1
|
Nonarath HJT, Simpson SL, Slobodianuk TL, Collery RF, Dinculescu A, Link BA. The USH3A causative gene clarin1 functions in Müller glia to maintain retinal photoreceptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.29.582878. [PMID: 38464015 PMCID: PMC10925332 DOI: 10.1101/2024.02.29.582878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Mutations in CLRN1 cause Usher syndrome type IIIA (USH3A), an autosomal recessive disorder characterized by hearing and vision loss, and often accompanied by vestibular balance issues. The identity of the cell types responsible for the pathology and mechanisms leading to vision loss in USH3A remains elusive. To address this, we employed CRISPR/Cas9 technology to delete a large region in the coding and untranslated (UTR) region of zebrafish clrn1. Retina of clrn1 mutant larvae exhibited sensitivity to cell stress, along with age-dependent loss of function and degeneration in the photoreceptor layer. Investigation revealed disorganization in the outer retina in clrn1 mutants, including actin-based structures of the Müller glia and photoreceptor cells. To assess cell-specific contributions to USH3A pathology, we specifically re-expressed clrn1 in either Müller glia or photoreceptor cells. Müller glia re-expression of clrn1 prevented the elevated cell death observed in larval clrn1 mutant zebrafish exposed to high-intensity light. Notably, the degree of phenotypic rescue correlated with the level of Clrn1 re-expression. Surprisingly, high levels of Clrn1 expression enhanced cell death in both wild-type and clrn1 mutant animals. However, rod- or cone-specific Clrn1 re-expression did not rescue the extent of cell death. Taken together, our findings underscore three crucial insights. First, clrn1 mutant zebrafish exhibit key pathological features of USH3A; second, Clrn1 within Müller glia plays a pivotal role in photoreceptor maintenance, with its expression requiring controlled regulation; third, the reliance of photoreceptors on Müller glia suggests a structural support mechanism, possibly through direct interactions between Müller glia and photoreceptors mediated in part by Clrn1 protein.
Collapse
Affiliation(s)
- Hannah J. T. Nonarath
- Department Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Samantha L. Simpson
- Department of Ophthalmology and Vision Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Tricia L. Slobodianuk
- Department Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Ross F. Collery
- Department of Ophthalmology and Vision Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Astra Dinculescu
- Department of Ophthalmology, University of Florida, Gainesville, Florida 32611
| | - Brian A. Link
- Department Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| |
Collapse
|
2
|
Tebbe L, Mwoyosvi ML, Crane R, Makia MS, Kakakhel M, Cosgrove D, Al-Ubaidi MR, Naash MI. The usherin mutation c.2299delG leads to its mislocalization and disrupts interactions with whirlin and VLGR1. Nat Commun 2023; 14:972. [PMID: 36810733 PMCID: PMC9944904 DOI: 10.1038/s41467-023-36431-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 02/01/2023] [Indexed: 02/24/2023] Open
Abstract
Usher syndrome (USH) is the leading cause of combined deafness-blindness with type 2 A (USH2A) being the most common form. Knockout models of USH proteins, like the Ush2a-/- model that develops a late-onset retinal phenotype, failed to mimic the retinal phenotype observed in patients. Since patient's mutations result in the expression of a mutant protein and to determine the mechanism of USH2A, we generated and evaluated an usherin (USH2A) knock-in mouse expressing the common human disease-mutation, c.2299delG. This mouse exhibits retinal degeneration and expresses a truncated, glycosylated protein which is mislocalized to the photoreceptor inner segment. The degeneration is associated with a decline in retinal function, structural abnormalities in connecting cilium and outer segment and mislocaliztion of the usherin interactors very long G-protein receptor 1 and whirlin. The onset of symptoms is significantly earlier compared to Ush2a-/-, proving expression of mutated protein is required to recapitulate the patients' retinal phenotype.
Collapse
Affiliation(s)
- Lars Tebbe
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Maggie L Mwoyosvi
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
- Department of Microbiology & Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Ryan Crane
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Mustafa S Makia
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Mashal Kakakhel
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | | | - Muayyad R Al-Ubaidi
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA.
| | - Muna I Naash
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
3
|
Xu NY, Liu ZY, Yang QM, Bian PP, Li M, Zhao X. Genomic Analyses for Selective Signatures and Genes Involved in Hot Adaptation Among Indigenous Chickens From Different Tropical Climate Regions. Front Genet 2022; 13:906447. [PMID: 35979430 PMCID: PMC9377314 DOI: 10.3389/fgene.2022.906447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
Climate change, especially weather extremes like extreme cold or extreme hot, is a major challenge for global livestock. One of the animal breeding goals for sustainable livestock production should be to breed animals with excellent climate adaptability. Indigenous livestock and poultry are well adapted to the local climate, and they are good resources to study the genetic footprints and mechanism of the resilience to weather extremes. In order to identify selection signatures and genes that might be involved in hot adaptation in indigenous chickens from different tropical climates, we conducted a genomic analysis of 65 indigenous chickens that inhabit different climates. Several important unique positively selected genes (PSGs) were identified for each local chicken group by the cross-population extended haplotype homozygosity (XP-EHH). These PSGs, verified by composite likelihood ratio, genetic differentiation index, nucleotide diversity, Tajima’s D, and decorrelated composite of multiple signals, are related to nerve regulation, vascular function, immune function, lipid metabolism, kidney development, and function, which are involved in thermoregulation and hot adaptation. However, one common PSG was detected for all three tropical groups of chickens via XP-EHH but was not confirmed by other five types of selective sweep analyses. These results suggest that the hot adaptability of indigenous chickens from different tropical climate regions has evolved in parallel by taking different pathways with different sets of genes. The results from our study have provided reasonable explanations and insights for the rapid adaptation of chickens to diverse tropical climates and provide practical values for poultry breeding.
Collapse
Affiliation(s)
- Nai-Yi Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Zhen-Yu Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Qi-Meng Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Pei-Pei Bian
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Ming Li
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Xin Zhao
- Department of Animal Science, McGill University, Montreal, QC, Canada
- *Correspondence: Xin Zhao,
| |
Collapse
|
4
|
Smirnov VM, Nassisi M, Mohand-Saïd S, Bonnet C, Aubois A, Devisme C, Dib T, Zeitz C, Loundon N, Marlin S, Petit C, Bodaghi B, Sahel JA, Audo I. Retinal Phenotype of Patients with CLRN1-Associated Usher 3A Syndrome in French Light4Deaf Cohort. Invest Ophthalmol Vis Sci 2022; 63:25. [PMID: 35481838 PMCID: PMC9055553 DOI: 10.1167/iovs.63.4.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Purpose Biallelic variants in CLRN1 are responsible for Usher syndrome 3A and non-syndromic rod–cone dystrophy (RCD). Retinal findings in Usher syndrome 3A have not been well defined. We report the detailed phenotypic description of RCD associated with CLRN1 variants in a prospective cohort. Methods Patients were clinically investigated at the National Reference Center for rare ocular diseases at the Quinze-Vingts Hospital, Paris, France. Best-corrected visual acuity (BCVA) tests, Goldmann perimetry, full-field electroretinography (ffERG), retinal photography, near-infrared reflectance, short-wavelength and near-infrared autofluorescence, and optical coherence tomography (OCT) were performed for all patients. Results Four patients from four unrelated families were recruited. Mean follow-up was 11 years for three patients, and only baseline data were available for one subject. Median BCVA at baseline was 0.2 logMAR (range, 0.3–0). ffERG responses were undetectable in all subjects. The III4e isopter of the Goldmann visual field was constricted to 10°. The retinal phenotype was consistent in all patients: small whitish granular atrophic areas were organized in a network pattern around the macula and in the midperiphery. OCT showed intraretinal microcysts in all patients. Upon follow-up, all patients experienced a progressive BCVA loss and further visual field constriction. Four distinct pathogenic variants were identified in our patients: two missense (c.144T>G, p.(Asn48Lys) and c.368C>A, p.(Ala123Asp)) and two frameshift variants (c.176del, p.(Gly59Valfs*13) and c.230dup, p.(Ala78Serfs*52)). Conclusions RCD in Usher 3A syndrome has some distinctive features. It is a severe photoreceptor dystrophy with whitish granular posterior pole appearance and cystic maculopathy.
Collapse
Affiliation(s)
- Vasily M Smirnov
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.,Université de Lille, Faculté de Médecine, Lille, France
| | - Marco Nassisi
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Saddek Mohand-Saïd
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.,CHNO des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and DHU Sight Restore, INSERM-DGOS CIC1423, Paris, France
| | - Crystel Bonnet
- Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, Paris, France.,Unité Mixte de Recherche en Santé 1120, INSERM, Paris, France.,Institut de l'Audition, Paris, France
| | - Anne Aubois
- CHNO des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and DHU Sight Restore, INSERM-DGOS CIC1423, Paris, France
| | - Céline Devisme
- CHNO des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and DHU Sight Restore, INSERM-DGOS CIC1423, Paris, France
| | - Thilissa Dib
- CHNO des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and DHU Sight Restore, INSERM-DGOS CIC1423, Paris, France
| | - Christina Zeitz
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Natalie Loundon
- Otorhinolaryngologie Pédiatrique, APHP Hôpital Necker, Paris, France.,Centre de référence des Surdités Génétiques, Service de Génétique, APHP Hôpital Necker, Paris, France
| | - Sandrine Marlin
- Centre de référence des Surdités Génétiques, Service de Génétique, APHP Hôpital Necker, Paris, France
| | - Christine Petit
- Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, Paris, France.,Unité Mixte de Recherche en Santé 1120, INSERM, Paris, France.,Institut de l'Audition, Paris, France.,Collège de France, Paris, France
| | | | - José-Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.,CHNO des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and DHU Sight Restore, INSERM-DGOS CIC1423, Paris, France.,Fondation Ophtalmologique Adolphe de Rothschild, Paris, France.,Department of Ophthalmology, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, United States.,Académie des Sciences-Institut de France, Paris, France
| | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.,CHNO des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and DHU Sight Restore, INSERM-DGOS CIC1423, Paris, France.,Institute of Ophthalmology, University College of London, London, United Kingdom
| |
Collapse
|
5
|
Usher syndrome IIIA: a review of the disorder and preclinical research advances in therapeutic approaches. Hum Genet 2022; 141:759-783. [PMID: 35320418 DOI: 10.1007/s00439-022-02446-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/03/2022] [Indexed: 12/27/2022]
Abstract
Usher syndrome (USH) is an autosomal recessive disorder characterized by sensorineural hearing loss, progressive pigmentary retinopathy, and vestibular dysfunction. The degree and onset of hearing loss vary among subtypes I, II, and III, while blindness often occurs in the second to fourth decades of life. Usher type III (USH3), characterized by postlingual progressive sensorineural hearing loss, varying levels of vestibular dysfunction, and varying degrees of visual impairment, typically manifests in the first to second decades of life. While USH3 is rare, it is highly prevalent in certain populations. RP61, USH3, and USH3A symbolize the same disorder, with the latter symbol used more frequently in recent literature. This review focuses on the clinical features, epidemiology, molecular genetics, treatment, and research advances for sensory deficits in USH3A.
Collapse
|
6
|
Yusuf IH, Garrett A, MacLaren RE, Issa PC. Retinal cadherins and the retinal cadherinopathies: Current concepts and future directions. Prog Retin Eye Res 2022; 90:101038. [DOI: 10.1016/j.preteyeres.2021.101038] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 12/13/2021] [Accepted: 12/20/2021] [Indexed: 12/18/2022]
|
7
|
Dinculescu A, Link BA, Saperstein DA. Retinal Gene Therapy for Usher Syndrome: Current Developments, Challenges, and Perspectives. Int Ophthalmol Clin 2021; 61:109-124. [PMID: 34584048 PMCID: PMC8478317 DOI: 10.1097/iio.0000000000000378] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
8
|
Fuster-García C, García-Bohórquez B, Rodríguez-Muñoz A, Aller E, Jaijo T, Millán JM, García-García G. Usher Syndrome: Genetics of a Human Ciliopathy. Int J Mol Sci 2021; 22:6723. [PMID: 34201633 PMCID: PMC8268283 DOI: 10.3390/ijms22136723] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/21/2022] Open
Abstract
Usher syndrome (USH) is an autosomal recessive syndromic ciliopathy characterized by sensorineural hearing loss, retinitis pigmentosa and, sometimes, vestibular dysfunction. There are three clinical types depending on the severity and age of onset of the symptoms; in addition, ten genes are reported to be causative of USH, and six more related to the disease. These genes encode proteins of a diverse nature, which interact and form a dynamic protein network called the "Usher interactome". In the organ of Corti, the USH proteins are essential for the correct development and maintenance of the structure and cohesion of the stereocilia. In the retina, the USH protein network is principally located in the periciliary region of the photoreceptors, and plays an important role in the maintenance of the periciliary structure and the trafficking of molecules between the inner and the outer segments of photoreceptors. Even though some genes are clearly involved in the syndrome, others are controversial. Moreover, expression of some USH genes has been detected in other tissues, which could explain their involvement in additional mild comorbidities. In this paper, we review the genetics of Usher syndrome and the spectrum of mutations in USH genes. The aim is to identify possible mutation associations with the disease and provide an updated genotype-phenotype correlation.
Collapse
Affiliation(s)
- Carla Fuster-García
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
- Biomedical Research Network for Rare Diseases, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - Belén García-Bohórquez
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
| | - Ana Rodríguez-Muñoz
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
| | - Elena Aller
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
- Biomedical Research Network for Rare Diseases, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Genetics Unit, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - Teresa Jaijo
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
- Biomedical Research Network for Rare Diseases, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Genetics Unit, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - José M. Millán
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
- Biomedical Research Network for Rare Diseases, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - Gema García-García
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
- Biomedical Research Network for Rare Diseases, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| |
Collapse
|
9
|
Whatley M, Francis A, Ng ZY, Khoh XE, Atlas MD, Dilley RJ, Wong EYM. Usher Syndrome: Genetics and Molecular Links of Hearing Loss and Directions for Therapy. Front Genet 2020; 11:565216. [PMID: 33193648 PMCID: PMC7642844 DOI: 10.3389/fgene.2020.565216] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/21/2020] [Indexed: 12/19/2022] Open
Abstract
Usher syndrome (USH) is an autosomal recessive (AR) disorder that permanently and severely affects the senses of hearing, vision, and balance. Three clinically distinct types of USH have been identified, decreasing in severity from Type 1 to 3, with symptoms of sensorineural hearing loss (SNHL), retinitis pigmentosa (RP), and vestibular dysfunction. There are currently nine confirmed and two suspected USH-causative genes, and a further three candidate loci have been mapped. The proteins encoded by these genes form complexes that play critical roles in the development and maintenance of cellular structures within the inner ear and retina, which have minimal capacity for repair or regeneration. In the cochlea, stereocilia are located on the apical surface of inner ear hair cells (HC) and are responsible for transducing mechanical stimuli from sound pressure waves into chemical signals. These signals are then detected by the auditory nerve fibers, transmitted to the brain and interpreted as sound. Disease-causing mutations in USH genes can destabilize the tip links that bind the stereocilia to each other, and cause defects in protein trafficking and stereocilia bundle morphology, thereby inhibiting mechanosensory transduction. This review summarizes the current knowledge on Usher syndrome with a particular emphasis on mutations in USH genes, USH protein structures, and functional analyses in animal models. Currently, there is no cure for USH. However, the genetic therapies that are rapidly developing will benefit from this compilation of detailed genetic information to identify the most effective strategies for restoring functional USH proteins.
Collapse
Affiliation(s)
- Meg Whatley
- Ear Science Institute Australia, Nedlands, WA, Australia
| | - Abbie Francis
- Ear Science Institute Australia, Nedlands, WA, Australia
- Emergency Medicine, The University of Western Australia, Nedlands, WA, Australia
| | - Zi Ying Ng
- Ear Science Institute Australia, Nedlands, WA, Australia
| | - Xin Ee Khoh
- Ear Science Institute Australia, Nedlands, WA, Australia
- School of Human Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Marcus D. Atlas
- Ear Science Institute Australia, Nedlands, WA, Australia
- Ear Sciences Centre, The University of Western Australia, Nedlands, WA, Australia
| | - Rodney J. Dilley
- Ear Science Institute Australia, Nedlands, WA, Australia
- Ear Sciences Centre, The University of Western Australia, Nedlands, WA, Australia
- Centre for Cell Therapy and Regenerative Medicine, The University of Western Australia, Perth, WA, Australia
| | - Elaine Y. M. Wong
- Ear Science Institute Australia, Nedlands, WA, Australia
- Ear Sciences Centre, The University of Western Australia, Nedlands, WA, Australia
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin University, Bentley, WA, Australia
| |
Collapse
|
10
|
Duplication in ECR Near HMX1 and a SNP in GATA6 Genes Regulate Microtia in Awassi Sheep. Genes (Basel) 2020; 11:genes11060597. [PMID: 32481741 PMCID: PMC7349607 DOI: 10.3390/genes11060597] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/22/2020] [Accepted: 05/28/2020] [Indexed: 01/14/2023] Open
Abstract
Microtia and anotia are hereditary traits characterized by an underdevelopment or complete absence of the outer ear. These congenital malformations observed in many species can exist as part of various syndromes or as an isolated trait as seen in the fat-tailed Awassi sheep breed. Our study aims to identify the genetic mutations causing microtia in Awassi sheep by DNA sequencing. DNA was extracted from blood samples randomly collected from 84 Awassi sheep (16 earless, 41 short ear and 27 normal ear) across different farms. GATA6 exons 1, 2, 4, 6 and 7, CLRN1 intron 3, DCC intron 2, ECR near HMX1 and the intergenic region between GATA6 and MIB1 genes were screened, amplified and sequenced. Allele and genotype frequencies were calculated by direct counting. Association was performed using chi-squared test for goodness-of-fit. Results showed mutations in only two genes significantly associated with microtia in Awassi: duplication in part of ECR near HMX1 (6:114293121-6:114293196) and a SNP at GATA6 exon 7 (23:34498242). Association results revealed that the ECR locus accounts for the microtia phenotype, while GATA6 exon 7 acts as a modifier gene. Genetic screening for these loci can be used to improve selection against microtia in Awassi sheep.
Collapse
|
11
|
Xu L, Bolch SN, Santiago CP, Dyka FM, Akil O, Lobanova ES, Wang Y, Martemyanov KA, Hauswirth WW, Smith WC, Handa JT, Blackshaw S, Ash JD, Dinculescu A. Clarin-1 expression in adult mouse and human retina highlights a role of Müller glia in Usher syndrome. J Pathol 2019; 250:195-204. [PMID: 31625146 PMCID: PMC7003947 DOI: 10.1002/path.5360] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/17/2019] [Accepted: 10/16/2019] [Indexed: 02/06/2023]
Abstract
Usher syndrome type 3 (USH3) is an autosomal recessively inherited disorder caused by mutations in the gene clarin‐1 (CLRN1), leading to combined progressive hearing loss and retinal degeneration. The cellular distribution of CLRN1 in the retina remains uncertain, either because its expression levels are low or because its epitopes are masked. Indeed, in the adult mouse retina, Clrn1 mRNA is developmentally downregulated, detectable only by RT‐PCR. In this study we used the highly sensitive RNAscope in situ hybridization assay and single‐cell RNA‐sequencing techniques to investigate the distribution of Clrn1 and CLRN1 in mouse and human retina, respectively. We found that Clrn1 transcripts in mouse tissue are localized to the inner retina during postnatal development and in adult stages. The pattern of Clrn1 mRNA cellular expression is similar in both mouse and human adult retina, with CLRN1 transcripts being localized in Müller glia, and not photoreceptors. We generated a novel knock‐in mouse with a hemagglutinin (HA) epitope‐tagged CLRN1 and showed that CLRN1 is expressed continuously at the protein level in the retina. Following enzymatic deglycosylation and immunoblotting analysis, we detected a single CLRN1‐specific protein band in homogenates of mouse and human retina, consistent in size with the main CLRN1 isoform. Taken together, our results implicate Müller glia in USH3 pathology, placing this cell type to the center of future mechanistic and therapeutic studies to prevent vision loss in this disease. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Lei Xu
- Department of Ophthalmology, University of Florida, Gainesville, FL, USA
| | - Susan N Bolch
- Department of Ophthalmology, University of Florida, Gainesville, FL, USA
| | - Clayton P Santiago
- Solomon H Snyder Department of Neuroscience, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Frank M Dyka
- Department of Ophthalmology, University of Florida, Gainesville, FL, USA
| | - Omar Akil
- Department of Otolaryngology-HNS, University of California, San Francisco, CA, USA
| | | | - Yuchen Wang
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | | | | | - W Clay Smith
- Department of Ophthalmology, University of Florida, Gainesville, FL, USA
| | - James T Handa
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Seth Blackshaw
- Solomon H Snyder Department of Neuroscience, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.,Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Center for Human Systems Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John D Ash
- Department of Ophthalmology, University of Florida, Gainesville, FL, USA
| | - Astra Dinculescu
- Department of Ophthalmology, University of Florida, Gainesville, FL, USA
| |
Collapse
|
12
|
Dunbar LA, Patni P, Aguilar C, Mburu P, Corns L, Wells HRR, Delmaghani S, Parker A, Johnson S, Williams D, Esapa CT, Simon MM, Chessum L, Newton S, Dorning J, Jeyarajan P, Morse S, Lelli A, Codner GF, Peineau T, Gopal SR, Alagramam KN, Hertzano R, Dulon D, Wells S, Williams FM, Petit C, Dawson SJ, Brown SDM, Marcotti W, El‐Amraoui A, Bowl MR. Clarin-2 is essential for hearing by maintaining stereocilia integrity and function. EMBO Mol Med 2019; 11:e10288. [PMID: 31448880 PMCID: PMC6728604 DOI: 10.15252/emmm.201910288] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 07/23/2019] [Accepted: 07/26/2019] [Indexed: 11/21/2022] Open
Abstract
Hearing relies on mechanically gated ion channels present in the actin-rich stereocilia bundles at the apical surface of cochlear hair cells. Our knowledge of the mechanisms underlying the formation and maintenance of the sound-receptive structure is limited. Utilizing a large-scale forward genetic screen in mice, genome mapping and gene complementation tests, we identified Clrn2 as a new deafness gene. The Clrn2clarinet/clarinet mice (p.Trp4* mutation) exhibit a progressive, early-onset hearing loss, with no overt retinal deficits. Utilizing data from the UK Biobank study, we could show that CLRN2 is involved in human non-syndromic progressive hearing loss. Our in-depth morphological, molecular and functional investigations establish that while it is not required for initial formation of cochlear sensory hair cell stereocilia bundles, clarin-2 is critical for maintaining normal bundle integrity and functioning. In the differentiating hair bundles, lack of clarin-2 leads to loss of mechano-electrical transduction, followed by selective progressive loss of the transducing stereocilia. Together, our findings demonstrate a key role for clarin-2 in mammalian hearing, providing insights into the interplay between mechano-electrical transduction and stereocilia maintenance.
Collapse
Affiliation(s)
- Lucy A Dunbar
- Mammalian Genetics UnitMRC Harwell InstituteHarwellUK
| | - Pranav Patni
- Déficits Sensoriels ProgressifsInstitut PasteurINSERM UMR‐S 1120Sorbonne UniversitésParisFrance
| | | | | | - Laura Corns
- Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
| | - Helena RR Wells
- Department of Twin Research & Genetic EpidemiologyKing's College LondonLondonUK
| | - Sedigheh Delmaghani
- Déficits Sensoriels ProgressifsInstitut PasteurINSERM UMR‐S 1120Sorbonne UniversitésParisFrance
| | - Andrew Parker
- Mammalian Genetics UnitMRC Harwell InstituteHarwellUK
| | - Stuart Johnson
- Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
| | | | | | | | | | | | | | | | - Susan Morse
- Mammalian Genetics UnitMRC Harwell InstituteHarwellUK
| | - Andrea Lelli
- Génétique et Physiologie de l'AuditionInstitut PasteurINSERM UMR‐S 1120Collège de FranceSorbonne UniversitésParisFrance
| | | | - Thibault Peineau
- Laboratoire de Neurophysiologie de la Synapse AuditiveUniversité de BordeauxBordeauxFrance
| | - Suhasini R Gopal
- Department of Otolaryngology – Head and Neck SurgeryUniversity Hospitals Cleveland Medical CenterCase Western Reserve UniversityClevelandOHUSA
| | - Kumar N Alagramam
- Department of Otolaryngology – Head and Neck SurgeryUniversity Hospitals Cleveland Medical CenterCase Western Reserve UniversityClevelandOHUSA
| | - Ronna Hertzano
- Department of Otorhinolaryngology Head and Neck Surgery, Anatomy and Neurobiology and Institute for Genome SciencesUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Didier Dulon
- Laboratoire de Neurophysiologie de la Synapse AuditiveUniversité de BordeauxBordeauxFrance
| | - Sara Wells
- Mary Lyon CentreMRC Harwell InstituteHarwellUK
| | - Frances M Williams
- Department of Twin Research & Genetic EpidemiologyKing's College LondonLondonUK
| | - Christine Petit
- Génétique et Physiologie de l'AuditionInstitut PasteurINSERM UMR‐S 1120Collège de FranceSorbonne UniversitésParisFrance
| | | | | | - Walter Marcotti
- Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
| | - Aziz El‐Amraoui
- Déficits Sensoriels ProgressifsInstitut PasteurINSERM UMR‐S 1120Sorbonne UniversitésParisFrance
| | | |
Collapse
|
13
|
György B, Meijer EJ, Ivanchenko MV, Tenneson K, Emond F, Hanlon KS, Indzhykulian AA, Volak A, Karavitaki KD, Tamvakologos PI, Vezina M, Berezovskii VK, Born RT, O'Brien M, Lafond JF, Arsenijevic Y, Kenna MA, Maguire CA, Corey DP. Gene Transfer with AAV9-PHP.B Rescues Hearing in a Mouse Model of Usher Syndrome 3A and Transduces Hair Cells in a Non-human Primate. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 13:1-13. [PMID: 30581889 PMCID: PMC6297893 DOI: 10.1016/j.omtm.2018.11.003] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 11/14/2018] [Indexed: 12/01/2022]
Abstract
Hereditary hearing loss often results from mutation of genes expressed by cochlear hair cells. Gene addition using AAV vectors has shown some efficacy in mouse models, but clinical application requires two additional advances. First, new AAV capsids must mediate efficient transgene expression in both inner and outer hair cells of the cochlea. Second, to have the best chance of clinical translation, these new vectors must also transduce hair cells in non-human primates. Here, we show that an AAV9 capsid variant, PHP.B, produces efficient transgene expression of a GFP reporter in both inner and outer hair cells of neonatal mice. We show also that AAV9-PHP.B mediates almost complete transduction of inner and outer HCs in a non-human primate. In a mouse model of Usher syndrome type 3A deafness (gene CLRN1), we use AAV9-PHP.B encoding Clrn1 to partially rescue hearing. Thus, we have identified a vector with promise for clinical treatment of hereditary hearing disorders, and we demonstrate, for the first time, viral transduction of the inner ear of a primate with an AAV vector.
Collapse
Affiliation(s)
- Bence György
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.,Molecular Neurogenetics Unit, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02114, USA
| | - Elise J Meijer
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | - Killian S Hanlon
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.,Molecular Neurogenetics Unit, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02114, USA
| | - Artur A Indzhykulian
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.,Department of Otolaryngology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Adrienn Volak
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02114, USA
| | | | | | - Mark Vezina
- Charles River Laboratories, Senneville, QC, Canada
| | | | - Richard T Born
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | - Yvan Arsenijevic
- Unit of Retinal Degeneration and Regeneration, Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Lausanne, Switzerland
| | - Margaret A Kenna
- Department of Otolaryngology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Casey A Maguire
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02114, USA
| | - David P Corey
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
14
|
Mastrangelo S, Sottile G, Sutera AM, Di Gerlando R, Tolone M, Moscarelli A, Sardina MT, Portolano B. Genome-wide association study reveals the locus responsible for microtia in Valle del Belice sheep breed. Anim Genet 2018; 49:636-640. [PMID: 30160299 DOI: 10.1111/age.12719] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2018] [Indexed: 01/09/2023]
Abstract
Microtia is a congenital deformity of the outer ear with phenotypes varying from a small auricle to total absence (anotia). The genetic basis is still poorly understood, and very few studies have been performed in sheep. Valle del Belice sheep is a breed showing microtia. The aim of this study was to identify the potential genomic regions involved in microtia in sheep. A total of 40 individuals, 20 with microtia and 20 normal, were genotyped with the Illumina OvineSNP50 BeadChip. The comparison among the results from a genome-wide association study, Fisher's exact test and FST analysis revealed a single strong association signal: rs419889303 on chromosome 1, located within intron 3 of the CLRN1 gene. Our study suggests for the first time that this novel candidate gene is responsible for microtia in sheep. Additional analysis based on the sequencing would help confirm our findings and allow for the proposal of a precise genetic basis for microtia in sheep.
Collapse
Affiliation(s)
- S Mastrangelo
- Dipartimento Scienze Agrarie, Alimentari e Forestali, University of Palermo, 90128, Palermo, Italy
| | - G Sottile
- Dipartimento Scienze Economiche, Aziendali e Statistiche, University of Palermo, 90128, Palermo, Italy
| | - A M Sutera
- Dipartimento Scienze Agrarie, Alimentari e Forestali, University of Palermo, 90128, Palermo, Italy
| | - R Di Gerlando
- Dipartimento Scienze Agrarie, Alimentari e Forestali, University of Palermo, 90128, Palermo, Italy
| | - M Tolone
- Dipartimento Scienze Agrarie, Alimentari e Forestali, University of Palermo, 90128, Palermo, Italy
| | - A Moscarelli
- Dipartimento Scienze Agrarie, Alimentari e Forestali, University of Palermo, 90128, Palermo, Italy
| | - M T Sardina
- Dipartimento Scienze Agrarie, Alimentari e Forestali, University of Palermo, 90128, Palermo, Italy
| | - B Portolano
- Dipartimento Scienze Agrarie, Alimentari e Forestali, University of Palermo, 90128, Palermo, Italy
| |
Collapse
|
15
|
Dulon D, Papal S, Patni P, Cortese M, Vincent PF, Tertrais M, Emptoz A, Tlili A, Bouleau Y, Michel V, Delmaghani S, Aghaie A, Pepermans E, Alegria-Prevot O, Akil O, Lustig L, Avan P, Safieddine S, Petit C, El-Amraoui A. Clarin-1 gene transfer rescues auditory synaptopathy in model of Usher syndrome. J Clin Invest 2018; 128:3382-3401. [PMID: 29985171 DOI: 10.1172/jci94351] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/17/2018] [Indexed: 11/17/2022] Open
Abstract
Clarin-1, a tetraspan-like membrane protein defective in Usher syndrome type IIIA (USH3A), is essential for hair bundle morphogenesis in auditory hair cells. We report a new synaptic role for clarin-1 in mouse auditory hair cells elucidated by characterization of Clrn1 total (Clrn1ex4-/-) and postnatal hair cell-specific conditional (Clrn1ex4fl/fl Myo15-Cre+/-) knockout mice. Clrn1ex4-/- mice were profoundly deaf, whereas Clrn1ex4fl/fl Myo15-Cre+/- mice displayed progressive increases in hearing thresholds, with, initially, normal otoacoustic emissions and hair bundle morphology. Inner hair cell (IHC) patch-clamp recordings for the 2 mutant mice revealed defective exocytosis and a disorganization of synaptic F-actin and CaV1.3 Ca2+ channels, indicative of a synaptopathy. Postsynaptic defects were also observed, with an abnormally broad distribution of AMPA receptors associated with a loss of afferent dendrites and defective electrically evoked auditory brainstem responses. Protein-protein interaction assays revealed interactions between clarin-1 and the synaptic CaV1.3 Ca2+ channel complex via the Cavβ2 auxiliary subunit and the PDZ domain-containing protein harmonin (defective in Usher syndrome type IC). Cochlear gene therapy in vivo, through adeno-associated virus-mediated Clrn1 transfer into hair cells, prevented the synaptic defects and durably improved hearing in Clrn1ex4fl/fl Myo15-Cre+/- mice. Our results identify clarin-1 as a key organizer of IHC ribbon synapses, and suggest new treatment possibilities for USH3A patients.
Collapse
Affiliation(s)
- Didier Dulon
- UMRS 1120, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Université de Bordeaux, Laboratoire de Neurophysiologie de la Synapse Auditive, Bordeaux Neurocampus, Bordeaux, France
| | - Samantha Papal
- UMRS 1120, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, Paris, France.,Sorbonne Universités, Complexité du Vivant, Paris, France
| | - Pranav Patni
- UMRS 1120, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, Paris, France.,Sorbonne Universités, Complexité du Vivant, Paris, France
| | - Matteo Cortese
- UMRS 1120, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, Paris, France.,Sorbonne Universités, Complexité du Vivant, Paris, France
| | - Philippe Fy Vincent
- UMRS 1120, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Université de Bordeaux, Laboratoire de Neurophysiologie de la Synapse Auditive, Bordeaux Neurocampus, Bordeaux, France
| | - Margot Tertrais
- UMRS 1120, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Université de Bordeaux, Laboratoire de Neurophysiologie de la Synapse Auditive, Bordeaux Neurocampus, Bordeaux, France
| | - Alice Emptoz
- UMRS 1120, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, Paris, France.,Sorbonne Universités, Complexité du Vivant, Paris, France
| | - Abdelaziz Tlili
- UMRS 1120, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, Paris, France.,Sorbonne Universités, Complexité du Vivant, Paris, France
| | - Yohan Bouleau
- UMRS 1120, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Université de Bordeaux, Laboratoire de Neurophysiologie de la Synapse Auditive, Bordeaux Neurocampus, Bordeaux, France
| | - Vincent Michel
- UMRS 1120, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, Paris, France.,Sorbonne Universités, Complexité du Vivant, Paris, France
| | - Sedigheh Delmaghani
- UMRS 1120, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, Paris, France.,Sorbonne Universités, Complexité du Vivant, Paris, France
| | - Alain Aghaie
- UMRS 1120, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, Paris, France.,Sorbonne Universités, Complexité du Vivant, Paris, France
| | - Elise Pepermans
- UMRS 1120, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, Paris, France.,Sorbonne Universités, Complexité du Vivant, Paris, France
| | - Olinda Alegria-Prevot
- UMRS 1120, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, Paris, France.,Sorbonne Universités, Complexité du Vivant, Paris, France
| | - Omar Akil
- Department of Otolaryngology-Head and Neck Surgery, UCSF, San Francisco, California, USA
| | - Lawrence Lustig
- Department of Otolaryngology-Head and Neck Surgery, Columbia University Medical Center, New York, New York, USA
| | - Paul Avan
- Laboratoire de Biophysique Sensorielle, Faculté de Médecine, Université d'Auvergne; Biophysique Médicale, Centre Jean Perrin, Clermont-Ferrand, France
| | - Saaid Safieddine
- UMRS 1120, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, Paris, France.,Sorbonne Universités, Complexité du Vivant, Paris, France.,Centre National de la Recherche Scientifique, Paris, France
| | - Christine Petit
- UMRS 1120, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, Paris, France.,Sorbonne Universités, Complexité du Vivant, Paris, France.,Collège de France, Paris, France
| | - Aziz El-Amraoui
- UMRS 1120, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, Paris, France.,Sorbonne Universités, Complexité du Vivant, Paris, France
| |
Collapse
|
16
|
Rudman JR, Mei C, Bressler SE, Blanton SH, Liu XZ. Precision medicine in hearing loss. J Genet Genomics 2018; 45:99-109. [PMID: 29500086 DOI: 10.1016/j.jgg.2018.02.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 02/12/2018] [Accepted: 02/12/2018] [Indexed: 11/26/2022]
Abstract
Precision medicine (PM) proposes customized medical care based on a patient's unique genome, biomarkers, environment and behaviors. Hearing loss (HL) is the most common sensorineural disorder worldwide and is frequently caused by a single genetic mutation. With recent advances in PM tools such as genetic sequencing and data analysis, the field of HL is ideally positioned to adopt the strategies of PM. Here, we review current and future applications of PM in HL as they relate to the four core qualities of PM (P4): predictive, personalized, patient-centered, and participatory. We then introduce a strategy for effective incorporation of HL PM into the design of future research studies, electronic medical records, and clinical practice to improve diagnostics, prognostics, and, ultimately, individualized patient treatment. Finally, specific anticipated ethical and economic concerns in this growing era of genomics-based HL treatment are discussed. By integrating PM principles into translational HL research and clinical practice, hearing specialists are uniquely positioned to effectively treat the heterogeneous causes and manifestations of HL on an individualized basis.
Collapse
Affiliation(s)
- Jason R Rudman
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Christine Mei
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Sara E Bressler
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Susan H Blanton
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA; John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Xue-Zhong Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA; John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
17
|
Geng R, Omar A, Gopal SR, Chen DHC, Stepanyan R, Basch ML, Dinculescu A, Furness DN, Saperstein D, Hauswirth W, Lustig LR, Alagramam KN. Modeling and Preventing Progressive Hearing Loss in Usher Syndrome III. Sci Rep 2017; 7:13480. [PMID: 29044151 PMCID: PMC5647385 DOI: 10.1038/s41598-017-13620-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 09/29/2017] [Indexed: 02/07/2023] Open
Abstract
Usher syndrome type III (USH3) characterized by progressive loss of vision and hearing is caused by mutations in the clarin-1 gene (CLRN1). Clrn1 knockout (KO) mice develop hair cell defects by postnatal day 2 (P2) and are deaf by P21-P25. Early onset profound hearing loss in KO mice and lack of information about the cochlear cell type that requires Clrn1 expression pose challenges to therapeutic investigation. We generated KO mice harboring a transgene, TgAC1, consisting of Clrn1-UTR (Clrn1 cDNA including its 5' and 3' UTR) under the control of regulatory elements (Atoh1 3' enhancer/β-globin basal promoter) to direct expression of Clrn1 in hair cells during development and down regulate it postnatally. The KO-TgAC1 mice displayed delayed onset progressive hearing loss associated with deterioration of the hair bundle structure, leading to the hypothesis that hair cell expression of Clrn1 is essential for postnatal preservation of hair cell structure and hearing. Consistent with that hypothesis, perinatal transfection of hair cells in KO-TgAC1 mice with a single injection of AAV-Clrn1-UTR vector showed correlative preservation of the hair bundle structure and hearing through adult life. Further, the efficacy of AAV-Clrn1 vector was significantly attenuated, revealing the potential importance of UTR in gene therapy.
Collapse
Affiliation(s)
- Ruishuang Geng
- Department of Otolaryngology-Head and Neck Surgery, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, Ohio, 44016, USA
| | - Akil Omar
- Department of Otolaryngology-Head and Neck Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Suhasini R Gopal
- Department of Otolaryngology-Head and Neck Surgery, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, Ohio, 44016, USA
| | - Daniel H-C Chen
- Department of Otolaryngology-Head and Neck Surgery, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, Ohio, 44016, USA
| | - Ruben Stepanyan
- Department of Otolaryngology-Head and Neck Surgery, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, Ohio, 44016, USA
| | - Martin L Basch
- Department of Otolaryngology-Head and Neck Surgery, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, Ohio, 44016, USA
| | - Astra Dinculescu
- Department of Ophthalmology, University of Florida, Gainesville, FL, 32610, USA
| | - David N Furness
- School of Life Sciences, Keele University, Keele, Staffordshire, ST5 5BG, UK
| | | | - William Hauswirth
- Department of Ophthalmology, University of Florida, Gainesville, FL, 32610, USA
| | - Lawrence R Lustig
- Department of Otolaryngology-Head and Neck Surgery, University of California San Francisco, San Francisco, CA, 94143, USA.
- Department of Otolaryngology-Head and Neck Surgery, Columbia University, New York, NY, 10032, USA.
| | - Kumar N Alagramam
- Department of Otolaryngology-Head and Neck Surgery, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, Ohio, 44016, USA.
- Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, 44016, USA.
- Neurosciences, Case Western Reserve University, Cleveland, Ohio, 44016, USA.
| |
Collapse
|
18
|
Bujakowska KM, Liu Q, Pierce EA. Photoreceptor Cilia and Retinal Ciliopathies. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a028274. [PMID: 28289063 DOI: 10.1101/cshperspect.a028274] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Photoreceptors are sensory neurons designed to convert light stimuli into neurological responses. This process, called phototransduction, takes place in the outer segments (OS) of rod and cone photoreceptors. OS are specialized sensory cilia, with analogous structures to those present in other nonmotile cilia. Deficient morphogenesis and/or dysfunction of photoreceptor sensory cilia (PSC) caused by mutations in a variety of photoreceptor-specific and common cilia genes can lead to inherited retinal degenerations (IRDs). IRDs can manifest as isolated retinal diseases or syndromic diseases. In this review, we describe the structure and composition of PSC and different forms of ciliopathies with retinal involvement. We review the genetics of the IRDs, which are monogenic disorders but genetically diverse with regard to causality.
Collapse
Affiliation(s)
- Kinga M Bujakowska
- Ocular Genomics Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114
| | - Qin Liu
- Ocular Genomics Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114
| | - Eric A Pierce
- Ocular Genomics Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114
| |
Collapse
|
19
|
Alagramam KN, Gopal SR, Geng R, Chen DHC, Nemet I, Lee R, Tian G, Miyagi M, Malagu KF, Lock CJ, Esmieu WRK, Owens AP, Lindsay NA, Ouwehand K, Albertus F, Fischer DF, Bürli RW, MacLeod AM, Harte WE, Palczewski K, Imanishi Y. A small molecule mitigates hearing loss in a mouse model of Usher syndrome III. Nat Chem Biol 2016; 12:444-51. [PMID: 27110679 PMCID: PMC4871731 DOI: 10.1038/nchembio.2069] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 03/08/2016] [Indexed: 12/30/2022]
Abstract
Usher syndrome type III (USH3), characterized by progressive deafness, variable balance disorder and blindness, is caused by destabilizing mutations in the gene encoding the clarin-1 (CLRN1) protein. Here we report a new strategy to mitigate hearing loss associated with a common USH3 mutation CLRN1(N48K) that involves cell-based high-throughput screening of small molecules capable of stabilizing CLRN1(N48K), followed by a secondary screening to eliminate general proteasome inhibitors, and finally an iterative process to optimize structure-activity relationships. This resulted in the identification of BioFocus 844 (BF844). To test the efficacy of BF844, we developed a mouse model that mimicked the progressive hearing loss associated with USH3. BF844 effectively attenuated progressive hearing loss and prevented deafness in this model. Because the CLRN1(N48K) mutation causes both hearing and vision loss, BF844 could in principle prevent both sensory deficiencies in patients with USH3. Moreover, the strategy described here could help identify drugs for other protein-destabilizing monogenic disorders.
Collapse
Affiliation(s)
- Kumar N Alagramam
- Otolaryngology Head and Neck Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, Ohio, USA.,Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA.,Neurosciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Suhasini R Gopal
- Otolaryngology Head and Neck Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Ruishuang Geng
- Otolaryngology Head and Neck Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Daniel H-C Chen
- Otolaryngology Head and Neck Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Ina Nemet
- Pharmacology and Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Richard Lee
- Pharmacology and Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Guilian Tian
- Pharmacology and Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Masaru Miyagi
- Center for Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Karine F Malagu
- BioFocus, a Charles River company, Chesterford Research Park, Saffron Walden, UK
| | - Christopher J Lock
- BioFocus, a Charles River company, Chesterford Research Park, Saffron Walden, UK
| | - William R K Esmieu
- BioFocus, a Charles River company, Chesterford Research Park, Saffron Walden, UK
| | - Andrew P Owens
- BioFocus, a Charles River company, Chesterford Research Park, Saffron Walden, UK
| | - Nicola A Lindsay
- BioFocus, a Charles River company, Chesterford Research Park, Saffron Walden, UK
| | | | | | | | - Roland W Bürli
- BioFocus, a Charles River company, Chesterford Research Park, Saffron Walden, UK
| | - Angus M MacLeod
- BioFocus, a Charles River company, Chesterford Research Park, Saffron Walden, UK
| | - William E Harte
- Office of Translation and Innovation, Case Western Reserve University, Cleveland, Ohio, USA
| | - Krzysztof Palczewski
- Pharmacology and Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Yoshikazu Imanishi
- Pharmacology and Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
20
|
Dinculescu A, Stupay RM, Deng WT, Dyka FM, Min SH, Boye SL, Chiodo VA, Abrahan CE, Zhu P, Li Q, Strettoi E, Novelli E, Nagel-Wolfrum K, Wolfrum U, Smith WC, Hauswirth WW. AAV-Mediated Clarin-1 Expression in the Mouse Retina: Implications for USH3A Gene Therapy. PLoS One 2016; 11:e0148874. [PMID: 26881841 PMCID: PMC4755610 DOI: 10.1371/journal.pone.0148874] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 01/23/2016] [Indexed: 02/07/2023] Open
Abstract
Usher syndrome type III (USH3A) is an autosomal recessive disorder caused by mutations in clarin-1 (CLRN1) gene, leading to progressive retinal degeneration and sensorineural deafness. Efforts to develop therapies for preventing photoreceptor cell loss are hampered by the lack of a retinal phenotype in the existing USH3 mouse models and by conflicting reports regarding the endogenous retinal localization of clarin-1, a transmembrane protein of unknown function. In this study, we used an AAV-based approach to express CLRN1 in the mouse retina in order to determine the pattern of its subcellular localization in different cell types. We found that all major classes of retinal cells express AAV-delivered CLRN1 driven by the ubiquitous, constitutive small chicken β-actin promoter, which has important implications for the design of future USH3 gene therapy studies. Within photoreceptor cells, AAV-expressed CLRN1 is mainly localized at the inner segment region and outer plexiform layer, similar to the endogenous expression of other usher proteins. Subretinal delivery using a full strength viral titer led to significant loss of retinal function as evidenced by ERG analysis, suggesting that there is a critical limit for CLRN1 expression in photoreceptor cells. Taken together, these results suggest that CLRN1 expression is potentially supported by a variety of retinal cells, and the right combination of AAV vector dose, promoter, and delivery method needs to be selected to develop safe therapies for USH3 disorder.
Collapse
Affiliation(s)
- Astra Dinculescu
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
- * E-mail:
| | - Rachel M. Stupay
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
| | - Wen-Tao Deng
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
| | - Frank M. Dyka
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
| | - Seok-Hong Min
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
| | - Sanford L. Boye
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
| | - Vince A. Chiodo
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
| | - Carolina E. Abrahan
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
| | - Ping Zhu
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
| | - Qiuhong Li
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
| | | | | | - Kerstin Nagel-Wolfrum
- Cell and Matrix Biology, Institute of Zoology, Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Uwe Wolfrum
- Cell and Matrix Biology, Institute of Zoology, Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - W. Clay Smith
- Ophthalmology, University of Florida, Gainesville, FL, United States of America
| | | |
Collapse
|
21
|
Zebrafish Models for the Mechanosensory Hair Cell Dysfunction in Usher Syndrome 3 Reveal That Clarin-1 Is an Essential Hair Bundle Protein. J Neurosci 2015; 35:10188-201. [PMID: 26180195 DOI: 10.1523/jneurosci.1096-15.2015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Usher syndrome type III (USH3) is characterized by progressive loss of hearing and vision, and varying degrees of vestibular dysfunction. It is caused by mutations that affect the human clarin-1 protein (hCLRN1), a member of the tetraspanin protein family. The missense mutation CLRN1(N48K), which affects a conserved N-glycosylation site in hCLRN1, is a common causative USH3 mutation among Ashkenazi Jews. The affected individuals hear at birth but lose that function over time. Here, we developed an animal model system using zebrafish transgenesis and gene targeting to provide an explanation for this phenotype. Immunolabeling demonstrated that Clrn1 localized to the hair cell bundles (hair bundles). The clrn1 mutants generated by zinc finger nucleases displayed aberrant hair bundle morphology with diminished function. Two transgenic zebrafish that express either hCLRN1 or hCLRN1(N48K) in hair cells were produced to examine the subcellular localization patterns of wild-type and mutant human proteins. hCLRN1 localized to the hair bundles similarly to zebrafish Clrn1; in contrast, hCLRN1(N48K) largely mislocalized to the cell body with a small amount reaching the hair bundle. We propose that this small amount of hCLRN1(N48K) in the hair bundle provides clarin-1-mediated function during the early stages of life; however, the presence of hCLRN1(N48K) in the hair bundle diminishes over time because of intracellular degradation of the mutant protein, leading to progressive loss of hair bundle integrity and hair cell function. These findings and genetic tools provide an understanding and path forward to identify therapies to mitigate hearing loss linked to the CLRN1 mutation. SIGNIFICANCE STATEMENT Mutations in the clarin-1 gene affect eye and ear function in humans. Individuals with the CLRN1(N48K) mutation are born able to hear but lose that function over time. Here, we develop an animal model system using zebrafish transgenesis and gene targeting to provide an explanation for this phenotype. This approach illuminates the role of clarin-1 and the molecular mechanism linked to the CLRN1(N48K) mutation in sensory hair cells of the inner ear. Additionally, the investigation provided an in vivo model to guide future drug discovery to rescue the hCLRN1(N48K) in hair cells.
Collapse
|
22
|
Toms M, Bitner-Glindzicz M, Webster A, Moosajee M. Usher syndrome: a review of the clinical phenotype, genes and therapeutic strategies. EXPERT REVIEW OF OPHTHALMOLOGY 2015. [DOI: 10.1586/17469899.2015.1033403] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
23
|
Discovery of molecular markers to discriminate corneal endothelial cells in the human body. PLoS One 2015; 10:e0117581. [PMID: 25807145 PMCID: PMC4373821 DOI: 10.1371/journal.pone.0117581] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 12/29/2014] [Indexed: 12/13/2022] Open
Abstract
The corneal endothelium is a monolayer of hexagonal corneal endothelial cells (CECs) on the inner surface of the cornea. CECs are critical in maintaining corneal transparency through their barrier and pump functions. CECs in vivo have a limited capacity in proliferation, and loss of a significant number of CECs results in corneal edema called bullous keratopathy which can lead to severe visual loss. Corneal transplantation is the most effective method to treat corneal endothelial dysfunction, where it suffers from donor shortage. Therefore, regeneration of CECs from other cell types attracts increasing interests, and specific markers of CECs are crucial to identify actual CECs. However, the currently used markers are far from satisfactory because of their non-specific expression in other cell types. Here, we explored molecular markers to discriminate CECs from other cell types in the human body by integrating the published RNA-seq data of CECs and the FANTOM5 atlas representing diverse range of cell types based on expression patterns. We identified five genes, CLRN1, MRGPRX3, HTR1D, GRIP1 and ZP4 as novel markers of CECs, and the specificities of these genes were successfully confirmed by independent experiments at both the RNA and protein levels. Notably none of them have been documented in the context of CEC function. These markers could be useful for the purification of actual CECs, and also available for the evaluation of the products derived from other cell types. Our results demonstrate an effective approach to identify molecular markers for CECs and open the door for the regeneration of CECs in vitro.
Collapse
|
24
|
Brennenstuhl C, Tanimoto N, Burkard M, Wagner R, Bolz S, Trifunovic D, Kabagema-Bilan C, Paquet-Durand F, Beck SC, Huber G, Seeliger MW, Ruth P, Wissinger B, Lukowski R. Targeted ablation of the Pde6h gene in mice reveals cross-species differences in cone and rod phototransduction protein isoform inventory. J Biol Chem 2015; 290:10242-55. [PMID: 25739440 DOI: 10.1074/jbc.m114.611921] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Indexed: 11/06/2022] Open
Abstract
Phosphodiesterase-6 (PDE6) is a multisubunit enzyme that plays a key role in the visual transduction cascade in rod and cone photoreceptors. Each type of photoreceptor utilizes discrete catalytic and inhibitory PDE6 subunits to fulfill its physiological tasks, i.e. the degradation of cyclic guanosine-3',5'-monophosphate at specifically tuned rates and kinetics. Recently, the human PDE6H gene was identified as a novel locus for autosomal recessive (incomplete) color blindness. However, the three different classes of cones were not affected to the same extent. Short wave cone function was more preserved than middle and long wave cone function indicating that some basic regulation of the PDE6 multisubunit enzyme was maintained albeit by a unknown mechanism. To study normal and disease-related functions of cone Pde6h in vivo, we generated Pde6h knock-out (Pde6h(-/-)) mice. Expression of PDE6H in murine eyes was restricted to both outer segments and synaptic terminals of short and long/middle cone photoreceptors, whereas Pde6h(-/-) retinae remained PDE6H-negative. Combined in vivo assessment of retinal morphology with histomorphological analyses revealed a normal overall integrity of the retinal organization and an unaltered distribution of the different cone photoreceptor subtypes upon Pde6h ablation. In contrast to human patients, our electroretinographic examinations of Pde6h(-/-) mice suggest no defects in cone/rod-driven retinal signaling and therefore preserved visual functions. To this end, we were able to demonstrate the presence of rod PDE6G in cones indicating functional substitution of PDE6. The disparities between human and murine phenotypes caused by mutant Pde6h/PDE6H suggest species-to-species differences in the vulnerability of biochemical and neurosensory pathways of the visual signal transduction system.
Collapse
Affiliation(s)
- Christina Brennenstuhl
- From the Institute of Pharmacy, Department of Pharmacology, Toxicology and Clinical Pharmacy
| | | | - Markus Burkard
- From the Institute of Pharmacy, Department of Pharmacology, Toxicology and Clinical Pharmacy
| | - Rebecca Wagner
- From the Institute of Pharmacy, Department of Pharmacology, Toxicology and Clinical Pharmacy
| | | | | | - Clement Kabagema-Bilan
- From the Institute of Pharmacy, Department of Pharmacology, Toxicology and Clinical Pharmacy
| | | | | | | | | | - Peter Ruth
- From the Institute of Pharmacy, Department of Pharmacology, Toxicology and Clinical Pharmacy
| | - Bernd Wissinger
- the Molecular Genetics Laboratory, Centre for Ophthalmology, University of Tuebingen, 72076 Tuebingen, Germany
| | - Robert Lukowski
- From the Institute of Pharmacy, Department of Pharmacology, Toxicology and Clinical Pharmacy,
| |
Collapse
|
25
|
Mathur P, Yang J. Usher syndrome: Hearing loss, retinal degeneration and associated abnormalities. Biochim Biophys Acta Mol Basis Dis 2014; 1852:406-20. [PMID: 25481835 DOI: 10.1016/j.bbadis.2014.11.020] [Citation(s) in RCA: 222] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 11/25/2014] [Accepted: 11/26/2014] [Indexed: 02/06/2023]
Abstract
Usher syndrome (USH), clinically and genetically heterogeneous, is the leading genetic cause of combined hearing and vision loss. USH is classified into three types, based on the hearing and vestibular symptoms observed in patients. Sixteen loci have been reported to be involved in the occurrence of USH and atypical USH. Among them, twelve have been identified as causative genes and one as a modifier gene. Studies on the proteins encoded by these USH genes suggest that USH proteins interact among one another and function in multiprotein complexes in vivo. Although their exact functions remain enigmatic in the retina, USH proteins are required for the development, maintenance and function of hair bundles, which are the primary mechanosensitive structure of inner ear hair cells. Despite the unavailability of a cure, progress has been made to develop effective treatments for this disease. In this review, we focus on the most recent discoveries in the field with an emphasis on USH genes, protein complexes and functions in various tissues as well as progress toward therapeutic development for USH.
Collapse
Affiliation(s)
- Pranav Mathur
- Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah, Salt Lake City, UT 84132, USA; Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84132, USA
| | - Jun Yang
- Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah, Salt Lake City, UT 84132, USA; Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84132, USA; Department of Otolaryngology Head and Neck Surgery, University of Utah, Salt Lake City, UT 84132, USA.
| |
Collapse
|
26
|
Ogun O, Zallocchi M. Clarin-1 acts as a modulator of mechanotransduction activity and presynaptic ribbon assembly. ACTA ACUST UNITED AC 2014; 207:375-91. [PMID: 25365995 PMCID: PMC4226736 DOI: 10.1083/jcb.201404016] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Clarin-1 is a four-transmembrane protein expressed by hair cells and photoreceptors. Mutations in its corresponding gene are associated with Usher syndrome type 3, characterized by late-onset and progressive hearing and vision loss in humans. Mice carrying mutations in the clarin-1 gene have hair bundle dysmorphology and a delay in synapse maturation. In this paper, we examined the expression and function of clarin-1 in zebrafish hair cells. We observed protein expression as early as 1 d postfertilization. Knockdown of clarin-1 resulted in inhibition of FM1-43 incorporation, shortening of the kinocilia, and mislocalization of ribeye b clusters. These phenotypes were fully prevented by co-injection with clarin-1 transcript, requiring its C-terminal tail. We also observed an in vivo interaction between clarin-1 and Pcdh15a. Altogether, our results suggest that clarin-1 is functionally important for mechanotransduction channel activity and for proper localization of synaptic components, establishing a critical role for clarin-1 at the apical and basal poles of hair cells.
Collapse
Affiliation(s)
- Oluwatobi Ogun
- Sensory Neuroscience Department, Boys Town National Research Hospital, Omaha, NE 68131
| | - Marisa Zallocchi
- Sensory Neuroscience Department, Boys Town National Research Hospital, Omaha, NE 68131
| |
Collapse
|
27
|
The retinal phenotype of Usher syndrome: pathophysiological insights from animal models. C R Biol 2014; 337:167-77. [PMID: 24702843 DOI: 10.1016/j.crvi.2013.12.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 12/03/2013] [Indexed: 01/26/2023]
Abstract
The Usher syndrome (USH) is the most prevalent cause of inherited deaf-blindness. Three clinical subtypes, USH1-3, have been defined, and ten USH genes identified. The hearing impairment due to USH gene defects has been shown to result from improper organisation of the hair bundle, the sound receptive structure of sensory hair cells. In contrast, the cellular basis of the visual defect is less well understood as this phenotype is absent in almost all the USH mouse models that faithfully mimic the human hearing impairment. Structural and molecular interspecies discrepancies regarding photoreceptor calyceal processes and the association with the distribution of USH1 proteins have recently been unravelled, and have led to the conclusion that a defect in the USH1 protein complex-mediated connection between the photoreceptor outer segment and the surrounding calyceal processes (in both rods and cones), and the inner segment (in rods only), probably causes the USH1 retinal dystrophy in humans.
Collapse
|
28
|
Wahlin KJ, Maruotti J, Zack DJ. Modeling retinal dystrophies using patient-derived induced pluripotent stem cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 801:157-64. [PMID: 24664693 DOI: 10.1007/978-1-4614-3209-8_20] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Retinal degenerative disease involving photoreceptor (PR) cell loss results in permanent vision loss and often blindness. Generation of induced pluripotent stem cell (iPSC)-derived retinal cells and tissues from individuals with retinal dystrophies is a relatively new and promising method for studying retinal degeneration mechanisms in vitro. Recent advancements in strategies to differentiate human iPSCs (hiPSCs) into 3D retinal eyecups with a strong resemblance to the mature retina raise the possibility that this system could offer a reliable model for translational drug studies. However, despite the potential benefits, there are challenges that remain to be overcome before stem-cell-derived retinal eyecups can be routinely used to model human retinal diseases. This chapter will discuss both the potential of these 3D eyecup approaches and the nature of some of the challenges that remain.
Collapse
Affiliation(s)
- Karl J Wahlin
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Smith Building Rm 3029, 400 N Broadway, Baltimore, MD, USA,
| | | | | |
Collapse
|
29
|
Han J, Dinculescu A, Dai X, Du W, Smith WC, Pang J. Review: the history and role of naturally occurring mouse models with Pde6b mutations. Mol Vis 2013; 19:2579-89. [PMID: 24367157 PMCID: PMC3869645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 12/18/2013] [Indexed: 11/08/2022] Open
Abstract
Mouse models are useful tools for developing potential therapies for human inherited retinal diseases, such as retinitis pigmentosa (RP), since more strains are being identified with the same mutant genes and phenotypes as humans with corresponding retinal degenerative diseases. Mutations in the beta subunit of the human rod phosphodiesterase (PDE6B) gene are a common cause of autosomal recessive RP (arRP). This article focuses on two well-established naturally occurring mouse models of arRP caused by spontaneous mutations in Pde6b, their discovery, phenotype, mechanism of degeneration, strengths and limitations, and therapeutic approaches to restore vision and delay disease progression. Viral vector, especially adeno-associated viral vector (AAV) -mediated gene replacement therapy, pharmacological treatment, cell-based therapy and other approaches that extend the therapeutic window of treatment, is a potentially promising strategy for improving photoreceptor function and significantly slowing the process of retinal degeneration.
Collapse
Affiliation(s)
- Juanjuan Han
- Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Astra Dinculescu
- Department of Ophthalmology, University of Florida, Gainesville, FL
| | - Xufeng Dai
- Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wei Du
- Department of Ophthalmology, University of Florida, Gainesville, FL
| | - W. Clay Smith
- Department of Ophthalmology, University of Florida, Gainesville, FL
| | - Jijing Pang
- Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China,Department of Ophthalmology, University of Florida, Gainesville, FL
| |
Collapse
|
30
|
Phillips JB, Västinsalo H, Wegner J, Clément A, Sankila EM, Westerfield M. The cone-dominant retina and the inner ear of zebrafish express the ortholog of CLRN1, the causative gene of human Usher syndrome type 3A. Gene Expr Patterns 2013; 13:473-81. [PMID: 24045267 PMCID: PMC3888827 DOI: 10.1016/j.gep.2013.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 09/05/2013] [Accepted: 09/06/2013] [Indexed: 01/13/2023]
Abstract
Clarin-1 (CLRN1) is the causative gene in Usher syndrome type 3A, an autosomal recessive disorder characterized by progressive vision and hearing loss. CLRN1 encodes Clarin-1, a glycoprotein with homology to the tetraspanin family of proteins. Previous cell culture studies suggest that Clarin-1 localizes to the plasma membrane and interacts with the cytoskeleton. Mouse models demonstrate a role for the protein in mechanosensory hair bundle integrity, but the function of Clarin-1 in hearing remains unclear. Even less is known of its role in vision, because the Clrn1 knockout mouse does not exhibit a retinal phenotype and expression studies in murine retinas have provided conflicting results. Here, we describe cloning and expression analysis of the zebrafish clrn1 gene, and report protein localization of Clarin-1 in auditory and visual cells from embryonic through adult stages. We detect clrn1 transcripts as early as 24h post-fertilization, and expression is maintained through adulthood. In situ hybridization experiments show clrn1 transcripts enriched in mechanosensory hair cells and supporting cells of the inner ear and lateral line organ, photoreceptors, and cells of the inner retina. In mechanosensory hair cells, Clarin-1 is polarized to the apical cell body and the synapses. In the retina, Clarin-1 localizes to lateral cell contacts between photoreceptors and is associated with the outer limiting membrane and subapical processes emanating from Müller glial cells. We also find Clarin-1 protein in the outer plexiform, inner nuclear and ganglion cell layers of the retina. Given the importance of Clarin-1 function in the human retina, it is imperative to find an animal model with a comparable requirement. Our data provide a foundation for exploring the role of Clarin-1 in retinal cell function and survival in a diurnal, cone-dominant species.
Collapse
Affiliation(s)
- Jennifer B Phillips
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR 97403-1254, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Novack GD. Translating Drugs From Animals to Humans: Do We Need to Prove Efficacy? Transl Vis Sci Technol 2013; 2:1. [PMID: 24078898 PMCID: PMC3783258 DOI: 10.1167/tvst.2.6.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 08/02/2013] [Indexed: 01/30/2023] Open
Affiliation(s)
- Gary D Novack
- PharmaLogic Development, Inc., San Rafael, California
| |
Collapse
|
32
|
Piret G, Perez MT, Prinz CN. Neurite outgrowth and synaptophysin expression of postnatal CNS neurons on GaP nanowire arrays in long-term retinal cell culture. Biomaterials 2012; 34:875-87. [PMID: 23131535 DOI: 10.1016/j.biomaterials.2012.10.042] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Accepted: 10/16/2012] [Indexed: 12/18/2022]
Abstract
We have established long-term cultures of postnatal retinal cells on arrays of gallium phosphide nanowires of different geometries. Rod and cone photoreceptors, ganglion cells and bipolar cells survived on the substrates for at least 18 days in vitro. Glial cells were also observed, but these did not overgrow the neuronal population. On nanowires, neurons extended numerous long and branched neurites that expressed the synaptic vesicle marker synaptophysin. The longest nanowires (4 μm long) allowed a greater attachment and neurite elongation and our analysis suggests that the length of the nanowire per se and/or the adsorption of biomolecules on the nanowires may have been important factors regulating the observed cell behavior. The study thus shows that CNS neurons are amenable to gallium phosphide nanowires, probably as they create conditions that more closely resemble those encountered in the in vivo environment. These findings suggest that gallium phosphide nanowires may be considered as a material of interest when improving existing or designing the next generation of implantable devices. The features of gallium phosphide nanowires can be precisely controlled, making them suitable for this purpose.
Collapse
Affiliation(s)
- Gaëlle Piret
- Division of Solid State Physics, The Nanometer Structure Consortium, Lund University, Sweden
| | | | | |
Collapse
|
33
|
Estrada-Cuzcano A, Roepman R, Cremers FPM, den Hollander AI, Mans DA. Non-syndromic retinal ciliopathies: translating gene discovery into therapy. Hum Mol Genet 2012; 21:R111-24. [PMID: 22843501 DOI: 10.1093/hmg/dds298] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Homozygosity mapping and exome sequencing have accelerated the discovery of gene mutations and modifier alleles implicated in inherited retinal degeneration in humans. To date, 158 genes have been found to be mutated in individuals with retinal dystrophies. Approximately one-third of the gene defects underlying retinal degeneration affect the structure and/or function of the 'connecting cilium' in photoreceptors. This structure corresponds to the transition zone of a prototypic cilium, a region with increasing relevance for ciliary homeostasis. The connecting cilium connects the inner and outer segments of the photoreceptor, mediating bi-directional transport of phototransducing proteins required for vision. In fact, the outer segment, connecting cilium and associated basal body, forms a highly specialized sensory cilium, fully dedicated to photoreception and subsequent signal transduction to the brain. At least 21 genes that encode ciliary proteins are implicated in non-syndromic retinal dystrophies such as cone dystrophy, cone-rod dystrophy, Leber congenital amaurosis (LCA), macular degeneration or retinitis pigmentosa (RP). The generation and characterization of vertebrate retinal ciliopathy animal models have revealed insights into the molecular disease mechanism which are indispensable for the development and evaluation of therapeutic strategies. Gene augmentation therapy has proven to be safe and successful in restoring long-term sight in mice, dogs and humans suffering from LCA or RP. Here, we present a comprehensive overview of the genes, mutations and modifier alleles involved in non-syndromic retinal ciliopathies, review the progress in dissecting the associated retinal disease mechanisms and evaluate gene augmentation approaches to antagonize retinal degeneration in these ciliopathies.
Collapse
|
34
|
Zallocchi M, Meehan DT, Delimont D, Rutledge J, Gratton MA, Flannery J, Cosgrove D. Role for a novel Usher protein complex in hair cell synaptic maturation. PLoS One 2012; 7:e30573. [PMID: 22363448 PMCID: PMC3281840 DOI: 10.1371/journal.pone.0030573] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 12/22/2011] [Indexed: 12/22/2022] Open
Abstract
The molecular mechanisms underlying hair cell synaptic maturation are not well understood. Cadherin-23 (CDH23), protocadherin-15 (PCDH15) and the very large G-protein coupled receptor 1 (VLGR1) have been implicated in the development of cochlear hair cell stereocilia, while clarin-1 has been suggested to also play a role in synaptogenesis. Mutations in CDH23, PCDH15, VLGR1 and clarin-1 cause Usher syndrome, characterized by congenital deafness, vestibular dysfunction and retinitis pigmentosa. Here we show developmental expression of these Usher proteins in afferent spiral ganglion neurons and hair cell synapses. We identify a novel synaptic Usher complex comprised of clarin-1 and specific isoforms of CDH23, PCDH15 and VLGR1. To establish the in vivo relevance of this complex, we performed morphological and quantitative analysis of the neuronal fibers and their synapses in the Clrn1−/− mouse, which was generated by incomplete deletion of the gene. These mice showed a delay in neuronal/synaptic maturation by both immunostaining and electron microscopy. Analysis of the ribbon synapses in Ames waltzerav3J mice also suggests a delay in hair cell synaptogenesis. Collectively, these results show that, in addition to the well documented role for Usher proteins in stereocilia development, Usher protein complexes comprised of specific protein isoforms likely function in synaptic maturation as well.
Collapse
Affiliation(s)
- Marisa Zallocchi
- Boys Town National Research Hospital, Omaha, Nebraska, United States of America
| | - Daniel T. Meehan
- Boys Town National Research Hospital, Omaha, Nebraska, United States of America
| | - Duane Delimont
- Boys Town National Research Hospital, Omaha, Nebraska, United States of America
| | - Joseph Rutledge
- Otolaryngology-Head, Neck Surgery, St Louis University, St Louis, Missouri, United States of America
| | - Michael Anne Gratton
- Otolaryngology-Head, Neck Surgery, St Louis University, St Louis, Missouri, United States of America
| | - John Flannery
- Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States of America
| | - Dominic Cosgrove
- Boys Town National Research Hospital, Omaha, Nebraska, United States of America
- University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail:
| |
Collapse
|
35
|
Zallocchi M, Meehan DT, Delimont D, Rutledge J, Gratton MA, Flannery J, Cosgrove D. Role for a novel Usher protein complex in hair cell synaptic maturation. PLoS One 2012; 7:e30573. [PMID: 22363448 DOI: 10.1371/journal.pone.0030573pone-d-11-06651[pii]] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 12/22/2011] [Indexed: 05/19/2023] Open
Abstract
The molecular mechanisms underlying hair cell synaptic maturation are not well understood. Cadherin-23 (CDH23), protocadherin-15 (PCDH15) and the very large G-protein coupled receptor 1 (VLGR1) have been implicated in the development of cochlear hair cell stereocilia, while clarin-1 has been suggested to also play a role in synaptogenesis. Mutations in CDH23, PCDH15, VLGR1 and clarin-1 cause Usher syndrome, characterized by congenital deafness, vestibular dysfunction and retinitis pigmentosa. Here we show developmental expression of these Usher proteins in afferent spiral ganglion neurons and hair cell synapses. We identify a novel synaptic Usher complex comprised of clarin-1 and specific isoforms of CDH23, PCDH15 and VLGR1. To establish the in vivo relevance of this complex, we performed morphological and quantitative analysis of the neuronal fibers and their synapses in the Clrn1-/- mouse, which was generated by incomplete deletion of the gene. These mice showed a delay in neuronal/synaptic maturation by both immunostaining and electron microscopy. Analysis of the ribbon synapses in Ames waltzer(av3J) mice also suggests a delay in hair cell synaptogenesis. Collectively, these results show that, in addition to the well documented role for Usher proteins in stereocilia development, Usher protein complexes comprised of specific protein isoforms likely function in synaptic maturation as well.
Collapse
Affiliation(s)
- Marisa Zallocchi
- Boys Town National Research Hospital, Omaha, Nebraska, United States of America
| | | | | | | | | | | | | |
Collapse
|
36
|
Zou J, Luo L, Shen Z, Chiodo VA, Ambati BK, Hauswirth WW, Yang J. Whirlin replacement restores the formation of the USH2 protein complex in whirlin knockout photoreceptors. Invest Ophthalmol Vis Sci 2011; 52:2343-51. [PMID: 21212183 DOI: 10.1167/iovs.10-6141] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Whirlin is the causative gene for Usher syndrome type IID (USH2D), a condition manifested as both retinitis pigmentosa and congenital deafness. Mutations in this gene cause disruption of the USH2 protein complex composed of USH2A and VLGR1 at the periciliary membrane complex (PMC) in photoreceptors. In this study, the adeno-associated virus (AAV)-mediated whirlin replacement was evaluated as a treatment option. METHODS Murine whirlin cDNA driven by the human rhodopsin kinase promoter (hRK) was packaged as an AAV2/5 vector and delivered into the whirlin knockout retina through subretinal injection. The efficiency, efficacy, and safety of this treatment were examined using immunofluorescent staining, confocal imaging, immunoelectron microscopy, Western blot analysis, histologic analysis, and electroretinogram. RESULTS The AAV-mediated whirlin expression started at two weeks, reached its maximum level at 10 weeks, and lasted up to six months post injection. The transgenic whirlin product had a molecular size and an expression level comparable to the wild-type. It was distributed at the PMC in both rod and cone photoreceptors from the central to peripheral retina. Importantly, the transgenic whirlin restored the cellular localization and expression level of both USH2A and VLGR1 and did not cause defects in the retinal histology and function in the whirlin knockout mouse. CONCLUSIONS Whirlin transgene recruits USH2A and VLGR1 to the PMC and is sufficient for the formation of the USH2 protein complex in photoreceptors. The combined hRK and AAV gene delivery system could be an effective gene therapy approach to treat retinal degeneration in USH2D patients.
Collapse
Affiliation(s)
- Junhuang Zou
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah, Salt Lake City, Utah 84132, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Clarin 1 (CLRN1) is a four-transmembrane protein expressed in cochlear hair cells and neural retina, and when mutated it causes Usher syndrome type 3 (USH3). The main human splice variant of CLRN1 is composed of three exons that code for a 232-aa protein. In this study, we aimed to refine the structure of CLRN1 by an examination of transcript splice variants and promoter regions. Analysis of human retinal cDNA revealed 11 CLRN1 splice variants, of which 5 have not been previously reported. We studied the regulation of gene expression by several promoter domains using a luciferase assay, and identified 1000 nt upstream of the translation start site of the primary CLRN1 splice variant as the principal promoter region. Our results suggest that the CLRN1 gene is significantly more complex than previously described. The complexity of the CLRN1 gene and the identification of multiple splice variants may partially explain why mutations in CLRN1 result in substantial variation in clinical phenotype.
Collapse
|
38
|
Yang J, Liu X, Zhao Y, Adamian M, Pawlyk B, Sun X, McMillan DR, Liberman MC, Li T. Ablation of whirlin long isoform disrupts the USH2 protein complex and causes vision and hearing loss. PLoS Genet 2010; 6:e1000955. [PMID: 20502675 PMCID: PMC2873905 DOI: 10.1371/journal.pgen.1000955] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Accepted: 04/19/2010] [Indexed: 12/25/2022] Open
Abstract
Mutations in whirlin cause either Usher syndrome type II (USH2), a deafness-blindness disorder, or nonsyndromic deafness. The molecular basis for the variable disease expression is unknown. We show here that only the whirlin long isoform, distinct from a short isoform by virtue of having two N-terminal PDZ domains, is expressed in the retina. Both long and short isoforms are expressed in the inner ear. The N-terminal PDZ domains of the long whirlin isoform mediates the formation of a multi-protein complex that includes usherin and VLGR1, both of which are also implicated in USH2. We localized this USH2 protein complex to the periciliary membrane complex (PMC) in mouse photoreceptors that appears analogous to the frog periciliary ridge complex. The latter is proposed to play a role in photoreceptor protein trafficking through the connecting cilium. Mice carrying a targeted disruption near the N-terminus of whirlin manifest retinal and inner ear defects, reproducing the clinical features of human USH2 disease. This is in contrast to mice with mutations affecting the C-terminal portion of whirlin in which the phenotype is restricted to the inner ear. In mice lacking any one of the USH2 proteins, the normal localization of all USH2 proteins is disrupted, and there is evidence of protein destabilization. Taken together, our findings provide new insights into the pathogenic mechanism of Usher syndrome. First, the three USH2 proteins exist as an obligatory functional complex in vivo, and loss of one USH2 protein is functionally close to loss of all three. Second, defects in the three USH2 proteins share a common pathogenic process, i.e., disruption of the PMC. Third, whirlin mutations that ablate the N-terminal PDZ domains lead to Usher syndrome, but non-syndromic hearing loss will result if they are spared.
Collapse
Affiliation(s)
- Jun Yang
- The Berman-Gund Laboratory for the Study of Retinal Degenerations, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States of America
| | - Xiaoqing Liu
- The Berman-Gund Laboratory for the Study of Retinal Degenerations, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States of America
| | - Yun Zhao
- The Berman-Gund Laboratory for the Study of Retinal Degenerations, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States of America
| | - Michael Adamian
- The Berman-Gund Laboratory for the Study of Retinal Degenerations, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States of America
| | - Basil Pawlyk
- The Berman-Gund Laboratory for the Study of Retinal Degenerations, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States of America
| | - Xun Sun
- The Berman-Gund Laboratory for the Study of Retinal Degenerations, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States of America
| | - D. Randy McMillan
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - M. Charles Liberman
- Department of Otology and Laryngology, Harvard Medical School and Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States of America
| | - Tiansen Li
- The Berman-Gund Laboratory for the Study of Retinal Degenerations, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States of America
| |
Collapse
|
39
|
Cosgrove D, Zallocchi M. Clarin-1 protein expression in photoreceptors. Hear Res 2009; 259:117. [PMID: 19772908 DOI: 10.1016/j.heares.2009.09.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Accepted: 09/16/2009] [Indexed: 11/26/2022]
|