Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Biochem Pharmacol ; 197: 114917, 2022 03.
Article in English | MEDLINE | ID: mdl-35041813

ABSTRACT

Noninfectious (autoimmune and immune-mediated) uveitis is one of the primary diseases leading to blindness in the world. Due to the limitation of current first-line drugs for clinical uveitis, novel drugs and targets against uveitis are urgently needed. Ganciclovir (GCV), an FDA-approved antiviral drug, is often used to treat cytomegalovirus-induced retinitis in clinical patients. Recently, GCV was found to suppress neuroinflammation via targeting STING signaling because the STING pathway plays a pivotal role in autoimmune diseases. However, until now, the effect of GCV on non-infectious uveitis has never been explored. In this work, using the rat experimental autoimmune uveitis (EAU) model, we first found STING to be highly expressed in infiltrating cells (CD68+, CD45+, and CD4+) and retinal glial cells (Iba1+ and GFAP+) of the immunized retina. More importantly, GCV treatment can significantly suppress the initiation and progression of EAU by inhibiting infiltration of Th17 and inflammatory cells into the retina. Mechanistically, we found that GCV could reverse the levels of pro-inflammatory factors (such as IL-1ß) and chemokine-related factors (such as Cxcr3), possibly via targeting the STING pathway. The present results suggest that GCV may be considered as a novel therapeutic strategy against human uveitis.


Subject(s)
Autoimmune Diseases/prevention & control , Ganciclovir/therapeutic use , Inflammation Mediators/antagonists & inhibitors , Retina/drug effects , Th17 Cells/drug effects , Uveitis/prevention & control , Animals , Autoimmune Diseases/chemically induced , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Disease Progression , Dose-Response Relationship, Drug , Eye Proteins/toxicity , Ganciclovir/pharmacology , Humans , Inflammation Mediators/immunology , Male , Rats , Rats, Inbred Lew , Retina/immunology , Retina/pathology , Retinol-Binding Proteins/toxicity , Th17 Cells/immunology , Th17 Cells/pathology , Uveitis/chemically induced , Uveitis/immunology , Uveitis/pathology
2.
Biochem Pharmacol ; 193: 114793, 2021 11.
Article in English | MEDLINE | ID: mdl-34600916

ABSTRACT

Uveitis is one of the most common blindness-causing ocular disorders. Due to its complicated pathogenesis, the treatment of uveitis has been widely recognized as a challenge for ophthalmologists. Recently, the anti-inflammatory properties of the antibiotic Azithromycin (AZM) have been reported. However, the therapeutic effects of Azithromycin in experimental autoimmune uveitis (EAU), a representative model of human AU, have not been elucidated till date. We conducted this study to examine the therapeutic effects and potential mechanisms of Azithromycin in EAU. We observed that Azithromycin significantly attenuated retinal inflammation in EAU mice at day 14 after immunization along with a significantly decreased inflammatory cell infiltration and cytokine production in the retina. Furthermore, we observed that Azithromycin increased the number of regulatory T cells (Treg) and decreased the number of effector T cells (Teff) in both the draining lymph nodes and spleen of EAU mice. Additionally, Azithromycin suppressed the proliferation and activation of CD4 + T cells, and induced the apoptosis of CD4 + CD44 + memory T and CD4 + CXCR3 + Th1 cells. Mechanistically, we proved that Azithromycin could regulate Teff/Treg balance by inhibiting the phosphorylation of S6 ribosomal protein, a downstream target of mammalian target of rapamycin (mTOR). Together, our findings revealed that Azithromycin alleviated EAU by regulating the Teff/Treg balance through the mTOR signaling pathway, suggesting that Azithromycin could be a promising therapeutic candidate for AU.


Subject(s)
Azithromycin/pharmacology , Inflammation/chemically induced , Inflammation/drug therapy , Retinal Diseases/drug therapy , T-Lymphocyte Subsets/drug effects , TOR Serine-Threonine Kinases/metabolism , Adoptive Transfer , Animals , Anti-Bacterial Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Female , Gene Expression Regulation/drug effects , Mice , Mice, Inbred C57BL , Peptide Fragments/toxicity , Retinal Diseases/chemically induced , Retinol-Binding Proteins/toxicity , T-Lymphocyte Subsets/physiology , TOR Serine-Threonine Kinases/genetics , Transcriptome
3.
Exp Eye Res ; 203: 108406, 2021 02.
Article in English | MEDLINE | ID: mdl-33347870

ABSTRACT

Experimental autoimmune uveoretinitis (EAU) in mice provides a useful platform to study the pathogenesis and experimental therapeutics of human uveitis. One often used EAU model employs C57BL/6 (B6) mice sensitized with a peptide residue having 1 to 20 amino acids of human interphotoreceptor retinoid binding protein (hIRBP1-20). The model using the B6 background has permitted a liberal use of genetically engineered strains and has provided insights for understanding uveoretinitis. However, this is usually acute/monophasic and does not represent human uveoretinitis that is characterized as a chronic/recurrent disease. Several chronic/recurrent EAU models have been developed; of these, we employed administration of staphylococcal enterotoxin B (SEB) for relapse in the present study, and found that recurrence was induced at day 24 after primary immunization, which is thought to be the convalescent phase. We reported the activation of invariant natural killer T (iNKT)-cells upon primary immunization of the EAU model mice with the ligand RCAI-56, which was found to mitigate the disease in our previous study. Here, we first attempted to ameliorate EAU in the relapse model using a preventive regimen by activating iNKT cells at the same time relapse induction (day 24) or in a regimen after 3 days of relapse induction (day 27). The preventive as well as post-inductive regimens were successful in reducing histopathological scores by inhibiting the Ag-specific Th17-biased response. Collectively, activation of iNKT cells may be useful to mitigate the relapse response of EAU induced with SEB.


Subject(s)
Autoimmune Diseases/prevention & control , Disease Models, Animal , Natural Killer T-Cells/physiology , Retinitis/prevention & control , Uveitis/prevention & control , Animals , Autoimmune Diseases/immunology , Cell Proliferation , Eye Proteins/toxicity , Female , Flow Cytometry , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Recurrence , Retinitis/immunology , Retinol-Binding Proteins/toxicity , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Uveitis/immunology
4.
Biochem Pharmacol ; 170: 113645, 2019 12.
Article in English | MEDLINE | ID: mdl-31545975

ABSTRACT

Autoimmune uveitis (AU), a sight-threatening intraocular disorder, is still a challenge for ophthalmologists in clinic. Teriflunomide has been approved for multiple sclerosis (MS) in 2012 for its immunoregulatory function. However, the effect and mechanisms of teriflunomide in uveitis are still unknown. In this investigation, we used a murine model of non-infectious uveitis, experimental autoimmune uveitis (EAU), to explore the anti-inflammatory features of teriflunomide. Treatment with teriflunomide resulted in reduced clinical and pathological scores of retinal inflammations, accompanied by decreased intraocular infiltration of Th17 and Th1 cells in EAU mice. Meanwhile, teriflunomide treatment inhibited the proliferation and polarization of CD4+ T cells to Th17 and Th1 cells. Moreover, adoptive transfer of teriflunomide primed IRBP1-20-T cells failed to induce EAU. Interestingly, we found that teriflunomide suppressed the maturation and function of dendritic cells (DCs) both in vivo and in vitro. In conclusion, our findings suggest that teriflunomide alleviates inflammation in EAU mice by down-regulating Th17 and Th1 cells and suppresses the maturation and function of DCs for the first time.


Subject(s)
Crotonates/therapeutic use , Dendritic Cells/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Toluidines/therapeutic use , Uveitis/drug therapy , Uveitis/immunology , Animals , Coculture Techniques , Crotonates/pharmacology , Dendritic Cells/drug effects , Disease Models, Animal , Eye Proteins/toxicity , Female , Hydroxybutyrates , Mice , Mice, Inbred C57BL , Nitriles , Retinol-Binding Proteins/toxicity , Th1 Cells/drug effects , Th17 Cells/drug effects , Toluidines/pharmacology , Uveitis/chemically induced
5.
Clin Exp Immunol ; 190(3): 293-303, 2017 12.
Article in English | MEDLINE | ID: mdl-28763100

ABSTRACT

Uveitis (intraocular inflammation) is a leading cause of loss of vision. Although its aetiology is largely speculative, it is thought to arise from complex genetic-environmental interactions that break immune tolerance to generate eye-specific autoreactive T cells. Experimental autoimmune uveitis (EAU), induced by immunization with the ocular antigen, interphotoreceptor retinoid binding protein (IRBP), in combination with mycobacteria-containing complete Freund's adjuvant (CFA), has many clinical and histopathological features of human posterior uveitis. Studies in EAU have focused on defining pathogenic CD4+ T cell effector responses, such as those of T helper type 17 (Th17) cells, but the innate receptor pathways precipitating development of autoreactive, eye-specific T cells remain poorly defined. In this study, we found that fungal-derived antigens possess autoimmune uveitis-promoting function akin to CFA in conventional EAU. The capacity of commensal fungi such as Candida albicans or Saccharomyces cerevisae to promote IRBP-triggered EAU was mediated by Card9. Because Card9 is an essential signalling molecule of a subgroup of C-type lectin receptors (CLRs) important in host defence, we evaluated further the proximal Card9-activating CLRs. Using single receptor-deficient mice we identified Dectin-2, but not Mincle or Dectin-1, as a predominant mediator of fungal-promoted uveitis. Conversely, Dectin-2 activation by α-mannan reproduced the uveitic phenotype of EAU sufficiently, in a process mediated by the Card9-coupled signalling axis and interleukin (IL)-17 production. Taken together, this report relates the potential of the Dectin-2/Card9-coupled pathway in ocular autoimmunity. Not only does it contribute to understanding of how innate immune receptors orchestrate T cell-mediated autoimmunity, it also reveals a previously unappreciated ability of fungal-derived signals to promote autoimmunity.


Subject(s)
Autoimmune Diseases/immunology , CARD Signaling Adaptor Proteins/immunology , Candida albicans/immunology , Candidiasis/immunology , Lectins, C-Type/immunology , Saccharomyces cerevisiae/immunology , Uveitis/immunology , Animals , Autoimmune Diseases/chemically induced , Autoimmune Diseases/pathology , CARD Signaling Adaptor Proteins/genetics , Candidiasis/chemically induced , Candidiasis/pathology , Eye Proteins/toxicity , Lectins, C-Type/genetics , Mice , Mice, Mutant Strains , Retinol-Binding Proteins/toxicity , Th17 Cells/immunology , Th17 Cells/pathology , Uveitis/chemically induced , Uveitis/genetics , Uveitis/pathology
6.
Invest Ophthalmol Vis Sci ; 57(8): 3747-58, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27415793

ABSTRACT

PURPOSE: To investigate the contribution of the gut microbiota to the pathogenesis of uveitis. METHODS: Experimental autoimmune uveitis (EAU) in B10.RIII mice was induced using interphotoreceptor binding protein peptide. Mice were treated with oral or intraperitoneal (IP) antibiotics. Effector (Teff) and regulatory (Treg) T lymphocytes were identified using flow cytometry; 16S rRNA gene sequencing and qPCR were performed on gastrointestinal (GI) contents. RESULTS: Broad-spectrum (four antibiotics given simultaneously) oral, but not IP, antibiotics reduced mean uveitis clinical scores significantly compared with water-treated animals (0.5 vs. 3.0, P < 0.0001 for oral; 3.4 vs. 3.4, P > 0.99 for IP). Both oral metronidazole (P = 0.02) and vancomycin (P < 0.0001) alone decreased inflammation, whereas neomycin (P = 0.7) and ampicillin (P = 0.4) did not change mean uveitis scores. Oral broad-spectrum antibiotics increased Tregs in the GI lamina propria of EAU animals at 1 week, and in extraintestinal lymphoid tissues later, whereas Teff and inflammatory cytokines were reduced. 16S sequencing of GI contents revealed altered microbiota in immunized mice compared with nonimmunized mice, and microbial diversity clustering in EAU mice treated with uveitis-protective antibiotics. Experimental autoimmune uveitis mice also demonstrated gut microbial diversity clustering associated with clinical score severity. CONCLUSIONS: Oral antibiotics modulate the severity of inducible EAU by increasing Tregs in the gut and extraintestinal tissues, as well as decreasing effector T cells and cytokines. 16S sequencing suggests that there may be protective and, conversely, potentially uveitogenic, gut microbiota. These findings may lead to a better understanding of how uveitis can be treated or prevented by modulating the gut microbiome.


Subject(s)
Autoimmune Diseases/microbiology , Gastrointestinal Microbiome/immunology , Uveitis/microbiology , Administration, Oral , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Autoimmune Diseases/prevention & control , Biomarkers/metabolism , Cells, Cultured , Cytokines/metabolism , Drug Combinations , Eye Proteins/toxicity , Female , Gastrointestinal Microbiome/drug effects , Injections, Intraperitoneal , Mice, Inbred Strains , Retina/immunology , Retina/microbiology , Retinol-Binding Proteins/toxicity , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Uveitis/prevention & control
7.
Cesk Slov Oftalmol ; 72(1): 276-82, 2016 Feb.
Article in Czech | MEDLINE | ID: mdl-27041283

ABSTRACT

INTRODUCTION: Autoimmune uveitis is a sight threatening disease which in many cases fails to respond to conventional immunosuppressive or biological therapy. The research in experimental models of autoimmune uveitis helps to find new therapeutical strategies. The aim of this study is to present the clinical and histological signs of experimental autoimmune uveitis (EAU) in mice. METHODS: EAU was induced in C57BL/6 mice by subcutaneous application of IRBP (interphotoreceptor retinoid binding protein) in complete Freunds adjuvant and intraperitoneal application of pertussis toxin. Clinical evaluation of uveitis was performed in vivo using special imaging system with otoscope. Histological evaluation of uveitis was performed at day 35 post induction of EAU on hematoxylin and eosin stained frozen sections. Clinical and histological grading was used to assess the inflammation intensity of EAU. RESULTS: The intensity of inflammation is depicted on representative fundus images and histological images of retina at day 35 post induction. CONCLUSION: The model of EAU is robust and reproducible and allows us to study the immunopathological mechanisms of inflammation and its regulation. The inflammatory signs in our model are similar to findings of posterior uveitis of autoimmune etiology in humans, thus we may apply our experimental results in human medicine.


Subject(s)
Autoimmune Diseases/diagnosis , Disease Models, Animal , Retina/pathology , Uveitis/diagnosis , Animals , Autoimmune Diseases/chemically induced , Eye Proteins/toxicity , Fundus Oculi , Immunosuppressive Agents , Mice, Inbred C57BL , Retinol-Binding Proteins/toxicity , Uveitis/chemically induced
8.
PLoS One ; 11(2): e0150078, 2016.
Article in English | MEDLINE | ID: mdl-26919582

ABSTRACT

γδ T cells can either enhance or inhibit an adaptive immune response, but the mechanisms involved are not fully understood. Given that CD73 is the main enzyme responsible for conversion of AMP into the immunosuppressive molecule adenosine, we investigated its role in the regulatory function of γδ T cells in experimental autoimmune uveitis (EAU). We found that γδ T cells expressed different amounts of CD73 during the different stages of EAU and that low CD73 expression on γδ T cells correlated with enhanced Th17 response-promoting activity. Functional comparison of CD73-deficient and wild-type B6 (CD73+/+) mice showed that failure to express CD73 decreased both the enhancing and suppressive effects of γδ T cells on EAU. We also demonstrated that γδ T cells expressed different amounts of CD73 when activated by different pathways, which enabled them to either enhance or inhibit an adaptive immune response. Our results demonstrate that targeting CD73 expression on γδ T cells may allow us to manipulate their pro- or anti-inflammatory effect on Th17 responses.


Subject(s)
5'-Nucleotidase/physiology , Nervous System Autoimmune Disease, Experimental/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Uveitis/immunology , 5'-Nucleotidase/biosynthesis , 5'-Nucleotidase/deficiency , 5'-Nucleotidase/genetics , Adenosine/metabolism , Adenosine Monophosphate/metabolism , Animals , Cells, Cultured , Dendritic Cells/immunology , Eye Proteins/immunology , Eye Proteins/toxicity , Female , Gene Expression Regulation/immunology , Interferon-gamma/blood , Interferon-gamma/deficiency , Interleukin-17/blood , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Nervous System Autoimmune Disease, Experimental/enzymology , Peptide Fragments/immunology , Peptide Fragments/toxicity , Receptors, Antigen, T-Cell, gamma-delta/analysis , Receptors, Antigen, T-Cell, gamma-delta/deficiency , Retinol-Binding Proteins/immunology , Retinol-Binding Proteins/toxicity , T-Lymphocyte Subsets/enzymology , T-Lymphocytes, Regulatory/enzymology , Th1 Cells/immunology , Th17 Cells/immunology , Uveitis/enzymology
9.
Clin Immunol ; 157(2): 198-204, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25701800

ABSTRACT

Uveitis is a sight-threatening inflammatory disease of the eye which represents the third leading cause of blindness in the developed countries. The conventional pharmacological treatment includes corticosteroids and immunosuppressive agents, which are limited by their side effects. New therapeutic strategies are thus strongly needed. Exogenously-administered carbon monoxide (CO) may represent an effective treatment for conditions characterized by a dysregulated inflammatory response. Carbon monoxide-releasing molecules (CORMs) are a novel group of compounds capable of carrying and liberating controlled quantities of CO. Among CORMs, CORM-A1 represents the first example of water soluble CO releaser. We show here that CORM-A1 under a late prophylactic regime is able to significantly ameliorate the natural course of experimental autoimmune uveoretinitis, a rodent model of immunoinflammatory posterior uveitis. The present study strongly supports the development of CORM-A1 as a potential new drug for treatment of patients with non-infectious posterior uveitis.


Subject(s)
Autoimmune Diseases/immunology , Boranes/pharmacology , Carbonates/pharmacology , RNA, Messenger/drug effects , Retina/drug effects , Retinitis/immunology , T-Lymphocytes, Regulatory/drug effects , Uvea/drug effects , Uveitis/immunology , Animals , Autoimmune Diseases/chemically induced , Autoimmune Diseases/pathology , Cytokines/drug effects , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Female , Gene Expression/drug effects , Peptide Fragments/toxicity , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Retina/immunology , Retina/pathology , Retinitis/chemically induced , Retinitis/pathology , Retinol-Binding Proteins/toxicity , Spleen/drug effects , Spleen/metabolism , T-Lymphocytes, Regulatory/immunology , Uvea/immunology , Uvea/pathology , Uveitis/chemically induced , Uveitis/pathology
10.
J Neuroinflammation ; 12: 17, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25623142

ABSTRACT

BACKGROUND: Experimental autoimmune uveoretinitis (EAU) is a widely used experimental animal model of human endogenous posterior uveoretinitis. In the present study, we performed in vivo imaging of the retina in transgenic reporter mice to investigate dynamic changes in exogenous inflammatory cells and endogenous immune cells during the disease process. METHODS: Transgenic mice (C57Bl/6 J Cx 3 cr1 (GFP/+) , C57Bl/6 N CD11c-eYFP, and C57Bl/6 J LysM-eGFP) were used to visualize the dynamic changes of myeloid-derived cells, putative dendritic cells and neutrophils during EAU. Transgenic mice were monitored with multi-modal fundus imaging camera over five time points following disease induction with the retinal auto-antigen, interphotoreceptor retinoid binding protein (IRBP1-20). Disease severity was quantified with both clinical and histopathological grading. RESULTS: In the normal C57Bl/6 J Cx 3 cr1 (GFP/+) mouse Cx3cr1-expressing microglia were evenly distributed in the retina. In C57Bl/6 N CD11c-eYFP mice clusters of CD11c-expressing cells were noted in the retina and in C57Bl/6 J LysM-eGFP mice very low numbers of LysM-expressing neutrophils were observed in the fundus. Following immunization with IRBP1-20, fundus examination revealed accumulations of Cx3cr1-GFP(+) myeloid cells, CD11c-eYFP(+) cells and LysM-eGFP(+) myelomonocytic cells around the optic nerve head and along retinal vessels as early as day 14 post-immunization. CD11c-eYFP(+) cells appear to resolve marginally earlier (day 21 post-immunization) than Cx3cr1-GFP(+) and LysM-eGFP(+) cells. The clinical grading of EAU in transgenic mice correlated closely with histopathological grading. CONCLUSIONS: These results illustrate that in vivo fundus imaging of transgenic reporter mice allows direct visualization of various exogenously and endogenously derived leukocyte types during EAU progression. This approach acts as a valuable adjunct to other methods of studying the clinical course of EAU.


Subject(s)
Autoimmune Diseases , Disease Models, Animal , Multimodal Imaging , Retinitis/pathology , Uveitis/complications , Uveitis/genetics , Uveitis/pathology , Animals , CD11c Antigen/genetics , CX3C Chemokine Receptor 1 , Disease Progression , Eye Proteins/toxicity , Freund's Adjuvant/toxicity , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Macrophages , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muramidase/genetics , Peptide Fragments/toxicity , Receptors, Chemokine/genetics , Retinal Vessels , Retinitis/chemically induced , Retinitis/complications , Retinitis/genetics , Retinol-Binding Proteins/toxicity , Time Factors , Uveitis/chemically induced
11.
J Leukoc Biol ; 96(6): 1077-85, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25201803

ABSTRACT

AGEs are permanently modified macromolecule derivatives that form through nonenzymatic glycation of amino groups of proteins. Glycer-AGEs are highly toxic and play an important role in the pathogenesis of chronic inflammatory diseases. However, the contribution of glycer-AGEs to the pathogenesis of uveitis is unclear. In this study, we measured serum levels of glycer-AGEs in 100 patients with endogenous uveitis (22 with HLA-B27-associated uveitis, 20 with VKH disease, 14 with Behçet's disease, and 44 with sarcoidosis) and 33 healthy volunteers. We then examined the effect of the AGE inhibitor in a mouse model of human endogenous uveitis (EAU) by continuous oral administration of pyridoxamine at 200 or 400 mg/kg/day. Regardless of the etiology, serum glycer-AGE levels were significantly higher in patients with uveitis than in healthy subjects. Treatment with 400 mg/kg pyridoxamine significantly reduced the clinical and histological severity of EAU and was accompanied by a significant decrease in serum and retinal glycer-AGE levels and suppression of translocation of NF-κB p65 into the nucleus of retinal cells. Serum glycer-AGE levels may therefore serve as a biomarker of human uveitis, as well as systemic inflammation, and may contribute to the progression of uveitis, including diabetic iritis, via the activation of NF-κB.


Subject(s)
Autoimmune Diseases/drug therapy , Glycation End Products, Advanced/antagonists & inhibitors , Pyridoxamine/therapeutic use , Retinitis/drug therapy , Uveitis/drug therapy , Administration, Oral , Adult , Amino Acid Sequence , Animals , Autoimmune Diseases/blood , Autoimmune Diseases/pathology , Behcet Syndrome/blood , Behcet Syndrome/complications , Disease Models, Animal , Drug Evaluation, Preclinical , Eye Proteins/immunology , Eye Proteins/metabolism , Eye Proteins/toxicity , Female , HLA-B27 Antigen/immunology , Humans , Male , Mice , Middle Aged , Molecular Sequence Data , Peptide Fragments/immunology , Peptide Fragments/toxicity , Protein Transport/drug effects , Pyridoxamine/administration & dosage , Pyridoxamine/pharmacology , Retina/metabolism , Retinitis/blood , Retinitis/etiology , Retinitis/pathology , Retinol-Binding Proteins/immunology , Retinol-Binding Proteins/toxicity , Sarcoidosis/blood , Sarcoidosis/complications , Uveitis/blood , Uveitis/etiology , Uveitis/pathology , Uveomeningoencephalitic Syndrome/blood , Uveomeningoencephalitic Syndrome/complications
12.
Invest Ophthalmol Vis Sci ; 52(11): 8076-85, 2011 Oct 17.
Article in English | MEDLINE | ID: mdl-21911582

ABSTRACT

PURPOSE: To investigate the effect of aldose reductase (AR) deficiency in protecting the chronic experimental autoimmune (EAU) and acute endotoxin-induced uveitis (EIU) in c57BL/6 mice. METHODS: The WT and AR-null (ARKO) mice were immunized with human interphotoreceptor retinoid-binding peptide (hIRPB-1-20), to induce EAU, or were injected subcutaneously with lipopolysaccharide (LPS; 100 µg) to induce EIU. The mice were killed on day 21 for EAU and at 24 hours for EIU, when the disease was at its peak, and the eyes were immediately enucleated for histologic and biochemical studies. Spleen-derived T-lymphocytes were used to study the antigen-specific immune response in vitro and in vivo. RESULTS: In WT-EAU mice, severe damage to the retinal wall, especially to the photoreceptor layer was observed, corresponding to a pathologic score of ∼2, which was significantly prevented in the ARKO or AR inhibitor-treated mice. The levels of cytokines and chemokines increased markedly in the whole-eye homogenates of WT-EAU mice, but not in ARKO-EAU mice. Further, expression of inflammatory marker proteins such as inducible nitric oxide synthase (iNOS), cyclooxygenase (COX)-2, tumor necrosis factor (TNF)-α, and vascular cell adhesion molecule (VCAM)-1 was increased in the WT-EIU mouse eyes but not in the ARKO-EIU eyes. The T cells proliferated vigorously when exposed to the hIRPB antigen in vitro and secreted various cytokines and chemokines, which were significantly inhibited in the T cells isolated from the ARKO mice. CONCLUSIONS: These findings suggest that AR-deficiency/inhibition protects against acute as well as chronic forms of ocular inflammatory complications such as uveitis.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Autoimmune Diseases/prevention & control , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Imidazolidines/pharmacology , Uveitis/prevention & control , Adoptive Transfer , Animals , Aqueous Humor/metabolism , Autoimmune Diseases/chemically induced , Autoimmune Diseases/enzymology , Autoimmune Diseases/pathology , Blotting, Western , Cells, Cultured , Chemokines/metabolism , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Eye Proteins/toxicity , Fluorescent Antibody Technique, Indirect , Leukocytes/physiology , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Reactive Oxygen Species/metabolism , Retinol-Binding Proteins/toxicity , T-Lymphocytes/immunology , Uveitis/chemically induced , Uveitis/enzymology , Uveitis/pathology
13.
Invest Ophthalmol Vis Sci ; 51(1): 383-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19696173

ABSTRACT

PURPOSE: To investigate the role of CD4(+)CD25(+) Treg cells in the development of experimental autoimmune uveoretinitis (EAU). METHODS: EAU was induced in B10RIII mice by immunization with IRBP(161-180) in complete Freund's adjuvant and evaluated clinically and pathologically on days 0, 7, 14, 21, and 28. Lymphocytes from draining lymph nodes (LNs) were subjected to flow cytometry to analyze the frequency of CD4(+)CD25(+) Treg cells. CD4(+)CD25(+) Treg cells and CD4(+)CD25(-) T cells were separated by means of magnetic-assisted cell sorting and cocultured or crossover cultured for 3 days. Proliferation of CD4(+)CD25(-) T cells was measured using a modified MTT assay. The levels of IFN-gamma and IL-17 in the supernatants were determined by enzyme-linked immunosorbent assay. RESULTS: Clinical and histopathologic results showed a severe intraocular inflammation in the immunized mice. The frequency of CD4(+)Foxp3(+) T cells and CD4(+)CD25(+)Foxp3(+) T cells in the draining LN lymphocytes was increased on day 7, reached its peak on day 14, and maintained a high level up to day 42. CD4(+)CD25(+) Treg cells obtained from mice on days 14 and 28 after immunization showed a stronger inhibitory effect on the proliferation of CD4(+)CD25(-) T cells and the production of IFN-gamma by CD4(+)CD25(-) T cells compared with those obtained from control mice. CD4(+)CD25(+) Treg cells did not affect IL-17 production. Transfer of CD4(+)CD25(+) Treg cells obtained from EAU mice was able to suppress EAU induction by IRBP(161-180) that was not observed after transfer of cells from mice that had received CFA alone, suggesting antigen specificity of the Treg response. CONCLUSIONS: A significantly increased frequency and immunoregulatory action of CD4(+)CD25(+) Treg cells is associated with the development and regression of EAU, suggesting that CD4(+)CD25(+) Treg cells are induced during EAU and may be involved in its regression.


Subject(s)
Autoimmune Diseases/immunology , CD4 Antigens/metabolism , Disease Models, Animal , Interleukin-2 Receptor alpha Subunit/metabolism , Retinitis/immunology , T-Lymphocytes, Regulatory/physiology , Uveitis/immunology , Adoptive Transfer , Animals , Autoimmune Diseases/chemically induced , Autoimmune Diseases/pathology , Autoimmune Diseases/prevention & control , Cell Separation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Forkhead Transcription Factors/metabolism , Interferon-gamma , Interleukin-17 , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Mice , Peptide Fragments/toxicity , Retinitis/chemically induced , Retinitis/pathology , Retinitis/prevention & control , Retinol-Binding Proteins/toxicity , Uveitis/chemically induced , Uveitis/pathology , Uveitis/prevention & control
14.
Invest Ophthalmol Vis Sci ; 49(1): 282-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18172104

ABSTRACT

PURPOSE: To determine the involvement of retinal astrocytes (RACs) in T cell-mediated experimental autoimmune uveitis (EAU). METHODS: Frozen sections of eyes from naive mice or mice with EAU were stained for glial fibrillary acidic protein (GFAP) or major histocompatibility complex (MHC) class II molecules and were examined by confocal microscopy. RACs were isolated and cocultured with interphotoreceptor retinoid-binding protein (IRBP) peptide-specific T cells. The proliferation and cytokine production of responder T cells were determined by [(3)H]-thymidine incorporation and ELISA, respectively. RESULTS: The development of intraocular inflammation was associated with increased GFAP-positive cells in the retina. RACs from EAU-prone mice (B10RIII) activated uveitogenic T cells in vitro, enhanced T-cell proliferation and the production of proinflammatory cytokines, and increased the numbers of IL-17(+) IRBP T cells in the inflamed eye. The interaction between local RACs and IRBP-specific T cells was regulated by a distinct pattern of costimulatory molecules. In addition, the ability of IRBP-specific T cells to interact with RACs was dependent on whether the latter were derived from EAU-prone (B10RIII) or EAU-low susceptible (C57Bl/6) strains of mice. CONCLUSIONS: This study suggests that the RACs in EAU-prone mice contribute to the reactivation of pathogenic T cells in the eye, leading to intraocular inflammation and tissue damage.


Subject(s)
Astrocytes/physiology , Autoimmune Diseases/immunology , Lymphocyte Activation/immunology , Retina/immunology , T-Lymphocytes/immunology , Uveitis/immunology , Adoptive Transfer , Animals , Coculture Techniques , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Eye Proteins/toxicity , Female , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Glial Fibrillary Acidic Protein/metabolism , Histocompatibility Antigens Class II/metabolism , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Retinol-Binding Proteins/toxicity , Specific Pathogen-Free Organisms
15.
Int Immunol ; 20(2): 209-14, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18156624

ABSTRACT

IL-17-producing CD4(+) T cells, so called T(h)17 cells, constitute a newly identified inflammatogenic cell population, which is critically involved in some inflammatory diseases. To explore the role of T(h)17 cells in murine experimental autoimmune uveoretinitis (EAU), a model of human autoimmune uveitis where T(h)1 responses predominantly participate in the pathogenesis, IL-17(-/-) mice were immunized with interphotoreceptor retinoid-binding protein peptide 1-20 for disease induction. Funduscopic examination revealed that EAU was induced in IL-17(-/-) mice just like in wild-type (WT) mice at early phases of the disease. However, at later/maintenance phases, the severity was significantly reduced in IL-17(-/-) mice. Expression of IFN-gamma and MCP-1 was comparable between WT and IL-17(-/-) mice during the time course. In vivo blockade of IFN-gamma and IL-4 resulted in exacerbation of EAU at later phases with augmented IL-17 production. Taken together, our data demonstrated that IL-17/T(h)17 participates in the late phases of EAU and also that T(h)1 and T(h)17 responses are differentially required for EAU.


Subject(s)
Autoimmune Diseases , Interferon-gamma/metabolism , Interleukin-17/metabolism , Retinitis , Uveitis , Amino Acid Sequence , Animals , Autoimmune Diseases/chemically induced , Autoimmune Diseases/immunology , Autoimmune Diseases/physiopathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Eye Proteins/administration & dosage , Eye Proteins/chemistry , Eye Proteins/toxicity , Humans , Inflammation/immunology , Inflammation/physiopathology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Retinitis/chemically induced , Retinitis/immunology , Retinitis/physiopathology , Retinol-Binding Proteins/administration & dosage , Retinol-Binding Proteins/chemistry , Retinol-Binding Proteins/toxicity , Th1 Cells , Uveitis/chemically induced , Uveitis/immunology , Uveitis/physiopathology
16.
Curr Eye Res ; 32(11): 917-22, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18027167

ABSTRACT

PURPOSE: The complement system plays important roles in a variety of chronic ocular diseases such as age-related macular degeneration. Here we examined the deposition of complement components in mouse eyes damaged by various mechanisms. METHODS: Mouse eyes were damaged by light or by three models of inflammation, i.e., local transgenic expression of cytokines, interleukin-1 or -7, or by induction of experimental autoimmune uveitis. Eye tissues obtained from each model were immunostained with antibodies against complement components C1q, C3, and C4. RESULTS: No complement deposition was seen in light damaged eyes, while in inflamed eyes we found complement deposition at sites of tissue damage and cellular infiltration. In addition to affected tissues, intense immunoreactivity against complement was unexpectedly observed in corneal tissues and lens capsule, despite lack of inflammation in these tissues. CONCLUSION: Our observations suggest that ocular tissues adjacent to inflammatory sites undergo changes that facilitate complement deposition.


Subject(s)
Autoimmune Diseases/metabolism , Complement C1q/metabolism , Complement C3/metabolism , Complement C4/metabolism , Inflammation/metabolism , Uveitis/metabolism , Animals , Autoimmune Diseases/chemically induced , Cornea/metabolism , Eye Proteins/toxicity , Female , Gene Expression/physiology , Interleukin-1/genetics , Interleukin-7/genetics , Lens Capsule, Crystalline/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Microscopy, Fluorescence , Photoreceptor Cells, Vertebrate/radiation effects , Radiation Injuries, Experimental/metabolism , Retinal Diseases/metabolism , Retinol-Binding Proteins/toxicity , Uveitis/chemically induced
17.
Invest Ophthalmol Vis Sci ; 46(10): 3753-60, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16186359

ABSTRACT

PURPOSE: Experimental autoimmune uveoretinitis (EAU) is an organ-specific, Th1-cell-mediated disease that targets the neural retina. CCR5 is a chemokine receptor expressed on Th1 cells that promotes their migration. In CCR5-deficient mice, we examined the role of CCR5 in the development of EAU induced by immunization with interphotoreceptor retinoid-binding protein (IRBP) peptide. METHODS: Wild-type or CCR5-deficient B6 mice were immunized with human IRBP peptide 1-20 (hIRBP-p), and the severity of EAU was assessed clinically and histologically. Splenocytes and cells of regional lymph nodes near the eye were collected and their proliferation and production of IL-6, IL-10, IFN-gamma, and CCL2 (MCP-1) in response to hIRBP-p stimulation were measured. Moreover, the intraocular levels of these cytokines were analyzed. RESULTS: Immunization with hIRBP-p induced EAU in CCR5-deficient mice with a severity comparable to that in wild-type mice. Histologically, T-cell infiltration of the eye was reduced, but granulocyte infiltration was augmented in CCR5-deficient mice. Although splenic T cells from CCR5-deficient mice produced IFN-gamma but not IL-10 on stimulation by hIRBP-p, T cells from the regional lymph nodes failed to produce both cytokines. IL-6 production in the eye and IL-6 and CCL2 production by splenic T cells were predominantly augmented in CCR5-deficient mice. CONCLUSIONS: The development of EAU is not prevented in CCR5-deficient mice. Although T-cell infiltration into the eye is apparently reduced in CCR5-deficient mice, the defect is compensated for by granulocyte infiltration, supposedly mediated by augmented intraocular production of IL-6.


Subject(s)
Autoimmune Diseases/immunology , Receptors, CCR5/physiology , Retinitis/immunology , Uveitis/immunology , Animals , Autoimmune Diseases/chemically induced , Autoimmune Diseases/pathology , Cytokines/biosynthesis , Disease Models, Animal , Eye Proteins/toxicity , Female , Immunization , Lymph Nodes/cytology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Retinitis/chemically induced , Retinitis/pathology , Retinol-Binding Proteins/toxicity , Spleen/cytology , T-Lymphocytes/immunology , Uveitis/chemically induced , Uveitis/pathology
18.
Arch Ophthalmol ; 123(7): 957-63, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16009838

ABSTRACT

OBJECTIVE: To evaluate the effects of an interleukin 1 receptor antagonist (IL-1RA) on the development of immune-mediated ocular inflammation in mice. METHODS: Recombinant, human, nonglycosylated IL-1RA (anakinra [kineret]) was tested for its inhibitory effects in 2 systems: (1) experimental autoimmune uveitis induced by interphotoreceptor retinoid-binding protein in B10.A mice using routine procedures and evaluated by clinical and histological examination, and (2) ocular inflammation in mice induced by transfer of hen egg lysozyme-specific T cells to hen egg lysozyme-transgenic mice. Treatment with IL-1RA included daily subcutaneous injections of the drug, at 300 and 500 mg/kg, or phosphate-buffered saline as control. RESULTS: Mean +/- SE experimental autoimmune uveitis scores of histological ocular changes of the mice at day 14 postimmunization with interphotoreceptor retinoid-binding protein were 1.5 +/- 0.3 in control mice; 1.0 +/- 0.4 in 300-mg/kg anakinra-treated mice; and 0.5 +/- 0.2 in 500- mg/kg anakinra-treated mice (P = .004). There was a corresponding decrease in the cellular immune response and cytokine production of immune cells in treated mice. Suppression of ocular inflammation by anakinra in the transfer system was also observed (P = .04). CONCLUSION: Human IL-1RA suppresses immune-mediated ocular inflammation in mice, affecting both the afferent and efferent components of the pathogenic immune response.Clinical Relevance Systemic administration of IL-1RA may have clinical application in the management of patients with uveitis.


Subject(s)
Autoimmune Diseases/prevention & control , Recombinant Proteins/administration & dosage , Sialoglycoproteins/administration & dosage , Uveitis/prevention & control , Adoptive Transfer , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Cytokines/biosynthesis , Disease Models, Animal , Eye Proteins/immunology , Eye Proteins/toxicity , Female , Immunity, Cellular/drug effects , Immunosuppression Therapy , Immunotherapy, Adoptive , Injections, Subcutaneous , Interleukin 1 Receptor Antagonist Protein , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Muramidase/immunology , Retinol-Binding Proteins/immunology , Retinol-Binding Proteins/toxicity , Th1 Cells/immunology , Uveitis/immunology , Uveitis/pathology
19.
J Neuroimmunol ; 164(1-2): 22-30, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15921764

ABSTRACT

Lymphocyte trafficking is controlled in part by the actions of chemokines. In rat experimental autoimmune uveitis (EAU) we observed differential therapeutic effects of Met-RANTES, a CCR1/CCR5 receptor antagonist, depending on the retinal antigen peptides inducing the disease and the time of application during the afferent or efferent immune response. CCR1 and/or CCR5 blockade may have inhibitory effects on different phases of the autoimmune response, depending on the antigen specificity of T cells in EAU. In contrast, Met-RANTES enhanced therapeutic oral tolerance independently of orally applied antigen.


Subject(s)
Autoimmune Diseases/drug therapy , Chemokine CCL5/analogs & derivatives , Chemokine CCL5/therapeutic use , Chemokines, CC/antagonists & inhibitors , Uveitis/drug therapy , Animals , Arrestin/chemistry , Arrestin/toxicity , Autoimmune Diseases/chemically induced , Cytokines/metabolism , Drug Interactions , Ectodysplasins , Eye/drug effects , Eye/metabolism , Eye/pathology , Eye Proteins/chemistry , Eye Proteins/toxicity , Immunohistochemistry/methods , Membrane Proteins/metabolism , Peptides/toxicity , RNA, Messenger/biosynthesis , Rats , Rats, Inbred Lew , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Retinol-Binding Proteins/chemistry , Retinol-Binding Proteins/toxicity , Reverse Transcriptase Polymerase Chain Reaction/methods , Severity of Illness Index , T-Lymphocytes/drug effects , Time Factors , Uveitis/chemically induced , Vaccination/methods
20.
Eur J Immunol ; 32(9): 2598-606, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12207344

ABSTRACT

Equine recurrent uveitis (ERU) is an inflammatory eye disease with high similarity to uveitis in man. It is the only spontaneous animal model for uveitis and the most frequent eye disease in horses affecting up to 10% of the population. To further investigate the pathophysiology of ERU we now report the establishment of an inducible uveitis model in horses. An ERU-like disease was elicited in seven out of seven horses by injection of interphotoreceptor retinoid-binding protein (IRBP) in complete Freund's adjuvant. Control horses did not develop uveitis. The disease model is characterized by a highly reproducible disease course and recurrent episodes with an identical time course elicited in all horses by repeated IRBP injections. The histology revealed the formation of lymphoid follicle-like structures in the eyes and an intraocular infiltration dominated by CD3(+) lymphocytes, morphological patterns typical for the spontaneous disease. Antigen-specific T cell proliferation of PBL was monitored prior to clinical uveitis and during disease episodes. An initial T cell response to IRBP-derived peptides was followed by epitope spreading to S-antigen-derived peptides in response to subsequent immunizations. Thus, horse experimental uveitis represents a valuable disease model for comparative studies with the spontaneous disease and the investigation of immunomodulatory therapeutic approaches after onset of the disease.


Subject(s)
Autoantigens/immunology , Autoimmune Diseases/veterinary , Eye Proteins , Horse Diseases/immunology , Retinol-Binding Proteins/immunology , Uveitis/veterinary , Amino Acid Sequence , Animals , Autoantigens/administration & dosage , Autoantigens/toxicity , Autoimmune Diseases/etiology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , CD3 Complex/analysis , Cattle , Disease Models, Animal , Freund's Adjuvant , Horse Diseases/etiology , Horse Diseases/pathology , Horses , Humans , Immunization , Immunization, Secondary , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/drug effects , Molecular Sequence Data , Peptide Fragments/immunology , Peptide Fragments/toxicity , Pertussis Toxin/immunology , Recurrence , Reproducibility of Results , Retinol-Binding Proteins/administration & dosage , Retinol-Binding Proteins/toxicity , Species Specificity , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Uveitis/etiology , Uveitis/immunology , Uveitis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...