Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Vet Pathol ; 59(2): 310-318, 2022 03.
Article in English | MEDLINE | ID: mdl-34974772

ABSTRACT

Twelve cases of adult-onset blindness were identified in a flock of 130 polled Wiltshire sheep in New Zealand over a 3-year period. Affected sheep developed night blindness between 2 and 3 years of age, which progressed to complete blindness by 4 to 5 years of age. Fundic examination findings included progressive tapetal hyperreflectivity and attenuation of retinal blood vessels. Histologically, the retinas had a selective loss of rod photoreceptors with initial preservation of cone photoreceptors. Retinal degeneration was not accompanied by any other ocular or central nervous system abnormalities, and pedigree analysis suggested an inherited basis for the disease. Mating an affected Wiltshire ram to 2 affected Wiltshire ewes resulted in 6 progeny that all developed retinal degeneration by 2 years of age, while mating of the same affected ram to 6 unaffected ewes resulted in 8 unaffected progeny, consistent with autosomal recessive inheritance. Homozygosity mapping of 5 affected Wiltshire sheep and 1 unaffected Wiltshire sheep using an OvineSNP50 Genotyping BeadChip revealed an identical-by-descent region on chromosome 5, but none of the genes within this region were considered plausible candidate genes. Whole-genome sequencing of 2 affected sheep did not reveal any significant mutations in any of the genes associated with retinitis pigmentosa in humans or progressive retinal atrophy in dogs. Inherited progressive retinal degeneration affecting rod photoreceptors has not been previously reported in sheep, but this disease has several similarities to inherited retinal dystrophies in other species.


Subject(s)
Night Blindness , Retinal Degeneration , Retinitis Pigmentosa , Sheep Diseases , Animals , Dogs , Female , Male , Night Blindness/genetics , Night Blindness/pathology , Night Blindness/veterinary , Pedigree , Retina/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Degeneration/veterinary , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Retinitis Pigmentosa/veterinary , Sheep , Sheep Diseases/genetics , Sheep Diseases/pathology
2.
J Appl Genet ; 61(2): 179-186, 2020 May.
Article in English | MEDLINE | ID: mdl-32189222

ABSTRACT

Rapid progress in knowledge of the organization of the dog genome has facilitated the identification of the mutations responsible for numerous monogenic diseases, which usually present a breed-specific distribution. The majority of these diseases have clinical and molecular counterparts in humans. The affected dogs have thus become valuable models for preclinical studies of gene therapy for problems such as eye diseases, immunodeficiency, lysosomal storage diseases, hemophilia, and muscular dystrophy. Successful gene therapies in dogs have significantly contributed to decisions to run clinical trials for several human diseases, such as Leber's congenital amaurosis 2-LCA2 (caused by a mutation of RPE65), X-linked retinitis pigmentosa-XLRP (caused by mutation RPGR), and achromatopsia (caused by mutation of CNGB3). Promising results were also obtained for canine as follows: hemophilia (A and B), mucopolysaccharidoses (MPS I, MPS IIIB, MPS VII), leukocyte adhesion deficiency (CLAD), and muscular dystrophy (a counterpart of human Duchenne dystrophy). Present knowledge on molecular background of canine monogenic diseases and their successful gene therapies prove that dogs have an important contribution to preclinical studies.


Subject(s)
Dog Diseases/genetics , Eye Diseases/genetics , Genetic Diseases, Inborn/genetics , Genetic Therapy , Animals , Dog Diseases/therapy , Dogs , Eye Diseases/therapy , Eye Diseases/veterinary , Genetic Diseases, Inborn/therapy , Genetic Diseases, Inborn/veterinary , Genome/genetics , Hemophilia A/genetics , Hemophilia A/therapy , Hemophilia A/veterinary , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/therapy , Lysosomal Storage Diseases/veterinary , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/therapy , Mutation/genetics , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Retinitis Pigmentosa/veterinary
3.
Mol Vis ; 22: 319-31, 2016.
Article in English | MEDLINE | ID: mdl-27122963

ABSTRACT

PURPOSE: Canine X-linked progressive retinal atrophy 1 (XLPRA1) caused by a mutation in retinitis pigmentosa (RP) GTPase regulator (RPGR) exon ORF15 showed significant variability in disease onset in a colony of dogs that all inherited the same mutant X chromosome. Defective protein trafficking has been detected in XLPRA1 before any discernible degeneration of the photoreceptors. We hypothesized that the severity of the photoreceptor degeneration in affected dogs may be associated with defects in genes involved in ciliary trafficking. To this end, we examined six genes as potential disease modifiers. We also examined the expression levels of 24 genes involved in ciliary trafficking (seven), visual pathway (five), neuronal maintenance genes (six), and cellular stress response (six) to evaluate their possible involvement in early stages of the disease. METHODS: Samples from a pedigree derived from a single XLPRA1-affected male dog outcrossed to unrelated healthy mix-bred or purebred females were used for immunohistochemistry (IHC), western blot, mutational and haplotype analysis, and gene expression (GE). Cell-specific markers were used to examine retinal remodeling in the disease. Single nucleotide polymorphisms (SNPs) spanning the entire RPGR interacting and protein trafficking genes (RAB8A, RPGRIP1L, CEP290, CC2D2A, DFNB31, and RAB11B) were genotyped in the pedigree. Quantitative real-time PCR (qRT-PCR) was used to examine the expression of a total of 24 genes, including the six genes listed. RESULTS: Examination of cryosections from XLPRA1-affected animals of similar age (3-4 years) with different disease severity phenotype revealed mislocalization of opsins and upregulation of the Müller cell gliosis marker GFAP. Four to ten haplotypes per gene were identified in RAB8A, RPGRIP1L, CEP290, CC2D2A, DFNB31, and RAB11B for further assessment as potential genetic modifiers of XLPRA1. No correlation was found between the haplotypes and disease severity. During mutational analysis, several new variants, including a single intronic mutation in RAB8A and three mutations in exon 3 of DFNB31 were described (c.970G>A (V324I), c.978T>C (G326=), and c.985G>A (A329T)). Expression analysis of stress response genes in 16-week-old predisease XLPRA1 retinas revealed upregulation of GFAP but not HSPA5, DDIT3, HSPA4, HSP90B1, or HIF1A. Western blot analysis confirmed GFAP upregulation. In the same predisease group, no significant differences were found in the expression of 18 selected genes (RHO, OPN1LW, OPN1MW, RLBP1, RPGRORF15, RAB8A, RPGRIP1L, CEP290, CC2D2A, DFNB31, RAB11B, CRX, RCVRN, PVALB, CALB1, FGFR1, NTRK2, and NTRK3) involved in neuronal function. CONCLUSIONS: Lack of association between haplotypes of RAB8A, RPGRIP1L, CEP290, CC2D2A, DFNB31, and RAB11B and the disease phenotype suggests that these genes are not genetic modifiers of XLPRA1. Upregulation of GFAP, an established indicator of the Müller cell gliosis, manifests as an important early feature of the disease.


Subject(s)
Dog Diseases/genetics , Eye Proteins/genetics , Gene Expression Regulation/physiology , Genetic Diseases, X-Linked/veterinary , Guanine Nucleotide Exchange Factors/genetics , Polymorphism, Single Nucleotide , Retinitis Pigmentosa/veterinary , Animals , Atrophy , Blotting, Western , Dogs , Female , Genetic Association Studies , Genetic Diseases, X-Linked/genetics , Genotyping Techniques , Glial Fibrillary Acidic Protein/genetics , Haplotypes , Male , Molecular Biology , Opsins/genetics , Pedigree , Phenotype , Polymerase Chain Reaction , Retina/pathology , Retinitis Pigmentosa/genetics
4.
Vet Ophthalmol ; 19(5): 398-408, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26417729

ABSTRACT

PURPOSE: To conduct ophthalmic, behavioral, electrophysiological, and genetic testing on two related Gordon setters presented for day blindness and compare findings with those of nine related and unrelated Gordon setters. METHODS: All dogs underwent comprehensive ophthalmic examination. Maze testing was conducted under different light intensities. Rod and cone function was assessed electroretinographically. DNA samples were screened for five canine retinal disease gene mutations. RESULTS: Ophthalmic examination was unremarkable in all dogs. There was no notable difference between day blind dogs and the reference population in scotopic and mesopic maze tests. Day blind dogs performed worse in the photopic maze with slower course completion time and more obstacle collisions. Electroretinography revealed extinguished cone function in day blind dogs and depressed rod responses in all but two reference dogs. One reference population dog presented with day blindness 1 year after initial examination. Mutations that cause achromatopsia (in CNGB3) and cone-rod dystrophies (in ADAM9 and IQCB1) were not detected in any dog tested, although five reference dogs were carriers of the mutation in C2orf71 that causes rod-cone degeneration 4 (rcd4) in Gordon setters and in polski owczarek nizinny dogs. CONCLUSIONS: This report describes a novel retinopathy in related Gordon setters that has clinical signs and vision testing results consistent with achromatopsia but electroretinographic results suggestive of cone-rod dystrophy. The majority of Gordon setters in this study had low rod responses on electroretinography but it is unclear whether this was indicative of rod dysfunction or normal for the breed. Longer-term observation of affected individuals is warranted.


Subject(s)
Color Vision Defects/physiopathology , Color Vision Defects/veterinary , Dog Diseases/genetics , Dog Diseases/physiopathology , Retinal Diseases/veterinary , Animals , Behavior, Animal , Color Vision Defects/genetics , Dogs , Electroretinography/veterinary , Pedigree , Retinal Diseases/genetics , Retinal Diseases/physiopathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/physiopathology , Retinitis Pigmentosa/veterinary , Species Specificity
5.
Pesqui. vet. bras ; 35(2): 153-159, 02/2015. tab, graf
Article in Portuguese | LILACS | ID: lil-748882

ABSTRACT

A tomografia de coerência óptica (OCT) é um exame não invasivo e de não contato que permite avaliar a retina e o nervo óptico. As imagens da OCT fornecem informações da constituição da retina e sua integridade estrutural in vivo, gerando imagens de alta resolução, que se assemelham à microscopia óptica. Objetivou-se descrever a técnica de tomografia de coerência óptica (OCT) e sua utilização em cães. Foi possível diferenciar claramente as camadas retinianas de cães hígidos e compará-las com as de cães portadores de atrofia progressiva de retina, que apresentaram perda da estratificação e diminuição significativa das camadas. No descolamento de retina (DR) foi possível observar a separação entre a retina neurossensorial e o epitélio pigmentário da retina (EPR), além da presença de exsudatos intrarretinianos. Assim, a OCT mostrou-se eficaz no diagnóstico de retinopatias.


The OCT is a noninvasive and noncontact exam capable to evaluate the retina and optic nerve. The OCT images provide information of the constitution of the retina and its structural integrity in vivo, providing high-resolution images that resemble optical microscopy. The objective of this paper was to describe and document the use of the optical coherence tomography (OCT) in dogs. It was possible differentiate the retinal layers of healthy dogs and compare them with dogs with progressive retinal atrophy which showed altered stratification and significant reduce of the layers. In cases of retinal detachment was observed separation of neurosensory retina from the retinal pigment epithelium, and the presence of intrarretinal exudates. Thus, the OCT was effective in the diagnosis of retinopathy.


Subject(s)
Animals , Dogs , Dogs , Retinal Detachment/veterinary , Retinitis Pigmentosa/veterinary , Tomography, Optical/veterinary , Retinitis Pigmentosa/diagnosis
6.
Vet Ophthalmol ; 17(1): 76-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23998563

ABSTRACT

Cone-rod dystrophy is a progressive inherited retinal degenerative disorder that occurs in humans and dogs. The deletion in the nephronophthisis 4 (NPHP4) gene was established as a causative mutation in standard wire-haired Dachshunds. We analyzed all varieties of Dachshunds from the Czech Republic and five other dog breeds and found that the deletion in the NPHP4 (in heterozygous state) is present not only in standard-, but also in miniature wire-haired Dachshunds, but not in other varieties of Dachshunds or in other breeds.


Subject(s)
Dog Diseases/genetics , Gene Deletion , Kidney Diseases, Cystic/genetics , Retinitis Pigmentosa/veterinary , Animals , Dogs , Gene Expression Regulation/physiology , Genetic Predisposition to Disease , Kidney Diseases, Cystic/metabolism , Retinitis Pigmentosa/genetics
7.
PLoS One ; 7(12): e51598, 2012.
Article in English | MEDLINE | ID: mdl-23251588

ABSTRACT

UNLABELLED: Mutations in RPGRIP1 are associated with early onset retinal degenerations in humans and dogs. Dogs homozygous for a 44 bp insertion including a polyA(29) tract potentially leading to premature truncation of the protein, show cone rod degeneration. This is rapid and blinding in a colony of dogs in which the mutation was characterised but in dogs with the same mutation in the pet population there is very variable disease severity and rate of progression. OBJECTIVE: We hypothesized that this variability must be associated with leakiness of the RPGRIP1 mutation, allowing continued RPGRIP1 production. The study was designed to discover mechanisms that might allow such leakiness. METHODS: We analysed alternate start sites and splicing of RPGRIP1 transcripts; variability of polyA(n) length in the insertion and slippage at polyA(n) during transcription/translation. RESULTS AND SIGNIFICANCE: We observed a low rate of use of alternative start codons having potential to allow forms of transcript not including the insertion, with the possibility of encoding truncated functional RPGRIP1 protein isoforms. Complex alternative splicing was observed, but did not increase this potential. Variable polyA(n) length was confirmed in DNA from different RPGRIP1(-/-) dogs, yet polyA(n) variability did not correspond with the clinical phenotypes and no individual was found that carried a polyA(n) tract capable of encoding an in-frame variant. Remarkably though, in luciferase reporter gene assays, out-of-frame inserts still allowed downstream reporter gene expression at some 40% of the efficiency of in-frame controls. This indicates a major role of transcriptional or translational frameshifting in RPGRIP1 expression. The known slippage of reverse transcriptases as well as RNA polymerases and thermostable DNA polymerases on oligoA homopolymers meant that we could not distinguish whether the majority of slippage was transcriptional or translational. This leakiness at the mutation site may allow escape from severe effects of the mutation for some dogs.


Subject(s)
Dog Diseases/genetics , Frameshift Mutation/genetics , Retinitis Pigmentosa/veterinary , Alleles , Animals , Base Sequence , DNA, Complementary/genetics , Dogs , Electrophoresis, Capillary , Exons/genetics , Genes, Reporter , Haplotypes/genetics , Luciferases/metabolism , Molecular Sequence Data , Poly A/genetics , Polymerase Chain Reaction , Protein Biosynthesis/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proteins/genetics , Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinitis Pigmentosa/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
8.
Mamm Genome ; 23(1-2): 212-23, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22193413

ABSTRACT

Cone-rod dystrophy (CRD) is a form of inherited retinal degeneration (RD) causing blindness in man as well as in several breeds of dog. Previously, a 44 bp insertion in RPGRIP1 (retinitis pigmentosa GTPase regulator interacting protein-1) was associated with a recessive early-onset CRD (cone-rod dystrophy 1, cord1) in a Miniature longhaired dachshund (MLHD) research colony. Yet in the MLHD pet population, extensive range of the onset age has been observed among RD cases, with some RPGRIP1(-/-) dogs lacking obvious clinical signs. Phenotypic variation has been known in human homologous diseases, including retinitis pigmentosa and Leber congenital amaurosis, indicating possible involvement of modifiers. To explore additional genetic loci associated with the phenotypic variation observed in MLHDs, a genome-wide association study was carried out using Canine SNP20 arrays in 83 RPGRIP1(-/-) MLHDs with variable ages of onset or no clinical abnormality. Using these samples, comparison of 31 early-onset RD cases against 49 controls (15 late-onset RD and 34 normal dogs combined) identified a strong association (P = 5.05 × 10(-13)) at a single locus on canine chromosome 15. At this locus, the majority of early-onset RD cases but few of the controls were homozygous for a 1.49 Mb interval containing ~11 genes. We conclude that homozygosity at both RPGRIP1 and the newly mapped second locus is necessary to develop early-onset RD, whereas RPGRIP1(-/-) alone leads to late-onset RD or no apparent clinical phenotype. This study establishes a unique model of canine RD requiring homozygous mutations at two distinct genetic loci for the manifestation of early-onset RD.


Subject(s)
Blindness/veterinary , Dog Diseases/genetics , Retinitis Pigmentosa/veterinary , Animals , Animals, Genetically Modified , Blindness/genetics , Dogs , Female , Genome-Wide Association Study/veterinary , Genotype , Homozygote , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/veterinary , Male , Oligonucleotide Array Sequence Analysis , Phenotype , Polymorphism, Single Nucleotide , Proteins/genetics , Retinitis Pigmentosa/genetics , Sequence Analysis, DNA
9.
Vet Ophthalmol ; 14(3): 146-52, 2011 May.
Article in English | MEDLINE | ID: mdl-21521437

ABSTRACT

OBJECTIVE: To investigate ophthalmic and cone-derived electrodiagnostic findings in outbred Miniature Long-haired Dachshunds (MLHD) homozygous for a mutation in the RPGRIP1 gene previously associated with cone-rod dystrophy 1 (cord1). ANIMALS: A total of 36 MLHD homozygous for the RPGRIP1 mutation and 23 dogs clear of the mutation (control group). PROCEDURES: The dogs underwent ophthalmic examination and photopic electroretinogram (ERG) recordings. RESULTS: None of the control dogs presented with clinical or ophthalmic signs consistent with cord1. Amongst the dogs homozygous for the mutation one presented with bilateral symmetrical total retinal atrophy. None of the other dogs in this group showed signs consistent with cord1. Photopic ERG recordings were available in 23 control dogs and 34 dogs homozygous for the mutation. Photopic a- and b-waves following four light stimuli (3 cdS/m(2) ) at a rate of 5.1 Hz were not significantly different between groups. The amplitudes of the 30 Hz flicker (128 flashes, 3 cdS/m(2) ) response were significantly reduced in the dogs homozygous for the PRGRIP1 mutation. The difference in age between the two groups did not significantly affect the difference. CONCLUSION: Homozygosity of the RPGRIP1 mutation does not invariably result in early onset cord1. However, cone derived ERG recordings show evidence of a reduced cone or inner retinal function in homozygous but clinically normal MLHD. Modifying genes that have yet to be identified may influence an individual dog's risk of developing the blinding cord1 and also the age of onset and rate of progression.


Subject(s)
Dog Diseases/genetics , Dogs/genetics , Electroretinography/veterinary , Proteins/metabolism , Retinitis Pigmentosa/veterinary , Animals , Case-Control Studies , Dog Diseases/pathology , Genetic Predisposition to Disease , Genotype , Homozygote , Mutation , Proteins/genetics , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology
10.
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
11.
Invest Ophthalmol Vis Sci ; 51(11): 6038-50, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20574030

ABSTRACT

PURPOSE: To identify genes and molecular mechanisms associated with photoreceptor degeneration in a canine model of XLRP caused by an RPGR exon ORF15 microdeletion. Methods. Expression profiles of mutant and normal retinas were compared by using canine retinal custom cDNA microarrays. qRT-PCR, Western blot analysis, and immunohistochemistry (IHC) were applied to selected genes, to confirm and expand the microarray results. RESULTS: At 7 and 16 weeks, respectively, 56 and 18 transcripts were downregulated in the mutant retinas, but none were differentially expressed (DE) at both ages, suggesting the involvement of temporally distinct pathways. Downregulated genes included the known retina-relevant genes PAX6, CHML, and RDH11 at 7 weeks and CRX and SAG at 16 weeks. Genes directly or indirectly active in apoptotic processes were altered at 7 weeks (CAMK2G, NTRK2, PRKCB, RALA, RBBP6, RNF41, SMYD3, SPP1, and TUBB2C) and 16 weeks (SLC25A5 and NKAP). Furthermore, the DE genes at 7 weeks (ELOVL6, GLOD4, NDUFS4, and REEP1) and 16 weeks (SLC25A5 and TARS2) are related to mitochondrial functions. qRT-PCR of 18 genes confirmed the microarray results and showed DE of additional genes not on the array. Only GFAP was DE at 3 weeks of age. Western blot and IHC analyses also confirmed the high reliability of the transcriptomic data. CONCLUSIONS: Several DE genes were identified in mutant retinas. At 7 weeks, a combination of nonclassic anti- and proapoptosis genes appear to be involved in photoreceptor degeneration, whereas at both 7 and 16 weeks, the expression of mitochondria-related genes indicates that they may play a relevant role in the disease process.


Subject(s)
Dog Diseases/genetics , Eye Proteins/genetics , Frameshift Mutation/genetics , Gene Expression Regulation/physiology , Open Reading Frames/genetics , Retinitis Pigmentosa/veterinary , Animals , Blotting, Western , DNA Mutational Analysis , Disease Models, Animal , Dog Diseases/pathology , Dogs , Exons/genetics , Gene Expression Profiling , Immunohistochemistry , In Situ Hybridization , Oligonucleotide Array Sequence Analysis , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Reverse Transcriptase Polymerase Chain Reaction
12.
Mol Vis ; 15: 2287-305, 2009 Nov 11.
Article in English | MEDLINE | ID: mdl-19936303

ABSTRACT

PURPOSE: Previously, a 44 bp insertion in exon 2 of retinitis pigmentosa GTPase interacting protein 1 (RPGRIP1) was identified as the cause of cone-rod dystrophy 1 (cord1), a recessive form of progressive retinal atrophy (PRA) in the Miniature Longhaired Dachshund (MLHD), a dog model for Leber congenital amaurosis. The cord1 locus was mapped using MLHDs from an inbred colony with a homogeneous early onset disease phenotype. In this paper, the MLHD pet population was studied to investigate phenotypic variation and genotype-phenotype correlation. Further, the cord1 locus was fine-mapped using PRA cases from the MLHD pet population to narrow the critical region. Other dog breeds were also screened for the RGPRIP1 insertion. METHODS: This study examined phenotypic variation in an MLHD pet population that included 59 sporadic PRA cases and 18 members of an extended family with shared environment and having six PRA cases. Ophthalmologic evaluations included behavioral abnormalities, responses to menace and light, fundoscopy, and electroretinography (ERG). The RPGRIP1 insertion was screened for in all cases and 200 apparently normal control MLHDs and in 510 dogs from 66 other breed. To fine-map the cord1 locus in the MLHD, 74 PRA cases and 86 controls aged 4 years or more were genotyped for 24 polymorphic markers within the previously mapped cord1 critical region of 14.15 Mb. RESULTS: Among sporadic PRA cases from the MLHD pet population, the age of onset varied from 4 months to 15 years old; MLHDs from the extended family also showed variable onset and rate of progression. Screening for the insertion in RPGRIP1 identified substantial genotype-phenotype discordance: 16% of controls were homozygous for the insertion (RPGRIP1(-/-)), while 20% of PRA cases were not homozygous for it. Four other breeds were identified to carry the insertion including English Springer Spaniels and Beagles with insertion homozygotes. The former breed included both controls and PRA cases, yet in the latter breed, cone ERG was undetectable in two dogs with no clinically apparent visual dysfunction. Notably, the insertion in the Beagles was a longer variant of that seen in the other breeds. Fine-mapping of the cord1 locus narrowed the critical region on CFA15 from 14.15 Mb to 1.74 Mb which still contains the RPGRIP1 gene. CONCLUSIONS: Extensive phenotypic variations of onset age and progression rate were observed in PRA cases of the MLHD pet population. The insertion in RPGRIP1 showed the strongest association with the disease, yet additional as well as alternative factors may account for the substantial genotype-phenotype discordance.


Subject(s)
Dog Diseases/genetics , Mutation/genetics , Proteins/genetics , Retinitis Pigmentosa/veterinary , Age Distribution , Animals , Animals, Domestic/genetics , Base Pairing/genetics , Breeding , Case-Control Studies , Dog Diseases/pathology , Dogs , Electrophoresis, Capillary , Electroretinography , Female , Fundus Oculi , Genetic Loci/genetics , Genetic Predisposition to Disease , Genotype , Haplotypes/genetics , Male , Mutagenesis, Insertional/genetics , Pedigree , Phenotype , Physical Chromosome Mapping , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology
13.
Invest Ophthalmol Vis Sci ; 50(11): 5355-63, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19643965

ABSTRACT

PURPOSE: The cAMP response element binding protein 1 (CREB1) and activating transcription factor 1 (ATF1) are closely related members of the bZIP superfamily of transcription factors. Both are activated in response to a wide array of stimuli, including cellular stress. This study was conducted to assess the CREB1/ATF1 pathway in photoreceptor disease and protection. METHODS: The expression levels of p-CREB1, CREB1, and ATF1 were examined by immunoblot and immunohistochemistry in normal canine retina and retinas of several canine models of retinal degeneration (rcd1, rcd2, erd, prcd, XLPRA1, XLPRA2, T4R RHO). Humans retinas affected with age-related macular degeneration (AMD) were also examined. p-CREB1/ATF1 immunolabeling was assessed in normal and rcd1 dogs treated with ciliary neurotrophic factor (CNTF), to examine the effect of a neuroprotective stimulus on activation of CREB1/ATF1. RESULTS: Native CREB1 and ATF1 as well as phosphorylated CREB1/ATF1 was examined in normal canine retina by immunoblot. The p-CREB1 antibody identified phosphorylated CREB1 and ATF1 and labeled the inner retina only in normal dogs. In degenerate canine and human retinas, strong immunolabeling appeared in rod and cone photoreceptors, indicating increased expression of native CREB1 and ATF1, as well as increased phosphorylation of these proteins. Retinal protection by CNTF in rcd1 dogs was accompanied by a significant increase in the number of p-CREB1/ATF1-labeled photoreceptor nuclei. CONCLUSIONS: Positive association of CREB1/ATF1 phosphorylation with photoreceptor protection suggests that it may contribute to an innate protective response. These data identify a signaling mechanism in rods and cones of potential importance for therapies of RP and AMD.


Subject(s)
Activating Transcription Factor 1/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Dog Diseases/metabolism , Dog Diseases/prevention & control , Macular Degeneration/metabolism , Retinitis Pigmentosa/prevention & control , Retinitis Pigmentosa/veterinary , Aged , Aged, 80 and over , Animals , Arrestin/metabolism , Cell Count , Ciliary Neurotrophic Factor/therapeutic use , Dog Diseases/pathology , Dogs , Female , Genotype , Humans , Immunoblotting/veterinary , Immunoenzyme Techniques/veterinary , Macular Degeneration/pathology , Male , Phosphorylation , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , Rhodopsin/metabolism
14.
Anim Genet ; 40(4): 572-4, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19392817

ABSTRACT

Cone-rod dystrophy in the standard wire-haired dachshund (SWHD) is inherited as a simple autosomal recessive trait and the recently discovered mutation is widespread within the SWHD population in Norway and other Scandinavian countries. The gene frequency was estimated to be 4.8%. On the basis of the assumption that the size of the ancestral haplotype around a mutation is inversely correlated with the number of generations since the mutation arose, we have found that the mutation is of a relatively recent origin. The conserved haplotype was found to be 8 Mb in size and therefore we estimate that the mutation arose roughly eight generations (approximately 37 years) ago. This indicates that the mutation arose after breed separation.


Subject(s)
Dog Diseases/genetics , Dogs/genetics , Mutation , Retinitis Pigmentosa/veterinary , Alleles , Animals , Dogs/physiology , Gene Frequency , Linkage Disequilibrium , Pedigree , Retinitis Pigmentosa/genetics
15.
Invest Ophthalmol Vis Sci ; 50(8): 3985-95, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19255154

ABSTRACT

PURPOSE: To characterize the retinal histopathology in carriers of X-linked progressive retinal atrophy (XLPRA1 and XLPRA2), two canine models of X-linked retinitis pigmentosa caused, respectively, by a stop and a frameshift mutation in RPGRORF15. METHODS: Retinas of XLPRA2 and XLPRA1 carriers of different ages were processed for morphologic evaluation, TUNEL assay, and immunohistochemistry. Cell-specific markers were used to examine retinal remodeling events. RESULTS: A mosaic pattern composed of patches of diseased and normal retina was first detected in XLPRA2 carriers at 4.9 weeks of age. A peak of photoreceptor cell death led to focal rod loss; however, in these patches an increased density of cones was found to persist over time. Patches of disease gradually disappeared so that by 39 weeks of age the overall retinal morphology, albeit thinner, had improved lamination. In older XLPRA2 carriers (>or=8.8 years), extended regions of severe degeneration occurred in the peripheral/mid-peripheral retina. In XLPRA1 carriers, opsin mislocalization and rare events of rod death were detected by TUNEL assay at 20 weeks of age; however, only patchy degeneration was seen by 1.4 years and was still apparent at 7.8 years. CONCLUSIONS: The time of onset and the progression of the disease differed between the two models. In the early-onset form (XLPRA2) the morphologic appearance of the retinal mosaic changed as a function of age, suggesting that structural plasticity persists in the early postnatal canine retina as mutant photoreceptors die. In the late-onset form (XLPRA1), patches of disease persisted until later ages.


Subject(s)
Aging/physiology , Dog Diseases/pathology , Genetic Diseases, X-Linked/veterinary , Guanine Nucleotide Exchange Factors/genetics , Heterozygote , Photoreceptor Cells, Vertebrate/pathology , Retinitis Pigmentosa/veterinary , Animals , Dog Diseases/genetics , Dogs , Exons/genetics , Female , Fluorescent Antibody Technique, Indirect , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/pathology , In Situ Nick-End Labeling , Male , Mutation , Open Reading Frames/genetics , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology
16.
Invest Ophthalmol Vis Sci ; 50(2): 801-13, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18775863

ABSTRACT

PURPOSE: To characterize a canine model of autosomal recessive RP due to a PDE6A gene mutation. METHODS: Affected and breed- and age-matched control puppies were studied by electroretinography (ERG), light and electron microscopy, immunohistochemistry, and assay for retinal PDE6 levels and enzymatic activity. RESULTS: The mutant puppies failed to develop normal rod-mediated ERG responses and had reduced light-adapted a-wave amplitudes from an early age. The residual ERG waveforms originated primarily from cone-driven responses. Development of photoreceptor outer segments stopped, and rod cells were lost by apoptosis. Immunohistochemistry demonstrated a marked reduction in rod opsin immunostaining outer segments and relative preservation of cones early in the disease process. With exception of rod bipolar cells, which appeared to be reduced in number relatively early in the disease process, other inner retinal cells were preserved in the early stages of the disease, although there was marked and early activation of Müller glia. Western blot analysis showed that the PDE6A mutation not only resulted in a lack of PDE6A protein but the affected retinas also lacked the other PDE6 subunits, suggesting expression of PDE6A is essential for normal expression of PDE6B and PDE6G. Affected retinas lacked PDE6 enzymatic activity. CONCLUSIONS: This represents the first characterization of a PDE6A model of autosomal recessive retinitis pigmentosa, and the PDE6A mutant dog shows promise as a large animal model for investigation of therapies to rescue mutant rod photoreceptors and to preserve cone photoreceptors in the face of a rapid loss of rod cells.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Disease Models, Animal , Dog Diseases/genetics , Genes, Recessive , Point Mutation , Retinitis Pigmentosa/veterinary , Animals , Blotting, Western/veterinary , Breeding , Dog Diseases/physiopathology , Dogs , Electroretinography/veterinary , Female , Immunohistochemistry/veterinary , In Situ Nick-End Labeling , Male , Retina/physiopathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/physiopathology
17.
Genome Res ; 18(9): 1415-21, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18687878

ABSTRACT

Cone-rod dystrophy is a retinal degenerative disorder occurring naturally in man and dog. Here we identify a novel gene for early-onset cone-rod dystrophy in the wire-haired dachshund. For the first time, we use genome-wide association-based Sibling Transmission Disequilibrium Test (sibTDT) analysis of only 13 discordant sib-pairs to identify a single significantly associated 6.5-Mb region (PrawTDT = 4.8 x 10(-5), PgenomeTDT = 6 x 10(-4)) on canine chromosome 5, containing more than 70 genes. Segregation studies using microsatellites in the candidate region including additional meiosis supported the sibTDT analysis but could not further reduce the area. Candidate gene resequencing identified a 180-bp deletion in exon/intron 5 of NPHP4 (nephronophthisis 4, also known as nephroretinin). RT-PCR analysis of NPHP4 in cases and controls showed exon skipping of exon 5, resulting in a truncated protein that retains the binding domain interacting with nephronophthisis 1 (also known as nephrocystin-1) in the kidney but lacks the domain interacting with RPGRIP1 in retina. We suggest that this deletion in the canine NPHP4 gene is the cause of cone-rod dystrophy in the standard wire-haired dachshund. In humans, mutations in NPHP4 have been associated with simultaneous eye and kidney disease. Here we describe the first naturally occurring mutation in NPHP4 without additional kidney disease. Further studies will permit elucidation of the complex molecular mechanism of this retinopathy and the development of potential therapies.


Subject(s)
Dog Diseases/genetics , Gene Deletion , Retinitis Pigmentosa/veterinary , Amino Acid Sequence , Animals , Base Sequence , DNA Mutational Analysis , Dogs , Genes, Recessive , Microsatellite Repeats , Molecular Sequence Data , Mutation , Pedigree , Phenotype , Polymorphism, Single Nucleotide , Proteins/genetics , Retinitis Pigmentosa/genetics , src Homology Domains
18.
Arch Ophthalmol ; 126(5): 714-7, 2008 May.
Article in English | MEDLINE | ID: mdl-18474785

ABSTRACT

A standard operating microscope was modified with a bandpass infrared filter in the light path and infrared image intensifiers for each of the 2 eyepieces. We evaluated this system for subretinal injections in normal control dogs and those with a mutation in the rhodopsin gene. Rhodopsin-mutant dogs are a model for human autosomal dominant retinitis pigmentosa, and their retinas degenerate faster when exposed to modest light levels as used in routine clinical examinations. We showed that the mutant retinas developed severe generalized degeneration when exposed to the standard operating microscope light but not the infrared light. The modified operating microscope provided an excellent view of the ocular fundus under infrared illumination and allowed us to perform subretinal injections in the retinas of the rhodopsin-mutant dogs without any subsequent light-induced retinal degeneration.


Subject(s)
Dog Diseases/surgery , Light/adverse effects , Microscopy/veterinary , Radiation Injuries/veterinary , Retina/radiation effects , Retinitis Pigmentosa/veterinary , Animals , Dog Diseases/genetics , Dogs , Microscopy/instrumentation , Mutation , Ophthalmologic Surgical Procedures/instrumentation , Ophthalmologic Surgical Procedures/veterinary , Radiation Injuries/etiology , Radiation Injuries/prevention & control , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/surgery , Rhodopsin/genetics
19.
Exp Eye Res ; 86(3): 537-40, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18262181

ABSTRACT

Blood flow in the choroid of eyes with retinal degeneration is normally reduced. In the Abyssinian cat, where no changes in choroidal blood flow were observed recently, a closer morphological analysis revealed that two factors might have lead to this finding. (a) The cat's eye consists of a tapetum lucidum where 15.5 microm microspheres cannot reach the precapillary branching in contrast to the non-tapetal areas, where microspheres were located right next to the choriocapillaris. This fact has not yet been described and might be important for further physiological measurements in eyes of species with a tapetum lucidum. (b) The tapetal cells themselves might have a protective role for the choriocapillaris and RPE during retinal degeneration in this specific animal model. Even in late stages of retinal degeneration the RPE and choriocapillaris in areas covered by the tapetum lucidum were not affected whereas non-tapetal areas showed loss of RPE and capillaries.


Subject(s)
Cat Diseases/physiopathology , Choroid/blood supply , Retinitis Pigmentosa/physiopathology , Retinitis Pigmentosa/veterinary , Animals , Cat Diseases/pathology , Cats , Microcirculation , Retinitis Pigmentosa/pathology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...