33 results on '"Keiko Miyadera"'
Search Results
2. Recent advances in the understanding of cilia mechanisms and their applications as therapeutic targets
- Author
-
Masaki Saito, Wataru Otsu, Keiko Miyadera, and Yuhei Nishimura
- Subjects
ciliary receptors ,ciliogenesis ,ciliary transport ,ciliopathy ,inherited retinal dystrophies ,photoreceptor ,Biology (General) ,QH301-705.5 - Abstract
The primary cilium is a single immotile microtubule-based organelle that protrudes into the extracellular space. Malformations and dysfunctions of the cilia have been associated with various forms of syndromic and non-syndromic diseases, termed ciliopathies. The primary cilium is therefore gaining attention due to its potential as a therapeutic target. In this review, we examine ciliary receptors, ciliogenesis, and ciliary trafficking as possible therapeutic targets. We first discuss the mechanisms of selective distribution, signal transduction, and physiological roles of ciliary receptors. Next, pathways that regulate ciliogenesis, specifically the Aurora A kinase, mammalian target of rapamycin, and ubiquitin-proteasome pathways are examined as therapeutic targets to regulate ciliogenesis. Then, in the photoreceptors, the mechanism of ciliary trafficking which takes place at the transition zone involving the ciliary membrane proteins is reviewed. Finally, some of the current therapeutic advancements highlighting the role of large animal models of photoreceptor ciliopathy are discussed.
- Published
- 2023
- Full Text
- View/download PDF
3. Molecular characterization of MAP9 in the photoreceptor sensory cilia as a modifier in canine RPGRIP1-associated cone-rod dystrophy
- Author
-
Kei Takahashi, Jennifer C. Kwok, Yu Sato, Gustavo D. Aguirre, and Keiko Miyadera
- Subjects
animal model ,canine ,inherited retinal diseases ,MAP9 ,microtubule-binding protein ,photoreceptor ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Photoreceptors possess a highly specialized primary cilium containing expanded ciliary membrane discs called the outer segment. The photoreceptor cilium is essential for the maintenance of the outer segment, and pathogenic variants in more than 50 cilia-related genes have been identified as causing non-syndromic inherited retinal diseases in patients. The retinitis pigmentosa GTPase regulator interacting protein 1 (RPGRIP1) is a structural protein localized to the photoreceptor cilium and biallelic RPGRIP1 variants have been associated with non-syndromic human inherited retinal diseases. In a canine cone-rod dystrophy model, a naturally occurring 44-bp exonic insertion in RPGRIP1 (RPGRIP1ins44/ins44) is the primary disease locus while an additional homozygous variant in MAP9 (microtubule associated protein 9) (MAP9aff/aff) acts as a modifier associated with early disease onset. MAP9 was originally identified as a microtubule-binding protein stabilizing microtubule structure during both mitosis and interphase in human cell lines. However, the roles of MAP9 in primary cilia, including photoreceptor neurosensory cilia, have not been well understood. Hence, we characterized the pathogenic phenotypes associated with homozygous MAP9 variant, and investigated the molecular function of MAP9 in primary cilia using the RPGRIP1-associated oligogenic canine cone-rod dystrophy model as well as cultured cells. Both functionally and structurally, the RPGRIP1ins44/ins44MAP9aff/aff retina exhibited progressive cone photoreceptor degeneration starting earlier than the retina affected by RPGRIP1ins44/ins44 alone. Based on immunostaining of canine retinal sections and cultured cells, we found that MAP9 is prominently localized in the basal body of primary cilia and played an important role in maintaining the structure of ciliary microtubule axoneme. These findings suggest that the affected MAP9, together with mutant RPGRIP1, is deprived of critical roles in cilia organization and maintenance resulting in altered cilia structure and function giving rise to early onset and accelerated disease progression in the RPGRIP1ins44/ins44MAP9aff/aff double homozygote cone-rod dystrophy canine model.
- Published
- 2023
- Full Text
- View/download PDF
4. Canine CNGA3 Gene Mutations Provide Novel Insights into Human Achromatopsia-Associated Channelopathies and Treatment.
- Author
-
Naoto Tanaka, Emily V Dutrow, Keiko Miyadera, Lucie Delemotte, Christopher M MacDermaid, Shelby L Reinstein, William R Crumley, Christopher J Dixon, Margret L Casal, Michael L Klein, Gustavo D Aguirre, Jacqueline C Tanaka, and Karina E Guziewicz
- Subjects
Medicine ,Science - Abstract
Cyclic nucleotide-gated (CNG) ion channels are key mediators underlying signal transduction in retinal and olfactory receptors. Genetic defects in CNGA3 and CNGB3, encoding two structurally related subunits of cone CNG channels, lead to achromatopsia (ACHM). ACHM is a congenital, autosomal recessive retinal disorder that manifests by cone photoreceptor dysfunction, severely reduced visual acuity, impaired or complete color blindness and photophobia. Here, we report the first canine models for CNGA3-associated channelopathy caused by R424W or V644del mutations in the canine CNGA3 ortholog that accurately mimic the clinical and molecular features of human CNGA3-associated ACHM. These two spontaneous mutations exposed CNGA3 residues essential for the preservation of channel function and biogenesis. The CNGA3-R424W results in complete loss of cone function in vivo and channel activity confirmed by in vitro electrophysiology. Structural modeling and molecular dynamics (MD) simulations revealed R424-E306 salt bridge formation and its disruption with the R424W mutant. Reversal of charges in a CNGA3-R424E-E306R double mutant channel rescued cGMP-activated currents uncovering new insights into channel gating. The CNGA3-V644del affects the C-terminal leucine zipper (CLZ) domain destabilizing intersubunit interactions of the coiled-coil complex in the MD simulations; the in vitro experiments showed incompetent trimeric CNGA3 subunit assembly consistent with abnormal biogenesis of in vivo channels. These newly characterized large animal models not only provide a valuable system for studying cone-specific CNG channel function in health and disease, but also represent prime candidates for proof-of-concept studies of CNGA3 gene replacement therapy for ACHM patients.
- Published
- 2015
- Full Text
- View/download PDF
5. A Naturally Occurring Canine Model of Autosomal Recessive Congenital Stationary Night Blindness.
- Author
-
Mineo Kondo, Gautami Das, Ryoetsu Imai, Evelyn Santana, Tomio Nakashita, Miho Imawaka, Kosuke Ueda, Hirohiko Ohtsuka, Kazuhiko Sakai, Takehiro Aihara, Kumiko Kato, Masahiko Sugimoto, Shinji Ueno, Yuji Nishizawa, Gustavo D Aguirre, and Keiko Miyadera
- Subjects
Medicine ,Science - Abstract
Congenital stationary night blindness (CSNB) is a non-progressive, clinically and genetically heterogeneous disease of impaired night vision. We report a naturally-occurring, stationary, autosomal recessive phenotype in beagle dogs with normal daylight vision but absent night vision. Affected dogs had normal retinas on clinical examination, but showed no detectable rod responses. They had "negative-type" mixed rod and cone responses in full-field ERGs. Their photopic long-flash ERGs had normal OFF-responses associated with severely reduced ON-responses. The phenotype is similar to the Schubert-Bornschein form of complete CSNB in humans. Homozygosity mapping ruled out most known CSNB candidates as well as CACNA2D4 and GNB3. Three remaining genes were excluded based on sequencing the open reading frame and intron-exon boundaries (RHO, NYX), causal to a different form of CSNB (RHO) or X-chromosome (NYX, CACNA1F) location. Among the genes expressed in the photoreceptors and their synaptic terminals, and mGluR6 cascade and modulators, reduced expression of GNAT1, CACNA2D4 and NYX was observed by qRT-PCR in both carrier (n = 2) and affected (n = 2) retinas whereas CACNA1F was down-regulated only in the affecteds. Retinal morphology revealed normal cellular layers and structure, and electron microscopy showed normal rod spherules and synaptic ribbons. No difference from normal was observed by immunohistochemistry (IHC) for antibodies labeling rods, cones and their presynaptic terminals. None of the retinas showed any sign of stress. Selected proteins of mGluR6 cascade and its modulators were examined by IHC and showed that PKCα weakly labeled the rod bipolar somata in the affected, but intensely labeled axonal terminals that appeared thickened and irregular. Dendritic terminals of ON-bipolar cells showed increased Goα labeling. Both PKCα and Goα labeled the more prominent bipolar dendrites that extended into the OPL in affected but not normal retinas. Interestingly, RGS11 showed no labeling in the affected retina. Our results indicate involvement of a yet unknown gene in this canine model of complete CSNB.
- Published
- 2015
- Full Text
- View/download PDF
6. Multiple mechanisms contribute to leakiness of a frameshift mutation in canine cone-rod dystrophy.
- Author
-
Keiko Miyadera, Ian Brierley, Jesús Aguirre-Hernández, Cathryn S Mellersh, and David R Sargan
- Subjects
Medicine ,Science - Abstract
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.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.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.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.
- Published
- 2012
- Full Text
- View/download PDF
7. Natural disease history of a canine model of oligogenic RPGRIP1-cone-rod dystrophy establishes variable effects of previously and newly mapped modifier loci
- Author
-
Ana Ripolles-Garcia, Leonardo Murgiano, Natalia Ziolkowska, Felipe Pompeo Marinho, Karolina Roszak, Sommer Iffrig, Gustavo D Aguirre, and Keiko Miyadera
- Subjects
Genetics ,General Medicine ,Molecular Biology ,Genetics (clinical) - Abstract
Canine RPGRIP1-cone-rod dystrophy (CRD), a model for human inherited retinal diseases (IRDs), was originally identified as autosomal recessive early-onset blindness. However, later studies revealed extensive phenotypic variability among RPGRIP1 mutants. This led to the identification of a homozygous MAP9 variant as a modifier associated with early-onset disease. Based on further phenotypic variation affecting cone photoreceptor function, we report mapping of L3 as an additional modifier locus, within a 4.1-Mb locus on canine chromosome 30. We establish the natural disease history of RPGRIP1-CRD based on up to 9-year long-term functional and structural retinal data from 58 dogs including 44 RPGRIP1 mutants grouped according to the modifier status. RPGRIP1 mutants affected by both MAP9 and L3 modifiers exhibited the most severe phenotypes with rapid disease progression. MAP9 alone was found to act as an overall accelerator of rod and cone diseases, while L3 had a cone-specific effect. Ultrastructural analysis of photoreceptors revealed varying degrees of rod and cone damage, while the connecting cilia appeared structurally preserved in all groups. We conclude that RPGRIP1-CRD is an oligogenic disease with at least three loci contributing to the pathogenesis. While the RPGRIP1 variant is required for developing the disease, MAP9 and L3 modifiers exacerbate the phenotype, individually and cumulatively. Oligogenic canine RPGRIP1-CRD illustrates the impact of multiple genetic modifiers on disease phenotype and thus has the potential to reveal new targets for broad-spectrum therapies for oligogenic or polygenic forms of human IRDs.
- Published
- 2023
8. Monocular retinopathy of prematurity‐like retinal vasculopathy in a dog
- Author
-
Kei Takahashi, Alexa Gray, Gustavo D. Aguirre, Valerie L. Dufour, Yu Sato, Keiko Miyadera, and Gerard A. Lutty
- Subjects
medicine.medical_specialty ,genetic structures ,General Veterinary ,medicine.diagnostic_test ,business.industry ,Retinopathy of prematurity ,Histology ,Retinal ,Fundus (eye) ,medicine.disease ,Fluorescein angiography ,Article ,eye diseases ,Neovascularization ,chemistry.chemical_compound ,chemistry ,Ophthalmology ,Retinal vasculopathy ,Medicine ,Immunohistochemistry ,sense organs ,medicine.symptom ,business - Abstract
PURPOSE: To describe a case of monocular retinopathy of prematurity (ROP)-like vasculopathy without oxygen supplementation in the dog. METHODS: Fundus photographs (RetCam), spectral-domain optical coherence tomography (sdOCT), confocal scanning laser ophthalmoscopy (cSLO), and fluorescein angiography (FA), as well as postmortem histology and immunohistochemistry (Collagen IV and anti-vWF antibodies), were carried out to characterize the vascular abnormalities. RESULTS: Ophthalmic examination showed peripheral and mid-temporal avascular areas in the tapetal region, neovascularization and abnormally dilated and tortuous retinal vessels in the left eye. sdOCT demonstrated not only cross-sectional views of preretinal fibrovascular proliferation but also extensive proliferation extraretinally into the vitreous. FA emphasized demarcation of vascular and avascular zones with neovascular tufts “popcorns.” Histology and immunohistochemistry confirmed presence of abnormally dilated vessels and the intravitreal blood vessels. CONCLUSIONS: ROP is a disease of abnormally developed retinal vascularization associated with oxygen supplementation therapy, potentially causing blindness in premature infants. Although the mechanism of ROP-like vasculopathy in our case is unclear, it is important to appreciate that the abnormal vascular pattern seen in ROP in premature infants can occur in canines without oxygen administration.
- Published
- 2021
9. Extended functional rescue following AAV gene therapy in a canine model of LRIT3-congenital stationary night blindness
- Author
-
Kei Takahashi, Jennifer C. Kwok, Yu Sato, Gustavo D. Aguirre, and Keiko Miyadera
- Subjects
Ophthalmology ,Sensory Systems - Published
- 2023
10. Targeting ON-bipolar cells by AAV gene therapy stably reverses
- Author
-
Keiko, Miyadera, Evelyn, Santana, Karolina, Roszak, Sommer, Iffrig, Meike, Visel, Simone, Iwabe, Ryan F, Boyd, Joshua T, Bartoe, Yu, Sato, Alexa, Gray, Ana, Ripolles-Garcia, Valérie L, Dufour, Leah C, Byrne, John G, Flannery, William A, Beltran, and Gustavo D, Aguirre
- Subjects
Dogs ,Night Blindness ,Electroretinography ,Myopia ,Animals ,Humans ,Membrane Proteins ,Eye Diseases, Hereditary ,Genetic Diseases, X-Linked ,Genetic Therapy ,Dependovirus - Abstract
SignificanceCanine models of inherited retinal diseases have helped advance adeno-associated virus (AAV)-based gene therapies targeting specific cells in the outer retina for treating blinding diseases in patients. However, therapeutic targeting of diseases such as congenital stationary night blindness (CSNB) that exhibit defects in ON-bipolar cells (ON-BCs) of the midretina remains underdeveloped. Using a leucine-rich repeat, immunoglobulin-like and transmembrane domain 3 (
- Published
- 2022
11. Krabbe disease successfully treated via monotherapy of intrathecal gene therapy
- Author
-
Ernesto R. Bongarzone, Charles A. Assenmacher, G. Diane Shelton, Keiko Miyadera, Gary P. Swain, Charles H. Vite, Xuntian Jiang, Erik Lykken, Duc Nguyen, Jill Pesayco Salvador, Jessica H. Bagel, Patricia O'Donnell, Arielle Ostrager, Steven J. Gray, Rebecka S. Hess, Mark S. Sands, Allison M. Bradbury, Daniel S. Ory, and Ian J. Hendricks
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Genetic enhancement ,Inflammation ,Hematopoietic stem cell transplantation ,Cisterna magna ,03 medical and health sciences ,Dogs ,0302 clinical medicine ,Galactosylceramidase ,medicine ,Animals ,business.industry ,Leukodystrophy ,Genetic Therapy ,General Medicine ,Dependovirus ,medicine.disease ,Leukodystrophy, Globoid Cell ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Peripheral nervous system ,Immunology ,Krabbe disease ,medicine.symptom ,business ,Research Article - Abstract
Globoid cell leukodystrophy (GLD; Krabbe disease) is a progressive, incurable neurodegenerative disease caused by deficient activity of the hydrolytic enzyme galactosylceramidase (GALC). The ensuing cytotoxic accumulation of psychosine results in diffuse central and peripheral nervous system (CNS, PNS) demyelination. Presymptomatic hematopoietic stem cell transplantation (HSCT) is the only treatment for infantile-onset GLD; however, clinical outcomes of HSCT recipients often remain poor, and procedure-related morbidity is high. There are no effective therapies for symptomatic patients. Herein, we demonstrate in the naturally occurring canine model of GLD that presymptomatic monotherapy with intrathecal AAV9 encoding canine GALC administered into the cisterna magna increased GALC enzyme activity, normalized psychosine concentration, improved myelination, and attenuated inflammation in both the CNS and PNS. Moreover, AAV-mediated therapy successfully prevented clinical neurological dysfunction, allowing treated dogs to live beyond 2.5 years of age, more than 7 times longer than untreated dogs. Furthermore, we found that a 5-fold lower dose resulted in an attenuated form of disease, indicating that sufficient dosing is critical. Finally, postsymptomatic therapy with high-dose AAV9 also significantly extended lifespan, signifying a treatment option for patients for whom HSCT is not applicable. If translatable to patients, these findings would improve the outcomes of patients treated either pre- or postsymptomatically.
- Published
- 2020
12. Intrastromal Gene Therapy Prevents and Reverses Advanced Corneal Clouding in a Canine Model of Mucopolysaccharidosis I
- Author
-
Brian C. Gilger, Telmo Llanga, Kendall Carlin, Patricia O'Donnell, Keiko Miyadera, Matthew L. Hirsch, R. Jude Samulski, Joanne Kurtzberg, Liujiang Song, Jessica H. Bagel, and Laura Conatser
- Subjects
Male ,Pathology ,medicine.medical_specialty ,genetic structures ,Mucopolysaccharidosis I ,medicine.medical_treatment ,Genetic enhancement ,Genetic Vectors ,Fluorescent Antibody Technique ,Gene Expression ,Corneal Diseases ,Animals, Genetically Modified ,Glycosaminoglycan ,Iduronidase ,03 medical and health sciences ,Mucopolysaccharidosis type I ,Dogs ,0302 clinical medicine ,Stroma ,Genes, Reporter ,Cornea ,Drug Discovery ,Genetics ,Lysosomal storage disease ,medicine ,Animals ,Transgenes ,Molecular Biology ,Corneal transplantation ,030304 developmental biology ,Pharmacology ,0303 health sciences ,business.industry ,Gene Transfer Techniques ,Genetic Therapy ,Dependovirus ,medicine.disease ,eye diseases ,Disease Models, Animal ,Treatment Outcome ,medicine.anatomical_structure ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,Molecular Medicine ,Original Article ,Female ,sense organs ,business - Abstract
Mucopolysaccharidosis type I (MPS I) is an autosomal recessive lysosomal storage disease characterized by severe phenotypes, including corneal clouding. MPS I is caused by mutations in alpha-l-iduronidase (IDUA), a ubiquitous enzyme that catalyzes the hydrolysis of glycosaminoglycans. Currently, no treatment exists to address MPS I corneal clouding other than corneal transplantation, which is complicated by a high risk for rejection. Investigation of an adeno-associated virus (AAV) IDUA gene addition strategy targeting the corneal stroma addresses this deficiency. In MPS I canines with early or advanced corneal disease, a single intrastromal AAV8G9-IDUA injection was well tolerated at all administered doses. The eyes with advanced disease demonstrated resolution of corneal clouding as early as 1 week post-injection, followed by sustained corneal transparency until the experimental endpoint of 25 weeks. AAV8G9-IDUA injection in the MPS I canine eye with early corneal disease prevented the development of advanced corneal changes while restoring clarity. Biodistribution studies demonstrated vector genomes in ocular compartments other than the cornea and in some systemic organs; however, a capsid antibody response was detected in only the highest dosed subject. Collectively, the results suggest that intrastromal AAV8G9-IDUA therapy prevents and reverses visual impairment associated with MPS I corneal clouding.
- Published
- 2020
13. NOVEL INSIGHTS INTO CHORIORETINAL AND JUXTAPAPILLARY COLOBOMAS BY OPTICAL COHERENCE TOMOGRAPHY
- Author
-
Alexa P. Gray, Yu Sato, Keiko Miyadera, and Gustavo D. Aguirre
- Subjects
Coloboma ,Dogs ,General Veterinary ,Retinal Diseases ,Choroid ,Animals ,Dog Diseases ,Article ,Retina ,Tomography, Optical Coherence - Abstract
PURPOSE: To describe the in vivo microanatomy of typical and atypical chorioretinal and juxtapapillary colobomas in the dog. METHODS: Three crossbreed dogs were found to be affected with colobomas. Two of the cases were NEHJ1 homozygous and Collie Eye Anomaly (CEA) affected and had the typical optic nerve head colobomas seen with the disease. The third case had an unexpected atypical coloboma. In vivo retinal photography and non-invasive retinal imaging by confocal scanning laser ophthalmoscope (cSLO) and optical coherence tomography (OCT) was done, and the eye affected with the atypical coloboma was collected and processed for histopathological evaluation. RESULTS: The majority of the defining features within the CEA defects were similar, with the extent of change to the choroid being of note. Similar to the first two cases, the atypical coloboma demonstrated absent normal retina, RPE and choroid within the coloboma. Prominent intercalary membranes and vitreal strands attached to the depth of the coloboma were also apparent in all affected eyes. However, unlike the CEA associated colobomas, the atypical coloboma possessed normal choroid surrounding the lesion and the depth of the lesion was apparent throughout. CONCLUSIONS: Advanced retinal imaging enables the appreciation of microanatomical changes that occur in the living eye. The ability of OCT to enhance visualization of abnormal retinal structures and detect subtle neurosensory retinal defects has allowed for the in vivo characterization of features observed in typical and atypical colobomas, as well as the appreciation of some of the resulting structural changes not visible by ophthalmoscopy alone.
- Published
- 2022
14. Targeting ON-bipolar cells by AAV gene therapy stably reverses LRIT3-congenital stationary night blindness
- Author
-
Ana Ripolles Garcia, Gustavo D. Aguirre, Meike Visel, Valerie L. Dufour, Karolina Roszak, Yu Sato, Simone Iwabe, Evelyn Santana, Sommer M Iffrig, Keiko Miyadera, Leah C. Byrne, Ryan F. Boyd, William A. Beltran, Alexa Gray, Joshua T. Bartoe, and John G. Flannery
- Subjects
Congenital stationary night blindness ,Retina ,business.industry ,Genetic enhancement ,Transgene ,Retinal ,Transduction (genetics) ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,Cancer research ,Medicine ,Vector (molecular biology) ,business ,Tropism - Abstract
AAV gene therapies aimed at curing inherited retinal diseases to date have typically focused on photoreceptors and retinal pigmented epithelia within the relatively accessible outer retina. However, therapeutic targeting in diseases such as congenital stationary night blindness (CSNB) that involve defects in ON-bipolar cells (ON-BCs) within the mid-retina has been challenged by the relative inaccessibility of the target cell in intact retinas, the limited transduction efficiency of these cells by existing AAV serotypes, poor availability of established ON-BC-specific promoters, and absence of appropriate patient-relevant large animal models. Here, we demonstrate safe and effective ON-BC targeting by AAV gene therapy in a recently characterized naturally-occurring canine model of CSNB, LRIT3-CSNB. To effectively target ON-BCs, new AAV capsid variants with ON-BC tropism and ON-BC specific modified GRM6 promoters were adopted to ensure cell-specific transgene expression. Notably, subretinal injection of one vector, AAVK9#4-shGRM6-cLRIT3-WPRE, significantly recovered rod-derived b-wave in all treated eyes (6/6) of adult dogs injected at 1-3 years of age. The robust therapeutic effect was evident 7 weeks post-injection and was sustained for at least 1 year in all treated eyes. Scotopic vision was significantly improved in treated eyes based on visually-guided obstacle course navigation. Restoration of LRIT3 signals was confirmed by immunohistochemistry. Thus, we report on the first ON-BC functional rescue in a large animal model using a novel AAV capsid variant and modified promoter construct optimized for ON-BC specificity, thereby establishing both proof-of-concept and a novel translational platform for treatment of CSNB in patients with defects in photoreceptor-to-bipolar signaling.
- Published
- 2021
15. CCDC66 frameshift variant associated with a new form of early-onset progressive retinal atrophy in Portuguese Water Dogs
- Author
-
Sue Pearce-Kelling, Gustavo D. Aguirre, Vidhya Jagannathan, Leonardo Murgiano, Kendall Carlin, Evelyn Santana, Doreen Becker, Courtney Spector, Jessica K. Niggel, Keiko Miyadera, and Tosso Leeb
- Subjects
0301 basic medicine ,Male ,Molecular biology ,Genome-wide association study ,Ciliopathies ,0302 clinical medicine ,Genetics research ,Protein Isoforms ,Sequencing ,DNA sequencing ,Frameshift Mutation ,610 Medicine & health ,Genetics ,Progressive retinal atrophy ,education.field_of_study ,Multidisciplinary ,Cilium ,Retinal Degeneration ,Medical genetics ,Chromosome Mapping ,Disease gene identification ,Retinal diseases ,Pedigree ,Phenotype ,Genetic linkage study ,590 Animals (Zoology) ,Female ,Genotype ,Fundus Oculi ,Population ,Biology ,Retina ,Article ,Frameshift mutation ,03 medical and health sciences ,Dogs ,Genetic linkage ,medicine ,Animals ,Amino Acid Sequence ,RNA, Messenger ,education ,Eye Proteins ,Eye diseases ,Animal breeding ,Genetic association study ,Cell Nucleus ,Base Sequence ,Portugal ,Protein transport ,Molecular Sequence Annotation ,medicine.disease ,030104 developmental biology ,Mutation ,Next-generation sequencing ,Genetic markers ,570 Life sciences ,biology ,Mutant Proteins ,Atrophy ,030217 neurology & neurosurgery - Abstract
Aberrant photoreceptor function or morphogenesis leads to blinding retinal degenerative diseases, the majority of which have a genetic aetiology. A variant in PRCD previously identified in Portuguese Water Dogs (PWDs) underlies prcd (progressive rod-cone degeneration), an autosomal recessive progressive retinal atrophy (PRA) with a late onset at 3–6 years of age or older. Herein, we have identified a new form of early-onset PRA (EOPRA) in the same breed. Pedigree analysis suggested an autosomal recessive inheritance. Four PWD full-siblings affected with EOPRA diagnosed at 2–3 years of age were genotyped (173,661 SNPs) along with 2 unaffected siblings, 2 unaffected parents, and 15 unrelated control PWDs. GWAS, linkage analysis and homozygosity mapping defined a 26-Mb candidate region in canine chromosome 20. Whole-genome sequencing in one affected dog and its obligatory carrier parents identified a 1 bp insertion (CFA20:g.33,717,704_33,717,705insT (CanFam3.1); c.2262_c.2263insA) in CCDC66 predicted to cause a frameshift and truncation (p.Val747SerfsTer8). Screening of an extended PWD population confirmed perfect co-segregation of this genetic variant with the disease. Western blot analysis of COS-1 cells transfected with recombinant mutant CCDC66 expression constructs showed the mutant transcript translated into a truncated protein. Furthermore, in vitro studies suggest that the mutant CCDC66 is mislocalized to the nucleus relative to wild type CCDC66. CCDC66 variants have been associated with inherited retinal degenerations (RDs) including canine and murine ciliopathies. As genetic variants affecting the primary cilium can cause ciliopathies in which RD may be either the sole clinical manifestation or part of a syndrome, our findings further support a role for CCDC66 in retinal function and viability, potentially through its ciliary function.
- Published
- 2020
- Full Text
- View/download PDF
16. Canine inherited retinal degeneration as model to study disease mechanisms and therapy for ciliopathies
- Author
-
Keiko Miyadera
- Subjects
Applied Mathematics ,General Mathematics - Published
- 2022
17. Variabilities in retinal function and structure in a canine model of cone-rod dystrophy associated with RPGRIP1 support multigenic etiology
- Author
-
Rueben G. Das, Felipe Pompeo Marinho, Evelyn Santana, Gustavo D. Aguirre, Kendra McDaid, Simone Iwabe, and Keiko Miyadera
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,genetic structures ,lcsh:Medicine ,030105 genetics & heredity ,Biology ,Retinal Cone Photoreceptor Cells ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,medicine ,lcsh:Science ,Retina ,Multidisciplinary ,medicine.diagnostic_test ,lcsh:R ,Dystrophy ,Retinal ,eye diseases ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,lcsh:Q ,sense organs ,Retinal Rod Photoreceptor Cells ,Erg ,Electroretinography ,Photopic vision - Abstract
Defects in the cilia gene RPGRIP1 cause Leber congenital amaurosis and cone-rod dystrophy in humans. A form of canine cone-rod dystrophy (cord1) was originally associated with a homozygous insertion in RPGRIP1 (RPGRIP1ins/ins) as the primary disease locus while a homozygous deletion in MAP9 (MAP9del/del) was later identified as a modifier associated with the early onset form. However, we find further variability in cone electroretinograms (ERGs) ranging from normal to absent in an extended RPGRIP1ins/ins canine colony, irrespective of the MAP9 genotype. Ophthalmoscopically, cone ERGabsentRPGRIP1ins/ins eyes show discolouration of the tapetal fundus with varying onset and disease progression, while sd-OCT reveals atrophic changes. Despite marked changes in cone ERG and retinal morphology, photopic vision-guided behaviour is comparable between normal and cone ERGabsentRPGRIP1ins/ins littermates. Cone morphology of the dogs lacking cone ERG are truncated with shortened outer and inner segments. Immunohistochemically, cone ERGabsentRPGRIP1ins/ins retinas have extensive L/M-opsin mislocalization, lack CNGB3 labelling in the L/M-cones, and lack GC1 in all cones. Our results indicate that cord1 is a multigenic disease in which mutations in neither RPGRIP1 nor MAP9 alone lead to visual deficits, and additional gene(s) contribute to cone-specific functional and morphologic defects.
- Published
- 2017
18. Genome-wide association study and whole-genome sequencing identify a deletion in LRIT3 associated with canine congenital stationary night blindness
- Author
-
Kendall Carlin, Keiko Miyadera, Gustavo D. Aguirre, Raghavi Sudharsan, Tosso Leeb, Yuji Nishizawa, Orly Goldstein, Vidhya Jagannathan, Evelyn Santana, Rueben G. Das, Doreen Becker, and Mineo Kondo
- Subjects
0301 basic medicine ,Male ,Heterozygote ,Mutant ,lcsh:Medicine ,Locus (genetics) ,Genome-wide association study ,610 Medicine & health ,Biology ,Genome-wide association studies ,Chromosomes ,Retina ,Article ,03 medical and health sciences ,0302 clinical medicine ,Dogs ,Night Blindness ,medicine ,Myopia ,Animals ,Dog Diseases ,lcsh:Science ,Whole genome sequencing ,Genetics ,Congenital stationary night blindness ,Multidisciplinary ,Whole Genome Sequencing ,630 Agriculture ,lcsh:R ,Metabotropic glutamate receptor 6 ,Membrane Proteins ,Heterozygote advantage ,Eye Diseases, Hereditary ,Genetic Diseases, X-Linked ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,590 Animals (Zoology) ,570 Life sciences ,biology ,lcsh:Q ,Female ,030217 neurology & neurosurgery ,Gene Deletion ,Genome-Wide Association Study - Abstract
Congenital stationary night blindness (CSNB), in the complete form, is caused by dysfunctions in ON-bipolar cells (ON-BCs) which are secondary neurons of the retina. We describe the first disease causative variant associated with CSNB in the dog. A genome-wide association study using 12 cases and 11 controls from a research colony determined a 4.6 Mb locus on canine chromosome 32. Subsequent whole-genome sequencing identified a 1 bp deletion in LRIT3 segregating with CSNB. The canine mutant LRIT3 gives rise to a truncated protein with unaltered subcellular expression in vitro. Genetic variants in LRIT3 have been associated with CSNB in patients although there is limited evidence regarding its apparently critical function in the mGluR6 pathway in ON-BCs. We determine that in the canine CSNB retina, the mutant LRIT3 is correctly localized to the region correlating with the ON-BC dendritic tips, albeit with reduced immunolabelling. The LRIT3-CSNB canine model has direct translational potential enabling studies to help understand the CSNB pathogenesis as well as to develop new therapies targeting the secondary neurons of the retina.
- Published
- 2019
19. Mapping of Canine Models of Inherited Retinal Diseases
- Author
-
Keiko, Miyadera
- Subjects
Disease Models, Animal ,Dogs ,Retinal Diseases ,Genetic Linkage ,DNA Mutational Analysis ,Animals ,Dog Diseases ,Sequence Analysis, DNA ,Cone-Rod Dystrophies ,Genetic Association Studies ,Genome-Wide Association Study - Abstract
The gene/mutation discovery approaches for inherited retinal diseases (RDs) in the dog model have seen considerable development over the past 25 years. Initial attempts were focused on candidate genes, followed by genome-wide approaches including linkage analysis and DNA-chip-based genome-wide association study. Combined, there are as many as 32 mutations in 27 genes that have been associated with canine retinal diseases to date. More recently, next-generation sequencing has become one of the key methods of choice. With increasing knowledge of the molecular basis of RDs and follow-up surveys in different subpopulations, the conventional understanding of RDs as simple Mendelian traits is being challenged. Modifiers and involvement of multiple genes that alter the disease expression are complicating the prediction of the disease course. In this chapter, advances in the gene/mutation discovery approaches for canine RDs are reviewed, and a multigenic form of canine RD is discussed using a form of canine cone-rod dystrophy as an example.
- Published
- 2018
20. Mapping of Canine Models of Inherited Retinal Diseases
- Author
-
Keiko Miyadera
- Subjects
0301 basic medicine ,Mutation ,Candidate gene ,Dystrophy ,Genome-wide association study ,Computational biology ,Biology ,medicine.disease_cause ,DNA sequencing ,03 medical and health sciences ,symbols.namesake ,030104 developmental biology ,0302 clinical medicine ,Genetic linkage ,030221 ophthalmology & optometry ,Mendelian inheritance ,symbols ,medicine ,Gene - Abstract
The gene/mutation discovery approaches for inherited retinal diseases (RDs) in the dog model have seen considerable development over the past 25 years. Initial attempts were focused on candidate genes, followed by genome-wide approaches including linkage analysis and DNA-chip-based genome-wide association study. Combined, there are as many as 32 mutations in 27 genes that have been associated with canine retinal diseases to date. More recently, next-generation sequencing has become one of the key methods of choice. With increasing knowledge of the molecular basis of RDs and follow-up surveys in different subpopulations, the conventional understanding of RDs as simple Mendelian traits is being challenged. Modifiers and involvement of multiple genes that alter the disease expression are complicating the prediction of the disease course. In this chapter, advances in the gene/mutation discovery approaches for canine RDs are reviewed, and a multigenic form of canine RD is discussed using a form of canine cone-rod dystrophy as an example.
- Published
- 2018
21. Genome-wide association study in RPGRIP1 −/− dogs identifies a modifier locus that determines the onset of retinal degeneration
- Author
-
Cathryn S. Mellersh, Mike Boursnell, David R. Sargan, Kumiko Kato, and Keiko Miyadera
- Subjects
Male ,Retinal degeneration ,Genotype ,Leber Congenital Amaurosis ,Population ,Genome-wide association study ,Locus (genetics) ,Biology ,Blindness ,Polymorphism, Single Nucleotide ,Article ,Animals, Genetically Modified ,Chromosome 15 ,Dogs ,Retinitis pigmentosa ,Genetics ,medicine ,Animals ,Dog Diseases ,education ,Oligonucleotide Array Sequence Analysis ,education.field_of_study ,Homozygote ,Proteins ,Dystrophy ,Sequence Analysis, DNA ,Retinitis pigmentosa GTPase regulator ,medicine.disease ,Molecular biology ,Phenotype ,Female ,Retinitis Pigmentosa ,Genome-Wide Association Study - 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.
- Published
- 2011
22. Author Correction: Variabilities in retinal function and structure in a canine model of cone-rod dystrophy associated with RPGRIP1 support multigenic etiology
- Author
-
Simone Iwabe, Gustavo D. Aguirre, Rueben G. Das, Keiko Miyadera, Felipe Pompeo Marinho, Evelyn Santana, and Kendra McDaid
- Subjects
Male ,Multifactorial Inheritance ,Pathology ,medicine.medical_specialty ,lcsh:Medicine ,Retina ,03 medical and health sciences ,Dogs ,0302 clinical medicine ,Retinal Rod Photoreceptor Cells ,Electroretinography ,medicine ,Animals ,RNA, Messenger ,Author Correction ,Eye Proteins ,Cone-Rod Dystrophy ,lcsh:Science ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Behavior, Animal ,business.industry ,lcsh:R ,Rod Opsins ,Dendrites ,Retinal Photoreceptor Cell Outer Segment ,Pedigree ,Disease Models, Animal ,Protein Transport ,Gene Expression Regulation ,Retinal Cone Photoreceptor Cells ,Etiology ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,Female ,Retinal function ,lcsh:Q ,business ,Canine model ,Cone-Rod Dystrophies ,030217 neurology & neurosurgery - Abstract
Defects in the cilia gene RPGRIP1 cause Leber congenital amaurosis and cone-rod dystrophy in humans. A form of canine cone-rod dystrophy (cord1) was originally associated with a homozygous insertion in RPGRIP1 (RPGRIP1
- Published
- 2018
23. Clinical and Pathological Aspects of Hemophilia A in Japanese Brown Cattle
- Author
-
Keiko Miyadera, Hiroyuki Ogawa, Maryam Khalaj, Kenichi Shimojo, Yasuo Moritomo, Tetsuo Kunieda, and Yuka Asano
- Subjects
Male ,medicine.medical_specialty ,Cattle Diseases ,Hemophilia A ,Fatal Outcome ,hemic and lymphatic diseases ,von Willebrand Factor ,Coagulopathy ,Animals ,Medicine ,Reduced factor VIII activity ,Subcutaneous hematoma ,Pathological ,Normal range ,Factor VIII ,General Veterinary ,medicine.diagnostic_test ,business.industry ,medicine.disease ,Surgery ,Japanese Brown cattle ,Von Willebrand factor.activity ,Cattle ,Partial Thromboplastin Time ,business ,Partial thromboplastin time - Abstract
A coagulopathy with subcutaneous bleeding and muscular or peritracheal/periesophageal bleeding occurred in two male Japanese Brown calves of the same dam. One of the affected calves died three days after the onset of bleeding and the other survived normally until being slaughtered despite once suffering from subcutaneous hematoma. Hemostatic tests of the latter case showed prolonged activated partial thromboplastin time (APTT), and severely reduced factor VIII activity. In addition, von Willebrand factor activity, determined by the human platelet aggregation test, was within the normal range; therefore, the calf was diagnosed with hemophilia A. These are the first bovine cases of hemophilia A definitely diagnosed clinicopathologically.
- Published
- 2008
24. Marker-assisted selection for forelimb-girdle muscular anomaly of Japanese Black cattle
- Author
-
Keiko Miyadera, A. A. Masoudi, Tetsuo Kunieda, Hiroyuki Ogawa, Kazuyuki Uchida, Kou Yokouchi, and Yoshikazu Sugimoto
- Subjects
Genetics ,Offspring ,Haplotype ,Sire ,Locus (genetics) ,General Medicine ,Biology ,Marker-assisted selection ,medicine.disease ,Hypoplasia ,medicine ,Microsatellite ,Allele ,General Agricultural and Biological Sciences - Abstract
Forelimb-girdle muscular anomaly is a hereditary disorder of Japanese Black cattle characterized by tremors and astasia caused by hypoplasia of the forelimb-girdle muscles. The locus responsible for this disorder has been mapped on a middle region of bovine chromosome 26. In this study, we applied marker-assisted selection to identify the carriers of this disorder. Four microsatellite markers, DIK4440, BM4505, MOK2602 and IDVGA-59, linked to the disorder locus were genotyped in 37 unaffected offspring of a carrier sire. Transmission of the mutant or wild-type allele of the disorder locus of the sire to the 37 offspring was determined by examining the haplotypes of these markers. The results showed that nine and 18 of the 37 animals possessed the paternally transmitted mutant and wild-type alleles, respectively, and therefore, the nine animals with the mutant allele were identified as carriers. We concluded that the marker-assisted selection using these four markers can be applied for the identification of the carriers of forelimb-girdle muscular anomaly of Japanese Black cattle.
- Published
- 2007
25. Canine genome assembly correction facilitates identification of a MAP9 deletion as a potential age of onset modifier for RPGRIP1-associated canine retinal degeneration
- Author
-
Cathryn S. Mellersh, Mike Boursnell, Oliver P. Forman, David R. Sargan, Keiko Miyadera, Rebekkah J. Hitti, Sargan, David [0000-0001-9897-2489], Mellersh, Cathryn [0000-0002-2336-0370], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Retinal degeneration ,Population ,Late onset ,Locus (genetics) ,Biology ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Dogs ,Genetics ,medicine ,Animals ,Humans ,Dog Diseases ,education ,Sequence Deletion ,education.field_of_study ,Genome ,Homozygote ,Retinal Degeneration ,Dystrophy ,Proteins ,Molecular Sequence Annotation ,Exons ,medicine.disease ,Molecular biology ,Pedigree ,Cytoskeletal Proteins ,030104 developmental biology ,Mutation ,030221 ophthalmology & optometry ,Tandem exon duplication ,Age of onset ,Microtubule-Associated Proteins ,Genome-Wide Association Study - Abstract
Retinal degeneration (RD) in the Miniature Long Haired Dachshund (MLHD) is a cone-rod dystrophy resulting in eventual blindness in affected individuals. In a previous study, a 44-nucleotide insertion (ins44) in exon 2 of RPGRIP1 was associated with RD. However, results on an extended population of MLHD revealed a variable RD onset age for ins44 homozygous dogs. Further investigations using a genome-wide association study comparing early onset and late onset RD cases identified an age of onset modifying locus for RD, approximately 30 Mb upstream of RPGRIP1 on chr15. In this investigation, target enriched sequencing identified a MAP9 deletion spanning approximately 22 kb associated with early RD onset. Identification of the deletion required correction to the CanFam3.1 genome build as canine MAP9 is part of a historic tandem duplication, resulting in incomplete assembly of this genome region. The deletion breakpoints were identified in MAP9 intron 10 and in a downstream partial MAP9 pseudogene. The fusion of these two genes, which we have called MAP9 EORD (microtubule-associated protein, early onset retinal degeneration), is in frame and is expressed at the RNA level, with the 3' region containing several predicted deleterious variants. We speculate that MAP9 associates with α-tubulin in the basal body of the cilium. RPGRIP1 is also known to locate to the cilium, where it is closely associated with RPGR. RPGRIP1 mutations also cause redistribution of α-tubulin away from the ciliary region in photoreceptors. Hence, a MAP9 partial deficit is a particularly attractive candidate to synergise with a partial RPGRIP1 deficit to cause a more serious disease.
- Published
- 2015
26. Inherited retinal diseases in dogs: advances in gene/mutation discovery
- Author
-
Keiko Miyadera
- Subjects
Genetics ,Candidate gene ,education.field_of_study ,Mutation ,Genetic enhancement ,Population ,Gene mutation ,Biology ,medicine.disease_cause ,eye diseases ,Article ,Genetic linkage ,medicine ,sense organs ,education ,Gene ,Genetic association - Abstract
1. Inherited retinal diseases (RDs) are vision-threatening conditions affecting humans as well as many domestic animals. Through many years of clinical studies of the domestic dog population, a wide array of RDs has been phenotypically characterized. Extensive effort to map the causative gene and to identify the underlying mutation followed. Through candidate gene, linkage analysis, genome-wide association studies, and more recently, by means of next-generation sequencing, as many as 31 mutations in 24 genes have been identified as the underlying cause for canine RDs. Most of these genes have been associated with human RDs providing opportunities to study their roles in the disease pathogenesis and in normal visual function. The canine model has also contributed in developing new treatments such as gene therapy which has been clinically applied to human patients. Meanwhile, with increasing knowledge of the molecular architecture of RDs in different subpopulations of dogs, the conventional understanding of RDs as a simple monogenic disease is beginning to change. Emerging evidence of modifiers that alters the disease outcome is complicating the interpretation of DNA tests. In this review, advances in the gene/mutation discovery approaches and the emerging genetic complexity of canine RDs are discussed.
- Published
- 2015
27. Canine CNGA3 Gene Mutations Provide Novel Insights into Human Achromatopsia-Associated Channelopathies and Treatment
- Author
-
Lucie Delemotte, Emily V. Dutrow, Margret L. Casal, William R. Crumley, Jacqueline C. Tanaka, Christopher J. Dixon, Naoto Tanaka, Keiko Miyadera, Michael L. Klein, Gustavo D. Aguirre, Shelby L. Reinstein, Karina E Guziewicz, and Christopher M. MacDermaid
- Subjects
Achromatopsia ,Retinal Disorder ,Mutant ,Molecular Sequence Data ,Cyclic Nucleotide-Gated Cation Channels ,lcsh:Medicine ,Color Vision Defects ,Biology ,Gene mutation ,Molecular Dynamics Simulation ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Dogs ,Channelopathy ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Cyclic nucleotide-gated ion channel ,lcsh:Science ,Ion channel ,030304 developmental biology ,Genetics ,0303 health sciences ,Mutation ,Leucine Zippers ,Multidisciplinary ,lcsh:R ,medicine.disease ,Cell biology ,Channelopathies ,lcsh:Q ,Ion Channel Gating ,030217 neurology & neurosurgery ,Research Article - Abstract
Cyclic nucleotide-gated (CNG) ion channels are key mediators underlying signal transduction in retinal and olfactory receptors. Genetic defects in CNGA3 and CNGB3, encoding two structurally related subunits of cone CNG channels, lead to achromatopsia (ACHM). ACHM is a congenital, autosomal recessive retinal disorder that manifests by cone photoreceptor dysfunction, severely reduced visual acuity, impaired or complete color blindness and photophobia. Here, we report the first canine models for CNGA3-associated channelopathy caused by R424W or V644del mutations in the canine CNGA3 ortholog that accurately mimic the clinical and molecular features of human CNGA3-associated ACHM. These two spontaneous mutations exposed CNGA3 residues essential for the preservation of channel function and biogenesis. The CNGA3-R424W results in complete loss of cone function in vivo and channel activity confirmed by in vitro electrophysiology. Structural modeling and molecular dynamics (MD) simulations revealed R424-E306 salt bridge formation and its disruption with the R424W mutant. Reversal of charges in a CNGA3-R424E-E306R double mutant channel rescued cGMP-activated currents uncovering new insights into channel gating. The CNGA3-V644del affects the C-terminal leucine zipper (CLZ) domain destabilizing intersubunit interactions of the coiled-coil complex in the MD simulations; the in vitro experiments showed incompetent trimeric CNGA3 subunit assembly consistent with abnormal biogenesis of in vivo channels. These newly characterized large animal models not only provide a valuable system for studying cone-specific CNG channel function in health and disease, but also represent prime candidates for proof-of-concept studies of CNGA3 gene replacement therapy for ACHM patients.
- Published
- 2015
28. An insertion mutation of the bovine F11 gene is responsible for factor XI deficiency in Japanese black cattle
- Author
-
Maryam Khalaj, Keiko Miyadera, Hiroyuki Ogawa, Tetsuo Kunieda, Miho Ikeda, Abdol Rahim Abbasi, Masaki Kunieda, and Takehito Tsuji
- Subjects
Genetics ,Mutation ,Intron ,Biology ,medicine.disease_cause ,Molecular biology ,Exon ,medicine ,Coding region ,Insertion ,Factor XI ,Gene ,Genotyping - Abstract
Factor XI deficiency in Japanese black cattle is an hereditary mild bleeding disorder with an autosomal recessive mode of inheritance. To characterize the molecular lesion causing factor XI deficiency in cattle, we isolated an entire coding region of the bovine F11 gene, which comprises 15 exons and 14 introns, and determined its nucleotide sequences. Comparison of the nucleotide sequences of the F11 gene between affected and unaffected animals revealed an insertion of 15 nucleotides in exon 9 of the affected animals. The insertion results in a substitution of one amino acid with six amino acids in a highly conserved amino acid sequence in the fourth apple domain of factor XI protein. Genotyping of the F11 gene in 109 Japanese black cattle revealed that the insertion clearly corresponded to the factor XI activities of the animals. We therefore concluded that the insertion of 15 nucleotides in the F11 gene is the causative mutation for factor XI deficiency in Japanese black cattle. Genotyping of the F11gene by detecting the insertion will be an effective DNA-based diagnostic system to prevent incidence of the disease.
- Published
- 2005
29. Investigating the inheritance of prolapsed nictitating membrane glands in a large canine pedigree
- Author
-
András M. Komáromy, Keiko Miyadera, Simone Iwabe, and Michele L. Edelmann
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Dachshund ,biology.animal_breed ,Pedigree chart ,German shorthaired pointer ,Article ,symbols.namesake ,Inheritance (object-oriented programming) ,Second line ,Dogs ,Exocrine Glands ,Prolapse ,medicine ,Animals ,Genetic Predisposition to Disease ,Dog Diseases ,Nictitating Membrane ,General Veterinary ,biology ,biology.organism_classification ,Pedigree ,Mendelian inheritance ,symbols ,Eyelid Diseases ,Mongrel ,Female ,Nictitating membrane - Abstract
Objective To investigate the inheritance of prolapsed nictitating membrane glands (PNMG) in a large pedigree of purpose-bred mongrel dogs. Animals studied Two lines of purpose-bred mongrel dogs kept at a research facility with controlled environment were analyzed for frequent occurrences of PNMG. The first line (GS line) consisted of 201 dogs, derived from one German shorthaired pointer and seven mongrel dogs. The second line (M line) was established from one mongrel dog and three miniature longhaired dachshund (MLHD) dogs followed by closed breeding practice (n = 50). The two canine lines were connected by a female dog, which contributed genetically to both lines. Procedures Medical records of all dogs were reviewed retrospectively for signalment, parental data, and the presence of PNMG. Pedigrees were constructed to facilitate assessment of inheritance. Results The overall prevalence of PNMG in the GS line was 4.0% (8/201) over a 12-year period. The prevalence in the M line was 10.0% (5/50) over 6 years, which increased to 23.1% (3/13) when only dogs aged 2 years or older were considered. Analysis of the pedigrees ruled out simple modes of Mendelian inheritance in both canine lines. Conclusion The high prevalence of PNMG in two canine lines bred and maintained under a strictly controlled environment supported the involvement of genetic risk factors. The mode of inheritance remains to be determined, but it appears to be complex and potentially multigenic.
- Published
- 2012
30. Genetic and phenotypic variations of inherited retinal diseases in dogs: the power of within- and across-breed studies
- Author
-
Gustavo D. Aguirre, Keiko Miyadera, and Gregory M. Acland
- Subjects
Retinal degeneration ,Candidate gene ,Genotype ,Population ,Biology ,Article ,Dogs ,Retinal Diseases ,Genetic linkage ,Genetics ,medicine ,Animals ,Dog Diseases ,Allele ,education ,Vision, Ocular ,Genetic association ,Progressive retinal atrophy ,education.field_of_study ,Cyclic Nucleotide Phosphodiesterases, Type 6 ,Retinal Degeneration ,Genetic Variation ,medicine.disease ,Human genetics ,Phenotype ,Mutation ,Genome-Wide Association Study - Abstract
Considerable clinical and molecular variations have been known in retinal blinding diseases in man and also in dogs. Different forms of retinal diseases occur in specific breed(s) caused by mutations segregating within each isolated breeding population. While molecular studies to find genes and mutations underlying retinal diseases in dogs have benefited largely from the phenotypic and genetic uniformity within a breed, within- and across-breed variations have often played a key role in elucidating the molecular basis. The increasing knowledge of phenotypic, allelic, and genetic heterogeneities in canine retinal degeneration has shown that the overall picture is rather more complicated than initially thought. Over the past 20 years, various approaches have been developed and tested to search for genes and mutations underlying genetic traits in dogs, depending on the availability of genetic tools and sample resources. Candidate gene, linkage analysis, and genome-wide association studies have so far identified 24 mutations in 18 genes underlying retinal diseases in at least 58 dog breeds. Many of these genes have been associated with retinal diseases in humans, thus providing opportunities to study the role in pathogenesis and in normal vision. Application in therapeutic interventions such as gene therapy has proven successful initially in a naturally occurring dog model followed by trials in human patients. Other genes whose human homologs have not been associated with retinal diseases are potential candidates to explain equivalent human diseases and contribute to the understanding of their function in vision.
- Published
- 2011
31. A Naturally Occurring Canine Model of Autosomal Recessive Congenital Stationary Night Blindness
- Author
-
Takehiro Aihara, Gustavo D. Aguirre, Masahiko Sugimoto, Kumiko Kato, Kazuhiko Sakai, Miho Imawaka, Yuji Nishizawa, Mineo Kondo, Evelyn Santana, Ryoetsu Imai, Tomio Nakashita, Gautami Das, Shinji Ueno, Keiko Miyadera, Kosuke Ueda, and Hirohiko Ohtsuka
- Subjects
Male ,Calcium Channels, L-Type ,genetic structures ,Presynaptic Terminals ,lcsh:Medicine ,Biology ,Retinal Cone Photoreceptor Cells ,Retina ,chemistry.chemical_compound ,Dogs ,Night Blindness ,Myopia ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,lcsh:Science ,GNAT1 ,Congenital stationary night blindness ,Genetics ,Multidisciplinary ,lcsh:R ,Metabotropic glutamate receptor 6 ,Eye Diseases, Hereditary ,Genetic Diseases, X-Linked ,Retinal ,Heterotrimeric GTP-Binding Proteins ,Molecular biology ,Pedigree ,Disease Models, Animal ,medicine.anatomical_structure ,Gene Expression Regulation ,chemistry ,Immunohistochemistry ,Female ,Proteoglycans ,lcsh:Q ,sense organs ,RGS Proteins ,Research Article ,Photopic vision - Abstract
Congenital stationary night blindness (CSNB) is a non-progressive, clinically and genetically heterogeneous disease of impaired night vision. We report a naturally-occurring, stationary, autosomal recessive phenotype in beagle dogs with normal daylight vision but absent night vision. Affected dogs had normal retinas on clinical examination, but showed no detectable rod responses. They had “negative-type” mixed rod and cone responses in full-field ERGs. Their photopic long-flash ERGs had normal OFF-responses associated with severely reduced ON-responses. The phenotype is similar to the Schubert-Bornschein form of complete CSNB in humans. Homozygosity mapping ruled out most known CSNB candidates as well as CACNA2D4 and GNB3. Three remaining genes were excluded based on sequencing the open reading frame and intron-exon boundaries (RHO, NYX), causal to a different form of CSNB (RHO) or X-chromosome (NYX, CACNA1F) location. Among the genes expressed in the photoreceptors and their synaptic terminals, and mGluR6 cascade and modulators, reduced expression of GNAT1, CACNA2D4 and NYX was observed by qRT-PCR in both carrier (n = 2) and affected (n = 2) retinas whereas CACNA1F was down-regulated only in the affecteds. Retinal morphology revealed normal cellular layers and structure, and electron microscopy showed normal rod spherules and synaptic ribbons. No difference from normal was observed by immunohistochemistry (IHC) for antibodies labeling rods, cones and their presynaptic terminals. None of the retinas showed any sign of stress. Selected proteins of mGluR6 cascade and its modulators were examined by IHC and showed that PKCα weakly labeled the rod bipolar somata in the affected, but intensely labeled axonal terminals that appeared thickened and irregular. Dendritic terminals of ON-bipolar cells showed increased Goα labeling. Both PKCα and Goα labeled the more prominent bipolar dendrites that extended into the OPL in affected but not normal retinas. Interestingly, RGS11 showed no labeling in the affected retina. Our results indicate involvement of a yet unknown gene in this canine model of complete CSNB.
- Published
- 2015
32. An insertion mutation of the bovine Fii gene is responsible for factor XI deficiency in Japanese black cattle
- Author
-
Masaki, Kunieda, Takehito, Tsuji, Abdol Rahim, Abbasi, Maryam, Khalaj, Miho, Ikeda, Keiko, Miyadera, Hiroyuki, Ogawa, and Tetsuo, Kunieda
- Subjects
Base Sequence ,Sequence Homology, Amino Acid ,Factor XI Deficiency ,Molecular Sequence Data ,Cattle Diseases ,Chromosome Mapping ,Genes, Recessive ,Exons ,Polymerase Chain Reaction ,Introns ,Mice ,DNA Transposable Elements ,Animals ,Humans ,Cattle ,Amino Acid Sequence ,Sequence Alignment ,Factor XI ,DNA Primers - Abstract
Factor XI deficiency in Japanese black cattle is an hereditary mild bleeding disorder with an autosomal recessive mode of inheritance. To characterize the molecular lesion causing factor XI deficiency in cattle, we isolated an entire coding region of the bovine F11 gene, which comprises 15 exons and 14 introns, and determined its nucleotide sequences. Comparison of the nucleotide sequences of the F11 gene between affected and unaffected animals revealed an insertion of 15 nucleotides in exon 9 of the affected animals. The insertion results in a substitution of one amino acid with six amino acids in a highly conserved amino acid sequence in the fourth apple domain of factor XI protein. Genotyping of the F11 gene in 109 Japanese black cattle revealed that the insertion clearly corresponded to the factor XI activities of the animals. We therefore concluded that the insertion of 15 nucleotides in the F11 gene is the causative mutation for factor XI deficiency in Japanese black cattle. Genotyping of the F11gene by detecting the insertion will be an effective DNA-based diagnostic system to prevent incidence of the disease.
- Published
- 2005
33. Multiple Mechanisms Contribute to Leakiness of a Frameshift Mutation in Canine Cone-Rod Dystrophy
- Author
-
Cathryn S. Mellersh, Ian Brierley, Keiko Miyadera, Jesús Aguirre-Hernández, and David R. Sargan
- Subjects
Transcription, Genetic ,lcsh:Medicine ,Polymerase Chain Reaction ,Veterinary Opthalmology ,Gene Splicing ,Autosomal Recessive ,Genes, Reporter ,Protein Isoforms ,Dog Diseases ,Frameshift Mutation ,Luciferases ,lcsh:Science ,Animal Management ,Genetics ,education.field_of_study ,Translational frameshift ,Multidisciplinary ,Reverse Transcriptase Polymerase Chain Reaction ,Exons ,RNA splicing ,Medicine ,Retinal Disorders ,Retinitis Pigmentosa ,Research Article ,Veterinary Medicine ,Gene isoform ,DNA, Complementary ,Clinical Research Design ,Molecular Sequence Data ,DNA transcription ,Population ,Biology ,Frameshift mutation ,Dogs ,Genetic Mutation ,Animals ,RNA, Messenger ,Animal Models of Disease ,Insertion ,education ,Gene ,Alleles ,Clinical Genetics ,Base Sequence ,lcsh:R ,Alternative splicing ,Mutation Types ,Electrophoresis, Capillary ,Proteins ,Ophthalmology ,Haplotypes ,Protein Biosynthesis ,Genetics of Disease ,Veterinary Science ,lcsh:Q ,Gene expression ,Gene Function ,Poly A ,Animal Genetics - Abstract
Mutations in RPGRIP1 are associated with early onset retinal degenerations in humans and dogs. Dogs homozygous for a 44 bp insertion including a polyA29 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 polyAn length in the insertion and slippage at polyAn 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 polyAn length was confirmed in DNA from different RPGRIP1 2/2 dogs, yet polyAn variability did not correspond with the clinical phenotypes and no individual was found that carried a polyAn 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 inframe 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.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.