Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Genes (Basel) ; 14(11)2023 Nov 17.
Article in English | MEDLINE | ID: mdl-38003037

ABSTRACT

Hundreds of genetic variants associated with canine traits and disorders have been identified, with commercial tests offered. However, the geographic distributions and changes in allele and genotype frequencies over prolonged, continuous periods of time are lacking. This study utilized a large set of genotypes from dogs tested for the progressive rod-cone degeneration-progressive retinal atrophy (prcd-PRA) G>A missense PRCD variant (n = 86,667) and the collie eye anomaly (CEA)-associated NHEJ1 deletion (n = 33,834) provided by the commercial genetic testing company (Optigen/Wisdom Panel, Mars Petcare Science & Diagnostics). These data were analyzed using the chi-square goodness-of-fit test, time-trend graphical analysis, and regression modeling in order to evaluate how test results changed over time. The results span fifteen years, representing 82 countries and 67 breeds/breed mixes. Both diseases exhibited significant differences in genotype frequencies (p = 2.7 × 10-152 for prcd-PRA and 0.023 for CEA) with opposing graphical trends. Regression modeling showed time progression to significantly affect the odds of a dog being homozygous or heterozygous for either disease, as do variables including breed and breed popularity. This study shows that genetic testing informed breeding decisions to produce fewer affected dogs. However, the presence of dogs homozygous for the disease variant, especially for prcd-PRA, was still observed fourteen years after test availability, potentially due to crosses of unknown carriers. This suggests that genetic testing of dog populations should continue.


Subject(s)
Retinal Degeneration , Dogs , Animals , Pedigree , Retinal Degeneration/genetics , Retinal Degeneration/veterinary , Genetic Testing , Genotype , Atrophy
2.
PLoS Genet ; 16(9): e1008956, 2020 09.
Article in English | MEDLINE | ID: mdl-32911491

ABSTRACT

The genomic diversity of the domestic dog is an invaluable resource for advancing understanding of mammalian biology, evolutionary biology, morphologic variation, and behavior. There are approximately 350 recognized breeds in the world today, many established through hybridization and selection followed by intense breeding programs aimed at retaining or enhancing specific traits. As a result, many breeds suffer from an excess of particular diseases, one of many factors leading to the recent trend of "designer breed" development, i.e. crossing purebred dogs from existing breeds in the hope that offspring will be enriched for desired traits and characteristics of the parental breeds. We used a dense panel of 150,106 SNPs to analyze the population structure of the Australian labradoodle (ALBD), to understand how such breeds are developed. Haplotype and admixture analyses show that breeds other than the poodle (POOD) and Labrador retriever (LAB) contributed to ALBD formation, but that the breed is, at the genetic level, predominantly POOD, with all small and large varieties contributing to its construction. Allele frequency analysis reveals that the breed is enhanced for variants associated with a poodle-like coat, which is perceived by breeders to have an association with hypoallergenicity. We observed little enhancement for LAB-specific alleles. This study provides a blueprint for understanding how dog breeds are formed, highlighting the limited scope of desired traits in defining new breeds.


Subject(s)
Animals, Domestic/genetics , Dogs/genetics , Selection, Genetic/genetics , Alleles , Animals , Australia , Breeding/methods , Gene Frequency/genetics , Genetic Testing , Genetic Variation , Genomics , Genotype , Haplotypes , Phenotype , Phylogeny
3.
BMC Genet ; 14: 27, 2013 Apr 20.
Article in English | MEDLINE | ID: mdl-23601474

ABSTRACT

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.


Subject(s)
Color Vision Defects/genetics , Cyclic Nucleotide-Gated Cation Channels/genetics , Dog Diseases/genetics , Dogs/genetics , Animals , Breeding , Color Vision Defects/veterinary , DNA Mutational Analysis , Founder Effect , Genotype , Linkage Disequilibrium , Phenotype , Sequence Deletion
4.
Mol Vis ; 16: 1549-69, 2010 Aug 11.
Article in English | MEDLINE | ID: mdl-20806078

ABSTRACT

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.


Subject(s)
ADAM Proteins/genetics , Dog Diseases/enzymology , Dog Diseases/genetics , Mutation/genetics , Retinitis Pigmentosa/veterinary , ADAM Proteins/metabolism , Animals , Breeding , Computational Biology , DNA Mutational Analysis , Dog Diseases/physiopathology , Dogs , Electroretinography , Gene Expression Profiling , Gene Expression Regulation , Genetic Testing , Genome-Wide Association Study , Homozygote , Humans , Phenotype , Retina/enzymology , Retina/pathology , Retina/ultrastructure , Retinitis Pigmentosa/enzymology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/physiopathology
5.
Mamm Genome ; 21(7-8): 398-408, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20686772

ABSTRACT

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.


Subject(s)
Collagen Type IX/genetics , Dog Diseases/genetics , Dwarfism/genetics , Eye Diseases, Hereditary/genetics , Animals , Animals, Newborn , Arthritis/genetics , Arthritis/veterinary , Base Sequence , Cataract/genetics , Cataract/veterinary , Collagen Type XI/deficiency , Connective Tissue Diseases/genetics , Connective Tissue Diseases/veterinary , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/veterinary , Dogs , Dwarfism/complications , Eye Diseases, Hereditary/complications , Female , Genes, Recessive , Genetic Association Studies , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/veterinary , Humans , Male , Molecular Sequence Data , Mutation , Osteochondrodysplasias/genetics , Osteochondrodysplasias/veterinary , Pedigree , Retinal Detachment/genetics , Retinal Detachment/veterinary
6.
Invest Ophthalmol Vis Sci ; 51(12): 6793-802, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20671290

ABSTRACT

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.


Subject(s)
Biomarkers/metabolism , Carrier Proteins/genetics , Disease Models, Animal , Eye Proteins/genetics , Leber Congenital Amaurosis/metabolism , Retinal Degeneration/metabolism , Retinal Neurons/metabolism , Retinal Pigment Epithelium/metabolism , cis-trans-Isomerases/genetics , Animals , Dogs , Fluorescent Antibody Technique, Indirect , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/pathology , Microscopy, Confocal , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Neurons/pathology , Retinal Pigment Epithelium/pathology
7.
Mamm Genome ; 20(2): 109-23, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19130129

ABSTRACT

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.


Subject(s)
Dog Diseases/genetics , Eye Proteins/genetics , Nuclear Proteins/genetics , Retinal Dysplasia/veterinary , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Dogs , Humans , Linkage Disequilibrium , Mice , Molecular Sequence Data , Mutation , Pedigree , Retinal Dysplasia/genetics
8.
Mol Vis ; 13: 1094-105, 2007 Jul 11.
Article in English | MEDLINE | ID: mdl-17653054

ABSTRACT

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.


Subject(s)
Disease Models, Animal , Dog Diseases/genetics , Genetic Diseases, X-Linked , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/veterinary , ATP-Binding Cassette Transporters/genetics , Animals , Cyclic Nucleotide Phosphodiesterases, Type 6 , Dogs , Female , Gene Expression , Genotype , Humans , Male , Molecular Chaperones/genetics , Nuclear Pore Complex Proteins/genetics , Nuclear Proteins/genetics , Nucleophosmin , Pedigree , Phenotype , Phosphoric Diester Hydrolases/genetics , Polymorphism, Genetic , RNA, Messenger/metabolism , Retina/metabolism , Severity of Illness Index
9.
Hum Gene Ther ; 17(8): 845-58, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16942444

ABSTRACT

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.


Subject(s)
Blindness/therapy , Dependovirus , Eye Proteins , Genetic Therapy , Optic Atrophy, Hereditary, Leber/therapy , Animals , Blindness/etiology , Blindness/genetics , Blindness/pathology , Brain/metabolism , Brain/pathology , Brain/virology , Carrier Proteins , Dogs , Eye Proteins/genetics , Eye Proteins/metabolism , Humans , Macaca fascicularis , Mutation , Optic Atrophy, Hereditary, Leber/complications , Optic Atrophy, Hereditary, Leber/genetics , Optic Atrophy, Hereditary, Leber/pathology , Optic Nerve/metabolism , Optic Nerve/pathology , Optic Nerve/virology , Photoreceptor Cells/metabolism , Photoreceptor Cells/pathology , Photoreceptor Cells/virology , Retina/metabolism , Retina/pathology , Retina/virology , cis-trans-Isomerases
10.
Genomics ; 88(5): 551-63, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16938425

ABSTRACT

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.


Subject(s)
Dog Diseases/genetics , Dogs/genetics , Point Mutation , Retinal Degeneration/veterinary , Retinitis Pigmentosa/genetics , Amino Acid Sequence , Animals , Breeding , Cloning, Molecular , Conserved Sequence , DNA Mutational Analysis , DNA, Complementary/genetics , Expressed Sequence Tags , Eye Proteins/genetics , Female , Gene Library , Genes, Recessive , Genomics , Homozygote , Humans , Male , Mice , Molecular Sequence Data , Pedigree , Retinal Degeneration/genetics , Sequence Homology, Amino Acid , Species Specificity
11.
Mol Ther ; 13(6): 1074-84, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16644289

ABSTRACT

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.


Subject(s)
Dependovirus/genetics , Eye Proteins/genetics , Genetic Vectors/administration & dosage , Genetic Vectors/adverse effects , Retina/drug effects , Animals , Animals, Genetically Modified , Blindness/genetics , Blindness/therapy , Carrier Proteins , Dogs , Dose-Response Relationship, Drug , Drug-Related Side Effects and Adverse Reactions , Eye Proteins/administration & dosage , Female , Genetic Therapy/methods , Genetic Vectors/pharmacokinetics , Injections, Intralesional , Male , Optic Atrophy, Hereditary, Leber/genetics , Optic Atrophy, Hereditary, Leber/therapy , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Retina/pathology , Tissue Distribution , cis-trans-Isomerases
12.
Mol Ther ; 12(6): 1072-82, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16226919

ABSTRACT

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.


Subject(s)
Blindness/genetics , Blindness/therapy , Dependovirus/genetics , Genetic Therapy/methods , Retinal Cone Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/pathology , Animals , Animals, Genetically Modified , Blotting, Western , Carrier Proteins , Chromatography , DNA, Complementary/metabolism , Disease Models, Animal , Dogs , Electroretinography , Enzyme-Linked Immunosorbent Assay , Eye Proteins/genetics , Gene Deletion , Gene Transfer Techniques , Genetic Vectors , Homozygote , Humans , Immunohistochemistry , Mutation , Promoter Regions, Genetic , Retinal Degeneration/genetics , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transgenes , cis-trans-Isomerases
13.
Proc Natl Acad Sci U S A ; 102(14): 5233-8, 2005 Apr 05.
Article in English | MEDLINE | ID: mdl-15784735

ABSTRACT

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.


Subject(s)
Mutation , Retina/injuries , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Rhodopsin/genetics , Animals , Base Sequence , DNA/genetics , DNA/metabolism , Disease Models, Animal , Dogs , Humans , Light , Nerve Regeneration , Retina/metabolism , Retina/pathology , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinitis Pigmentosa/metabolism , Rhodopsin/metabolism , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
14.
J Biol Chem ; 279(51): 53828-39, 2004 Dec 17.
Article in English | MEDLINE | ID: mdl-15459196

ABSTRACT

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.


Subject(s)
Mutation , Receptors, G-Protein-Coupled/chemistry , Rhodopsin/analogs & derivatives , Rod Opsins/genetics , Alleles , Amino Acid Sequence , Animals , Chromatography, Liquid , Cytoplasm/metabolism , Detergents/pharmacology , Disease Models, Animal , Dogs , Electrophoresis, Polyacrylamide Gel , Glycosylation , Immunoblotting , Immunohistochemistry , Ligands , Light , Mass Spectrometry , Models, Molecular , Molecular Sequence Data , Peptides/chemistry , Protein Structure, Tertiary , Proteins/chemistry , Receptors, G-Protein-Coupled/metabolism , Retina/pathology , Retinitis Pigmentosa/genetics , Retinoids/metabolism , Rhodopsin/chemistry , Rod Cell Outer Segment , Rod Opsins/metabolism , Time Factors , Ultraviolet Rays , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
15.
Gene ; 294(1-2): 167-76, 2002 Jul 10.
Article in English | MEDLINE | ID: mdl-12234678

ABSTRACT

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.


Subject(s)
DNA-Binding Proteins/genetics , Dogs/genetics , Amino Acid Sequence , Animals , Chromosome Mapping , Cloning, Molecular , DNA/chemistry , DNA/genetics , DNA, Complementary/chemistry , DNA, Complementary/genetics , DNA-Binding Proteins/metabolism , Gene Expression , Immunohistochemistry , Molecular Sequence Data , Radiation Hybrid Mapping , Retina/metabolism , Retina/pathology , Retinal Degeneration/genetics , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid
16.
Proc Natl Acad Sci U S A ; 99(9): 6328-33, 2002 Apr 30.
Article in English | MEDLINE | ID: mdl-11972042

ABSTRACT

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.


Subject(s)
Mutation , Retinitis Pigmentosa/genetics , Rhodopsin/genetics , Animals , Cell Membrane/metabolism , DNA Mutational Analysis , Dogs , Electroretinography , Genes, Dominant , Genetic Linkage , Humans , Immunohistochemistry , Molecular Sequence Data , Phenotype , Photoreceptor Cells , Point Mutation , Protein Structure, Secondary , Time Factors , Tomography , X Chromosome
SELECTION OF CITATIONS
SEARCH DETAIL
...