Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Proc Natl Acad Sci U S A ; 113(26): E3764-72, 2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27298364

RESUMEN

Retinal degenerative diseases are major causes of untreatable blindness, and novel approaches to treatment are being sought actively. Here we explored the activation of a unique protein, sigma 1 receptor (Sig1R), in the treatment of PRC loss because of its multifaceted role in cellular survival. We used Pde6ß(rd10) (rd10) mice, which harbor a mutation in the rod-specific phosphodiesterase gene Pde6ß and lose rod and cone photoreceptor cells (PRC) within the first 6 wk of life, as a model for severe retinal degeneration. Systemic administration of the high-affinity Sig1R ligand (+)-pentazocine [(+)-PTZ] to rd10 mice over several weeks led to the rescue of cone function as indicated by electroretinographic recordings using natural noise stimuli and preservation of cone cells upon spectral domain optical coherence tomography and retinal histological examination. The protective effect appears to result from the activation of Sig1R, because rd10/Sig1R(-/-) mice administered (+)-PTZ exhibited no cone preservation. (+)-PTZ treatment was associated with several beneficial cellular phenomena including attenuated reactive gliosis, reduced microglial activation, and decreased oxidative stress in mutant retinas. To our knowledge, this is the first report that activation of Sig1R attenuates inherited PRC loss. The findings may have far-reaching therapeutic implications for retinal neurodegenerative diseases.


Asunto(s)
Receptores sigma/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Degeneración Retiniana/metabolismo , Animales , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Receptores sigma/genética , Degeneración Retiniana/congénito , Degeneración Retiniana/tratamiento farmacológico , Degeneración Retiniana/genética , Receptor Sigma-1
2.
Adv Exp Med Biol ; 964: 267-284, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28315277

RESUMEN

This review article focuses on studies of Sigma 1 Receptor (Sigma1R) and retina . It provides a brief overview of the earliest pharmacological studies performed in the late 1990s that provided evidence of the presence of Sigma1R in various ocular tissues. It then describes work from a number of labs concerning the location of Sigma1R in several retinal cell types including ganglion, Müller glia , and photoreceptors . The role of Sigma1R ligands in retinal neuroprotection is emphasized. Early studies performed in vitro clearly showed that targeting Sigma1R could attenuate stress-induced retinal cell loss. These studies were followed by in vivo experiments. Data about the usefulness of targeting Sigma1R to prevent ganglion cell loss associated with diabetic retinopathy are reviewed. Mechanisms of Sigma1R-mediated retinal neuroprotection involving Müller cells , especially in modulating oxidative stress are described along with information about the retinal phenotype of mice lacking Sigma1R (Sigma1R -/- mice). The retina develops normally in Sigma1R -/- mice, but after many months there is evidence of apoptosis in the optic nerve head, decreased ganglion cell function and eventual loss of these cells. Additional studies using the Sigma1R -/- mice provide strong evidence that in the retina, Sigma1R plays a key role in modulating cellular stress. Recent work has shown that targeting Sigma1R may extend beyond protection of ganglion cells to include photoreceptor cell degeneration as well.


Asunto(s)
Receptores sigma/metabolismo , Retina/metabolismo , Animales , Células Ependimogliales/metabolismo , Humanos , Estrés Oxidativo/fisiología , Células Fotorreceptoras/metabolismo , Degeneración Retiniana/metabolismo , Células Ganglionares de la Retina/metabolismo , Receptor Sigma-1
3.
Dev Biol ; 319(2): 359-69, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18514182

RESUMEN

The innermost layer of the Drosophila eggshell, the vitelline membrane, provides structural support and positional information to the embryo. It is assembled in an incompletely understood manner from four major proteins to form a homogeneous, transparent extracellular matrix. Here we show that RNAi knockdown or genetic deletion of a minor constituent of this matrix, Palisade, results in structural disruptions during the initial synthesis of the vitelline membrane by somatic follicle cells surrounding the oocyte, including wide size variation among the precursor vitelline bodies and disorganization of follicle cell microvilli. Loss of Palisade or the microvillar protein Cad99C results in abnormal uptake into the oocyte of sV17, a major vitelline membrane protein, and defects in non-disulfide cross-linking of sV17 and sV23, while loss of Palisade has additional effects on processing and disulfide cross-linking of these proteins. Embryos surrounded by the abnormal vitelline membranes synthesized when Palisade is reduced are fertilized but undergo developmental arrest, usually during the first 13 nuclear divisions, with a nuclear phenotype of chromatin margination similar to that described for wild-type embryos subjected to anoxia. Our results demonstrate that Palisade is involved in coordinating assembly of the vitelline membrane and is required for functional properties of the eggshell.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila/embriología , Ovario/fisiología , Membrana Vitelina/fisiología , Animales , Animales Modificados Genéticamente , Anticuerpos , Western Blotting , Proteínas de Drosophila/análisis , Proteínas de Drosophila/química , Femenino , Interferencia de ARN , Proteínas Recombinantes de Fusión/análisis , Proteínas Recombinantes de Fusión/química , Transfección
4.
Mol Vis ; 15: 362-72, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19223991

RESUMEN

PURPOSE: GPR109A has been identified as a G-protein-coupled receptor for niacin. beta-hydroxybutyrate (beta-HB) is a physiologic ligand for the receptor. beta-HB, the predominate ketone body in circulation, is an important energy source for neurons, including retinal neurons, under various physiologic and pathologic conditions. The identification of GPR109A as the receptor for beta-HB suggests additional, hitherto unknown, functions for this metabolite. The circulating levels of beta-HB increase in diabetes. Since retinopathy is a serious complication associated with diabetes, we investigated GPR109A expression in retina and in different retinal cell types to determine if the receptor may have a role in the pathophysiology of diabetic retinopathy. METHODS: RT-PCR, fluorescent in situ hybridization, and immunofluorescent techniques were used to analyze GPR109A expression in mouse retina and in three transformed retinal cell lines: ARPE-19 (RPE), RGC-5 (ganglion), and rMC-1 (Müller). Activation of GPR109A by niacin and beta-HB was demonstrated in ARPE-19 cells by cAMP assay. RESULTS: Studies conducted using mouse retinal tissues demonstrated that GPR109A is expressed in retina with its expression restricted to RPE, where it differentially polarizes to the basolateral membrane. These results were confirmed with cell lines, which demonstrated GPR109A expression in ARPE-19, but not in rMC-1 and RGC-5 cells. Primary cultures of mouse RPE also showed robust expression of GPR109A. cAMP assay demonstrated that GPR109A expressed in RPE is functional. CONCLUSIONS: These data represent the first report on GPR109A expression in retina. The exclusive expression of GPR109A in RPE basolateral membrane, which has access to beta-HB in blood, may have biologic importance in diabetic retinopathy.


Asunto(s)
Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Animales , Línea Celular Transformada , AMP Cíclico/metabolismo , Técnica del Anticuerpo Fluorescente , Expresión Génica/fisiología , Humanos , Hibridación Fluorescente in Situ , Ratones , Ratones Endogámicos BALB C , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Retina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Biochem J ; 411(1): 79-88, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18042040

RESUMEN

Hepcidin is a hormone central to the regulation of iron homeostasis in the body. It is believed to be produced exclusively by the liver. Ferroportin, an iron exporter, is the receptor for hepcidin. This transporter/receptor is expressed in Müller cells, photoreceptor cells and the RPE (retinal pigment epithelium) within the retina. Since the retina is protected by the retinal-blood barriers, we asked whether ferroportin in the retina is regulated by hepcidin in the circulation or whether the retina produces hepcidin for regulation of its own iron homeostasis. Here we show that hepcidin is expressed robustly in Müller cells, photoreceptor cells and RPE cells, closely resembling the expression pattern of ferroportin. We also show that bacterial LPS (lipopolysaccharide) is a regulator of hepcidin expression in Müller cells and the RPE, both in vitro and in vivo, and that the regulation occurs at the transcriptional level. The action of LPS on hepcidin expression is mediated by the TLR4 (Toll-like receptor-4). The upregulation of hepcidin by LPS occurs independent of Hfe (human leukocyte antigen-like protein involved in Fe homeostasis). The increase in hepcidin levels in retinal cells in response to LPS treatment is associated with a decrease in ferroportin levels. The LPS-induced upregulation of hepcidin and consequent down-regulation of ferroportin is associated with increased oxidative stress and apoptosis within the retina in vivo. We conclude that retinal iron homeostasis may be regulated in an autonomous manner by hepcidin generated within the retina and that chronic bacterial infection/inflammation of the retina may disrupt iron homeostasis and retinal function.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/genética , Regulación de la Expresión Génica/efectos de los fármacos , Antígenos de Histocompatibilidad Clase I , Lipopolisacáridos/farmacología , Proteínas de la Membrana , Retina/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos/inmunología , Apoptosis , Proteínas de Transporte de Catión/genética , Proteína de la Hemocromatosis , Hepcidinas , Homeostasis , Hierro/metabolismo , Ratones , Estrés Oxidativo
6.
J Neurochem ; 104(4): 970-8, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17976164

RESUMEN

D-Serine, the endogenous ligand for the glycine modulatory binding site of the NMDA receptor, and serine racemase, the enzyme that converts L-serine to D-serine, have been reported in vertebrate retina; initial reports suggested that localization was restricted to Müller glial cells. Recent reports, in which D-serine and serine racemase were detected in neurons of the brain, prompted the present investigation of neuronal expression of D-serine and serine racemase in retina and whether expression patterns were developmentally regulated. RT-PCR, in situ hybridization, western blotting, immunohistochemistry, and immunocytochemical methods were used to localize D-serine and serine racemase in intact retina obtained from 1 to 3 day, 3 week, and 18 week mouse retinas and in primary ganglion cells harvested by immunopanning from neonatal mouse retina. Results of these analyses revealed robust expression of D-serine and serine racemase in ganglion cells, both in intact retina and in cultured cells. The levels appear to be developmentally regulated with D-serine levels being quite high in ganglion cells of neonatal retinas and decreasing rapidly postnatally. Serine racemase levels are also developmentally regulated, with high levels detected during the early postnatal period, but diminishing considerably in the mature retina. This represents the first report of neuronal expression of D-serine and serine racemase in the vertebrate retina and suggests an important contribution of neuronal D-serine during retinal development.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/fisiología , Racemasas y Epimerasas/biosíntesis , Retina/crecimiento & desarrollo , Células Ganglionares de la Retina/fisiología , Serina/biosíntesis , Animales , Animales Recién Nacidos , Ratones , Ratones Endogámicos C57BL , Racemasas y Epimerasas/genética , Retina/metabolismo , Serina/química , Serina/genética
7.
Invest Ophthalmol Vis Sci ; 48(7): 3356-63, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17591909

RESUMEN

PURPOSE: Monocarboxylates are primary energy substrates in the retina. Recently, the authors identified two sodium-coupled monocarboxylate transporters (SMCTs), SMCT1 (a high-affinity transporter) and SMCT2 (a low-affinity transporter). Expression of SMCT1 and SMCT2 has been studied in several tissues; however, little is known about their expression in retina. In the present study, the authors asked whether SMCT1 and SMCT2 are also expressed in retina and, if so, in which particular retinal cell types. METHODS: SMCT1 and SMCT2 expression was analyzed in intact mouse retina and cultured retinal cells (ganglion, Müller, RPE) by RT-PCR, in situ hybridization, and immunofluorescence. Uptake assays were performed to demonstrate SMCT1 (RGC-5 and ARPE-19 cells) and SMCT2 (rMC-1 cells) expression at the functional level. RESULTS: SMCT1 mRNA and protein were detected in the ganglion cell layer, inner nuclear layer, inner/outer plexiform layers, photoreceptor inner segments, and RPE. In RPE, the expression of SMCT1 was restricted to the basolateral membrane. SMCT2 mRNA and protein were detected only in neural retina, with a pattern of protein localization consistent with labeling of Müller cells. In vitro studies confirmed the cell type-specific expression of SMCT1 and SMCT2. Uptake assays demonstrated Na(+)-coupled monocarboxylate transport in RGC-5, ARPE-19, and rMC-1 cells. CONCLUSIONS: These data provide the first evidence for the expression of SMCT1 and SMCT2 in the retina and for the cell-type specific distribution of these transporters within the retina. These studies suggest that SMCT1 and SMCT2 play a differential role in monocarboxylate transport in the retina in a cell type-specific manner.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Células del Tejido Conectivo/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Epitelio Pigmentado Ocular/metabolismo , Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Animales , Proteínas de Transporte de Catión/genética , Células Cultivadas , Técnica del Anticuerpo Fluorescente Indirecta , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Transportadores de Ácidos Monocarboxílicos/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Invest Ophthalmol Vis Sci ; 58(11): 4545-4558, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28877319

RESUMEN

Purpose: Sigma 1 Receptor (Sig1R) is a novel therapeutic target in neurodegenerative diseases, including retinal disease. Sig1R-/- mice have late-onset retinal degeneration with ganglion cell loss that worsens under stress. Whether Sig1R plays a role in maintaining other retinal neurons is unknown, but was investigated here using rd10 mice, a model of severe photoreceptor degeneration. Methods: Wild-type, rd10, and rd10/Sig1R-/- mice were subjected to ERG and spectral-domain optical coherence tomography (SD-OCT) to assess visual function/structure in situ. Retinas imaged microscopically were subjected to morphometric analysis, immunodetection of cones, and analysis of gliosis. Oxidative and endoplasmic reticulum (ER) stress was evaluated at mRNA/protein levels. Results: Photopic ERG responses were reduced significantly in rd10/Sig1R-/- versus rd10 mice at P28 (31 ± 6 vs. 56 ± 7 µV), indicating accelerated cone loss when Sig1R was absent. At P28, SD-OCT revealed reduced retinal thickness in rd10/Sig1R-/- mice (60% of WT) versus rd10 (80% of WT). Morphometric analysis disclosed profound photoreceptor nuclei loss in rd10/Sig1R-/- versus rd10 mice. rd10/Sig1R-/- mice had 35% and 60% fewer photoreceptors, respectively, at P28 and P35, than rd10. Peanut agglutinin cone labeling decreased significantly; gliosis increased significantly in rd10/Sig1R-/- versus rd10 mice. At P21, NRF2 levels increased in rd10/Sig1R-/- mice versus rd10 and downstream antioxidants increased indicating oxidative stress. At P28, ER stress genes/proteins, especially XBP1, a potent transcriptional activator of the unfolded protein response and CHOP, a proapoptotic transcription factor, increased significantly in rd10/Sig1R-/- mice versus rd10. Conclusions: Photoreceptor cell degeneration accelerates and cone function diminishes much earlier in rd10/Sig1R-/- than rd10 mice emphasizing the importance of Sig1R as a modulator of retinal cell survival.


Asunto(s)
Apoptosis , Modelos Animales de Enfermedad , Receptores sigma/fisiología , Células Fotorreceptoras Retinianas Conos/patología , Células Fotorreceptoras Retinianas Bastones/patología , Retinitis Pigmentosa/patología , Animales , Electrorretinografía , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo , Retina/fisiología , Tomografía de Coherencia Óptica , Receptor Sigma-1
9.
Invest Ophthalmol Vis Sci ; 58(4): 1954-1963, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28384716

RESUMEN

Purpose: Hyperhomocysteinemia (Hhcy) is implicated in certain retinal neurovascular diseases, although whether it is causative remains uncertain. In isolated ganglion cells (GCs), mild Hhcy induces profound death, whereas retinal phenotypes in Hhcy mice caused by mutations in remethylation (methylene tetrahydrofolatereductase [Mthfr+/-]) or transsulfuration pathways (cystathionine ß-synthase [Cbs+/-]) demonstrate mild GC loss and mild vasculopathy. The current work investigated compensation in vivo of one pathway for the other, and, because the transsulfuration pathway yields cysteine necessary for formation of glutathione (GSH), taurine, and hydrogen sulfide (H2S), they were analyzed also. Methods: Retinas isolated from wild-type (WT), Mthfr+/-, and Cbs+/- mice (12 and 22 weeks) were analyzed for methylene tetrahydrofolate reductase (MTHFR), cystathionine-ß-synthase (CBS), and cystathionase (CTH) RNA/protein levels. Retinas were evaluated for levels of reduced:oxidized GSH (GSH:GSSG), Slc7a11 (xCT), taurine, taurine transporter (TAUT), and H2S. Results: Aside from decreased CBS RNA/protein levels in Cbs+/- retinas, there were minimal alterations in remethylation/transsulfuration pathways in the two mutant mice strains. Glutathione and taurine levels in Mthfr+/- and Cbs+/- retinas were similar to WT, which may be due to robust levels of xCT and TAUT in mutant retinas. Interestingly, levels of H2S were markedly increased in retinas of Mthfr+/- and Cbs+/- mice compared with WT. Conclusions: Ganglion cell loss and vasculopathy observed in Mthfr+/- and Cbs+/- mouse retinas may be milder than expected, not because of compensatory increases of enzymes in remethylation/transsulfuration pathways, but because downstream transsulfuration pathway products GSH, taurine, and H2S are maintained at robust levels. Elevation of H2S is particularly intriguing owing to neuroprotective properties reported for this gasotransmitter.


Asunto(s)
Regulación de la Expresión Génica , Glutatión/metabolismo , Sulfuro de Hidrógeno/metabolismo , Hiperhomocisteinemia/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Células Ganglionares de la Retina/metabolismo , Taurina/metabolismo , Animales , Modelos Animales de Enfermedad , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/metabolismo , Metilenotetrahidrofolato Reductasa (NADPH2)/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Retina/metabolismo , Retina/patología , Enfermedades de la Retina/etiología , Enfermedades de la Retina/genética , Enfermedades de la Retina/metabolismo , Células Ganglionares de la Retina/patología
10.
Invest Ophthalmol Vis Sci ; 47(10): 4238-44, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17003411

RESUMEN

PURPOSE: Hereditary hemochromatosis is an autosomal recessive disorder of iron overload leading to oxidative stress. Mutations in HFE are responsible for approximately 90% of cases of this disease. HFE is the principal regulator of iron homeostasis, and the process involves interaction with transferrin receptor (TfR)-1, transferrin receptor (TfR)-2, and beta2-microglobulin (beta2M). Expression of HFE has not been investigated in the retina. In the present study, the expression of HFE and the HFE-interacting proteins TfR1, TfR2, and beta2M were analyzed in mouse retina. METHODS: RT-PCR was used to detect the expression of HFE mRNA in neural retina and the RPE-eyecup. Expression of HFE in intact retina was investigated by in situ hybridization, immunofluorescence, and immunogold electron microscopy. Expression of HFE-interacting proteins was also analyzed using similar techniques. RESULTS: RT-PCR showed predominant expression of HFE mRNA in the RPE-eyecup. In situ hybridization in intact retina revealed that HFE mRNA is expressed almost exclusively in RPE Immunofluorescence and immunogold electron microscopy showed that HFE protein was specifically associated with the basolateral membrane of RPE. Expression of the HFE-interacting proteins TfR1, TfR2, and beta2M was also evident in the retina. CONCLUSIONS: This is the first report on the expression of HFE in the retina. The specific localization of HFE and its interacting proteins, TfR1 and TfR2, at the basolateral membrane of RPE is relevant to the regulation of iron homeostasis in this cell. Patients with hemochromatosis may have impairment of iron homeostasis in RPE, potentially contributing to age-related RPE dysfunction and retinal degeneration.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Antígenos de Histocompatibilidad Clase I/genética , Proteínas de la Membrana/genética , Epitelio Pigmentado Ocular/metabolismo , Animales , Membrana Basal/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Proteína de la Hemocromatosis , Hibridación in Situ , Hierro/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microscopía Inmunoelectrónica , ARN Mensajero/metabolismo , Receptores de Transferrina/metabolismo , Retina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Microglobulina beta-2/metabolismo
11.
Invest Ophthalmol Vis Sci ; 57(6): 2770-81, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27206247

RESUMEN

PURPOSE: Sigma receptor 1 (Sigma1R), a nonopioid putative molecular chaperone, has neuroprotective properties in retina. This study sought to determine whether delaying administration of (+)-pentazocine, a high-affinity Sigma1R ligand after onset of diabetes in Ins2Akita/+ diabetic mice would afford retinal neuroprotection and to determine consequences on retinal phenotype in Ins2Akita/+ diabetic mice in the absence of Sigma1R. METHODS: Ins2Akita/+ diabetic and WT mice received intraperitoneal injections of (+)-pentazocine beginning 4 or 8 weeks after onset of diabetes; eyes were harvested at 25 weeks. Retinal histologic sections were analyzed to determine thicknesses of retinal layers, number of ganglion cells, and evidence of gliosis (increased glial fibrillary acidic protein levels). Ins2Akita/+/Sig1R-/-mice were generated and subjected to in vivo assessment of retinal architecture (optical coherence tomography [OCT]) and retinal vasculature using fluorescein angiography (FA) at 12 and 16 weeks compared with age-matched Ins2Akita/+ mice. Eyes were then harvested for retinal morphometric assessment and gliosis assessment. RESULTS: Wild-type mice had 13 ± 0.06 cells/100 µm retinal length; cell bodies in Ins2Akita/+ mice injected 4 and 8 weeks after onset of diabetes with (+)-pentazocine retained significantly more ganglion cells compared with Ins2Akita/+ mice (9 ± 0.04) and demonstrated significant attenuation of gliosis. Ins2Akita/+/Sig1R-/-mouse retinas, analyzed to determine whether the Ins2Akita/+ phenotype was accelerated when lacking Sigma1R, revealed increased nerve fiber layer thickness (OCT), evidence of vitreal opacities, and vessel beading (FA) compared with Ins2Akita/+ mice. Morphometric analysis revealed significantly fewer ganglion cells in Ins2Akita/+/Sig1R-/-mice compared with Ins2Akita/+ mice. CONCLUSIONS: Sigma1R may be a novel retinal stress modulator, and targeting it even after disease onset may afford retinal neuroprotection.


Asunto(s)
Diabetes Mellitus Experimental , Retinopatía Diabética/complicaciones , Regulación de la Expresión Génica , ARN/genética , Receptores sigma/genética , Degeneración Retiniana/etiología , Células Ganglionares de la Retina/metabolismo , Animales , Muerte Celular , Retinopatía Diabética/metabolismo , Femenino , Angiografía con Fluoresceína , Fondo de Ojo , Insulina/genética , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Receptores sigma/biosíntesis , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Células Ganglionares de la Retina/patología , Tomografía de Coherencia Óptica , Receptor Sigma-1
12.
Invest Ophthalmol Vis Sci ; 46(6): 2047-55, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15914622

RESUMEN

PURPOSE: Previous results suggest that retinal neurons are infected early during murine cytomegalovirus (MCMV) infection of the inner retina. The purposes of this study were to identify which retinal neurons are infected and to determine the routes by which MCMV spreads in the retina. METHODS: Immunosuppressed (IS) BALB/c mice were inoculated with 5 x 10(3) PFU of MCMV (k181) through the supraciliary route. Injected eyes were collected at several times after inoculation, sectioned, and examined by electron microscopy and by staining for retinal cell antigens and for MCMV early (EA) or late (LA) antigen. RESULTS: MCMV-infected cells were observed in the choroid and RPE by day 3 after infection (PI) and in the inner retina beginning at day 5 PI. At this time, many horizontal and bipolar cells were MCMV-antigen-positive but only rare MCMV-infected amacrine cells (glycine positive or gamma-aminobutyric acid [GABA] positive) or MCMV-infected ganglion cells (NF positive) were observed in the inner retina. At day 10 PI, most virus-infected cells were glial fibrillary acidic protein (GFAP)- and GABA-positive glia. Virions were observed by electron microscopy in the choroid, RPE, and inner nuclear layer of the retina. Although virions were observed in the endothelium of the retinal vessels and the nearby retinal cells, the endothelial cell lining of the retinal vessels remained intact. Both apoptotic cells and necrotic cells were seen in the inner retina. CONCLUSIONS: In the inner retina, horizontal and bipolar cells were the early (< or = day 7 PI) targets of MCMV infection. Virus spread from the RPE and the photoreceptor layer to the inner retina through infected Muller cells and within the inner retina horizontally through infected horizontal cells.


Asunto(s)
Infecciones Virales del Ojo/virología , Infecciones por Herpesviridae/virología , Muromegalovirus/patogenicidad , Neuronas/virología , Retina/virología , Retinitis/virología , Animales , Antígenos Virales/análisis , Calbindinas , Coroides/microbiología , Coroides/ultraestructura , Infecciones Virales del Ojo/patología , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Proteína Ácida Fibrilar de la Glía/metabolismo , Infecciones por Herpesviridae/patología , Ratones , Ratones Endogámicos BALB C , Epitelio Pigmentado Ocular/ultraestructura , Epitelio Pigmentado Ocular/virología , Retinitis/patología , Proteína G de Unión al Calcio S100/metabolismo , Virión/ultraestructura , Ácido gamma-Aminobutírico/metabolismo
13.
Genetics ; 201(2): 631-49, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26265702

RESUMEN

Dynactin is a multi-subunit complex that functions as a regulator of the Dynein motor. A central component of this complex is Dynamitin/p50 (Dmn). Dmn is required for endosome motility in mammalian cell lines. However, the extent to which Dmn participates in the sorting of cargo via the endosomal system is unknown. In this study, we examined the endocytic role of Dmn using the Drosophila melanogaster oocyte as a model. Yolk proteins are internalized into the oocyte via clathrin-mediated endocytosis, trafficked through the endocytic pathway, and stored in condensed yolk granules. Oocytes that were depleted of Dmn contained fewer yolk granules than controls. In addition, these oocytes accumulated numerous endocytic intermediate structures. Particularly prominent were enlarged endosomes that were relatively devoid of Yolk proteins. Ultrastructural and genetic analyses indicate that the endocytic intermediates are produced downstream of Rab5. Similar phenotypes were observed upon depleting Dynein heavy chain (Dhc) or Lis1. Dhc is the motor subunit of the Dynein complex and Lis1 is a regulator of Dynein activity. We therefore propose that Dmn performs its function in endocytosis via the Dynein motor. Consistent with a role for Dynein in endocytosis, the motor colocalized with the endocytic machinery at the oocyte cortex in an endocytosis-dependent manner. Our results suggest a model whereby endocytic activity recruits Dynein to the oocyte cortex. The motor along with its regulators, Dynactin and Lis1, functions to ensure efficient endocytic uptake and maturation.


Asunto(s)
Endocitosis/genética , Endosomas/genética , Proteínas Asociadas a Microtúbulos/genética , Oocitos/metabolismo , Animales , Citoesqueleto/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Complejo Dinactina , Dineínas/biosíntesis , Dineínas/genética , Endosomas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Oocitos/crecimiento & desarrollo , Transporte de Proteínas/genética
14.
Invest Ophthalmol Vis Sci ; 56(4): 2684-95, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25766590

RESUMEN

PURPOSE: Methylenetetrahydrofolate reductase (Mthfr) is a key enzyme in homocysteine-methionine metabolism. We investigated Mthfr expression in retina and asked whether mild hyperhomocysteinemia, due to Mthfr deficiency, alters retinal neurovascular structure and function. METHODS: Expression of Mthfr was investigated at the gene and protein level using quantitative (q) RT-PCR, in situ hybridization, immunoblotting, and immunohistochemistry (IHC). The Mthfr+/+ and Mthfr+/- mice were subjected to comprehensive evaluation using ERG, funduscopy, fluorescein angiography (FA), spectral-domain optical coherence tomography (SD-OCT), HPLC, and morphometric and IHC analysis of glial fibrillary acidic protein (GFAP) at 8 to 24 weeks. RESULTS: Gene and protein analyses disclosed widespread retinal expression of Mthfr. Electroretinography (ERG) revealed a significant decrease in positive scotopic threshold response in retinas of Mthfr+/- mice at 24 weeks. Fundus examination in mice from both groups was normal; FA revealed areas of focal vascular leakage in 20% of Mthfr+/- mice at 12 to 16 weeks and 60% by 24 weeks. The SD-OCT revealed a significant decrease in nerve fiber layer (NFL) thickness at 24 weeks in Mthfr+/- compared to Mthfr+/+ mice. There was a 2-fold elevation in retinal hcy at 24 weeks in Mthfr+/- mice by HPLC and IHC. Morphometric analysis revealed an approximately 20% reduction in cells in the ganglion cell layer of Mthfr+/- mice at 24 weeks. The IHC indicated significantly increased GFAP labeling suggestive of Müller cell activation. CONCLUSIONS: Mildly hyperhomocysteinemic Mthfr+/- mice demonstrate reduced ganglion cell function, thinner NFL, and mild vasculopathy by 24 weeks. The retinal phenotype is similar to that of hyperhomocysteinemic mice with deficiency of cystathionine-ß-synthase (Cbs) reported earlier. The data support the hypothesis that hyperhomocysteinemia may be causative in certain retinal neurovasculopathies.


Asunto(s)
ADN/genética , Regulación de la Expresión Génica , Homocistinuria/patología , Hiperhomocisteinemia/patología , Metilenotetrahidrofolato Reductasa (NADPH2)/deficiencia , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Espasticidad Muscular/patología , Células Ganglionares de la Retina/patología , Animales , Modelos Animales de Enfermedad , Electrorretinografía , Angiografía con Fluoresceína , Fondo de Ojo , Homocistinuria/genética , Homocistinuria/metabolismo , Hiperhomocisteinemia/genética , Hiperhomocisteinemia/metabolismo , Immunoblotting , Inmunohistoquímica , Metilenotetrahidrofolato Reductasa (NADPH2)/biosíntesis , Metilenotetrahidrofolato Reductasa (NADPH2)/metabolismo , Ratones , Ratones Transgénicos , Espasticidad Muscular/genética , Espasticidad Muscular/metabolismo , Trastornos Psicóticos/genética , Trastornos Psicóticos/metabolismo , Trastornos Psicóticos/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Tomografía de Coherencia Óptica
15.
BMC Dev Biol ; 3: 6, 2003 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-12887734

RESUMEN

BACKGROUND: Folate is essential for cellular proliferation and tissue regeneration. As mammalian cells cannot synthesize folates de novo, tightly regulated cellular uptake processes have evolved to sustain sufficient levels of intracellular tetrahydrofolate cofactors to support biosynthesis of purines, pyrimidines, and some amino acids (serine, methionine). Though reduced-folate carrier (RFC) is one of the major proteins mediating folate transport, knowledge of the developmental expression of RFC is lacking. We utilized in situ hybridization and immunolocalization to determine the developmental distribution of RFC message and protein, respectively. RESULTS: In the mouse, RFC transcripts and protein are expressed in the E10.0 placenta and yolk sac. In the E9.0 to E11.5 mouse embryo RFC is widely detectable, with intense signal localized to cell populations in the neural tube, craniofacial region, limb buds and heart. During early development, RFC is expressed throughout the eye, but by E12.5, RFC protein becomes localized to the retinal pigment epithelium (RPE). CONCLUSIONS: Clinical studies show a statistical decrease in the number of neural tube defects, craniofacial abnormalities, cardiovascular defects and limb abnormalities detected in offspring of female patients given supplementary folate during pregnancy. The mechanism, however, by which folate supplementation ameliorates the occurrence of developmental defects is unclear. The present work demonstrates that RFC is present in placenta and yolk sac and provides the first evidence that it is expressed in the neural tube, craniofacial region, limb buds and heart during organogenesis. These findings suggest that rapidly dividing cells in the developing neural tube, craniofacial region, limb buds and heart may be particularly susceptible to folate deficiency.


Asunto(s)
Proteínas Portadoras/biosíntesis , Embrión de Mamíferos/química , Proteínas de Transporte de Membrana , Animales , Proteínas Portadoras/química , Proteínas Portadoras/inmunología , Ojo/química , Ojo/embriología , Femenino , Corazón/embriología , Inmunohistoquímica , Esbozos de los Miembros/química , Esbozos de los Miembros/embriología , Masculino , Ratones , Ratones Endogámicos ICR , Datos de Secuencia Molecular , Cresta Neural/química , Cresta Neural/embriología , Péptidos/inmunología , Péptidos/metabolismo , Epitelio Pigmentado Ocular/química , Epitelio Pigmentado Ocular/embriología , Placenta/química , Placenta/embriología , Embarazo , Prosencéfalo/química , Prosencéfalo/embriología , ARN Mensajero/biosíntesis , Proteína Portadora de Folato Reducido , Rombencéfalo/química , Rombencéfalo/embriología , Útero/química , Saco Vitelino/química , Saco Vitelino/embriología
16.
Invest Ophthalmol Vis Sci ; 45(9): 3330-6, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15326158

RESUMEN

PURPOSE: Neuronal cell death has been reported in retinas of humans with diabetic retinopathy and in diabetic rat models. Little is known about neuronal cell death in mouse models of diabetic retinopathy. This study was designed to determine whether neurons are lost in diabetic mouse retinas and whether the loss involves an apoptotic process. METHODS: Three-week-old C57Bl/6 mice were made diabetic with streptozotocin. They were studied over the course of 14 weeks after onset of diabetes. Eyes were processed for morphometric analysis and detection of apoptotic cells by TUNEL analysis and activated caspase-3 and were subjected to electron microscopy. RESULTS: Morphometric analysis of retinal cross sections of mice that had been diabetic 14 weeks showed approximately 20% to 25% fewer cells in the ganglion cell layer compared with age-matched control mice. There was a modest, but significant, decrease in the thickness of the whole retina and the inner and outer nuclear layers in mice that had been diabetic for 10 weeks. TUNEL analysis and detection of active caspase-3 revealed that cells of the ganglion cell layer were dying by apoptosis. Electron microscopic analysis detected morphologic features characteristic of apoptosis, including margination of chromatin and crenated nuclei of cells in the ganglion cell layer. CONCLUSIONS: The data suggest that in diabetic mouse retinas, neurons in the ganglion cell layer die, and this death occurs through an apoptotic pathway. Diabetic mice may be appropriate and valuable models for studies of neuronal cell death in diabetes.


Asunto(s)
Apoptosis , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Retina/patología , Retina/fisiopatología , Células Ganglionares de la Retina , Animales , Caspasa 3 , Caspasas/metabolismo , Diabetes Mellitus Experimental/enzimología , Activación Enzimática , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica , Retina/enzimología , Células Ganglionares de la Retina/patología
17.
Mol Vis ; 8: 494-501, 2002 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-12500176

RESUMEN

PURPOSE: To understand molecular events that lead to retinal pigment epithelial (RPE) cell proliferation and migration during the early phases of proliferative vitreoretinopathy (PVR) in a rabbit model. METHODS: Retinal holes were created and interleukin-1beta(IL-1beta) was injected intravitreally. Eyes were examined by indirect ophthalmoscopy and eyecup pieces containing retinal holes were analyzed at different times after the surgery up to 4 weeks. RPE proliferation and migration were examined by immunohistochemistry. Tyrosine phosphorylation of extracellular signal regulated kinase (ERK) and hepatocyte growth factor receptor (HGFR or c-met) was determined by immunoprecipitation and western blot analysis. Tyrosine phosphorylation of c-met and morphological studies was performed on vitreous treated ARPE-19 cells. Expression of c-jun was determined by Northern blot analysis. Matrix metalloproteinase (MMP) content in vitreous was assessed by zymography. RESULTS: Indirect ophthalmoscopy identified formation of epiretinal membrane and immunohistochemistry identified proliferative and migratory RPE and other cells in the posterior segment containing retinal holes at 4 weeks post-surgery. Tyrosine phosphorylation of ERK and c-met occurred in this segment within 30 min of surgery. ARPE-19 cells treated with vitreous from the 24 h post-surgical eyes, but not with control vitreous or IL-1beta, showed morphological changes and tyrosine phosphorylation of c-met. Northern blot analysis in this segment identified upregulation of c-jun within 30 min of surgery and the expression peaked at 72 h. Zymographic analysis of vitreous identified MMP-9 in 12-72 h post-surgery. CONCLUSIONS: These data suggest that the presence of retinal holes and IL-1beta may lead to activation of HGF, mitogen activated protein kinases (MAPK), c-jun and extracellular matrix remodeling, resulting in proliferative and migratory cells in the wounded retina.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Interleucina-1/administración & dosificación , Epitelio Pigmentado Ocular/patología , Perforaciones de la Retina/patología , Vitreorretinopatía Proliferativa/patología , Animales , Northern Blotting , Western Blotting , División Celular , Movimiento Celular , Modelos Animales de Enfermedad , Queratinas/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Pruebas de Precipitina , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Conejos , Perforaciones de la Retina/complicaciones , Perforaciones de la Retina/metabolismo , Tirosina/metabolismo , Vitreorretinopatía Proliferativa/etiología , Vitreorretinopatía Proliferativa/metabolismo , Cuerpo Vítreo/efectos de los fármacos , Cuerpo Vítreo/enzimología
18.
Mol Vis ; 8: 483-93, 2002 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-12500177

RESUMEN

PURPOSE: Hepatocyte growth factor (HGF) has been implicated in retinal pigment epithelial (RPE) cell proliferation and migration that occurs in proliferative retinal diseases such as proliferative vitreoretinopathy (PVR). The aim of this study is to investigate HGF induced signaling pathways that lead to RPE cell migration. METHODS: Localization of beta-catenin was determined by immunofluorescence. HGF induced migration of ARPE-19 cells was studied using a quantitative migration assay after wounding in the presence of a DNA polymerase inhibitor, and in the presence or absence of a mitogen activated protein kinase (MAP kinase) kinase inhibitor. C-jun expression was determined by semi-quantitative RT-PCR and by Northern blot analysis. P42/p44 MAP kinase activity was determined by western blot and by an immunoprecipitation kinase assay. Tyrosine phosphorylation of the HGF receptor (HGFR or c-met) and beta-catenin was determined by immunoprecipitation and western blot analysis. Transactivation activity of beta-catenin was determined by luciferase reporter gene analysis. RESULTS: Beta-catenin and E-cadherin were co-localized on the basal surface of the RPE in vivo. Diffusion of the cell surface-localized beta-catenin occurs in migratory cells in vitro in the presence of HGF. HGF induced a MAP kinase dependent ARPE-19 cell migration, which is accompanied with a transient increase of c-jun expression and concomitant increases of MAP kinase activity, tyrosine phosphorylation of HGFR and beta-catenin, increased cytosolic levels of beta-catenin, and transactivation activity of beta-catenin. Tyrosine phosphorylation of HGFR and beta-catenin occurs in the primary or passaged RPE cultures or proliferative ARPE-19 cells, but not freshly isolated RPE or differentiated ARPE-19 cells. CONCLUSIONS: This study defines the signal transduction pathways activated by HGF in RPE cells, leading to an increase in the MAP kinase activity and free pool of beta-catenin, and changes in gene expression. These findings are consistent with the hypothesis that both beta-catenin and MAP kinases are components of the HGF induced RPE migration that occurs in proliferative retinal diseases.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proteínas del Citoesqueleto/metabolismo , Factor de Crecimiento de Hepatocito/farmacología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Epitelio Pigmentado Ocular/citología , Transducción de Señal/fisiología , Transactivadores/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Western Blotting , Cadherinas/metabolismo , Células Cultivadas , Humanos , Persona de Mediana Edad , Fosforilación , Epitelio Pigmentado Ocular/efectos de los fármacos , Epitelio Pigmentado Ocular/metabolismo , Pruebas de Precipitina , Proteínas Proto-Oncogénicas c-jun/biosíntesis , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , ARN Mensajero/biosíntesis , Conejos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tirosina/metabolismo , beta Catenina
19.
Brain Res Mol Brain Res ; 107(2): 97-107, 2002 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12425939

RESUMEN

The type 1 sigma receptor (sigmaR1) is a nonopiate and nonphencyclidine binding site that has numerous pharmacological and physiological functions. In some studies, agonists for sigmaR1 have been shown to afford neuroprotection against overstimulation of the NMDA receptor. sigmaR1 expression has been demonstrated recently in retinal ganglion cells (RGC). RGCs undergo apoptosis early in diabetic retinopathy via NMDA receptor overstimulation. In the present study we asked whether RGCs cultured under hyperglycemic conditions and RGCs of diabetic mice continue to express sigmaR1. RGCs were cultured 48 h in RPMI medium containing either 45 mM glucose or 11 mM glucose plus 34 mM mannitol (osmolar control). C57BL/6 mice were made diabetic using streptozotocin. The retina was dissected from normal and streptozotocin-induced diabetic mice 3, 6 and 12 weeks post-onset of diabetes. sigmaR1 was analyzed in cells using semiquantitative RT-PCR and in tissues by semiquantitative RT-PCR, in situ hybridization, Western blot analysis and immunolocalization. The RT-PCR analysis of cultured RGCs showed that sigmaR1 mRNA is expressed under hyperglycemic conditions at levels similar to control cells. Similarly, analysis of retinas of diabetic mice showed no difference in levels of mRNA encoding sigmaR1 compared to retinas of control mice. In situ hybridization analysis showed that expression patterns of sigmaR1 mRNA in the ganglion cell layer were similar between diabetic and control mice. Western blot analysis suggested that levels of sigmaR1 in retina were similar between diabetic and control retinas. Immunohistochemical analysis of sigmaR1 showed a similar pattern of sigmaR1 protein expression between control and diabetic retina. These studies demonstrate that sigmaR1 is expressed under hyperglycemic conditions in vitro and in vivo.


Asunto(s)
Diabetes Mellitus Experimental/genética , Retinopatía Diabética/genética , Receptores sigma/genética , Células Ganglionares de la Retina/metabolismo , Animales , Glucemia/genética , Western Blotting , Células Cultivadas , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Retinopatía Diabética/tratamiento farmacológico , Retinopatía Diabética/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/fisiología , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/fisiopatología , Degeneración Nerviosa/prevención & control , Fármacos Neuroprotectores/farmacología , ARN Mensajero/metabolismo , Ratas , Receptores sigma/metabolismo , Células Ganglionares de la Retina/citología
20.
Invest Ophthalmol Vis Sci ; 54(2): 939-49, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23307965

RESUMEN

PURPOSE: Mice with moderate/severe hyperhomocysteinemia due to deficiency or absence of the cbs gene encoding cystathionine-beta-synthase (CBS) have marked retinal disruption, ganglion cell loss, optic nerve mitochondrial dysfunction, and ERG defects; those with mild hyperhomocysteinemia have delayed retinal morphological/functional phenotype. Excess homocysteine is a risk factor for cardiovascular diseases; however, it is not known whether excess homocysteine alters retinal vasculature. METHODS: Cbs(+/+), cbs(+/-), and cbs(-/-) mice (age ∼3 weeks) were subjected to angiography; retinas were harvested for cryosections, flat-mount preparations, or trypsin digestion and subjected to immunofluorescence microscopy to visualize vessels using isolectin-B4, to detect angiogenesis using anti-VEGF and anti-endoglin (anti-CD105) and activated glial cells (anti-glial fibrillary acidic protein [anti-GFAP]) and to investigate the blood-retinal barrier using the tight junction markers zonula occludens-1 (ZO-1) and occludin. Expression of vegf was determined by quantitative RT-PCR (qRT-PCR) and immunoblotting. Human retinal endothelial cells (HRECs) were treated with excess homocysteine to analyze permeability. RESULTS: Angiography revealed vascular leakage in cbs(-/-) mice; immunohistochemical analysis demonstrated vascular patterns consistent with ischemia; isolectin-B4 labeling revealed a capillary-free zone centrally and new vessels with capillary tufts midperipherally. This was associated with increased vegf mRNA and protein, CD105, and GFAP in cbs(-/-) retinas concomitant with a marked decrease in ZO-1 and occludin. Homocysteine-treated HRECs showed increased permeability. CONCLUSIONS: Severe elevation of homocysteine in cbs(-/-) mutant mice is accompanied by alterations in retinal vasculature (ischemia, neovascularization, and incompetent blood-retinal barrier). The marked disruption of retinal structure and decreased visual function reported in cbs(-/-) mice may reflect vasculopathy as well as neuropathy.


Asunto(s)
Regulación de la Expresión Génica , Homocisteína/metabolismo , Hiperhomocisteinemia/genética , ARN Mensajero/genética , Retina/patología , Enfermedades de la Retina/genética , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Barrera Hematorretinal/metabolismo , Barrera Hematorretinal/fisiopatología , Permeabilidad Capilar , Cistationina betasintasa/deficiencia , Cistationina betasintasa/genética , Modelos Animales de Enfermedad , Angiografía con Fluoresceína , Fondo de Ojo , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/metabolismo , Inmunohistoquímica , Ratones , Ratones Mutantes , Microscopía Fluorescente , Retina/metabolismo , Retina/fisiopatología , Enfermedades de la Retina/etiología , Enfermedades de la Retina/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor A de Crecimiento Endotelial Vascular/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA