did not reduce their pathogenicity and instead elevated their expression of the Th17 cytokines GM-CSF and IL-17F. Mechanistic in vitro studies revealed a Th17 cell-intrinsic autocrine loop triggered by binding of IL-17A to its receptor, leading to activation of the transcription factor NF-κB and induction of IL-24, which repressed the Th17 cytokine program. In vivo, IL-24 treatment ameliorated Th17-induced EAU, whereas silencing of IL-24 in Th17 cells enhanced disease. This regulatory pathway also operated in human Th17 cells. Thus, IL-17A limits pathogenicity of Th17 cells by inducing IL-24. These findings may explain the disappointing therapeutic effect of targeting IL-17A in uveitis.


Subject(s)
Cytokines/metabolism , Interleukin-17/metabolism , Th17 Cells/pathology , Uveitis/pathology , Adult , Animals , Cytokines/genetics , Disease Models, Animal , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-17/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Th17 Cells/immunology , Uveitis/immunology , Young Adult
2.
Immunity ; 43(2): 343-53, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26287682

ABSTRACT

Activated retina-specific T cells that have acquired the ability to break through the blood-retinal barrier are thought to be causally involved in autoimmune uveitis, a major cause of human blindness. It is unclear where these autoreactive T cells first become activated, given that their cognate antigens are sequestered within the immune-privileged eye. We demonstrate in a novel mouse model of spontaneous uveitis that activation of retina-specific T cells is dependent on gut commensal microbiota. Retina-specific T cell activation involved signaling through the autoreactive T cell receptor (TCR) in response to non-cognate antigen in the intestine and was independent of the endogenous retinal autoantigen. Our findings not only have implications for the etiology of human uveitis, but also raise the possibility that activation of autoreactive TCRs by commensal microbes might be a more common trigger of autoimmune diseases than is currently appreciated.


Subject(s)
Intestines/immunology , Microbiota/immunology , Retina/immunology , T-Lymphocytes/immunology , Uveitis/immunology , Animals , Antigens, Bacterial/administration & dosage , Autoantigens/immunology , Autoimmunity , Blood-Retinal Barrier/immunology , Cells, Cultured , Disease Models, Animal , Eye Proteins/genetics , Eye Proteins/immunology , Eye Proteins/metabolism , Immune Tolerance , Intestines/microbiology , Lymphocyte Activation , Mice, Inbred Strains , Mice, Knockout , Receptors, Antigen, T-Cell/metabolism , Retinol-Binding Proteins/genetics , Retinol-Binding Proteins/immunology , Retinol-Binding Proteins/metabolism , Uveitis/microbiology
3.
J Autoimmun ; 114: 102507, 2020 11.
Article in English | MEDLINE | ID: mdl-32593472

ABSTRACT

IFN-γ and IL-17A can each elicit ocular autoimmunity independently of the other. Since absence of IFN-γ or IL-17A individually failed to abolish pathology of experimental autoimmune uveitis (EAU), we examined EAU development in the absence of both these cytokines. Ifng-/-Il17a-/- mice were fully susceptible to EAU with a characteristic eosinophilic ocular infiltrate, as opposed to a mononuclear infiltrate in WT mice. Retinal pathology in double-deficient mice was ameliorated when eosinophils were genetically absent or their migration was blocked, supporting a pathogenic role for eosinophils in EAU in the concurrent absence of IFN-γ and IL-17A. In EAU-challenged Ifng-/-Il17a-/- mice, ocular infiltrates contained increased GM-CSF-producing CD4+ T cells, and supernatants of retinal antigen-stimulated splenocytes contained enhanced levels of GM-CSF that contributed to activation and migration of eosinophils in vitro. Systemic or local blockade of GM-CSF ameliorated EAU in Ifng-/-Il17a-/- mice, reduced eosinophil peroxidase levels in the eye and in the serum and decreased eosinophil infiltration to the eye. These results support the interpretation that, in the concurrent absence of IFN-γ and IL-17A, GM-CSF takes on a major role as an inflammatory effector cytokine and drives an eosinophil-dominant pathology. Our findings may impact therapeutic strategies aiming to target IFN-γ and IL-17A in autoimmune uveitis.


Subject(s)
Autoimmunity , Eosinophilia/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interferon-gamma/metabolism , Interleukin-17/metabolism , Retinitis/etiology , Retinitis/metabolism , Animals , Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Disease Models, Animal , Disease Susceptibility/immunology , Eosinophils/immunology , Eosinophils/metabolism , Eosinophils/pathology , Interferon-gamma/genetics , Interleukin-17/genetics , Mice , Mice, Knockout , Retinitis/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
4.
J Autoimmun ; 100: 52-61, 2019 06.
Article in English | MEDLINE | ID: mdl-30853312

ABSTRACT

AS101 is an organotellurium compound with multifaceted immunoregulatory properties that is remarkable for its lack of toxicity. We tested the therapeutic effect of AS101 in experimental autoimmune uveitis (EAU), a model for human autoimmune uveitis. Unexpectedly, treatment with AS101 elicited Treg generation in vivo in otherwise unmanipulated mice. Mice immunized for EAU with the retinal antigen IRBP and treated with AS101 developed attenuated disease, as did AS101-treated recipients of retina-specific T cells activated in vitro. In both settings, eye-infiltrating effector T cells were decreased, whereas regulatory T (Treg) cells in the spleen were increased. Mechanistic studies in vitro revealed that AS101 restricted polarization of retina-specific T cells towards Th1 or Th17 lineage by repressing activation of their respective lineage-specific transcription factors and downstream signals. Retina-specific T cells polarized in vitro towards Th1 or Th17 in the presence of AS101 had impaired ability to induce EAU in naïve recipients. Finally, AS101 promoted differentiation of retina-specific T cells to Tregs in vitro independently of TGF-ß. We conclude that AS101 modulates autoimmune T cells by inhibiting acquisition and expression of effector function and by promoting Treg generation, and suggest that AS101 could be useful as a therapeutic approach for autoimmune uveitis.


Subject(s)
Autoimmune Diseases/drug therapy , Ethylenes/pharmacology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Uveitis/drug therapy , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Disease Models, Animal , Mice , Mice, Transgenic , T-Lymphocytes, Regulatory/pathology , Th1 Cells/pathology , Th17 Cells/pathology , Uveitis/genetics , Uveitis/immunology , Uveitis/pathology
5.
J Autoimmun ; 102: 65-76, 2019 08.
Article in English | MEDLINE | ID: mdl-31080013

ABSTRACT

IL-22 has opposing effects in different tissues, from pro-inflammatory (skin, joints) to protective (liver, intestine) but little is known about its effects on neuroinflammation. We examined the effect of IL-22 on retinal tissue by using the model of experimental autoimmune uveitis (EAU) in IL-22-/- mice, as well as by intraocular injections of recombinant IL-22 or anti-IL-22 antibodies in wild type animals. During EAU, IL-22 was produced in the eye by CD4+ eye-infiltrating T cells. EAU-challenged IL-22-/- mice, as well as WT mice treated systemically or intraocularly with anti-IL-22 antibodies during the expression phase of disease, developed exacerbated retinal damage. Furthermore, IL-22-/- mice were more susceptible than WT controls to glutamate-induced neurotoxicity, whereas local IL-22 supplementation was protective, suggesting direct or indirect neuroprotective effects. Mechanistic studies revealed that retinal glial Müller cells express IL-22rα1 in vivo, and in vitro IL-22 enhanced their ability to suppress proliferation of effector T cells. Finally, IL-22 injected into the eye concurrently with IL-1, inhibited the (IL-1-induced) expression of multiple proinflammatory and proapoptotic genes in retinal tissue. These findings suggest that IL-22 can function locally within the retina to reduce inflammatory damage and provide neuroprotection by affecting multiple molecular and cellular pathways.


Subject(s)
Autoimmunity , Central Nervous System/immunology , Central Nervous System/metabolism , Disease Susceptibility , Interleukins/metabolism , Animals , Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Autoimmunity/genetics , Central Nervous System/pathology , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Ependymoglial Cells/immunology , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Interleukins/genetics , Interleukins/pharmacology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Nervous System Diseases/etiology , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Neuroprotection/genetics , Severity of Illness Index , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Uveitis/etiology , Uveitis/metabolism , Uveitis/pathology , Interleukin-22
6.
J Immunol ; 196(7): 3148-58, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26921309

ABSTRACT

Uveitis, which occurs in association with systemic immunological diseases, presents a considerable medical challenge because of incomplete understanding of its pathogenesis. The signals that initiate T cells to target the eye, which may be of infectious or noninfectious origin, are poorly understood. Experimental autoimmune uveoretinitis (EAU) develops in mice immunized with the endogenous retinal protein interphotoreceptor retinoid binding protein in the presence of the adjuvant CFA. EAU manifests as posterior ocular inflammation consisting of vasculitis, granulomas, retinal damage, and invasion of self-reactive T cells, which are key clinical features of human uveitis. Our studies uncover Card9 as a critical genetic determinant for EAU. Card9 was responsible for Th17 polarization and Th17-associated Ag-specific responses, but not Th1-associated responses. Nonetheless, Card9 expression was essential for accumulation of both lineages within the eye. Consistent with its recently identified role as an intracellular signaling mediator for C-type lectin receptors (CLRs), a Card9-dependent transcriptional response in the neuroretina was observed involving genes encoding the CLRs Dectin-1, Dectin-2, and Mincle. Genetic deletion of these individual CLRs revealed an essential role for Mincle. Mincle activation was sufficient to generate the EAU phenotype, and this required activation of both Syk and Card9. In contrast, Dectin-1 contributed minimally and a possible repressive role was shown for Dectin-2. These findings extend our understanding of CLRs in autoimmune uveitis. The newly identified role of Mincle and Syk/Card9-coupled signaling axis in autoimmune uveitis could provide novel targets for treatment of patients with ocular inflammatory disease.


Subject(s)
Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , CARD Signaling Adaptor Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lectins, C-Type/metabolism , Membrane Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Uveitis/immunology , Uveitis/metabolism , Animals , Autoimmune Diseases/diagnosis , Autoimmune Diseases/genetics , CARD Signaling Adaptor Proteins/genetics , Calcitonin Receptor-Like Protein/genetics , Calcitonin Receptor-Like Protein/metabolism , Disease Models, Animal , Eye Proteins/metabolism , Gene Expression , Gene Expression Profiling , Lectins, C-Type/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout , Retina/immunology , Retina/metabolism , Retina/pathology , Retinol-Binding Proteins/metabolism , Syk Kinase , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Transcriptome , Uveitis/diagnosis , Uveitis/genetics
7.
J Immunol ; 194(7): 3011-9, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25716996

ABSTRACT

Experimental autoimmune uveitis (EAU) induced in mice by immunization with the retinal Ag interphotoreceptor retinoid-binding protein (IRBP) is a model of human autoimmune uveitis. We examined whether T regulatory cells (Tregs) found in uveitic eyes are IRBP specific, functionally suppressive, and play a role in natural resolution of disease and in maintenance of remission. Progressive increase of Foxp3(+) Treg to T effector cell (Teff) ratio in uveitic eyes correlated with resolution of disease. At peak disease, up to 20% of Tregs (CD4(+)Foxp3(+)) and up to 60% of Teffs (CD4(+)Foxp3(-)) were IRBP specific, whereas in lymphoid organs retina-specific T cells were undetectable. Tregs isolated from eyes of mice with EAU efficiently suppressed IRBP-specific responses of Teffs from the same eyes. Importantly, systemic depletion of Tregs at peak disease delayed resolution of EAU, and their depletion after resolution triggered a relapse. This could be partially duplicated by depletion of Tregs locally within the eye. Thus, the T cell infiltrate in uveitic eyes of normal mice with a polyclonal T cell repertoire is highly enriched in IRBP-specific Tregs and Teffs. Unlike what has been reported for Tregs in other inflammatory sites, Tregs from uveitic eyes appear unimpaired functionally. Finally, Foxp3(+) Tregs play a role in the natural resolution of uveitis and in the maintenance of remission, which occurs at least in part through an effect that is local to the eye.


Subject(s)
Autoimmune Diseases/immunology , Retina/immunology , T-Lymphocytes, Regulatory/immunology , Uveitis/immunology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , DNA Methylation , Disease Models, Animal , Eye Proteins/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Immunomodulation , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphocyte Depletion , Male , Mice , Mice, Transgenic , Promoter Regions, Genetic , Recurrence , Retina/pathology , Retinol-Binding Proteins/immunology , T-Cell Antigen Receptor Specificity , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , Uveitis/genetics , Uveitis/pathology
8.
J Autoimmun ; 50: 12-22, 2014 May.
Article in English | MEDLINE | ID: mdl-24021664

ABSTRACT

Central nervous system (CNS) autoimmunity such as uveitis and multiple sclerosis is accompanied by Th1 and Th17 responses. In their corresponding animal models, experimental autoimmune uveitis (EAU) and experimental autoimmune encephalomyelitis (EAE), both responses are induced and can drive disease independently. Because immune responses have inherent plasticity, therapeutic targeting of only one pathway could promote the other, without reducing pathology. IL-27p28 antagonizes gp130, required for signaling by IL-27 and IL-6, which respectively promote Th1 and Th17 responses. We therefore examined its ability to protect the CNS by concurrently targeting both effector responses. Overexpression of IL-27p28 in vivo ameliorated EAU as well as EAE pathology and reduced tissue infiltration by Th1 and Th17 cells in a disease prevention, as well as in a disease reversal protocol. Mechanistic studies revealed inhibition of Th1 and Th17 commitment in vitro and decreased lineage stability of pre-formed effectors in vivo, with reduction in expression of gp130-dependent transcription factors and cytokines. Importantly, IL-27p28 inhibited polarization of human T cells to the Th1 and Th17 effector pathways. The ability of IL-27p28 to inhibit generation as well as function of pathogenic Th1 and Th17 effector cells has therapeutic implications for controlling immunologically complex autoimmune diseases.


Subject(s)
Autoimmunity/drug effects , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukins/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Uveitis/immunology , Animals , Cell Lineage/immunology , Cell Movement/drug effects , Central Nervous System/immunology , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation , Humans , Interleukin-6/genetics , Interleukin-6/immunology , Interleukins/genetics , Interleukins/pharmacology , Mice , Mice, Transgenic , Signal Transduction , Th1 Cells/drug effects , Th1 Cells/pathology , Th17 Cells/drug effects , Th17 Cells/pathology , Transcription Factors/genetics , Transcription Factors/immunology , Uveitis/genetics , Uveitis/pathology
9.
J Immunol ; 188(4): 1742-50, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22238462

ABSTRACT

Immune privilege is used by the eye, brain, reproductive organs, and gut to preserve structural and functional integrity in the face of inflammation. The eye is arguably the most vulnerable and, therefore, also the most "privileged" of tissues; paradoxically, it remains subject to destructive autoimmunity. It has been proposed, although never proven in vivo, that the eye can induce T regulatory cells (Tregs) locally. Using Foxp3-GFP reporter mice expressing a retina-specific TCR, we now show that uncommitted T cells rapidly convert in the living eye to Foxp3(+) Tregs in a process involving retinal Ag recognition, de novo Foxp3 induction, and proliferation. This takes place within the ocular tissue and is supported by retinoic acid, which is normally present in the eye because of its function in the chemistry of vision. Nonconverted T cells showed evidence of priming but appeared restricted from expressing effector function in the eye. Pre-existing ocular inflammation impeded conversion of uncommitted T cells into Tregs. Importantly, retina-specific T cells primed in vivo before introduction into the eye were resistant to Treg conversion in the ocular environment and, instead, caused severe uveitis. Thus, uncommitted T cells can be disarmed, but immune privilege is unable to protect from uveitogenic T cells that have acquired effector function prior to entering the eye. These findings shed new light on the phenomenon of immune privilege and on its role, as well as its limitations, in actively controlling immune responses in the tissue.


Subject(s)
Autoimmunity , Eye/immunology , Forkhead Transcription Factors/analysis , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes/immunology , Tretinoin/metabolism , Uveitis/immunology , Animals , Cell Differentiation , Cell Proliferation , Eye Proteins/immunology , Forkhead Transcription Factors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/immunology , Retinol-Binding Proteins/immunology
10.
J Autoimmun ; 44: 21-33, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23810578

ABSTRACT

Despite presence of circulating retina-specific T cells in healthy individuals, ocular immune privilege usually averts development of autoimmune uveitis. To study the breakdown of immune privilege and development of disease, we generated transgenic (Tg) mice that express a T cell receptor (TCR) specific for interphotoreceptor retinoid-binding protein (IRBP), which serves as an autoimmune target in uveitis induced by immunization. Three lines of TCR Tg mice, with different levels of expression of the transgenic R161 TCR and different proportions of IRBP-specific CD4⁺ T cells in their peripheral repertoire, were successfully established. Importantly, two of the lines rapidly developed spontaneous uveitis, reaching 100% incidence by 2 and 3 months of age, respectively, whereas the third appeared "poised" and only developed appreciable disease upon immune perturbation. Susceptibility roughly paralleled expression of the R161 TCR. In all three lines, peripheral CD4⁺ T cells displayed a naïve phenotype, but proliferated in vitro in response to IRBP and elicited uveitis upon adoptive transfer. In contrast, CD4⁺ T cells infiltrating uveitic eyes mostly showed an effector/memory phenotype, and included Th1, Th17 as well as T regulatory cells that appeared to have been peripherally converted from conventional CD4⁺ T cells rather than thymically derived. Thus, R161 mice provide a new and valuable model of spontaneous autoimmune disease that circumvents the limitations of active immunization and adjuvants, and allows to study basic mechanisms involved in maintenance and breakdown of immune homeostasis affecting immunologically privileged sites such as the eye.


Subject(s)
Autoantigens/immunology , Autoimmunity/immunology , Receptors, Antigen, T-Cell/immunology , Retina/immunology , Animals , Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Cytokines/immunology , Eye Proteins/immunology , Humans , Immunologic Memory/immunology , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell/biosynthesis , Retinol-Binding Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Uveitis/immunology
11.
J Immunol ; 187(4): 1977-85, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21765017

ABSTRACT

Noninfectious uveitis is a leading cause of blindness and thought to involve autoimmune T cell responses to retinal proteins (e.g., retinal arrestin [soluble-Ag (S-Ag)]). There are no known biomarkers for the disease. Susceptibility is associated with HLA, but little is known about susceptible class II alleles or the potentially pathogenic epitopes that they present. Using a humanized HLA-transgenic mouse model of S-Ag-induced autoimmune uveitis, we identified several susceptible and resistant alleles of HLA-DR and -DQ genes and defined pathogenic epitopes of S-Ag presented by the susceptible alleles. The sequences of these epitopes overlap with some previously identified peptides of S-Ag ("M" and "N"), known to elicit memory responses in lymphocytes of uveitis patients. HLA-DR-restricted, S-Ag-specific CD4(+) T cells could be detected in blood and draining lymph nodes of uveitic mice with HLA class II tetramers and transferred the disease to healthy mice. Importantly, tetramer-positive cells were detected in peripheral blood of a uveitis patient. To our knowledge, these findings provide the first tangible evidence that an autoimmune response to retina is causally involved in pathogenesis of human uveitis, demonstrating the feasibility of identifying and isolating retinal Ag-specific T cells from uveitis patients and may facilitate their development as biomarkers for the disease.


Subject(s)
Autoantigens/immunology , Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Eye Proteins/immunology , HLA-DQ Antigens/immunology , HLA-DR Antigens/immunology , Retina/immunology , Uveitis/immunology , Alleles , Animals , Autoantigens/genetics , Autoimmune Diseases/genetics , Biomarkers , CD4-Positive T-Lymphocytes/pathology , Disease Models, Animal , Epitopes, T-Lymphocyte/genetics , Eye Proteins/genetics , Genetic Predisposition to Disease , HLA-DQ Antigens/genetics , HLA-DR Antigens/genetics , Humans , Immunologic Memory/genetics , Immunologic Memory/immunology , Mice , Mice, Transgenic , Retina/pathology , Uveitis/genetics , Uveitis/pathology
12.
J Exp Med ; 203(4): 851-6, 2006 Apr 17.
Article in English | MEDLINE | ID: mdl-16585264

ABSTRACT

Susceptibility to experimental autoimmune uveitis (EAU), a model for human uveitis induced in mice with the retinal antigen interphotoreceptor retinoid-binding protein (IRBP), is controlled by "natural" CD4+CD25+ regulatory T (T reg) cells. To examine whether endogenous expression of IRBP is necessary to generate these T reg cells, we studied responses of IRBP knockout (KO) versus wild-type (WT) mice. Unexpectedly, not only WT but also IRBP KO mice immunized with a uveitogenic regimen of IRBP in complete Freund's adjuvant (CFA) exhibited CD25+ regulatory cells that could be depleted by PC61 treatment, which suppressed development of uveitogenic effector T cells and decreased immunological responses to IRBP. These EAU-relevant T reg cells were not IRBP specific, as their activity was not present in IRBP KO mice immunized with IRBP in incomplete Freund's adjuvant (IFA), lacking mycobacteria (whereas the same mice exhibited normal T reg cell activity to retinal arrestin in IFA). We propose that mycobacterial components in CFA activate T reg cells of other specificities to inhibit generation of IRBP-specific effector T cells in a bystander fashion, indicating that effective T reg cells can be antigen nonspecific. Our data also provide the first evidence that generation of specific T reg cells to a native autoantigen in a mouse with a diverse T cell repertoire requires a cognate interaction.


Subject(s)
Autoimmune Diseases/prevention & control , Cell Differentiation/immunology , Eye Proteins/physiology , Retina/immunology , Retinol-Binding Proteins/physiology , T-Lymphocytes, Regulatory/immunology , Uveitis/prevention & control , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , CD4 Antigens/biosynthesis , Cattle , Eye Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-2/biosynthesis , Receptors, Interleukin-2/deficiency , Retina/pathology , Retinol-Binding Proteins/deficiency , Retinol-Binding Proteins/genetics , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Uveitis/genetics , Uveitis/immunology
14.
J Immunol ; 182(8): 4624-32, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19342637

ABSTRACT

Experimental autoimmune uveitis (EAU) serves as a model for human autoimmune uveitis and for cell-mediated autoimmunity in general. EAU induced in mice by immunization with the retinal Ag interphotoreceptor retinoid-binding protein in CFA is driven by the Th17 response. Oral calcitriol (1,25-dihydroxyvitamin D(3)) prevented as well as partly reversed disease and suppressed immunological responses. In vitro, calcitriol directly suppressed IL-17 induction in purified naive CD4(+) T cells without inhibiting Th17 lineage commitment, as reflected by unaltered RORgammat, STAT3, and FoxP3 expression. In contrast, in vivo treatment with calcitriol of mice challenged for EAU impaired commitment to the Th17 lineage, as judged by reduction of both RORgammat and IL-17 in CD4(+) T cells. Innate immune response parameters in draining lymph nodes of treated mice were suppressed, as was production of IL-1, IL-6, TNF-alpha, and IL-12/IL-23p40, but not IL-10, by explanted splenic dendritic cells (DC). Finally, supernatants of calcitriol-conditioned bone marrow-derived DC had reduced ability to support Th17 polarization of naive CD4(+) T cells in vitro and in vivo. Thus, calcitriol appears to suppress autoimmunity by inhibiting the Th17 response at several levels, including the ability of DC to support priming of Th17 cells, the ability of CD4(+) T cells to commit to the Th17 lineage, and the ability of committed Th17 T cells to produce IL-17.


Subject(s)
Autoimmunity/drug effects , Autoimmunity/immunology , Calcitriol/pharmacology , Interleukin-17/immunology , Retina/immunology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , Administration, Oral , Animals , Calcitriol/administration & dosage , Cell Lineage/drug effects , Cell Lineage/immunology , Cells, Cultured , Female , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mice , Mice, Knockout , Retina/drug effects , T-Lymphocytes, Helper-Inducer/cytology , Uveitis/immunology
15.
J Exp Med ; 198(11): 1665-76, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14657219

ABSTRACT

Immunologically privileged retinal antigens can serve as targets of experimental autoimmune uveitis (EAU), a model for human uveitis. The tolerance status of susceptible strains, whose target antigen is not expressed in the thymus at detectable levels, is unclear. Here, we address this issue directly by analyzing the consequences of genetic deficiency versus sufficiency of a uveitogenic retinal antigen, interphotoreceptor retinoid-binding protein (IRBP). IRBP-knockout (KO) and wild-type (WT) mice on a highly EAU-susceptible background were challenged with IRBP. The KO mice had greatly elevated responses to IRBP, an altered recognition of IRBP epitopes, and their primed T cells induced exacerbated disease in WT recipients. Ultrasensitive immunohistochemical staining visualized sparse IRBP-positive cells, undetectable by conventional assays, in thymi of WT (but not of KO) mice. IRBP message was PCR amplified from these cells after microdissection. Thymus transplantation between KO and WT hosts demonstrated that this level of expression is functionally relevant and sets the threshold of immune (and autoimmune) reactivity. Namely, KO recipients of WT thymi generated reduced IRBP-specific responses, and WT recipients of KO thymi developed enhanced responses and a highly exacerbated disease. Repertoire culling and thymus-dependent CD25+ T cells were implicated in this effect. Thus, uveitis-susceptible individuals display a detectable and functionally significant tolerance to their target antigen, in which central mechanisms play a prominent role.


Subject(s)
Antigens/immunology , Eye Proteins , Immune Tolerance/immunology , Retina/immunology , Animals , Autoimmune Diseases/immunology , Mice , Mice, Knockout , Retinal Diseases/immunology , Retinol-Binding Proteins/genetics , Retinol-Binding Proteins/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology
16.
J Immunol ; 181(7): 4791-7, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18802082

ABSTRACT

Invariant NKT cells (iNKT cells) have been reported to play a role not only in innate immunity but also to regulate several models of autoimmunity. Furthermore, iNKT cells are necessary for the generation of the prototypic eye-related immune regulatory phenomenon, anterior chamber associated immune deviation (ACAID). In this study, we explore the role of iNKT cells in regulation of autoimmunity to retina, using a model of experimental autoimmune uveitis (EAU) in mice immunized with a uveitogenic regimen of the retinal Ag, interphotoreceptor retinoid-binding protein. Natural strain-specific variation in iNKT number or induced genetic deficiencies in iNKT did not alter baseline susceptibility to EAU. However, iNKT function seemed to correlate with susceptibility and its pharmacological enhancement in vivo by treatment with iNKT TCR ligands at the time of uveitogenic immunization reproducibly ameliorated disease scores. Use of different iNKT TCR ligands revealed dependence on the elicited cytokine profile. Surprisingly, superior protection against EAU was achieved with alpha-C-GalCer, which induces a strong IFN-gamma but only a weak IL-4 production by iNKT cells, in contrast to the ligands alpha-GalCer (both IFN-gamma and IL-4) and OCH (primarily IL-4). The protective effect of alpha-C-GalCer was associated with a reduction of adaptive Ag-specific IFN-gamma and IL-17 production and was negated by systemic neutralization of IFN-gamma. These data suggest that pharmacological activation of iNKT cells protects from EAU at least in part by a mechanism involving innate production of IFN-gamma and a consequent dampening of the Th1 as well as the Th17 effector responses.


Subject(s)
Autoimmune Diseases/therapy , Interferon-gamma/biosynthesis , Interleukin-17/physiology , Lymphocyte Activation/immunology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Th1 Cells/immunology , Uveitis/therapy , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/prevention & control , Cattle , Disease Susceptibility/immunology , Eye Proteins/administration & dosage , Eye Proteins/immunology , Immunity, Innate , Interferon-gamma/metabolism , Interferon-gamma/physiology , Interleukin-17/antagonists & inhibitors , Interleukin-4/metabolism , Ligands , Mice , Mice, Inbred AKR , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Retinol-Binding Proteins/administration & dosage , Retinol-Binding Proteins/immunology , Species Specificity , Th1 Cells/metabolism , Uveitis/immunology , Uveitis/prevention & control
17.
J Leukoc Biol ; 84(2): 577-85, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18495789

ABSTRACT

We identified inhibitory peptide analogs (IPAs), capable of immunomodulating experimental autoimmune uveitis (EAU), induced in B10.RIII mice by immunization with the retinal antigen interphotoreceptor-binding protein in CFA. Alanine-substituted peptides of the major pathogenic epitope, residues 161-180, were synthesized. They were tested for immunogenicity, cross-reactivity with the native 161-180 epitope, pathogenicity, and ability to prevent EAU when given in IFA before EAU challenge with native murine (m)161-180. Two peptides, 169A and 171A, were unable to elicit disease but cross-reacted with m161-180 by lymphocyte proliferation. Mice pretreated with either of the substituted peptides failed to develop EAU after challenge with the native epitope, m161-180, and had reduced cellular responses by lymphocyte proliferation and by delayed hypersensitivity. Their cytokine response profile to m161-180 showed reduced antigen-specific IFN-gamma and IL-17, whereas IL-4, IL-5, IL-10, and IL-13 from IPA-protected mice were increased, and serum antibody titers to m161-180 revealed reduced IgG2a and elevated IgG1 isotypes, suggesting a Th2 shift in the response. Protection was transferable with lymphoid cells from protected donors to naïve recipients, who were subsequently immunized for EAU. Thus, IPA pretreatment prevents induction of EAU by skewing the response to a subsequent uveitogenic challenge with the native peptide to a nonpathogenic phenotype, as well as by eliciting transferable regulatory cells.


Subject(s)
Autoimmune Diseases/drug therapy , Peptides/therapeutic use , Retina/immunology , T-Lymphocytes, Regulatory/immunology , Uveitis/drug therapy , Uveitis/immunology , Adoptive Transfer , Amino Acid Sequence , Animals , Autoimmunity , Cytokines/metabolism , Immune Tolerance , Lymphocyte Activation , Mice , Models, Animal , Molecular Sequence Data , Peptides/chemistry , Structure-Activity Relationship
18.
Invest Ophthalmol Vis Sci ; 49(5): 1946-56, 2008 May.
Article in English | MEDLINE | ID: mdl-18436827

ABSTRACT

PURPOSE: Interphotoreceptor retinoid binding protein (IRBP) is the major uveitogenic retinal antigen eliciting experimental autoimmune uveoretinitis (EAU) in mice. The most frequently used mouse strains are B10.RIII and C57BL/6, but to date only one uveitogenic epitope for each has been identified. The purpose of this study was to identify and characterize additional uveitogenic epitopes in B10.RIII and C57BL/6 mice and to compare epitope recognition in wild-type versus IRBP-deficient mice on both backgrounds. METHODS: Mice were immunized with IRBP. Spleen cells were stimulated in culture with overlapping peptides representing the entire IRBP molecule, and lymphocyte proliferative responses were measured. Peptides determined to be immunodominant were used to immunize mice for EAU. Cytokine profile and proliferation of the CD4 versus CD8 subsets were analyzed for the most pathogenic peptides. RESULTS: Two new major pathogenic epitopes were identified in WT C57BL/6 mice, residues 461-480 and 651-670. These epitopes induced EAU of severity similar to that induced by the previously known peptide, 1-20. Several other peptides elicited mild disease with lower incidence. Some peptides elicited EAU only in WT recipients of IRBP KO splenocytes. In the B10.RIII strain, two major new uveitogenic peptides were identified, 171-190 and 541-560, and several others elicited moderate disease. Unlike in C57BL/6 mice, adoptive transfer of WT B10.RIII with IRBP KO splenocytes did not reveal additional uveitogenic epitopes. Both CD4 and CD8 lymphocyte subsets proliferated to pathogenic peptides. CONCLUSIONS: Several new pathogenic peptides of IRBP were identified in C57BL/6 and B10.RIII mice. Differences in epitope recognition between WT and IRBP KO mice were observed in C57BL/6 mice, but not in B10.RIII mice, suggesting more extensive culling of the repertoire in C57BL/6 mice by endogenously expressed IRBP.


Subject(s)
Autoimmune Diseases/immunology , Eye Proteins/immunology , H-2 Antigens/immunology , Immunodominant Epitopes/immunology , Peptide Fragments/immunology , Retinitis/immunology , Retinol-Binding Proteins/immunology , Uveitis/immunology , Amino Acid Sequence , Animals , Autoimmune Diseases/pathology , CD4-CD8 Ratio , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Female , Haplotypes , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Retinitis/pathology , Uveitis/pathology
19.
Ophthalmic Res ; 40(3-4): 169-74, 2008.
Article in English | MEDLINE | ID: mdl-18421234

ABSTRACT

The mouse model of experimental autoimmune uveitis, induced by immunization of mice with the retinal protein IRBP, was developed in our laboratory 20 years ago and published in 1988. Since that time it has been adopted by many investigators and has given rise to many studies that helped elucidate genetic influences, dissect the basic mechanisms of pathogenesis and test novel immunotherapeutic paradigms. The current overview will summarize the salient features of the experimental autoimmune uveitis model and discuss its mechanisms.


Subject(s)
Autoimmune Diseases/immunology , Disease Models, Animal , Uveitis/immunology , Animals , Autoantigens/immunology , Autoimmune Diseases/genetics , Autoimmunity/genetics , Epitopes/immunology , Genetic Predisposition to Disease , Haplotypes , Major Histocompatibility Complex/genetics , Major Histocompatibility Complex/immunology , Mice , Mice, Inbred Strains , Retina/immunology , T-Lymphocytes/immunology , Uveitis/genetics
20.
J Clin Invest ; 111(8): 1171-80, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12697736

ABSTRACT

Experimental autoimmune uveitis (EAU) is a disease of the neural retina induced by immunization with retinal antigens, such as interphotoreceptor retinoid-binding protein (IRBP) and arrestin (retinal soluble antigen, S-Ag). EAU serves as a model for human autoimmune uveitic diseases associated with major histocompatibility complex (HLA) genes, in which patients exhibit immunological responses to retinal antigens. Here we report the development of a humanized EAU model in HLA transgenic (TG) mice. HLA-DR3, -DR4, -DQ6, and -DQ8 TG mice were susceptible to IRBP-induced EAU. Importantly, HLA-DR3 TG mice developed severe EAU with S-Ag, to which wild-type mice are highly resistant. Lymphocyte proliferation was blocked by anti-HLA antibodies, confirming that antigen is functionally presented by the human MHC molecules. Disease could be transferred by immune cells with a Th1-like cytokine profile. Antigen-specific T cell repertoire, as manifested by responses to overlapping peptides derived from S-Ag or IRBP, differed from that of wild-type mice. Interestingly, DR3 TG mice, but not wild-type mice, recognized an immunodominant S-Ag epitope between residues 291 and 310 that overlaps with a region of S-Ag recognized by uveitis patients. Thus, EAU in HLA TG mice offers a new model of uveitis that should represent human disease more faithfully than currently existing models.


Subject(s)
Autoimmune Diseases/etiology , Eye Proteins , Histocompatibility Antigens Class II/physiology , Uveitis/etiology , Amino Acid Sequence , Animals , Antigen Presentation , Arrestin/immunology , Disease Models, Animal , Epitopes, T-Lymphocyte , HLA-DR3 Antigen/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Retinol-Binding Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL