Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Ther Methods Clin Dev ; 18: 582-594, 2020 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-32775493

RESUMO

No treatment is available for nicotinamide mononucleotide adenylyltransferase 1 (NMNAT1)-associated retinal degeneration, an inherited disease that leads to severe vision loss early in life. Although the causative gene, NMNAT1, plays an essential role in nuclear nicotinamide adenine dinucleotide (NAD)+ metabolism in tissues throughout the body, NMNAT1-associated disease is isolated to the retina. Since this condition is recessive, supplementing the retina with a normal copy of NMNAT1 should protect vulnerable cells from disease progression. We tested this hypothesis in a mouse model that harbors the p.Val9Met mutation in Nmnat1 and consequently develops a retinal degenerative phenotype that recapitulates key features of the human disease. Gene augmentation therapy, delivered by subretinal injection of adeno-associated virus (AAV) carrying a normal human copy of NMNAT1, rescued retinal structure and function. Due to the early-onset profile of the phenotype, a rapidly activating self-complementary AAV was required to initiate transgene expression during the narrow therapeutic window. These data represent the first proof of concept for a therapy to treat patients with NMNAT1-associated disease.

2.
Adv Exp Med Biol ; 1185: 91-96, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31884594

RESUMO

Mutations in USH2A gene account for most cases of Usher syndrome type II (USH2), characterized by a combination of congenital hearing loss and progressive vision loss. In particular, approximately 30% of USH2A patients harbor a single base pair deletion, c.2299delG, in exon 13 that creates a frameshift and premature stop codon, leading to a nonfunctional USH2A protein. The USH2A protein, also known as usherin, is an extremely large transmembrane protein (5202 aa), which limits the use of conventional AAV-mediated gene therapy; thus development of alternative approaches is required for the treatment of USH2A patients. As usherin contains multiple repetitive domains, we hypothesize that removal of one or more of those domains encoded by mutant exon(s) in the USH2A gene may reconstitute the reading frame and restore the production of a shortened yet adequately functional protein. In this study, we deleted the exon 12 of mouse Ush2a gene (corresponding to exon 13 of human USH2A) using CRISPR/Cas9-based exon-skipping approach and revealed that a shortened form of Ush2a that lacks exon 12 (Ush2a-∆Ex12) is produced and localized correctly in the cochlea. When the Ush2a-∆Ex12 allele is expressed on an Ush2a null background, the Ush2a-∆Ex12 protein can successfully restore the impaired hair cell structure and the auditory function in the Ush2a-/- mice. These results demonstrate that CRISPR/Cas9-based exon-skipping strategy holds a great therapeutic potential for the treatment of USH2A patients.


Assuntos
Proteínas da Matriz Extracelular/genética , Síndromes de Usher/terapia , Animais , Sistemas CRISPR-Cas , Éxons , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Mutação , Síndromes de Usher/genética
3.
Curr Eye Res ; 41(11): 1498-1506, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27158920

RESUMO

PURPOSE: Anterior ischemic optic neuropathy (AION) is the most common cause of non-glaucomatous optic nerve head (ONH) injury among older adults. AION results from a sudden ischemic insult to the proximal portion of the optic nerve, typically leading to visual impairment. Here, we present an experimental model of photodynamically induced ONH injury that can be used to study neuroprotective modalities. METHODS: Intraperitoneal injection of mesoporphyrin IX was followed by photodynamic treatment of the ONH in one eye of Brown-Norway rats; the fellow eye received the reverse sequence as a sham control. Fluorescein angiography (FA), spectral domain optical coherence tomography (SD-OCT), and visual evoked potential (VEP) recordings were performed at different time points following laser treatment. Immunohistochemistry was used to monitor apoptotic cell death (TUNEL) and macrophage infiltration (CD68). Cytokine levels were evaluated using enzyme-linked immunosorbent assay (ELISA). RESULTS: FA showed early hyperfluorescence and late leakage of the ONH, while SD-OCT revealed optic nerve edema. No leakage or other abnormalities were detected in control eyes. VEPs were significantly reduced in amplitude and showed prolonged responses compared to sham eyes. The number of apoptotic retinal ganglion cells was elevated one day after laser treatment (13.77 ± 4.49, p < 0.01) and peaked on day 7 (57.22 ± 11.34, p < 0.01). ONH macrophage infiltration also peaked on day 7 (101.8 ± 9.8, p < 0.05). ELISAs performed showed upregulation of macrophage chemoattractant protein-1 and macrophage inflammatory protein-2 on days 3 and 1, respectively. CONCLUSIONS: Photodynamic treatment of the ONH after administration of mesoporphyrin IX leads to macroscopic, histologic, and physiologic evidence of ONH injury. Given the long half-life of mesoporphyrin IX and the ease of intraperitoneal injections, this new model of photodynamically induced ONH injury may be a useful tool for studying optic nerve injury and possible neuroprotective treatments.


Assuntos
Disco Óptico/patologia , Neuropatia Óptica Isquêmica/patologia , Fotoquimioterapia/efeitos adversos , Células Ganglionares da Retina/patologia , Animais , Apoptose , Modelos Animais de Doenças , Potenciais Evocados Visuais , Angiofluoresceinografia , Fundo de Olho , Masculino , Neuropatia Óptica Isquêmica/etiologia , Neuropatia Óptica Isquêmica/fisiopatologia , Ratos , Ratos Endogâmicos BN , Tomografia de Coerência Óptica/métodos
4.
Hum Gene Ther ; 21(8): 993-1004, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20384479

RESUMO

RPGR-interacting protein-1 (RPGRIP1) is localized in the photoreceptor-connecting cilium, where it anchors the RPGR (retinitis pigmentosa GTPase regulator) protein, and its function is essential for photoreceptor maintenance. Genetic defect in RPGRIP1 is a known cause of Leber congenital amaurosis (LCA), a severe, early-onset form of retinal degeneration. We evaluated the efficacy of replacement gene therapy in a murine model of LCA carrying a targeted disruption of RPGRIP1. The replacement construct, packaged in an adeno-associated virus serotype 8 (AAV8) vector, used a rhodopsin kinase gene promoter to drive RPGRIP1 expression. Both promoter and transgene were of human origin. After subretinal delivery of the replacement gene in the mutant mice, human RPGRIP1 was expressed specifically in photoreceptors, localized correctly in the connecting cilia, and restored the normal localization of RPGR. Electroretinogram and histological examinations showed better preservation of rod and cone photoreceptor function and improved photoreceptor survival in the treated eyes. This study demonstrates the efficacy of human gene replacement therapy and validates a gene therapy design for future clinical trials in patients afflicted with this condition. Our results also have therapeutic implications for other forms of retinal degenerations attributable to a ciliary defect.


Assuntos
Terapia Genética , Amaurose Congênita de Leber/terapia , Cílio Conector dos Fotorreceptores/metabolismo , Proteínas/genética , Animais , Proteínas do Citoesqueleto , Modelos Animais de Doenças , Eletrorretinografia , Receptor Quinase 1 Acoplada a Proteína G/metabolismo , Vetores Genéticos , Humanos , Amaurose Congênita de Leber/genética , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Proteínas/uso terapêutico , Degeneração Retiniana/genética , Degeneração Retiniana/terapia
5.
PLoS One ; 4(12): e8438, 2009 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-20041177

RESUMO

BACKGROUND: In one group of gene mutations that cause photoreceptor degeneration in human patients, guanylyl cyclase is overactive in the dark. The ensuing excess opening of cGMP-gated cation channels causes intracellular calcium to rise to toxic levels. The Y99C mutation in guanylate cyclase-activating protein 1 (GCAP1) has been shown to act this way. We determined whether prolonged light exposure, which lowers cGMP levels through activation of phototransduction, might protect photoreceptors in a line of transgenic mice carrying the GCAP1-Y99C. METHODOLOGY/PRINCIPAL FINDINGS: We reared cohorts of GCAP1-Y99C transgenic mice under standard cyclic, constant dark and constant light conditions. Mouse eyes were analyzed by histology and by immunofluorescence for GFAP upregulation, a non-specific marker for photoreceptor degeneration. Full-field electroretinograms (ERGs) were recorded to assess retinal function. Consistent with our hypothesis, constant darkness accelerated disease, while continuous lighting arrested photoreceptor degeneration. CONCLUSIONS/SIGNIFICANCE: In contrast to most forms of retinal degeneration, which are exacerbated by increased exposure to ambient light, a subset with mutations that cause overly active guanylyl cyclase and high intracellular calcium benefitted from prolonged light exposure. These findings may have therapeutic implications for patients with these types of genetic defects.


Assuntos
Cálcio/metabolismo , Escuridão , Células Fotorreceptoras de Vertebrados/patologia , Animais , Modelos Animais de Doenças , Proteínas Ativadoras de Guanilato Ciclase/metabolismo , Camundongos , Camundongos Mutantes , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/efeitos da radiação , Retina/metabolismo , Retina/fisiopatologia , Retina/efeitos da radiação
6.
Invest Ophthalmol Vis Sci ; 48(9): 3954-61, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17724172

RESUMO

PURPOSE: Gene therapy for retinal degeneration requires well-defined promoters that drive expression in rod and cone photoreceptors. This study was undertaken to develop short, active derivatives of the human rhodopsin kinase (RK) gene promoter for targeting transgene expression in rods and cones. RK, also known as G protein-coupled receptor kinase 1 (GRK1), is a component of the light adaptation pathway expressed in rods and cones. METHODS: Human RK (hRK) promoter and its concatemers or derivatives extending into the conserved 5' untranslated region (5'-UTR) were assayed for promoter activity in WERI retinoblastoma or Crx/Sp1-supplemented HEK-293 cells. The derivative displaying the highest activity was linked to a GFP reporter and packaged in a pseudotyped adenoassociated viral vector (AAV2/5). The AAV vector was tested in vivo by subretinal injections in wild-type mice, in the all-cone Nrl(-/-) mice, and in the cone-rich diurnal Nile grass rat (Arvicanthis niloticus). Control eyes received a similar AAV2/5 vector carrying a mouse rod opsin (mOps) promoter-controlled GFP reporter. RESULTS: The hRK promoter with the full 5' untranslated sequence (-112 to +180) was the most active in cell culture. Delivered by the AAV2/5 vector, RK promoter drove GFP expression specifically in photoreceptors. In rods, hRK promoter-mediated expression was as efficient as, but appeared more uniform than, mOps promoter-mediated expression. In cones, the hRK promoter drove expression, whereas the mOps promoter did not. CONCLUSIONS: The hRK promoter is active and specific for rod and cone photoreceptors. Because of its small size and proven activity in cones, it is a promoter of choice for somatic gene transfer and gene therapy targeting rods and cones.


Assuntos
Dependovirus/genética , Receptor Quinase 1 Acoplada a Proteína G/genética , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Regiões Promotoras Genéticas/genética , Animais , Marcação de Genes , Vetores Genéticos , Rim/embriologia , Substâncias Luminescentes , Camundongos , Plasmídeos , Ratos , Neoplasias da Retina/genética , Retinoblastoma/genética , Opsinas de Bastonetes/genética , Transfecção , Transgenes , Células Tumorais Cultivadas
7.
J Neurochem ; 101(5): 1294-302, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17442056

RESUMO

Retinal abnormalities are well documented in patients with ganglioside storage diseases. The total content and distribution of retinal glycosphingolipids was studied for the first time in control mice and in Sandhoff disease (SD) and GM1 gangliosidosis mice. Light and electron microscopy of the SD and the GM1 retinas revealed storage in ganglion cells. Similar to previous findings in rat retina, GD3 was the major ganglioside in mouse retina, while GM2 and GM1 were minor species. Total ganglioside content was 44% and 40% higher in the SD and the GM1 retinas, respectively, than in the control retinas. Furthermore, GM2 and GM1 content were 11-fold and 51-fold higher in the SD and the GM1 retinas than in the control retinas, respectively. High concentrations of asialo-GM2 and asialo-GM1 were found in the SD and the GM1 retinas, respectively, but were undetectable in the control retinas. The GSL abnormalities in the SD and the GM1 retinas reflect significant reductions in beta-hexosaminidase and beta-galactosidase enzyme activities, respectively. Although electroretinograms appeared normal in the SD and the GM1 mice, visual evoked potentials were subnormal in both mutants, indicating visual impairments. Our findings present a model system for assessing retinal pathobiology and therapies for the gangliosidoses.


Assuntos
Gangliosidose GM1/genética , Retina/metabolismo , Retina/patologia , Doença de Sandhoff/patologia , Doença de Sandhoff/fisiopatologia , Fatores Etários , Animais , Cromatografia Líquida de Alta Pressão/métodos , Modelos Animais de Doenças , Eletrorretinografia , Potenciais Evocados Visuais/genética , Feminino , Gangliosidoses/metabolismo , Regulação Enzimológica da Expressão Gênica/genética , Masculino , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão/métodos , Retina/fisiopatologia , Retina/ultraestrutura , Doença de Sandhoff/enzimologia , Doença de Sandhoff/genética , beta-Galactosidase/genética , beta-N-Acetil-Hexosaminidases/deficiência , beta-N-Acetil-Hexosaminidases/metabolismo
8.
Am J Ophthalmol ; 142(3): 515-8, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16935610

RESUMO

PURPOSE: We investigated whether opsin mislocalization occurs in photoreceptors in a female carrier of X-linked retinitis pigmentosa with a Gly436Asp mutation in the retinitis pigmentosa GTPase regulator gene (RPGR). DESIGN: Histologic findings in autopsy eyes from a carrier were compared with those from a normal female. METHODS: Frozen retinal sections from the periphery of one eye of the carrier and the normal were stained with antibodies against either human red or green opsins, blue cone opsin, or rhodopsin and labeled with fluorochrome conjugated secondary antibodies. Cell nuclei were counterstained with Hoechst dye. Fellow eyes were evaluated with light microscopy. RESULTS: Fluorescent labeling showed mislocalized cone and rod opsins in photoreceptor cells only in the carrier. The carrier also showed some loss of photoreceptor nuclei. CONCLUSIONS: A defect in trafficking of opsins to outer segments exists in a carrier with the RPGR Gly436Asp mutation.


Assuntos
Proteínas do Olho/genética , Doenças Genéticas Ligadas ao Cromossomo X/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Mutação Puntual , Retinose Pigmentar/metabolismo , Opsinas de Bastonetes/metabolismo , Idoso , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Doenças Genéticas Ligadas ao Cromossomo X/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Heterozigoto , Humanos , Retinose Pigmentar/genética
9.
Cancer Cell ; 9(1): 13-22, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16413468

RESUMO

Cyclin D1 is a multifunctional protein that activates CDK4 and CDK6, titrates Cip/Kip CDK inhibitors to increase CDK2 activity, and modulates the function of certain transcription factors. To specifically test the importance of cyclin D1-associated kinase activity, we generated "knockin" mice expressing mutant cyclin D1 deficient in activating CDK4/6. The development of several cyclin D1-dependent compartments, including mammary glands, proceeds relatively normally in these animals, demonstrating that cyclin D1-associated kinase activity is largely dispensable for development of these tissues. Strikingly, knockin mice were resistant to breast cancers initiated by ErbB-2. These results demonstrate a differential requirement for cyclin D1-CDK4/6 kinase activity in development versus tumorigenesis and strongly support cyclin D1-dependent kinase activity as a specific therapeutic target in breast cancer.


Assuntos
Ciclina D1/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Quinase 6 Dependente de Ciclina/metabolismo , Glândulas Mamárias Animais/crescimento & desenvolvimento , Neoplasias Mamárias Experimentais/metabolismo , Animais , Ciclina D1/genética , Quinase 4 Dependente de Ciclina/genética , Quinase 6 Dependente de Ciclina/genética , Ativação Enzimática , Feminino , Genes erbB-2 , Glândulas Mamárias Animais/enzimologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Mutantes , Mutação , Ligação Proteica , Retina/enzimologia , Retina/crescimento & desenvolvimento
10.
Invest Ophthalmol Vis Sci ; 46(9): 3039-45, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16123399

RESUMO

PURPOSE: Retinitis pigmentosa GTPase regulator (RPGR) is a photoreceptor protein anchored in the connecting cilia by an RPGR-interacting protein (RPGRIP). Loss of RPGRIP causes Leber congenital amaurosis (LCA), a severe form of photoreceptor degeneration. The current study was an investigation of whether somatic gene replacement could rescue degenerating photoreceptors in a murine model of LCA due to a defect in RPGRIP. METHODS: An RPGRIP expression cassette, driven by a mouse opsin promoter, was packaged into recombinant adeno-associated virus (AAV). The AAV vector was delivered into the right eyes of RPGRIP(-/-) mice by a single subretinal injection into the superior hemisphere. The left eyes received a saline injection as a control. Full-field electroretinograms (ERGs) were recorded from both eyes at 2, 3, 4, and 5 months after injection. After the final follow-up, retinas were analyzed by immunostaining or by light and electron microscopy. RESULTS: Delivery of the AAV vector led to RPGRIP expression and restoration of normal RPGR localization at the connecting cilia. Photoreceptor preservation was evident by a thicker cell layer and well-developed outer segments in the treated eyes. Rescue was more pronounced in the superior hemisphere coincident with the site of delivery. Functional preservation was demonstrated by ERG. CONCLUSIONS: AAV-mediated RPGRIP gene replacement preserves photoreceptor structure and function in a mouse model of LCA, despite ongoing cell loss at the time of intervention. These results indicate that gene replacement therapy may be effective in patients with LCA due to a defect in RPGRIP and suggest that further preclinical development of gene therapy for this disorder is warranted.


Assuntos
Cegueira/terapia , Regulação da Expressão Gênica/fisiologia , Terapia Genética , Células Fotorreceptoras de Vertebrados/metabolismo , Proteínas/genética , Degeneração Retiniana/terapia , Animais , Cegueira/congênito , Cegueira/metabolismo , Proteínas do Citoesqueleto , Dependovirus/genética , Modelos Animais de Doenças , Eletrorretinografia , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Camundongos , Camundongos Knockout , Células Fotorreceptoras de Vertebrados/ultraestrutura , Regiões Promotoras Genéticas , Proteínas/metabolismo , Degeneração Retiniana/metabolismo , Degeneração Retiniana/patologia , Opsinas de Bastonetes/genética , Transfecção
11.
Invest Ophthalmol Vis Sci ; 46(2): 435-41, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15671266

RESUMO

PURPOSE: The retinitis pigmentosa GTPase regulator (RPGR) is essential for the maintenance of photoreceptor viability. RPGR is expressed as constitutive and ORF15 variants because of alternative splicing. This study was designed to examine whether the retina-specific ORF15 variant alone could substantially substitute for RPGR function. A further objective was to test whether the highly repetitive purine-rich region of ORF15 could be abbreviated without ablating the function, so as to accommodate RPGR replacement genes in adenoassociated virus (AAV) vectors. METHODS: A cDNA representing RPGR-ORF15 but shortened by 654 bp in the repetitive region was placed under the control of a chicken beta-actin (CBA) hybrid promoter. The resultant construct was transfected into mouse embryonic stem cells. Clones expressing the transgene were selected and injected into mouse blastocysts. Transgenic chimeras were crossed with RPGR knockout (KO) mice. Mice expressing the transgene but null for endogenous RPGR (Tg/KO) were studied from 1 month to 18 months of age by light and electron microscopy, immunofluorescence, and electroretinography (ERG). The results were compared with those of wild-type (WT) and RPGR-null control mice. RESULTS: Transgenic RPGR-ORF15 was found in the connecting cilia of rod and cone photoreceptors, at approximately 20% of the WT level. Photoreceptor morphology, cone opsin localization, expression of GFAP (a marker for retinal degeneration) and ERGs were consistent with the transgene exerting substantial rescue of retinal degeneration due to loss of endogenous RPGR. CONCLUSIONS: RPGR-ORF15 is the functionally significant variant in photoreceptors. The length of its repetitive region can be reduced while preserving its function. The current findings should facilitate the design of gene replacement therapy for RPGR-null mutations.


Assuntos
Proteínas de Transporte/genética , Proteínas do Olho/genética , Regulação da Expressão Gênica/fisiologia , Células Fotorreceptoras de Vertebrados/metabolismo , Proteínas Recombinantes de Fusão/genética , Retinose Pigmentar/prevenção & controle , Animais , Proteínas do Citoesqueleto , Dependovirus/genética , Eletrorretinografia , Éxons/genética , Técnica Indireta de Fluorescência para Anticorpo , Vetores Genéticos , Proteína Glial Fibrilar Ácida/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Immunoblotting , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fases de Leitura Aberta/genética , Proteínas/metabolismo , Retinose Pigmentar/metabolismo , Retinose Pigmentar/patologia , Opsinas de Bastonetes/metabolismo , Transfecção , Transgenes
12.
Invest Ophthalmol Vis Sci ; 45(1): 36-41, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14691151

RESUMO

PURPOSE: The retinitis pigmentosa GTPase regulator (RPGR) is essential in the maintenance of photoreceptor viability. Mutations in the X-linked RPGR gene have generally been assumed to be recessive. This study was undertaken to investigate whether certain mutant RPGR alleles may act dominantly. METHODS: An RPGR transgene representing the RPGR ORF15 variant was placed under a non-tissue-specific promoter and introduced into transgenic mice. The transgene was crossed into both a wild type (WT) and an RPGR null background. Its expression was analyzed by RT-PCR, immunoblot analysis, and immunofluorescence. Photoreceptor survival was assessed by electroretinography and histology. RESULTS: The RPGR transgene transcript underwent photoreceptor-specific, alternative splicing involving the purine-rich region of the ORF15 exon, generating a shortened mRNA and a premature stop codon. This truncation mutant caused more rapid photoreceptor degeneration than that in the RPGR null (knockout) mutant. The disease course was similar, whether the transgene was coexpressed with WT RPGR or expressed alone in the RPGR null background. CONCLUSIONS: Certain truncated forms of RPGR can behave as a dominant, gain-of-function mutant. These data suggest that human RPGR mutations are not necessarily null and some may also act as dominant alleles, leading to a more severe phenotype than a null mutant.


Assuntos
Proteínas de Transporte/genética , Proteínas do Olho , Doenças Genéticas Ligadas ao Cromossomo X/genética , Retinose Pigmentar/genética , Processamento Alternativo/genética , Animais , Células COS , Eletrorretinografia , Regulação da Expressão Gênica/fisiologia , Genes Dominantes , Doenças Genéticas Ligadas ao Cromossomo X/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Fases de Leitura Aberta/genética , Células Fotorreceptoras de Vertebrados/patologia , RNA Mensageiro/metabolismo , Retinose Pigmentar/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Transgenes
13.
EMBO J ; 23(1): 89-99, 2004 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-14685281

RESUMO

Age-related macular degeneration (ARMD) with abnormal deposit formation under the retinal pigment epithelium (RPE) is the major cause of blindness in the Western world. basal laminar deposits are found in early ARMD and are composed of excess basement membrane material produced by the RPE. Here, we demonstrate that mice lacking the basement membrane component collagen XVIII/endostatin have massive accumulation of sub-RPE deposits with striking similarities to basal laminar deposits, abnormal RPE, and age-dependent loss of vision. The progressive attenuation of visual function results from decreased retinal rhodopsin content as a consequence of abnormal vitamin A metabolism in the RPE. In addition, aged mutant mice show photoreceptor abnormalities and increased expression of glial fibrillary acidic protein in the neural retina. Our data demonstrate that collagen XVIII/endostatin is essential for RPE function, and suggest an important role of this collagen in Bruch's membrane. Consistent with such a role, the ultrastructural organization of collagen XVIII/endostatin in basement membranes, including Bruch's membrane, shows that it is part of basement membrane molecular networks.


Assuntos
Colágeno Tipo XVIII/deficiência , Endostatinas/deficiência , Epitélio Pigmentado Ocular/patologia , Visão Ocular , Envelhecimento , Animais , Western Blotting , Lâmina Basilar da Corioide/química , Lâmina Basilar da Corioide/ultraestrutura , Galinhas , Colágeno Tipo XVIII/química , Colágeno Tipo XVIII/genética , Colágeno Tipo XVIII/isolamento & purificação , Colágeno Tipo XVIII/metabolismo , Colágeno Tipo XVIII/ultraestrutura , Eletrorretinografia/efeitos dos fármacos , Endostatinas/genética , Endostatinas/metabolismo , Endostatinas/ultraestrutura , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Degeneração Macular/patologia , Camundongos , Camundongos Mutantes , Modelos Biológicos , Epitélio Pigmentado Ocular/metabolismo , Epitélio Pigmentado Ocular/ultraestrutura , Estrutura Terciária de Proteína , Retina/química , Retina/patologia , Retina/fisiologia , Rodopsina/análise , Pele/química , Pele/citologia , Vitamina A/farmacologia
14.
Invest Ophthalmol Vis Sci ; 43(6): 1912-5, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12036998

RESUMO

PURPOSE: Because of a previous report suggesting that D-cis-diltiazem slows retinal degeneration in rd mice, this study was undertaken to examine the effect of D-cis-diltiazem on photoreceptor structure and function in this line of mice. METHODS: Mice were randomly assigned to daily intraperitoneal injections of D-cis-diltiazem or saline between postnatal days 9 and 24. On postnatal day 26 or 27, retinal function was assessed by recording dark-adapted bright-flash ERGs in all animals. Retinal morphology was examined in fixed sections and in immunolabeled frozen sections. Examiners were masked to the treatment group assignment. RESULTS: On postnatal days 26 and 27, diltiazem- and saline-treated mice had only one row of remaining photoreceptor cells throughout most of the central retina. Cone cells in the periphery had remnants of inner segments. Total cell counts and separate counts of rod and cone photoreceptor cells by immunostaining were similar in the diltiazem- versus saline-treated mice. Both groups of mice had, on average, comparable subnormal ERG amplitudes. CONCLUSIONS: D-cis-Diltiazem had no detectable effect on preservation of photoreceptor structure and function in rd mice.


Assuntos
Bloqueadores dos Canais de Cálcio/uso terapêutico , Diltiazem/uso terapêutico , Células Fotorreceptoras de Vertebrados/efeitos dos fármacos , Retinose Pigmentar/tratamento farmacológico , 3',5'-GMP Cíclico Fosfodiesterases/genética , Animais , Contagem de Células , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6 , Adaptação à Escuridão , Eletrorretinografia , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos C3H , Estimulação Luminosa , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/fisiologia , Retinose Pigmentar/metabolismo , Retinose Pigmentar/fisiopatologia , Rodopsina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...