Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Development ; 150(5)2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36762625

RESUMO

Microglia, a resident immune cell of the central nervous system (CNS), play a pivotal role in facilitating neurovascular development through mechanisms that are not fully understood. Previous reports indicate a role for microglia in regulating astrocyte density. This current work resolves the mechanism through which microglia facilitate astrocyte spatial patterning and superficial vascular bed formation in the neuroretina during development. Ablation of microglia increased astrocyte density and altered spatial patterning. Mechanistically, we show that microglia regulate the formation of the spatially organized astrocyte template required for subsequent vascular growth, through the complement C3/C3aR axis during neuroretinal development. Lack of C3 or C3aR hindered the developmental phagocytic removal of astrocyte bodies and resulted in increased astrocyte density. In addition, increased astrocyte density was associated with elevated proangiogenic extracellular matrix gene expression in C3- and C3aR-deficient retinas, resulting in increased vascular density. These data demonstrate that microglia regulate developmental astrocyte and vascular network spatial patterning in the neuroretina via the complement axis.


Assuntos
Complemento C3 , Microglia , Astrócitos , Complemento C3/genética , Retina
2.
Proc Natl Acad Sci U S A ; 116(20): 9989-9998, 2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-31023885

RESUMO

Autoimmune uveitis is a sight-threatening ocular inflammatory condition in which the retina and uveal tissues become a target of autoreactive immune cells. While microglia have been studied extensively in autoimmune uveitis, their exact function remains uncertain. The objective of the current study was to determine whether resident microglia are necessary and sufficient to initiate and amplify retinal inflammation in autoimmune uveitis. In this study, we clearly demonstrate that microglia are essential for initiating infiltration of immune cells utilizing a murine model of experimental autoimmune uveoretinitis (EAU) and the recently identified microglia-specific marker P2ry12. Initiating disease is the primary function of microglia in EAU, since eliminating microglia during the later stages of EAU had little effect, indicating that the function of circulating leukocytes is to amplify and sustain destructive inflammation once microglia have triggered disease. In the absence of microglia, uveitis does not develop, since leukocytes cannot gain entry through the blood-retinal barrier, illustrating that microglia play a critical role in regulating infiltration of inflammatory cells into the retina.


Assuntos
Doenças Autoimunes/etiologia , Microglia/fisiologia , Doenças Retinianas/imunologia , Uveíte/imunologia , Animais , Modelos Animais de Doenças , Feminino , Camundongos , Compostos Orgânicos , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/antagonistas & inibidores
3.
Proc Natl Acad Sci U S A ; 115(27): E6264-E6273, 2018 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-29915052

RESUMO

Retinal detachment (RD) is a sight-threatening complication common in many highly prevalent retinal disorders. RD rapidly leads to photoreceptor cell death beginning within 12 h following detachment. In patients with sustained RD, progressive visual decline due to photoreceptor cell death is common, leading to significant and permanent loss of vision. Microglia are the resident immune cells of the central nervous system, including the retina, and function in the homeostatic maintenance of the neuro-retinal microenvironment. It is known that microglia become activated and change their morphology in retinal diseases. However, the function of activated microglia in RD is incompletely understood, in part because of the lack of microglia-specific markers. Here, using the newly identified microglia marker P2ry12 and microglial depletion strategies, we demonstrate that retinal microglia are rapidly activated in response to RD and migrate into the injured area within 24 h post-RD, where they closely associate with infiltrating macrophages, a population distinct from microglia. Once in the injured photoreceptor layer, activated microglia can be observed to contain autofluorescence within their cell bodies, suggesting they function to phagocytose injured or dying photoreceptors. Depletion of retinal microglia results in increased disease severity and inhibition of macrophage infiltration, suggesting that microglia are involved in regulating neuroinflammation in the retina. Our work identifies that microglia mediate photoreceptor survival in RD and suggests that this effect may be due to microglial regulation of immune cells and photoreceptor phagocytosis.


Assuntos
Macrófagos/imunologia , Microglia/imunologia , Células Fotorreceptoras de Vertebrados/imunologia , Receptores Purinérgicos P2Y12/imunologia , Descolamento Retiniano/imunologia , Animais , Morte Celular/genética , Morte Celular/imunologia , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Macrófagos/patologia , Camundongos , Camundongos Transgênicos , Microglia/patologia , Células Fotorreceptoras de Vertebrados/patologia , Receptores Purinérgicos P2Y12/genética , Descolamento Retiniano/genética , Descolamento Retiniano/patologia
4.
Clin Immunol ; 214: 108391, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32229292

RESUMO

Age-related macular degeneration (AMD) is a leading cause of irreversible vision loss among the elderly population. Genetic studies in susceptible individuals have linked this ocular disease to deregulated complement activity that culminates in increased C3 turnover, retinal inflammation and photoreceptor loss. Therapeutic targeting of C3 has therefore emerged as a promising strategy for broadly intercepting the detrimental proinflammatory consequences of complement activation in the retinal tissue. In this regard, a PEGylated second-generation derivative of the compstatin family of C3-targeted inhibitors is currently in late-stage clinical development as a treatment option for geographic atrophy, an advanced form of AMD which lacks approved therapy. While efficacy has been strongly suggested in phase 2 clinical trials, crucial aspects still remain to be defined with regard to the ocular bioavailability, tissue distribution and residence, and dosing frequency of such inhibitors in AMD patients. Here we report the intraocular distribution and pharmacokinetic profile of the fourth-generation compstatin analog, Cp40-KKK in cynomolgus monkeys following a single intravitreal injection. Using a sensitive surface plasmon resonance (SPR)-based competition assay and ELISA, we have quantified both the amount of inhibitor and the concentration of C3 retained in the vitreous of Cp40-KKK-injected animals. Cp40-KKK displays prolonged intraocular residence, being detected at C3-saturating levels for over 3 months after a single intravitreal injection. Moreover, we have probed the distribution of Cp40-KKK within the ocular tissue by means of immunohistochemistry and highly specific anti-Cp40-KKK antibodies. Both C3 and Cp40-KKK were detected in the retinal tissue of inhibitor-injected animals, with prominent co-localization in the choroid one-month post intravitreal injection. These results attest to the high retinal tissue penetrance and target-driven distribution of Cp40-KKK. Given its subnanomolar binding affinity and prolonged ocular residence, Cp40-KKK constitutes a promising drug candidate for ocular pathologies underpinned by deregulated C3 activation.


Assuntos
Complemento C3/antagonistas & inibidores , Olho/química , Idoso , Animais , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Humanos , Injeções Intravítreas , Macaca fascicularis , Retina/química , Fatores de Tempo , Distribuição Tecidual
5.
Proc Natl Acad Sci U S A ; 114(36): E7545-E7553, 2017 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-28827330

RESUMO

Age-related macular degeneration (AMD) is the most common cause of blindness for individuals age 50 and above in the developed world. Abnormal growth of choroidal blood vessels, or choroidal neovascularization (CNV), is a hallmark of the neovascular (wet) form of advanced AMD and leads to significant vision loss. A growing body of evidence supports a strong link between neovascular disease and inflammation. Metabolites of long-chain polyunsaturated fatty acids derived from the cytochrome P450 (CYP) monooxygenase pathway serve as vital second messengers that regulate a number of hormones and growth factors involved in inflammation and vascular function. Using transgenic mice with altered CYP lipid biosynthetic pathways in a mouse model of laser-induced CNV, we characterized the role of these lipid metabolites in regulating neovascular disease. We discovered that the CYP-derived lipid metabolites epoxydocosapentaenoic acids (EDPs) and epoxyeicosatetraenoic acids (EEQs) are vital in dampening CNV severity. Specifically, overexpression of the monooxygenase CYP2C8 or genetic ablation or inhibition of the soluble epoxide hydrolase (sEH) enzyme led to increased levels of EDP and EEQ with attenuated CNV development. In contrast, when we promoted the degradation of these CYP-derived metabolites by transgenic overexpression of sEH, the protective effect against CNV was lost. We found that these molecules work in part through their ability to regulate the expression of key leukocyte adhesion molecules, on both leukocytes and endothelial cells, thereby mediating leukocyte recruitment. These results suggest that CYP lipid signaling molecules and their regulators are potential therapeutic targets in neovascular diseases.


Assuntos
Neovascularização de Coroide/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Metabolismo dos Lipídeos/fisiologia , Sistemas do Segundo Mensageiro/fisiologia , Animais , Citocromo P-450 CYP2C8/metabolismo , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Epóxido Hidrolases/metabolismo , Ácidos Graxos Insaturados/metabolismo , Leucócitos/metabolismo , Degeneração Macular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
6.
Am J Pathol ; 188(10): 2142-2146, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30220553

RESUMO

This commentary highlights the article by Lin et al that demonstrates the therapeutic potential of small-molecule atypical protein kinase C inhibitors in inflammatory ocular disease.


Assuntos
Permeabilidade Capilar , Oftalmopatias , Humanos , Inflamação , Proteína Quinase C
7.
FASEB J ; 30(3): 1300-5, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26631482

RESUMO

Proliferative retinopathic diseases often progress in 2 phases: initial regression of retinal vasculature (phase 1) followed by subsequent neovascularization (NV) (phase 2). The immune system has been shown to aid in vascular pruning in such retinopathies; however, little is known about the role of the alternative complement pathway in the initial vascular regression phase. Using a mouse model of oxygen-induced retinopathy (OIR), we observed that alternative complement pathway-deficient mice (Fb(-/-)) exhibited a mild decrease in vascular loss at postnatal day (P)8 compared with age- and strain-matched controls (P = 0.035). Laser capture microdissection was used to isolate the retinal blood vessels. Expression of the complement inhibitors Cd55 and Cd59 was significantly decreased in blood vessels isolated from hyperoxic retinas compared with those from normoxic control mice. Vegf expression was measured at P8 and found to be significantly lower in OIR mice than in normoxic control mice (P = 0.0048). Further examination of specific Vegf isoform expression revealed a significant decrease in Vegf120 (P = 0.00032) and Vegf188 (P = 0.0092). In conjunction with the major modulating effects of Vegf during early retinal vascular development, our data suggest a modest involvement of the alternative complement pathway in targeting vessels for regression in the initial vaso-obliteration stage of OIR.


Assuntos
Via Alternativa do Complemento/imunologia , Neovascularização Patológica/imunologia , Retina/imunologia , Neovascularização Retiniana/imunologia , Vitreorretinopatia Proliferativa/imunologia , Animais , Animais Recém-Nascidos/imunologia , Animais Recém-Nascidos/metabolismo , Modelos Animais de Doenças , Hiperóxia/imunologia , Hiperóxia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/patologia , Oxigênio/metabolismo , Isoformas de Proteínas/metabolismo , Retina/metabolismo , Neovascularização Retiniana/metabolismo , Vasos Retinianos/imunologia , Vasos Retinianos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Vitreorretinopatia Proliferativa/metabolismo
8.
Proc Natl Acad Sci U S A ; 111(26): 9603-8, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24979774

RESUMO

Ocular neovascularization, including age-related macular degeneration (AMD), is a primary cause of blindness in individuals of industrialized countries. With a projected increase in the prevalence of these blinding neovascular diseases, there is an urgent need for new pharmacological interventions for their treatment or prevention. Increasing evidence has implicated eicosanoid-like metabolites of long-chain polyunsaturated fatty acids (LCPUFAs) in the regulation of neovascular disease. In particular, metabolites generated by the cytochrome P450 (CYP)-epoxygenase pathway have been shown to be potent modulators of angiogenesis, making this pathway a reasonable previously unidentified target for intervention in neovascular ocular disease. Here we show that dietary supplementation with ω-3 LCPUFAs promotes regression of choroidal neovessels in a well-characterized mouse model of neovascular AMD. Leukocyte recruitment and adhesion molecule expression in choroidal neovascular lesions were down-regulated in mice fed ω-3 LCPUFAs. The serum of these mice showed increased levels of anti-inflammatory eicosanoids derived from eicosapentaenoic acid and docosahexaenoic acid. 17,18-epoxyeicosatetraenoic acid and 19,20-epoxydocosapentaenoic acid, the major CYP-generated metabolites of these primary ω-3 LCPUFAs, were identified as key lipid mediators of disease resolution. We conclude that CYP-derived bioactive lipid metabolites from ω-3 LCPUFAs are potent inhibitors of intraocular neovascular disease and show promising therapeutic potential for resolution of neovascular AMD.


Assuntos
Neovascularização de Coroide/tratamento farmacológico , Sistema Enzimático do Citocromo P-450/metabolismo , Ácidos Graxos Ômega-3/farmacologia , Alimentos Fortificados , Degeneração Macular/fisiopatologia , Animais , Ácidos Araquidônicos , Cromatografia Líquida , DNA Complementar/genética , Ensaio de Imunoadsorção Enzimática , Ácidos Graxos Ômega-3/uso terapêutico , Citometria de Fluxo , Immunoblotting , Microdissecção e Captura a Laser , Degeneração Macular/tratamento farmacológico , Camundongos , PPAR gama/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Espectrometria de Massas em Tandem
9.
FASEB J ; 28(7): 3171-82, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24668752

RESUMO

A defining feature in proliferative retinopathies is the formation of pathological neovessels. In these diseases, the balance between neovessel formation and regression determines blindness, making the modulation of neovessel growth highly desirable. The role of the immune system in these retinopathies is of increasing interest, but it is not completely understood. We investigated the role of the alternative complement pathway during the formation and resolution of aberrant neovascularization. We used alternative complement pathway-deficient (Fb(-/-)) mice and age- and strain-matched control mice to assess neovessel development and regression in an oxygen-induced retinopathy (OIR) mouse model. In the control mice, we found increased transcription of Fb after OIR treatment. In the Fb(-/-) mice, we prepared retinal flatmounts and identified an increased number of neovessels, peaking at postnatal day 17 (P17; P=0.001). Subjecting human umbilical vein endothelial cells (HUVECs) to low oxygen, mimicking a characteristic of neovessels, decreased the expression of the complement inhibitor Cd55. Finally, using laser capture microdissection (LCM) to isolate the neovessels after OIR, we found decreased expression of Cd55 (P=0.005). Together, our data implicate the alternative complement pathway in facilitating neovessel clearance by down-regulating the complement inhibitor Cd55 specifically on neovessels, allowing for their targeted removal while leaving the established vasculature intact.-Sweigard, J. H., Yanai, R., Gaissert, P., Saint-Geniez, M., Kataoka, K., Thanos, A., Stahl, G. L., Lambris, J. D., Connor, K. M. The alternative complement pathway regulates pathological angiogenesis in the retina.


Assuntos
Via Alternativa do Complemento/fisiologia , Neovascularização Patológica/patologia , Neovascularização Retiniana/patologia , Animais , Apoptose/fisiologia , Antígenos CD55/metabolismo , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo/fisiologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/metabolismo , Oxigênio/metabolismo , Retina , Doenças Retinianas/metabolismo , Doenças Retinianas/patologia , Neovascularização Retiniana/metabolismo
10.
Nature ; 457(7233): 1103-8, 2009 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-19242469

RESUMO

Angiogenesis is controlled by physical interactions between cells and extracellular matrix as well as soluble angiogenic factors, such as VEGF. However, the mechanism by which mechanical signals integrate with other microenvironmental cues to regulate neovascularization remains unknown. Here we show that the Rho inhibitor, p190RhoGAP (also known as GRLF1), controls capillary network formation in vitro in human microvascular endothelial cells and retinal angiogenesis in vivo by modulating the balance of activities between two antagonistic transcription factors, TFII-I (also known as GTF2I) and GATA2, that govern gene expression of the VEGF receptor VEGFR2 (also known as KDR). Moreover, this new angiogenesis signalling pathway is sensitive to extracellular matrix elasticity as well as soluble VEGF. This is, to our knowledge, the first known functional cross-antagonism between transcription factors that controls tissue morphogenesis, and that responds to both mechanical and chemical cues.


Assuntos
Neovascularização Fisiológica/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Animais , Animais Recém-Nascidos , Linhagem Celular , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/crescimento & desenvolvimento , Matriz Extracelular/metabolismo , Fator de Transcrição GATA2/metabolismo , Técnicas de Silenciamento de Genes , Fatores de Troca do Nucleotídeo Guanina/deficiência , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/fisiologia , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Vasos Retinianos/crescimento & desenvolvimento , Vasos Retinianos/metabolismo , Transdução de Sinais , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Fatores de Transcrição TFII/metabolismo , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/biossíntese , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
11.
Am J Pathol ; 182(1): 255-65, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23141926

RESUMO

Neovascular diseases of the eye are the most common causes of blindness worldwide. The mechanisms underlying pathological neovascularization in the retina remain incompletely understood. PGC-1α is a transcriptional coactivator that plays a central role in the regulation of cellular metabolism. In skeletal muscle, PGC-1α induces VEGFA expression and powerfully promotes angiogenesis, suggesting a similar role in other tissues. This study investigates the role of PGC-1α during normal and pathological vascularization in the retina. We show that PGC-1α induces the expression of VEGFA in numerous retinal cells, and that PGC-1α expression is strongly induced during postnatal retinal development, coincident with VEGFA expression and angiogenesis. PGC-1α(-/-) mice have a significant reduction of early retinal vascular outgrowth, and reduced density of capillaries and number of main arteries and veins as adults. In the oxygen-induced retinopathy model of retinopathy of prematurity, PGC-1α expression is dramatically induced in the inner nuclear layer of the retina, suggesting that PGC-1α drives pathological neovascularization. In support of this, PGC-1α(-/-) mice subjected to oxygen-induced retinopathy had decreased expression of VEGFA and were protected against pathological neovascularization. These results demonstrate that PGC-1α regulates VEGFA in the retina and is required for normal vessel development and for pathological neovascularization. The data highlight PGC-1α as a novel target in the treatment of neovascular diseases of the eye.


Assuntos
Neovascularização Fisiológica/fisiologia , Neovascularização Retiniana/metabolismo , Vasos Retinianos/metabolismo , Transativadores/fisiologia , Animais , Modelos Animais de Doenças , Regulação da Expressão Gênica/fisiologia , Humanos , Recém-Nascido , Camundongos , Camundongos Knockout , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Retina/crescimento & desenvolvimento , Retina/metabolismo , Retina/patologia , Neovascularização Retiniana/patologia , Neovascularização Retiniana/prevenção & controle , Vasos Retinianos/crescimento & desenvolvimento , Vasos Retinianos/patologia , Retinopatia da Prematuridade/metabolismo , Retinopatia da Prematuridade/prevenção & controle , Transativadores/biossíntese , Transativadores/deficiência , Fatores de Transcrição , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
12.
Nat Med ; 13(7): 868-873, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17589522

RESUMO

Many sight-threatening diseases have two critical phases, vessel loss followed by hypoxia-driven destructive neovascularization. These diseases include retinopathy of prematurity and diabetic retinopathy, leading causes of blindness in childhood and middle age affecting over 4 million people in the United States. We studied the influence of omega-3- and omega-6-polyunsaturated fatty acids (PUFAs) on vascular loss, vascular regrowth after injury, and hypoxia-induced pathological neovascularization in a mouse model of oxygen-induced retinopathy. We show that increasing omega-3-PUFA tissue levels by dietary or genetic means decreased the avascular area of the retina by increasing vessel regrowth after injury, thereby reducing the hypoxic stimulus for neovascularization. The bioactive omega-3-PUFA-derived mediators neuroprotectinD1, resolvinD1 and resolvinE1 also potently protected against neovascularization. The protective effect of omega-3-PUFAs and their bioactive metabolites was mediated, in part, through suppression of tumor necrosis factor-alpha. This inflammatory cytokine was found in a subset of microglia that was closely associated with retinal vessels. These findings indicate that increasing the sources of omega-3-PUFA or their bioactive products reduces pathological angiogenesis. Western diets are often deficient in omega-3-PUFA, and premature infants lack the important transfer from the mother to the infant of omega-3-PUFA that normally occurs in the third trimester of pregnancy. Supplementing omega-3-PUFA intake may be of benefit in preventing retinopathy.


Assuntos
Ácidos Graxos Ômega-3/farmacologia , Neovascularização Patológica/induzido quimicamente , Vasos Retinianos/efeitos dos fármacos , Animais , Dieta , Gorduras na Dieta , Ácidos Graxos Ômega-6/farmacologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doenças Retinianas/induzido quimicamente , Doenças Retinianas/tratamento farmacológico
13.
bioRxiv ; 2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38585800

RESUMO

Autoimmune uveitis is a major cause of blindness in the working-age population of developed countries. Experimental autoimmune uveitis (EAU) depends on activation of interphotoreceptor retinoid-binding protein (IRBP) specific CD4 + effector T cells that migrate systemically and infiltrate into the retina. Following systemic induction of retinal antigen-specific T cells, the development of EAU can be broken down into three phases: early phase when inflammatory cells begin to infiltrate the retina, amplification phase, and peak phase. Although studied extensively, the function of local antigen-presenting cells (APCs) within the retina remains unclear. Two potential types of APCs are present during uveitis, resident microglia and infiltrating CD11c + dendritic cells (DCs). MHC class II (MHC II) is expressed within the retina on both CD11c + DCs and microglia during the amplification phase of EAU. Therefore, we used microglia specific (P2RY12 and TMEM119) and CD11c + DC specific MHC II knockout mice to study the function of APCs within the retina using the conventional and adoptive transfer methods of inducing EAU. Microglia were essential during all phases of EAU development: the early phase when microglia were MHC Il negative, and amplification and peak phases when microglia were MHC II positive. Unexpectedly, retinal infiltrating MHC Il + CD11c + DCs were present within the retina but their antigen-presenting function was not required for all phases of uveitis. Our data indicate microglia are the critical APCs within the retina and an important therapeutic target that can prevent and/or diminish uveitis even in the presence of circulating IRBP-specific CD4 + effector T cells.

14.
Cells ; 12(13)2023 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-37443787

RESUMO

Retinal detachment (RD) is a neurodegenerative blinding disease caused by plethora of clinical conditions. RD is characterized by the physical separation of retina from the underlying retinal pigment epithelium (RPE), eventually leading to photoreceptor cell death, inflammation, and vision loss. Albeit the activation of complement plays a critical role in the pathogenesis of RD, the retinal cellular source for complement production remains elusive. Here, using C3 tdTomato reporter mice we show that retinal injury upregulates C3 expression, specifically in Müller cells. Activation of the complement cascade results in the generation of proinflammatory cleaved products, C3a and C5a, that bind C3aR and C5aR1, respectively. Our flow cytometry data show that retinal injury significantly upregulated C3aR and C5aR1 in microglia and resulted in the infiltration of peripheral immune cells. Loss of C3, C5, C3aR or C5aR1 reduced photoreceptor cell death and infiltration of microglia and peripheral immune cells into the sub-retinal space. These results indicate that C3/C3aR and C5/C5aR1 play a crucial role in eliciting photoreceptor degeneration and inflammatory responses in RD.


Assuntos
Células Ependimogliais , Descolamento Retiniano , Camundongos , Animais , Células Ependimogliais/patologia , Doenças Neuroinflamatórias , Células Fotorreceptoras/patologia , Morte Celular , Retina/metabolismo , Descolamento Retiniano/metabolismo , Proteínas do Sistema Complemento/metabolismo
15.
Circ Res ; 107(4): 495-500, 2010 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-20634487

RESUMO

RATIONALE: Omega3 long-chain polyunsaturated fatty acids (omega3-PUFAs) are powerful modulators of angiogenesis. However, little is known about the mechanisms governing omega3-PUFA-dependent attenuation of angiogenesis. OBJECTIVE: This study aims to identify a major mechanism by which omega3-PUFAs attenuate retinal neovascularization. METHODS AND RESULTS: Administering omega3-PUFAs exclusively during the neovascular stage of the mouse model of oxygen-induced retinopathy induces a direct neovascularization reduction of more than 40% without altering vasoobliteration or the regrowth of normal vessels. Cotreatment with an inhibitor of peroxisome proliferator-activated receptor (PPAR)gamma almost completely abrogates this effect. Inhibition of PPARgamma also reverses the omega3-PUFA-induced reduction of retinal tumor necrosis factor-alpha, intercellular adhesion molecule-1, vascular cell adhesion molecule-1, endothelial selectin, and angiopoietin 2 but not vascular endothelial growth factor. CONCLUSIONS: These results identify a direct, PPARgamma-mediated effect of omega3-PUFAs on retinal neovascularization formation and retinal angiogenic activation that is independent of vascular endothelial growth factor.


Assuntos
Inibidores da Angiogênese/fisiologia , Ácidos Graxos Ômega-3/administração & dosagem , Neovascularização Patológica/metabolismo , PPAR gama/fisiologia , Doenças Retinianas/metabolismo , Inibidores da Angiogênese/administração & dosagem , Animais , Animais Recém-Nascidos , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/dietoterapia , Neovascularização Patológica/prevenção & controle , Doenças Retinianas/dietoterapia , Doenças Retinianas/prevenção & controle , Fator A de Crescimento do Endotélio Vascular/fisiologia
16.
Adv Exp Med Biol ; 946: 161-83, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21948368

RESUMO

Pathological neovascularization (NV) is a hallmark of late stage neovascular age-related macular degeneration (AMD), diabetic retinopathy (DR), and retinopathy of prematurity (ROP). There is accumulating evidence that alterations in inflammatory and immune system pathways that arise from genetic differences, injury, and disease can predispose individuals to retinal neovascular eye diseases. Yet the mechanism of disease progression with respect to the complement system in these maladies is not fully understood. Recent studies have implicated the complement system as an emerging player in the etiology of several retinal diseases. We will summarize herein several of the complement system pathways known to be involved in ocular neovascular pathologies. Current treatment for many neovascular eye diseases focuses on suppression of NV with laser ablation, photodynamic therapy, or anti-VEGF angiogenic inhibitors. However, these treatments do not address the underlying cause of many of these diseases. A clear understanding of the cellular and molecular mechanisms could bring a major shift in our approach to disease treatment and prevention.


Assuntos
Neovascularização de Coroide/imunologia , Proteínas do Sistema Complemento/imunologia , Retinopatia Diabética/imunologia , Degeneração Macular/imunologia , Retinopatia da Prematuridade/imunologia , Humanos , Recém-Nascido
17.
J Clin Invest ; 118(2): 526-33, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18219389

RESUMO

Erythropoietin (Epo), a hormone known to stimulate bone marrow erythrocyte production, is widely used to treat anemia in patients at risk for vascular disease. However, the effects of Epo on angiogenesis are not well defined. We studied the role of Epo in a mouse model of retinopathy characterized by oxygen-induced vascular loss followed by hypoxia-induced pathological neovascularization. Without treatment, local retinal Epo levels were suppressed during the vessel loss phase. Administration of exogenous Epo prevented both vessel dropout and subsequent hypoxia-induced neovascularization. Early use of Epo also protected against hypoxia-induced retinal neuron apoptosis. In contrast, retinal Epo mRNA levels were highly elevated during the retinopathy neovascular phase. Exogenous late Epo treatment did not protect the retina, but rather enhanced pathological neovascularization. Epo's early protective effect occurred through both systemic retinal recruitment of proangiogenic bone marrow-derived progenitor cells and activation of prosurvival NF-kappaB via Epo receptor activation on retinal vessels and neurons. Thus early retinal Epo suppression contributed to retinal vascular instability, and elevated Epo levels during the proliferation stage contributed to neovascularization and disease. Understanding the role of Epo in angiogenesis is critical to timing its intervention in patients with retinopathy or other diseases in which pathological angiogenesis plays a significant role.


Assuntos
Eritropoetina/administração & dosagem , Eritropoetina/fisiologia , Neovascularização Patológica/genética , Neovascularização Patológica/prevenção & controle , Doenças Retinianas/tratamento farmacológico , Vasos Retinianos/efeitos dos fármacos , Vasos Retinianos/crescimento & desenvolvimento , Animais , Apoptose/efeitos dos fármacos , Eritropoetina/genética , Hipóxia , Camundongos , NF-kappa B/metabolismo , Neurônios/efeitos dos fármacos , Oxigênio/toxicidade , Receptores da Eritropoetina/agonistas , Retina/efeitos dos fármacos , Retina/patologia , Doenças Retinianas/induzido quimicamente , Doenças Retinianas/patologia , Vasos Retinianos/patologia , Células-Tronco/efeitos dos fármacos
18.
Front Immunol ; 12: 680568, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34093583

RESUMO

Autoimmune uveitis is a sight-threatening ocular inflammatory condition in which the retina and uveal tissues become a target of autoreactive immune cells. The CD47 is a ubiquitously expressed transmembrane protein which plays multiple roles in fundamental cellular functions including phagocytosis, proliferation, and adhesion. Signal regulatory protein alpha (SIRPα), one of the CD47 ligands, is predominantly expressed in myeloid lineage cells such as dendritic cells (DCs) or macrophages, and CD47-SIRPα signaling pathway is implicated in the development of autoimmune diseases. Our current study demonstrates how CD47 depletion is effective in the prevention of experimental autoimmune uveitis (EAU), an animal model of human autoimmune uveitis, in animals deficient of CD47 (CD47-/- ). Systemic suppression of SIRPα+ DCs in animals deficient in CD47 resulted in the inability of autoreactive CD4+ T cells to develop, which is crucial to induction of EAU. Of interest, retinal microglia, the resident immune cell of the retina, express SIRPα, however these cells were not operative in EAU suppression in response to CD47 depletion. These results identify CD47 as a significant regulator in the development of SIRPα+ DCs that is vital to disease induction in EAU.


Assuntos
Doenças Autoimunes/etiologia , Antígeno CD47/deficiência , Suscetibilidade a Doenças , Oftalmopatias/etiologia , Animais , Doenças Autoimunes/diagnóstico , Doenças Autoimunes/metabolismo , Autoimunidade/genética , Biomarcadores , Modelos Animais de Doenças , Oftalmopatias/diagnóstico , Oftalmopatias/metabolismo , Feminino , Imunomodulação/genética , Imunofenotipagem , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos Knockout , Retina/imunologia , Retina/metabolismo , Retina/patologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Uveíte/diagnóstico , Uveíte/etiologia , Uveíte/metabolismo
19.
J Clin Invest ; 117(10): 2758-62, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17909623

RESUMO

Age-related macular degeneration (AMD), the most common cause of blindness in the elderly, is characterized by degeneration of the macula and can lead to loss of fine color vision. Alterations in inflammatory and immune system pathways, which arise from genetic differences, predispose individuals to AMD. Yet the mechanism of disease progression with respect to inflammation is not fully understood. In this issue of the JCI, the study by Combadière and colleagues shows that CX3C chemokine receptor 1-deficient (CX3CR1-deficient) mice have abnormal microglia that accumulate beneath the retina and contribute to the progression of AMD.


Assuntos
Predisposição Genética para Doença , Degeneração Macular/genética , Degeneração Macular/patologia , Microglia/patologia , Receptores de Quimiocinas/fisiologia , Animais , Receptor 1 de Quimiocina CX3C , Humanos , Degeneração Macular/terapia , Camundongos , Receptores de Quimiocinas/genética , Retina/patologia , Retina/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA