Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
BMC Genet ; 14: 27, 2013 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-23601474

RESUMEN

BACKGROUND: Achromatopsia is an autosomal recessive disease characterized by the loss of cone photoreceptor function that results in day-blindness, total colorblindness, and decreased central visual acuity. The most common causes for the disease are mutations in the CNGB3 gene, coding for the beta subunit of the cyclic nucleotide-gated channels in cones. CNGB3-achromatopsia, or cone degeneration (cd), is also known to occur in two canine breeds, the Alaskan malamute (AM) and the German shorthaired pointer. RESULTS: Here we report an in-depth characterization of the achromatopsia phenotype in a new canine breed, the miniature Australian shepherd (MAS). Genotyping revealed that the dog was homozygous for a complete genomic deletion of the CNGB3 gene, as has been previously observed in the AM. Identical breakpoints on chromosome 29 were identified in both the affected AM and MAS with a resulting deletion of 404,820 bp. Pooled DNA samples of unrelated purebred Australian shepherd, MAS, Siberian husky, Samoyed and Alaskan sled dogs were screened for the presence of the affected allele; one Siberian husky and three Alaskan sled dogs were identified as carriers. The affected chromosomes from the AM, MAS, and Siberian husky were genotyped for 147 SNPs in a 3.93 Mb interval within the cd locus. An identical shared affected haplotype, 0.5 Mb long, was observed in all three breeds and defined the minimal linkage disequilibrium (LD) across breeds. This supports the idea that the mutated allele was identical by descent (IBD). CONCLUSION: We report the occurrence of CNGB3-achromatopsia in a new canine breed, the MAS. The CNGB3-deletion allele previously described in the AM was also observed in a homozygous state in the affected MAS, as well as in a heterozygous carrier state in a Siberian husky and Alaskan sled dogs. All affected alleles were shown to be IBD, strongly suggesting an affected founder effect. Since the MAS is not known to be genetically related to the AM, other breeds may potentially carry the same cd-allele and be affected by achromatopsia.


Asunto(s)
Defectos de la Visión Cromática/genética , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Enfermedades de los Perros/genética , Perros/genética , Animales , Cruzamiento , Defectos de la Visión Cromática/veterinaria , Análisis Mutacional de ADN , Efecto Fundador , Genotipo , Desequilibrio de Ligamiento , Fenotipo , Eliminación de Secuencia
2.
Mamm Genome ; 21(7-8): 398-408, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20686772

RESUMEN

Oculoskeletal dysplasia segregates as an autosomal recessive trait in the Labrador retriever and Samoyed canine breeds, in which the causative loci have been termed drd1 and drd2, respectively. Affected dogs exhibit short-limbed dwarfism and severe ocular defects. The disease phenotype resembles human hereditary arthro-ophthalmopathies such as Stickler and Marshall syndromes, although these disorders are usually dominant. Linkage studies mapped drd1 to canine chromosome 24 and drd2 to canine chromosome 15. Positional candidate gene analysis then led to the identification of a 1-base insertional mutation in exon 1 of COL9A3 that cosegregates with drd1 and a 1,267-bp deletion mutation in the 5' end of COL9A2 that cosegregates with drd2. Both mutations affect the COL3 domain of the respective gene. Northern analysis showed that RNA expression of the respective genes was reduced in affected retinas. These models offer potential for studies such as protein-protein interactions between different members of the collagen gene family, regulation and expression of these genes in retina and cartilage, and even opportunities for gene therapy.


Asunto(s)
Colágeno Tipo IX/genética , Enfermedades de los Perros/genética , Enanismo/genética , Enfermedades Hereditarias del Ojo/genética , Animales , Animales Recién Nacidos , Artritis/genética , Artritis/veterinaria , Secuencia de Bases , Catarata/genética , Catarata/veterinaria , Colágeno Tipo XI/deficiencia , Enfermedades del Tejido Conjuntivo/genética , Enfermedades del Tejido Conjuntivo/veterinaria , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/veterinaria , Perros , Enanismo/complicaciones , Enfermedades Hereditarias del Ojo/complicaciones , Femenino , Genes Recesivos , Estudios de Asociación Genética , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/veterinaria , Humanos , Masculino , Datos de Secuencia Molecular , Mutación , Osteocondrodisplasias/genética , Osteocondrodisplasias/veterinaria , Linaje , Desprendimiento de Retina/genética , Desprendimiento de Retina/veterinaria
3.
Mol Vis ; 16: 1549-69, 2010 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-20806078

RESUMEN

PURPOSE: To identify the causative mutation in a canine cone-rod dystrophy (crd3) that segregates as an adult onset disorder in the Glen of Imaal Terrier breed of dog. METHODS: Glen of Imaal Terriers were ascertained for crd3 phenotype by clinical ophthalmoscopic examination, and in selected cases by electroretinography. Blood samples from affected cases and non-affected controls were collected and used, after DNA extraction, to undertake a genome-wide association study using Affymetrix Version 2 Canine single nucleotide polymorphism chips and 250K Sty Assay protocol. Positional candidate gene analysis was undertaken for genes identified within the peak-association signal region. Retinal morphology of selected crd3-affected dogs was evaluated by light and electron microscopy. RESULTS: A peak association signal exceeding genome-wide significance was identified on canine chromosome 16. Evaluation of genes in this region suggested A Disintegrin And Metalloprotease domain, family member 9 (ADAM9), identified concurrently elsewhere as the cause of human cone-rod dystrophy 9 (CORD9), as a strong positional candidate for canine crd3. Sequence analysis identified a large genomic deletion (over 20 kb) that removed exons 15 and 16 from the ADAM9 transcript, introduced a premature stop, and would remove critical domains from the encoded protein. Light and electron microscopy established that, as in ADAM9 knockout mice, the primary lesion in crd3 appears to be a failure of the apical microvilli of the retinal pigment epithelium to appropriately invest photoreceptor outer segments. By electroretinography, retinal function appears normal in very young crd3-affected dogs, but by 15 months of age, cone dysfunction is present. Subsequently, both rod and cone function degenerate. CONCLUSIONS: Identification of this ADAM9 deletion in crd3-affected dogs establishes this canine disease as orthologous to CORD9 in humans, and offers opportunities for further characterization of the disease process, and potential for genetic therapeutic intervention.


Asunto(s)
Proteínas ADAM/genética , Enfermedades de los Perros/enzimología , Enfermedades de los Perros/genética , Mutación/genética , Retinitis Pigmentosa/veterinaria , Proteínas ADAM/metabolismo , Animales , Cruzamiento , Biología Computacional , Análisis Mutacional de ADN , Enfermedades de los Perros/fisiopatología , Perros , Electrorretinografía , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Pruebas Genéticas , Estudio de Asociación del Genoma Completo , Homocigoto , Humanos , Fenotipo , Retina/enzimología , Retina/patología , Retina/ultraestructura , Retinitis Pigmentosa/enzimología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/fisiopatología
4.
Mol Vis ; 13: 1094-105, 2007 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-17653054

RESUMEN

PURPOSE: Canine X-linked progressive retinal atrophy (XLPRA) is caused by mutations in RPGR exon ORF15, which is also a mutation hotspot in human X-linked retinitis pigmentosa 3 (RP3). The XLPRA1 form of disease has shown extensive phenotypic variability in a colony of dogs that all inherited the same mutant X-chromosome. This variability in onset and severity makes XLPRA1 a valuable model to use to identify genes influencing photoreceptors degeneration in dog and to elucidate molecular mechanisms underlying RP in its human homolog. In this study, RPGRIP1, RANBP2, NPM1, PDE6D, NPHP5, and ABCA4 genes were selected on the basis of interaction with RPGR or RPGRIP1 or their implication in related retinal diseases, and were investigated as candidate genetic modifiers of XLPRA1. METHODS: A pedigree derived from an affected male dog outcrossed to unrelated normal mix bred or purebred females was used. Morphologic examination revealed phenotypic variability in the affected dogs characterized as mild, moderate, or severe. Single nucleotide polymorphisms (SNPs) and indel-containing markers spanning the entire genes were designed, based on the canine sequence and the Broad Institute SNP library, and genotyped on the pedigree. For each candidate gene, haplotypes were identified and their frequencies in severely and moderately affected dogs were compared to detect a putative correlation between a gene-specific haplotype(s), and severity level of the disease. Primers were derived from expressed sequence tags (ESTs) and predicted transcripts to assess the relative retinal expression of the six genes of interest in normal and affected retinas of different ages. RESULTS: Four to seven haplotypes per gene were identified. None of the haplotypes of RPGRIP1, NPM1, PDE6D, NPHP5, RANBP2, and ABCA4 were found to co-segregate with the moderate or severe phenotype. No significant difference in the retinal expression levels of the candidate genes was observed between normal and affected dogs. CONCLUSIONS: The haplotype distribution of RPGRIP1, NPM1, PDE6D, NPHP5, RANBP2, and ABCA4 suggests these genes are not modifiers of the disease phenotype observed in the XLPRA1 pedigree. The RPGRORF15 stop mutation does not affect the retinal expression of these genes at the mRNA level in the pre-degenerate stage of disease, but no conclusions can be made at this time about changes that may occur at the protein level.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedades de los Perros/genética , Enfermedades Genéticas Ligadas al Cromosoma X , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/veterinaria , Transportadoras de Casetes de Unión a ATP/genética , Animales , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6 , Perros , Femenino , Expresión Génica , Genotipo , Humanos , Masculino , Chaperonas Moleculares/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas Nucleares/genética , Nucleofosmina , Linaje , Fenotipo , Hidrolasas Diéster Fosfóricas/genética , Polimorfismo Genético , ARN Mensajero/metabolismo , Retina/metabolismo , Índice de Severidad de la Enfermedad
5.
Hum Gene Ther ; 17(8): 845-58, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16942444

RESUMEN

Leber congenital amaurosis (LCA) is a molecularly heterogeneous disease group that leads to blindness. LCA caused by RPE65 mutations has been studied in animal models and vision has been restored by subretinal delivery of AAV-RPE65 vector. Human ocular gene transfer trials are being considered. Our safety studies of subretinal AAV-2/2.RPE65 in RPE65-mutant dogs showed evidence of modest photoreceptor loss in the injection region in some animals at higher vector doses. We now test the hypothesis that there can be vectorrelated toxicity to the normal monkey, with its human-like retina. Good Laboratory Practice safety studies following single intraocular injections of AAV-2/2.RPE65 in normal cynomolgus monkeys were performed for 1-week and 3-month durations. Systemic toxicity was not identified. Ocular-specific studies included clinical examinations, electroretinography, and retinal histopathology. Signs of ocular inflammation postinjection had almost disappeared by 1 week. At 3 months, electroretinography in vector-injected eyes was no different than in vehicle-injected control eyes or compared with presurgical recordings. Healed sites of retinal perforation from subretinal injections were noted clinically and by histopathology. Foveal architecture in subretinally injected eyes, vector or vehicle, could be abnormal. Morphometry of central retina showed no photoreceptor layer thickness abnormalities occurring in a dose-dependent manner. Vector sequences were present in the injected retina, vitreous, and optic nerve at 1 week but not consistently in the brain. At 3 months, there were no vector sequences in optic nerve and brain. The results allow for consideration of an upper range for no observed adverse effect level in future human trials of subretinal AAV-2/2.RPE65. The potential value of foveal treatment for LCA and other retinal degenerations warrants further research into how to achieve gene transfer without retinal injury from surgical detachment of the retina.


Asunto(s)
Ceguera/terapia , Dependovirus , Proteínas del Ojo , Terapia Genética , Atrofia Óptica Hereditaria de Leber/terapia , Animales , Ceguera/etiología , Ceguera/genética , Ceguera/patología , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/virología , Proteínas Portadoras , Perros , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Humanos , Macaca fascicularis , Mutación , Atrofia Óptica Hereditaria de Leber/complicaciones , Atrofia Óptica Hereditaria de Leber/genética , Atrofia Óptica Hereditaria de Leber/patología , Nervio Óptico/metabolismo , Nervio Óptico/patología , Nervio Óptico/virología , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patología , Células Fotorreceptoras/virología , Retina/metabolismo , Retina/patología , Retina/virología , cis-trans-Isomerasas
6.
Gene ; 294(1-2): 167-76, 2002 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-12234678

RESUMEN

Progressive rod-cone degeneration (prcd) is an autosomal recessive retinal degeneration of dogs that maps to chromosome 9 (CFA9). Positional cloning and candidate gene approaches are presently used to identify the disease-causing gene. To complement these strategies and identify novel candidate genes, we have used a subtraction approach to detect modified gene expression caused by prcd that may be causally associated with the disease, or, alternatively, be involved in the molecular mechanisms leading to the disease phenotype. With this technique we characterized a 4503 nucleotide open reading frame (ORF) within a 5.6 kb cDNA that predicts a protein of 1500 amino acids. The gene shows about 90% homology to the human and rat glucocorticoid receptor DNA binding factor 1 (GRLF1) gene, also known as p190-A. The transcript was detected in several tissues, including retina, and the protein was localized to the photoreceptor cell layer. The canine GRLF1 maps near the telomere of CFA1 close to CRX, a region synteny to human chromosome 19q13 (HSA19q13). Based on its chromosomal location, GRLF1 has been excluded as a candidate gene for prcd. Northern blot analysis also failed to prove down-regulation of the gene in early stages of disease in six different non-allelic canine retinal degenerations. However, we were able to show that in advanced stages of prcd, GRLF1 is expressed in remaining photoreceptor cells, thus, providing a challenging task to uncover the gene's exact function in the retina and degenerative processes.


Asunto(s)
Proteínas de Unión al ADN/genética , Perros/genética , Secuencia de Aminoácidos , Animales , Mapeo Cromosómico , Clonación Molecular , ADN/química , ADN/genética , ADN Complementario/química , ADN Complementario/genética , Proteínas de Unión al ADN/metabolismo , Expresión Génica , Inmunohistoquímica , Datos de Secuencia Molecular , Mapeo de Híbrido por Radiación , Retina/metabolismo , Retina/patología , Degeneración Retiniana/genética , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido
7.
Invest Ophthalmol Vis Sci ; 51(12): 6793-802, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20671290

RESUMEN

PURPOSE: Leber congenital amaurosis (LCA) is a group of childhood-onset retinal diseases characterized by severe visual impairment or blindness. One form is caused by mutations in the RPE65 gene, which encodes the retinal pigment epithelium (RPE) isomerase. In this study, the retinal structure and expression of molecular markers for different retinal cell types were characterized, and differences between control and RPE65 mutant dogs during the temporal evolution of the disease were analyzed. METHODS: Retinas from normal and mutant dogs of different ages were examined by immunofluorescence with a panel of 16 different antibodies. RESULTS: Cones and rods were preserved in the mutant retinas, and the number of cones was normal. However, there was altered expression of cone arrestin and delocalization of rod opsin. The ON bipolar cells showed sprouting of the dendritic arbors toward the outer nuclear layer (ONL) and retraction of their axons in the inner nuclear layer (INL). A decreased expression of GABA, and an increased expression of intermediate filament glial markers was also found in the mutant retinas. These changes were more evident in the adult than the young mutant retinas. CONCLUSIONS: The structure of the retina is well preserved in the mutant retina, but several molecular changes take place in photoreceptors and in bipolar and amacrine cells. Some of these changes are structural, whereas others reflect a change in localization of the examined proteins. This study provides new information that can be applied to the interpretation of outcomes of retinal gene therapy in animal models and humans.


Asunto(s)
Biomarcadores/metabolismo , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Proteínas del Ojo/genética , Amaurosis Congénita de Leber/metabolismo , Degeneración Retiniana/metabolismo , Neuronas Retinianas/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , cis-trans-Isomerasas/genética , Animales , Perros , Técnica del Anticuerpo Fluorescente Indirecta , Amaurosis Congénita de Leber/genética , Amaurosis Congénita de Leber/patología , Microscopía Confocal , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Neuronas Retinianas/patología , Epitelio Pigmentado de la Retina/patología
8.
Mamm Genome ; 20(2): 109-23, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19130129

RESUMEN

Rod-cone dysplasia type 2 (rcd2) is an autosomal recessive disorder that segregates in collie dogs. Linkage disequilibrium and meiotic linkage mapping were combined to take advantage of population structure within this breed and to fine map rcd2 to a 230-kb candidate region that included the gene C1orf36 responsible for human and murine rd3, and within which all affected dogs were homozygous for one haplotype. In one of three identified canine retinal RD3 splice variants, an insertion was found that cosegregates with rcd2 and is predicted to alter the last 61 codons of the normal open reading frame and further extend the open reading frame. Thus, combined meiotic linkage and LD mapping within a single canine breed can yield critical reduction of the disease interval when appropriate advantage is taken of within-breed population structure. This should permit a similar approach to tackle other hereditary traits that segregate in single closed populations.


Asunto(s)
Enfermedades de los Perros/genética , Proteínas del Ojo/genética , Proteínas Nucleares/genética , Displasia Retiniana/veterinaria , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Mapeo Cromosómico , Perros , Humanos , Desequilibrio de Ligamiento , Ratones , Datos de Secuencia Molecular , Mutación , Linaje , Displasia Retiniana/genética
9.
Genomics ; 88(5): 551-63, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16938425

RESUMEN

Progressive rod-cone degeneration (prcd) is a late-onset, autosomal recessive photoreceptor degeneration of dogs and a homolog for some forms of human retinitis pigmentosa (RP). Previously, the disease-relevant interval was reduced to a 106-kb region on CFA9, and a common phenotype-specific haplotype was identified in all affected dogs from several different breeds and breed varieties. Screening of a canine retinal EST library identified partial cDNAs for novel candidate genes in the disease-relevant interval. The complete cDNA of one of these, PRCD, was cloned in dog, human, and mouse. The gene codes for a 54-amino-acid (aa) protein in dog and human and a 53-aa protein in the mouse; the first 24 aa, coded for by exon 1, are highly conserved in 14 vertebrate species. A homozygous mutation (TGC --> TAC) in the second codon shows complete concordance with the disorder in 18 different dog breeds/breed varieties tested. The same homozygous mutation was identified in a human patient from Bangladesh with autosomal recessive RP. Expression studies support the predominant expression of this gene in the retina, with equal expression in the retinal pigment epithelium, photoreceptor, and ganglion cell layers. This study provides strong evidence that a mutation in the novel gene PRCD is the cause of autosomal recessive retinal degeneration in both dogs and humans.


Asunto(s)
Enfermedades de los Perros/genética , Perros/genética , Mutación Puntual , Degeneración Retiniana/veterinaria , Retinitis Pigmentosa/genética , Secuencia de Aminoácidos , Animales , Cruzamiento , Clonación Molecular , Secuencia Conservada , Análisis Mutacional de ADN , ADN Complementario/genética , Etiquetas de Secuencia Expresada , Proteínas del Ojo/genética , Femenino , Biblioteca de Genes , Genes Recesivos , Genómica , Homocigoto , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Linaje , Degeneración Retiniana/genética , Homología de Secuencia de Aminoácido , Especificidad de la Especie
10.
Mol Ther ; 13(6): 1074-84, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16644289

RESUMEN

AAV2 delivery of the RPE65 gene to the retina of blind RPE65-deficient animals restores vision. This strategy is being considered for human trials in RPE65-associated Leber congenital amaurosis (LCA), but toxicity and dose efficacy have not been defined. We studied ocular delivery of AAV-2/2.RPE65 in RPE65-mutant dogs. There was no systemic toxicity. Ocular examinations showed mild or moderate inflammation that resolved over 3 months. Retinal histopathology indicated that traumatic lesions from the injection were common, but thinning within the injection region occurred only at the two highest vector doses. Biodistribution studies at 3 months postinjection showed no vector in optic nerve or visual centers in the brain and only isolated non-dose-related detection in other organs. We also performed biodistribution studies in normal rats at about 2 weeks and 2 months postinjection and vector was not widespread outside the injected eye. Dose-response results in RPE65-mutant dogs indicated that the highest 1.5-log unit range of vector doses proved efficacious. The efficacy and toxicity limits defined in this study lead to suggestions for the design of a subretinal AAV-2/2.RPE65 human trial of RPE65-associated LCA.


Asunto(s)
Dependovirus/genética , Proteínas del Ojo/genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Retina/efectos de los fármacos , Animales , Animales Modificados Genéticamente , Ceguera/genética , Ceguera/terapia , Proteínas Portadoras , Perros , Relación Dosis-Respuesta a Droga , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Proteínas del Ojo/administración & dosificación , Femenino , Terapia Genética/métodos , Vectores Genéticos/farmacocinética , Inyecciones Intralesiones , Masculino , Atrofia Óptica Hereditaria de Leber/genética , Atrofia Óptica Hereditaria de Leber/terapia , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Retina/patología , Distribución Tisular , cis-trans-Isomerasas
11.
Proc Natl Acad Sci U S A ; 102(14): 5233-8, 2005 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-15784735

RESUMEN

Genetic and environmental factors modify the severity of human neurodegenerations. Retinal degenerations caused by rhodopsin gene mutations show severity differences within and between families and even within regions of the same eye. Environmental light is thought to contribute to this variation. In the naturally occurring dog model of the human disorder, we found that modest light levels, as used in routine clinical practice, dramatically accelerated the neurodegeneration. Dynamics of acute retinal injury (consisting of abnormal intraretinal light scattering) were visualized in vivo in real time with high-resolution optical imaging. Long term consequences included fast or slow retinal degeneration or repair of injury depending on the dose of light exposure. These experiments provide a platform to study mechanisms of neuronal injury, repair, compensation, and degeneration. The data also argue for a gene-specific clinical trial of light reduction in human rhodopsin disease.


Asunto(s)
Mutación , Retina/lesiones , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología , Rodopsina/genética , Animales , Secuencia de Bases , ADN/genética , ADN/metabolismo , Modelos Animales de Enfermedad , Perros , Humanos , Luz , Regeneración Nerviosa , Retina/metabolismo , Retina/patología , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Retinitis Pigmentosa/metabolismo , Rodopsina/metabolismo , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo
12.
Mol Ther ; 12(6): 1072-82, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16226919

RESUMEN

The short- and long-term effects of gene therapy using AAV-mediated RPE65 transfer to canine retinal pigment epithelium were investigated in dogs affected with disease caused by RPE65 deficiency. Results with AAV 2/2, 2/1, and 2/5 vector pseudotypes, human or canine RPE65 cDNA, and constitutive or tissue-specific promoters were similar. Subretinally administered vectors restored retinal function in 23 of 26 eyes, but intravitreal injections consistently did not. Photoreceptoral and postreceptoral function in both rod and cone systems improved with therapy. In dogs followed electroretinographically for 3 years, responses remained stable. Biochemical analysis of retinal retinoids indicates that mutant dogs have no detectable 11-cis-retinal, but markedly elevated retinyl esters. Subretinal AAV-RPE65 treatment resulted in detectable 11-cis-retinal expression, limited to treated areas. RPE65 protein expression was limited to retinal pigment epithelium of treated areas. Subretinal AAV-RPE65 vector is well tolerated and does not elicit high antibody levels to the vector or the protein in ocular fluids or serum. In long-term studies, wild-type cDNA is expressed only in target cells. Successful, stable restoration of rod and cone photoreceptor function in these dogs has important implications for treatment of human patients affected with Leber congenital amaurosis caused by RPE65 mutations.


Asunto(s)
Ceguera/genética , Ceguera/terapia , Dependovirus/genética , Terapia Genética/métodos , Células Fotorreceptoras Retinianas Conos/patología , Células Fotorreceptoras Retinianas Bastones/patología , Animales , Animales Modificados Genéticamente , Western Blotting , Proteínas Portadoras , Cromatografía , ADN Complementario/metabolismo , Modelos Animales de Enfermedad , Perros , Electrorretinografía , Ensayo de Inmunoadsorción Enzimática , Proteínas del Ojo/genética , Eliminación de Gen , Técnicas de Transferencia de Gen , Vectores Genéticos , Homocigoto , Humanos , Inmunohistoquímica , Mutación , Regiones Promotoras Genéticas , Degeneración Retiniana/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Transgenes , cis-trans-Isomerasas
13.
J Biol Chem ; 279(51): 53828-39, 2004 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-15459196

RESUMEN

Rho (rhodopsin; opsin plus 11-cis-retinal) is a prototypical G protein-coupled receptor responsible for the capture of a photon in retinal photoreceptor cells. A large number of mutations in the opsin gene associated with autosomal dominant retinitis pigmentosa have been identified. The naturally occurring T4R opsin mutation in the English mastiff dog leads to a progressive retinal degeneration that closely resembles human retinitis pigmentosa caused by the T4K mutation in the opsin gene. Using genetic approaches and biochemical assays, we explored the properties of the T4R mutant protein. Employing immunoaffinity-purified Rho from affected RHO(T4R/T4R) dog retina, we found that the mutation abolished glycosylation at Asn(2), whereas glycosylation at Asn(15) was unaffected, and the mutant opsin localized normally to the rod outer segments. Moreover, we found that T4R Rho(*) lost its chromophore faster as measured by the decay of meta-rhodopsin II and that it was less resistant to heat denaturation. Detergent-solubilized T4R opsin regenerated poorly and interacted abnormally with the G protein transducin (G(t)). Structurally, the mutation affected mainly the "plug" at the intradiscal (extracellular) side of Rho, which is possibly responsible for protecting the chromophore from the access of bulk water. The T4R mutation may represent a novel molecular mechanism of degeneration where the unliganded form of the mutant opsin exerts a detrimental effect by losing its structural integrity.


Asunto(s)
Mutación , Receptores Acoplados a Proteínas G/química , Rodopsina/análogos & derivados , Opsinas de Bastones/genética , Alelos , Secuencia de Aminoácidos , Animales , Cromatografía Liquida , Citoplasma/metabolismo , Detergentes/farmacología , Modelos Animales de Enfermedad , Perros , Electroforesis en Gel de Poliacrilamida , Glicosilación , Immunoblotting , Inmunohistoquímica , Ligandos , Luz , Espectrometría de Masas , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/química , Estructura Terciaria de Proteína , Proteínas/química , Receptores Acoplados a Proteínas G/metabolismo , Retina/patología , Retinitis Pigmentosa/genética , Retinoides/metabolismo , Rodopsina/química , Segmento Externo de la Célula en Bastón , Opsinas de Bastones/metabolismo , Factores de Tiempo , Rayos Ultravioleta , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
14.
Proc Natl Acad Sci U S A ; 99(9): 6328-33, 2002 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-11972042

RESUMEN

Rhodopsin is the G protein-coupled receptor that is activated by light and initiates the transduction cascade leading to night (rod) vision. Naturally occurring pathogenic rhodopsin (RHO) mutations have been previously identified only in humans and are a common cause of dominantly inherited blindness from retinal degeneration. We identified English Mastiff dogs with a naturally occurring dominant retinal degeneration and determined the cause to be a point mutation in the RHO gene (Thr4Arg). Dogs with this mutant allele manifest a retinal phenotype that closely mimics that in humans with RHO mutations. The phenotypic features shared by dog and man include a dramatically slowed time course of recovery of rod photoreceptor function after light exposure and a distinctive topographic pattern to the retinal degeneration. The canine disease offers opportunities to explore the basis of prolonged photoreceptor recovery after light in RHO mutations and determine whether there are links between the dysfunction and apoptotic retinal cell death. The RHO mutant dog also becomes the large animal needed for preclinical trials of therapies for a major subset of human retinopathies.


Asunto(s)
Mutación , Retinitis Pigmentosa/genética , Rodopsina/genética , Animales , Membrana Celular/metabolismo , Análisis Mutacional de ADN , Perros , Electrorretinografía , Genes Dominantes , Ligamiento Genético , Humanos , Inmunohistoquímica , Datos de Secuencia Molecular , Fenotipo , Células Fotorreceptoras , Mutación Puntual , Estructura Secundaria de Proteína , Factores de Tiempo , Tomografía , Cromosoma X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA