Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Exp Eye Res ; 203: 108406, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33347870

RESUMO

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.


Assuntos
Doenças Autoimunes/prevenção & controle , Modelos Animais de Doenças , Células T Matadoras Naturais/fisiologia , Retinite/prevenção & controle , Uveíte/prevenção & controle , Animais , Doenças Autoimunes/imunologia , Proliferação de Células , Proteínas do Olho/toxicidade , Feminino , Citometria de Fluxo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Recidiva , Retinite/imunologia , Proteínas de Ligação ao Retinol/toxicidade , Células Th1/imunologia , Células Th17/imunologia , Células Th2/imunologia , Uveíte/imunologia
2.
Exp Eye Res ; 190: 107885, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31758977

RESUMO

Diabetic retinopathy (DR) is triggered by retinal cell damage stimulated by the diabetic milieu, including increased levels of intraocular free fatty acids. Free fatty acids may serve as an initiator of inflammatory cytokine release from Müller cells, and the resulting cytokines are potent stimulators of retinal endothelial pathology, such as leukostasis, vascular permeability, and basement membrane thickening. Our previous studies have elucidated a role for peroxisome proliferator-activated receptor-ß/δ (PPARß/δ) in promoting several steps in the pathologic cascade in DR, including angiogenesis and expression of inflammatory mediators. Furthermore, PPARß/δ is a known target of lipid signaling, suggesting a potential role for this transcription factor in fatty acid-induced retinal inflammation. Therefore, we hypothesized that PPARß/δ stimulates both the induction of inflammatory mediators by Müller cells as well the paracrine induction of leukostasis in endothelial cells (EC) by Müller cell inflammatory products. To test this, we used the PPARß/δ inhibitor, GSK0660, in primary human Müller cells (HMC), human retinal microvascular endothelial cells (HRMEC) and mouse retina. We found that palmitic acid (PA) activation of PPARß/δ in HMC leads to the production of pro-angiogenic and/or inflammatory cytokines that may constitute DR-relevant upstream paracrine inflammatory signals to EC and other retinal cells. Downstream, EC transduce these signals and increase their synthesis and release of chemokines such as CCL8 and CXCL10 that regulate leukostasis and other cellular events related to vascular inflammation in DR. Our results indicate that PPARß/δ inhibition mitigates these upstream (MC) as well as downstream (EC) inflammatory signaling events elicited by metabolic stimuli and inflammatory cytokines. Therefore, our data suggest that PPARß/δ inhibition is a potential therapeutic strategy against early DR pathology.


Assuntos
Células Ependimogliais/efeitos dos fármacos , Leucostasia/prevenção & controle , PPAR delta/antagonistas & inibidores , PPAR beta/antagonistas & inibidores , Retinite/prevenção & controle , Sulfonas/farmacologia , Tiofenos/farmacologia , Adulto , Animais , Células Cultivadas , Citocinas/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Ependimogliais/metabolismo , Humanos , Inflamação , Leucostasia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ácidos Palmíticos/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Retina/efeitos dos fármacos , Retina/metabolismo , Retinite/metabolismo
3.
Vet Ophthalmol ; 23(5): 797-805, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32618114

RESUMO

OBJECTIVE: To investigate the effects of CD40 on ocular inflammation in experimental autoimmune uveoretinitis (EAU) in B10.RIII mice. ANIMALS STUDIED: EAU-susceptible B10.RIII mice were subcutaneously immunized with interphotoreceptor retinoid-binding protein (IRBP) 161-180 in complete Freund's adjuvant and evaluated clinically and pathologically on days 7, 14, 21, 28, and 35 postimmunization. Anti-CD40 antibody was intraperitoneally injected into mice every other day from days 7 to 14 postimmunization. Phosphate-buffered saline (PBS)-injected EAU mice were used as the controls. PROCEDURES: The frequencies of CD11c+ CD40+ dendritic cells (DCs), CD11c+ MHC-II+ DCs, and CD11c+ CD40+ MHC-II+ DCs in splenocytes were evaluated by flow cytometry on days 0, 7, 14, and 21 after immunization. Tumor necrosis factor (TNF)-α and interleukin (IL)-6 production in CD11c+ DCs was assessed by ELISA. IRBP-specific lymphocyte proliferation was assessed using a modified MTT cell proliferation assay. RESULTS: The number of CD11c+ CD40+ DCs, CD11c+ MHC-II+ DCs, and CD11c+ CD40+ MHC-II+ DCs increased at the onset of EAU, peaked at the height of disease severity, and was sustained at a high level until day 21. Treatment with anti-CD40 antibody significantly alleviated clinical and pathological activities related to EAU. Compared with the control mice, antibody-treated EAU mice showed few CD11c+ CD40+ DC and CD11c+ CD40+ MHC-II+ DC frequencies in splenocytes. The anti-CD40 antibody significantly suppressed IRBP-specific lymphocyte proliferation and TNF-α and IL-6 production by DCs in EAU mice. CONCLUSIONS: The increased expression of CD40 and major histocompatibility complex (MHC) class II molecules in the splenocytes of EAU mice were correlated with inflammatory activity. Anti-CD40 treatment can significantly attenuate EAU activity by inhibiting systemic IRBP-specific immune responses.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Autoantígenos/imunologia , Doenças Autoimunes/terapia , Retinite/prevenção & controle , Fator de Necrose Tumoral alfa/imunologia , Uveíte Posterior/prevenção & controle , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Mutantes
4.
Exp Eye Res ; 178: 27-36, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30240585

RESUMO

Vascular inflammation plays a critical role in the pathogenesis of diabetic retinopathy. Recently, Interleukin-6 (IL-6) trans-signaling via soluble IL-6 receptor (sIL-6R) has emerged as a prominent regulator of inflammation in endothelial cells. This study was designed to test the hypothesis that selective inhibition of the IL-6 trans-signaling pathway will attenuate inflammation and subsequent barrier disruption in retinal endothelial cells. Human retinal endothelial cells (HRECs) were exposed to IL-6 and sIL-6R to induce IL-6 trans-signaling and the commercially available compound sgp130Fc (soluble gp-130 fused chimera) was used to selectively inhibit IL-6 trans-signaling. IL-6 trans-signaling activation caused a significant increase in STAT3 phosphorylation, expression of adhesion molecules, ROS production and apoptosis in HRECs whereas a significant decrease in mitochondrial membrane potential and NO production was observed in IL-6 trans-signaling activated cells. These changes were not observed in cells pre-treated with sgp130Fc. IL-6 trans-signaling activation was sufficient to cause barrier disruption in endothelial monolayers and pre-treatment of HRECs with sgp130Fc, maintained endothelial barrier function similar to that of untreated cells. Thus, in conclusion, these results indicate that IL-6 trans-signaling is an important mediator of inflammation, apoptosis and barrier disruptive effects in the retinal endothelial cells and inhibition of the IL-6 trans-signaling pathway using sgp130-Fc attenuates vascular inflammation and endothelial barrier disruption.


Assuntos
Barreira Hematorretiniana/efeitos dos fármacos , Células Endoteliais/metabolismo , Inflamação/prevenção & controle , Interleucina-6/antagonistas & inibidores , Proteínas Recombinantes de Fusão/farmacologia , Transdução de Sinais/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Western Blotting , Células Cultivadas , Células Endoteliais/patologia , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Peróxido de Hidrogênio/metabolismo , Interleucina-6/metabolismo , Potencial da Membrana Mitocondrial , Microscopia Confocal , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Óxido Nítrico/metabolismo , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Receptores de Interleucina-6/metabolismo , Retinite/prevenção & controle , Fator de Transcrição STAT3/metabolismo
5.
Vis Neurosci ; 34: E009, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28965505

RESUMO

Chronic low grade inflammation is considered to contribute to the development of experimental diabetic retinopathy (DR). We recently demonstrated that lack of CD40 in mice ameliorates the upregulation of inflammatory molecules in the diabetic retina and prevented capillary degeneration, a hallmark of experimental diabetic retinopathy. Herein, we investigated the contribution of CD40 to diabetes-induced reductions in retinal function via the electroretinogram (ERG) to determine if inflammation plays a role in the development of ERG defects associated with diabetes. We demonstrate that diabetic CD40-/- mice are not protected from reduction to the ERG b-wave despite failing to upregulate inflammatory molecules in the retina. Our data therefore supports the hypothesis that retinal dysfunction found in diabetics occurs independent of the induction of inflammatory processes.


Assuntos
Antígenos CD40/fisiologia , Diabetes Mellitus Experimental/prevenção & controle , Retinopatia Diabética/prevenção & controle , Retina/fisiopatologia , Retinite/prevenção & controle , Animais , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/fisiopatologia , Retinopatia Diabética/genética , Retinopatia Diabética/fisiopatologia , Eletrorretinografia , Feminino , Molécula 1 de Adesão Intercelular/genética , Interleucina-1beta/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Retinite/genética , Retinite/fisiopatologia , Fator de Necrose Tumoral alfa/genética , Regulação para Cima
6.
J Neuroinflammation ; 12: 136, 2015 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-26219952

RESUMO

BACKGROUND: Inflammation and the excess production of reactive oxygen species (ROS) contribute significantly to the pathogenesis of ischemic retinopathies such as diabetic retinopathy and retinopathy of prematurity. We hypothesized that GKT137831, a dual inhibitor of NADPH oxidases (NOX) 1 and NOX4, reduces inflammation in the ischemic retina by dampening the pro-inflammatory phenotype of retinal immune cells as well as macroglial Müller cells and neurons. METHODS: Ischemic retinopathy was induced in Sprague-Dawley rats by exposure to 80 % O2 cycled with 21 % O2 for 3 h per day from postnatal day (P) 0 to P11, followed by room air (P12 to P18). GKT137831 was administered P12 to P18 (60 mg/kg, subcutaneous) and comparisons were to room air controls. Retinal inflammation was examined by measuring leukocyte adherence to the retinal vasculature, ionized calcium-binding adaptor protein-1-positive microglia/macrophages, and the mRNA and protein levels of key inflammatory factors involved in retinal disease. Damage to Müller cells was evaluated by quantitating glial fibrillary acidic protein-positive cells and vascular leakage with an albumin ELISA. To verify the anti-inflammatory actions of GKT137831 on glia and neurons involved in ischemic retinopathy, primary cultures of rat retinal microglia, Müller cells, and ganglion cells were exposed to the in vitro counterpart of ischemia, hypoxia (0.5 %), and treated with GKT137831 for up to 72 h. ROS levels were evaluated with dihydroethidium and the protein and gene expression of inflammatory factors with quantitative PCR, ELISA, and a protein cytokine array. RESULTS: In the ischemic retina, GKT137831 reduced the increased leukocyte adherence to the vasculature, the pro-inflammatory phenotype of microglia and macroglia, the increased gene and protein expression of vascular endothelial growth factor, monocyte chemoattractant protein-1, and leukocyte adhesion molecules as well as vascular leakage. In all cultured cell types, GKT137831 reduced the hypoxia-induced increase in ROS levels and protein expression of various inflammatory mediators. CONCLUSIONS: NOX1/4 enzyme inhibition with GKT137831 has potent anti-inflammatory effects in the retina, indicating its potential as a treatment for a variety of vision-threatening retinopathies.


Assuntos
NADH NADPH Oxirredutases/antagonistas & inibidores , NADPH Oxidases/antagonistas & inibidores , Neuroglia/efeitos dos fármacos , Pirazóis/farmacologia , Pirazóis/uso terapêutico , Piridinas/farmacologia , Piridinas/uso terapêutico , Retinite/prevenção & controle , Animais , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Quimiocina CCL2/metabolismo , Modelos Animais de Doenças , Hipóxia/complicações , Técnicas In Vitro , Molécula 1 de Adesão Intercelular/metabolismo , Isquemia/etiologia , Isquemia/patologia , Isquemia/prevenção & controle , NADH NADPH Oxirredutases/efeitos dos fármacos , NADPH Oxidase 1 , NADPH Oxidase 4 , NADPH Oxidases/efeitos dos fármacos , Neuroglia/metabolismo , Neuroglia/patologia , Pirazolonas , Piridonas , Ratos , Espécies Reativas de Oxigênio/metabolismo , Retinite/metabolismo , Retinite/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Life Sci ; 355: 122996, 2024 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-39173995

RESUMO

AIMS: To investigate the therapeutic potential of visual stimulation (VS) and BDNF in murine experimental autoimmune uveoretinitis (EAU). MAIN METHODS: Mice were immunized by subcutaneous injection of interphotoreceptor retinoid-binding protein in Freund's complete adjuvant and intravenous injection of pertussis toxin, and were then exposed to high-contrast VS 12 h/day (days 1-14 post-immunization). EAU severity was assessed by examining clinical score, visual acuity, inflammatory markers, and immune cells in the retina. The transcriptome of activated retinal cells was determined by RNA-seq using RNA immunoprecipitated in complex with phosphorylated ribosomal protein S6. The retinal levels of protein products of relevant upregulated genes were quantified. The effect of BDNF on EAU was tested in unstimulated mice by its daily topical ocular administration (days 8-14 post-immunization). KEY FINDINGS: VS attenuated EAU development and decreased the expression of pro-inflammatory cytokines/chemokines and numbers of immune cells in the retina (n = 10-20 eyes/group for each analysis). In activated retinal cells of control mice (n = 30 eyes/group), VS upregulated genes encoding immunomodulatory neuropeptides, of which BDNF and vasoactive intestinal peptide (VIP) also showed increased mRNA and protein levels in the retina of VS-treated EAU mice (n = 6-10 eyes/group for each analysis). In unstimulated EAU mice, BDNF treatment mimicked the protective effects of VS by modulating the inflammatory and stem cell properties of Müller cells (n = 5 eyes/group for each analysis). SIGNIFICANCE: VS effectively suppresses EAU, at least through enhancing retinal levels of anti-inflammatory and neuroprotective factors, VIP and BDNF. Our findings also suggest BDNF as a promising therapeutic agent for uveitis treatment.


Assuntos
Doenças Autoimunes , Fator Neurotrófico Derivado do Encéfalo , Retinite , Uveíte , Animais , Camundongos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Uveíte/metabolismo , Uveíte/tratamento farmacológico , Uveíte/imunologia , Retinite/tratamento farmacológico , Retinite/prevenção & controle , Retinite/imunologia , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Feminino , Retina/metabolismo , Retina/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Peptídeo Intestinal Vasoativo/farmacologia , Modelos Animais de Doenças , Citocinas/metabolismo
8.
Am J Pathol ; 180(2): 672-81, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22119714

RESUMO

Fingolimod (FTY720) is an FDA-approved therapeutic drug with efficacy demonstrated in experimental models of multiple sclerosis and in phase III human multiple sclerosis trials. Fingolimod prevents T-cell migration to inflammatory sites by decreasing expression of the sphingosine-1 phosphate receptor normally required for egress from secondary lymphoid tissue. As a preclinical model of human uveitis, experimental autoimmune uveoretinitis permits assessment of immunotherapeutic efficacy. Murine experimental autoimmune uveoretinitis is induced by activation of retinal antigen-specific CD4(+) T cells that infiltrate the eye. High-dose fingolimod treatment administered before disease onset reduces ocular infiltration within hours of administration and suppresses clinicopathologic expression of experimental autoimmune uveoretinitis. In the present investigation of the efficacy of fingolimod treatment for established disease, single-dose treatment was effective and immunosuppressive ability was maintained through a dose range, demonstrating significant and rapid reduction in CD4(+) cell infiltration at clinically relevant therapeutic doses of fingolimod. A repeated-treatment regimen using a dose similar to that in current multiple sclerosis patient protocols significantly reduced infiltration within 24 hours of administration; importantly, repeated doses did not compromise the vascular integrity of the blood-ocular barrier. On withdrawal of fingolimod, drug-induced remission was lost and recrudescence of clinical disease was observed. These results support a strong therapeutic potential for fingolimod as an acute rescue therapy for the treatment of ocular immune-mediated inflammation.


Assuntos
Doenças Autoimunes/prevenção & controle , Barreira Hematorretiniana/efeitos dos fármacos , Imunossupressores/farmacologia , Propilenoglicóis/farmacologia , Retinite/prevenção & controle , Esfingosina/análogos & derivados , Uveíte/prevenção & controle , Animais , Apoptose , Doenças Autoimunes/patologia , Barreira Hematorretiniana/patologia , Relação Dose-Resposta a Droga , Feminino , Cloridrato de Fingolimode , Imunossupressores/administração & dosagem , Camundongos , Microvasos/patologia , Permeabilidade , Propilenoglicóis/administração & dosagem , Retinite/patologia , Esfingosina/administração & dosagem , Esfingosina/farmacologia , Uveíte/patologia
9.
Exp Eye Res ; 105: 43-52, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23059401

RESUMO

The efficacy and action mechanism of everolimus in the treatment of experimental autoimmune uveoretinitis (EAU) was analyzed. Disease was induced in B10.RIII mice by immunization with human interphotoreceptor-retinoid-binding protein peptide 161-180 (hIRBPp161-180). Everolimus was administered by oral gavage (5 mg/kg/d) beginning either two days before or 14 days after immunization. Everolimus significantly reduced the histopathological uveitis score compared to sham-treated mice. To examine the effect on the antigen-specific immune response, proliferation ([(3)H]-thymidine test) and delayed-type hypersensitivity (DTH) response were measured. Furthermore, content of T-helper-1, -2, and -17 cytokines were analyzed intraocularly (Bead Array) and in cell culture supernatants from splenocytes (sandwich ELISA). To study the effect on the humoral immune response the presence of antigen-specific serum antibodies was tested (indirect ELISA). The DTH, the humoral immune response, the proliferation of splenocytes and the intraocular Th1, Th2, Th17 cytokine content and in vitro production of Th1 and Th17 cytokines were impaired after everolimus treatment. The study of CD4+CD25+FoxP3+ regulatory T cells (T(reg)) in peripheral blood, draining lymph nodes, and spleen by flow cytometry showed an increased number of splenic T(reg) in mice of the everolimus therapy group. Furthermore the T(reg) of these mice had a higher suppressive capacity than cells from sham-treated mice. These results indicate that the immunosuppressive effect of everolimus on EAU was associated with the suppression of pathogenic effector responses and induction of regulatory T cells.


Assuntos
Doenças Autoimunes/prevenção & controle , Modelos Animais de Doenças , Imunossupressores/uso terapêutico , Retinite/prevenção & controle , Sirolimo/análogos & derivados , Uveíte Posterior/prevenção & controle , Animais , Anticorpos/sangue , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Proliferação de Células , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Everolimo , Proteínas do Olho/imunologia , Citometria de Fluxo , Fatores de Transcrição Forkhead/metabolismo , Hipersensibilidade Tardia/tratamento farmacológico , Hipersensibilidade Tardia/imunologia , Camundongos , Retinite/imunologia , Retinite/patologia , Proteínas de Ligação ao Retinol/imunologia , Sirolimo/uso terapêutico , Baço/citologia , Linfócitos T Reguladores/imunologia , Uveíte Posterior/imunologia , Uveíte Posterior/patologia
10.
Sci Rep ; 11(1): 9677, 2021 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-33958662

RESUMO

Free fatty acid dysregulation in diabetics may elicit the release of inflammatory cytokines from Müller cells (MC), promoting the onset and progression of diabetic retinopathy (DR). Palmitic acid (PA) is elevated in the sera of diabetics and stimulates the production of the DR-relevant cytokines by MC, including IL-1ß, which induces the production of itself and other inflammatory cytokines in the retina as well. In this study we propose that experimental elevation of cytochrome P450 epoxygenase (CYP)-derived epoxygenated fatty acids, epoxyeicosatrienoic acid (EET) and epoxydocosapentaenoic acid (EDP), will reduce PA- and IL-1ß-induced MC inflammation. Broad-spectrum CYP inhibition by SKF-525a increased MC expression of inflammatory cytokines. Exogenous 11,12-EET and 19,20-EDP significantly decreased PA- and IL-1ß-induced MC expression of IL-1ß and IL-6. Both epoxygenated fatty acids significantly decreased IL-8 expression in IL-1ß-induced MC and TNFα in PA-induced MC. Interestingly, 11,12-EET and 19,20-EDP significantly increased TNFα in IL-1ß-treated MC. GSK2256294, a soluble epoxide hydrolase (sEH) inhibitor, significantly reduced PA- and IL-1ß-stimulated MC cytokine expression. 11,12-EET and 19,20-EDP were also found to decrease PA- and IL-1ß-induced NFκB-dependent transcriptional activity. These data suggest that experimental elevation of 11,12-EET and 19,20-EDP decreases MC inflammation in part by blocking NFκB-dependent transcription and may represent a viable therapeutic strategy for inhibition of early retinal inflammation in DR.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Células Ependimogliais/metabolismo , Epóxido Hidrolases/metabolismo , Ácidos Graxos/metabolismo , Neuroglia/patologia , Retinite/prevenção & controle , Células Cultivadas , Cicloexilaminas/farmacologia , Retinopatia Diabética/complicações , Células Ependimogliais/patologia , Epóxido Hidrolases/antagonistas & inibidores , Humanos , Mediadores da Inflamação/metabolismo , NF-kappa B/genética , Regiões Promotoras Genéticas , Retinite/complicações , Retinite/patologia , Triazinas/farmacologia
11.
Exp Eye Res ; 90(1): 41-8, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19766630

RESUMO

Osteopontin (OPN) is elevated during the progression of experimental autoimmune uveoretinitis (EAU) in C57BL/6 (B6) mice. Furthermore, EAU symptoms are ameliorated in OPN knockout mice or in B6 mice treated with anti-OPN antibody (M5). Recently, OPN has been shown to promote the Th1 response not only in the extracellular space as a secretory protein but also in cytosol as a signaling component. Thus, we attempted to reduce OPN in both compartments by using a small interfering RNA (siRNA) targeting the OPN coding sequence (OPN-siRNA). EAU was induced in B6 mice by immunization with human interphotoreceptor retinoid-binding protein (hIRBP) peptide sequence 1-20. The OPN- or control-siRNA was administered with hydrodynamic methods 24 h before and simultaneously with immunization (prevention regimen). When plasma OPN levels were quantified following siRNA administration with the prevention regimen, the level in the OPN-siRNA-treated group was significantly lower than that in the control-siRNA-treated group. Accordingly, the clinical and histopathological scores of EAU were significantly reduced in B6 mice when siRNA caused OPN blockade. Furthermore, TNF-alpha, IFN-gamma, IL-2, GM-CSF and IL-17 levels in the culture supernatants were markedly suppressed in the OPN-siRNA-treated group, whereas the proliferative responses of T lymphocytes from regional lymph nodes against immunogenic peptides was not significantly reduced. On the other hand, the protection was not significant if the mice received the OPN-siRNA treatment on day 7 and day 8 after immunization when the clinical symptoms appeared overt (reversal regimen). Our results suggest that OPN blockade with OPN-siRNA can be an alternative choice for the usage of anti-OPN antibody and controlling uveoretinitis in the preventive regimen.


Assuntos
Doenças Autoimunes/prevenção & controle , Osteopontina/antagonistas & inibidores , RNA Interferente Pequeno/farmacologia , Retinite/prevenção & controle , Uveíte/prevenção & controle , Animais , Doenças Autoimunes/sangue , Doenças Autoimunes/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Osteopontina/sangue , Interferência de RNA , Retinite/sangue , Retinite/imunologia , Linfócitos T/imunologia , Uveíte/sangue , Uveíte/imunologia
12.
Exp Eye Res ; 91(2): 162-70, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20420831

RESUMO

The aim of this study was to investigate the effect of anti-mouse IL-6 receptor monoclonal antibody (MR16-1) treatment on CD4 T cell differentiation and compared it to the effect of anti-TNF mAb treatment with using a murine model of experimental autoimmune uveoretinitis (EAU). C57BL/6 mice were immunized with interphotoreceptor retinoid-binding protein (IRBP) to induce ocular inflammation treatment with control IgG or MR16-1 or anti-TNF mAb. Helper T cells differentiation was analyzed during the development of EAU. Immunization with IRBP increased the frequency of Th17 cells rather than Th1 cells in the early stage of EAU. Treatment with MR16-1 on the same day as immunization (day 0) or one day after (day 1) suppressed ocular inflammation in EAU mice. Treatment with MR16-1 on day 0 inhibited the induction of Th17 cells in vivo, and inhibited not only IRBP-responsive Th17 cells but also their Th1 counterparts and induced IRBP-responsive regulatory T (Treg) cells in vitro. The administration of anti-TNF mAb had no significant protective effect in EAU mice. The protective effect of anti-IL-6R mAb treatment, but not anti-TNF mAb treatment on EAU correlated with the inhibition of Th17 differentiation. This finding suggests that IL-6 blockade may have a therapeutic effect on human ocular inflammation which is mediated via mechanisms distinct from those of TNF blockade. IL-6 blockade may thus represent an alternative therapy for patients with ocular inflammation who are refractory to anti-TNF mAb therapy.


