Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 24(13): 3699-707, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25855802

RESUMEN

The CNGA3(-/-)/Nrl(-/-) mouse is a cone-dominant model with Cnga3 channel deficiency, which partially mimics the all cone foveal structure of human achromatopsia 2 with CNGA3 mutations. Although subretinal (SR) AAV vector administration can transfect retinal cells efficiently, the injection-induced retinal detachment can cause retinal damage, particularly when SR vector bleb includes the fovea. We therefore explored whether cone function-structure could be rescued in CNGA3(-/-)/Nrl(-/-) mice by intravitreal (IVit) delivery of tyrosine to phenylalanine (Y-F) capsid mutant AAV8. We find that AAV-mediated CNGA3 expression can restore cone function and rescue structure following IVit delivery of AAV8 (Y447, 733F) vector. Rescue was assessed by restoration of the cone-mediated electroretinogram (ERG), optomotor responses, and cone opsin immunohistochemistry. Demonstration of gene therapy in a cone-dominant mouse model by IVit delivery provides a potential alternative vector delivery mode for safely transducing foveal cones in achromatopsia patients and in other human retinal diseases affecting foveal function.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Defectos de la Visión Cromática/genética , Defectos de la Visión Cromática/terapia , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Proteínas del Ojo/genética , Terapia Genética , Células Fotorreceptoras Retinianas Conos/fisiología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Defectos de la Visión Cromática/metabolismo , Defectos de la Visión Cromática/fisiopatología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Proteínas del Ojo/metabolismo , Femenino , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
2.
Nat Med ; 13(6): 685-7, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17515894

RESUMEN

Loss of cone function in the central retina is a pivotal event in the development of severe vision impairment for many prevalent blinding diseases. Complete achromatopsia is a genetic defect resulting in cone vision loss in 1 in 30,000 individuals. Using adeno-associated virus (AAV) gene therapy, we show that it is possible to target cones and rescue both the cone-mediated electroretinogram response and visual acuity in the Gnat2 ( cpfl3 ) mouse model of achromatopsia.


Asunto(s)
Defectos de la Visión Cromática/terapia , Modelos Animales de Enfermedad , Terapia Genética , Células Fotorreceptoras Retinianas Conos/fisiología , Animales , Defectos de la Visión Cromática/genética , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Proteínas de Unión al GTP Heterotriméricas/genética , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Ratones , Ratones Transgénicos
3.
Zhonghua Yan Ke Za Zhi ; 49(6): 521-5, 2013 Jun.
Artículo en Zh | MEDLINE | ID: mdl-24119965

RESUMEN

OBJECTIVE: To investigate the distribution and biological roles of voltage-dependent calcium channel (VDCC) α1F subunit in murine retina. METHODS: Experimental study.α1F(-/-) (homozygous mutant) mice (n = 35) and α1F(+/+) (wild type) mice (n = 35) were used in this study. Immunohistochemistry was performed to determine the expression of VDCC α1F subunit in the mouse retina. Retinae in α1F(-/-) mice and age-matched control mice at 3, 6, 9, 14-day and 3-month after birth were paraffin embedded, sectioned and HE stained, and full-field electroretinogram (ERG) were also recorded at these time points.Statistics were based on independent samples t-test. RESULTS: (1) α1F subunit was absent in α1F(-/-) mice retina. But in α1F(+/+) mice retina, α1F subunit was expressed most strongly in the outer plexiform layer (OPL), less in the inner plexiform layer (IPL) and ganglion cell layer (GCL). (2) OPL thickness in the subunit deficient mice gradually reduced after birth and lost at adult age. (3) In dark-adapted ERGs,standard response showed that the b-wave amplitude of α1F(-/-) mice [(163.8 ± 26.7) µV] significantly decreased compared with that of α1F(+/+) mice [(408.4 ± 54.5) µV] (t = -9.017, P = 0.000), whereas the a-wave amplitude of α1F(-/-) group [(208.2 ± 27.3) µV] was similar to that of control group [(196.0 ± 24.2) µV] (t = 0.748, P = 0.476). CONCLUSION: This study demonstrates that the lack of VDCC α1F subunit affect the structure and function in the OPL of the murine retina.


Asunto(s)
Canales de Calcio Tipo L/genética , Retina/patología , Retina/fisiopatología , Animales , Electrorretinografía , Homocigoto , Ratones , Ratones Noqueados , Mutación
4.
Mol Ther ; 19(2): 234-42, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21139570

RESUMEN

The retinal degeneration 10 (rd10) mouse is a well-characterized model of autosomal recessive retinitis pigmentosa (RP), which carries a spontaneous mutation in the ß subunit of rod cGMP-phosphodiesterase (PDEß). Rd10 mouse exhibits photoreceptor dysfunction and rapid rod photoreceptor degeneration followed by cone degeneration and remodeling of the inner retina. Here, we evaluate whether gene replacement using the fast-acting tyrosine-capsid mutant AAV8 (Y733F) can provide long-term therapy in this model. AAV8 (Y733F)-smCBA-PDEß was subretinally delivered to postnatal day 14 (P14) rd10 mice in one eye only. Six months after injection, spectral domain optical coherence tomography (SD-OCT), electroretinogram (ERG), optomotor behavior tests, and immunohistochemistry showed that AAV8 (Y733F)-mediated PDEß expression restored retinal function and visual behavior and preserved retinal structure in treated rd10 eyes for at least 6 months. This is the first demonstration of long-term phenotypic rescue by gene therapy in an animal model of PDEß-RP. It is also the first example of tyrosine-capsid mutant AAV8 (Y733F)-mediated correction of a retinal phenotype. These results lay the groundwork for the development of PDEß-RP gene therapy trial and suggest that tyrosine-capsid mutant AAV vectors may be effective for treating other rapidly degenerating models of retinal degeneration.


Asunto(s)
Cápside/metabolismo , Dependovirus/genética , Vectores Genéticos/genética , Retinitis Pigmentosa/terapia , Animales , Western Blotting , Modelos Animales de Enfermedad , Electrorretinografía , Terapia Genética , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología
5.
Mol Ther ; 19(2): 293-301, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21045809

RESUMEN

Vectors based on adeno-associated virus serotype 2 (AAV2) have been used extensively in many gene-delivery applications, including several successful clinical trials for one type of Leber congenital amaurosis in the retina. Many studies have focused on improving AAV2 transduction efficiency and cellular specificity by genetically engineering its capsid. We have previously shown that vectors-containing single-point mutations of capsid surface tyrosines in serotypes AAV2, AAV8, and AAV9 displayed significantly increased transduction efficiency in the retina compared with their wild-type counterparts. In the present study, we evaluated the transduction characteristics of AAV2 vectors containing combinations of multiple tyrosine to phenylalanine mutations in seven highly conserved surface-exposed capsid tyrosine residues following subretinal or intravitreal delivery in adult mice. The multiply mutated vectors exhibited different in vivo transduction properties, with some having a unique ability of transgene expression in all retinal layers. Such novel vectors may be useful in developing valuable new therapeutic strategies for the treatment of many genetic diseases.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/genética , Retina/metabolismo , Tirosina/genética , Animales , Ratones , Ratones Endogámicos C57BL , Mutagénesis Sitio-Dirigida , Mutación , Mutación Puntual/genética , Retina/patología
6.
Zhonghua Yan Ke Za Zhi ; 48(8): 755-8, 2012 Aug.
Artículo en Zh | MEDLINE | ID: mdl-23141518

RESUMEN

Achromatopsia is an early onset retinal dystrophy that causes severe visual impairment. To date, four genes have been found to be implicated in achromatopsia-associated mutations: guanine nucleotide-binding protein (GNAT2), cyclic nucleotide-gated channel alpha-3 (CNGA3), cyclic nucleotide-gated channel beta-3 (CNGB3) and phosphodiesterase 6C (PDE6C). Even with early onset, the slow progress and the good responses to gene therapy in animal models render achromatopsia a very attractive candidate for human gene therapy after the successful of the Phase I clinical trials of Leber's congenital amaurosis. With the development of molecular genetics and the therapeutic gene replacement technology, the adeno-associated viral (AAV) vector-mediated gene therapy for achromatopsia in the preclinical animal experiments achieved encouraging progress in the past years. This article briefly reviews the recent research achievements of achromatopsia with gene therapy.


Asunto(s)
Defectos de la Visión Cromática/terapia , Terapia Genética , Humanos
7.
Proc Natl Acad Sci U S A ; 105(39): 15112-7, 2008 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-18809924

RESUMEN

The RPE65 gene encodes the isomerase of the retinoid cycle, the enzymatic pathway that underlies mammalian vision. Mutations in RPE65 disrupt the retinoid cycle and cause a congenital human blindness known as Leber congenital amaurosis (LCA). We used adeno-associated virus-2-based RPE65 gene replacement therapy to treat three young adults with RPE65-LCA and measured their vision before and up to 90 days after the intervention. All three patients showed a statistically significant increase in visual sensitivity at 30 days after treatment localized to retinal areas that had received the vector. There were no changes in the effect between 30 and 90 days. Both cone- and rod-photoreceptor-based vision could be demonstrated in treated areas. For cones, there were increases of up to 1.7 log units (i.e., 50 fold); and for rods, there were gains of up to 4.8 log units (i.e., 63,000 fold). To assess what fraction of full vision potential was restored by gene therapy, we related the degree of light sensitivity to the level of remaining photoreceptors within the treatment area. We found that the intervention could overcome nearly all of the loss of light sensitivity resulting from the biochemical blockade. However, this reconstituted retinoid cycle was not completely normal. Resensitization kinetics of the newly treated rods were remarkably slow and required 8 h or more for the attainment of full sensitivity, compared with <1 h in normal eyes. Cone-sensitivity recovery time was rapid. These results demonstrate dramatic, albeit imperfect, recovery of rod- and cone-photoreceptor-based vision after RPE65 gene therapy.


Asunto(s)
Ceguera/terapia , Proteínas Portadoras/genética , Proteínas del Ojo/genética , Terapia Genética , Isomerasas/genética , Células Fotorreceptoras Retinianas Bastones/fisiopatología , Retinoides/metabolismo , Ceguera/patología , Ceguera/fisiopatología , Dependovirus/genética , Humanos , Células Fotorreceptoras Retinianas Conos/enzimología , Células Fotorreceptoras Retinianas Conos/patología , Células Fotorreceptoras Retinianas Conos/fisiopatología , Células Fotorreceptoras Retinianas Bastones/enzimología , Células Fotorreceptoras Retinianas Bastones/patología , Visión Ocular/fisiología , cis-trans-Isomerasas
8.
Zhonghua Yan Ke Za Zhi ; 47(1): 83-7, 2011 Jan.
Artículo en Zh | MEDLINE | ID: mdl-21418933

RESUMEN

Leber congenital amaurosis (LCA) is an early onset retinal dystrophy that causes severe visual impairment. With the development of molecular genetics and the therapeutic gene replacement technology, the adeno-associated viral (AAV) vector-mediated gene therapy for LCA achieved encouraging progress in the past decade. The success of the Phase I clinical trials of human RPE65 gene therapy for LCA II patients makes it a pioneer in the field of retinal gene therapy and brings light to the cure of other hereditary retinopathy. This article briefly reviews the recent developments in the preclinical animal experiments and Phase I clinical trials for LCA.


Asunto(s)
Terapia Genética , Amaurosis Congénita de Leber/terapia , Ensayos Clínicos Fase I como Asunto , Humanos
9.
Exp Eye Res ; 90(5): 546-54, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20138034

RESUMEN

To clarify whether transduction efficiency and cell type specificity of self-complementary (sc) AAV5 vectors are similar to those of standard, single-stranded AAV5 vectors in normal retina, one micro liter of scAAV5-smCBA-GFP vector (1 x 10(12) genome-containing particles/ml) and AAV5-smCBA-GFP vector (1 x 10(12) genome-containing particles/ml) were subretinally or intravitreally (in both cases through the cornea) injected into the right and left eyes of adult C57BL/6J mice, respectively. On post-injection day (PID) 1, 2, 5, 7, 10, 14, 21, 28 and 35, eyes were enucleated; retinal pigment epithelium (RPE) wholemounts, neuroretinal wholemounts and eyecup sections were prepared to evaluate green fluorescent protein (GFP) expression by fluorescent microscopy. GFP expression following trans-cornea subretinal injection of scAAV5-smCBA-GFP vector was first detected in RPE wholemounts around PID 1 and in neuroretinal wholemounts between PID 2 and 5; GFP expression peaked and stabilized between PID 10-14 in RPE wholemounts and between P14 and P21 in neuroretinal wholemounts with strong, homogeneous green fluorescence covering the entire wholemounts. The frozen sections supported the following findings from the wholemounts: GFP expression appeared first in RPE around PID 1-2 and soon spread to photoreceptors (PR) cells; by PID 7, moderate GFP expression was found mainly in PR and RPE layers; between PID 14 and 21, strong and homogenous GFP expression was observed in RPE and PR cells. GFP expression following subretinal injection of AAV5-smCBA-GFP was first detected in RPE wholemounts around PID 5-7 and in neuroretinal wholemounts around PID 7-10; ssAAV5-mediated GFP expression peaked at PID 21 in RPE wholemounts and around PID 28 in neuroretinal wholemounts; sections from AAV5 treated eyes also supported findings obtained from wholemounts: GFP expression was first detected in RPE and then spread to the PR cells. Peak GFP expression in RPE mediated by scAAV5 was similar to that mediated by AAV5. However, peak GFP expression mediated by scAAV5 in PR cells was stronger than that mediated by AAV5. No GFP fluorescence was detected in any retinal cells (RPE wholemounts, neuroretinal wholemounts and retinal sections) after trans-cornea intravitreal delivery of either scAAV5-GFP or AAV5-GFP. Neither scAAV5 nor AAV5 can transduce retinal cells following trans-cornea intravitreal injection. The scAAV5 vector used in this study directs an earlier onset of transgene expression than the matched AAV5 vector, and has stronger transgene expression in PR cells following subretinal injection. Our data confirm the previous reports that scAAV vectors have an earlier onset than the standard, single strand AAV vectors (Natkunarajah et al., 2008; Yokoi et al., 2007). scAAV5 vectors may be more useful than standard, single-stranded AAV vector when addressing certain RPE and/or PR cell-related models of retinal dystrophy, particularly for mouse models of human retinitis pigmentosa that require rapid and robust transgene expression to prevent early degeneration in PR cells.


Asunto(s)
Dependovirus/genética , Regulación de la Expresión Génica/fisiología , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Animales , Inyecciones , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Retina/efectos de los fármacos , Transgenes , Cuerpo Vítreo
10.
Mol Ther ; 17(3): 463-71, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19066593

RESUMEN

Vectors derived from adeno-associated viruses (AAVs) have become important gene delivery tools for the treatment of many inherited ocular diseases in well-characterized animal models. Previous studies have determined that the viral capsid plays an essential role in the cellular tropism and efficiency of transgene expression. Recently, it was shown that phosphorylation of surface-exposed tyrosine residues from AAV2 capsid targets the viral particles for ubiquitination and proteasome- mediated degradation, and mutations of these tyrosine residues lead to highly efficient vector transduction in vitro and in vivo. Because the tyrosine residues are highly conserved in other AAV serotypes, in this study we evaluated the intraocular transduction characteristics of vectors containing point mutations in surface- exposed capsid tyrosine residues in AAV serotypes 2, 8, and 9. Several of these novel AAV mutants were found to display a strong and widespread transgene expression in many retinal cells after subretinal or intravitreal delivery compared with their wild-type counterparts. For the first time, we show efficient transduction of the ganglion cell layer by AAV serotype 8 or 9 mutant vectors, thus providing additional tools besides AAV2 for targeting these cells. These enhanced AAV vectors have a great potential for future therapeutic applications for retinal degenerations and ocular neovascular diseases.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/genética , Retina/metabolismo , Transgenes/genética , Animales , Dependovirus/clasificación , Ganglión , Vectores Genéticos/administración & dosificación , Inyecciones , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Tirosina/genética , Tirosina/metabolismo
11.
Adv Exp Med Biol ; 664: 639-46, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20238068

