Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Biochem Biophys Res Commun ; 530(2): 367-373, 2020 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-32800337

RESUMO

Choroidal neovascularization (CNV) is the hallmark of wet age-related macular degeneration (AMD), a leading cause of irreversible blindness in the modern world. The objective for this study was to investigate the therapeutic potential of known antiangiogenic agents: thalidomide, senicapoc, and sodium butyrate. Dose-dependent effect of the agents on growth of ARPE-19 cells and human umbilical vein endothelial cells (HUVECs) was investigated with cell counting assays. Half-maximal inhibitory concentrations of thalidomide (765 µM and 1520 µM), senicapoc (50 µM and 79 µM), and sodium butyrate (933 µM and 557 µM) were determined for HUVECs and ARPE-19 cells, respectively. Immunofluorescence analysis showed decrease of VEGFA expression in both ARPE-19 cells and HUVECs after treatment only with thalidomide but not with senicapoc or sodium butyrate. Efficacy of the agents was studied in vivo with laser-induced CNV in C57BL/6 mice. Thalidomide (24 µg), senicapoc (4 µg), or sodium butyrate (100 µg) was intravitreally injected the day after CNV induction. Thalidomide, senicapoc, and sodium butyrate inhibited CNV size by 56%, 24%, and 21% respectively on day 7 post-laser. Thalidomide also reduced cobalt chloride induced increase of VEGFA mRNA in ARPE-19 (-33%) and protein in culture medium (-20%). Our results suggest that thalidomide may have more therapeutic potential than senicapoc or sodium butyrate for treatment of CNV or wet AMD.


Assuntos
Acetamidas/uso terapêutico , Inibidores da Angiogênese/uso terapêutico , Ácido Butírico/uso terapêutico , Neovascularização de Coroide/tratamento farmacológico , Talidomida/uso terapêutico , Compostos de Tritil/uso terapêutico , Acetamidas/farmacologia , Inibidores da Angiogênese/farmacologia , Animais , Ácido Butírico/farmacologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Neovascularização de Coroide/patologia , Modelos Animais de Doenças , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos Endogâmicos C57BL , Talidomida/farmacologia , Compostos de Tritil/farmacologia
2.
Graefes Arch Clin Exp Ophthalmol ; 257(5): 953-960, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30719689

RESUMO

PURPOSE: Experimental autoimmune anterior uveitis (EAAU) is a clinically relevant animal model for human idiopathic anterior uveitis (IAU). The role of the immunomodulator transforming growth factor ß2 (TGF-ß2) in EAAU pathology is unknown. In this study, we investigated the regulatory role of TGF-ß2 in EAAU. METHODS: EAAU was induced in male Lewis rats by footpad injection of melanin-associated antigen (MAA). TGF-ß2 was administered intravenously (iv) in MAA-sensitized rats during the induction of EAAU, or after the clinical onset of uveitis. MAA-sensitized rats injected similarly with an equal volume of PBS served as control. Animals were examined daily between days 7 and 30 post-injection for the clinical signs of uveitis using slit lamp biomicroscopy. Animals were sacrificed at various time points and eyes were harvested for histological analysis to assess the course and severity of inflammation. For histopathological analysis, paraffin sections of harvested eyes were stained with hematoxylin and eosin. Popliteal lymph nodes (LNs) were used for CD4+CD25+FoxP3+ T regulatory (Tregs) population analysis and for CD4+ T cell proliferation assay. RESULTS: Administration of recombinant TGF-ß2 during the early stages of EAAU prevented the induction of uveitis. Compared to PBS, the presence of TGF-ß2 in the cell culture significantly (p < 0.05) inhibited the proliferation of CD4+ T cells in response to MAA. In MAA-sensitized Lewis rats, iv treatment with recombinant TGF-ß2 resulted in significantly (p < 0.05) increased percentage of Tregs compared to animals treated similarly with PBS. Thus, TGF-ß2 inhibited the induction of EAAU by inhibiting CD4+ T cell proliferation and increasing the number of Tregs. Injection of TGF-ß2 in rats with active EAAU resulted in diminished disease activity. Unfortunately, this treatment did not lead to the early resolution of EAAU. CONCLUSIONS: TGF-ß2 plays a critical role in regulation of intraocular inflammation in EAAU. Findings reported in this study improve our understanding of immunopathology of IAU and suggest that recombinant TGF-ß2 may be a promising therapeutic agent for human IAU.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Fator de Crescimento Transformador beta2/farmacologia , Uveíte Anterior/tratamento farmacológico , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Proliferação de Células , Modelos Animais de Doenças , Injeções Intraoculares , Masculino , Ratos , Ratos Endogâmicos Lew , Proteínas Recombinantes , Linfócitos T/imunologia , Linfócitos T/patologia , Uveíte Anterior/imunologia , Uveíte Anterior/patologia
3.
Am J Pathol ; 186(8): 2088-2104, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27295359

RESUMO

In the mouse, membrane cofactor protein (CD46), a key regulator of the alternative pathway of the complement system, is only expressed in the eye and on the inner acrosomal membrane of spermatozoa. We noted that although Cd46(-/-) mice have normal systemic alternative pathway activating ability, lack of CD46 leads to dysregulated complement activation in the eye, as evidenced by increased deposition of C5b-9 in the retinal pigment epithelium (RPE) and choroid. A knockout of CD46 induced the following cardinal features of human dry age-related macular degeneration (AMD) in 12-month-old male and female mice: accumulation of autofluorescent material in and hypertrophy of the RPE, dense deposits in and thickening of Bruch's membrane, loss of photoreceptors, cells in subretinal space, and a reduction of choroidal vessels. Collectively, our results demonstrate spontaneous age-related degenerative changes in the retina, RPE, and choroid of Cd46(-/-) mice that are consistent with human dry AMD. These findings provide the exciting possibility of using Cd46(-/-) mice as a convenient and reliable animal model for dry AMD. Having such a relatively straight-forward model for dry AMD should provide valuable insights into pathogenesis and a test model system for novel drug targets. More important, tissue-specific expression of CD46 gives the Cd46(-/-) mouse model of dry AMD a unique advantage over other mouse models using knockout strains.


Assuntos
Modelos Animais de Doenças , Atrofia Geográfica/genética , Degeneração Macular/genética , Proteína Cofatora de Membrana/deficiência , Animais , Western Blotting , Feminino , Atrofia Geográfica/patologia , Degeneração Macular/patologia , Masculino , Proteína Cofatora de Membrana/genética , Camundongos , Camundongos Knockout
4.
Clin Exp Ophthalmol ; 44(7): 610-617, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26861462

RESUMO

BACKGROUND: To characterize the effect of aspirin (ASA) in mouse models of choroidal neovascularization (CNV) and retinal degeneration. METHODS: In vivo: Male C57BL/6 mice were given ASA in food or regular rodent diet. CNV was induced by argon laser photocoagulation. Subretinal injections of polyethylene glycol 400 (PEG-400) were administered to induce retinal degeneration. CNV size, laser spot area and mean intensity of VEGF in the laser injured zones were measured. In the PEG injected eyes the thickness of retinal pigment epithelium (RPE) and choroid was measured. In vitro: Human ARPE-19 cells were treated with 0.5 or 2.0 mM/L of ASA for 72 h. ELISA was used to measure the concentration of VEGF and CCL-2 in the supernatants. Additionally, damaged RPE monolayer was treated with ASA (0.5 or 2.0 mM/L) and vehicle separately. Size of damaged area was measured. ELISA was used to measure secretion of VEGF-A and CCL-2 by damaged cells after 24 h. RESULTS: No statistically significant effect of ASA on CNV size, laser spot size or VEGF expression was noted in CNV model. In the PEG model, ASA did not have any effect on RPE and choroid thickness; however, a significant increase in RPE atrophy was observed (P = 0.02 + 38%). In addition, ASA had a significant effect on the ability of the RPE cells to regenerate and become confluent after mechanical damage. CONCLUSIONS: ASA at doses consumed clinically for various medical causes may not worsen CNV in human subjects. However, ASA may increase RPE atrophy when consumed over long periods of time.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Aspirina/farmacologia , Neovascularização de Coroide/tratamento farmacológico , Modelos Animais de Doenças , Degeneração Retiniana/tratamento farmacológico , Epitélio Pigmentado da Retina/efeitos dos fármacos , Animais , Linhagem Celular , Quimiocina CCL2/metabolismo , Neovascularização de Coroide/metabolismo , Neovascularização de Coroide/patologia , Dieta , Ensaio de Imunoadsorção Enzimática , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Degeneração Retiniana/metabolismo , Degeneração Retiniana/patologia , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Cicatrização/efeitos dos fármacos
5.
Am J Pathol ; 184(9): 2537-48, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25019227

RESUMO

Dysregulation of the complement system is increasingly recognized as a contributing factor in age-related macular degeneration. Although the complement regulator CD46 is expressed ubiquitously in humans, in mouse it was previously thought to be expressed only on spermatozoa. We detected CD46 mRNA and protein in the posterior ocular segment (neuronal retina, retinal pigment epithelium, and choroid) of wild-type (WT) C57BL/6J mice. Cd46(-/-) knockout mice exhibited increased levels of the membrane attack complex and of vascular endothelial growth factor (VEGF) in the retina and choroid. The Cd46(-/-) mice were also more susceptible to laser-induced choroidal neovascularization (CNV). In Cd46(-/-) mice, 19% of laser spots were positive for CNV at day 2 after treatment, but no positive spots were detected in WT mice. At day 3, 42% of laser spots were positive in Cd46(-/-) mice, but only 11% in WT mice. A fully developed CNV complex was noted in both Cd46(-/-) and WT mice at day 7; however, lesion size was significantly (P < 0.05) increased in Cd46(-/-) mice. Our findings provide evidence for expression of CD46 in the mouse eye and a role for CD46 in protection against laser-induced CNV. We propose that the Cd46(-/-) mouse has a greater susceptibility to experimental CNV because of insufficient complement inhibition, which leads to increased membrane attack complex deposition and VEGF expression.


Assuntos
Neovascularização de Coroide/metabolismo , Proteína Cofatora de Membrana/metabolismo , Animais , Western Blotting , Ensaio de Imunoadsorção Enzimática , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Exp Eye Res ; 127: 143-52, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25088354

RESUMO

Age-related macular degeneration (AMD) is a leading cause of irreversible blindness. This study was done to characterize dry AMD-like changes in mouse retinal pigment epithelium (RPE) and retina after polyethylene glycol (PEG) treatment. We injected male C57BL/6 mice subretinally with PBS, 0.025, 0.25, 0.5 and 1.0 mg of PEG-400 and the animals were sacrificed on day 5. Eyes were harvested and processed for histological analysis. In all other experiments 0.5 mg PEG was injected and animals were sacrificed on days 1, 3, 5 or 14. Paraffin, 5 µm and plastic, 1 µm and 80 nm sections were used for further analysis. Subretinal injection of 0.5 mg PEG induced a 32% reduction of outer nuclear layer (ONL) thickness, 61% decrease of photoreceptor outer and inner segment length, 49% decrease of nuclear density in the ONL and 31% increase of RPE cell density by day 5 after injection. The maximum level of TUNEL positive nuclei in the ONL (6.8 + 1.99%) was detected at day 5 after PEG injection and co-localized with Casp3act. Histological signs of apoptosis were observed in the ONL by light or electron microscopy. Degeneration of RPE cells was found in PEG injected eyes. Gene expression data identified several genes reported to be involved in human AMD. C3, Cfi, Serping1, Mmp9, Htra1 and Lpl were up-regulated in PEG injected eyes compared to PBS controls. PEG leads to morphological and gene expression changes in RPE and retina consistent with dry AMD. This model will be useful to investigate dry AMD pathogenesis and treatment.


Assuntos
Modelos Animais de Doenças , Atrofia Geográfica/patologia , Células Fotorreceptoras de Vertebrados/patologia , Polietilenoglicóis/toxicidade , Epitélio Pigmentado da Retina/patologia , Aminopeptidases/genética , Animais , Apoptose , Autofagia , Proteína Inibidora do Complemento C1/genética , Complemento C3/genética , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/fisiologia , Atrofia Geográfica/induzido quimicamente , Atrofia Geográfica/genética , Serina Peptidase 1 de Requerimento de Alta Temperatura A , Marcação In Situ das Extremidades Cortadas , Injeções Intraoculares , Masculino , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Endogâmicos C57BL , Serina Endopeptidases/genética , Fatores de Tempo
7.
Biochim Biophys Acta ; 1823(8): 1264-72, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22633972

RESUMO

Age-related macular degeneration (AMD) is a leading cause of central blindness in the elderly population. The wet type of AMD is characterized by extensive growth of new vessels. One of the effective strategies to treat wet AMD is to limit the choroidal neovascularization (CNV). We studied the effects of adiponectin peptide I (APNpI) on new vessel growth in laser-induced rat model of wet AMD and on rat choroidal endothelial cell (CEC) culture. CNV size and vessel density were investigated by microscopy. Immunohistochemical staining (IHC) for von Willebrand Factor (vWF), APN, APN receptors 1 (AdipoR1), 2 (AdipoR2), VEGF, VEGF receptor 2 (VEGF-R2), proliferating cell nuclear antigen (PCNA) was performed in CNV area. The mRNA expression of VEGF and VEGF-R2 in RPE-choroid was investigated by RT-PCR and real-time PCR. APNpI inhibited area of CNV by 4 fold, number of vWF positive vessels by 99% and area of subretinal tissue by 40%. The expression of VEGF and VEGF-R2 at mRNA and protein levels decreased after APNpI treatment in vivo. Proliferative index (PCNA) was 5 folds less in laser spots of APNpI treated rats compared to controls. In conclusion, APNpI inhibited formation of new vessels in rat model of CNV by decreasing VEGF, VEGF-R2 expression and cell proliferation. Thus, APNpI may have potential therapeutic use for AMD treatment since it significantly inhibited CNV.


Assuntos
Adiponectina/farmacologia , Corioide/irrigação sanguínea , Neovascularização de Coroide/prevenção & controle , Fragmentos de Peptídeos/farmacologia , Degeneração Macular Exsudativa/tratamento farmacológico , Adiponectina/uso terapêutico , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Corioide/efeitos dos fármacos , Corioide/patologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Expressão Gênica/efeitos dos fármacos , Masculino , Fragmentos de Peptídeos/uso terapêutico , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ratos Endogâmicos BN , Receptores de Adiponectina/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
8.
Biomedicines ; 11(11)2023 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-38002042

RESUMO

In recent years, there has been a captivating focus of interest in elucidating the intricate crosstalk between adiponectin (APN), a versatile fat-associated adipokine and ocular pathologies. Unveiling the intricate relationship between adipocytokine APN and its receptors (AdipoRs) with aging eye disorders has emerged as a fascinating frontier in medical research. This review article delves into this connection, illuminating the hidden influence of APN on retinal health. This comprehensive review critically examines the latest findings and breakthroughs that underscore the pivotal roles of APN/AdipoRs signaling in maintaining ocular homeostasis and protecting against eye ailments. Here, we meticulously explore the intriguing mechanisms by which APN protein influences retinal function and overall visual acuity. Drawing from an extensive array of cutting-edge studies, the article highlights APN's multifaceted functions, ranging from anti-inflammatory properties and oxidative stress reduction to angiogenic regulation within retinal and macula tissues. The involvement of APN/AdipoRs in mediating these effects opens up novel avenues for potential therapeutic interventions targeting prevalent aging eye conditions. Moreover, this review unravels the interplay between APN signaling pathways and age-related macular degeneration (AMD). The single-cell RNA-seq results validate the expression of both the receptor isoforms (AdipoR1/R2) in retinal cells. The transcriptomic analysis showed lower expression of AdipoR1/2 in dry AMD pathogenesis compared to healthy subjects. The inhibitory adiponectin peptide (APN1) demonstrated over 75% suppression of CNV, whereas the control peptide did not exert any inhibitory effect on choroidal neovascularization (CNV). The elucidation of these relationships fosters a deeper understanding of adipose tissue's profound influence on ocular health, presenting new prospects for personalized treatments and preventative measures. Because APN1 inhibits CNV and leakage, it can be used to treat human AMD, although the possibility to treat human AMD is in the early stage and more clinical research is needed. In conclusion, this review provides a captivating journey into the enthralling world of APN, intertwining the realms of adipose biology and ophthalmology in aging.

9.
J Biol Chem ; 286(18): 16229-37, 2011 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-21454496

RESUMO

In this study, we describe a new method for inducing choroidal neovascularization (CNV) in C57BL/6 mice, an animal model of wet age-related macular degeneration (AMD). AMD is a disease that causes central blindness in humans. We injected PEG-8 subretinally in different doses (0.125-2 mg) to induce CNV. After PEG-8 injection, we examined CNV at several time points (days 3-42). We also used Western blotting, immunohistochemistry, and ELISA to examine the complement component C3 split products, C9, VEGF, TGF-ß2, and basic FGF. As early as day 1 after treatment, we found that a single subretinal injection of 1 mg of PEG-8 increased the C3 split products and the C9, TGF-ß2, and basic FGF levels in the retinal pigment epithelium-choroid tissue. By day 3 after PEG-8 injection, the intraocular activation of the complement system caused induction and progression of CNV, including new vessels penetrating the Bruch's membrane. At day 5 after PEG-8 injection, we observed a fully developed CNV and retinal degeneration. Thus, in this study, we present a new, inexpensive, and accelerated mouse model of CNV that may be useful to study AMD.


Assuntos
Neovascularização de Coroide/induzido quimicamente , Modelos Animais de Doenças , Portadores de Fármacos/efeitos adversos , Degeneração Macular , Polietilenoglicóis/efeitos adversos , Animais , Neovascularização de Coroide/metabolismo , Neovascularização de Coroide/patologia , Complemento C3/metabolismo , Complemento C9/metabolismo , Portadores de Fármacos/farmacologia , Camundongos , Polietilenoglicóis/farmacologia , Fatores de Tempo , Fator de Crescimento Transformador beta2/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
J Biol Chem ; 286(10): 8472-8480, 2011 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-21216963

RESUMO

The objective of the current study was to delineate the pathway of complement activation that is crucial for the induction of experimental autoimmune anterior uveitis (EAAU). We studied the development of EAAU in melanin-associated antigen (MAA)-sensitized Lewis rats treated with antibody against C4 or factor B. Control animals received isotype IgG control. Antibody against C4 had no effect on EAAU, and all of the animals developed EAAU similar to those injected with control IgG. In contrast, EAAU was completely inhibited in all MAA-sensitized Lewis rats injected with factor B antibody. Treatment with anti-factor B antibody resulted in suppression of ocular complement activation. Adoptive transfer of T lymphocytes harvested from draining lymph nodes of donor animals treated with anti-factor B did not transfer EAAU to naïve syngenic rats. Anti-factor B antibody inhibited the ability of MAA-specific CD4(+) T cells to proliferate (in vitro) in response to MAA in a dose-dependent manner. Level of TNF-α and IFN-γ decreased in the presence of anti-factor B. Collectively, our results provide the novel finding that complement activation via the alternative pathway contributes to intraocular inflammation in EAAU, and anti-factor B-mediated inhibition of EAAU is due to diminished antigen-specific CD4(+) T cell responses to MAA. Our findings explain the interactions between the complement system and T cells that are critical for the induction of EAAU and may lead to the development of therapy for idiopathic anterior uveitis based on selective blockade of the alternative pathway.


Assuntos
Doenças Autoimunes/imunologia , Linfócitos T CD4-Positivos/imunologia , Complemento C4/imunologia , Fator B do Complemento/imunologia , Via Alternativa do Complemento/imunologia , Uveíte Anterior/imunologia , Transferência Adotiva , Animais , Autoanticorpos/imunologia , Autoanticorpos/farmacologia , Doenças Autoimunes/induzido quimicamente , Via Alternativa do Complemento/efeitos dos fármacos , Modelos Animais de Doenças , Imunoglobulina G/imunologia , Imunoglobulina G/farmacologia , Masculino , Melaninas/efeitos adversos , Melaninas/imunologia , Melaninas/farmacologia , Ratos , Ratos Endogâmicos Lew , Uveíte Anterior/induzido quimicamente
11.
J Biol Chem ; 286(23): 20991-1001, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21515678

RESUMO

The present study investigated the interactions among the complement membrane attack complex (MAC), CCL2, and VEGF that occur in vivo during the development of choroidal neovascularization (CNV). We first investigated the sequential expression of MAC, CCL2, and VEGF during laser-induced CNV in C57BL/6 mice. Increased MAC deposition was detected at 1 h, CCL2 increased at 3 h, and VEGF was up-regulated at day 3 post-laser treatment. These results suggested that during laser-induced CNV, MAC, CCL2 and VEGF are formed and/or expressed in the following order: MAC → CCL2 → VEGF. To determine the cross-talk between MAC, CCL2, and VEGF during laser-induced CNV, neutralizing antibodies were injected both systemically and locally to block the bioactivity of each molecule. Blocking MAC formation inhibited CCL2 and VEGF expression and also limited CNV formation, whereas neutralization of CCL2 bioactivity did not affect MAC deposition; however, it reduced VEGF expression and CNV formation. When bioactivity of VEGF was blocked, CNV formation was significantly inhibited, but MAC deposition was not affected. Together, our results demonstrate that MAC is an upstream mediator and effect of MAC on the development of laser-induced CNV can be attributed to its direct effect on VEGF as well as its effect on VEGF that is mediated by CCL2. Understanding the interplay between immune mediators is critical to gain insight into the pathogenesis of CNV.


Assuntos
Quimiocina CCL2/biossíntese , Neovascularização de Coroide/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Regulação da Expressão Gênica , Lasers/efeitos adversos , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Anticorpos Neutralizantes/farmacologia , Neovascularização de Coroide/etiologia , Neovascularização de Coroide/patologia , Modelos Animais de Doenças , Camundongos , Fatores de Tempo
12.
Retina ; 32(6): 1171-80, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22088983

RESUMO

PURPOSE: The purpose of this article is to evaluate the effect of nicotine on anti-vascular endothelial growth factor therapy in the treatment of neovascular age-related macular degeneration. METHODS: One group of mice received nicotine in drinking water and the other group received water only. Choroidal neovascularization (CNV) was induced with a laser. Nicotinic acetylcholine receptor-α7 (nAChRα7) expression was evaluated by immunohistochemistry. Bevacizumab or adiponectin peptide II (APNpII) was injected intravitreally on Day 7 postlaser, and the effects were evaluated on Days 14 and 21. α-Bungarotoxin was injected intraperitoneally on Days 2 to 5, and its effect was evaluated on Day 14. RESULTS: Expression of nAChRα7 was 2 to 7 times higher between Days 3 and 7 postlaser compared with naive mice. In water-fed mice, APNpII, bevacizumab, and α-bungarotoxin significantly reduced CNV size. In nicotine-fed mice, treatment with APNpII or bevacizumab did not significantly reduce CNV size, whereas α-bungarotoxin did have an effect. Comparing water- and nicotine-fed mice, CNV size was 61% to 86% smaller in water-fed mice except for the α-bungarotoxin group, where there was no difference. Platelet-derived growth factor and vascular endothelial growth factor expression was 1.5- to 2.5-fold higher at Day 14 in nicotine-treated mice. CONCLUSION: Nicotine significantly blocks the effect of anti-vascular endothelial growth factor therapy in the treatment of laser-induced neovascular age-related macular degeneration. nAChRα7 is significantly upregulated during the formation of CNV, and treatment with an nAChRα7 antagonist decreases CNV size irrespective of nicotine administration.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Neovascularização de Coroide/tratamento farmacológico , Nicotina/farmacologia , Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Adiponectina/uso terapêutico , Análise de Variância , Animais , Bevacizumab , Bungarotoxinas/farmacologia , Corioide/metabolismo , Neovascularização de Coroide/metabolismo , Neovascularização de Coroide/patologia , Modelos Animais de Doenças , Imuno-Histoquímica , Injeções Intravítreas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Nicotínicos/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor Nicotínico de Acetilcolina alfa7
13.
Biomolecules ; 12(9)2022 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-36139070

RESUMO

Age-related macular degeneration (AMD) is an eye disease that can cause central vision loss, particularly in the elderly population. There are 2 classes of AMD, wet-type and dry-type. Wet-type involves excess angiogenesis around the macula, referred to as choroidal neovascularization (CNV). This can result in leaky vessels, often causing more severe vision loss than dry-type AMD. Adiponectin peptide 1 (APNp1) has been shown to slow the progression of CNV. Here, we used a mouse model and FITC-labeled APNp1 to determine if APNp1 could be delivered effectively as an eye drop. Our experiment revealed that topically applied FITC-APNp1 could reach the macula of the eye, which is crucial for treating wet-type AMD. We also tested delivery of APNp1 via injection of an adeno-associated virus (AAV) vector in a mouse model of CNV. AAV is a harmless virus easy to manipulate and is very often used for protein or peptide deliveries. Results revealed an increase in the expression of APNp1 in the retina and choroid over a 28-day period. Finally, we investigated the mechanism by which APNp1 affects CNV by examining the expression of adiponectin receptor 1 (AdipoR1) and proliferating cell nuclear antigen (PCNA) in the retinal and choroidal tissue of the mouse eyes. AdipoR1 and PCNA were overexpressed in these tissues in mice with laser-induced CNV compared to naïve mice. Based on our data shown here, we think it will enhance our understanding of APNp1 as a therapeutic agent for wet-type AMD and possible treatment alternatives that could be more beneficial for patients.


Assuntos
Neovascularização de Coroide , Degeneração Macular Exsudativa , Adiponectina/genética , Idoso , Animais , Neovascularização de Coroide/genética , Neovascularização de Coroide/metabolismo , Modelos Animais de Doenças , Fluoresceína-5-Isotiocianato , Humanos , Camundongos , Soluções Oftálmicas , Peptídeos , Antígeno Nuclear de Célula em Proliferação/metabolismo , Receptores de Quinase C Ativada , Receptores de Adiponectina/genética , Receptores de Adiponectina/metabolismo
14.
J Biol Chem ; 285(44): 33826-33, 2010 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-20736175

RESUMO

This study was designed to explore the effect of recombinant, membrane-targeted CD59 (rCD59-APT542) on the growth and size of fully developed neovascular complex using the murine model of laser-induced choroidal neovascularization (CNV). CNV was induced by laser photocoagulation in C57BL/6 mice using an argon laser, and the animals received rCD59-APT542 via intravitreal (ivt) route. Western blot analysis, immunohistochemistry, and total complement hemolytic assay demonstrated that exogenously administered rCD59-APT542 was incorporated as well as retained in RPE and choroid and was functionally active in vivo. Single ivt injection during the growth of the CNV (i.e. at day 3 post-laser) resulted in ∼79% inhibition of the further growth of neovascular complex. The size of the CNV complex was significantly (p < 0.05) reduced by the administration of rCD59-APT542 after the CNV complex has fully developed (i.e. at day 7 post-laser). Treatment with rCD59-APT542 blocked the formation of membrane attack complex (MAC), increased apoptosis and decreased cell proliferation in the neovascular complex. On the basis of results presented here we conclude that recombinant membrane targeted CD59 inhibited the growth of the CNV complex and reduced the size of fully developed CNV in the laser-induced mouse model. We propose that a combination of two mechanisms: increased apoptosis and decreased cell proliferation, both resulting from local inhibition of MAC, may be responsible for inhibition of CNV by rCD59-APT542.


Assuntos
Antígenos CD59/metabolismo , Neovascularização de Coroide/metabolismo , Neovascularização Patológica/metabolismo , Proteínas Recombinantes/química , Animais , Apoptose , Membrana Celular/metabolismo , Proliferação de Células , Proteínas do Sistema Complemento/química , Imuno-Histoquímica , Inflamação , Degeneração Macular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL
15.
Am J Pathol ; 177(4): 1870-80, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20813971

RESUMO

The objective of this study was to explore the relationship between local (ie, ocular) complement factor H (CFH) and choroidal neovascularization (CNV) associated with wet age-related macular degeneration (AMD), a leading cause of irreversible blindness, in laser-treated C57BL/6 mice. Immunohistochemical and RT-PCR analysis of retinal pigmented epithelium (RPE)-choroid sclera revealed that the expression of CFH was down-regulated on day 1 with a dramatic increase on days 5 and 7 postlaser injury. Flat mount and Western blot analysis further revealed that membrane attack complex (MAC) expression was up-regulated on days 1 and 3 postlaser injury; however, MAC was down-regulated on days 5 and 7 postinjury but was still higher than in non-injured mice. Similar patterns for CFH and MAC were observed for RPE cells when serial paraffin sections of the laser spots were analyzed. Subretinal injection of siRNA directed against CFH resulted in a threefold suppression of CFH in the RPE and choroid without affecting either CFH levels in the liver or the functional activity of the alternative pathway in the peripheral blood. Ocular knock-down of CFH resulted in increased MAC deposition, which leads to the early onset as well as exacerbation of laser-induced CNV. In conclusion, our findings provide evidence that CFH present on RPE and choroid regulates local MAC formation that is critical for the development of laser-induced CNV.


Assuntos
Neovascularização de Coroide/etiologia , Neovascularização de Coroide/metabolismo , Fator H do Complemento/fisiologia , Modelos Animais de Doenças , Lasers/efeitos adversos , Epitélio Pigmentado Ocular/metabolismo , Animais , Western Blotting , Neovascularização de Coroide/patologia , Fator H do Complemento/antagonistas & inibidores , Técnicas Imunoenzimáticas , Degeneração Macular/etiologia , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Epitélio Pigmentado Ocular/patologia , RNA Mensageiro/genética , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Visão Ocular/fisiologia
16.
Nat Med ; 9(2): 206-12, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12514742

RESUMO

Systemic tolerance can be induced by the introduction of antigen into an immune-privileged site. Here we investigated the role of complement in the induction of tolerance after intraocular injection. We found that the development of antigen-specific tolerance is dependent on a complement activation product. The ligation of the complement C3 activation product iC3b to complement receptor type 3 (the iC3b receptor) on antigen-presenting cells resulted in the sequential production of transforming growth factor-beta2 and interleukin-10, which is essential for the induction of tolerance. These observations may extend to the development of both neonatal tolerance and other forms of acquired tolerance.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Antígenos CD , Antígenos de Neoplasias , Antígenos de Superfície , Proteínas Aviárias , Proteínas Sanguíneas , Complemento C3b/imunologia , Tolerância Imunológica/imunologia , Animais , Células Apresentadoras de Antígenos/metabolismo , Basigina , Hipersensibilidade Tardia/imunologia , Interleucina-10/antagonistas & inibidores , Interleucina-10/biossíntese , Interleucina-12/biossíntese , Glicoproteínas de Membrana/imunologia , Ratos , Ratos Endogâmicos Lew , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta2
17.
Ophthalmol Glaucoma ; 4(3): 330-335, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33080409

RESUMO

PURPOSE: Intra-Tenon or subconjunctival injection of a solution of mitomycin C (MMC) and 1% preservative-free lidocaine (as an anesthetic) has gained popularity for its use in trabeculectomy, a filtering surgery for glaucoma. To our knowledge, no studies have analyzed the impact of lidocaine on the cytotoxic effects of MMC in this setting. This study was conducted to evaluate in vitro fibroblast cytotoxicity to a solution of MMC (0.2 mg/ml) and 1% preservative-free lidocaine. DESIGN: Experimental study. PARTICIPANTS: Nonhuman subject research. METHODS: Cultured human conjunctival fibroblasts were incubated in phosphate-buffered saline (PBS) (control), MMC (0.2 mg/ml), a mixture of 0.2 mg/ml MMC + 1% preservative-free lidocaine, or 1% preservative-free lidocaine. Samples were taken at 2, 5, 10, 30, and 60 minutes, and a 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) assay with photoabsorbance testing was used to assess conjunctival cell viability. MAIN OUTCOME MEASURE: Mean photoabsorbance. RESULTS: Mean photoabsorbance across all time intervals was 0.680 for solutions incubated in PBS, 0.642 for MMC, 0.612 for MMC + 1% preservative-free lidocaine, and 0.605 for 1% preservative-free lidocaine. A 2-way analysis of variance analyzing solution, time, and solution-time interaction on photoabsorbance showed that PBS was least cytotoxic and an optimal control for this study. Tukey post hoc comparisons showed that MMC was more cytotoxic than PBS (P < 0.001). However, both MMC + 1% preservative-free lidocaine and 1% preservative-free lidocaine were more cytotoxic than MMC and PBS (P < 0.01 for all). No significant differences in cytotoxicity comparing lidocaine-containing solutions were observed. CONCLUSIONS: In this in vitro study, we found an increase in cytotoxicity when MMC (0.2 mg/ml) was combined with 1% preservative-free lidocaine. We note that lidocaine did not inhibit MMC cytotoxicity and exhibited a significant cytotoxic effect on its own.


Assuntos
Glaucoma , Trabeculectomia , Glaucoma/cirurgia , Humanos , Lidocaína/farmacologia , Mitomicina/farmacologia , Sais de Tetrazólio
18.
J Biol Chem ; 284(45): 31401-11, 2009 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-19755419

RESUMO

This study was initiated to induce experimental autoimmune anterior uveitis (EAAU) in Lewis rats by melanin-associated antigen (MAA; 22-kDa fragment of type I collagen alpha2 chain) derived from rat iris and ciliary body (CB), to localize MAA within the eye, and to investigate the possible mechanism of MAA generation in vivo. The EAAU model replicates idiopathic human anterior uveitis. Lewis rats sensitized to rat MAA developed anterior uveitis, and EAAU induced by rat MAA can be adoptively transferred to naive syngenic rats by MAA-primed T cells. Animals immunized with rat MAA developed cellular immunity to the antigen. MAA was detected only in the iris and CB of the eye. Iris and CB were the major source of matrix metalloproteinase-1 (MMP-1) in the naive eye, and ocular expression of MMP-1 was up-regulated, whereas expression of tissue inhibitor of metalloproteinase 1 decreased before the onset of EAAU. These results demonstrated that EAAU can be induced by autologous MAA. Uveitogenic antigen is present only in the iris and CB of the eye, and the imbalance between MMP-1 and tissue inhibitor of metalloproteinase 1 may play a role in the generation of MAA in vivo. Collectively, the evidence presented here suggests that MAA is an autoantigen in EAAU. These observations may extend to idiopathic human anterior uveitis and facilitate the development of antigen-specific therapy.


Assuntos
Autoantígenos/imunologia , Doenças Autoimunes/metabolismo , Colágeno Tipo I/metabolismo , Processamento de Proteína Pós-Traducional , Uveíte Anterior/metabolismo , Animais , Autoantígenos/metabolismo , Doenças Autoimunes/imunologia , Bovinos , Colágeno Tipo I/imunologia , Modelos Animais de Doenças , Humanos , Masculino , Ratos , Ratos Endogâmicos Lew , Organismos Livres de Patógenos Específicos , Uveíte Anterior/imunologia
19.
Immunol Cell Biol ; 88(2): 187-96, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19884905

RESUMO

The objective of this study was to inhibit experimental autoimmune anterior uveitis (EAAU) by establishing antigen-specific immune tolerance in animals pre-sensitized with melanin-associated antigen (MAA). Intravenous administration of MAA on days 6, 7, 8 and 9 post-immunization induced tolerance and inhibited EAAU in all Lewis rats. The number of cells (total T cells, CD4(+) T cells and CD8(+) T cells) undergoing apoptosis dramatically increased in the popliteal lymph nodes (LNs) of the tolerized animals compared with non-tolerized animals. In addition, Fas ligand (FasL), TNF receptor 1 (TNFR1) and caspase-8 were upregulated in tolerized rats. Proliferation of total lymphocytes, CD4(+)T cells and CD8(+) T cells (harvested from the popliteal LNs) in response to antigenic stimulation was drastically reduced in the state of tolerance compared with the cells from non-tolerized animals. The level of interferon (IFN)-gamma and IL-2 decreased, whereas TGF-beta2 was elevated in the state of tolerance. Furthermore, the number of CD4(+)CD25(+)FoxP3(+) regulatory T cells (Tregs) increased in the popliteal LNs of tolerized animals compared with non-tolerized animals. In conclusion, our results suggest that deletion of antigen-specific T cells by apoptosis and active suppression mediated by Tregs has an important role in the induction of antigen specific immune tolerance in animals with an established immune response against MAA.


Assuntos
Autoantígenos/imunologia , Doenças Autoimunes/prevenção & controle , Epitopos/imunologia , Tolerância Imunológica/imunologia , Subunidade alfa de Receptor de Interleucina-2/imunologia , Linfócitos T Reguladores/imunologia , Uveíte/prevenção & controle , Transferência Adotiva , Animais , Apoptose , Autoantígenos/administração & dosagem , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Bovinos , Proliferação de Células , Citocinas/biossíntese , Fatores de Transcrição Forkhead/metabolismo , Imunização , Injeções Intravenosas , Linfonodos/imunologia , Linfonodos/patologia , Depleção Linfocítica , Masculino , Ratos , Ratos Endogâmicos Lew , Uveíte/imunologia , Uveíte/patologia
20.
Am J Pathol ; 173(5): 1440-54, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18832572

RESUMO

Experimental autoimmune anterior uveitis (EAAU) serves as an animal model for human idiopathic AU, the most common form of intraocular inflammation of significant morbidity whose recurrence can lead to permanent vision loss. This study was undertaken to inhibit EAAU by inducing tolerance to melanin-associated antigen (MAA) and to investigate the underlying mechanisms responsible for tolerance induction. Intravenous administration of MAA both induced tolerance and inhibited EAAU in Lewis rats. Flow cytometric analysis revealed that the proliferation of lymph node cells in response to antigenic stimulation was drastically reduced in the state of tolerance both in vivo and in vitro. Our results from co-culture experiments demonstrated that intravenous administration of MAA led to the generation of T-regulatory cells that suppress T-cell proliferative responses and induce tolerance. Expression levels of both interleukin-10 and transforming growth factor-beta2 were elevated whereas reduced levels of tumor necrosis factor-alpha, interferon-gamma, and interleukin-2 were detected in tolerance-induced animals. Tolerance was reversed by replenishing these animals with recombinant interleukin-2. Tolerance could be adoptively transferred by removing lymph node cells from tolerance-induced donors and giving them to recipient rats. Interestingly, adoptive transfer of tolerance failed when lymph nodes cells were depleted of CD4(+)CD25(+) T cells. In conclusion, T-cell nonresponsiveness because of active suppression mediated by T-regulatory cells facilitates the development of tolerance to MAA in EAAU.


Assuntos
Antígenos de Neoplasias/imunologia , Autoantígenos/imunologia , Doenças Autoimunes/imunologia , Antígenos CD4/metabolismo , Tolerância Imunológica/imunologia , Linfócitos T Reguladores/imunologia , Uveíte/imunologia , Transferência Adotiva , Animais , Antígenos de Neoplasias/administração & dosagem , Antígenos de Neoplasias/farmacologia , Autoantígenos/administração & dosagem , Autoantígenos/farmacologia , Doenças Autoimunes/patologia , Bovinos , Proliferação de Células/efeitos dos fármacos , Técnicas de Cocultura , Tolerância Imunológica/efeitos dos fármacos , Injeções Intravenosas , Interleucina-2/farmacologia , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Linfonodos/efeitos dos fármacos , Linfonodos/patologia , Contagem de Linfócitos , Masculino , Ratos , Ratos Endogâmicos Lew , Linfócitos T Reguladores/efeitos dos fármacos , Uveíte/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA