Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Exp Eye Res ; 235: 109630, 2023 10.
Article in English | MEDLINE | ID: mdl-37625575

ABSTRACT

CRX is a transcription factor essential for normal photoreceptor development and survival. The CRXRdy cat has a naturally occurring truncating mutation in CRX and is a large animal model for dominant Leber congenital amaurosis. This study investigated retinal remodeling that occurs as photoreceptors degenerate. CRXRdy/+ cats from 6 weeks to 10 years of age were investigated. In vivo structural changes of retinas were analyzed by fundus examination, confocal scanning laser ophthalmoscopy and spectral domain optical coherence tomography. Histologic analyses included immunohistochemistry for computational molecular phenotyping with macromolecules and small molecules. Affected cats had a cone-led photoreceptor degeneration starting in the area centralis. Initially there was preservation of inner retinal cells such as bipolar, amacrine and horizontal cells but with time migration of the deafferented neurons occurred. Early in the process of degeneration glial activation occurs ultimately resulting in formation of a glial seal. With progression the macula-equivalent area centralis developed severe atrophy including loss of retinal pigmentary epithelium. Microneuroma formation occured in advanced stages as more marked retinal remodeling occurred. This study indicates that retinal degeneration in the CrxRdy/+ cat retina follows the progressive, phased revision of retina that have been previously described for retinal remodeling. These findings suggest that therapy dependent on targeting inner retinal cells may be useful in young adults with preserved inner retinas prior to advanced stages of retinal remodeling and neuronal cell loss.


Subject(s)
Leber Congenital Amaurosis , Retinal Degeneration , Animals , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/metabolism , Leber Congenital Amaurosis/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism
2.
Transl Vis Sci Technol ; 12(6): 15, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37351895

ABSTRACT

Purpose: Mutations in the CRX transcription factor are associated with dominant retinopathies often with more severe macular changes. The CRX-mutant cat (Rdy-A182d2) is the only animal model with the equivalent of the critical retinal region for high-acuity vision, the macula. Heterozygous cats (CRXRdy/+) have a severe phenotype modeling Leber congenital amaurosis. This study reports the distinct ocular phenotype of homozygous cats (CRXRdy/Rdy). Methods: Gene expression changes were assessed at both mRNA and protein levels. Changes in globe morphology and retinal structure were analyzed. Results: CRXRdy/Rdy cats had high levels of mutant CRX mRNA and protein. The expression of photoreceptor target genes was severely impaired although there were variable effects on the expression of other transcription factors. The photoreceptor cells remained immature and failed to elaborate outer segments consistent with the lack of retinal function. The retinal layers displayed a progressive remodeling with cell loss but maintained overall retinal thickness due to gliosis. Rapid photoreceptor loss largely occurred in the macula-equivalent retinal region. The homozygous cats developed markedly increased ocular globe length. Conclusions: The phenotype of CRXRdy/Rdy cats was more severe compared to CRXRdy/+ cats by several metrics. Translational Relevance: The CRX-mutant cat is the only model for CRX-retinopathies with a macula-equivalent region. A prominent feature of the CRXRdy/Rdy cat phenotype not detectable in homozygous mouse models was the rapid degeneration of the macula-equivalent retinal region highlighting the value of this large animal model and its future importance in the testing of translational therapies aiming to restore vision.


Subject(s)
Retinal Diseases , Trans-Activators , Animals , Mice , Trans-Activators/genetics , Homeodomain Proteins/genetics , Mutation , Phenotype , RNA, Messenger
3.
Mol Ther ; 31(7): 2028-2041, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37056049

ABSTRACT

In this study, we investigate a gene augmentation therapy candidate for the treatment of retinitis pigmentosa (RP) due to cyclic nucleotide-gated channel beta 1 (CNGB1) mutations. We use an adeno-associated virus serotype 5 with transgene under control of a novel short human rhodopsin promoter. The promoter/capsid combination drives efficient expression of a reporter gene (AAV5-RHO-eGFP) exclusively in rod photoreceptors in primate, dog, and mouse following subretinal delivery. The therapeutic vector (AAV5-RHO-CNGB1) delivered to the subretinal space of CNGB1 mutant dogs restores rod-mediated retinal function (electroretinographic responses and vision) for at least 12 months post treatment. Immunohistochemistry shows human CNGB1 is expressed in rod photoreceptors in the treated regions as well as restoration of expression and trafficking of the endogenous alpha subunit of the rod CNG channel required for normal channel formation. The treatment reverses abnormal accumulation of the second messenger, cyclic guanosine monophosphate, which occurs in rod photoreceptors of CNGB1 mutant dogs, confirming formation of a functional CNG channel. In vivo imaging shows long-term preservation of retinal structure. In conclusion, this study establishes the long-term efficacy of subretinal delivery of AAV5-RHO-CNGB1 to rescue the disease phenotype in a canine model of CNGB1-RP, confirming its suitability for future clinical development.


Subject(s)
Parvovirinae , Retinitis Pigmentosa , Humans , Animals , Dogs , Mice , Cyclic Nucleotide-Gated Cation Channels/genetics , Cyclic Nucleotide-Gated Cation Channels/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Retinitis Pigmentosa/metabolism , Retina/metabolism , Electroretinography , Rhodopsin/metabolism
4.
Methods Mol Biol ; 2560: 233-248, 2023.
Article in English | MEDLINE | ID: mdl-36481900

ABSTRACT

Large animal models are valuable for developing and testing translational therapies for inherited retinal dystrophies such as retinitis pigmentosa (RP). Gene augmentation therapy has been developed utilizing such models. Adeno-associated viral (AAV) vectors have been frequently utilized and delivered by intravitreal or subretinal injection. In vivo longitudinal assessments of therapeutic outcomes are essential. These include regular ophthalmic examinations as well as detailed fundus assessments including confocal scanning laser ophthalmoscopy (cSLO) and high-resolution cross-sectional imaging of the retina by spectral domain-optical coherence tomography (SD-OCT). Retinal function assessment includes vision testing and electroretinography (ERG).


Subject(s)
Retinitis Pigmentosa , Animals , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Models, Animal
5.
PLoS One ; 17(12): e0279437, 2022.
Article in English | MEDLINE | ID: mdl-36584140

ABSTRACT

PURPOSE: To investigate whether raised levels of retinal cyclic guanosine monophosphate (cGMP) was reflected in plasma levels in PDE6A-/- dogs. MATERIALS AND METHODS: Retina was collected from 2-month-old wildtype dogs (PDE6A+/+, N = 6), heterozygous dogs (PDE6A+/-, N = 4) and affected dogs (PDE6A-/-, N = 3) and plasma was collected from 2-month-old wildtype dogs (PDE6A+/+, N = 5), heterozygous dogs (PDE6A+/-, N = 5) and affected dogs (PDE6A-/-, N = 5). Retina and plasma samples were measured by ELISA. RESULTS: cGMP levels in retinal samples of PDE6A-/- dogs at 2 months of age were significantly elevated. There was no significant difference in plasma cGMP levels between wildtype and PDE6A-/- or PDE6A+/- puppies. However, the plasma cGMP levels of the PDE6A-/- puppies were significantly lower than that of PDE6A+/- puppies. CONCLUSION: cGMP levels in the plasma from PDE6A-/- was not elevated when compared to control dogs. At the 2-month timepoint, cGMP plasma levels would not be a useful biomarker for disease.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 6 , Retinitis Pigmentosa , Dogs , Animals , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Retina , Cyclic GMP , Heterozygote
6.
Doc Ophthalmol ; 145(3): 237-246, 2022 12.
Article in English | MEDLINE | ID: mdl-36107278

ABSTRACT

PURPOSE: Mutations in the cyclic nucleotide-gated (CNG) channel beta subunit (CNGB1) are an important cause of recessive retinitis pigmentosa. We identified a large animal model with a truncating mutation of CNGB1. This study reports the persistence of small, desensitized rod ERG responses in this model. METHODS: Dark-, light-adapted and chromatic ERGs were recorded in CNGB1 mutant dogs and age and breed matched controls. Comparisons were made with a dog model known to completely lack rod function; young dogs with a mutation in the rod phosphodiesterase 6 alpha subunit (PDE6A-/-). Immunohistochemistry (IHC) to label the rod CNG alpha (CNGA1) and CNGB1 subunits was performed. RESULTS: The dark-adapted ERG of CNGB1 mutant dogs had a raised response threshold with lack of normal rod response and a remaining cone response. Increasing stimulus strength resulted in the appearance of a separate, slower positive waveform following the dark-adapted cone b-wave. With increasing stimulus strength this increased in amplitude and became faster to merge with the initial b-wave. Comparison of responses from PDE6A-/- (cone only dogs) with CNGB1 mutant dogs to red and blue flashes and between dark-adapted and light-adapted responses supported the hypothesis that the CNGB1 mutant dog had residual desensitized rod responses. CNGB1 mutant dogs had a small amount of CNGA1 detectable in the outer segments. CONCLUSIONS: CNGB1 mutant dogs have a residual ERG response from desensitized rods. This may be due to low levels of CNGA1 in outer segments.


Subject(s)
Electroretinography , Retinitis Pigmentosa , Dogs , Animals , Cyclic Nucleotide-Gated Cation Channels/genetics , Retinitis Pigmentosa/genetics , Retinal Cone Photoreceptor Cells , Disease Models, Animal
7.
Vet Ophthalmol ; 25(5): 385-397, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35713167

ABSTRACT

OBJECTIVES: To assess an inherited abnormal negative response electroretinogram (NRE) that originated in a family of Papillon dogs. ANIMALS STUDIED: Thirty-eight dogs (Papillons, or Papillon cross Beagles or Beagles). PROCEDURES: Dogs underwent routine ophthalmic examination and a detailed dark-adapted, light-adapted and On-Off electroretinographic study. Vision was assessed using a four-choice exit device. Spectral-domain optical coherence tomography (SD-OCT) was performed on a subset of dogs. Two affected males were outcrossed to investigate the mode of inheritance of the phenotype. RESULTS: The affected dogs had an increased underlying negative component to the ERG. This was most pronounced in the light-adapted ERG, resulting in a reduced b-wave and an exaggerated photopic negative response (PhNR). Changes were more pronounced with stronger flashes. Similarly, the On-response of the On-Off ERG had a reduced b-wave and a large post-b-wave negative component. The dark-adapted ERG had a significant increase in the scotopic threshold response (STR) and a significant reduction in the b:a-wave ratio. Significant changes could be detected at 2 months of age but became more pronounced with age. Vision testing using a four-choice device showed affected dogs had reduced visual performance under the brightest light condition. There was no evidence of a degenerative process in the affected dogs up to 8.5 years of age. Test breeding results suggested the NRE phenotype had an autosomal dominant mode of inheritance. CONCLUSIONS: We describe an inherited ERG phenotype in Papillon dogs characterized by an underlying negative component affecting both dark- and light-adapted ERG responses.


Subject(s)
Dog Diseases , Retinal Diseases , Animals , Dog Diseases/genetics , Dogs , Electroretinography/methods , Electroretinography/veterinary , Male , Retina/physiology , Retinal Diseases/veterinary , Tomography, Optical Coherence/veterinary
8.
BMC Vet Res ; 18(1): 182, 2022 May 16.
Article in English | MEDLINE | ID: mdl-35578341

ABSTRACT

BACKGROUND: A number of etiologies for different canine chorioretinal lesions have been proved or suggested but some fundic lesions remain unclear in terms of an etiologic diagnosis, treatment options and prognosis. The purpose of this case series is to describe atypical chorioretinal lesions observed in dogs with primary angle-closure glaucoma (PACG). CASE PRESENTATION: Two spayed-female Siberian Huskies (3- and 4-year-old) and one Siberian Husky/Australian Shepherd mixed breed dog (11-month-old) that had multifocal depigmented retinal lesions and PACG were included. PROCEDURES: Ophthalmic examination, gross, and histopathologic examination findings are described. One of the dogs underwent further clinical diagnostics. Advanced clinical diagnostics on the fellow, presumed to be non-glaucomatous eye of a dog revealed: pectinate ligament dysplasia by gonioscopy, retinal thinning in the depigmented area and wedge shaped retinal thinning with delayed choroidal vascular perfusion by optical coherence tomography, confocal scanning laser ophthalmoscopy, fluorescein and indocyanine green angiography. Quantifiable maze testing for the same eye revealed mild nyctalopia but the full-field electroretinogram showed no generalized decrease of retinal function. Genetic testing for mutations within the retinitis pigmentosa GTPase regulator gene causing X-linked progressive retinal atrophy in Siberian Huskies was negative. Histopathologic evaluations on enucleated eyes in two dogs confirmed goniodysgenesis, PACG with optic nerve head cupping, and diffuse inner retinal atrophy. In addition, segmental profound retinal atrophy, loss of retinal pigment epithelium, and adhesion of the retina to Bruch's membrane was observed and coincided with multifocal depigmented lesions noted on fundic examination. CONCLUSIONS: To our knowledge, this is the first case series with clinical and histopathologic data of chorioretinal lesions, most likely caused by severely impaired choroidal perfusion. Further studies are warranted to elucidate the etiology and pathophysiology, including its possible association with PACG.


Subject(s)
Dog Diseases , Glaucoma, Angle-Closure , Optic Disk , Animals , Atrophy/complications , Atrophy/pathology , Atrophy/veterinary , Australia , Choroid/pathology , Dog Diseases/diagnosis , Dog Diseases/genetics , Dog Diseases/pathology , Dogs , Female , Glaucoma, Angle-Closure/diagnosis , Glaucoma, Angle-Closure/genetics , Glaucoma, Angle-Closure/veterinary , Optic Disk/pathology
9.
Doc Ophthalmol ; 144(2): 81-97, 2022 04.
Article in English | MEDLINE | ID: mdl-35247111

ABSTRACT

PURPOSE: In this study, we assessed several extended electroretinographic protocols using nonstandard stimuli. Our aim was to separate and quantify the contributions of different populations of retinal cells to the overall response, both to assess normal function and characterize dogs with inherited retinal disease. METHODS: We investigated three different protocols for measuring the full-field flash electroretinogram-(1) chromatic dark-adapted red and blue flashes, (2) increasing luminance blue-background, (3) flicker with fixed frequency and increasing luminance, and flicker with increasing frequency at a fixed luminance-to assess rod and cone contributions to electroretinograms recorded in phenotypically normal control dogs and dogs lacking rod function. RESULTS: Temporal separation of the rod- and cone-driven responses is possible in the fully dark-adapted eye using dim red flashes. A- and b-wave amplitudes decrease at different rates with increasing background luminance in control dogs. Flicker responses elicited with extended flicker protocols are well fit with mathematical models in control dogs. Dogs lacking rod function demonstrated larger amplitude dark-adapted compared to light-adapted flicker responses. CONCLUSIONS: Using extended protocols of the full-field electroretinogram provides additional characterization of the health and function of different populations of cells in the normal retina and enables quantifiable comparison between phenotypically normal dogs and those with retinal disease.


Subject(s)
Electroretinography , Retinal Diseases , Animals , Dark Adaptation , Dogs , Electroretinography/methods , Photic Stimulation , Retinal Cone Photoreceptor Cells/physiology
10.
Vet Ophthalmol ; 25 Suppl 1: 164-178, 2022 May.
Article in English | MEDLINE | ID: mdl-35156737

ABSTRACT

PURPOSE: While the retinal vasculature can be assessed by simple funduscopy, a more detailed assessment can be performed by conventional angiography using dyes such as fluorescein or indocyanine green. The development of optical coherence tomography angiography (OCT-A) allows a non-invasive detailed examination of posterior segment vasculature. The purpose of this prospective study was to compare imaging of posterior segment vasculature in normal dogs and cats using OCT-A, fluorescein angiography (FA), and indocyanine green angiography (ICGA). METHODS: Eight adult funduscopically normal dogs and 13 funduscopically normal cats were included in the study. Retinal vasculature was assessed by OCT-A followed by ICGA then FA. Regular fundus imaging was also performed. RESULTS: High-resolution images of the different vascular layers within the retina and choroid could be acquired using OCT-A in both dogs and cats. The technique provided more detail than obtained with FA/ICGA. However, artifacts/errors can occur during OCT-A image acquisition/analysis/interpretation and must be considered. Furthermore, OCT-A only allows for a limited field of view compared to FA/ICGA. CONCLUSIONS: Optical coherence tomography angiography is a new non-invasive posterior segment imaging technique that is complementary to traditional dye-based angiographic techniques. Detailed imaging of the dog and cat posterior segment can be achieved under general anesthesia. OCT-A provides additional detail of the vasculature and can clearly demonstrate the anatomical depth of the imaged vessels. There are, however, some limitations to this new technique that may be overcome by future technological advances.


Subject(s)
Cat Diseases , Dog Diseases , Animals , Cats , Choroid/blood supply , Coloring Agents , Dog Diseases/diagnostic imaging , Dogs , Fluorescein , Fluorescein Angiography/methods , Fluorescein Angiography/veterinary , Indocyanine Green , Prospective Studies , Tomography, Optical Coherence/methods , Tomography, Optical Coherence/veterinary
11.
Vet Ophthalmol ; 25(2): 109-117, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34708922

ABSTRACT

OBJECTIVE: To report the development of focal bullous retinal detachments (bullae) in dogs with different forms of progressive retinal atrophy (PRA). PROCEDURES: Dogs with three distinct forms of PRA (PRA-affected Whippets, German Spitzes and CNGB1-mutant Papillon crosses) were examined by indirect ophthalmoscopy and spectral domain optical coherence tomography (SD-OCT). Retinal bullae were monitored over time. One CNGB1-mutant dog was treated with gene augmentation therapy. The canine BEST1 gene coding region and flanking intronic sequence was sequenced in at least one affected dog of each breed. RESULTS: Multiple focal bullous retinal detachments (bullae) were identified in PRA-affected dogs of all three types. They developed in 4 of 5 PRA-affected Whippets, 3 of 8 PRA-affected Germans Spitzes and 15 of 20 CNGB1-mutant dogs. The bullae appeared prior to marked retinal degeneration and became less apparent as retinal degeneration progressed. Bullae were not seen in any heterozygous animals of any of the types of PRA. Screening of the coding region and flanking intronic regions of the canine BEST1 gene failed to reveal any associated pathogenic variants. Retinal gene augmentation therapy in one of the CNGB1-mutant dogs appeared to prevent formation of bullae. CONCLUSIONS: Retinal bullae were identified in dogs with three distinct forms of progressive retinal atrophy. The lesions develop prior to retinal thinning. This clinical change should be monitored for in dogs with PRA.


Subject(s)
Dog Diseases , Retinal Degeneration , Animals , Atrophy/pathology , Atrophy/veterinary , Blister/pathology , Blister/veterinary , Dog Diseases/genetics , Dog Diseases/pathology , Dogs , Retina/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Degeneration/veterinary
12.
Hum Mol Genet ; 31(8): 1263-1277, 2022 04 22.
Article in English | MEDLINE | ID: mdl-34726233

ABSTRACT

Pathogenic variants in retinol dehydrogenase 5 (RDH5) attenuate supply of 11-cis-retinal to photoreceptors leading to a range of clinical phenotypes including night blindness because of markedly slowed rod dark adaptation and in some patients, macular atrophy. Current animal models (such as Rdh5-/- mice) fail to recapitulate the functional or degenerative phenotype. Addressing this need for a relevant animal model we present a new domestic cat model with a loss-of-function missense mutation in RDH5 (c.542G > T; p.Gly181Val). As with patients, affected cats have a marked delay in recovery of dark adaptation. In addition, the cats develop a degeneration of the area centralis (equivalent to the human macula). This recapitulates the development of macular atrophy that is reported in a subset of patients with RDH5 mutations and is shown in this paper in seven patients with biallelic RDH5 mutations. There is notable variability in the age at onset of the area centralis changes in the cat, with most developing changes as juveniles but some not showing changes over the first few years of age. There is similar variability in development of macular atrophy in patients and while age is a risk factor, it is hypothesized that genetic modifying loci influence disease severity, and we suspect the same is true in the cat model. This novel cat model provides opportunities to improve molecular understanding of macular atrophy and test therapeutic interventions for RDH5-associated retinopathies.


Subject(s)
Macular Degeneration , Retinal Diseases , Alcohol Oxidoreductases/genetics , Animals , Atrophy , Cats , Electroretinography , Humans , Mice , Models, Animal , Phenotype , Retinal Diseases/genetics
13.
BMC Vet Res ; 17(1): 366, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34847929

ABSTRACT

BACKGROUND: Despite the common use of topical ophthalmic corticosteroids in dogs, detailed reports on systemic and dermatologic adverse effects are limited. RESULTS: Nine purpose-bred research Beagles were treated with difluprednate 0.05% ophthalmic emulsion in one or both eyes 2-3 times daily. Some difluprednate treated dogs developed mild to severe alopecia of the periocular region, face, and distal pinna (5/9). The median duration of treatment prior to onset of dermatologic signs for difluprednate treated dogs was 550 days (453-1160 days). Diagnostic testing included complete blood count (CBC) and serum biochemistry, adrenocorticotropic hormone (ACTH) stimulation testing combined with endogenous ACTH measurement, and skin biopsy. The CBC and chemistry were within normal limits for all dogs. There were varying degrees of suppression of the hypothalamic-pituitary-adrenocortical (HPA) axis with difluprednate treatment. Dogs with the most profound alopecic changes had less pronounced HPA axis suppression compared to dogs with no integumentary changes. Skin biopsies demonstrated follicular atrophy and follicular keratosis. When topical difluprednate was reduced to unilateral therapy, the hair regrew on the untreated side of the face. In addition to the affected research dogs, a 7-year old female spayed Chihuahua that was being treated as a clinical patient with long-term difluprednate 0.05% ophthalmic emulsion developed generalized hypotrichosis on the head and body and a potbellied appearance. ACTH stimulation testing revealed suppression of the HPA axis with a mild increase in serum alkaline phosphatase (ALP) activity and a urine specific gravity of 1.016. The combination of clinical signs and laboratory abnormalities was supportive of iatrogenic hyperadrenocorticism. CONCLUSIONS: In dogs long-term use of difluprednate ophthalmic emulsion results in HPA axis suppression and in some cases iatrogenic hyperadrenocorticism. A novel pattern of localized alopecia is suspected to be related to dermal absorption and local action due to superior potency and penetration compared to other commonly utilized ophthalmic corticosteroids.


Subject(s)
Alopecia , Dog Diseases , Fluprednisolone/analogs & derivatives , Hypothalamo-Hypophyseal System , Pituitary-Adrenal System , Adrenocorticotropic Hormone/therapeutic use , Alopecia/chemically induced , Alopecia/drug therapy , Alopecia/veterinary , Animals , Cushing Syndrome/veterinary , Dog Diseases/chemically induced , Dog Diseases/drug therapy , Dogs , Emulsions , Female , Fluprednisolone/therapeutic use
14.
Int J Mol Sci ; 22(22)2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34830323

ABSTRACT

Sporadic occurrence of inherited eye disorders has been reported in cattle but so far pathogenic variants were found only for rare forms of cataract but not for retinopathies. The aim of this study was to characterize the phenotype and the genetic aetiology of a recessive form of congenital day-blindness observed in several cases of purebred Original Braunvieh cattle. Electroretinography in an affected calf revealed absent cone-mediated function, whereas the rods continue to function normally. Brain areas involved in vision were morphologically normal. When targeting cones by immunofluorescence, a decrease in cone number and an accumulation of beta subunits of cone cyclic-nucleotide gated channel (CNGB3) in the outer plexiform layer of affected animals was obvious. Achromatopsia is a monogenic Mendelian disease characterized by the loss of cone photoreceptor function resulting in day-blindness, total color-blindness, and decreased central visual acuity. After SNP genotyping and subsequent homozygosity mapping with twelve affected cattle, we performed whole-genome sequencing and variant calling of three cases. We identified a single missense variant in the bovine CNGB3 gene situated in a ~2.5 Mb homozygous genome region on chromosome 14 shared between all cases. All affected cattle were homozygous carriers of the p.Asp251Asn mutation that was predicted to be deleterious, affecting an evolutionary conserved residue. In conclusion, we have evidence for the occurrence of a breed-specific novel CNGB3-related form of recessively inherited achromatopsia in Original Braunvieh cattle which we have designated OH1 showing an allele frequency of the deleterious allele of ~8%. The identification of carriers will enable selection against this inherited disorder. The studied cattle might serve as an animal model to further elucidate the function of CNGB3 in mammals.


Subject(s)
Alleles , Color Vision Defects/genetics , Cyclic Nucleotide-Gated Cation Channels/genetics , Mutation, Missense , Protein Subunits/genetics , Retinal Cone Photoreceptor Cells/metabolism , Amino Acid Substitution , Animals , Asparagine/metabolism , Aspartic Acid/metabolism , Cattle , Color Vision Defects/diagnostic imaging , Color Vision Defects/metabolism , Color Vision Defects/pathology , Cyclic Nucleotide-Gated Cation Channels/deficiency , Electroretinography , Female , Gene Expression , Gene Frequency , Homozygote , Male , Phenotype , Protein Subunits/deficiency , Retinal Cone Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/metabolism , Whole Genome Sequencing
15.
J Vis Exp ; (174)2021 08 05.
Article in English | MEDLINE | ID: mdl-34424232

ABSTRACT

Retinal degenerative (RD) conditions associated with photoreceptor loss such as age-related macular degeneration (AMD), retinitis pigmentosa (RP) and Leber Congenital Amaurosis (LCA) cause progressive and debilitating vision loss. There is an unmet need for therapies that can restore vision once photoreceptors have been lost. Transplantation of human pluripotent stem cell (hPSC)-derived retinal tissue (organoids) into the subretinal space of an eye with advanced RD brings retinal tissue sheets with thousands of healthy mutation-free photoreceptors and has a potential to treat most/all blinding diseases associated with photoreceptor degeneration with one approved protocol. Transplantation of fetal retinal tissue into the subretinal space of animal models and people with advanced RD has been developed successfully but cannot be used as a routine therapy due to ethical concerns and limited tissue supply. Large eye inherited retinal degeneration (IRD) animal models are valuable for developing vision restoration therapies utilizing advanced surgical approaches to transplant retinal cells/tissue into the subretinal space. The similarities in globe size, and photoreceptor distribution (e.g., presence of macula-like region area centralis) and availability of IRD models closely recapitulating human IRD would facilitate rapid translation of a promising therapy to the clinic. Presented here is a surgical technique of transplanting hPSC-derived retinal tissue into the subretinal space of a large animal model allowing assessment of this promising approach in animal models.


Subject(s)
Human Embryonic Stem Cells , Pluripotent Stem Cells , Retinal Degeneration , Animals , Cats , Disease Models, Animal , Humans , Retina , Stem Cell Transplantation
16.
Doc Ophthalmol ; 143(2): 171-184, 2021 10.
Article in English | MEDLINE | ID: mdl-33818677

ABSTRACT

PURPOSE: To analyze ERG responses from two dog models of retinitis pigmentosa, one due to a PDE6A mutation and the other a CNGB1 mutation, both to assess the effect of these mutations on retinal function and the ability of gene augmentation therapy to restore normal function. METHODS: Scotopic and photopic ERGs from young affected and normal control dogs and affected dogs following AAV-mediated gene augmentation therapy were analyzed. Parameters reflecting rod and cone function were collected by modeling the descending slope of the a-wave to measure receptor response and sensitivity. Rod-driven responses were further assessed by Naka-Rushton fitting of the first limb of the scotopic b-wave luminance-response plot. RESULTS: PDE6A-/- dogs showed a dramatic decrease in rod-driven responses with very reduced rod maximal responses and sensitivity. There was a minor reduction in the amplitude of maximal cone responses. In contrast, CNGB1-/- dogs had some residual rod responses with reduced amplitude and sensitivity and normal cone responses. Following gene augmentation therapy, rod parameters were substantially improved in both models with restoration of sensitivity parameters log S and log K and a large increase in log Rmax in keeping with rescue of normal rod phototransduction in the treated retinal regions. CONCLUSIONS: Modeling of rod and cone a-waves and the luminance-response function of the scotopic b-wave characterized the loss of rod photoreceptor function in two dog models of retinitis pigmentosa and showed the effectiveness of gene augmentation therapy in restoring normal functional parameters.


Subject(s)
Color Vision , Retinitis Pigmentosa , Animals , Dogs , Electroretinography , Retina , Retinal Cone Photoreceptor Cells , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy
17.
EMBO Mol Med ; 13(4): e13392, 2021 04 09.
Article in English | MEDLINE | ID: mdl-33616280

ABSTRACT

Gene therapy using recombinant adeno-associated virus (rAAV) vectors to treat blinding retinal dystrophies has become clinical reality. Therapeutically impactful targeting of photoreceptors still relies on subretinal vector delivery, which detaches the retina and harbours substantial risks of collateral damage, often without achieving widespread photoreceptor transduction. Herein, we report the development of novel engineered rAAV vectors that enable efficient targeting of photoreceptors via less invasive intravitreal administration. A unique in vivo selection procedure was performed, where an AAV2-based peptide-display library was intravenously administered in mice, followed by isolation of vector DNA from target cells after only 24 h. This stringent selection yielded novel vectors, termed AAV2.GL and AAV2.NN, which mediate widespread and high-level retinal transduction after intravitreal injection in mice, dogs and non-human primates. Importantly, both vectors efficiently transduce photoreceptors in human retinal explant cultures. As proof-of-concept, intravitreal Cnga3 delivery using AAV2.GL lead to cone-specific expression of Cnga3 protein and rescued photopic cone responses in the Cnga3-/- mouse model of achromatopsia. These novel rAAV vectors expand the clinical applicability of gene therapy for blinding human retinal dystrophies.


Subject(s)
Color Vision Defects , Dependovirus , Animals , Capsid , Color Vision Defects/therapy , Dependovirus/genetics , Dogs , Genetic Therapy , Genetic Vectors , Mice , Retina
18.
Cells ; 10(1)2021 01 09.
Article in English | MEDLINE | ID: mdl-33435495

ABSTRACT

The Rpe65-deficient dog has been important for development of translational therapies of Leber congenital amaurosis type 2 (LCA2). The purpose of this study was to provide a comprehensive report of the natural history of retinal changes in this dog model. Rpe65-deficient dogs from 2 months to 10 years of age were assessed by fundus imaging, electroretinography (ERG) and vision testing (VT). Changes in retinal layer thickness were assessed by optical coherence tomography and on plastic retinal sections. ERG showed marked loss of retinal sensitivity, with amplitudes declining with age. Retinal thinning initially developed in the area centralis, with a slower thinning of the outer retina in other areas starting with the inferior retina. VT showed that dogs of all ages performed well in bright light, while at lower light levels they were blind. Retinal pigment epithelial (RPE) inclusions developed and in younger dogs and increased in size with age. The loss of photoreceptors was mirrored by a decline in ERG amplitudes. The slow degeneration meant that sufficient photoreceptors, albeit very desensitized, remained to allow for residual bright light vision in older dogs. This study shows the natural history of the Rpe65-deficient dog model of LCA2.


Subject(s)
Retina/enzymology , Retina/pathology , cis-trans-Isomerases/deficiency , Adaptation, Ocular/radiation effects , Aging/pathology , Animals , Dogs , Electroretinography , Fundus Oculi , Light , Phenotype , Retina/diagnostic imaging , Retina/physiopathology , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/physiopathology , Tomography, Optical Coherence , Vision, Ocular , cis-trans-Isomerases/metabolism
19.
BMC Vet Res ; 16(1): 225, 2020 Jun 30.
Article in English | MEDLINE | ID: mdl-32605619

ABSTRACT

BACKGROUND: Retinal diseases are common in dogs. Some hereditary retinal dystrophies in dogs are important not only because they lead to vision loss but also because they show strong similarities to the orthologous human conditions. Advances in in vivo non-invasive retinal imaging allow the capture of retinal cross-section images that parallel low power microscopic examination of histological sections. Spectral domain - optical coherence tomography (SD-OCT) allows the measurement of retinal layer thicknesses and gives the opportunity for repeat examination to investigate changes in thicknesses in health (such as changes with maturation and age) and disease (following the course of retinal degenerative conditions). The purpose of this study was to use SD-OCT to measure retinal layer thicknesses in the dog during retinal maturation and over the first year of life. SD-OCT was performed on normal beagle cross dogs from 4 weeks of age to 52 weeks of age. To assess changes in layer thickness with age, measurements were taken from fixed regions in each of the 4 quadrants and the area centralis (the region important for most detailed vision). Additionally, changes in retinal layer thickness along vertical and horizontal planes passing through the optic nerve head were assessed. RESULTS: In the four quadrants an initial thinning of retinal layers occurred over the first 12 to 15 weeks of life after which there was little change in thickness. However, in the area centralis there was a thickening of the photoreceptor layer over this time period which was mostly due to a lengthening of the photoreceptor inner/outer segment layer. The retina thinned with greater distances from the optic nerve head in both vertical and horizontal planes with the dorsal retina being thicker than the ventral retina. Most of the change in thickness with distance from the optic nerve head was due to difference in thickness of the inner retinal layers. The outer retinal layers remained more constant in thickness, particularly in the horizontal plane and dorsal to the optic nerve head. CONCLUSIONS: These measurements will provide normative data for future studies.


Subject(s)
Dogs/anatomy & histology , Retina/diagnostic imaging , Tomography, Optical Coherence/veterinary , Animals , Female , Male , Retina/growth & development , Tomography, Optical Coherence/methods
20.
Am J Hum Genet ; 106(6): 893-904, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32386558

ABSTRACT

Kinesin-2 enables ciliary assembly and maintenance as an anterograde intraflagellar transport (IFT) motor. Molecular motor activity is driven by a heterotrimeric complex comprised of KIF3A and KIF3B or KIF3C plus one non-motor subunit, KIFAP3. Using exome sequencing, we identified heterozygous KIF3B variants in two unrelated families with hallmark ciliopathy phenotypes. In the first family, the proband presents with hepatic fibrosis, retinitis pigmentosa, and postaxial polydactyly; he harbors a de novo c.748G>C (p.Glu250Gln) variant affecting the kinesin motor domain encoded by KIF3B. The second family is a six-generation pedigree affected predominantly by retinitis pigmentosa. Affected individuals carry a heterozygous c.1568T>C (p.Leu523Pro) KIF3B variant segregating in an autosomal-dominant pattern. We observed a significant increase in primary cilia length in vitro in the context of either of the two mutations while variant KIF3B proteins retained stability indistinguishable from wild type. Furthermore, we tested the effects of KIF3B mutant mRNA expression in the developing zebrafish retina. In the presence of either missense variant, rhodopsin was sequestered to the photoreceptor rod inner segment layer with a concomitant increase in photoreceptor cilia length. Notably, impaired rhodopsin trafficking is also characteristic of recessive KIF3B models as exemplified by an early-onset, autosomal-recessive, progressive retinal degeneration in Bengal cats; we identified a c.1000G>A (p.Ala334Thr) KIF3B variant by genome-wide association study and whole-genome sequencing. Together, our genetic, cell-based, and in vivo modeling data delineate an autosomal-dominant syndromic retinal ciliopathy in humans and suggest that multiple KIF3B pathomechanisms can impair kinesin-driven ciliary transport in the photoreceptor.


Subject(s)
Ciliopathies/genetics , Ciliopathies/pathology , Genes, Dominant/genetics , Kinesins/genetics , Mutation , Retina/pathology , Amino Acid Sequence , Animals , Cats , Child, Preschool , Cilia/pathology , Female , Genome-Wide Association Study , Heterozygote , Humans , Kinesins/chemistry , Kinesins/metabolism , Larva , Male , Middle Aged , Pedigree , Phenotype , Photoreceptor Cells/metabolism , Retina/cytology , Retina/growth & development , Retina/metabolism , Rhodopsin/metabolism , Young Adult , Zebrafish/genetics , Zebrafish/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...