RESUMEN

Achromatopsia is an autosomal recessive retinal disease involving loss of cone function that afflicts approximately 1 in 30,000 individuals. Patients with achromatopsia usually have visual acuities lower than 20/200 because of the central vision loss, photophobia, complete color blindness and reduced cone-mediated electroretinographic (ERG) amplitudes. Mutations in three genes have been found to be the primary causes of achromatopsia, including CNGB3 (beta subunit of the cone cyclic nucleotide-gated cation channel), CNGA3 (alpha subunit of the cone cyclic nucleotide-gated cation channel), and GNAT2 (cone specific alpha subunit of transducin). Naturally occurring mouse models with mutations in Cnga3 (cpfl5 mice) and Gnat2 (cpfl3 mice) were discovered at The Jackson Laboratory. A natural occurring canine model with CNGB3 mutations has also been found. These animal models have many of the central phenotypic features of the corresponding human diseases. Using adeno-associated virus (AAV)-mediated gene therapy, we and others show that cone function can be restored in all three models. These data suggest that human achromatopsia may be a good candidate for corrective gene therapy.


Asunto(s)
Defectos de la Visión Cromática/genética , Defectos de la Visión Cromática/terapia , Terapia Genética , Animales , Defectos de la Visión Cromática/fisiopatología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Canales Catiónicos Regulados por Nucleótidos Cíclicos/uso terapéutico , Perros , Proteínas de Unión al GTP Heterotriméricas/genética , Proteínas de Unión al GTP Heterotriméricas/uso terapéutico , Humanos , Ratones , Mutación/genética
12.
Zhonghua Yan Ke Za Zhi ; 46(2): 103-5, 2010 Feb.
Artículo en Zh | MEDLINE | ID: mdl-20388341

RESUMEN

Hereditary retinal diseases (HRD) are a group of both common and severe ocular hereditary diseases. HRD are a major cause of blindness. Human genetic resources in China are plentiful. Through the accomplishment of the human genome project and the widespread application of related genetic techniques, the methods for detecting the mutant genes of HRD has been established, and important findings in the treatment of HRD through gene therapy studies have been obtained. In the past ten years, HRD studies in China have gained significant progress in this field, including several pioneer results in the detection of autosomal dominant retinitis pigmentosa mutant gene. However, significant difference still exists in basic and clinical studies of HRD between the western countries and China. At present, HRD are transforming from "the incurable disease" to "the curable disease". Advancements in gene therapy studies are making the treatment of HRD possible, we should take advantage of these technological innovations to launch our own clinical trials using gene therapy in the treatment of HRD in Chinese patients as soon as possible.


Asunto(s)
Terapia Genética , Enfermedades de la Retina/terapia , China , Humanos , Enfermedades de la Retina/genética
13.
Zhonghua Yan Ke Za Zhi ; 46(2): 186-92, 2010 Feb.
Artículo en Zh | MEDLINE | ID: mdl-20388354

RESUMEN

Hereditary retinal diseases (HRDs), including retinitis pigmentosa, Leber's congenital amaurosis, congenital stationary night blindness, vitelliform macular dystrophy, Stargardt macular dystrophy, etc., are the most common and severe hereditary ocular diseases, which are closely associated with blindness. With the accomplishment of human genome project and the widespread application of genetic study techniques, the way leading to understanding of gene mutations of HRDs has been paved. Many encouraging breakthroughs of gene therapy studies have been made. Among them, the arrayed primer extension chip (Apex) technology greatly improved the efficiency of mutant gene screening of HRDs. Till now, 46 pathogenic genes and 2497 mutation loci have been identified to be related to HRDs. Gene therapy is one of the key treatments for HRDs. The disease causing mutant gene must be detected before the application of gene therapy. This paper reviews the latest progress in the study of gene mutations in HRDs.


Asunto(s)
Enfermedades Genéticas Congénitas/genética , Enfermedades de la Retina/genética , Análisis Mutacional de ADN , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos
14.
Mol Vis ; 15: 267-75, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19190735

RESUMEN

PURPOSE: In an earlier study we found normal adeno-associated viral vector type 2 (AAV2)-mediated GFP expression after intravitreal injection to one eye of normal C57BL/6J mice. However, GFP expression was very poor in the partner eye of the same mouse if this eye received an intravitreal injection of the same vector one month after the initial intravitreal injection. We also found both injections worked well if they were subretinal. In this study, we tested whether the efficiency of subretinal AAV vector transduction is altered by a previous intravitreal injection in the partner eye and more importantly whether therapeutic efficiency is altered in the rd12 mouse (with a recessive RPE65 mutation) after the same injection series. METHODS: One microl of scAAV5-smCBA-GFP (1 x 10(13) genome containing viral particles per ml) was intravitreally injected into the right eyes of four-week-old C57BL/6J mice and 1 microl of scAAV5-smCBA-hRPE65 (1 x 10(13) genome containing viral particles per ml) was intravitreally injected into the right eyes of four-week-old rd12 mice Four weeks later, the same vectors were subretinally injected into the left eyes of the same C57BL/6J and rd12 mice. Left eyes of another cohort of eight-week-old rd12 mice received a single subretinal injection of the same scAAV5-smCBA-hRPE65 vector as the positive control. Dark-adapted electroretinograms (ERGs) were recorded five months after the subretinal injections. AAV-mediated GFP expression in C57BL/6J mice and RPE65 expression and ERG restoration in rd12 mice were evaluated five months after the second subretinal injection. Frozen section analysis was performed for GFP fluorescence in C57BL/6J mice and immunostaining for RPE65 in rd12 eyes. RESULTS: In rd12 mice, dark-adapted ERGs were minimal following the first intravitreal injection of scAAV5-smCBA-RPE65. Following subsequent subretinal injection in the partner eye, dramatic ERG restoration was recorded in that eye. In fact, ERG b-wave amplitudes were statistically similar to those from the eyes that received the initial subretinal injection at a similar age. In C57BL/6J mice, GFP positive cells were detected in eyes following the first intravitreal injection around the injection site. Strong GFP expression in both the retinal pigment epithelium (RPE) and photoreceptor (PR) cells was detected in the partner eyes following the subsequent subretinal injection. Immunostaining of retinal sections with anti-RPE65 antibody showed strong RPE65 expression mainly in the RPE cells of subretinally injected eyes but not in the intravitreally injected eyes except minimally around the injection site. CONCLUSIONS: These results show that an initial intravitreal injection of AAV vectors to one eye of a mouse does not influence AAV-mediated gene expression or related therapeutic effects in the other eye when vectors are administered to the subretinal space. This suggests that the subretinal space possesses a unique immune privilege relative to the vitreous cavity.


Asunto(s)
Proteínas Portadoras/genética , Dependovirus/genética , Proteínas del Ojo/genética , Terapia Genética/métodos , Vectores Genéticos , Retina/metabolismo , Cuerpo Vítreo/metabolismo , Análisis de Varianza , Animales , Proteínas Portadoras/metabolismo , Electrorretinografía , Proteínas del Ojo/metabolismo , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/patología , Epitelio Pigmentado de la Retina/metabolismo , cis-trans-Isomerasas
15.
Vision Res ; 48(3): 377-85, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17950399

RESUMEN

The specificity of retinal cells transduced by AAV serotype 1, 2 or 5 vectors was determined in vivo versus in vitro in the normal P7 mouse in order to develop a rapid and accurate way to anticipate the behavior of AAV vectors in the retina. In vivo results confirm that AAV1 transduces retinal pigment epithelial cells, while AAV2 and AAV5 transduce both RPE and photoreceptor cells by subretinal injection. AAV2 was the only serotype to efficiently transduce inner retinal cells by intravitreal injection. Parallel analysis employing in vitro retinal organ culture showed qualitatively similar AAV-mediated GFP expression as seen in vivo suggesting that organ culture substitute is a useful method to screen new vector transduction patterns, particular in retinal cells in neonatal mice.


Asunto(s)
Dependovirus/clasificación , Dependovirus/genética , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Retina/metabolismo , Animales , Inyecciones , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Células Fotorreceptoras de Vertebrados/metabolismo , Epitelio Pigmentado Ocular/metabolismo , Serotipificación , Transducción Genética , Cuerpo Vítreo
16.
Sci Rep ; 8(1): 4807, 2018 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-29540812

RESUMEN

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

17.
Invest Ophthalmol Vis Sci ; 48(10): 4407-20, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17898259

RESUMEN

PURPOSE: To test the hypothesis that oxidative injury to the retinal pigment epithelium (RPE) may lead to retinal damage similar to that associated with the early stages of age-related macular degeneration (AMD). METHODS: A ribozyme that targets the protective enzyme manganese superoxide dismutase (MnSOD) was expressed in RPE-J cells, and adeno-associated virus (AAV) expressing the ribozyme gene was injected beneath the retinas of adult C57BL/6 mice. The RPE/choroid complex was examined for SOD2 protein levels and protein markers of oxidative damage using immunoblot analysis and LC MS/MS-identification of proteins and nitration sites. Lipids were extracted from retinal tissue and analyzed for the bis-retinoid compounds A2E and iso-A2E. The mice were analyzed by full-field electroretinography (ERG) for light response. Light and electron microscopy were used to measure cytological changes in the retinas. RESULTS: The treatment of RPE-J cells with Rz432 resulted in decreased MnSOD mRNA and protein as well as increased levels of superoxide anion and apoptotic cell death. When delivered by AAV, Rz432 reduced MnSOD protein and increased markers of oxidative damage, including nitrated and carboxyethylpyrrole-modified proteins in the RPE-choroid of mice. Ribozyme delivery caused a progressive loss of electroretinograph response, vacuolization, degeneration of the RPE, thickening of Bruch's membrane, and shortening and disorganization of the photoreceptor outer and inner segments. Progressive thinning of the photoreceptor outer nuclear layer resulted from apoptotic cell death. Similar to the eyes of patients with AMD, ribozyme-treated eyes exhibited increased autofluorescence and elevated levels of A2E and iso-A2E, major bis-retinoid pigments of lipofuscin. CONCLUSIONS: These results support the hypothesis that oxidative damage to the RPE may play a role in some of the key features of AMD.


Asunto(s)
Modelos Animales de Enfermedad , Silenciador del Gen , Degeneración Macular/genética , Superóxido Dismutasa/genética , Animales , Apoptosis , Dependovirus/genética , Electrorretinografía , Vectores Genéticos , Degeneración Macular/metabolismo , Degeneración Macular/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Estrés Oxidativo , Estimulación Luminosa , Epitelio Pigmentado Ocular/efectos de los fármacos , Epitelio Pigmentado Ocular/enzimología , Compuestos de Piridinio/metabolismo , ARN Catalítico/farmacología , ARN Mensajero/metabolismo , Retina/metabolismo , Retina/ultraestructura , Retinoides/metabolismo , Superóxido Dismutasa/metabolismo , Espectrometría de Masas en Tándem
18.
Mol Vis ; 13: 1701-10, 2007 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-17960108

RESUMEN

PURPOSE: Dramatic restoration of retinal function has followed subretinal viral-mediated gene therapy in RPE65-deficient animal models of human Leber congenital amaurosis (LCA) caused by RPE65 mutations. Progress in early-phase clinical trials of RPE65-LCA prompted us to begin development of an in vivo bioassay of clinical grade vector stability for later-phase trials. METHODS: Naturally-occurring Rpe65-mutant rd12 mice (2-4 mo of age) were studied with full-field electroretinograms (ERGs). Flash stimuli (range, -4.1 to 3.6 log scot-cd x s x m(-2)) were used to evoke ERGs in anesthetized, dark-adapted mice. B-wave amplitudes were measured conventionally and luminance-response functions were fit. Leading edges of photoresponses were analyzed with a model of rod phototransduction activation. A unilateral subretinal injection of AAV2-CB(SB)-hRPE65 vector was delivered and therapeutic efficacy of 4 doses spanning a 2 log unit range was studied with ERGs performed about 6 weeks after injection. Uninjected rd12 eyes and wild-type (wt) mice served as controls. RESULTS: Rd12 mice showed substantially smaller amplitudes and lower sensitivities than wt mice for all measured ERG b-wave and photoresponse parameters. For the dose-response study, there was no difference between 0.01X-dosed mice and untreated mutants. Improved receptoral and post-receptoral function was evident for 0.1X, 0.3X, 1X doses: b-wave semi-saturation constants decreased, b-wave amplitudes increased with dose; photoresponses showed faster kinetics and higher maximum amplitudes. ERG b-wave amplitude to a selected stimulus light intensity could provide evidence of biologic activity of the vector; interocular differences in b-wave amplitude comparing treated versus untreated eyes in the same animal also revealed vector efficacy. CONCLUSIONS: We have taken the first steps toward developing an ERG assay of biologic activity of human grade vector for future clinical trials of RPE65-LCA. Faithful murine models of treatable human disease tested with specific ERG protocols may emerge as valuable in vivo bioassays for future human clinical trials of therapy in many retinal degenerative diseases.


Asunto(s)
Proteínas Portadoras/genética , Electrorretinografía , Proteínas del Ojo/genética , Terapia Genética , Mutación , Degeneración Retiniana/genética , Animales , Bioensayo , Ceguera/congénito , Ceguera/terapia , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Mutantes , Atrofia Óptica Hereditaria de Leber/terapia , Estimulación Luminosa , Degeneración Retiniana/diagnóstico , Degeneración Retiniana/fisiopatología , Degeneración Retiniana/terapia , cis-trans-Isomerasas
19.
PLoS One ; 12(11): e0188032, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29131863

RESUMEN

Adeno-associated virus (AAV) vectors are important gene delivery tools for the treatment of many recessively inherited retinal diseases. For example, a wild-type (WT) AAV5 vector can deliver a full-length Cnga3 (cyclic nucleotide-gated channel alpha-3) cDNA to target cells of the cone photoreceptor function loss 5 (cpfl5) mouse, a spontaneous animal model of achromatopsia with a Cnga3 mutation. Gene therapy restores cone-mediated function and blocks cone degeneration in the mice. However, since transgene expression delivered by an AAV vector shows relatively short-term effectiveness, this cannot be regarded as a very successful therapy. AAV2 and AAV8 vectors with capsid mutations have significantly enhanced transduction efficiency in retinas compared to WT AAV controls. In this study, AAV8 (Y447, 733F+T494V)-treated cpfl5 retinas showed greater preservation of short-term cone electroretinogram (ERG) responses than AAV8 (Y447, 733F)- or AAV2 (Y272, 444, 500, 730F+T491V)-mediated treatments. To explore the long-term rescue effect, AAV8 (Y447, 733F+T494V)-treated cpfl5 retinas were evaluated at 9 months following postnatal day 14 (P14) treatment. Rescued ERG responses in the cones of treated cpfl5 eyes decreased with increasing age, but still maintained more than 60% of the WT mouse responses at the oldest time point examined. Expression of CNGA3 and M/S-opsins was maintained in cone outer segments of the treated cpfl5 eyes and was equal to expression in age-matched WT retinas. Near-normal cone-mediated water maze behavior was observed in the treated cpfl5 mice. As these are the longest follow-up data reported thus far, AAV8 with capsid Y-F and T-V mutations may be one of the most effective AAV vectors for long-term treatment in a naturally occurring mouse model of CNGA3 achromatopsia.


Asunto(s)
Cápside , Defectos de la Visión Cromática/terapia , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Dependovirus/genética , Modelos Animales de Enfermedad , Vectores Genéticos , Mutación , Retina/fisiopatología , Animales , Conducta Animal , Defectos de la Visión Cromática/genética , Defectos de la Visión Cromática/fisiopatología , Electrorretinografía , Terapia Genética , Ratones
20.
J Ophthalmol ; 2017: 9721362, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28168050

RESUMEN

The retinal degeneration 11 (rd11) mouse is a new animal model with rapid photoreceptor degeneration. The long-term efficacy of gene therapy has a direct relationship with the onset of photoreceptor degeneration or apoptosis, whereas the degeneration or apoptosis patterns of photoreceptors are still unclear in rd11 mice. The distribution patterns of cone function-related L- and S-opsin were examined by immunofluorescence staining, and the apoptosis was performed by TUNEL assay in rd11 mice. The expression pattern of L-opsin or S-opsin in rd11 retina at postnatal day (P) 14 was similar to the pattern observed in wildtype retina. With increasing age, the expression of L-opsin and S-opsin, especially S-opsin, decreased significantly in rd11 mice. The degeneration of L-opsin began around the optic nerve and expanded to the periphery of the retina, from the ventral/nasal to dorsal/temporal retina, whereas the expression of S-opsin gradually decreased from the dorsal/temporal to ventral/nasal retina. Apoptotic signal appeared at P14 and was strongest at P28 of rd11 mice. The key genes associated with apoptosis confirmed those changes. These indicated that the degeneration and apoptosis of cone photoreceptors began at P14 of rd11 mice, which was a key point for gene therapy.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA