Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 13(1): 7037, 2022 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-36396639

RESUMEN

Ciliary neurotrophic factor (CNTF) acts as a potent neuroprotective cytokine in multiple models of retinal degeneration. To understand mechanisms underlying its broad neuroprotective effects, we have investigated the influence of CNTF on metabolism in a mouse model of photoreceptor degeneration. CNTF treatment improves the morphology of photoreceptor mitochondria, but also leads to reduced oxygen consumption and suppressed respiratory chain activities. Molecular analyses show elevated glycolytic pathway gene transcripts and active enzymes. Metabolomics analyses detect significantly higher levels of ATP and the energy currency phosphocreatine, elevated glycolytic pathway metabolites, increased TCA cycle metabolites, lipid biosynthetic pathway intermediates, nucleotides, and amino acids. Moreover, CNTF treatment restores the key antioxidant glutathione to the wild type level. Therefore, CNTF significantly impacts the metabolic status of degenerating retinas by promoting aerobic glycolysis and augmenting anabolic activities. These findings reveal cellular mechanisms underlying enhanced neuronal viability and suggest potential therapies for treating retinal degeneration.


Asunto(s)
Factor Neurotrófico Ciliar , Degeneración Retiniana , Ratones , Animales , Factor Neurotrófico Ciliar/genética , Factor Neurotrófico Ciliar/metabolismo , Degeneración Retiniana/terapia , Neuroprotección , Retina/metabolismo , Glucólisis
2.
Redox Biol ; 37: 101787, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33214125

RESUMEN

Stargardt macular degeneration (STGD) is a central blinding disease caused by loss of or dysfunctional ABCA4 transporter in both photoreceptors and retinal pigment epithelial (RPE) cells. Toxic bisretinoid-lipofuscin buildup in the RPE cells is a pathological hallmark of STGD patients and its mouse model, the Abca4-/-. These vitamin A-derived fluorophores have been shown to induce oxidative stress, stimulate complement activity, and cause chronic inflammation of the RPE. In vivo modulation of complement regulatory pathway in the STGD mouse model has partially rescued the STGD phenotype suggesting that complement attack on the RPE is an important etiologic factor in disease pathogenesis. While bisretinoid-dependent complement activation was further evidenced in cultured RPE cells, this pathway has never been investigated directly in the context of RPE from STGD donor eyes. In the current study, we evaluate the complement reactivity in postmortem donor eyes of clinically diagnosed STGD patients. All three STGD donor eyes RPE displayed strong immunoreactivity for an antibody specific to 4-Hydroxynonenal, a lipid peroxidation byproduct. Also, unlike the control eyes, all three STGD donor eyes showed significantly increased membrane attack complex deposition on the RPE cells. In STGD eyes, increased MAC accumulation was mirrored by elevated C3 fragments internalized by the RPE and inversely correlated with the levels of complement factor H, a major complement regulatory protein. Here, we report the first direct evidence of RPE complement dysregulation as a causative factor in developing Stargardt phenotype.


Asunto(s)
Degeneración Macular , Epitelio Pigmentado de la Retina , Transportadoras de Casetes de Unión a ATP , Animales , Humanos , Degeneración Macular/genética , Ratones , Retina , Enfermedad de Stargardt
3.
Proc Natl Acad Sci U S A ; 117(18): 9857-9864, 2020 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-32300017

RESUMEN

Vitamin A has diverse biological functions and is essential for human survival at every point from embryogenesis to adulthood. Vitamin A and its derivatives have been used to treat human diseases including vision diseases, skin diseases, and cancer. Both insufficient and excessive vitamin A uptake are detrimental, but how its transport is regulated is poorly understood. STRA6 is a multitransmembrane domain cell-surface receptor and mediates vitamin A uptake from plasma retinol binding protein (RBP). STRA6 can mediate both cellular vitamin A influx and efflux, but what regulates these opposing activities is unknown. To answer this question, we purified and identified STRA6-associated proteins in a native mammalian cell type that takes up vitamin A through STRA6 using mass spectrometry. We found that the major protein repeatedly identified as STRA6-associated protein is calmodulin, consistent with the cryogenic electron microscopy (cryo-EM) study of zebrafish STRA6 associated with calmodulin. Using radioactivity-based, high-performance liquid chromatography (HPLC)-based and real-time fluorescence techniques, we found that calmodulin profoundly affects STRA6's vitamin A transport activity. Increased calcium/calmodulin promotes cellular vitamin A efflux and suppresses vitamin A influx through STRA6. Further mechanistic studies revealed that calmodulin enhances the binding of apo-RBP to STRA6, and this enhancement is much more pronounced for apo-RBP than holo-RBP. This study revealed that calmodulin regulates STRA6's vitamin A influx or efflux activity by modulating its preferential interaction with apo-RBP or holo-RBP. This molecular mechanism of regulating vitamin A transport may point to new directions to treat human diseases associated with insufficient or excessive vitamin A uptake.


Asunto(s)
Transporte Biológico/genética , Calmodulina/genética , Proteínas de la Membrana/genética , Proteínas Plasmáticas de Unión al Retinol/genética , Vitamina A/metabolismo , Animales , Apoproteínas/genética , Apoproteínas/metabolismo , Calcio/metabolismo , Bovinos , Línea Celular , Cromatografía Líquida de Alta Presión , Microscopía por Crioelectrón , Humanos , Proteínas de la Membrana/metabolismo , Unión Proteica/genética , Receptores de Superficie Celular/genética , Proteínas Plasmáticas de Unión al Retinol/metabolismo , Vitamina A/genética , Pez Cebra/genética
4.
Commun Biol ; 2: 186, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31123710

RESUMEN

The retinal pigment epithelium (RPE) serves vital roles in ocular development and retinal homeostasis but has limited representation in large-scale functional genomics datasets. Understanding how common human genetic variants affect RPE gene expression could elucidate the sources of phenotypic variability in selected monogenic ocular diseases and pinpoint causal genes at genome-wide association study (GWAS) loci. We interrogated the genetics of gene expression of cultured human fetal RPE (fRPE) cells under two metabolic conditions and discovered hundreds of shared or condition-specific expression or splice quantitative trait loci (e/sQTLs). Co-localizations of fRPE e/sQTLs with age-related macular degeneration (AMD) and myopia GWAS data suggest new candidate genes, and mechanisms by which a common RDH5 allele contributes to both increased AMD risk and decreased myopia risk. Our study highlights the unique transcriptomic characteristics of fRPE and provides a resource to connect e/sQTLs in a critical ocular cell type to monogenic and complex eye disorders.


Asunto(s)
Epitelio Pigmentado de la Retina/metabolismo , Oxidorreductasas de Alcohol/genética , Células Cultivadas , Mapeo Cromosómico , Metabolismo Energético , Feto/citología , Feto/metabolismo , Expresión Génica , Variación Genética , Estudio de Asociación del Genoma Completo , Humanos , Degeneración Macular/genética , Miopía/genética , Degradación de ARNm Mediada por Codón sin Sentido , Sitios de Carácter Cuantitativo , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/embriología , Factores de Riesgo , Transcriptoma
5.
Proc Natl Acad Sci U S A ; 115(47): E11120-E11127, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30397118

RESUMEN

Recessive Stargardt disease (STGD1) is an inherited blinding disorder caused by mutations in the Abca4 gene. ABCA4 is a flippase in photoreceptor outer segments (OS) that translocates retinaldehyde conjugated to phosphatidylethanolamine across OS disc membranes. Loss of ABCA4 in Abca4-/- mice and STGD1 patients causes buildup of lipofuscin in the retinal pigment epithelium (RPE) and degeneration of photoreceptors, leading to blindness. No effective treatment currently exists for STGD1. Here we show by several approaches that ABCA4 is additionally expressed in RPE cells. (i) By in situ hybridization analysis and by RNA-sequencing analysis, we show the Abca4 mRNA is expressed in human and mouse RPE cells. (ii) By quantitative immunoblotting, we show that the level of ABCA4 protein in homogenates of wild-type mouse RPE is about 1% of the level in neural retina homogenates. (iii) ABCA4 immunofluorescence is present in RPE cells of wild-type and Mertk-/- but not Abca4-/- mouse retina sections, where it colocalizes with endolysosomal proteins. To elucidate the role of ABCA4 in RPE cells, we generated a line of genetically modified mice that express ABCA4 in RPE cells but not in photoreceptors. Mice from this line on the Abca4-/- background showed partial rescue of photoreceptor degeneration and decreased lipofuscin accumulation compared with nontransgenic Abca4-/- mice. We propose that ABCA4 functions to recycle retinaldehyde released during proteolysis of rhodopsin in RPE endolysosomes following daily phagocytosis of distal photoreceptor OS. ABCA4 deficiency in the RPE may play a role in the pathogenesis of STGD1.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Degeneración Macular/congénito , Células Fotorreceptoras/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Retinaldehído/metabolismo , Transportadoras de Casetes de Unión a ATP/biosíntesis , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Lipofuscina/metabolismo , Lisosomas/metabolismo , Degeneración Macular/genética , Degeneración Macular/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Fagocitosis/inmunología , Retina/patología , Degeneración Retiniana/patología , Rodopsina/metabolismo , Enfermedad de Stargardt , Tirosina Quinasa c-Mer/genética
6.
PLoS One ; 13(7): e0200417, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30001398

RESUMEN

Rbfox1 is a splicing regulator that has been associated with various neurological conditions such as autism spectrum disorder, mental retardation, epilepsy, attention-deficit/hyperactivity disorder and schizophrenia. We show that in adult rodent retinas, Rbfox1 is expressed in all types of retinal ganglion cells (RGCs) and in certain subsets of amacrine cells (ACs), within the inner nuclear (INL) and ganglion cell (GCL) layers. In the INL, all Rbfox1-positive cells were colocalized with GABAergic ACs, however not all GABAergic ACs were immunostained for Rbfox1. In the GCL, a vast majority of GABAergic dACs were Rbfox1-immunopositive. Furthermore, all cholinergic starburst ACs (SACs) in the INL (type a) and in the GCL (type b) were Rbfox1 positive. The expression of Rbfox1 in the retina significantly overlapped with expression of Rbfox2, another member of Rbfox family of proteins. Rbfox2, in addition to RGCs and ACs, was also expressed in horizontal cells. In developing retinas at E12 and E15, Rbfox1 is localized to the cytoplasm of differentiating RGCs and ACs. Between P0 and P5, Rbfox1 subcellular localization switched from cytoplasmic to predominantly nuclear. Downregulation of Rbfox1 in adult Rbfox1loxP/loxP mice had no detectable effect on retinal gross morphology. However, the visual cliff test revealed marked abnormalities of depth perception of these animals. RNA sequencing of retinal transcriptomes of control and Rbfox1 knockout animals identified a number of Rbfox1-regulated genes that are involved in establishing neuronal circuits and synaptic transmission, including Vamp1, Vamp2, Snap25, Trak2, and Slc1A7, suggesting the role of Rbfox1 in facilitating synaptic communications between ACs and RGCs.


Asunto(s)
Percepción de Profundidad/fisiología , Factores de Empalme de ARN/metabolismo , Células Amacrinas/citología , Células Amacrinas/metabolismo , Animales , Regulación hacia Abajo , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones Endogámicos C57BL , Ratones Transgénicos , Factores de Empalme de ARN/genética , Retina/citología , Retina/crecimiento & desarrollo , Retina/metabolismo , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/metabolismo , Conducta Espacial/fisiología , Transcriptoma
7.
J Biol Chem ; 292(52): 21407-21416, 2017 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-29109151

RESUMEN

Peropsin is a non-visual opsin in both vertebrate and invertebrate species. In mammals, peropsin is present in the apical microvilli of retinal pigment epithelial (RPE) cells. These structures interdigitate with the outer segments of rod and cone photoreceptor cells. RPE cells play critical roles in the maintenance of photoreceptors, including the recycling of visual chromophore for the opsin visual pigments. Here, we sought to identify the function of peropsin in the mouse eye. To this end, we generated mice with a null mutation in the peropsin gene (Rrh). These mice exhibited normal retinal histology, normal morphology of outer segments and RPE cells, and no evidence of photoreceptor degeneration. Biochemically, Rrh-/- mice had ∼2-fold higher vitamin A (all-trans-retinol (all-trans-ROL)) in the neural retina following a photobleach and 5-fold lower retinyl esters in the RPE. This phenotype was similar to those reported in mice that lack interphotoreceptor retinoid-binding protein (IRBP) or cellular retinol-binding protein, suggesting that peropsin plays a role in the movement of all-trans-ROL from photoreceptors to the RPE. We compared the phenotypes in mice lacking both peropsin and IRBP with those of mice lacking peropsin or IRBP alone and found that the retinoid phenotype was similarly severe in each of these knock-out mice. We conclude that peropsin controls all-trans-ROL movement from the retina to the RPE or may regulate all-trans-ROL storage within the RPE. We propose that peropsin affects light-dependent regulation of all-trans-ROL uptake from photoreceptors into RPE cells through an as yet undefined mechanism.


Asunto(s)
Rodopsina/metabolismo , Vitamina A/fisiología , Animales , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Ratones , Ratones Noqueados , Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Pigmentos Retinianos/metabolismo , Retinaldehído/metabolismo , Retinoides/metabolismo , Proteínas de Unión al Retinol/genética , Proteínas de Unión al Retinol/metabolismo , Proteínas Celulares de Unión al Retinol/metabolismo , Rodopsina/genética , Rodopsina/fisiología , Opsinas de Bastones/metabolismo , Vitamina A/metabolismo
8.
Proc Natl Acad Sci U S A ; 114(15): 3987-3992, 2017 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-28348233

RESUMEN

Recessive Stargardt macular degeneration (STGD1) is caused by mutations in the gene for the ABCA4 transporter in photoreceptor outer segments. STGD1 patients and Abca4-/- (STGD1) mice exhibit buildup of bisretinoid-containing lipofuscin pigments in the retinal pigment epithelium (RPE), increased oxidative stress, augmented complement activation and slow degeneration of photoreceptors. A reduction in complement negative regulatory proteins (CRPs), possibly owing to bisretinoid accumulation, may be responsible for the increased complement activation seen on the RPE of STGD1 mice. CRPs prevent attack on host cells by the complement system, and complement receptor 1-like protein y (CRRY) is an important CRP in mice. Here we attempted to rescue the phenotype in STGD1 mice by increasing expression of CRRY in the RPE using a gene therapy approach. We injected recombinant adeno-associated virus containing the CRRY coding sequence (AAV-CRRY) into the subretinal space of 4-wk-old Abca4-/- mice. This resulted in sustained, several-fold increased expression of CRRY in the RPE, which significantly reduced the complement factors C3/C3b in the RPE. Unexpectedly, AAV-CRRY-treated STGD1 mice also showed reduced accumulation of bisretinoids compared with sham-injected STGD1 control mice. Furthermore, we observed slower photoreceptor degeneration and increased visual chromophore in 1-y-old AAV-CRRY-treated STGD1 mice. Rescue of the STGD1 phenotype by AAV-CRRY gene therapy suggests that complement attack on the RPE is an important etiologic factor in STGD1. Modulation of the complement system by locally increasing CRP expression using targeted gene therapy represents a potential treatment strategy for STGD1 and other retinopathies associated with complement dysregulation.


Asunto(s)
Complemento C3/metabolismo , Degeneración Macular/congénito , Células Fotorreceptoras de Vertebrados/patología , Receptores de Complemento/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Animales , Autofagia , Dependovirus/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Inyecciones Intraoculares , Lipofuscina/metabolismo , Degeneración Macular/metabolismo , Degeneración Macular/patología , Ratones Endogámicos BALB C , Ratones Mutantes , Estrés Oxidativo , Células Fotorreceptoras de Vertebrados/metabolismo , Receptores de Complemento/genética , Receptores de Complemento 3b , Epitelio Pigmentado de la Retina/patología , Retinoides/metabolismo , Enfermedad de Stargardt
9.
Adv Exp Med Biol ; 854: 525-32, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26427455

RESUMEN

More than 100 different mutations in the RPE65 gene are associated with inherited retinal degeneration. Although some missense mutations have been shown to abolish isomerase activity of RPE65, the molecular bases leading to loss of function and retinal degeneration remain incompletely understood. Here we show that several missense mutations resulted in significant decrease in expression level of RPE65 in the human retinal pigment epithelium cells. The 26S proteasome non-ATPase regulatory subunit 13, a newly identified negative regulator of RPE65, mediated degradation of mutant RPE65s, which were misfolded and formed aggregates in the cells. Many mutations, including L22P, T101I, and L408P, were mapped on nonactive sites of RPE65. Enzyme activities of these mutant RPE65s were significantly rescued at low temperature, whereas mutant RPE65s with a distinct active site mutation could not be rescued under the same conditions. 4-phenylbutyrate (PBA) displayed a significant synergistic effect on the low temperature-mediated rescue of the mutant RPE65s. Our results suggest that a low temperature eye mask and PBA, a FDA-approved oral medicine, may provide a promising "protein repair therapy" that can enhance the efficacy of gene therapy for delaying retinal degeneration caused by RPE65 mutations.


Asunto(s)
Proteínas Mutantes/genética , Mutación , Degeneración Retiniana/genética , cis-trans-Isomerasas/genética , Western Blotting , Dominio Catalítico/genética , Línea Celular , Células Cultivadas , Frío , Células HEK293 , Humanos , Microscopía Confocal , Proteínas Mutantes/metabolismo , Fenilbutiratos/farmacología , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Interferencia de ARN , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , cis-trans-Isomerasas/metabolismo
10.
Genome Med ; 7(1): 58, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26150894

RESUMEN

BACKGROUND: Age-related macular degeneration (AMD) is a leading cause of blindness. Most vision loss occurs following the transition from a disease of deposit formation and inflammation to a disease of neovascular fibrosis and/or cell death. Here, we investigate how repeated wound stimulus leads to seminal changes in gene expression and the onset of a perpetual state of stimulus-independent wound response in retinal pigmented epithelial (RPE) cells, a cell-type central to the etiology of AMD. METHODS: Transcriptome wide expression profiles of human fetal RPE cell cultures as a function of passage and time post-plating were determined using Agilent 44 K whole genome microarrays and RNA-Seq. Using a systems level analysis, differentially expressed genes and pathways of interest were identified and their role in the establishment of a persistent mesenchymal state was assessed using pharmacological-based experiments. RESULTS: Using a human fetal RPE cell culture model that considers monolayer disruption and subconfluent culture as a proxy for wound stimulus, we show that prolonged wound stimulus leads to terminal acquisition of a mesenchymal phenotype post-confluence and altered expression of more than 40 % of the transcriptome. In contrast, at subconfluence fewer than 5 % of expressed transcripts have two-fold or greater expression differences after repeated passage. Protein-protein and pathway interaction analysis of the genes with passage-dependent expression levels in subconfluent cultures reveals a 158-node interactome comprised of two interconnected modules with functions pertaining to wound response and cell division. Among the wound response genes are the TGFß pathway activators: TGFB1, TGFB2, INHBA, INHBB, GDF6, CTGF, and THBS1. Significantly, inhibition of TGFBR1/ACVR1B mediated signaling using receptor kinase inhibitors both forestalls and largely reverses the passage-dependent loss of epithelial potential; thus extending the effective lifespan by at least four passages. Moreover, a disproportionate number of RPE wound response genes have altered expression in neovascular and geographic AMD, including key members of the TGFß pathway. CONCLUSIONS: In RPE cells the switch to a persistent mesenchymal state following prolonged wound stimulus is driven by lasting activation of the TGFß pathway. Targeted inhibition of TGFß signaling may be an effective approach towards retarding AMD progression and producing RPE cells in quantity for research and cell-based therapies.

11.
PLoS One ; 10(5): e0125921, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25974161

RESUMEN

Retinyl esters represent an insoluble storage form of vitamin A and are substrates for the retinoid isomerase (Rpe65) in cells of the retinal pigment epithelium (RPE). The major retinyl-ester synthase in RPE cells is lecithin:retinol acyl-transferase (LRAT). A second palmitoyl coenzyme A-dependent retinyl-ester synthase activity has been observed in RPE homogenates but the protein responsible has not been identified. Here we show that diacylglycerol O-acyltransferase-1 (DGAT1) is expressed in multiple cells of the retina including RPE and Müller glial cells. DGAT1 catalyzes the synthesis of retinyl esters from multiple retinol isomers with similar catalytic efficiencies. Loss of DGAT1 in dgat1(-/-) mice has no effect on retinal anatomy or the ultrastructure of photoreceptor outer-segments (OS) and RPE cells. Levels of visual chromophore in dgat1(-/-) mice were also normal. However, the normal build-up of all-trans-retinyl esters (all-trans-RE's) in the RPE during the first hour after a deep photobleach of visual pigments in the retina was not seen in dgat1(-/-) mice. Further, total retinyl-ester synthase activity was reduced in both dgat1(-/-) retina and RPE.


Asunto(s)
Diacilglicerol O-Acetiltransferasa/metabolismo , Retina/citología , Retina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Aciltransferasas/metabolismo , Animales , Células Cultivadas , Diacilglicerol O-Acetiltransferasa/análisis , Diacilglicerol O-Acetiltransferasa/genética , Ésteres/metabolismo , Eliminación de Gen , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Palmitoil Coenzima A/metabolismo , Retina/ultraestructura , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/ultraestructura , Retinaldehído/metabolismo , Vitamina A/metabolismo , cis-trans-Isomerasas/metabolismo
12.
J Biochem ; 158(2): 115-25, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25752820

RESUMEN

RPE65 is a membrane-associated retinoid isomerase involved in the visual cycle responsible for sustaining vision. Many mutations in the human RPE65 gene are associated with distinct forms of retinal degenerative diseases. The pathogenic mechanisms for most of these mutations remain poorly understood. Here, we show that three Leber congenital amaurosis -associated RPE65 mutants (R91W, Y249C and R515W) undergo rapid proteasomal degradation mediated by the 26 S proteasome non-ATPase regulatory subunit 13 (PSMD13) in cultured human retinal pigment epithelium (RPE) cells. These mutant proteins formed cytosolic inclusion bodies or high molecular weight complexes via disulfide bonds. The mutations are mapped on non-active sites but severely reduced isomerase activity of RPE65. At 30°C, however, the enzymatic function and membrane-association of the mutant RPE65s are significantly rescued possibly due to proper folding. In addition, PSMD13 displayed a drastically decreased effect on degradation of the mutant proteins in the cells grown at 30°C. These results suggest that PSMD13 plays a critical role in regulating pathogenicity of the mutations and the molecular basis for the PSMD13-mediated rapid degradation and loss of function of the mutants is misfolding of RPE65.


Asunto(s)
Predisposición Genética a la Enfermedad , Amaurosis Congénita de Leber/enzimología , Amaurosis Congénita de Leber/genética , Mutación/genética , Temperatura , cis-trans-Isomerasas/genética , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Disulfuros/metabolismo , Humanos , Cuerpos de Inclusión/metabolismo , Modelos Moleculares , Peso Molecular , Proteínas Mutantes/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis
13.
J Biol Chem ; 289(27): 18943-56, 2014 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-24849605

RESUMEN

Over 70 different missense mutations, including a dominant mutation, in RPE65 retinoid isomerase are associated with distinct forms of retinal degeneration; however, the disease mechanisms for most of these mutations have not been studied. Although some mutations have been shown to abolish enzyme activity, the molecular mechanisms leading to the loss of enzymatic function and retinal degeneration remain poorly understood. Here we show that the 26 S proteasome non-ATPase regulatory subunit 13 (PSMD13), a newly identified negative regulator of RPE65, plays a critical role in regulating pathogenicity of three mutations (L22P, T101I, and L408P) by mediating rapid degradation of mutated RPE65s via a ubiquitination- and proteasome-dependent non-lysosomal pathway. These mutant RPE65s were misfolded and formed aggregates or high molecular complexes via disulfide bonds. Interaction of PSMD13 with mutant RPE65s promoted degradation of misfolded but not properly folded mutant RPE65s. Many mutations, including L22P, T101I, and L408P, were mapped on non-active sites. Although their activities were very low, these mutant RPE65s were catalytically active and could be significantly rescued at low temperature, whereas mutant RPE65s with a distinct active site mutation could not be rescued under the same conditions. Sodium 4-phenylbutyrate and glycerol displayed a significant synergistic effect on the low temperature rescue of the mutant RPE65s by promoting proper folding, reducing aggregation, and increasing membrane association. Our results suggest that a low temperature eye mask and sodium 4-phenylbutyrate, a United States Food and Drug Administration-approved oral medicine, may provide a promising "protein repair therapy" that can enhance the efficacy of gene therapy by reducing the cytotoxic effect of misfolded mutant RPE65s.


Asunto(s)
Dominio Catalítico , Enfermedad/genética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación Missense , cis-trans-Isomerasas/genética , cis-trans-Isomerasas/metabolismo , Animales , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glicerol/farmacología , Células HEK293 , Humanos , Ratones , Modelos Moleculares , Proteínas Mutantes/química , Fenilbutiratos/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Pliegue de Proteína/efectos de los fármacos , Proteolisis/efectos de los fármacos , Epitelio Pigmentado de la Retina/citología , Temperatura , Ubiquitinación/efectos de los fármacos , cis-trans-Isomerasas/química
14.
J Biol Chem ; 289(13): 9113-20, 2014 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-24550392

RESUMEN

Age-related macular degeneration (AMD) is a common central blinding disease of the elderly. Homozygosity for a sequence variant causing Y402H and I62V substitutions in the gene for complement factor H (CFH) is strongly associated with risk of AMD. CFH, secreted by many cell types, including those of the retinal pigment epithelium (RPE), is a regulatory protein that inhibits complement activation. Recessive Stargardt maculopathy is another central blinding disease caused by mutations in the gene for ABCA4, a transporter in photoreceptor outer segments (OS) that clears retinaldehyde and prevents formation of toxic bisretinoids. Photoreceptors daily shed their distal OS, which are phagocytosed by the RPE cells. Here, we investigated the relationship between the CFH haplotype of human RPE (hRPE) cells, exposure to OS containing bisretinoids, and complement activation. We show that hRPE cells of the AMD-predisposing CFH haplotype (HH402/VV62) are attacked by complement following exposure to bisretinoid-containing Abca4(-/-) OS. This activation was dependent on factor B, indicating involvement of the alternative pathway. In contrast, hRPE cells of the AMD-protective CFH haplotype (YY402/II62) showed no complement activation following exposure to either Abca4(-/-) or wild-type OS. The AMD-protective YY402/II62 hRPE cells were more resistant to the membrane attack complex, whereas HH402/VV62 hRPE cells showed significant membrane attack complex deposition following ingestion of Abca4(-/-) OS. These results suggest that bisretinoid accumulation in hRPE cells stimulates activation and dysregulation of complement. Cells with an intact complement negative regulatory system are protected from complement attack, whereas cells with reduced CFH synthesis because of the Y402H and I62V substitutions are vulnerable to disease.


Asunto(s)
Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Haplotipos , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/metabolismo , Retinoides/metabolismo , Transportadoras de Casetes de Unión a ATP/deficiencia , Animales , Membrana Celular/metabolismo , Complemento C3b/metabolismo , Factor H de Complemento/biosíntesis , Predisposición Genética a la Enfermedad/genética , Humanos , Degeneración Macular/genética , Degeneración Macular/metabolismo , Degeneración Macular/patología , Ratones , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Segmento Externo de las Células Fotorreceptoras Retinianas/patología , Epitelio Pigmentado de la Retina/patología
15.
Exp Eye Res ; 126: 46-50, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24060345

RESUMEN

Human fetal retinal pigment epithelium (hfRPE), when harvested by mechanical dissection and cultured initially under low calcium conditions, will proliferate and tolerate cryopreservation for future use. Cryopreserved cells can be subsequently thawed and cultured in standard calcium and in the presence of appropriate nutrients to a high state of differentiation, allowing recapitulation of multiple in vivo functions. In this review we briefly discuss some of our previous studies of the classical retinoid visual cycle and introduce current studies in our laboratory that involve two new areas of investigation; the dynamic response of the receptor for retinol binding protein, STRA6 to the addition of holo-retinol binding protein to the culture medium and the protective complement-based response of hfRPE to the ingestion of toxic byproducts of the visual cycle. This response is studied in the context of genotyped hfRPE expressing either predisposing or protective variants of complement factor H.


Asunto(s)
Enfermedades de la Retina/fisiopatología , Epitelio Pigmentado de la Retina/citología , Retinoides/metabolismo , Técnicas de Cultivo de Célula , Células Cultivadas , Activación de Complemento/fisiología , Feto/citología , Humanos , Modelos Biológicos , Epitelio Pigmentado de la Retina/fisiología , Proteínas de Unión al Retinol/fisiología
17.
Proc Natl Acad Sci U S A ; 110(47): E4520-9, 2013 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-24191003

RESUMEN

Ciliary neurotrophic factor (CNTF) acts as a potent neuroprotective agent in multiple retinal degeneration animal models. Recently, CNTF has been evaluated in clinical trials for the inherited degenerative disease retinitis pigmentosa (RP) and for dry age-related macular degeneration (AMD). Despite its potential as a broad-spectrum therapeutic treatment for blinding diseases, the target cells of exogenous CNTF and its mechanism of action remain poorly understood. We have shown previously that constitutive expression of CNTF prevents photoreceptor death but alters the retinal transcriptome and suppresses visual function. Here, we use a lentivirus to deliver the same secreted human CNTF used in clinical trials to a mouse model of RP. We found that low levels of CNTF halt photoreceptor death, improve photoreceptor morphology, and correct opsin mislocalization. However, we did not detect corresponding improvement of retinal function as measured by the electroretinogram. Disruption of the cytokine receptor gp130 gene in Müller glia reduces CNTF-dependent photoreceptor survival and prevents phosphorylation of STAT3 and ERK in Müller glia and the rest of the retina. Targeted deletion of gp130 in rods also demolishes neuroprotection by CNTF and prevents further activation of Müller glia. Moreover, CNTF elevates the expression of LIF and endothelin 2, thus positively promoting Müller and photoreceptor interactions. We propose that exogenous CNTF initially targets Müller glia, and subsequently induces cytokines acting through gp130 in photoreceptors to promote neuronal survival. These results elucidate a cellular mechanism for exogenous CNTF-triggered neuroprotection and provide insight into the complex cellular responses induced by CNTF in diseased retinas.


Asunto(s)
Factor Neurotrófico Ciliar/metabolismo , Receptor gp130 de Citocinas/metabolismo , Células Ependimogliales/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Degeneración Retiniana/tratamiento farmacológico , Transducción de Señal/fisiología , Análisis de Varianza , Animales , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Inmunohistoquímica , Lentivirus , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Reacción en Cadena en Tiempo Real de la Polimerasa , Degeneración Retiniana/genética
18.
J Biol Chem ; 288(16): 11395-406, 2013 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-23486466

RESUMEN

Interphotoreceptor retinoid-binding protein (IRBP) secreted by photoreceptors plays a pivotal role in photoreceptor survival and function. Recently, a D1080N mutation in IRBP was found in patients with retinitis pigmentosa, a frequent cause of retinal degeneration. The molecular and cellular bases for pathogenicity of the mutation are unknown. Here, we show that the mutation abolishes secretion of IRBP and results in formation of insoluble high molecular weight complexes via disulfide bonds. Co-expression of protein disulfide isomerase A2 that regulates disulfide bond formation or introduction of double Cys-to-Ala substitutions at positions 304 and 1175 in D1080N IRBP promoted secretion of the mutated IRBP. D1080N IRBP was not transported to the Golgi apparatus, but accumulated in the endoplasmic reticulum (ER), bound with the ER-resident chaperone proteins such as BiP, protein disulfide isomerase, and heat shock proteins. Splicing of X-box-binding protein-1 mRNA, expression of activating transcription factor 4 (ATF4), and cleavage of ATF6 were significantly increased in cells expressing D1080N IRBP. Moreover, D1080N IRBP induced up-regulation and nuclear translocation of the C/EBP homologous protein, a proapoptotic transcription factor associated with the unfolded protein response. These results indicate that loss of normal function (nonsecretion) and gain of cytotoxic function (ER stress) are involved in the disease mechanisms of D1080N IRBP. Chemical chaperones and low temperature, which help proper folding of many mutated proteins, significantly rescued secretion of D1080N IRBP, suggesting that misfolding is the molecular basis for pathogenicity of D1080N substitution and that chemical chaperones are therapeutic candidates for the mutation-caused blinding disease.


Asunto(s)
Proteínas del Ojo/metabolismo , Mutación Missense , Pliegue de Proteína , Retinitis Pigmentosa/metabolismo , Proteínas de Unión al Retinol/metabolismo , Respuesta de Proteína Desplegada , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Transporte Activo de Núcleo Celular/genética , Sustitución de Aminoácidos , Animales , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Chaperón BiP del Retículo Endoplásmico , Proteínas del Ojo/genética , Aparato de Golgi/genética , Aparato de Golgi/metabolismo , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Ratones , Proteína Disulfuro Isomerasas/biosíntesis , Proteína Disulfuro Isomerasas/genética , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología , Proteínas de Unión al Retinol/genética
19.
Invest Ophthalmol Vis Sci ; 53(6): 3027-39, 2012 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-22467576

RESUMEN

PURPOSE: We report generation of a mouse model in which the STRA6 gene has been disrupted functionally to facilitate the study of visual responses, changes in ocular morphology, and retinoid processing under STRA6 protein deficiency. METHODS: A null mouse line, stra6 -/-, was generated. Western Blot and immunocytochemistry were used to determine expression of STRA6 protein. Visual responses and morphological studies were performed on 6-week, 5-month and 10-month-old mice. The retinoid content of eye tissues was evaluated in dark-adapted mice by high performance liquid chromatography. RESULTS: STRA6 protein was not detectable in stra6 -/- null mice, which had a consistent reduction, but not total ablation of their visual responses. The mice also showed significant depletion of their retinoid content in retinal pigment epithelium (RPE) and neurosensory retina, including a 95% reduction in retinyl esters. At the morphological level, a reduction in thickness of the neurosensory retina due to shortening of the rod outer and inner segments was observed when compared to control litter mates with a commensurate reduction in rod a- and b-wave amplitudes. In addition, there was a reduction in cone photoreceptor cell number and cone b-wave amplitude. A typical hallmark in stra6 -/- null eyes was the presence of a persistent primary hypertrophic vitreous, an optically dense vascularized structure located in the vitreous humor between the posterior surface of the lens and neurosensory retina. CONCLUSIONS: Our studies of stra6 -/- null mice established the importance of the STRA6 protein for the uptake, intracellular transport, and processing of retinol by the RPE. In its absence, rod photoreceptor outer and inner segment length was reduced, and cone cell numbers were reduced, as were scotopic and photopic responses. STRA6 also was required for dissolution of the primary vitreous. However, it was clear from these studies that STRA6 is not the only pathway for retinol uptake by the RPE.


Asunto(s)
Proteínas de la Membrana/deficiencia , Retinoides/metabolismo , Aciltransferasas/metabolismo , Animales , Recuento de Células , Línea Celular , Cromatografía Líquida de Alta Presión , Adaptación a la Oscuridad , Electrorretinografía , Proteínas del Ojo/metabolismo , Hipertrofia , Ratones , Ratones Endogámicos C57BL , Receptores Acoplados a Proteínas G/metabolismo , Retina/metabolismo , Retina/patología , Células Fotorreceptoras Retinianas Conos/citología , Epitelio Pigmentado de la Retina/metabolismo , Tomografía de Coherencia Óptica , Cuerpo Vítreo/patología , cis-trans-Isomerasas/metabolismo
20.
Proc Natl Acad Sci U S A ; 108(45): 18277-82, 2011 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-21969589

RESUMEN

We introduce a human retinal pigmented epithelial (RPE) cell-culture model that mimics several key aspects of early stage age-related macular degeneration (AMD). These include accumulation of sub-RPE deposits that contain molecular constituents of human drusen, and activation of complement leading to formation of deposit-associated terminal complement complexes. Abundant sub-RPE deposits that are rich in apolipoprotein E (APOE), a prominent drusen constituent, are formed by RPE cells grown on porous supports. Exposure to human serum results in selective, deposit-associated accumulation of additional known drusen components, including vitronectin, clusterin, and serum amyloid P, thus suggesting that specific protein-protein interactions contribute to the accretion of plasma proteins during drusen formation. Serum exposure also leads to complement activation, as evidenced by the generation of C5b-9 immunoreactive terminal complement complexes in association with APOE-containing deposits. Ultrastructural analyses reveal two morphologically distinct forms of deposits: One consisting of membrane-bounded multivesicular material, and the other of nonmembrane-bounded particle conglomerates. Collectively, these results suggest that drusen formation involves the accumulation of sub-RPE material rich in APOE, a prominent biosynthetic product of the RPE, which interacts with a select group of drusen-associated plasma proteins. Activation of the complement cascade appears to be mediated via the classical pathway by the binding of C1q to ligands in APOE-rich deposits, triggering direct activation of complement by C1q, deposition of terminal complement complexes and inflammatory sequelae. This model system will facilitate the analysis of molecular and cellular aspects of AMD pathogenesis, and the testing of new therapeutic agents for its treatment.


Asunto(s)
Activación de Complemento , Degeneración Macular/patología , Modelos Biológicos , Drusas Retinianas/patología , Apolipoproteínas E/metabolismo , Técnicas de Cultivo de Célula , Humanos , Inmunohistoquímica , Degeneración Macular/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...