Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 149(4): 847-59, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22541070

RESUMEN

Alu RNA accumulation due to DICER1 deficiency in the retinal pigmented epithelium (RPE) is implicated in geographic atrophy (GA), an advanced form of age-related macular degeneration that causes blindness in millions of individuals. The mechanism of Alu RNA-induced cytotoxicity is unknown. Here we show that DICER1 deficit or Alu RNA exposure activates the NLRP3 inflammasome and triggers TLR-independent MyD88 signaling via IL18 in the RPE. Genetic or pharmacological inhibition of inflammasome components (NLRP3, Pycard, Caspase-1), MyD88, or IL18 prevents RPE degeneration induced by DICER1 loss or Alu RNA exposure. These findings, coupled with our observation that human GA RPE contains elevated amounts of NLRP3, PYCARD, and IL18 and evidence of increased Caspase-1 and MyD88 activation, provide a rationale for targeting this pathway in GA. Our findings also reveal a function of the inflammasome outside the immune system and an immunomodulatory action of mobile elements.


Asunto(s)
Elementos Alu , ARN Helicasas DEAD-box/metabolismo , Atrofia Geográfica/inmunología , Atrofia Geográfica/patología , Inflamasomas/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Ribonucleasa III/metabolismo , Animales , Proteínas Portadoras/metabolismo , Atrofia Geográfica/metabolismo , Humanos , Inflamasomas/metabolismo , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , Epitelio Pigmentado de la Retina/patología , Receptores Toll-Like/metabolismo
2.
Proc Natl Acad Sci U S A ; 117(5): 2579-2587, 2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-31964819

RESUMEN

Degeneration of the retinal pigmented epithelium (RPE) and aberrant blood vessel growth in the eye are advanced-stage processes in blinding diseases such as age-related macular degeneration (AMD), which affect hundreds of millions of people worldwide. Loss of the RNase DICER1, an essential factor in micro-RNA biogenesis, is implicated in RPE atrophy. However, the functional implications of DICER1 loss in choroidal and retinal neovascularization are unknown. Here, we report that two independent hypomorphic mouse strains, as well as a separate model of postnatal RPE-specific DICER1 ablation, all presented with spontaneous RPE degeneration and choroidal and retinal neovascularization. DICER1 hypomorphic mice lacking critical inflammasome components or the innate immune adaptor MyD88 developed less severe RPE atrophy and pathological neovascularization. DICER1 abundance was also reduced in retinas of the JR5558 mouse model of spontaneous choroidal neovascularization. Finally, adenoassociated vector-mediated gene delivery of a truncated DICER1 variant (OptiDicer) reduced spontaneous choroidal neovascularization in JR5558 mice. Collectively, these findings significantly expand the repertoire of DICER1 in preserving retinal homeostasis by preventing both RPE degeneration and pathological neovascularization.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Degeneración Macular/metabolismo , Epitelio Pigmentado de la Retina/irrigación sanguínea , Ribonucleasa III/metabolismo , Animales , Neovascularización Coroidal/genética , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/patología , Neovascularización Coroidal/fisiopatología , ARN Helicasas DEAD-box/genética , Humanos , Degeneración Macular/genética , Degeneración Macular/patología , Degeneración Macular/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Degeneración Retiniana/fisiopatología , Neovascularización Retiniana/genética , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/parasitología , Neovascularización Retiniana/fisiopatología , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Ribonucleasa III/genética
3.
Exp Eye Res ; 178: 91-98, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30268699

RESUMEN

Optical coherence tomography (OCT) angiography is a dye-free and non-invasive angiography which allows visualization of retinal and choroid vascular flow, enabling observation of highly permeable and three dimensional vasculature. Although OCT angiography is providing new insights in human retinal and choroidal diseases, a few studies have been reported in experimental mice. In this study, to determine the potential of OCT angiography in experimental mice, we sought to examine whether OCT angiography can detect vascular change in type I diabetic mice. To conduct age dependent analysis, 2 and 6 month old male type 1 diabetic Ins2Akita/+ and age matched C57BL/6J mice were used. OCT angiography was performed by Heidelberg Spectralis OCT Angiography Module with 30° lens + mouse adapter lens. We acquired the OCT angiography image from the peripheral nasal position. For analysis of OCT angiography images, OCT angiography positive area were used for vascular density. We analyzed vascular density from the retinal surface (inner limiting membrane) to 120 µm depth with 4 µm steps in order to correlate vascular density vs depth (N = 4 per group). Vascular density of both mouse strains demonstrated three different peaks. By comparing with the OCT image, the first peak (superficial), second peak (intermediate) and third peak (deep) were located in nerve fiber layer/ganglion cell layer, inner plexiform layer/inner nuclear layer and outer plexiform layer/outer nuclear layer, respectively. We calculated vascular density of these peaks separately. In C57BL/6J mice, the vascular density in all three layers do not show significant difference between 2- and 6-month-old. On the other hand, 6-month-old Ins2Akita/+ mice showed a significant decrease of the vascular density in all three layers compared to 2-month-old Ins2Akita/+ mice. Also, the vascular density of 6-month-old Ins2Akita/+ mice in the deep layer showed a significant decrease compared to 2- and 6-month-old C57BL/6J mice. Thus, OCT angiography successfully detects retinal vascular difference between type I diabetic mice and control mice, and age-dependent vasculature change in type I diabetic mice. The diabetic mice demonstrated reduced vascular density due to reduced density of flowing deep vessels. Importantly, we observed this difference without retinal blood leakage, hemorrhage or neovascularization. Our analysis (vascular density vs retinal depth) suggests that OCT angiography is useful for in vivo detection of retinal vasculature alteration in experimental mice.


Asunto(s)
Diabetes Mellitus Tipo 1/diagnóstico , Retinopatía Diabética/diagnóstico , Vasos Retinianos/patología , Envejecimiento/fisiología , Animales , Diabetes Mellitus Experimental/diagnóstico , Angiografía con Fluoresceína , Masculino , Ratones , Ratones Endogámicos C57BL , Microvasos/patología , Vasos Retinianos/diagnóstico por imagen , Tomografía de Coherencia Óptica
4.
Int Ophthalmol ; 39(12): 2975-2983, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31313070

RESUMEN

PURPOSE: To describe and evaluate the value of nutritional supplements in the management of age-related macular degeneration (AMD) through a review of the current literature. METHODS: An extensive literature search was performed, and key research articles exploring AREDS and AREDS-2 formulations, genetics, omega fatty acids, calcium and folic acid in high-risk women were reviewed. PubMed and Web of Science databases were used for generating articles to review. RESULTS: The AREDS and AREDS-2 trials, while difficult to validate, show support for antioxidant supplementation in reducing AMD progression in Caucasian populations. While genetic guided personalized medicine has been studied mainly with complement factor H and age-related maculopathy susceptibility 2 risk alleles, the data have not been reproducible. Women at a higher risk of cardiovascular disease may benefit from antioxidant therapies in preventing AMD. Omega 3 fatty acid supplementation has been widely supported through observational studies; however, randomized controlled trials have not shown benefit in disease progression. Calcium exposure has been linked to increased mechanisms in cell death and may be detrimental to older individuals with AMD. CONCLUSION: The data regarding nutritional supplements in preventing AMD progression are inconclusive, and therefore recommendations should be based on risk factors and demographic data.


Asunto(s)
Antioxidantes/uso terapéutico , Suplementos Dietéticos , Degeneración Macular/terapia , Vitaminas/uso terapéutico , Progresión de la Enfermedad , Humanos
5.
Mol Ther ; 25(7): 1606-1615, 2017 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-28236576

RESUMEN

Neovascular age-related macular degeneration (AMD) is treated with anti-VEGF intravitreal injections, which can cause geographic atrophy, infection, and retinal fibrosis. To minimize these toxicities, we developed a nanoparticle delivery system for recombinant Flt23k intraceptor plasmid (RGD.Flt23k.NP) to suppress VEGF intracellularly within choroidal neovascular (CNV) lesions in a laser-induced CNV mouse model through intravenous administration. In the current study, we examined the efficacy and safety of RGD.Flt23k.NP in mice. The effect of various doses was determined using fluorescein angiography and optical coherence tomography to evaluate CNV leakage and volume. Efficacy was determined by the rate of inhibition of CNV volume at 2 weeks post-treatment. RGD.Flt23k.NP had peak efficacy at a dose range of 30-60 µg pFlt23k/mouse. Using the lower dose (30 µg pFlt23k/mouse), RGD.Flt23k.NP safety was determined both in single-dose groups and in repeat-dose (three times) groups by measuring body weight, organ weight, hemoglobin levels, complement C3 levels, and histological changes in vital organs. Neither toxicity nor inflammation from RGD.Flt23k.NP was detected. No side effect was detected on visual function. Thus, systemic RGD.Flt23k.NP may be an alternative to standard intravitreal anti-VEGF therapy for the treatment of neovascular AMD.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Neovascularización Coroidal/terapia , Portadores de Fármacos , Degeneración Macular/terapia , Plásmidos/metabolismo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/química , Animales , Coroides/irrigación sanguínea , Coroides/metabolismo , Coroides/patología , Neovascularización Coroidal/genética , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/patología , Complemento C3/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Femenino , Regulación de la Expresión Génica , Hemoglobinas/metabolismo , Humanos , Inyecciones Intravenosas , Inyecciones Intravítreas , Rayos Láser , Degeneración Macular/genética , Degeneración Macular/metabolismo , Degeneración Macular/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Nanopartículas/administración & dosificación , Nanopartículas/química , Plásmidos/química , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
Int Ophthalmol ; 38(5): 2257-2266, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28852910

RESUMEN

PURPOSE: To describe the epidemiology and prevalence, rates of progression, difference between adult and pediatric populations, and therapeutic approaches to pediatric keratoconus from documented literature. METHODS: A literature search was done on PubMed using key words including pediatric keratoconus, children with keratoconus, adult keratoconus, penetrating keratoplasty, corneal cross-linking and intracorneal ring segments. The literature was reviewed and reported to explore the key epidemiological differences between the pediatric and adult population with regards to presentation and treatment options. RESULTS: Pediatric keratoconus is more aggressive than adult keratoconus, which has been explained by structural differences in the cornea between both populations. High rates of progression were documented in pediatric populations. While corneal collagen cross-linking, intracorneal ring segments and penetrating keratoplasties have been used as therapies in the pediatric population, the literature overwhelmingly shows higher rates of failure and progression despite these measures as compared to adults. CONCLUSION: Pediatric keratoconus is more aggressive than adult keratoconus, and current therapies used in adults may not be sufficient for the pediatric population.


Asunto(s)
Colágeno/uso terapéutico , Sustancia Propia/patología , Reactivos de Enlaces Cruzados/uso terapéutico , Queratocono , Queratoplastia Penetrante/métodos , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/uso terapéutico , Niño , Salud Global , Humanos , Incidencia , Queratocono/diagnóstico , Queratocono/epidemiología , Queratocono/terapia
7.
Nature ; 471(7338): 325-30, 2011 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-21297615

RESUMEN

Geographic atrophy (GA), an untreatable advanced form of age-related macular degeneration, results from retinal pigmented epithelium (RPE) cell degeneration. Here we show that the microRNA (miRNA)-processing enzyme DICER1 is reduced in the RPE of humans with GA, and that conditional ablation of Dicer1, but not seven other miRNA-processing enzymes, induces RPE degeneration in mice. DICER1 knockdown induces accumulation of Alu RNA in human RPE cells and Alu-like B1 and B2 RNAs in mouse RPE. Alu RNA is increased in the RPE of humans with GA, and this pathogenic RNA induces human RPE cytotoxicity and RPE degeneration in mice. Antisense oligonucleotides targeting Alu/B1/B2 RNAs prevent DICER1 depletion-induced RPE degeneration despite global miRNA downregulation. DICER1 degrades Alu RNA, and this digested Alu RNA cannot induce RPE degeneration in mice. These findings reveal a miRNA-independent cell survival function for DICER1 involving retrotransposon transcript degradation, show that Alu RNA can directly cause human pathology, and identify new targets for a major cause of blindness.


Asunto(s)
Elementos Alu/genética , ARN Helicasas DEAD-box/deficiencia , Degeneración Macular/genética , Degeneración Macular/patología , ARN/genética , ARN/metabolismo , Ribonucleasa III/deficiencia , Animales , Muerte Celular , Supervivencia Celular , Células Cultivadas , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Ratones , MicroARNs/metabolismo , Datos de Secuencia Molecular , Oligonucleótidos Antisentido , Fenotipo , Epitelio Pigmentado de la Retina/enzimología , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Ribonucleasa III/genética , Ribonucleasa III/metabolismo
8.
Proc Natl Acad Sci U S A ; 111(45): 16082-7, 2014 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-25349431

RESUMEN

Geographic atrophy, an advanced form of age-related macular degeneration (AMD) characterized by death of the retinal pigmented epithelium (RPE), causes untreatable blindness in millions worldwide. The RPE of human eyes with geographic atrophy accumulates toxic Alu RNA in response to a deficit in the enzyme DICER1, which in turn leads to activation of the NLRP3 inflammasome and elaboration of IL-18. Despite these recent insights, it is still unclear how RPE cells die during the course of the disease. In this study, we implicate the involvement of Caspase-8 as a critical mediator of RPE degeneration. Here we show that DICER1 deficiency, Alu RNA accumulation, and IL-18 up-regulation lead to RPE cell death via activation of Caspase-8 through a Fas ligand-dependent mechanism. Coupled with our observation of increased Caspase-8 expression in the RPE of human eyes with geographic atrophy, our findings provide a rationale for targeting this apoptotic pathway in this disease.


Asunto(s)
Elementos Alu , Apoptosis , Caspasa 8/metabolismo , ARN Helicasas DEAD-box/metabolismo , Proteínas del Ojo/metabolismo , Degeneración Macular/metabolismo , ARN/metabolismo , Ribonucleasa III/metabolismo , Animales , Caspasa 8/genética , ARN Helicasas DEAD-box/genética , Proteínas del Ojo/genética , Humanos , Interleucina-18/genética , Interleucina-18/metabolismo , Degeneración Macular/patología , Ratones , Ratones Noqueados , ARN/genética , Ribonucleasa III/genética , Regulación hacia Arriba/genética
9.
Exp Eye Res ; 145: 248-257, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26775053

RESUMEN

To assess whether Tie2-mediated vascular stabilization ameliorates neovascular age-related macular degeneration (AMD), we investigated the impact of adeno-associated virus-mediated gene therapy with cartilage oligomeric matrix protein angiopoietin-1 (AAV2.COMP-Ang1) on choroidal neovascularization (CNV), vascular endothelial growth factor (VEGF), and hypoxia-inducible factor (HIF) in a mouse model of the disease. We treated mice with subretinal injections of AAV2.COMP-Ang1 or control (AAV2.AcGFP, AAV2.LacZ, and phosphate-buffered saline). Subretinal AAV2 localization and plasmid protein expression was verified in the retinal pigment epithelium (RPE)/choroid of mice treated with all AAV2 constructs. Laser-assisted simulation of neovascular AMD was performed and followed by quantification of HIF, VEGF, and CNV in each experimental group. We found that AAV2.COMP-Ang1 was associated with a significant reduction in VEGF levels (29-33%, p < 0.01) and CNV volume (60-70%, p < 0.01), without a concomitant decrease in HIF1-α, compared to all controls. We concluded that a) AAV2 is a viable vector for delivering COMP-Ang1 to subretinal tissues, b) subretinal COMP-Ang1 holds promise as a prospective treatment for neovascular AMD, and c) although VEGF suppression in the RPE/choroid may be one mechanism by which AAV2.COMP-Ang1 reduces CNV, this therapeutic effect may be hypoxia-independent. Taken together, these findings suggest that AAV2.COMP-Ang1 has potential to serve as an alternative or complementary option to anti-VEGF agents for the long-term amelioration of neovascular AMD.


Asunto(s)
Proteína de la Matriz Oligomérica del Cartílago/uso terapéutico , Neovascularización Coroidal/terapia , Terapia Genética/métodos , Degeneración Macular/terapia , Factor A de Crecimiento Endotelial Vascular/metabolismo , Angiopoyetina 1/metabolismo , Animales , Western Blotting , Proteína de la Matriz Oligomérica del Cartílago/metabolismo , Neovascularización Coroidal/metabolismo , Dependovirus/genética , Modelos Animales de Enfermedad , Vectores Genéticos/administración & dosificación , Factor 1 Inducible por Hipoxia/metabolismo , Degeneración Macular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Epitelio Pigmentado de la Retina/metabolismo
10.
Mol Ther ; 23(2): 226-34, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25306972

RESUMEN

Long-term inhibition of extracellular vascular endothelial growth factor (VEGF) in the treatment of age-related macular degeneration (AMD) may induce retinal neuronal toxicity and risk other side effects. We developed a novel strategy which inhibits retinal pigment epithelium (RPE)-derived VEGF, sparing other highly sensitive retinal tissues. Flt23k, an intraceptor inhibitor of VEGF, was able to inhibit VEGF in vitro. Adeno-associated virus type 2 (AAV2)-mediated expression of Flt23k was maintained for up to 6 months postsubretinal injection in mice. Flt23k was able to effectively inhibit laser-induced murine choroidal neovascularization (CNV). VEGF levels in the RPE/choroid complex decreased significantly in AAV2.Flt23k treated eyes. Neither retinal structure detected by Heidelberg Spectralis nor function measured by electroretinography (ERG) was adversely affected by treatment with AAV2.Flt23k. Hence AAV2.Flt23k can effectively maintain long-term expression and inhibit laser-induced CNV in mice through downregulation of VEGF while maintaining a sound retinal safety profile. These findings suggest a promising novel approach for the treatment of CNV.


Asunto(s)
Neovascularización Coroidal/genética , Dependovirus/genética , Vectores Genéticos/genética , Dominios y Motivos de Interacción de Proteínas/genética , Proteínas Recombinantes de Fusión , Transducción Genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Animales , Apoptosis , Coroides/metabolismo , Neovascularización Coroidal/patología , Neovascularización Coroidal/terapia , Modelos Animales de Enfermedad , Expresión Génica , Genes Reporteros , Terapia Genética , Vectores Genéticos/administración & dosificación , Humanos , Ratones , Retina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA