Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
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.
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
3.
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
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 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
6.
Bio Protoc ; 6(18)2016 Sep 20.
Article in English | MEDLINE | ID: mdl-28191487

ABSTRACT

The contribution of microbiota in regulating multiple physiological and pathological host responses has been studied intensively in recent years. Evidence suggests that commensal microbiota can directly modulate different populations of cells of the immune system (e.g., Ivanov et al., 2008; Atarashi et al., 2011). Recently, we showed that protein extracts from gut commensal microbiota can activate retina-specific T cells, allowing these autoreactive T cells to then break through the blood-retinal barrier and trigger autoimmune uveitis in the recipient (Horai et al., 2015). The protocol below describes the method to prepare intestinal protein-rich extracts that can be used in various in vitro andin vivo immunological studies.

7.
J Exp Med ; 212(10): 1739-52, 2015 Sep 21.
Article in English | MEDLINE | ID: mdl-26347474

ABSTRACT

IFN-γ is a pathogenic cytokine involved in inflammation. Paradoxically, its deficiency exacerbates experimental autoimmune encephalomyelitis, uveitis, and arthritis. Here, we demonstrate using IFN-γ(-/-) mice repleted with IFN-γ +/+: NK cells that innate production of IFN-γ from NK cells is necessary and sufficient to trigger an endogenous regulatory circuit that limits autoimmunity. After immunization, DCs recruited IFN-γ-producing NK cells to the draining lymph node and interacted with them in a CXCR3-dependent fashion. The interaction caused DCs to produce IL-27, which in turn enhanced IFN-γ production by NK cells, forming a self-amplifying positive feedback loop. IL-10, produced by the interacting cells themselves, was able to limit this process. The NK-DC-dependent IL-27 inhibited development of the adaptive pathogenic IL-17 response and induced IL-10-producing Tr1-like cells, which ameliorated disease in an IL-10-dependent manner. Our data reveal that an early NK-DC interaction controls the adaptive Th17 response and limits tissue-specific autoimmunity through an innate IFN-γ-IL-27 axis.


Subject(s)
Dendritic Cells/immunology , Interferon-gamma/immunology , Interleukins/immunology , Killer Cells, Natural/immunology , Th17 Cells/immunology , Uveitis/immunology , Animals , Autoimmune Diseases/immunology , Feedback, Physiological , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukins/metabolism , Killer Cells, Natural/metabolism , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Receptors, CXCR3/metabolism , Uveitis/pathology
8.
Invest Ophthalmol Vis Sci ; 56(9): 5439-49, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26284549

ABSTRACT

PURPOSE: Experimental autoimmune uveitis (EAU) induced in mice using the retinal antigen interphotoreceptor retinoid binding protein (IRBP) is an animal model for posterior uveitis in humans. However, EAU induced by native IRBP protein or its widely used epitope amino acid residues 1 to 20 of human IRBP (hIRBP1-20) is inconsistent, often showing low scores and incidence. We found an urgent need to identify a better pathogenic epitope for the C57BL/6 strain. METHODS: Mice were immunized with uveitogenic peptides or with native bovine IRBP. Clinical and histological disease and associated immunological responses were evaluated. Truncated and substituted peptides, as well as bioinformatic analyses, were used to identify critical major histocompatibility complex (MHC)/T cell receptor (TCR) contact residues and the minimal core epitope. RESULTS: The new uveitogenic epitope of IRBP, amino acid residues 651 to 670 of human IRBP (LAQGAYRTAVDLESLASQLT [hIRBP651-670]) is uveitogenic for mice of the H-2b haplotype and elicits EAU with a higher severity and incidence in C57BL/6 mice than the previously characterized hIRBP1-20 epitope. Using truncated and substituted peptides, as well as bioinformatic analysis, we identified the critical contact residues with MHC/TCR and defined the minimal core epitope. This made it possible to design MHC tetramers and use them to detect epitope-specific T cells in the uveitic eye and in lymphoid organs of hIRBP651-670-immunized mice. CONCLUSIONS: Data suggest that hIRBP651-670 is an epitope naturally processed from a conserved region of native IRBP, potentially explaining its relatively high uveitogenicity. This epitope should be useful for basic and preclinical studies of uveitis in the C57BL/6 model and gives access to genetically engineered mice available on this background.


Subject(s)
Autoimmune Diseases/immunology , Eye Proteins/immunology , Histocompatibility Antigens Class II/immunology , Immunity, Cellular , Retinitis/immunology , Retinol-Binding Proteins/immunology , T-Lymphocytes/immunology , Uveitis, Posterior/immunology , Animals , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Cattle , Cells, Cultured , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Eye Proteins/metabolism , Haplotypes , Histocompatibility Antigens Class II/metabolism , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Retinitis/metabolism , Retinitis/pathology , Retinol-Binding Proteins/metabolism , Severity of Illness Index , Uveitis, Posterior/metabolism , Uveitis, Posterior/pathology
9.
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
11.
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
12.
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
13.
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
14.
Methods Mol Biol ; 900: 443-69, 2012.
Article in English | MEDLINE | ID: mdl-22933083

ABSTRACT

The model of experimental autoimmune uveitis (EAU) in mice and in rats is described. EAU targets immunologically privileged retinal antigens and serves as a model of autoimmune uveitis in humans as well as a model for autoimmunity in a more general sense. EAU is a well-characterized, robust, and reproducible model that is easily followed and quantitated. It is inducible with synthetic peptides derived from retinal autoantigens in commonly available strains of rats and mice. The ability to induce EAU in various gene-manipulated, including HLA-transgenic, mouse strains makes the EAU model suitable for the study of basic mechanisms as well as in clinically relevant interventions.


Subject(s)
Autoimmune Diseases/pathology , Disease Models, Animal , Uveitis/pathology , Amino Acid Sequence , Animals , Disease Progression , Disease Susceptibility , Humans , Mice , Molecular Sequence Data , Peptides/chemistry , Rats
15.
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
16.
Int J Mol Med ; 28(5): 727-31, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21887476

ABSTRACT

Uveitis is an inflammation of the middle layer of the eye with a high risk of blindness. The Gi protein associated A3 adenosine receptor (A3AR) is highly expressed in inflammatory cells whereas low expression is found in normal cells. CF101 is a highly specific agonist at the A3AR known to induce a robust anti-inflammatory effect in different experimental animal models. The CF101 mechanism of action entails down-regulation of the NF-κB-TNF-α signaling pathway, resulting in inhibition of pro-inflammatory cytokine production and apoptosis of inflammatory cells. In this study the effect of CF101 on the development of retinal antigen interphotoreceptor retinoid-binding protein (IRBP)-induced experimental autoimmune uveitis (EAU) was assessed. Oral treatment with CF101 (10 µg/kg, twice daily), initiated upon disease onset, improved uveitis clinical score measured by fundoscopy and ameliorated the pathological manifestations of the disease. Shortly after treatment with CF101 A3AR expression levels were down-regulated in the lymph node and spleen cells pointing towards receptor activation. Downstream events included a decrease in PI3K and STAT-1 and proliferation inhibition of IRPB auto-reactive T cells ex vivo. Inhibition of interleukin-2, tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) production and up-regulation of interleukin-10 was found in cultured splenocytes derived from CF101-treated animals. Overall, the present study data point towards a marked anti-inflammatory effect of CF101 in EAU and support further exploration of this small molecule drug for the treatment of uveitis.


Subject(s)
Adenosine A3 Receptor Agonists/therapeutic use , Adenosine/analogs & derivatives , Uveitis/drug therapy , Uveitis/immunology , Adenosine/therapeutic use , Animals , Cell Proliferation/drug effects , Cells, Cultured , Interleukin-10/metabolism , Interleukin-2/metabolism , Lymph Nodes/metabolism , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Receptor, Adenosine A3/metabolism , STAT1 Transcription Factor/metabolism , Spleen/metabolism , Tumor Necrosis Factor-alpha/metabolism , Uveitis/metabolism
17.
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
18.
Invest Ophthalmol Vis Sci ; 51(6): 3092-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20107166

ABSTRACT

PURPOSE: Induction of tissue-specific experimental autoimmune diseases involves the use of complete Freund adjuvant containing Mycobacterium tuberculosis, whose recognition by the innate immune system depends on Toll-like receptors (TLRs) that signal through the adaptor molecule MyD88. The authors' previous study showed that MyD88(-/-) mice, but not TLR2(-/-), TLR4(-/-), or TLR9(-/-) mice, were resistant to experimental autoimmune uveitis (EAU). METHODS: The EAU induction in mice deficient in TLR3 or mice double deficient in TLR2+4, TLR2+9, and TLR4+9 was examined and the role of the TLR agonists in the adjuvant effect involved in the induction of EAU was assessed. RESULTS: TLR3-deficient and TLR2+4, TLR2+9, and TLR4+9 double-deficient mice were as susceptible to EAU as their control littermates. However, in mice immunized with a low-dose EAU regimen, TLR4 agonist lipopolysaccharide (LPS) enhanced EAU scores, delayed-type hypersensitivity responses, and antigen-specific T-cell proliferation. Antigen-specific IL-17 and IFN-gamma production by T lymphocytes was markedly increased in the LPS-treated group. The effects of LPS on EAU were abolished by treatment with an LPS deactivator polymyxin B. Inclusion of agonists for TLR2, TRL3, or TRL9 in immunization also enhanced EAU scores. CONCLUSIONS: These results suggest that signaling of TLR2, TRL3, TRL4, and TRL9 is highly redundant in the adjuvant effect needed to induce EAU and that diverse microbial infections may contribute to the pathogenesis of diseases such as uveitis.


Subject(s)
Autoimmune Diseases/immunology , Autoimmunity/immunology , Disease Models, Animal , Myeloid Differentiation Factor 88/physiology , Toll-Like Receptors/physiology , Uveitis/immunology , Animals , Autoantigens/immunology , Eye Proteins/immunology , Female , Gene Silencing/physiology , Hypersensitivity, Delayed/immunology , Interferon-gamma/metabolism , Interleukin-17/metabolism , Lipopolysaccharides/pharmacology , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Retinol-Binding Proteins/immunology , Signal Transduction/physiology , T-Lymphocytes/immunology , Toll-Like Receptor 2/physiology , Toll-Like Receptor 3/physiology , Toll-Like Receptor 4/physiology , Toll-Like Receptor 9/physiology , Toll-Like Receptors/agonists
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...