Assuntos
Anticorpos Monoclonais/farmacologia , Doenças Autoimunes/prevenção & controle , Interleucina-6/metabolismo , Retinite/prevenção & controle , Transdução de Sinais/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/imunologia , Uveíte/prevenção & controle , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Proteínas do Olho , Feminino , Interleucina-17/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Interleucina-6/imunologia , Retinite/imunologia , Retinite/patologia , Proteínas de Ligação ao Retinol , Fator de Necrose Tumoral alfa/imunologia , Uveíte/imunologia , Uveíte/patologia
13.
Dermatol Ther ; 23(1): 31-47, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20136907

RESUMO

Exposure to ultraviolet (UV) radiation is associated with a variety of harmful effects ranging from photoaging to skin cancer. UVB (290 to 320 nm) directly damages the cellular DNA leading to the formation of the 6-4 cyclobutane pyrimidine dimmers, and UVA (320 to 400 nm) indirectly damages the DNA via the production of oxygen radical species. In this review, we focused on the technological and scientific aspects of photoprotection using sunglasses and clothing while attempting to dispel some of the misconceptions. In addition to the basic knowledge relating to sunscreens, we reviewed the current guidelines for testing and labeling UVA protection around the world, controversies associated with nanoparticles, and future sunscreens actives waiting for the Food and Drug Administration approval. Lastly, we reviewed alternative agents, such as antioxidants, that can be used to supplement and augment photoprotection provided by sunscreens.


Assuntos
Vestuário , Dispositivos de Proteção dos Olhos , Proteção Radiológica/métodos , Pele/efeitos da radiação , Protetores Solares/administração & dosagem , Raios Ultravioleta/efeitos adversos , Animais , Catarata/etiologia , Catarata/prevenção & controle , Humanos , Ceratoconjuntivite/etiologia , Ceratoconjuntivite/prevenção & controle , Degeneração Macular/etiologia , Degeneração Macular/prevenção & controle , Camundongos , Nanopartículas , Pterígio/etiologia , Pterígio/prevenção & controle , Proteção Radiológica/instrumentação , Retinite/etiologia , Retinite/prevenção & controle , Estados Unidos
14.
Graefes Arch Clin Exp Ophthalmol ; 248(2): 277-81, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19821164

RESUMO

BACKGROUND: Bacterial endophthalmitis is a serious complication of penetrating ocular trauma and cataract surgery. The purpose of this study is to assess the ability of an anterior chamber air bubble to prevent experimental Staphylococcus epidermidis endophthalmitis. METHODS: The anterior chamber was opened with a clear corneal incision to drain the aqueous humor in one eye of 24 rabbits. S. epidermidis was injected into the anterior chamber. The infected rabbits were randomly divided into two groups. Animals in group 1 (n = 12) received an air bubble to reconstruct the anterior chamber, and animals in Group 2 (n = 12) received Balanced Salt Solution (BSS) for reconstruction. Clinical examination in each group was performed 2, 3 and 7 days after inoculation of the bacteria. Clinical examination scores were recorded. All eyes were enucleated for histophatological examination. RESULTS: Clinical endophthalmitis was seen in 7/12 (58.3%) in group 2, but in only 1/12 (8.3%) in group 1 (p = 0.027). Mean clinical scores of groups 1and 2 on the 3rd day were 2.6 +/- 1.4 and 7.2 +/- 1.7 respectively. (p < 0.001). The mean clinical scores of groups 1and 2 on the 7th day were 2.3 +/- 1.2 and 6.8 +/- 1.5 respectively (p < 0.001). Histopathological examination confirmed clinical findings. CONCLUSION: An air bubble in the anterior chamber may have protective effects against the development of experimental S. epidermidis endophthalmitis.


Assuntos
Ar , Câmara Anterior/microbiologia , Endoftalmite/prevenção & controle , Retinite/prevenção & controle , Infecções Estafilocócicas/prevenção & controle , Staphylococcus epidermidis , Animais , Modelos Animais de Doenças , Endoftalmite/microbiologia , Endoftalmite/patologia , Coelhos , Retina/patologia , Retinite/microbiologia , Retinite/patologia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia
15.
Cell Rep ; 30(11): 3806-3820.e6, 2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-32187551

RESUMO

The cross-talk between mesenchymal stem and stromal cells (MSCs) and macrophages is critical for the restoration of tissue homeostasis after injury. Here, we demonstrate a pathway through which MSCs instruct macrophages to resolve inflammation and preserve tissue-specific stem cells, leading to homeostasis in mice with autoimmune uveoretinitis and sterile-injury-induced corneal epithelial stem cell deficiency. Distinct from their conventional role in macrophage reprogramming to anti-inflammatory phenotype by a PGE2-dependent mechanism, MSCs enhance the phagocytic activity of macrophages, which partly depends on the uptake of MSC mitochondria-containing extracellular vesicles. The MSC-primed macrophages increase the secretion of amphiregulin (AREG) in a phagocytosis-dependent manner. AREG is essential for MSC-primed macrophages to suppress immune responses through regulatory T (Treg) cells and to protect corneal epithelial stem cells via apoptosis inhibition and proliferation promotion. Hence, the data reveal that MSCs harness macrophage-derived AREG to maintain tissue homeostasis after injury and provide a therapeutic target in immune-mediated disease and regenerative medicine.


Assuntos
Anfirregulina/metabolismo , Homeostase , Macrófagos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Especificidade de Órgãos , Animais , Anti-Inflamatórios/metabolismo , Doenças Autoimunes/prevenção & controle , Polaridade Celular , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Epitélio Corneano/citologia , Receptores ErbB/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Macrófagos/citologia , Camundongos , Mitocôndrias/metabolismo , Fagocitose , Fenótipo , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Retinite/prevenção & controle , Transdução de Sinais , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Transcrição Gênica
16.
Cell Death Dis ; 11(2): 101, 2020 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-32029703

RESUMO

N-Methyl-D-aspartate (NMDA)-induced neuronal cell death is involved in a large spectrum of diseases affecting the brain and the retina such as Alzheimer's disease and diabetic retinopathy. Associated neurological impairments may result from the inhibition of neuronal plasticity by Nogo-A. The objective of the current study was to determine the contribution of Nogo-A to NMDA excitotoxicity in the mouse retina. We observed that Nogo-A is upregulated in the mouse vitreous during NMDA-induced inflammation. Intraocular injection of a function-blocking antibody specific to Nogo-A (11C7) was carried out 2 days after NMDA-induced injury. This treatment significantly enhanced visual function recovery in injured animals. Strikingly, the expression of potent pro-inflammatory molecules was downregulated by 11C7, among which TNFα was the most durably decreased cytokine in microglia/macrophages. Additional analyses suggest that TNFα downregulation may stem from cofilin inactivation in microglia/macrophages. 11C7 also limited gliosis presumably via P.Stat3 downregulation. Diabetic retinopathy was associated with increased levels of Nogo-A in the eyes of donors. In summary, our results reveal that Nogo-A-targeting antibody can stimulate visual recovery after retinal injury and that Nogo-A is a potent modulator of excitotoxicity-induced neuroinflammation. These data may be used to design treatments against inflammatory eye diseases.


Assuntos
Células Amácrinas/efeitos dos fármacos , Anti-Inflamatórios/farmacologia , Anticorpos Neutralizantes/farmacologia , Proteínas Nogo/antagonistas & inibidores , Células Ganglionares da Retina/efeitos dos fármacos , Retinite/prevenção & controle , Visão Ocular/efeitos dos fármacos , Idoso , Idoso de 80 Anos ou mais , Células Amácrinas/metabolismo , Células Amácrinas/patologia , Animais , Retinopatia Diabética/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/metabolismo , N-Metilaspartato , Plasticidade Neuronal/efeitos dos fármacos , Proteínas Nogo/metabolismo , Fosforilação , Recuperação de Função Fisiológica , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Retinite/induzido quimicamente , Retinite/metabolismo , Retinite/fisiopatologia , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
17.
Graefes Arch Clin Exp Ophthalmol ; 247(6): 755-65, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19125271

RESUMO

BACKGROUND: RNA interference (RNAi) is now being exploited as a powerful tool for gene knockdown. Recently, we had shown that inducible co-stimulator (ICOS) was up-regulated in experimental autoimmune uveoretinitis (EAU). The aim of this study was to investigate whether intravitreal injection of small interfering RNA (siRNA) plasmid, targeting ICOS, suppresses the ongoing experimental autoimmune uveoretinitis (EAU) in rats. METHODS: Oligonucleotide targeting ICOS was cloned into linearized pRNAT-U6.1/Neo eukaryotic expression vector to construct the recombinant plasmid (pRNAT-U6.1/Neo-ICOS). After transfecting activated rat T cells with the recombinant plasmid, ICOS mRNA and protein expression levels were determined by real-time RT-PCR and Western blot analysis respectively. Rats were immunized with IRBP R16 peptide emulsified in complete Freund's adjuvant (CFA) and given an intravitreal injection of pRNAT-U6.1/Neo-ICOS on day 6 after immunization. After 13days of immunization, the ICOS protein expression and CD4(+) ICOS (+) T cells were identified in retinae through Western blot analysis and flow cytometry respectively. Intraocular inflammation was assessed by the scores of the clinical and histological appearances. Delayed-type hypersensitivity (DTH) and lymphocyte proliferation were detected to evaluate the systemic effect of intravitreal injection of pRNAT-U6.1/Neo-ICOS. RESULT: The recombinant plasmid (pRNAT-U6.1/Neo-ICOS) for the ICOS siRNA was successfully constructed. In vitro studies using the recombinant plasmid has showed the down-regulation of ICOS gene expression both at the mRNA and protein levels. Clinical and pathological scores showed that ocular inflammation of pRNAT-U6.1/Neo-ICOS-treated eyes was markedly less than that of vehicle-treated eyes. The expression of ICOS protein and the amount of CD4(+) ICOS(+) T cells in retinae significantly decreased by intravitreal injection of the recombinant plasmid, whereas delayed-type hypersensitivity response and lymphocyte proliferation were not impaired in rats treated with the recombinant plasmid. CONCLUSION: Intravitreal injection of siRNA plasmid targeting ICOS effectively down-regulated the expression of ICOS, and was highly effective in suppressing the ongoing process of EAU without any side-effects on systemic cellular immunity.


Assuntos
Antígenos de Diferenciação de Linfócitos T/genética , Doenças Autoimunes/prevenção & controle , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , RNA Interferente Pequeno/administração & dosagem , Retinite/prevenção & controle , Uveíte/prevenção & controle , Animais , Antígenos de Diferenciação de Linfócitos T/metabolismo , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Western Blotting , Linfócitos T CD4-Positivos/imunologia , Técnicas de Cultura de Células , Feminino , Citometria de Fluxo , Inativação Gênica , Hipersensibilidade Tardia/imunologia , Proteína Coestimuladora de Linfócitos T Induzíveis , Injeções , Ativação Linfocitária , Fragmentos de Peptídeos , Plasmídeos , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Ratos , Ratos Endogâmicos Lew , Retinite/imunologia , Retinite/patologia , Proteínas de Ligação ao Retinol , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Uveíte/imunologia , Uveíte/patologia , Corpo Vítreo
18.
Lasers Med Sci ; 24(4): 653-7, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18566852

RESUMO

The effect of trientine hydrochloride (TRIEN), a copper-selective chelating agent, on retinal inflammation induced by photocoagulation laser treatment was studied. Nine Long-Evans rats were treated with TRIEN (0.5 mmol/kg per day, intraperitoneal injection) for 9 days. On day 8, each animal underwent unilateral photocoagulation laser treatment with an argon dye laser. On day 9, animals were killed and the eyes processed for immunohistochemistry and light microscopy. In the TRIEN-treated group, retinal thickness and number of macrophages (ED-1) were both significantly lower than in the saline-treated, control group exposed to laser photocoagulation. The results support the hypothesis that selective copper chelation prior to laser treatment may inhibit ocular inflammation. The results suggest that pretreatment with a selective copper-chelating compound can minimize retinal inflammation secondary to laser photocoagulation treatment, which may improve overall outcome of photocoagulation treatment for diabetic retinopathy.


Assuntos
Quelantes/farmacologia , Cobre/metabolismo , Fotocoagulação a Laser/métodos , Retinite/prevenção & controle , Trientina/farmacologia , Animais , Retinopatia Diabética/cirurgia , Humanos , Radical Hidroxila/metabolismo , Fotocoagulação a Laser/efeitos adversos , Macrófagos/patologia , Microglia/patologia , Modelos Biológicos , Ratos , Ratos Long-Evans , Retina/metabolismo , Retina/patologia , Retina/cirurgia , Retinite/etiologia , Retinite/metabolismo , Retinite/patologia
20.
Invest Ophthalmol Vis Sci ; 60(1): 294-303, 2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30657854

RESUMO

Purpose: In this study, we aim to investigate whether mesenchymal stem cell (MSC)-derived exosomes (MSC-Exos) could regulate hyperglycemia-induced retinal inflammation by transferring microRNA-126 (miR-126). Methods: MSC-Exos were isolated from the media of human umbilical cord-derived mesenchymal stem cells (hUCMSCs), and this isolation was followed by the transfer of miR-126. MSC-Exos or MSC-Exos overexpressing miR-126 were intravitreally injected into diabetic rats in vivo and were cocultured with high glucose-affected human retinal endothelial cells (HRECs) in vitro. Plasma samples were obtained from the vitreous of rats and from HREC cells after treatment for ELISA assay. Retinal sections were examined using immunohistochemistry. RT-PCR and Western blotting were conducted to assess the levels of high-mobility group box 1 (HMGB1), NLRP3 inflammasome, and NF-κB/P65 in retinas and HRECs. Results: Our results showed that hyperglycemia greatly increased inflammation in diabetic rats or HRECs exposed to high glucose, increasing the levels of caspase-1, interleukin-1ß (IL-1ß) and IL-18. The administration of MSC-Exos could effectively reverse this reaction. Compared to control MSC-Exos, MSC-Exos overexpressing miR-126 more successfully suppressed the HMGB1 signaling pathway and suppressed inflammation in diabetic rats. The administration of miR-126-expressing MSC-Exos significantly reduced high glucose-induced HMGB1 expression and the activity of the NLRP3 inflammasome in HRECs. Conclusions: miR-126 expression in MSC-Exos reduces hyperglycemia-induced retinal inflammation by downregulating the HMGB1 signaling pathway.


Assuntos
Exossomos/metabolismo , Proteína HMGB1/metabolismo , Hiperglicemia/prevenção & controle , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , Retinite/prevenção & controle , Animais , Western Blotting , Caspase 1/metabolismo , Técnicas de Cocultura , Diabetes Mellitus Experimental/etiologia , Ensaio de Imunoadsorção Enzimática , Hiperglicemia/complicações , Hiperglicemia/metabolismo , Imuno-Histoquímica , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Retinite/etiologia , Retinite/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA