Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 136
Filtrar
1.
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
2.
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
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.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
J Diabetes Complications ; 31(10): 1481-1490, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28847447

RESUMO

AIM: Calcium dobesilate (CaD) is beneficial in early stages of diabetic retinopathy (DR), but its mechanisms of action remains to be elucidated. The aim was to investigate the effect of CaD on proinflammatory cytokines and oxidative stress. METHODS: db/db mice were randomly assigned to daily oral treatment with CaD (200mg/kg/day) or vehicle for 15days. Biomarkers of oxidative stress (dihydroethidium, malondialdehyde), NF-κB, and proinflammatory cytokines (IL-1ß, IL-6, IL-8, TNF-α, MCP-1) were examined in the retina by immunohistochemical analysis. Cultures of human retinal endothelial cells (HRECs) were used for complementary experiments. RESULTS: CaD significantly reduced the biomarkers of oxidative stress in the retina of db/db mice. In addition, CaD prevented the increase of NF-κB, IL-6, IL-8, TNF-α and MCP-1 induced by diabetes. CaD inhibited the activation of NF-kß induced by IL-1ß by preventing IKKB-α phosphorylation in HRECs and reduced the upregulation of IL-6 and IL-18 induced by TNF-α in a dose-dependent manner. CONCLUSION: Our results suggest that antioxidant and antiinflammatory effects are crucial in accounting for the effectiveness of CaD for treating DR.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Dobesilato de Cálcio/uso terapêutico , Retinopatia Diabética/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Retina/efeitos dos fármacos , Retinite/prevenção & controle , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Biomarcadores/metabolismo , Dobesilato de Cálcio/farmacologia , Células Cultivadas , Cruzamentos Genéticos , Citocinas/antagonistas & inibidores , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/tratamento farmacológico , Retinopatia Diabética/imunologia , Retinopatia Diabética/metabolismo , Retinopatia Diabética/patologia , Humanos , Quinase I-kappa B/metabolismo , Masculino , Camundongos Mutantes , Camundongos Transgênicos , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Distribuição Aleatória , Retina/imunologia , Retina/metabolismo , Retina/patologia , Retinite/complicações , Retinite/imunologia , Retinite/metabolismo
11.
Front Biosci (Landmark Ed) ; 21(5): 958-72, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27100484

RESUMO

Interphotoreceptor retinoid-binding protein (IRBP) is a classic inducer of experimental autoimmune uveoretinitis (EAU). Although IRBP causes neuronal loss in susceptible animals, resistant animals such as Sprague-Dawley (SPD) rats can benefit from the evoked protective autoimmune responses. The aim of the present study was to analyze the neuroprotective effects of IRBP against light-induced photoreceptor degeneration. We immunized 75 male SPD rats with IRBP and the rats were then exposed to blue light for 24 hours (IRBP group). Seventy five rats were included in the control group. We found that the number of apoptotic cells in the outer nuclear layer (ONL) peaked on 1 day after light exposure, and the ONL thickness decreased significantly on day 3. OX42-positive cells appeared in the ONL immediately after light exposure, and their number peaked on day 3, and changed from resting ramified cells to activated amoeboid cells. Compared with the control group (n=75), the IRBP group showed less apoptotic cells, a thicker ONL, and reduced expression of tumor necrosis factor-alpha. These outcomes indicate the IRPB might protect retinal photoreceptors against light-induced injury.


Assuntos
Luz/efeitos adversos , Fragmentos de Peptídeos/imunologia , Células Fotorreceptoras de Vertebrados/imunologia , Células Fotorreceptoras de Vertebrados/efeitos da radiação , Proteínas de Ligação ao Retinol/imunologia , Animais , Apoptose/imunologia , Apoptose/efeitos da radiação , Doenças Autoimunes/etiologia , Doenças Autoimunes/prevenção & controle , Modelos Animais de Doenças , Masculino , Microglia/imunologia , Microglia/patologia , Microglia/efeitos da radiação , Fármacos Neuroprotetores/imunologia , Células Fotorreceptoras de Vertebrados/patologia , Ratos , Ratos Sprague-Dawley , Retinite/etiologia , Retinite/prevenção & controle , Fator de Necrose Tumoral alfa/metabolismo , Uveíte/etiologia , Uveíte/prevenção & controle , Vacinação
12.
PLoS One ; 10(8): e0135126, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26273824

RESUMO

The aim of this preliminary study was to determine the effectiveness of short pulse duration, high-power laser photocoagulation (PC) during vitrectomy for diabetic retinopathy (DR).The effects of short pulse duration PC with power of 340-360 mW and duration of 0.02 second were compared to conventional PC with power of 120-150 mW and duration of 0.2 second. The degree of inflammation was quantified by laser flare cell photometry before and at 1 day, 1 week, 4 week, and 12 weeks postoperatively. Twenty-two eyes of 22 consecutive patients were studied. Ten eyes were treated with short pulse duration PC and 12 eyes with conventional PC. The total energy was significantly lower in the short pulse duration PC group than in the conventional PC group (P = 0.007). The flare cell values were not significantly different between the two groups after 1 day, but at 1 week, the flare cell value was significantly lower in the short pulse duration PC group than in the conventional PC group (P = 0.04). This difference was also present at 4 and 12 weeks (P<0.05). The significant lower inflammation after short pulse duration PC than conventional PC indicates that the short pulse duration PC protocol should be considered to treat DR.


Assuntos
Retinopatia Diabética/cirurgia , Inflamação/prevenção & controle , Fotocoagulação a Laser/métodos , Complicações Pós-Operatórias/prevenção & controle , Vitrectomia/efeitos adversos , Idoso , Humanos , Inflamação/epidemiologia , Fotocoagulação a Laser/reabilitação , Pessoa de Meia-Idade , Complicações Pós-Operatórias/epidemiologia , Período Pós-Operatório , Retinite/epidemiologia , Retinite/prevenção & controle , Fatores de Tempo , Resultado do Tratamento , Acuidade Visual , Vitrectomia/métodos , Vitrectomia/reabilitação
13.
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
14.
PLoS One ; 10(4): e0122093, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25874928

RESUMO

Macrophage activation is, in part, regulated via hydrolysis of oxidised low density lipoproteins by Lipoprotein-Associated phospholipase A2 (Lp-PLA2), resulting in increased macrophage migration, pro-inflammatory cytokine release and chemokine expression. In uveitis, tissue damage is mediated as a result of macrophage activation; hence inhibition of Lp-PLA2 may limit macrophage activation and protect the tissue. Utilising Lp-PLA2 gene-deficient (KO) mice and a pharmacological inhibitor of Lp-PLA2 (SB-435495) we aimed to determine the effect of Lp-PLA2 suppression in mediating retinal protection in a model of autoimmune retinal inflammation, experimental autoimmune uveoretinitis (EAU). Following immunisation with RBP-3 (IRBP) 1-20 or 161-180 peptides, clinical disease was monitored and severity assessed, infiltrating leukocytes were enumerated by flow cytometry and tissue destruction quantified by histology. Despite ablation of Lp-PLA2 enzyme activity in Lp-PLA2 KO mice or wild-type mice treated with SB-435495, the number of infiltrating CD45+ cells in the retina was equivalent to control EAU animals, and there was no reduction in disease severity. Thus, despite the reported beneficial effects of therapeutic Lp-PLA2 depletion in a variety of vascular inflammatory conditions, we were unable to attenuate disease, show delayed disease onset or prevent progression of EAU in Lp-PLA2 KO mice. Although EAU exhibits inflammatory vasculopathy there is no overt defect in lipid metabolism and given the lack of effect following Lp-PLA2 suppression, these data support the hypothesis that sub-acute autoimmune inflammatory disease progresses independently of Lp-PLA2 activity.


Assuntos
1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Doenças Autoimunes/metabolismo , Retinite/metabolismo , Uveíte/metabolismo , 1-Alquil-2-acetilglicerofosfocolina Esterase/antagonistas & inibidores , 1-Alquil-2-acetilglicerofosfocolina Esterase/genética , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/prevenção & controle , Compostos de Bifenilo/farmacologia , Células Cultivadas , Modelos Animais de Doenças , Expressão Gênica/genética , Imunização , Leucócitos/efeitos dos fármacos , Leucócitos/metabolismo , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Peptídeos/imunologia , Fosfolipases A2/genética , Fosfolipases A2/metabolismo , Pirimidinonas/farmacologia , Retinite/genética , Retinite/prevenção & controle , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Uveíte/genética , Uveíte/prevenção & controle
15.
Cir Cir ; 82(5): 477-88, 2014.
Artigo em Espanhol | MEDLINE | ID: mdl-25259426

RESUMO

BACKGROUND: Photocoagulation reduces the incidence of visual loss in diabetic patients with focal macular edema, but it can induce it for Efficacy of topical ketorolac for improving visual function after photocoagulation in diabetic patients with focal macular edema 6 weeks after treatment and produces visual improvement in some cases. Topical ketorolac may reduce the inflammation caused by photocoagulation and improve visual outcome. PURPOSE: To determine the efficacy of topical ketorolac for improving visual function after photocoagulation in diabetic patients with focal macular edema. METHODS: An experimental, comparative, prospective, longitudinal study in diabetic patients with focal macular edema was conducted. Eyes were randomized into two groups of topical treatment for 3 weeks after photocoagulation (A: ketorolac, B: placebo). Best corrected visual acuity before and after treatment was compared in each group (paired t test), and the proportion of eyes with visual improvement was compared between groups (χ(2)). The evaluation was repeated after stratifying for initial visual acuity (≥ 0.5, < 0.5). RESULTS: There were 105 eyes included. In group A (n= 46) mean visual acuity changed from 0.50 to 0.58 (p= 0.003), and from 0.55 to 0.55 in group B (n= 59, p= 0.83); mean percent change was 22.3% in group A and 3.5% in group B (p= 0.03). Visual improvement was identified in 25 eyes from group A (54.3%) and 19 from group B (32.2%, p= 0.019, RR 1.65); the difference only persisted when initial visual acuity was ≥ 0.5 (10 [40%], group A, 5 [14.7%], group B, p= 0.02, RR 2.72). CONCLUSION: Topical ketorolac was more effective than placebo to improve best corrected visual acuity in diabetic patients with focal macular edema.


Antecedentes: la fotocoagulación reduce la incidencia de pérdida visual en diabéticos con edema macular focal, aunque puede inducirla durante 6 semanas; la mejoría visual después del tratamiento es excepcional. El ketorolaco tópico puede limitar la inflamación causada por la fotocoagulación, y mejorar el desenlace visual. Objetivo: determinar la eficacia del ketorolaco tópico en la mejoría de la función visual después de la fotocoagulación, en diabéticos con edema macular focal. Material y métodos: estudio experimental, comparativo, prospectivo, longitudinal efectuado en diabéticos con edema macular focal, asignados al azar a dos grupos de tratamiento tópico durante 3 semanas después de la fotocoagulación (A: ketorolaco, B: placebo). En cada grupo se comparó la agudeza visual antes y después del tratamiento (t pareada) y entre grupos la proporción de ojos con mejoría visual (χ2). La evaluación se repitió con estratificación por agudeza visual inicial (≥ 0.5, < 0.5). Resultados: se analizaron 105 ojos; en el grupo A (n= 46) el promedio de agudeza visual cambió de 0.50 a 0.58 (p= 0.003), en el B (n= 59) de 0.55 a 0.55 (p= 0.83); el promedio del cambio porcentual fue 22.3% en el grupo A y 3.5% en el B (p= 0.03). Hubo mejoría visual en 25 ojos del grupo A (54.3%) y 19 del B (32.2%, p= 0.019, RR 1.65); la diferencia persistió cuando la agudeza visual inicial era ≥ 0.5 (10 [40%], grupo A, 5 [14.7%], grupo B, p= 0.02, RR 2.72). Conclusiones: el ketorolaco fue más eficaz que el placebo para mejorar la agudeza visual en pacientes diabéticos con edema macular focal.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Retinopatia Diabética/cirurgia , Cetorolaco/uso terapêutico , Fotocoagulação/efeitos adversos , Edema Macular/cirurgia , Complicações Pós-Operatórias/prevenção & controle , Retinite/prevenção & controle , Administração Oftálmica , Adulto , Idoso , Idoso de 80 Anos ou mais , Anti-Inflamatórios não Esteroides/administração & dosagem , Diabetes Mellitus Tipo 2/complicações , Dinoprostona/antagonistas & inibidores , Dinoprostona/biossíntese , Feminino , Humanos , Cetorolaco/administração & dosagem , Macula Lutea/ultraestrutura , Masculino , Pessoa de Meia-Idade , Tamanho do Órgão/efeitos dos fármacos , Complicações Pós-Operatórias/tratamento farmacológico , Complicações Pós-Operatórias/etiologia , Estudos Prospectivos , Retinite/tratamento farmacológico , Retinite/etiologia , Tomografia de Coerência Óptica , Resultado do Tratamento , Acuidade Visual/efeitos dos fármacos
16.
Biomed Res Int ; 2014: 250323, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24800216

RESUMO

The application of perfluorocarbon liquids has been well acclaimed in vitreoretinal surgery. Its unique physical properties make it an ideal intraoperative tool to improve the efficiency and safety of surgical procedures in complicated cases. The main functions of perfluorocarbon liquids in vitreoretinal surgery include relocating and fixing the detached retina, displacing the subretinal and subchoroidal to fluid anteriorly, revealing proliferative vitreous retinopathy (PVR) for further maneuvers, protecting the macula from exposure to chemicals with potential toxicity, and assisting the removal of foreign body. The related clinical applications include retinal detachment with severe proliferative vitreoretinopathy, giant tear, diabetic retinopathy (DR), retinopathy of prematurity (ROP), and posterior dislocated crystalline and intraocular lenses. The application of perfluorocarbon liquids has been expended over the past fewer years. Several PFCLs related ocular inflammations have been observed in in vitro studies, animal studies, and clinical follow-up. The complete removal of PFCLs is recommended at the end of the surgery in most cases.


Assuntos
Fluorocarbonos/administração & dosagem , Pré-Medicação/métodos , Descolamento Retiniano/terapia , Retinite/tratamento farmacológico , Retinite/prevenção & controle , Cirurgia Vitreorretiniana/métodos , Humanos , Soluções Oftálmicas/administração & dosagem , Retinite/etiologia , Soluções , Resultado do Tratamento , Cirurgia Vitreorretiniana/efeitos adversos
18.
J Neuroimmunol ; 263(1-2): 98-107, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-24029580

RESUMO

We report here identification of novel mimicry epitopes for interphotoreceptor retinoid-binding protein (IRBP) 201-216, a candidate ocular antigen that causes experimental autoimmune uveoretinitis (EAU) in A/J mice. One mimicry epitope from Ehrlichia canis (EHC), designated EHC 44-59, induced cross-reactive T cells for IRBP 201-216 capable of producing T helper (Th)1 and Th17 cytokines, but failed to induce EAU in A/J mice. In addition, animals first primed with suboptimal doses of IRBP 201-216 and subsequently immunized with EHC 44-59 did not develop EAU; rather, the mimicry epitope prevented the disease induced by IRBP 201-216. However, alteration in the composition of EHC 44-59 by substituting alanine with valine at position 49, similar to the composition of IRBP 201-216, enabled the mimicry epitope to acquire uveitogenicity. The data provide new insights as to how microbes containing mimicry sequences for retinal antigens can prevent ocular inflammation by acting as naturally occurring altered peptide ligands.


Assuntos
Doenças Autoimunes do Sistema Nervoso/prevenção & controle , Ehrlichia canis/imunologia , Ehrlichiose/prevenção & controle , Mimetismo Molecular/imunologia , Retinite/prevenção & controle , Uveíte/prevenção & controle , Sequência de Aminoácidos , Animais , Doenças Autoimunes do Sistema Nervoso/imunologia , Doenças Autoimunes do Sistema Nervoso/microbiologia , Bovinos , Ehrlichia canis/genética , Ehrlichiose/imunologia , Ehrlichiose/microbiologia , Proteínas do Olho/administração & dosagem , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Feminino , Ligantes , Camundongos , Camundongos Endogâmicos A , Dados de Sequência Molecular , Retinite/imunologia , Retinite/microbiologia , Proteínas de Ligação ao Retinol/administração & dosagem , Proteínas de Ligação ao Retinol/genética , Proteínas de Ligação ao Retinol/metabolismo , Uveíte/imunologia , Uveíte/microbiologia
19.
Invest Ophthalmol Vis Sci ; 54(1): 39-46, 2013 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-23211816

RESUMO

PURPOSE: To investigate the effect of systemic or local TNF-α inhibition with etanercept on experimental autoimmune uveoretinitis (EAU). METHODS: EAU was induced by immunizing B10.RIII mice with IRBPp161-180 or by adoptively transferring uveitogenic splenocytes. Mice received systemic or local treatment with etanercept in the afferent or efferent phase. For systemic treatment, mice were injected intraperitoneally. For local treatment, etanercept was injected intravitreally or subconjunctivally. Control mice received PBS. EAU scores were determined histologically. Splenic cells were assessed for [(3)H]thymidine incorporation. ELISA was performed to measure levels of cytokines produced by splenocytes. Vitreous cavity-associated immune deviation (VCAID) was induced by intravitreally injecting ovalbumin and evaluated by measuring DTH reaction. RESULTS: After systemic treatment with etanercept in the afferent phase, EAU disease scores, IRBP-specific cell proliferation, and production of Th1, Th2, and Th17 cytokines were reduced. EAU also improved after intravitreal etanercept treatment in the afferent phase, with unaltered IRBP-specific proliferation, reduced IFN-γ, but increased IL-6 and IL-10 secretion. VCAID induction was impaired after intravitreal etanercept treatment. No amelioration of EAU or reduction in IRBP-specific cell response was found after systemic or intravitreal treatment in the efferent phase or after subconjunctival treatment. After adoptive transfer, etanercept- and PBS-treated recipients showed similar disease severity and antigen-specific proliferation of splenocytes. CONCLUSIONS: It can be concluded that TNF-α participates mainly in the immunopathology in the induction phase of EAU. The mechanism of action underlying EAU improvement may be different for local and systemic etanercept treatment.


Assuntos
Doenças Autoimunes/prevenção & controle , Modelos Animais de Doenças , Imunoglobulina G/farmacologia , Imunossupressores/farmacologia , Retinite/prevenção & controle , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Uveíte Posterior/prevenção & controle , Transferência Adotiva , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Proliferação de Células , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Etanercepte , Proteínas do Olho , Imunoglobulina G/administração & dosagem , Imunossupressores/administração & dosagem , Injeções Intraperitoneais , Injeções Intravítreas , Camundongos , Ovalbumina , Receptores do Fator de Necrose Tumoral/administração & dosagem , Retinite/imunologia , Retinite/patologia , Proteínas de Ligação ao Retinol , Linfócitos T Auxiliares-Indutores/imunologia , Uveíte Posterior/imunologia , Uveíte Posterior/patologia , Corpo Vítreo/metabolismo
20.
PLoS One ; 7(9): e46479, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23029526

RESUMO

PURPOSE: Ischemia and reperfusion injury (I/R) of neuronal structures and organs is associated with increased morbidity and mortality due to neuronal cell death. We hypothesized that inhalation of carbon monoxide (CO) after I/R injury ('postconditioning') would protect retinal ganglion cells (RGC). METHODS: Retinal I/R injury was performed in Sprague-Dawley rats (n = 8) by increasing ocular pressure (120 mmHg, 1 h). Rats inhaled room air or CO (250 ppm) for 1 h immediately following ischemia or with 1.5 and 3 h latency. Retinal tissue was harvested to analyze Bcl-2, Bax, Caspase-3, HO-1 expression and phosphorylation of the nuclear transcription factor (NF)-κB, p38 and ERK-1/2 MAPK. NF-κB activation was determined and inhibition of ERK-1/2 was performed using PD98059 (2 mg/kg). Densities of fluorogold prelabeled RGC were analyzed 7 days after injury. Microglia, macrophage and Müller cell activation and proliferation were evaluated by Iba-1, GFAP and Ki-67 staining. RESULTS: Inhalation of CO after I/R inhibited Bax and Caspase-3 expression (Bax: 1.9 ± 0.3 vs. 1.4 ± 0.2, p = 0.028; caspase-3: 2.0 ± 0.2 vs. 1.5 ± 0.1, p = 0.007; mean ± S.D., fold induction at 12 h), while expression of Bcl-2 was induced (1.2 ± 0.2 vs. 1.6 ± 0.2, p = 0.001; mean ± S.D., fold induction at 12 h). CO postconditioning suppressed retinal p38 phosphorylation (p = 0.023 at 24 h) and induced the phosphorylation of ERK-1/2 (p<0.001 at 24 h). CO postconditioning inhibited the expression of HO-1. The activation of NF-κB, microglia and Müller cells was potently inhibited by CO as well as immigration of proliferative microglia and macrophages into the retina. CO protected I/R-injured RGC with a therapeutic window at least up to 3 h (n = 8; RGC/mm(2); mean ± S.D.: 1255 ± 327 I/R only vs. 1956 ± 157 immediate CO treatment, vs. 1830 ± 109 1.5 h time lag and vs. 1626 ± 122 3 h time lag; p<0.001). Inhibition of ERK-1/2 did not counteract the CO effects (RGC/mm(2): 1956 ± 157 vs. 1931 ± 124, mean ± S.D., p = 0.799). CONCLUSION: Inhaled CO, administered after retinal ischemic injury, protects RGC through its strong anti-apoptotic and anti-inflammatory effects.


Assuntos
Anti-Inflamatórios/administração & dosagem , Apoptose/efeitos dos fármacos , Monóxido de Carbono/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Traumatismo por Reperfusão/tratamento farmacológico , Retinite/prevenção & controle , Administração por Inalação , Animais , Caspase 3/genética , Caspase 3/metabolismo , Movimento Celular/efeitos dos fármacos , Ativação Enzimática , Feminino , Expressão Gênica/efeitos dos fármacos , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Isquemia/tratamento farmacológico , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Neuroglia/efeitos dos fármacos , Neuroglia/fisiologia , Fosforilação , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Sprague-Dawley , Retina/efeitos dos fármacos , Retina/patologia , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/patologia , Vasos Retinianos/patologia , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA