Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Angiogenesis ; 23(3): 385-394, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32140799

RESUMEN

To examine whether free fatty acid receptor 4 (FFAR4) activation can protect against choroidal neovascularization (CNV), which is a common cause of blindness, and to elucidate the mechanism underlying the inhibition, we used the mouse model of laser-induced CNV to mimic angiogenic aspects of age-related macular degeneration (AMD). Laser-induced CNV was compared between groups treated with an FFAR4 agonist or vehicle, and between FFAR4 wild-type (Ffar4+/+) and knock out (Ffar4-/-) mice on a C57BL/6J/6N background. The ex vivo choroid-sprouting assay, including primary retinal pigment epithelium (RPE) and choroid, without retina was used to investigate whether FFAR4 affects choroidal angiogenesis. Western blotting for pNF-ĸB/NF-ĸB and qRT-PCR for Il-6, Il-1ß, Tnf-α, Vegf, and Nf-ĸb were used to examine the influence of FFAR4 on inflammation, known to influence CNV. RPE isolated from Ffar4+/+ and Ffar4-/- mice were used to assess RPE contribution to inflammation. The FFAR4 agonist suppressed laser-induced CNV in C57BL/6J mice, and CNV increased in Ffar4-/- compared to Ffar4+/+ mice. We showed that the FFAR4 agonist acted through the FFAR4 receptor. The FFAR4 agonist suppressed mRNA expression of inflammation markers (Il-6, Il-1ß) via the NF-ĸB pathway in the retina, choroid, RPE complex. The FFAR4 agonist suppressed neovascularization in the choroid-sprouting ex vivo assay and FFAR4 deficiency exacerbated sprouting. Inflammation markers were increased in primary RPE cells of Ffar4-/- mice compared with Ffar4+/+ RPE. In this mouse model, the FFAR4 agonist suppressed CNV, suggesting FFAR4 to be a new molecular target to reduce pathological angiogenesis in CNV.


Asunto(s)
Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/prevención & control , Receptores Acoplados a Proteínas G/metabolismo , Animales , Neovascularización Coroidal/genética , Citocinas/genética , Citocinas/metabolismo , Masculino , Ratones , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/metabolismo , Receptores Acoplados a Proteínas G/genética
2.
Int J Mol Sci ; 21(2)2020 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-31963809

RESUMEN

Since the discovery of the first microRNA (miRNA) decades ago, studies of miRNA biology have expanded in many biomedical research fields, including eye research. The critical roles of miRNAs in normal development and diseases have made miRNAs useful biomarkers or molecular targets for potential therapeutics. In the eye, ocular neovascularization (NV) is a leading cause of blindness in multiple vascular eye diseases. Current anti-angiogenic therapies, such as anti-vascular endothelial growth factor (VEGF) treatment, have their limitations, indicating the need for investigating new targets. Recent studies established the roles of various miRNAs in the regulation of pathological ocular NV, suggesting miRNAs as both biomarkers and therapeutic targets in vascular eye diseases. This review summarizes the biogenesis of miRNAs, and their functions in the normal development and diseases of the eye, with a focus on clinical and experimental retinopathies in both human and animal models. Discovery of novel targets involving miRNAs in vascular eye diseases will provide insights for developing new treatments to counter ocular NV.


Asunto(s)
Neovascularización Coroidal/genética , MicroARNs/genética , Neovascularización Retiniana/genética , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Neovascularización Coroidal/tratamiento farmacológico , Regulación de la Expresión Génica/efectos de los fármacos , Marcadores Genéticos/efectos de los fármacos , Predisposición Genética a la Enfermedad , Humanos , MicroARNs/efectos de los fármacos , Terapia Molecular Dirigida , Neovascularización Retiniana/tratamiento farmacológico
3.
Int J Mol Sci ; 21(4)2020 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-32098361

RESUMEN

The tightly structured neural retina has a unique vascular network comprised of three interconnected plexuses in the inner retina (and choroid for outer retina), which provide oxygen and nutrients to neurons to maintain normal function. Clinical and experimental evidence suggests that neuronal metabolic needs control both normal retinal vascular development and pathological aberrant vascular growth. Particularly, photoreceptors, with the highest density of mitochondria in the body, regulate retinal vascular development by modulating angiogenic and inflammatory factors. Photoreceptor metabolic dysfunction, oxidative stress, and inflammation may cause adaptive but ultimately pathological retinal vascular responses, leading to blindness. Here we focus on the factors involved in neurovascular interactions, which are potential therapeutic targets to decrease energy demand and/or to increase energy production for neovascular retinal disorders.


Asunto(s)
Células Fotorreceptoras de Vertebrados/metabolismo , Retina/metabolismo , Enfermedades de la Retina/metabolismo , Neovascularización Retiniana/metabolismo , Vasos Retinianos/metabolismo , Animales , Velocidad del Flujo Sanguíneo , Humanos , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Enfermedades de la Retina/fisiopatología , Neovascularización Retiniana/fisiopatología , Vasos Retinianos/fisiología
4.
FASEB J ; 31(11): 4665-4681, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28739642

RESUMEN

Pathological angiogenesis in the eye is an important feature in the pathophysiology of many vision-threatening diseases, including retinopathy of prematurity, diabetic retinopathy, and age-related macular degeneration, as well as corneal diseases with abnormal angiogenesis. Development of reproducible and reliable animal models of ocular angiogenesis has advanced our understanding of both the normal development and the pathobiology of ocular neovascularization. These models have also proven to be valuable experimental tools with which to easily evaluate potential antiangiogenic therapies beyond eye research. This review summarizes the current available animal models of ocular angiogenesis. Models of retinal and choroidal angiogenesis, including oxygen-induced retinopathy, laser-induced choroidal neovascularization, and transgenic mouse models with deficient or spontaneous retinal/choroidal neovascularization, as well as models with induced corneal angiogenesis, are widely used to investigate the molecular and cellular basis of angiogenic mechanisms. Theoretical concepts and experimental protocols of these models are outlined, as well as their advantages and potential limitations, which may help researchers choose the most suitable models for their investigative work.-Liu, C.-H., Wang, Z., Sun, Y., Chen, J. Animal models of ocular angiogenesis: from development to pathologies.


Asunto(s)
Neovascularización Coroidal , Retinopatía Diabética , Modelos Animales de Enfermedad , Neovascularización Retiniana , Animales , Neovascularización Coroidal/genética , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/patología , Neovascularización Coroidal/fisiopatología , Retinopatía Diabética/genética , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Retinopatía Diabética/fisiopatología , Humanos , Ratones , Ratones Transgénicos , Neovascularización Retiniana/genética , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología , Neovascularización Retiniana/fisiopatología
5.
FASEB J ; 31(10): 4492-4502, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28646017

RESUMEN

Pathological proliferation of retinal blood vessels commonly causes vision impairment in proliferative retinopathies, including retinopathy of prematurity. Dysregulated crosstalk between the vasculature and retinal neurons is increasingly recognized as a major factor contributing to the pathogenesis of vascular diseases. Class 3 semaphorins (SEMA3s), a group of neuron-secreted axonal and vascular guidance factors, suppress pathological vascular growth in retinopathy. However, the upstream transcriptional regulators that mediate the function of SEMA3s in vascular growth are poorly understood. Here we showed that retinoic acid receptor-related orphan receptor α (RORα), a nuclear receptor and transcription factor, is a novel transcriptional regulator of SEMA3E-mediated neurovascular coupling in a mouse model of oxygen-induced proliferative retinopathy. We found that genetic deficiency of RORα substantially induced Sema3e expression in retinopathy. Both RORα and SEMA3E were expressed in retinal ganglion cells. RORα directly bound to a specific ROR response element on the promoter of Sema3e and negatively regulated Sema3e promoter-driven luciferase expression. Suppression of Sema3e using adeno-associated virus 2 carrying short hairpin RNA targeting Sema3e promoted disoriented pathological neovascularization and partially abolished the inhibitory vascular effects of RORα deficiency in retinopathy. Our findings suggest that RORα is a novel transcriptional regulator of SEMA3E-mediated neurovascular coupling in pathological retinal angiogenesis.-Sun, Y., Liu, C.-H., Wang, Z., Meng, S. S., Burnim, S. B., SanGiovanni, J. P., Kamenecka, T. M., Solt, L. A., Chen, J. RORα modulates semaphorin 3E transcription and neurovascular interaction in pathological retinal angiogenesis.


Asunto(s)
Glicoproteínas/genética , Proteínas de la Membrana/genética , Neovascularización Patológica/metabolismo , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Neovascularización Retiniana/metabolismo , Vasos Retinianos/metabolismo , Animales , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas del Citoesqueleto , Células Endoteliales/metabolismo , Glicoproteínas/metabolismo , Proteínas de la Membrana/metabolismo , Ratones Transgénicos , Neovascularización Patológica/genética , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Células Ganglionares de la Retina , Neovascularización Retiniana/genética , Semaforinas
6.
Proc Natl Acad Sci U S A ; 112(39): 12163-8, 2015 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-26374840

RESUMEN

Pathologic ocular neovascularization commonly causes blindness. It is critical to identify the factors altered in pathologically proliferating versus normally quiescent vessels to develop effective targeted therapeutics. MicroRNAs regulate both physiological and pathological angiogenesis through modulating expression of gene targets at the posttranscriptional level. However, it is not completely understood if specific microRNAs are altered in pathologic ocular blood vessels, influencing vascular eye diseases. Here we investigated the potential role of a specific microRNA, miR-150, in regulating ocular neovascularization. We found that miR-150 was highly expressed in normal quiescent retinal blood vessels and significantly suppressed in pathologic neovessels in a mouse model of oxygen-induced proliferative retinopathy. MiR-150 substantially decreased endothelial cell function including cell proliferation, migration, and tubular formation and specifically suppressed the expression of multiple angiogenic regulators, CXCR4, DLL4, and FZD4, in endothelial cells. Intravitreal injection of miR-150 mimic significantly decreased pathologic retinal neovascularization in vivo in both wild-type and miR-150 knockout mice. Loss of miR-150 significantly promoted angiogenesis in aortic rings and choroidal explants ex vivo and laser-induced choroidal neovascularization in vivo. In conclusion, miR-150 is specifically enriched in quiescent normal vessels and functions as an endothelium-specific endogenous inhibitor of pathologic ocular neovascularization.


Asunto(s)
Neovascularización Coroidal/genética , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , MicroARNs/metabolismo , MicroARNs/farmacología , Neovascularización Retiniana/genética , Vasos Retinianos/citología , Regiones no Traducidas 3'/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Secuencia de Bases , Proteínas de Unión al Calcio , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neovascularización Coroidal/metabolismo , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Inyecciones Intravítreas , Captura por Microdisección con Láser , Luciferasas , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , MicroARNs/administración & dosificación , MicroARNs/genética , Datos de Secuencia Molecular , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Neovascularización Retiniana/metabolismo , Vasos Retinianos/metabolismo
7.
Proc Natl Acad Sci U S A ; 112(33): 10401-6, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26243880

RESUMEN

Pathologic ocular angiogenesis is a leading cause of blindness, influenced by both dysregulated lipid metabolism and inflammation. Retinoic-acid-receptor-related orphan receptor alpha (RORα) is a lipid-sensing nuclear receptor with diverse biologic function including regulation of lipid metabolism and inflammation; however, its role in pathologic retinal angiogenesis remains poorly understood. Using a mouse model of oxygen-induced proliferative retinopathy, we showed that RORα expression was significantly increased and genetic deficiency of RORα substantially suppressed pathologic retinal neovascularization. Loss of RORα led to decreased levels of proinflammatory cytokines and increased levels of antiinflammatory cytokines in retinopathy. RORα directly suppressed the gene transcription of suppressors of cytokine signaling 3 (SOCS3), a critical negative regulator of inflammation. Inhibition of SOCS3 abolished the antiinflammatory and vasoprotective effects of RORα deficiency in vitro and in vivo. Moreover, treatment with a RORα inverse agonist SR1001 effectively protected against pathologic neovascularization in both oxygen-induced retinopathy and another angiogenic model of very-low-density lipoprotein receptor (Vldlr)-deficient (Vldlr (-/-) ) mice with spontaneous subretinal neovascularization, whereas a RORα agonist worsened oxygen-induced retinopathy. Our data demonstrate that RORα is a novel regulator of pathologic retinal neovascularization, and RORα inhibition may represent a new way to treat ocular neovascularization.


Asunto(s)
Neovascularización Patológica , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Retina/patología , Neovascularización Retiniana , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Animales , Antiinflamatorios/química , Proliferación Celular , Inmunoprecipitación de Cromatina , Citocinas/metabolismo , Heterocigoto , Homocigoto , Inflamación/patología , Lípidos/química , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Oxígeno/química , Unión Proteica , ARN Mensajero/metabolismo , Receptores de LDL/genética , Proteína 3 Supresora de la Señalización de Citocinas
8.
Am J Pathol ; 186(10): 2588-600, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27524797

RESUMEN

Familial exudative vitreoretinopathy (FEVR) is characterized by delayed retinal vascular development, which promotes hypoxia-induced pathologic vessels. In severe cases FEVR may lead to retinal detachment and visual impairment. Genetic studies linked FEVR with mutations in Wnt signaling ligand or receptors, including low-density lipoprotein receptor-related protein 5 (LRP5) gene. Here, we investigated ocular pathologies in a Lrp5 knockout (Lrp5(-/-)) mouse model of FEVR and explored whether treatment with a pharmacologic Wnt activator lithium could bypass the genetic defects, thereby protecting against eye pathologies. Lrp5(-/-) mice displayed significantly delayed retinal vascular development, absence of deep layer retinal vessels, leading to increased levels of vascular endothelial growth factor and subsequent pathologic glomeruloid vessels, as well as decreased inner retinal visual function. Lithium treatment in Lrp5(-/-) mice significantly restored the delayed development of retinal vasculature and the intralaminar capillary networks, suppressed formation of pathologic glomeruloid structures, and promoted hyaloid vessel regression. Moreover, lithium treatment partially rescued inner-retinal visual function and increased retinal thickness. These protective effects of lithium were largely mediated through restoration of canonical Wnt signaling in Lrp5(-/-) retina. Lithium treatment also substantially increased vascular tubular formation in LRP5-deficient endothelial cells. These findings suggest that pharmacologic activation of Wnt signaling may help treat ocular pathologies in FEVR and potentially other defective Wnt signaling-related diseases.


Asunto(s)
Litio/farmacología , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Enfermedades de la Retina/tratamiento farmacológico , Vía de Señalización Wnt/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Enfermedades Hereditarias del Ojo , Vitreorretinopatías Exudativas Familiares , Femenino , Humanos , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Retina/efectos de los fármacos , Retina/embriología , Enfermedades de la Retina/genética , Enfermedades de la Retina/patología , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/embriología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 36(9): 1919-27, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27417579

RESUMEN

OBJECTIVE: Pathological ocular neovascularization is a major cause of blindness. Increased dietary intake of ω-3 long-chain polyunsaturated fatty acids (LCPUFA) reduces retinal neovascularization and choroidal neovascularization (CNV), but ω-3 LCPUFA metabolites of a major metabolizing pathway, cytochrome P450 oxidase (CYP) 2C, promote ocular pathological angiogenesis. We hypothesized that inhibition of CYP2C activity will add to the protective effects of ω-3 LCPUFA on neovascular eye diseases. APPROACH AND RESULTS: The mouse models of oxygen-induced retinopathy and laser-induced CNV were used to investigate pathological angiogenesis in the retina and choroid, respectively. The plasma levels of ω-3 LCPUFA metabolites of CYP2C were determined by mass spectroscopy. Aortic ring and choroidal explant sprouting assays were used to investigate the effects of CYP2C inhibition and ω-3 LCPUFA-derived CYP2C metabolic products on angiogenesis ex vivo. We found that inhibition of CYP2C activity by montelukast added to the protective effects of ω-3 LCPUFA on retinal neovascularization and CNV by 30% and 20%, respectively. In CYP2C8-overexpressing mice fed a ω-3 LCPUFA diet, montelukast suppressed retinal neovascularization and CNV by 36% and 39% and reduced the plasma levels of CYP2C8 products. Soluble epoxide hydrolase inhibition, which blocks breakdown and inactivation of CYP2C ω-3 LCPUFA-derived active metabolites, increased oxygen-induced retinopathy and CNV in vivo. Exposure to selected ω-3 LCPUFA metabolites of CYP2C significantly reversed the suppression of both angiogenesis ex vivo and endothelial cell functions in vitro by the CYP2C inhibitor montelukast. CONCLUSIONS: Inhibition of CYP2C activity adds to the protective effects of ω-3 LCPUFA on pathological retinal neovascularization and CNV.


Asunto(s)
Acetatos/farmacología , Inhibidores de la Angiogénesis/farmacología , Neovascularización Coroidal/prevención & control , Inhibidores del Citocromo P-450 CYP2C8/farmacología , Citocromo P-450 CYP2C8/metabolismo , Ácidos Grasos Omega-3/farmacología , Quinolinas/farmacología , Neovascularización Retiniana/prevención & control , Retinopatía de la Prematuridad/prevención & control , Animales , Aorta/efectos de los fármacos , Aorta/enzimología , Células Cultivadas , Neovascularización Coroidal/enzimología , Neovascularización Coroidal/genética , Neovascularización Coroidal/fisiopatología , Ciclopropanos , Citocromo P-450 CYP2C8/genética , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Ácidos Grasos Omega-3/metabolismo , Genotipo , Humanos , Hiperoxia/complicaciones , Rayos Láser , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Fisiológica/efectos de los fármacos , Fenotipo , Neovascularización Retiniana/enzimología , Neovascularización Retiniana/genética , Neovascularización Retiniana/fisiopatología , Retinopatía de la Prematuridad/enzimología , Retinopatía de la Prematuridad/genética , Retinopatía de la Prematuridad/fisiopatología , Sulfuros , Técnicas de Cultivo de Tejidos
10.
Exp Eye Res ; 110: 18-25, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23500521

RESUMEN

α-Internexin, a neuronal intermediate filament (IF) protein that is expressed in most neurons at the beginning of differentiation, has been studied in the developing vertebrate retina. It has been suggested that α-internexin plays a role in neuroplasticity during development. However, the expression of α-internexin in the chicken retina has not been investigated. In this study, we identified the expression patterns of the chicken α-internexin (chkINA) and neurofilament (NF) proteins in the developing chicken retina. These proteins exhibited dynamic patterns of expression with the progression of retinal development. Expression of chkINA was detected in the processes of ganglion, amacrine, and horizontal cells throughout development. chkINA was also transiently expressed in photoreceptor and bipolar cell lineages. NF triplet proteins exhibited more restricted expression patterns than did chkINA during development. Our observations suggest the coexpression of chkINA and NF triplet proteins in ganglion and amacrine cells in the developing retina, as well as in horizontal cell processes in the adult chicken retina. Our results indicate that chkINA is transiently expressed in all neuronal lineages in the developing chicken retina. These findings provide novel information on chicken retinal cell development and suggest that chkINA is a good neuronal marker for the study of retinal differentiation.


Asunto(s)
Proteínas del Ojo/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Proteínas de Neurofilamentos/metabolismo , Retina/embriología , Neuronas Retinianas/metabolismo , Células Amacrinas/metabolismo , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Linaje de la Célula , Embrión de Pollo , Técnica del Anticuerpo Fluorescente Indirecta , Proteínas de Homeodominio/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/metabolismo , Proteína Quinasa C-alfa/metabolismo , Proteínas Represoras/metabolismo , Retina/citología , Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Células Horizontales de la Retina/metabolismo , Neuronas Retinianas/citología , Proteínas Supresoras de Tumor/metabolismo
11.
Neural Regen Res ; 18(10): 2173-2181, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37056126

RESUMEN

Age-related macular degeneration is a primary cause of blindness in the older adult population. Past decades of research in the pathophysiology of the disease have resulted in breakthroughs in the form of anti-vascular endothelial growth factor therapies against neovascular age-related macular degeneration; however, effective treatment is not yet available for geographical atrophy in dry age-related macular degeneration or for preventing the progression from early or mid to the late stage of age-related macular degeneration. Both clinical and experimental investigations involving human age-related macular degeneration retinas and animal models point towards the atrophic alterations in retinal pigment epithelium as a key feature in age-related macular degeneration progression. Retinal pigment epithelium cells are primarily responsible for cellular-structural maintenance and nutrition supply to keep photoreceptors healthy and functional. The retinal pigment epithelium constantly endures a highly oxidative environment that is balanced with a cascade of antioxidant enzyme systems regulated by nuclear factor erythroid-2-related factor 2 as a main redox sensing transcription factor. Aging and accumulated oxidative stress triggers retinal pigment epithelium dysfunction and eventually death. Exposure to both environmental and genetic factors aggravates oxidative stress damage in aging retinal pigment epithelium and accelerates retinal pigment epithelium degeneration in age-related macular degeneration pathophysiology. The present review summarizes the role of oxidative stress in retinal pigment epithelium degeneration, with potential impacts from both genetic and environmental factors in age-related macular degeneration development and progression. Potential strategies to counter retinal pigment epithelium damage and protect the retinal pigment epithelium through enhancing its antioxidant capacity are also discussed, focusing on existing antioxidant nutritional supplementation, and exploring nuclear factor erythroid-2-related factor 2 and its regulators including REV-ERBα as therapeutic targets to protect against age-related macular degeneration development and progression.

12.
Aging (Albany NY) ; 15(1): 37-52, 2023 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-36626253

RESUMEN

Choroidal neovascularization (CNV) causes acute vision loss in neovascular age-related macular degeneration (AMD). Genetic variations of the nuclear receptor RAR-related orphan receptor alpha (RORα) have been linked with neovascular AMD, yet its specific role in pathological CNV development is not entirely clear. In this study, we showed that Rora was highly expressed in the mouse choroid compared with the retina, and genetic loss of RORα in Staggerer mice (Rorasg/sg) led to increased expression levels of Vegfr2 and Tnfa in the choroid and retinal pigment epithelium (RPE) complex. In a mouse model of laser-induced CNV, RORα expression was highly increased in the choroidal/RPE complex post-laser, and loss of RORα in Rorasg/sg eyes significantly worsened CNV with increased lesion size and vascular leakage, associated with increased levels of VEGFR2 and TNFα proteins. Pharmacological inhibition of RORα also worsened CNV. In addition, both genetic deficiency and inhibition of RORα substantially increased vascular growth in isolated mouse choroidal explants ex vivo. RORα inhibition also promoted angiogenic function of human choroidal endothelial cell culture. Together, our results suggest that RORα negatively regulates pathological CNV development in part by modulating angiogenic response of the choroidal endothelium and inflammatory environment in the choroid/RPE complex.


Asunto(s)
Neovascularización Coroidal , Degeneración Macular Húmeda , Ratones , Humanos , Animales , Inhibidores de la Angiogénesis , Factor A de Crecimiento Endotelial Vascular/metabolismo , Agudeza Visual , Degeneración Macular Húmeda/complicaciones , Neovascularización Coroidal/genética , Neovascularización Coroidal/tratamiento farmacológico , Rayos Láser , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
13.
Redox Biol ; 51: 102261, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35176707

RESUMEN

Retinal pigment epithelium (RPE) dysfunction and atrophy occur in dry age-related macular degeneration (AMD), often leading to photoreceptor degeneration and vision loss. Accumulated oxidative stress during aging contributes to RPE dysfunction and degeneration. Here we show that the nuclear receptor REV-ERBα, a redox sensitive transcription factor, protects RPE from age-related degeneration and oxidative stress-induced damage. Genetic deficiency of REV-ERBα leads to accumulated oxidative stress, dysfunction and degeneration of RPE, and AMD-like ocular pathologies in aging mice. Loss of REV-ERBα exacerbates chemical-induced RPE damage, and pharmacological activation of REV-ERBα protects RPE from oxidative damage both in vivo and in vitro. REV-ERBα directly regulates transcription of nuclear factor erythroid 2-related factor 2 (NRF2) and its downstream antioxidant enzymes superoxide dismutase 1 (SOD1) and catalase to counter oxidative damage. Moreover, aged mice with RPE specific knockout of REV-ERBα also exhibit accumulated oxidative stress and fundus and RPE pathologies. Together, our results suggest that REV-ERBα is a novel intrinsic protector of the RPE against age-dependent oxidative stress and a new molecular target for developing potential therapies to treat age-related retinal degeneration.


Asunto(s)
Degeneración Macular , Degeneración Retiniana , Animales , Degeneración Macular/genética , Degeneración Macular/patología , Ratones , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares , Estrés Oxidativo/fisiología , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Epitelio Pigmentado de la Retina/metabolismo
14.
Elife ; 112022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36454214

RESUMEN

Amino acid (AA) metabolism in vascular endothelium is important for sprouting angiogenesis. SLC38A5 (solute carrier family 38 member 5), an AA transporter, shuttles neutral AAs across cell membrane, including glutamine, which may serve as metabolic fuel for proliferating endothelial cells (ECs) to promote angiogenesis. Here, we found that Slc38a5 is highly enriched in normal retinal vascular endothelium, and more specifically, in pathological sprouting neovessels. Slc38a5 is suppressed in retinal blood vessels from Lrp5-/- and Ndpy/- mice, both genetic models of defective retinal vascular development with Wnt signaling mutations. Additionally, Slc38a5 transcription is regulated by Wnt/ß-catenin signaling. Genetic deficiency of Slc38a5 in mice substantially delays retinal vascular development and suppresses pathological neovascularization in oxygen-induced retinopathy modeling ischemic proliferative retinopathies. Inhibition of SLC38A5 in human retinal vascular ECs impairs EC proliferation and angiogenic function, suppresses glutamine uptake, and dampens vascular endothelial growth factor receptor 2. Together these findings suggest that SLC38A5 is a new metabolic regulator of retinal angiogenesis by controlling AA nutrient uptake and homeostasis in ECs.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros , Células Endoteliales , Humanos , Ratones , Animales , Glutamina , Factor A de Crecimiento Endotelial Vascular , Neovascularización Patológica/genética , Sistemas de Transporte de Aminoácidos
15.
Artículo en Inglés | MEDLINE | ID: mdl-31835092

RESUMEN

Deficiency in retinoid acid receptor-related orphan receptor alpha (RORα) of staggerer mice results in extensive granule and Purkinje cell loss in the cerebellum as well as in learned motor deficits, cognition impairments and perseverative tendencies that are commonly observed in autistic spectrum disorder (ASD). The effects of RORα on brain lipid metabolism associated with cerebellar atrophy remain unexplored. The aim of this study is to examine the effects of RORα deficiency on brain phospholipid fatty acid concentrations and compositions. Staggerer mice (Rorasg/sg) and wildtype littermates (Rora+/+) were fed n-3 polyunsaturated fatty acids (PUFA) containing diets ad libitum. At 2 months and 7 or more months old, brain total phospholipid fatty acids were quantified by gas chromatography-flame ionization detection. In the cerebellum, all fatty acid concentrations were reduced in 2 months old mice. Since total fatty acid concentrations were significantly different at 2-month-old, we examined changes in fatty acid composition. The composition of ARA was not significantly different between genotypes; though DHA composition remained significantly lowered. Despite cerebellar atrophy at >7-months-old, cerebellar fatty acid concentrations had recovered comparably to wildtype control. Therefore, RORα may be necessary for fatty acid accretions during neurodevelopment. Specifically, the effects of RORα on PUFA metabolisms are region-specific and age-dependent.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Ácidos Docosahexaenoicos/metabolismo , Ácidos Grasos Omega-3/administración & dosificación , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/deficiencia , Animales , Encéfalo/metabolismo , Cromatografía de Gases , Femenino , Metabolismo de los Lípidos , Masculino , Ratones , Fosfolípidos/metabolismo
16.
Sci Adv ; 6(35): eaba7457, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32923627

RESUMEN

Breakdown of the blood-retinal barrier (BRB) causes retinal edema and vision loss. We investigated the role of Wnt signaling in maintaining the BRB by limiting transcytosis. Mice lacking either the Wnt co-receptor low-density lipoprotein receptor-related protein 5 (Lrp5-/- ) or the Wnt ligand Norrin (Ndpy/- ) exhibit increased retinal vascular leakage and enhanced endothelial transcytosis. Wnt signaling directly controls the transcription of an endothelium-specific transcytosis inhibitor, major facilitator superfamily domain-containing protein 2a (MFSD2A), in a ß-catenin-dependent manner. MFSD2A overexpression reverses Wnt deficiency-induced transcytosis in endothelial cells and in retinas. Moreover, Wnt signaling mediates MFSD2A-dependent vascular endothelium transcytosis through a caveolin-1 (CAV-1)-positive caveolae pathway. In addition, levels of omega-3 fatty acids are also decreased in Wnt signaling-deficient retinas, reflecting the basic function of MFSD2A as a lipid transporter. Our findings uncovered the Wnt/ß-catenin/MFSD2A/CAV-1 axis as a key pathway governing endothelium transcytosis and inner BRB integrity.

17.
Prog Retin Eye Res ; 70: 110-133, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30513356

RESUMEN

The Wnt signaling pathway plays a pivotal role in vascular morphogenesis in various organs including the eye. Wnt ligands and receptors are key regulators of ocular angiogenesis both during the eye development and in vascular eye diseases. Wnt signaling participates in regulating multiple vascular beds in the eye including regression of the hyaloid vessels, and development of structured layers of vasculature in the retina. Loss-of-function mutations in Wnt signaling components cause rare genetic eye diseases in humans such as Norrie disease, and familial exudative vitreoretinopathy (FEVR) with defective ocular vasculature. On the other hand, experimental studies in more prevalent vascular eye diseases, such as wet age-related macular degeneration (AMD), diabetic retinopathy (DR), retinopathy of prematurity (ROP), and corneal neovascularization, suggest that aberrantly increased Wnt signaling is one of the causations for pathological ocular neovascularization, indicating the potential of modulating Wnt signaling to ameliorate pathological angiogenesis in eye diseases. This review recapitulates the key roles of the Wnt signaling pathway during ocular vascular development and in vascular eye diseases, and pharmaceutical approaches targeting the Wnt signaling as potential treatment options.


Asunto(s)
Enfermedades de la Retina/fisiopatología , Vasos Retinianos/fisiopatología , Vía de Señalización Wnt/fisiología , Animales , Humanos
18.
J Vis Exp ; (147)2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31157789

RESUMEN

In the eye, the embryonic hyaloid vessels nourish the developing lens and retina and regress when the retinal vessels develop. Persistent or failed regression of hyaloid vessels can be seen in diseases such as persistent hyperplastic primary vitreous (PHPV), leading to an obstructed light path and impaired visual function. Understanding the mechanisms underlying the hyaloid vessel regression may lead to new molecular insights into the vascular regression process and potential new ways to manage diseases with persistent hyaloid vessels. Here we describe the procedures for imaging hyaloid in live mice with optical coherence tomography (OCT) and fundus fluorescein angiography (FFA) and a detailed technical protocol of isolating and flat-mounting hyaloid ex vivo for quantitative analysis. Low-density lipoprotein receptor-related protein 5 (LRP5) knockout mice were used as an experimental model of persistent hyaloid vessels, to illustrate the techniques. Together, these techniques may facilitate a thorough assessment of hyaloid vessels as an experimental model of vascular regression and studies on the mechanism of persistent hyaloid vessels.


Asunto(s)
Vasos Retinianos/anatomía & histología , Animales , Angiografía con Fluoresceína , Imagenología Tridimensional , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/deficiencia , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Vasos Retinianos/diagnóstico por imagen , Tomografía de Coherencia Óptica
19.
Mol Ther Nucleic Acids ; 16: 335-347, 2019 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-30981984

RESUMEN

Pathological angiogenesis is a hallmark of various vascular diseases, including vascular eye disorders. Dysregulation of microRNAs (miRNAs), a group of small regulatory RNAs, has been implicated in the regulation of ocular neovascularization. This study investigated the specific role of microRNA-145 (miR-145) in regulating vascular endothelial cell (EC) function and pathological ocular angiogenesis in a mouse model of oxygen-induced retinopathy (OIR). Expression of miR-145 was significantly upregulated in OIR mouse retinas compared with room air controls. Treatment with synthetic miR-145 inhibitors drastically decreased levels of pathological neovascularization in OIR, without substantially affecting normal developmental angiogenesis. In cultured human retinal ECs, treatment with miR-145 mimics significantly increased the EC angiogenic function, including proliferation, migration, and tubular formation, whereas miR-145 inhibitors attenuated in vitro angiogenesis. Tropomodulin3 (TMOD3), an actin-capping protein, is a direct miR-145 target and is downregulated in OIR retinas. Treatment with miR-145 mimic led to TMOD3 inhibition, altered actin cytoskeletal architecture, and elongation of ECs. Moreover, inhibition of TMOD3 promoted EC angiogenic function and pathological neovascularization in OIR and abolished the vascular effects of miR-145 inhibitors in vitro and in vivo. Overall, our findings indicate that miR-145 is a novel regulator of TMOD3-dependent cytoskeletal architecture and pathological angiogenesis and a potential target for development of treatments for neovascular eye disorders.

20.
Nat Commun ; 10(1): 804, 2019 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-30778060

RESUMEN

Choroidal neovascularization (CNV) is the major cause of vision loss in wet age-related macular degeneration (AMD). Current therapies require repeated intravitreal injections, which are painful and can cause infection, bleeding, and retinal detachment. Here we develop nanoparticles (NP-[CPP]) that can be administered intravenously and allow local drug delivery to the diseased choroid via light-triggered targeting. NP-[CPP] is formed by PEG-PLA chains modified with a cell penetrating peptide (CPP). Attachment of a DEACM photocleavable group to the CPP inhibits cellular uptake of NP-[CPP]. Irradiation with blue light cleaves DEACM from the CPP, allowing the CPP to migrate from the NP core to the surface, rendering it active. In mice with laser-induced CNV, intravenous injection of NP-[CPP] coupled to irradiation of the eye allows NP accumulation in the neovascular lesions. When loaded with doxorubicin, irradiated NP-[CPP] significantly reduces neovascular lesion size. We propose a strategy for non-invasive treatment of CNV and enhanced drug accumulation specifically in diseased areas of the eye.


Asunto(s)
Neovascularización Coroidal/tratamiento farmacológico , Nanopartículas/administración & dosificación , Nanopartículas/química , Administración Intravenosa , Animales , Péptidos de Penetración Celular/química , Neovascularización Coroidal/terapia , Cumarinas/química , Modelos Animales de Enfermedad , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacocinética , Sistemas de Liberación de Medicamentos/métodos , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Luz , Ratones Endogámicos C57BL , Fototerapia/métodos , Polietilenglicoles , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA