1
|
Foster T, Lim P, Wagle SR, Ionescu CM, Kovacevic B, McLenachan S, Carvalho L, Brunet A, Mooranian A, Al-Salami H. Nanoparticle-Based gene therapy strategies in retinal delivery. J Drug Target 2025; 33:508-527. [PMID: 39749456 DOI: 10.1080/1061186x.2024.2433563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/31/2024] [Accepted: 11/17/2024] [Indexed: 01/04/2025]
Abstract
Vision loss and blindness are significant issues in both developed and developing countries. There are a wide variety of aetiologies that can cause vision loss, which are outlined in this review. Although treatment has significantly improved over time for some conditions, nearly half of all people with vision impairment are left untreated. Gene delivery is an emerging field that may assist with the treatment of some of these difficult to manage forms of vision loss. Here we review how a component of nanotechnology-based, non-viral gene delivery systems are being applied to help resolve vision impairment. This review focuses on the use of lipid and polymer nanoparticles, and quantum dots as gene delivery vectors to the eye. Finally, we also highlight some emerging technologies that may be useful in this discipline.
Collapse
Affiliation(s)
- Thomas Foster
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
- Department of Clinical Biochemistry, Pathwest Laboratory Medicine, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Patrick Lim
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Susbin Raj Wagle
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Corina Mihaela Ionescu
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Bozica Kovacevic
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Samuel McLenachan
- Centre for Ophthalmology and Visual Science (incorporating the Lions Eye Institute), The University of Western Australia, Crawley, Western Australia, Australia
| | - Livia Carvalho
- Centre for Ophthalmology and Visual Science (incorporating the Lions Eye Institute), The University of Western Australia, Crawley, Western Australia, Australia
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Alicia Brunet
- Centre for Ophthalmology and Visual Science (incorporating the Lions Eye Institute), The University of Western Australia, Crawley, Western Australia, Australia
| | - Armin Mooranian
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
- School of Pharmacy, University of Otago, Dunedin, Otago, New Zealand
| | - Hani Al-Salami
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
- Medical School, The University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
2
|
Miller C, Bohnsack BL, Drackley A, Ing A, Rahmani S, Ralay Ranaivo H, Allegretti V, Rossen JL. Genetic testing results of retinal dystrophies in a diverse population: impact of race and ethnicity. Ophthalmic Genet 2025; 46:160-165. [PMID: 39763302 DOI: 10.1080/13816810.2024.2446549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 09/26/2024] [Accepted: 11/20/2024] [Indexed: 04/17/2025]
Abstract
INTRODUCTION Due to the recent advent of gene-targeted retinal therapies, the clinical value of high-yield genetic testing for inherited retinal dystrophies (IRDs) has increased considerably. However, diagnostic yield is limited by the reported patient populations in allele frequency databases. This study aimed to determine the effect of race and ethnicity on diagnostic yield in IRDs. METHODS Retrospective review of individuals with suspected IRD based on clinical findings or diagnosis of associated syndrome who underwent genetic testing between 2009 and 2021. Self-reported race and ethnicity, ophthalmic examination findings, ERG results, and genetic testing findings were collected and analyzed. RESULTS In 93 individuals (90 families) with suspected IRD, the diagnostic yield was 72% (67 individuals). The rate of diagnostic yield was not significantly associated with family history, associated syndromes, age at testing, ERG results, or ophthalmic exam findings. Further, higher rates of positive diagnostic yield were not associated with more recent genetic testing. There was a trend toward differences in diagnostic yield between races (77% White, 65% Other, 64% Asian, 50% Black) and ratio of pathogenic/likely pathogenic (P/LP) variants to variants of unknown significance (46%:54% White, 36%:64% Other, 31%:69% Asian, 30%:70% Black). CONCLUSIONS Current genetic testing for IRDs trends toward higher diagnostic yield and identification of P/LP variants in patients identifying as White compared to other races. In order to prevent negative impacts on access to gene-targeted trials and treatments for non-White patients, wider genetic testing in diverse populations is required to create comprehensive catalogs of gene variants associated with IRDs.
Collapse
Affiliation(s)
- Charles Miller
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Brenda L Bohnsack
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Division of Ophthalmology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - Andy Drackley
- Division of Genetics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - Alexander Ing
- Division of Genetics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - Safa Rahmani
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Division of Ophthalmology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - Hantamalala Ralay Ranaivo
- Division of Ophthalmology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - Valerie Allegretti
- Division of Genetics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - Jennifer L Rossen
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Division of Ophthalmology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| |
Collapse
|
3
|
Kaminer Abargel M, Macarov M, Hendler K, Yahalom C. The importance of genetic counselling and testing in inherited eye diseases: A population-based retrospective study. PLoS One 2025; 20:e0318492. [PMID: 39946424 PMCID: PMC11825002 DOI: 10.1371/journal.pone.0318492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 01/16/2025] [Indexed: 02/16/2025] Open
Abstract
PURPOSE Inherited eye diseases (IEDs) are among the main causes of visual impairment and blindness in children and young people worldwide. The objective of our study was to characterize the prevalence and distribution of the most common IEDs and causative genes in our population. STUDY DESIGN Retrospective study based on medical records of patients with IEDs who underwent genetic counselling through our multidisciplinary low vision center from 2018 to 2020. METHODS Data retrieved from medical files included: year of consultation, age, gender, ethnicity, diagnosis, gene variants and mode of inheritance. RESULTS 228 patients were included in our study. The most common diagnoses were inherited retinal diseases (IRDs) (41.2%) and albinism (32%). In 2018 and 2019 the number of patients reaching out for genetic counselling was between 50 and 60; this number doubled by 2020. The rate of pathogenic variant detection was 65.3%. The most common genes identified were TYR (29.2%), OCA2 (7.9%), ABCA4 (5.3%), TRPM1 (5.3%) and USH2A (4.4%). CONCLUSION Genetic counselling and testing became an essential part of caregiving for patients and families affected by these severe IEDs. The most common IEDs were IRDs in the Muslim population and albinism in the Jewish population. Pathogenic variants in the TYR gene were the most common in our cohort, OCA2 gene was the second in frequency, followed by ABCA4, TRPM1 and USH2A genes. We detected an increasing trend over the studied time in the number of patients reaching out for genetic counselling.
Collapse
Affiliation(s)
| | - Michal Macarov
- Department of Ophthalmology, Hadassah Medical Center, Jerusalem, Israel
| | - Karen Hendler
- Department of Ophthalmology, Hadassah Medical Center, Jerusalem, Israel
| | - Claudia Yahalom
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Ophthalmology, Hadassah Medical Center, Jerusalem, Israel
| |
Collapse
|
4
|
Tharmarajah B, Cornish EE, Nguyen J, Barnes E, Leahy KE, Vaze A, Jamieson RV, Grigg JR. Hardy-Rand-Rittler colour vision testing in cone and cone-rod dystrophies: correlation with structural and functional outcome measures. Eye (Lond) 2025; 39:527-532. [PMID: 39821145 PMCID: PMC11794465 DOI: 10.1038/s41433-024-03584-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 11/22/2024] [Accepted: 12/20/2024] [Indexed: 01/19/2025] Open
Abstract
PURPOSE To determine how Hardy-Rand-Rittler (HRR) colour vision testing correlates with visual functional and structural assessments in Cone and Cone-Rod Dystrophy. METHODS Thirty-four Cone and 69 Cone-Rod Dystrophy patients diagnosed by electroretinography (ERG) at the Save Sight Institute in Sydney were included in a retrospective analysis. Each patient's HRR colour vision test scores were compared with markers of cone and rod system function including visual acuity (VA), ERG responses, changes on Spectral Domain Optical Coherence Tomography (OCT) and Fundus Autofluorescence. RESULTS The number of plates identified on HRR testing correlated with logMAR best-corrected distance VA; r(101) = -0.49, p < 0.0001. HRR scores correlated with markers of cone and macula function including OCT Ellipsoid Zone Gap Width, Central Macular and Outer Nuclear Layer Thickness, Full Field ERG 30 Hz flicker amplitudes, light adapted 3.0 b-wave amplitudes and Pattern ERG 15- and 30-degree p50 amplitudes. CONCLUSION HRR colour vision testing correlates with structural and functional measures in Cone and Cone-Rod Dystrophy. HRR colour vision testing provides a simple clinic-based option to monitor disease changes in Cone and Cone-Rod Dystrophy patients, especially when ERG testing is not available.
Collapse
Affiliation(s)
- Brindhan Tharmarajah
- Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Sydney Eye Hospital, Sydney, NSW, Australia
| | - Elisa E Cornish
- Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Sydney Eye Hospital, Sydney, NSW, Australia
- Eye Genetics Research Unit, Children's Medical Research Institute, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Jonathan Nguyen
- Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Sydney Eye Hospital, Sydney, NSW, Australia
| | - Elizabeth Barnes
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Kate E Leahy
- Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Sydney Eye Hospital, Sydney, NSW, Australia
- Eye Genetics Research Unit, Children's Medical Research Institute, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Anagha Vaze
- Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Sydney Eye Hospital, Sydney, NSW, Australia
- Eye Genetics Research Unit, Children's Medical Research Institute, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Robyn V Jamieson
- Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Eye Genetics Research Unit, Children's Medical Research Institute, The Children's Hospital at Westmead, Sydney, NSW, Australia
- Disciplines of Genomic Medicine & Child and Adolescent Health, The University of Sydney, Sydney, NSW, Australia
| | - John R Grigg
- Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
- Sydney Eye Hospital, Sydney, NSW, Australia.
- Eye Genetics Research Unit, Children's Medical Research Institute, The Children's Hospital at Westmead, Sydney, NSW, Australia.
| |
Collapse
|
5
|
Barabino A, Mellal K, Hamam R, Polosa A, Griffith M, Bouchard JF, Kalevar A, Hanna R, Bernier G. Molecular characterization and sub-retinal transplantation of hypoimmunogenic human retinal sheets in a minipig model of severe photoreceptor degeneration. Development 2024; 151:dev203071. [PMID: 39633598 DOI: 10.1242/dev.203071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 10/08/2024] [Indexed: 12/07/2024]
Abstract
Retinal degenerative diseases affect millions of people worldwide, and legal blindness is generally associated with the loss of cone photoreceptors located in the central region of the retina called the macula. Currently, there is no treatment to replace the macula. Addressing this unmet need, we employed control isogenic and hypoimmunogenic induced pluripotent stem cell lines to generate spontaneously polarized retinal sheets (RSs). RSs were enriched in retinal progenitor and cone precursor cells, which could differentiate into mature S- and M/L-cones in long-term cultures. Single-cell RNA-seq analysis showed that RSs recapitulate the ontogeny of the developing human retina. Isolation of neural rosettes for sub-retinal transplantation effectively eliminated unwanted cells such as RPE cells. In a porcine model of chemically induced retinal degeneration, grafts integrated the host retina and formed a new, yet immature, photoreceptor layer. In one transplanted animal, functional and immunohistochemical assays suggest that grafts exhibited responsiveness to light stimuli and established putative synaptic connections with host bipolar neurons. This study underscores the potential and challenges of RSs for clinical applications.
Collapse
Affiliation(s)
- Andrea Barabino
- Stem Cell and Developmental Biology Laboratory, Hôpital Maisonneuve-Rosemont, 5690 Boul. Rosemont, Montreal, QC H1T 2H2, Canada
| | - Katia Mellal
- Stem Cell and Developmental Biology Laboratory, Hôpital Maisonneuve-Rosemont, 5690 Boul. Rosemont, Montreal, QC H1T 2H2, Canada
| | - Rimi Hamam
- Stem Cell and Developmental Biology Laboratory, Hôpital Maisonneuve-Rosemont, 5690 Boul. Rosemont, Montreal, QC H1T 2H2, Canada
| | - Anna Polosa
- Stem Cell and Developmental Biology Laboratory, Hôpital Maisonneuve-Rosemont, 5690 Boul. Rosemont, Montreal, QC H1T 2H2, Canada
| | - May Griffith
- Department of Ophthalmology, University of Montreal, Montreal, QC H3T 1J4, Canada
| | | | - Ananda Kalevar
- Department of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Roy Hanna
- Stem Cell and Developmental Biology Laboratory, Hôpital Maisonneuve-Rosemont, 5690 Boul. Rosemont, Montreal, QC H1T 2H2, Canada
| | - Gilbert Bernier
- Stem Cell and Developmental Biology Laboratory, Hôpital Maisonneuve-Rosemont, 5690 Boul. Rosemont, Montreal, QC H1T 2H2, Canada
- Department of Neurosciences, University of Montreal, Montreal, QC H3C 3J7, Canada
| |
Collapse
|
6
|
Zhu V, Huang T, Wang D, Colville D, Mack H, Savige J. Ocular manifestations of the genetic causes of focal and segmental glomerulosclerosis. Pediatr Nephrol 2024; 39:655-679. [PMID: 37578539 PMCID: PMC10817844 DOI: 10.1007/s00467-023-06073-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 06/11/2023] [Accepted: 06/19/2023] [Indexed: 08/15/2023]
Abstract
Genetic forms of focal and segmental glomerulosclerosis (FSGS) often have extra-renal manifestations. This study examined FSGS-associated genes from the Genomics England Renal proteinuria panel for reported and likely ocular features. Thirty-two of the 55 genes (58%) were associated with ocular abnormalities in human disease, and a further 12 (22%) were expressed in the retina or had an eye phenotype in mouse models. The commonest genes affected in congenital nephrotic syndrome (NPHS1, NPHS2, WT1, LAMB2, PAX2 but not PLCE1) may have ocular manifestations . Many genes affected in childhood-adolescent onset FSGS (NPHS1, NPHS2, WT1, LAMB2, SMARCAL1, NUP107 but not TRPC6 or PLCE1) have ocular features. The commonest genes affected in adult-onset FSGS (COL4A3-COL4A5, GLA ) have ocular abnormalities but not the other frequently affected genes (ACTN4, CD2AP, INF2, TRPC6). Common ocular associations of genetic FSGS include cataract, myopia, strabismus, ptosis and retinal atrophy. Mitochondrial forms of FSGS (MELAS, MIDD, Kearn's Sayre disease) are associated with retinal atrophy and inherited retinal degeneration. Some genetic kidney diseases (CAKUT, ciliopathies, tubulopathies) that result in secondary forms of FSGS also have ocular features. Ocular manifestations suggest a genetic basis for FSGS, often help identify the affected gene, and prompt genetic testing. In general, ocular abnormalities require early evaluation by an ophthalmologist, and sometimes, monitoring or treatment to improve vision or prevent visual loss from complications. In addition, the patient should be examined for other syndromic features and first degree family members assessed.
Collapse
Affiliation(s)
- Victor Zhu
- Department of Medicine (Melbourne Health and Northern Health), Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - Tess Huang
- Department of Medicine (Melbourne Health and Northern Health), Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - David Wang
- Department of Medicine (Melbourne Health and Northern Health), Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - Deb Colville
- Department of Surgery, Royal Victorian Eye and Ear Hospital, The University of Melbourne, East Melbourne, VIC, 3002, Australia
| | - Heather Mack
- Department of Surgery, Royal Victorian Eye and Ear Hospital, The University of Melbourne, East Melbourne, VIC, 3002, Australia
| | - Judy Savige
- Department of Medicine (Melbourne Health and Northern Health), Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3050, Australia.
| |
Collapse
|
7
|
Drinking hydrogen water improves photoreceptor structure and function in retinal degeneration 6 mice. Sci Rep 2022; 12:13610. [PMID: 35948585 PMCID: PMC9365798 DOI: 10.1038/s41598-022-17903-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 08/02/2022] [Indexed: 11/08/2022] Open
Abstract
Retinitis pigmentosa (RP) is a genetically heterogeneous group of inherited retinal disorders involving the progressive dysfunction of photoreceptors and the retinal pigment epithelium, for which there is currently no treatment. The rd6 mouse is a natural model of autosomal recessive retinal degeneration. Given the known contributions of oxidative stress caused by reactive oxygen species (ROS) and selective inhibition of potent ROS peroxynitrite and OH·by H2 gas we have previously demonstrated, we hypothesized that ingestion of H2 water may delay the progression of photoreceptor death in rd6 mice. H2 mice showed significantly higher retinal thickness as compared to controls on optical coherence tomography. Histopathological and morphometric analyses revealed higher thickness of the outer nuclear layer for H2 mice than controls, as well as higher counts of opsin red/green-positive cells. RNA sequencing (RNA-seq) analysis of differentially expressed genes in the H2 group versus control group revealed 1996 genes with significantly different expressions. Gene and pathway ontology analysis showed substantial upregulation of genes responsible for phototransduction in H2 mice. Our results show that drinking water high in H2 (1.2-1.6 ppm) had neuroprotective effects and inhibited photoreceptor death in mice, and suggest the potential of H2 for the treatment of RP.
Collapse
|
8
|
Panikker P, Roy S, Ghosh A, Poornachandra B, Ghosh A. Advancing precision medicines for ocular disorders: Diagnostic genomics to tailored therapies. Front Med (Lausanne) 2022; 9:906482. [PMID: 35911417 PMCID: PMC9334564 DOI: 10.3389/fmed.2022.906482] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/29/2022] [Indexed: 11/20/2022] Open
Abstract
Successful sequencing of the human genome and evolving functional knowledge of gene products has taken genomic medicine to the forefront, soon combining broadly with traditional diagnostics, therapeutics, and prognostics in patients. Recent years have witnessed an extraordinary leap in our understanding of ocular diseases and their respective genetic underpinnings. As we are entering the age of genomic medicine, rapid advances in genome sequencing, gene delivery, genome surgery, and computational genomics enable an ever-increasing capacity to provide a precise and robust diagnosis of diseases and the development of targeted treatment strategies. Inherited retinal diseases are a major source of blindness around the world where a large number of causative genes have been identified, paving the way for personalized diagnostics in the clinic. Developments in functional genetics and gene transfer techniques has also led to the first FDA approval of gene therapy for LCA, a childhood blindness. Many such retinal diseases are the focus of various clinical trials, making clinical diagnoses of retinal diseases, their underlying genetics and the studies of natural history important. Here, we review methodologies for identifying new genes and variants associated with various ocular disorders and the complexities associated with them. Thereafter we discuss briefly, various retinal diseases and the application of genomic technologies in their diagnosis. We also discuss the strategies, challenges, and potential of gene therapy for the treatment of inherited and acquired retinal diseases. Additionally, we discuss the translational aspects of gene therapy, the important vector types and considerations for human trials that may help advance personalized therapeutics in ophthalmology. Retinal disease research has led the application of precision diagnostics and precision therapies; therefore, this review provides a general understanding of the current status of precision medicine in ophthalmology.
Collapse
Affiliation(s)
| | - Shomereeta Roy
- Grow Research Laboratory, Narayana Nethralaya Foundation, Bengaluru, India
| | - Anuprita Ghosh
- Grow Research Laboratory, Narayana Nethralaya Foundation, Bengaluru, India
| | | | - Arkasubhra Ghosh
- Grow Research Laboratory, Narayana Nethralaya Foundation, Bengaluru, India
| |
Collapse
|
9
|
Whole Genome Sequencing, Focused Assays and Functional Studies Increasing Understanding in Cryptic Inherited Retinal Dystrophies. Int J Mol Sci 2022; 23:ijms23073905. [PMID: 35409265 PMCID: PMC8999823 DOI: 10.3390/ijms23073905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/24/2022] [Accepted: 03/30/2022] [Indexed: 12/15/2022] Open
Abstract
The inherited retinal dystrophies (IRDs) are a clinically and genetically complex group of disorders primarily affecting the rod and cone photoreceptors or other retinal neuronal layers, with emerging therapies heralding the need for accurate molecular diagnosis. Targeted capture and panel-based strategies examining the partial or full exome deliver molecular diagnoses in many IRD families tested. However, approximately one in three families remain unsolved and unable to obtain personalised recurrence risk or access to new clinical trials or therapy. In this study, we investigated whole genome sequencing (WGS), focused assays and functional studies to assist with unsolved IRD cases and facilitate integration of these approaches to a broad molecular diagnostic clinical service. The WGS approach identified variants not covered or underinvestigated by targeted capture panel-based clinical testing strategies in six families. This included structural variants, with notable benefit of the WGS approach in repetitive regions demonstrated by a family with a hybrid gene and hemizygous missense variant involving the opsin genes, OPN1LW and OPN1MW. There was also benefit in investigation of the repetitive GC-rich ORF15 region of RPGR. Further molecular investigations were facilitated by focused assays in these regions. Deep intronic variants were identified in IQCB1 and ABCA4, with functional RNA based studies of the IQCB1 variant revealing activation of a cryptic splice acceptor site. While targeted capture panel-based methods are successful in achieving an efficient molecular diagnosis in a proportion of cases, this study highlights the additional benefit and clinical value that may be derived from WGS, focused assays and functional genomics in the highly heterogeneous IRDs.
Collapse
|
10
|
Nash BM, Loi TH, Fernando M, Sabri A, Robinson J, Cheng A, Eamegdool SS, Farnsworth E, Bennetts B, Grigg JR, Chung SK, Gonzalez-Cordero A, Jamieson RV. Evaluation for Retinal Therapy for RPE65 Variation Assessed in hiPSC Retinal Pigment Epithelial Cells. Stem Cells Int 2021; 2021:4536382. [PMID: 34938339 PMCID: PMC8687838 DOI: 10.1155/2021/4536382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 11/02/2021] [Indexed: 12/14/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) generated from patients and the derivative retinal cells enable the investigation of pathological and novel variants in relevant cell populations. Biallelic pathogenic variants in RPE65 cause early-onset severe retinal dystrophy (EOSRD) or Leber congenital amaurosis (LCA). Increasingly, regulatory-approved in vivo RPE65 retinal gene replacement therapy is available for patients with these clinical features, but only if they have biallelic pathological variants and sufficient viable retinal cells. In our cohort of patients, we identified siblings with early-onset severe retinal degeneration where genomic studies revealed compound heterozygous variants in RPE65, one a known pathogenic missense variant and the other a novel synonymous variant of uncertain significance. The synonymous variant was suspected to affect RNA splicing. Since RPE65 is very poorly expressed in all tissues except the retinal pigment epithelium (RPE), we generated hiPSC-derived RPE cells from the parental carrier of the synonymous variant. Sequencing of RNA obtained from hiPSC-RPE cells demonstrated heterozygous skipping of RPE65 exon 2 and the introduction of a premature stop codon in the mRNA. Minigene studies confirmed the splicing aberration. Results from this study led to reclassification of the synonymous variant to a pathogenic variant, providing the affected patients with access to RPE65 gene replacement therapy.
Collapse
Affiliation(s)
- Benjamin M. Nash
- Eye Genetics Research Unit, Sydney Children's Hospitals Network-Westmead, Save Sight Institute, Children's Medical Research Institute, University of Sydney, Sydney, New South Wales, Australia
- Specialty of Genomic Medicine, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
- Sydney Genome Diagnostics, Western Sydney Genetics Program, Sydney Children's Hospitals Network-Westmead, Sydney, New South Wales, Australia
| | - To Ha Loi
- Eye Genetics Research Unit, Sydney Children's Hospitals Network-Westmead, Save Sight Institute, Children's Medical Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Milan Fernando
- Stem Cell Medicine Group and Stem Cell and Organoid Facility, Children's Medical Research Institute, University of Sydney, Faculty of Medicine & Health, Sydney NSW, Australia
| | - Amin Sabri
- Eye Genetics Research Unit, Sydney Children's Hospitals Network-Westmead, Save Sight Institute, Children's Medical Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - James Robinson
- Department of Ophthalmology, Sydney Children's Hospitals Network-Westmead, Sydney, New South Wales, Australia
- Specialty of Ophthalmology, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Anson Cheng
- Eye Genetics Research Unit, Sydney Children's Hospitals Network-Westmead, Save Sight Institute, Children's Medical Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Steven S. Eamegdool
- Eye Genetics Research Unit, Sydney Children's Hospitals Network-Westmead, Save Sight Institute, Children's Medical Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Elizabeth Farnsworth
- Sydney Genome Diagnostics, Western Sydney Genetics Program, Sydney Children's Hospitals Network-Westmead, Sydney, New South Wales, Australia
| | - Bruce Bennetts
- Specialty of Genomic Medicine, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
- Sydney Genome Diagnostics, Western Sydney Genetics Program, Sydney Children's Hospitals Network-Westmead, Sydney, New South Wales, Australia
| | - John R. Grigg
- Eye Genetics Research Unit, Sydney Children's Hospitals Network-Westmead, Save Sight Institute, Children's Medical Research Institute, University of Sydney, Sydney, New South Wales, Australia
- Department of Ophthalmology, Sydney Children's Hospitals Network-Westmead, Sydney, New South Wales, Australia
- Specialty of Ophthalmology, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Seo-Kyung Chung
- Translational Neurogenomics Group, Kids Research, Sydney Children's Hospitals Network-Westmead, Sydney NSW, Australia
- Brain and Mind Centre, Faculty of Medicine & Health, University of Sydney, Sydney NSW, Australia
| | - Anai Gonzalez-Cordero
- Stem Cell Medicine Group and Stem Cell and Organoid Facility, Children's Medical Research Institute, University of Sydney, Faculty of Medicine & Health, Sydney NSW, Australia
| | - Robyn V. Jamieson
- Eye Genetics Research Unit, Sydney Children's Hospitals Network-Westmead, Save Sight Institute, Children's Medical Research Institute, University of Sydney, Sydney, New South Wales, Australia
- Specialty of Genomic Medicine, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
- Department of Clinical Genetics, Western Sydney Genetics Program, Sydney Children's Hospitals Network-Westmead, Sydney, New South Wales, Australia
| |
Collapse
|
11
|
Shalabi O, Nazzal Z, Natsheh M, Iriqat S, Michaelides M, Ghanem M, Aslanian A, Alswaiti Y, AlTalbishi A. Swept-source optical coherence tomography changes and visual acuity among Palestinian retinitis Pigmentosa patients: a cross-sectional study. BMC Ophthalmol 2021; 21:289. [PMID: 34320936 PMCID: PMC8320214 DOI: 10.1186/s12886-021-02047-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 07/09/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Retinitis pigmentosa (RP) is a heterogeneous group of inherited ocular diseases that result in progressive retinal degeneration. This study aims to describe different Swept-source Optical Coherence Tomographic (SS-OCT) changes in Palestinian RP patients and to explore possible correlations with Visual Acuity (VA). METHODS A cross-sectional observational study was conducted on Retinitis Pigmentosa patients diagnosed with RP in a tertiary eye hospital. Full history and ocular examination were made. SS-OCT imaging was done for all eyes assessing the presence of cystoid macular edema, epiretinal membrane, macular holes, and external limiting membrane, ellipsoid zone status. Also, central macular thickness and choroidal vascular thickness were measured. RESULTS The study was run on 161 eyes of 81 patients; 53 males and 28 females. The average age at examination was 26.1 (6-78) years. Twenty-six eyes (16.1%) were of syndromic RP patients, mostly Usher syndrome; 20 eyes (12.4%). The mean Logaritmic minimal angle of resolution (LogMAR) of Best Corrected Visual Acuity (BCVA)of the study sample was 0.66 ± 0.7. The most prevalent change was cystoid macular edema [28 eyes, (17.4%)], followed by epiretinal membrane [17eye, (10.6%)]. A macular hole was noted only in one eye (0.6%). Ellipsoid zone and external limiting membrane were absent in 55 eyes (35.0%) and 60 eyes 37.5%. Vitreous hyperreflective foci were found in 35 eyes (43.8%). LogMAR of BCVA was associated significantly with cystoid macular edema (p = 0.001), ellipsoid zone(p = 0.001), and external limiting membrane (p = 0.001). CONCLUSIONS Detailed SS-OCT assessment in Palestinian patients diagnosed with RP identified different morphologies from other populations. Cystoid macular edema and vitreous hyperreflective foci may reflect signs of early or intermediate stages of the disease. Disease progression can be monitored by measuring the length/width (area) of ellipsoid zone +/- external limiting membrane and choroidal vascular thickness, which should be evaluated serially using high-resolution OCT.
Collapse
Affiliation(s)
- Orjowan Shalabi
- St. John of Jerusalem Eye Hospital Group, East Jerusalem, 91198, Palestine
| | - Zaher Nazzal
- Department of Family and Community Medicine, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Muath Natsheh
- St. John of Jerusalem Eye Hospital Group, East Jerusalem, 91198, Palestine
| | - Salam Iriqat
- St. John of Jerusalem Eye Hospital Group, East Jerusalem, 91198, Palestine
| | - Michel Michaelides
- Department of genetics, Moorfields Eye Hospital and UCL Institute of Ophthalmology, London, UK
| | - Muyassar Ghanem
- St. John of Jerusalem Eye Hospital Group, East Jerusalem, 91198, Palestine
| | - Alice Aslanian
- St. John of Jerusalem Eye Hospital Group, East Jerusalem, 91198, Palestine
| | - Yahya Alswaiti
- St. John of Jerusalem Eye Hospital Group, East Jerusalem, 91198, Palestine
| | - Alaa AlTalbishi
- St. John of Jerusalem Eye Hospital Group, East Jerusalem, 91198, Palestine.
| |
Collapse
|
12
|
The electroretinogram in the genomics era: outer retinal disorders. Eye (Lond) 2021; 35:2406-2418. [PMID: 34234290 DOI: 10.1038/s41433-021-01659-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/03/2021] [Accepted: 06/18/2021] [Indexed: 11/08/2022] Open
Abstract
The inherited retinal diseases (IRDs) have traditionally been described phenotypically with the description evolving to incorporate more sophisticated structural and functional assessments. In the last 25 years there has been considerable advances in the understanding of underlying genetic aetiologies. The role of the ophthalmologist is now to work in a multi-disciplinary team to identify the disease-causing genotype, which might be amenable to gene-directed intervention. Visual electrophysiology is an important tool to assist the ophthalmologist in guiding the clinical geneticist to reach a final molecular diagnosis. This review outlines the physiological basis for the ISCEV standard electrophysiology tests, the role of electrophysiology in localising the functional deficit, correlation with structural findings to guide diagnosis and finally management of IRDs in the era of genomics with emphasis on the outer retina.
Collapse
|
13
|
Iancu IF, Avila-Fernandez A, Arteche A, Trujillo-Tiebas MJ, Riveiro-Alvarez R, Almoguera B, Martin-Merida I, Del Pozo-Valero M, Perea-Romero I, Corton M, Minguez P, Ayuso C. Prioritizing variants of uncertain significance for reclassification using a rule-based algorithm in inherited retinal dystrophies. NPJ Genom Med 2021; 6:18. [PMID: 33623043 PMCID: PMC7902814 DOI: 10.1038/s41525-021-00182-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Inherited retinal dystrophies (IRD) are a highly heterogeneous group of rare diseases with a molecular diagnostic rate of >50%. Reclassification of variants of uncertain significance (VUS) poses a challenge for IRD diagnosis. We collected 668 IRD cases analyzed by our geneticists using two different clinical exome-sequencing tests. We identified 114 unsolved cases pending reclassification of 125 VUS and studied their genomic, functional, and laboratory-specific features, comparing them to pathogenic and likely pathogenic variants from the same cohort (N = 390). While the clinical exome used did not show differences in diagnostic rate, the more IRD-experienced geneticist reported more VUS (p = 4.07e-04). Significantly fewer VUS were reported in recessive cases (p = 2.14e-04) compared to other inheritance patterns, and of all the genes analyzed, ABCA4 and IMPG2 had the lowest and highest VUS frequencies, respectively (p = 3.89e-04, p = 6.93e-03). Moreover, few frameshift and stop-gain variants were found to be informed VUS (p = 6.73e-08 and p = 2.93e-06). Last, we applied five pathogenicity predictors and found there is a significant proof of deleteriousness when all score for pathogenicity in missense variants. Altogether, these results provided input for a set of rules that correctly reclassified ~70% of VUS as pathogenic in validation datasets. Disease- and setting-specific features influence VUS reporting. Comparison with pathogenic and likely pathogenic variants can prioritize VUS more likely to be reclassified as causal.
Collapse
Affiliation(s)
- Ionut-Florin Iancu
- Department of Genetics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain.,Center for Biomedical Network Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Almudena Avila-Fernandez
- Department of Genetics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain.,Center for Biomedical Network Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Ana Arteche
- Department of Genetics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Maria Jose Trujillo-Tiebas
- Department of Genetics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain.,Center for Biomedical Network Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Rosa Riveiro-Alvarez
- Department of Genetics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain.,Center for Biomedical Network Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Berta Almoguera
- Department of Genetics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain.,Center for Biomedical Network Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Inmaculada Martin-Merida
- Department of Genetics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain.,Center for Biomedical Network Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Marta Del Pozo-Valero
- Department of Genetics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain.,Center for Biomedical Network Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Irene Perea-Romero
- Department of Genetics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain.,Center for Biomedical Network Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Marta Corton
- Department of Genetics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain.,Center for Biomedical Network Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Pablo Minguez
- Department of Genetics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain. .,Center for Biomedical Network Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain.
| | - Carmen Ayuso
- Department of Genetics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain. .,Center for Biomedical Network Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain.
| |
Collapse
|
14
|
Habibi I, Falfoul Y, Tran HV, El Matri K, Chebil A, El Matri L, Schorderet DF. Different Phenotypes in Pseudodominant Inherited Retinal Dystrophies. Front Cell Dev Biol 2021; 9:625560. [PMID: 33634125 PMCID: PMC7902019 DOI: 10.3389/fcell.2021.625560] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/11/2021] [Indexed: 11/29/2022] Open
Abstract
Retinal dystrophies (RD) are a group of Mendelian disorders caused by rare genetic variations leading to blindness. A pathogenic variant may manifest in both dominant or recessive mode and clinical and genetic heterogeneity makes it difficult to establish a precise diagnosis. In this study, families with autosomal dominant RD in successive generations were identified, and we aimed to determine the disease's molecular origin in these consanguineous families. Whole exome sequencing was performed in the index patient of each family. The aim was to determine whether these cases truly represented examples of dominantly inherited RD, or whether another mode of inheritance might be applicable. Six potentially pathogenic variants in four genes were identified in four families. In index patient with enhanced S-cone syndrome in F1, we identified a new digenetic combination: a heterozygous variant p.[G51A];[=] in RHO and a homozygous pathogenic variant p.[R311Q];[R311Q] in NR2E3. Helicoid subretinal fibrosis associated with recessive NR2E3 variant p.[R311Q];[R311Q] was identified in F2. A new frameshift variant c.[105delG];[105delG] in RDH12 was found in F3 with cone-rod dystrophy. In F4, the compound heterozygous variants p.[R964*];[W758*] were observed in IMPG2 with a retinitis pigmentosa (RP) phenotype. We showed that both affected parents and the offspring, were homozygous for the same variants in all four families. Our results provide evidence that in consanguineous families, autosomal recessive can be transmitted as pseudodominant inheritance in RD patients, and further extend our knowledge of pathogenic variants in RD genes.
Collapse
Affiliation(s)
- Imen Habibi
- IRO-Institute for Research in Ophthalmology, Sion, Switzerland
| | - Yosra Falfoul
- Oculogenetic Laboratory LR14SP01, Faculty of Medicine of Tunis, Hedi Rais Institute of Ophthalmology (Department B), Tunis El Manar University, Tunis, Tunisia
| | - Hoai Viet Tran
- Hôpital Ophtalmique Jules-Gonin, Unité d'oculogénétique, Lausanne, Switzerland
| | - Khaled El Matri
- Oculogenetic Laboratory LR14SP01, Faculty of Medicine of Tunis, Hedi Rais Institute of Ophthalmology (Department B), Tunis El Manar University, Tunis, Tunisia
| | - Ahmed Chebil
- Oculogenetic Laboratory LR14SP01, Faculty of Medicine of Tunis, Hedi Rais Institute of Ophthalmology (Department B), Tunis El Manar University, Tunis, Tunisia
| | - Leila El Matri
- Oculogenetic Laboratory LR14SP01, Faculty of Medicine of Tunis, Hedi Rais Institute of Ophthalmology (Department B), Tunis El Manar University, Tunis, Tunisia
| | - Daniel F Schorderet
- IRO-Institute for Research in Ophthalmology, Sion, Switzerland.,Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.,Faculty of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
15
|
Gauvain G, Akolkar H, Chaffiol A, Arcizet F, Khoei MA, Desrosiers M, Jaillard C, Caplette R, Marre O, Bertin S, Fovet CM, Demilly J, Forster V, Brazhnikova E, Hantraye P, Pouget P, Douar A, Pruneau D, Chavas J, Sahel JA, Dalkara D, Duebel J, Benosman R, Picaud S. Optogenetic therapy: high spatiotemporal resolution and pattern discrimination compatible with vision restoration in non-human primates. Commun Biol 2021; 4:125. [PMID: 33504896 PMCID: PMC7840970 DOI: 10.1038/s42003-020-01594-w] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 12/09/2020] [Indexed: 01/06/2023] Open
Abstract
Vision restoration is an ideal medical application for optogenetics, because the eye provides direct optical access to the retina for stimulation. Optogenetic therapy could be used for diseases involving photoreceptor degeneration, such as retinitis pigmentosa or age-related macular degeneration. We describe here the selection, in non-human primates, of a specific optogenetic construct currently tested in a clinical trial. We used the microbial opsin ChrimsonR, and showed that the AAV2.7m8 vector had a higher transfection efficiency than AAV2 in retinal ganglion cells (RGCs) and that ChrimsonR fused to tdTomato (ChR-tdT) was expressed more efficiently than ChrimsonR. Light at 600 nm activated RGCs transfected with AAV2.7m8 ChR-tdT, from an irradiance of 1015 photons.cm−2.s−1. Vector doses of 5 × 1010 and 5 × 1011 vg/eye transfected up to 7000 RGCs/mm2 in the perifovea, with no significant immune reaction. We recorded RGC responses from a stimulus duration of 1 ms upwards. When using the recorded activity to decode stimulus information, we obtained an estimated visual acuity of 20/249, above the level of legal blindness (20/400). These results lay the groundwork for the ongoing clinical trial with the AAV2.7m8 - ChR-tdT vector for vision restoration in patients with retinitis pigmentosa. Gauvain et al demonstrate that optogenetic therapy using the AAV2.7m8- ChR-tdT construct can partially restore vision in non-human primates to levels above those considered legally-blind. This study enables the identification of the most suitable construct for ongoing clinical trials attempting vision restoration in patients with retinitis pigmentosa.
Collapse
Affiliation(s)
- Gregory Gauvain
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France.
| | - Himanshu Akolkar
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France.,Department of Ophthalmology, University Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Antoine Chaffiol
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Fabrice Arcizet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Mina A Khoei
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Mélissa Desrosiers
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Céline Jaillard
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Romain Caplette
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Olivier Marre
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Stéphane Bertin
- CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, F-75012, Paris, France
| | - Claire-Maelle Fovet
- Département des Sciences du Vivant (DSV), MIRcen, Institut d'imagerie Biomédicale (I2BM), Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), 92260, Fontenay-aux-Roses, France
| | - Joanna Demilly
- Département des Sciences du Vivant (DSV), MIRcen, Institut d'imagerie Biomédicale (I2BM), Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), 92260, Fontenay-aux-Roses, France
| | - Valérie Forster
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Elena Brazhnikova
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Philippe Hantraye
- Département des Sciences du Vivant (DSV), MIRcen, Institut d'imagerie Biomédicale (I2BM), Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), 92260, Fontenay-aux-Roses, France
| | - Pierre Pouget
- ICM, UMRS 1127 UPMC - U 1127 INSERM - UMR 7225 CNRS, Paris, France
| | - Anne Douar
- Gensight Biologics, 74 rue du faubourg Saint Antoine, F-75012, Paris, France
| | - Didier Pruneau
- Gensight Biologics, 74 rue du faubourg Saint Antoine, F-75012, Paris, France
| | - Joël Chavas
- Gensight Biologics, 74 rue du faubourg Saint Antoine, F-75012, Paris, France
| | - José-Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France.,Department of Ophthalmology, University Pittsburgh Medical Center, Pittsburgh, PA, USA.,CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, F-75012, Paris, France
| | - Deniz Dalkara
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Jens Duebel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Ryad Benosman
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France.,Department of Ophthalmology, University Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Serge Picaud
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France.
| |
Collapse
|
16
|
Bi-Allelic Pathogenic Variations in MERTK Including Deletions Are Associated with an Early Onset Progressive Form of Retinitis Pigmentosa. Genes (Basel) 2020; 11:genes11121517. [PMID: 33353011 PMCID: PMC7766129 DOI: 10.3390/genes11121517] [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: 09/28/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/20/2022] Open
Abstract
Bi-allelic pathogenic variants in MERTK cause retinitis pigmentosa (RP). Since deletions of more than one exon have been reported repeatedly for MERTK, CNV (copy number variation) analysis of next-generation sequencing (NGS) data has proven important in molecular genetic diagnostics of MERTK. CNV analysis was performed on NGS data of 677 individuals with inherited retinal diseases (IRD) and confirmed by quantitative RT-PCR analysis. Clinical evaluation was based on retrospective records. Clinical re-examination included visual field examination, dark adaption, scotopic and photopic full-field electroretinograms (ffERG), multifocal ERG (mfERG) and optic coherence tomography (OCT). Fourteen variants were detected in MERTK in six individuals, three of which were deletions of more than one exon. Clinical examinations of five out of six individuals revealed a severe phenotype with early-onset generalized retinal dystrophy with night blindness and progressive visual field loss; however, one individual had a milder phenotype. Three individuals had hearing impairments. We show that deletions represent a substantial part of the causative variants in MERTK and emphasize that CNV analysis should be included in the molecular genetic diagnostics of IRDs.
Collapse
|
17
|
Abstract
PURPOSE Choroideremia is an incurable, X-linked, recessive retinal dystrophy caused by loss of function mutations in the CHM gene. It is estimated to affect approximately 1 in 50,000 male patients. It is characterized by progressive degeneration of the retinal pigment epithelium, choroid, and photoreceptors, resulting in visual impairment and blindness. There is an unmet need in choroideremia, because currently, there are no approved treatments available for patients with the disease. METHODS We review the patient journey, societal impact, and emerging treatments for patients with choroideremia. RESULTS Its relative rarity and similarities with other retinal diseases in early years mean that diagnosis of choroideremia can often be delayed. Furthermore, its impact on affected individuals, and wider society, is also likely underestimated. AAV2-mediated gene therapy is an investigational treatment that aims to replace the faulty CHM gene. Early-phase studies reported potentially important visual acuity gains and maintenance of vision in some patients, and a large Phase 3 program is now underway. CONCLUSION Choroideremia is a disease with a significant unmet need. Interventions that can treat progression of the disease and improve visual and functional outcomes have the potential to reduce health care costs and enhance patient quality of life.
Collapse
|
18
|
Wang L, Zou T, Lin Y, Li L, Zhang P, Gong B, Hao J, Zhang H. Identification of a novel homozygous variant in the CNGA1 gene in a Chinese family with autosomal recessive retinitis pigmentosa. Mol Med Rep 2020; 22:2516-2520. [PMID: 32705276 PMCID: PMC7411332 DOI: 10.3892/mmr.2020.11331] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 06/12/2020] [Indexed: 11/30/2022] Open
Abstract
Retinitis pigmentosa (RP) is a complex group of hereditary retinal dystrophies. Although >60 genes have been identified to be associated with non-syndromic RP, the exact genetic variant remains elusive in numerous cases of RP. In the present study, a Chinese pedigree affected by RP with autosomal recessive inheritance, including a total of seven members with one affected patient and six unaffected individuals, was recruited. Comprehensive ophthalmic examinations were performed on the proband and the proband's unaffected daughter. Genomic DNA was extracted from peripheral blood. Whole-exome sequencing (WES) was performed for the affected individual. The candidate pathogenic variant was verified by direct Sanger sequencing. The affected individual presented with classical clinical symptoms of RP. A novel homozygous variant, c.265delC (p.L89Ffs*3) in the cyclic nucleotide-gated channel subunit α 1 gene was identified in the affected patient. This homozygous variant was absent in other unaffected family members and 600 ethnicity-matched healthy controls. The variant was co-segregated with the disease phenotype in an autosomal recessive manner.
Collapse
Affiliation(s)
- Le Wang
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun, Jilin 130103, P.R. China
| | - Tongdan Zou
- Key Laboratory for Human Disease Gene Study of Sichuan Province and Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, P.R. China
| | - Yongqiong Lin
- Key Laboratory for Human Disease Gene Study of Sichuan Province and Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, P.R. China
| | - Ling Li
- Key Laboratory for Human Disease Gene Study of Sichuan Province and Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, P.R. China
| | - Peng Zhang
- Diagnosis Center, The First Hospital of Jilin University, Changchun, Jilin 130103, P.R. China
| | - Bo Gong
- Key Laboratory for Human Disease Gene Study of Sichuan Province and Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, P.R. China
| | - Jilong Hao
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun, Jilin 130103, P.R. China
| | - Houbin Zhang
- Key Laboratory for Human Disease Gene Study of Sichuan Province and Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, P.R. China
| |
Collapse
|
19
|
Olsen TW, Dyer RB, Mano F, Boatright JH, Chrenek MA, Paley D, Wabner K, Schmit J, Chae JB, Sellers JT, Singh RJ, Wiedmann TS. Drug Tissue Distribution of TUDCA From a Biodegradable Suprachoroidal Implant versus Intravitreal or Systemic Delivery in the Pig Model. Transl Vis Sci Technol 2020; 9:11. [PMID: 32821508 PMCID: PMC7408862 DOI: 10.1167/tvst.9.6.11] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/10/2020] [Indexed: 12/14/2022] Open
Abstract
Purpose To determine local ocular tissue levels of the bile acid, tauroursodeoxycholic acid (TUDCA), in the pig model using oral, intravenous (IV), intravitreal injection (IVitI) and low- and high-dose suprachoroidal, sustained-release implants (SCI-L or SCI-H). Methods Forty-six pigs (92 globes) were included in the study. TUDCA was delivered orally in 5 pigs, IV in 4, IVitI in 6, SCI-L in 17, and SCI-H in 14. Testing timeframes varied from the same day (within minutes) for IV; 1 to 6 days, oral; and 1 to 4 weeks, IVitI and SCI. Enucleated globes were dissected, specimens from specific tissues were separated, and TUDCA was extracted and quantified using mass spectrometry. Results The highest TUDCA tissue levels occurred after IV delivery in the macula (252 ± 238 nM) and peripheral retina (196 ± 171 nM). Macular choroid and peripheral choroid levels were also high (1032 ± 1269 and 1219 ± 1486 nM, respectively). For IVitI delivery, macular levels at day 6 were low (0.5 ± 0.5 nM), whereas peripheral choroid was higher (15.3 ± 16.7 nM). Neither the SCI-L nor SCI-H implants delivered meaningful macular doses (≤1 nM); however, peripheral retina and choroid levels were significantly higher. Bile acid isoforms were found in the serum specimens. Conclusions The highest TUDCA tissue levels in the pig model were obtained using IV delivery. Oral delivery was associated with reasonable tissue levels. Local delivery (IVitI and SCI) was able to achieve measurable local ocular tissue levels. Translational Relevance Diffusional kinetics from the suprachoroidal space follow the choroidal blood flow, away from the macula and toward the periphery.
Collapse
Affiliation(s)
- Timothy W Olsen
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, USA.,Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA, USA
| | - Roy B Dyer
- Division of Clinical Biochemistry and Immunology, Mayo Clinic, Rochester, MN, USA
| | - Fukutaro Mano
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, USA
| | - Jeffrey H Boatright
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA, USA
| | - Micah A Chrenek
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA, USA
| | - Daniel Paley
- Department of Civil Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Kathy Wabner
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | - Jenn Schmit
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | - Ju Byung Chae
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA, USA.,Department of Ophthalmology, Chungbuk National University, Chungbuk, South Korea
| | - Jana T Sellers
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | - Ravinder J Singh
- Division of Clinical Biochemistry and Immunology, Mayo Clinic, Rochester, MN, USA.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Timothy S Wiedmann
- Department of Civil Engineering, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
20
|
García-Ayuso D, Di Pierdomenico J, Vidal-Sanz M, Villegas-Pérez MP. Retinal Ganglion Cell Death as a Late Remodeling Effect of Photoreceptor Degeneration. Int J Mol Sci 2019; 20:ijms20184649. [PMID: 31546829 PMCID: PMC6770703 DOI: 10.3390/ijms20184649] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/09/2019] [Accepted: 09/17/2019] [Indexed: 12/16/2022] Open
Abstract
Inherited or acquired photoreceptor degenerations, one of the leading causes of irreversible blindness in the world, are a group of retinal disorders that initially affect rods and cones, situated in the outer retina. For many years it was assumed that these diseases did not spread to the inner retina. However, it is now known that photoreceptor loss leads to an unavoidable chain of events that cause neurovascular changes in the retina including migration of retinal pigment epithelium cells, formation of “subretinal vascular complexes”, vessel displacement, retinal ganglion cell (RGC) axonal strangulation by retinal vessels, axonal transport alteration and, ultimately, RGC death. These events are common to all photoreceptor degenerations regardless of the initial trigger and thus threaten the outcome of photoreceptor substitution as a therapeutic approach, because with a degenerating inner retina, the photoreceptor signal will not reach the brain. In conclusion, therapies should be applied early in the course of photoreceptor degeneration, before the remodeling process reaches the inner retina.
Collapse
Affiliation(s)
- Diego García-Ayuso
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Virgen de la Arrixaca), 30120 Murcia, Spain.
| | - Johnny Di Pierdomenico
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Virgen de la Arrixaca), 30120 Murcia, Spain.
| | - Manuel Vidal-Sanz
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Virgen de la Arrixaca), 30120 Murcia, Spain.
| | - María P Villegas-Pérez
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Virgen de la Arrixaca), 30120 Murcia, Spain.
| |
Collapse
|
21
|
Liu B, Calton MA, Abell NS, Benchorin G, Gloudemans MJ, Chen M, Hu J, Li X, Balliu B, Bok D, Montgomery SB, Vollrath D. Genetic analyses of human fetal retinal pigment epithelium gene expression suggest ocular disease mechanisms. Commun Biol 2019; 2:186. [PMID: 31123710 PMCID: PMC6527609 DOI: 10.1038/s42003-019-0430-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 04/17/2019] [Indexed: 02/07/2023] Open
Abstract
The retinal pigment epithelium (RPE) serves vital roles in ocular development and retinal homeostasis but has limited representation in large-scale functional genomics datasets. Understanding how common human genetic variants affect RPE gene expression could elucidate the sources of phenotypic variability in selected monogenic ocular diseases and pinpoint causal genes at genome-wide association study (GWAS) loci. We interrogated the genetics of gene expression of cultured human fetal RPE (fRPE) cells under two metabolic conditions and discovered hundreds of shared or condition-specific expression or splice quantitative trait loci (e/sQTLs). Co-localizations of fRPE e/sQTLs with age-related macular degeneration (AMD) and myopia GWAS data suggest new candidate genes, and mechanisms by which a common RDH5 allele contributes to both increased AMD risk and decreased myopia risk. Our study highlights the unique transcriptomic characteristics of fRPE and provides a resource to connect e/sQTLs in a critical ocular cell type to monogenic and complex eye disorders.
Collapse
Affiliation(s)
- Boxiang Liu
- Department of Biology, Stanford University, Stanford, CA 94305 USA
| | - Melissa A. Calton
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Nathan S. Abell
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Gillie Benchorin
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Michael J. Gloudemans
- Program in Biomedical Informatics, Stanford University School of Medicine, Stanford, 94305 CA USA
| | - Ming Chen
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Jane Hu
- Department of Ophthalmology, Jules Stein Eye Institute, UCLA, Los Angeles, 90095 CA USA
| | - Xin Li
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Brunilda Balliu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Dean Bok
- Department of Ophthalmology, Jules Stein Eye Institute, UCLA, Los Angeles, 90095 CA USA
| | - Stephen B. Montgomery
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305 USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Douglas Vollrath
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305 USA
| |
Collapse
|
22
|
Donato L, Scimone C, Nicocia G, D'Angelo R, Sidoti A. Role of oxidative stress in Retinitis pigmentosa: new involved pathways by an RNA-Seq analysis. Cell Cycle 2018; 18:84-104. [PMID: 30569795 DOI: 10.1080/15384101.2018.1558873] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Retinitis pigmentosa (RP) is a very heterogeneous inherited ocular disorder group characterized by progressive retinal disruption. Retinal pigment epithelium (RPE) degeneration, due to oxidative stress which arrests the metabolic support to photoreceptors, represents one of the principal causes of RP. Here, the role of oxidative stress in RP onset and progression was analyzed by a comparative whole transcriptome analysis of human RPE cells, treated with 100 µg/ml of oxLDL and untreated, at different time points. Experiment was thrice repeated and performed on Ion ProtonTM sequencing system. Data analysis, including low quality reads trimming and gene expression quantification, was realized by CLC Genomics Workbench software. The whole analysis highlighted 14 clustered "macro-pathways" and many sub-pathways, classified by selection of 5271 genes showing the highest alteration of expression. Among them, 23 genes were already known to be RP causative ones (15 over-expressed and 8 down-expressed), and their enrichment and intersection analyses highlighted new 77 candidate related genes (49 over-expressed and 28 down-expressed). A final filtering analysis then highlighted 29 proposed candidate genes. This data suggests that many new genes, not yet associated with RP, could influence its etiopathogenesis.
Collapse
Affiliation(s)
- Luigi Donato
- a Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine , University of Messina , Messina , Italy.,b Department of Cutting-Edge Medicine and Therapies, Biomolecular Strategies and Neuroscience, Section of Applied Neuroscience, Molecular Genetics and Predictive Medicine , I.E.ME.S.T. ., Palermo , Italy
| | - Concetta Scimone
- a Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine , University of Messina , Messina , Italy.,b Department of Cutting-Edge Medicine and Therapies, Biomolecular Strategies and Neuroscience, Section of Applied Neuroscience, Molecular Genetics and Predictive Medicine , I.E.ME.S.T. ., Palermo , Italy
| | - Giacomo Nicocia
- c Department of Clinical and Experimental Medicine , University of Messina , Messina , Italy
| | - Rosalia D'Angelo
- a Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine , University of Messina , Messina , Italy.,b Department of Cutting-Edge Medicine and Therapies, Biomolecular Strategies and Neuroscience, Section of Applied Neuroscience, Molecular Genetics and Predictive Medicine , I.E.ME.S.T. ., Palermo , Italy
| | - Antonina Sidoti
- a Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine , University of Messina , Messina , Italy.,b Department of Cutting-Edge Medicine and Therapies, Biomolecular Strategies and Neuroscience, Section of Applied Neuroscience, Molecular Genetics and Predictive Medicine , I.E.ME.S.T. ., Palermo , Italy
| |
Collapse
|
23
|
Motta FL, Martin RP, Filippelli-Silva R, Salles MV, Sallum JMF. Relative frequency of inherited retinal dystrophies in Brazil. Sci Rep 2018; 8:15939. [PMID: 30374144 PMCID: PMC6206004 DOI: 10.1038/s41598-018-34380-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 10/15/2018] [Indexed: 11/09/2022] Open
Abstract
Among the Brazilian population, the frequency rates of inherited retinal dystrophies and their causative genes are underreported. To increase the knowledge about these dystrophies in our population, we retrospectively studied the medical records of 1,246 Brazilian patients with hereditary retinopathies during 20 years of specialized outpatient clinic care. Of these patients, 559 had undergone at least one genetic test. In this cohort, the most prevalent dystrophies were non-syndromic retinitis pigmentosa (35%), Stargardt disease (21%), Leber congenital amaurosis (9%), and syndromic inherited retinal dystrophies (12%). Most patients had never undergone genetic testing (55%), and among the individuals with molecular test results, 28.4% had negative or inconclusive results compared to 71.6% with a conclusive molecular diagnosis. ABCA4 was the most frequent disease-causing gene, accounting for 20% of the positive cases. Pathogenic variants also occurred frequently in the CEP290, USH2A, CRB1, RPGR, and CHM genes. The relative frequency rates of different inherited retinal dystrophies in Brazil are similar to those found globally. Although mutations in more than 250 genes lead to hereditary retinopathies, only 66 genes were responsible for 70% of the cases, which indicated that smaller and cheaper gene panels can be just as effective and provide more affordable solutions for implementation by the Brazilian public health system.
Collapse
Affiliation(s)
- Fabiana Louise Motta
- Department of Ophthalmology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Renan Paulo Martin
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo, Brazil.,Institute of Genetic Medicine, Johns Hopkins Medicine, Baltimore, USA
| | | | | | - Juliana Maria Ferraz Sallum
- Department of Ophthalmology, Universidade Federal de São Paulo, São Paulo, Brazil. .,Instituto de Genética Ocular, Sao Paulo, Brazil.
| |
Collapse
|
24
|
Fiorentino A, Yu J, Arno G, Pontikos N, Halford S, Broadgate S, Michaelides M, Carss KJ, Raymond FL, Cheetham ME, Webster AR, Downes SM, Hardcastle AJ, for the NIHR-BioResource Rare Diseases Consortium, the U.K. Inherited Retinal Dystrophy Consortium. Novel homozygous splicing mutations in ARL2BP cause autosomal recessive retinitis pigmentosa. Mol Vis 2018; 24:603-612. [PMID: 30210231 PMCID: PMC6128700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 08/29/2018] [Indexed: 12/02/2022] Open
Abstract
Purpose Mutations in ARL2BP, encoding ADP-ribosylation factor-like 2 binding protein, have recently been implicated as a cause of autosomal recessive retinitis pigmentosa (arRP), with three homozygous variants identified to date. In this study, we performed next-generation sequencing to reveal additional arRP cases associated with ARL2BP variants. Methods Whole-genome sequencing (WGS) or whole-exome sequencing (WES) was performed in 1,051 unrelated individuals recruited for the UK Inherited Retinal Disease Consortium and NIHR-BioResource Rare Diseases research studies. Sanger sequencing was used to validate the next-generation sequencing data, and reverse transcriptase (RT)-PCR analysis was performed on RNA extracted from blood from affected individuals to test for altered splicing of ARL2BP. Detailed phenotyping was performed, including clinical evaluation, electroretinography, fundus photography, fundus autofluorescence imaging, and spectral-domain optical coherence tomography. Results Homozygous variants in ARL2BP (NM_012106.3) were identified in two unrelated individuals with RP. The variants, c.207+1G>A and c.390+5G>A, at conserved splice donor sites for intron 3 and intron 5, respectively, were predicted to alter the pre-mRNA splicing of ARL2BP. RT-PCR spanning the affected introns revealed that both variants caused abnormal splicing of ARL2BP in samples from affected individuals. Conclusions This study identified two homozygous variants in ARL2BP as a rare cause of arRP. Further studies are required to define the underlying disease mechanism causing retinal degeneration as a result of mutations in ARL2BP and any phenotype-genotype correlation associated with residual levels of the wild-type transcript.
Collapse
Affiliation(s)
| | - Jing Yu
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Gavin Arno
- UCL Institute of Ophthalmology, London, UK
- Moorfields Eye Hospital, London, UK
| | | | - Stephanie Halford
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Suzanne Broadgate
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Michel Michaelides
- UCL Institute of Ophthalmology, London, UK
- Moorfields Eye Hospital, London, UK
| | - Keren J. Carss
- Department of Haematology, University of Cambridge, NHS Blood and Transplant Centre, Cambridge, UK
- NIHR BioResource - Rare Diseases, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK
| | - F. Lucy Raymond
- NIHR BioResource - Rare Diseases, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | | | - Andrew R. Webster
- UCL Institute of Ophthalmology, London, UK
- Moorfields Eye Hospital, London, UK
| | | | | | - for the NIHR-BioResource Rare Diseases Consortium
- UCL Institute of Ophthalmology, London, UK
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
- Moorfields Eye Hospital, London, UK
- Department of Haematology, University of Cambridge, NHS Blood and Transplant Centre, Cambridge, UK
- NIHR BioResource - Rare Diseases, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
- Oxford Eye Hospital, John Radcliffe Hospital, Oxford, UK
| | - the U.K. Inherited Retinal Dystrophy Consortium
- UCL Institute of Ophthalmology, London, UK
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
- Moorfields Eye Hospital, London, UK
- Department of Haematology, University of Cambridge, NHS Blood and Transplant Centre, Cambridge, UK
- NIHR BioResource - Rare Diseases, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
- Oxford Eye Hospital, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
25
|
Developing an item bank to measure the coping strategies of people with hereditary retinal diseases. Graefes Arch Clin Exp Ophthalmol 2018; 256:1291-1298. [PMID: 29730797 DOI: 10.1007/s00417-018-3998-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/10/2018] [Accepted: 04/20/2018] [Indexed: 01/09/2023] Open
Abstract
PURPOSE Our understanding of the coping strategies used by people with visual impairment to manage stress related to visual loss is limited. This study aims to develop a sophisticated coping instrument in the form of an item bank implemented via Computerised adaptive testing (CAT) for hereditary retinal diseases. METHODS Items on coping were extracted from qualitative interviews with patients which were supplemented by items from a literature review. A systematic multi-stage process of item refinement was carried out followed by expert panel discussion and cognitive interviews. The final coping item bank had 30 items. Rasch analysis was used to assess the psychometric properties. A CAT simulation was carried out to estimate an average number of items required to gain precise measurement of hereditary retinal disease-related coping. RESULTS One hundred eighty-nine participants answered the coping item bank (median age = 58 years). The coping scale demonstrated good precision and targeting. The standardised residual loadings for items revealed six items grouped together. Removal of the six items reduced the precision of the main coping scale and worsened the variance explained by the measure. Therefore, the six items were retained within the main scale. Our CAT simulation indicated that, on average, less than 10 items are required to gain a precise measurement of coping. CONCLUSIONS This is the first study to develop a psychometrically robust coping instrument for hereditary retinal diseases. CAT simulation indicated that on an average, only four and nine items were required to gain measurement at moderate and high precision, respectively.
Collapse
|
26
|
Sun C, Galicia C, Stenkamp DL. Transcripts within rod photoreceptors of the Zebrafish retina. BMC Genomics 2018; 19:127. [PMID: 29422031 PMCID: PMC5806438 DOI: 10.1186/s12864-018-4499-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 01/28/2018] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND The purpose of this study was to identify transcripts of retinal rod photoreceptors of the zebrafish. The zebrafish is an important animal model for vision science due to rapid and tractable development, persistent neurogenesis of rods throughout the lifespan, and capacity for functional retinal regeneration. RESULTS Zebrafish rods, and non-rod retinal cells of the xops:eGFP transgenic line, were separated by cell dissociation and fluorescence-activated cell sorting (FACS), followed by RNA-seq. At a false discovery rate of < 0.01, 597 transcripts were upregulated ("enriched") in rods vs. other retinal cells, and 1032 were downregulated ("depleted"). Thirteen thousand three hundred twenty four total transcripts were detected in rods, including many not previously known to be expressed by rods. Forty five transcripts were validated by qPCR in FACS-sorted rods vs. other retinal cells. Transcripts enriched in rods from adult retinas were also enriched in rods from larval and juvenile retinas, and were also enriched in rods sorted from retinas subjected to a neurotoxic lesion and allowed to regenerate. Many transcripts enriched in rods were upregulated in retinas of wildtype retinas vs. those of a zebrafish model for rod degeneration. CONCLUSIONS We report the generation and validation of an RNA-seq dataset describing the rod transcriptome of the zebrafish, which is now available as a resource for further studies of rod photoreceptor biology and comparative transcriptomics.
Collapse
Affiliation(s)
- Chi Sun
- Department of Biological Sciences, University of Idaho, 875 Perimeter Drive, MS 3051, Moscow, ID 83844-3051 USA
| | - Carlos Galicia
- Department of Biological Sciences, University of Idaho, 875 Perimeter Drive, MS 3051, Moscow, ID 83844-3051 USA
| | - Deborah L. Stenkamp
- Department of Biological Sciences, University of Idaho, 875 Perimeter Drive, MS 3051, Moscow, ID 83844-3051 USA
| |
Collapse
|
27
|
Eblimit A, Agrawal SA, Thomas K, Anastassov IA, Abulikemu T, Moayedi Y, Mardon G, Chen R. Conditional loss of Spata7 in photoreceptors causes progressive retinal degeneration in mice. Exp Eye Res 2018; 166:120-130. [PMID: 29100828 PMCID: PMC5756513 DOI: 10.1016/j.exer.2017.10.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 10/10/2017] [Accepted: 10/13/2017] [Indexed: 12/31/2022]
Abstract
The mammalian retina consists of multiple cell layers including photoreceptor cells, which are light sensing neurons that play essential functions in the visual process. Previously, we identified mutations in SPATA7, encoding spermatogenesis associated protein 7, in families with Leber Congenital Amaurosis (LCA) and juvenile Retinitis Pigmentosa (RP), and showed that Spata7 null mice recapitulate the human disease phenotype of retinal degeneration. SPATA7 is expressed in the connecting cilium of photoreceptor (PR) cells in the mouse retina, as well as in retinal pigment epithelium (RPE) cells, but the functional role of Spata7 in the RPE remains unknown. To investigate whether Spata7 is required in PRs, the RPE, or both, we conditionally knocked out Spata7 in photoreceptors and RPE cells using Crx-Cre and Best1-Cre transgenic mouse lines, respectively. In Spata7 photoreceptor-specific conditional (cKO) mice, both rod and cone photoreceptor dysfunction and degeneration is observed, characterized by progressive thinning of the outer nuclear layer and reduced response to light; however, RPE-specific deletion of Spata7 does not impair retinal function or cell survival. Furthermore, our findings show that both Rhodopsin and RPGRIP1 are mislocalized in the Spata7Flox/-; Crx-Cre cKO mice, suggesting that loss of Spata7 in photoreceptors alone can result in altered trafficking of these proteins in the connecting cilium. Together, our findings suggest that loss of Spata7 in photoreceptors alone is sufficient to cause photoreceptor degeneration, but its function in the RPE is not required for photoreceptor survival; therefore, loss of Spata7 in photoreceptors alters both rod and cone function and survival, consistent with the clinical phenotypes observed in LCA and RP patients with mutations in SPATA7.
Collapse
Affiliation(s)
- Aiden Eblimit
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030-3411, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030-3411, USA
| | - Smriti Akshay Agrawal
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030-3411, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030-3411, USA
| | - Kandace Thomas
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030-3411, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030-3411, USA
| | - Ivan Assenov Anastassov
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030-3411, USA
| | - Tajiguli Abulikemu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030-3411, USA; The Key Laboratory of Plant Resources and Chemistry of Arid Zone, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, Xinjiang, 830011, China
| | | | - Graeme Mardon
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030-3411, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030-3411, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030-3411, USA.
| | - Rui Chen
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030-3411, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030-3411, USA.
| |
Collapse
|
28
|
Bryant L, Lozynska O, Maguire AM, Aleman TS, Bennett J. Prescreening whole exome sequencing results from patients with retinal degeneration for variants in genes associated with retinal degeneration. Clin Ophthalmol 2017; 12:49-63. [PMID: 29343940 PMCID: PMC5749571 DOI: 10.2147/opth.s147684] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Background Accurate clinical diagnosis and prognosis of retinal degeneration can be aided by the identification of the disease-causing genetic variant. It can confirm the clinical diagnosis as well as inform the clinician of the risk for potential involvement of other organs such as kidneys. It also aids in genetic counseling for affected individuals who want to have a child. Finally, knowledge of disease-causing variants informs laboratory investigators involved in translational research. With the advent of next-generation sequencing, identifying pathogenic mutations is becoming easier, especially the identification of novel pathogenic variants. Methods We used whole exome sequencing on a cohort of 69 patients with various forms of retinal degeneration and in whom screens for previously identified disease-causing variants had been inconclusive. All potential pathogenic variants were verified by Sanger sequencing and, when possible, segregation analysis of immediate relatives. Potential variants were identified by using a semi-masked approach in which rare variants in candidate genes were identified without knowledge of the clinical diagnosis (beyond "retinal degeneration") or inheritance pattern. After the initial list of genes was prioritized, genetic diagnosis and inheritance pattern were taken into account. Results We identified the likely pathogenic variants in 64% of the subjects. Seven percent had a single heterozygous mutation identified that would cause recessive disease and 13% had no obviously pathogenic variants and no family members available to perform segregation analysis. Eleven subjects are good candidates for novel gene discovery. Two de novo mutations were identified that resulted in dominant retinal degeneration. Conclusion Whole exome sequencing allows for thorough genetic analysis of candidate genes as well as novel gene discovery. It allows for an unbiased analysis of genetic variants to reduce the chance that the pathogenic mutation will be missed due to incomplete or inaccurate family history or analysis at the early stage of a syndromic form of retinal degeneration.
Collapse
Affiliation(s)
- Laura Bryant
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Olga Lozynska
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Albert M Maguire
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Ophthalmology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tomas S Aleman
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Ophthalmology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jean Bennett
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Ophthalmology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
29
|
Porto FBO, Jones EM, Branch J, Soens ZT, Maia IM, Sena IFG, Sampaio SAM, Simões RT, Chen R. Molecular Screening of 43 Brazilian Families Diagnosed with Leber Congenital Amaurosis or Early-Onset Severe Retinal Dystrophy. Genes (Basel) 2017; 8:genes8120355. [PMID: 29186038 PMCID: PMC5748673 DOI: 10.3390/genes8120355] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 11/20/2017] [Accepted: 11/21/2017] [Indexed: 12/17/2022] Open
Abstract
Leber congenital amaurosis (LCA) is a severe disease that leads to complete blindness in children, typically before the first year of life. Due to the clinical and genetic heterogeneity among LCA and other retinal diseases, providing patients with a molecular diagnosis is essential to assigning an accurate clinical diagnosis. Using our gene panel that targets 300 genes that are known to cause retinal disease, including 24 genes reported to cause LCA, we sequenced 43 unrelated probands with Brazilian ancestry. We identified 42 unique variants and were able to assign a molecular diagnosis to 30/43 (70%) Brazilian patients. Among these, 30 patients were initially diagnosed with LCA or a form of early-onset retinal dystrophy, 17 patients harbored mutations in LCA-associated genes, while 13 patients had mutations in genes that were reported to cause other diseases involving the retina.
Collapse
Affiliation(s)
- Fernanda B O Porto
- INRET Clínica e Centro de Pesquisa, Belo Horizonte, 30150290 Minas Gerais, Brazil.
- Centro Oftalmológico de Minas Gerais, COMG, Belo Horizonte, 30150290 Minas Gerais, Brazil.
| | - Evan M Jones
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Justin Branch
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Zachry T Soens
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Igor Mendes Maia
- Instituto de Ensino e Pesquisa da Santa Casa de Belo Horizonte, IEP/SCBH, Belo Horizonte, 30150290 Minas Gerais, Brazil.
| | - Isadora F G Sena
- Instituto de Ensino e Pesquisa da Santa Casa de Belo Horizonte, IEP/SCBH, Belo Horizonte, 30150290 Minas Gerais, Brazil.
| | - Shirley A M Sampaio
- INRET Clínica e Centro de Pesquisa, Belo Horizonte, 30150290 Minas Gerais, Brazil.
| | - Renata T Simões
- Instituto de Ensino e Pesquisa da Santa Casa de Belo Horizonte, IEP/SCBH, Belo Horizonte, 30150290 Minas Gerais, Brazil.
| | - Rui Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA.
- Structural and Computational Biology & Molecular Biophysics, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
30
|
Nash BM, Symes R, Goel H, Dinger ME, Bennetts B, Grigg JR, Jamieson RV. NMNAT1 variants cause cone and cone-rod dystrophy. Eur J Hum Genet 2017; 26:428-433. [PMID: 29184169 DOI: 10.1038/s41431-017-0029-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/13/2017] [Accepted: 10/10/2017] [Indexed: 11/09/2022] Open
Abstract
Cone and cone-rod dystrophies (CD and CRD, respectively) are degenerative retinal diseases that predominantly affect the cone photoreceptors. The underlying disease gene is not known in approximately 75% of autosomal recessive cases. Variants in NMNAT1 cause a severe, early-onset retinal dystrophy called Leber congenital amaurosis (LCA). We report two patients where clinical phenotyping indicated diagnoses of CD and CRD, respectively. NMNAT1 variants were identified, with Case 1 showing an extremely rare homozygous variant c.[271G > A] p.(Glu91Lys) and Case 2 compound heterozygous variants c.[53 A > G];[769G > A] p.(Asn18Ser);(Glu257Lys). The detailed variant analysis, in combination with the observation of an associated macular atrophy phenotype, indicated that these variants were disease-causing. This report demonstrates that the variants in NMNAT1 may cause CD or CRD associated with macular atrophy. Genetic investigations of the patients with CD or CRD should include NMNAT1 in the genes examined.
Collapse
Affiliation(s)
- Benjamin M Nash
- Eye Genetics Research, The Children's Hospital at Westmead, Save Sight Institute, Children's Medical Research Institute, University of Sydney, Sydney, NSW, Australia.,Disciplines of Genetic Medicine, and Child and Adolescent Health, Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Sydney Genome Diagnostics, Western Sydney Genetics Program, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Richard Symes
- Discipline of Ophthalmology, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | | | - Marcel E Dinger
- Kinghorn Centre for Clinical Genomics, Garvan Institute for Medical Research, Sydney, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Bruce Bennetts
- Sydney Genome Diagnostics, Western Sydney Genetics Program, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | - John R Grigg
- Eye Genetics Research, The Children's Hospital at Westmead, Save Sight Institute, Children's Medical Research Institute, University of Sydney, Sydney, NSW, Australia.,Discipline of Ophthalmology, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Robyn V Jamieson
- Eye Genetics Research, The Children's Hospital at Westmead, Save Sight Institute, Children's Medical Research Institute, University of Sydney, Sydney, NSW, Australia. .,Disciplines of Genetic Medicine, and Child and Adolescent Health, Sydney Medical School, University of Sydney, Sydney, NSW, Australia. .,Department of Clinical Genetics, Western Sydney Genetics Program, The Children's Hospital at Westmead, Sydney, NSW, Australia.
| |
Collapse
|
31
|
Hartl D, Krebs AR, Jüttner J, Roska B, Schübeler D. Cis-regulatory landscapes of four cell types of the retina. Nucleic Acids Res 2017; 45:11607-11621. [PMID: 29059322 PMCID: PMC5714137 DOI: 10.1093/nar/gkx923] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/28/2017] [Accepted: 10/02/2017] [Indexed: 12/18/2022] Open
Abstract
The retina is composed of ∼50 cell-types with specific functions for the process of vision. Identification of the cis-regulatory elements active in retinal cell-types is key to elucidate the networks controlling this diversity. Here, we combined transcriptome and epigenome profiling to map the regulatory landscape of four cell-types isolated from mouse retinas including rod and cone photoreceptors as well as rare inter-neuron populations such as horizontal and starburst amacrine cells. Integration of this information reveals sequence determinants and candidate transcription factors for controlling cellular specialization. Additionally, we refined parallel reporter assays to enable studying the transcriptional activity of large collection of sequences in individual cell-types isolated from a tissue. We provide proof of concept for this approach and its scalability by characterizing the transcriptional capacity of several hundred putative regulatory sequences within individual retinal cell-types. This generates a catalogue of cis-regulatory regions active in retinal cell types and we further demonstrate their utility as potential resource for cellular tagging and manipulation.
Collapse
Affiliation(s)
- Dominik Hartl
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, CH 4058 Basel, Switzerland
- University of Basel, Faculty of Sciences, Petersplatz 1, CH 4003 Basel, Switzerland
| | - Arnaud R. Krebs
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, CH 4058 Basel, Switzerland
| | - Josephine Jüttner
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, CH 4058 Basel, Switzerland
| | - Botond Roska
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, CH 4058 Basel, Switzerland
- University of Basel, Department of Ophthalmology, Mittlere Strasse 91, CH 4031 Basel, Switzerland
| | - Dirk Schübeler
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, CH 4058 Basel, Switzerland
- University of Basel, Faculty of Sciences, Petersplatz 1, CH 4003 Basel, Switzerland
| |
Collapse
|
32
|
Llavona P, Pinelli M, Mutarelli M, Marwah VS, Schimpf-Linzenbold S, Thaler S, Yoeruek E, Vetter J, Kohl S, Wissinger B. Allelic Expression Imbalance in the Human Retinal Transcriptome and Potential Impact on Inherited Retinal Diseases. Genes (Basel) 2017; 8:genes8100283. [PMID: 29053642 PMCID: PMC5664133 DOI: 10.3390/genes8100283] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 10/11/2017] [Accepted: 10/16/2017] [Indexed: 01/19/2023] Open
Abstract
Inherited retinal diseases (IRDs) are often associated with variable clinical expressivity (VE) and incomplete penetrance (IP). Underlying mechanisms may include environmental, epigenetic, and genetic factors. Cis-acting expression quantitative trait loci (cis-eQTLs) can be implicated in the regulation of genes by favoring or hampering the expression of one allele over the other. Thus, the presence of such loci elicits allelic expression imbalance (AEI) that can be traced by massive parallel sequencing techniques. In this study, we performed an AEI analysis on RNA-sequencing (RNA-seq) data, from 52 healthy retina donors, that identified 194 imbalanced single nucleotide polymorphisms(SNPs) in 67 IRD genes. Focusing on SNPs displaying AEI at a frequency higher than 10%, we found evidence of AEI in several IRD genes regularly associated with IP and VE (BEST1, RP1, PROM1, and PRPH2). Based on these SNPs commonly undergoing AEI, we performed pyrosequencing in an independent sample set of 17 healthy retina donors in order to confirm our findings. Indeed, we were able to validate CDHR1, BEST1, and PROM1 to be subjected to cis-acting regulation. With this work, we aim to shed light on differentially expressed alleles in the human retina transcriptome that, in the context of autosomal dominant IRD cases, could help to explain IP or VE.
Collapse
Affiliation(s)
- Pablo Llavona
- Institute for Ophthalmic Research, Centre for Ophthalmology, 72076 Tuebingen, Germany.
| | - Michele Pinelli
- Telethon Institute of Genetics and Medicine, 80078 Pozzuoli, Italy.
| | | | - Veer Singh Marwah
- Telethon Institute of Genetics and Medicine, 80078 Pozzuoli, Italy.
- Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland.
| | | | - Sebastian Thaler
- Center for Ophthalmology, 72076 Tuebingen, Germany. --unituebingen-i09f.de
| | - Efdal Yoeruek
- Center for Ophthalmology, 72076 Tuebingen, Germany.
- Augenklinik Mülheim, 45468 Mülheim an der Ruhr, Germany.
| | - Jan Vetter
- Universitäts-Augenklinik, 55131 Mainz, Germany.
| | - Susanne Kohl
- Institute for Ophthalmic Research, Centre for Ophthalmology, 72076 Tuebingen, Germany.
| | - Bernd Wissinger
- Institute for Ophthalmic Research, Centre for Ophthalmology, 72076 Tuebingen, Germany.
| |
Collapse
|
33
|
A Brain-Derived Neurotrophic Factor Mimetic Is Sufficient to Restore Cone Photoreceptor Visual Function in an Inherited Blindness Model. Sci Rep 2017; 7:11320. [PMID: 28900183 PMCID: PMC5595969 DOI: 10.1038/s41598-017-11513-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 08/25/2017] [Indexed: 01/25/2023] Open
Abstract
Controversially, histone deacetylase inhibitors (HDACi) are in clinical trial for the treatment of inherited retinal degeneration. Utilizing the zebrafish dyeucd6 model, we determined if treatment with HDACi can rescue cone photoreceptor-mediated visual function. dye exhibit defective visual behaviour and retinal morphology including ciliary marginal zone (CMZ) cell death and decreased photoreceptor outer segment (OS) length, as well as gross morphological defects including hypopigmentation and pericardial oedema. HDACi treatment of dye results in significantly improved optokinetic (OKR) (~43 fold, p < 0.001) and visualmotor (VMR) (~3 fold, p < 0.05) responses. HDACi treatment rescued gross morphological defects and reduced CMZ cell death by 80%. Proteomic analysis of dye eye extracts suggested BDNF-TrkB and Akt signaling as mediators of HDACi rescue in our dataset. Co-treatment with the TrkB antagonist ANA-12 blocked HDACi rescue of visual function and associated Akt phosphorylation. Notably, sole treatment with a BDNF mimetic, 7,8-dihydroxyflavone hydrate, significantly rescued dye visual function (~58 fold increase in OKR, p < 0.001, ~3 fold increase in VMR, p < 0.05). In summary, HDACi and a BDNF mimetic are sufficient to rescue retinal cell death and visual function in a vertebrate model of inherited blindness.
Collapse
|
34
|
Agrawal SA, Burgoyne T, Eblimit A, Bellingham J, Parfitt DA, Lane A, Nichols R, Asomugha C, Hayes MJ, Munro PM, Xu M, Wang K, Futter CE, Li Y, Chen R, Cheetham ME. REEP6 deficiency leads to retinal degeneration through disruption of ER homeostasis and protein trafficking. Hum Mol Genet 2017; 26:2667-2677. [PMID: 28475715 PMCID: PMC5808736 DOI: 10.1093/hmg/ddx149] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 04/12/2017] [Accepted: 04/14/2017] [Indexed: 01/09/2023] Open
Abstract
Retinitis pigmentosa (RP) is the most common form of inherited retinal dystrophy. We recently identified mutations in REEP6, which encodes the receptor expression enhancing protein 6, in several families with autosomal recessive RP. REEP6 is related to the REEP and Yop1p family of ER shaping proteins and potential receptor accessory proteins, but the role of REEP6 in the retina is unknown. Here we characterize the disease mechanisms associated with loss of REEP6 function using a Reep6 knockout mouse generated by CRISPR/Cas9 gene editing. In control mice REEP6 was localized to the inner segment and outer plexiform layer of rod photoreceptors. The Reep6-/- mice exhibited progressive photoreceptor degeneration from P20 onwards. Ultrastructural analyses at P20 by transmission electron microscopy and 3View serial block face scanning EM revealed an expansion of the distal ER in the Reep6-/- rods and an increase in their number of mitochondria. Electroretinograms revealed photoreceptor dysfunction preceded degeneration, suggesting potential defects in phototransduction. There was no effect on the traffic of rhodopsin, Rom1 or peripherin/rds; however, the retinal guanylate cyclases GC1 and GC2 were severely affected in the Reep6 knockout animals, with almost undetectable expression. These changes correlated with an increase in C/EBP homologous protein (CHOP) expression and the activation of caspase 12, suggesting that ER stress contributes to cell death. Collectively, these data suggest that REEP6 plays an essential role in maintaining cGMP homeostasis though facilitating the stability and/or trafficking of guanylate cyclases and maintaining ER and mitochondrial homeostasis.
Collapse
Affiliation(s)
- Smriti A. Agrawal
- Department of Molecular and Human Genetics
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030-3411, USA
| | - Thomas Burgoyne
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Aiden Eblimit
- Department of Molecular and Human Genetics
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030-3411, USA
| | - James Bellingham
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - David A. Parfitt
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Amelia Lane
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | | | - Chinwe Asomugha
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030-3411, USA
| | - Matthew J. Hayes
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Peter M. Munro
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Mingchu Xu
- Department of Molecular and Human Genetics
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030-3411, USA
| | - Keqing Wang
- Department of Molecular and Human Genetics
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030-3411, USA
| | - Clare E. Futter
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Yumei Li
- Department of Molecular and Human Genetics
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030-3411, USA
| | - Rui Chen
- Department of Molecular and Human Genetics
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030-3411, USA
| | | |
Collapse
|
35
|
Whole exome sequencing using Ion Proton system enables reliable genetic diagnosis of inherited retinal dystrophies. Sci Rep 2017; 7:42078. [PMID: 28181551 PMCID: PMC5299602 DOI: 10.1038/srep42078] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/05/2017] [Indexed: 01/17/2023] Open
Abstract
Inherited retinal dystrophies (IRD) comprise a wide group of clinically and genetically complex diseases that progressively affect the retina. Over recent years, the development of next-generation sequencing (NGS) methods has transformed our ability to diagnose heterogeneous diseases. In this work, we have evaluated the implementation of whole exome sequencing (WES) for the molecular diagnosis of IRD. Using Ion ProtonTM system, we simultaneously analyzed 212 genes that are responsible for more than 25 syndromic and non-syndromic IRD. This approach was used to evaluate 59 unrelated families, with the pathogenic variant(s) successfully identified in 71.18% of cases. Interestingly, the mutation detection rate varied substantially depending on the IRD subtype. Overall, we found 63 different mutations (21 novel) in 29 distinct genes, and performed in vivo functional studies to determine the deleterious impact of variants identified in MERTK, CDH23, and RPGRIP1. In addition, we provide evidences that support CDHR1 as a gene responsible for autosomal recessive retinitis pigmentosa with early macular affectation, and present data regarding the disease mechanism of this gene. Altogether, these results demonstrate that targeted WES of all IRD genes is a reliable, hypothesis-free approach, and a cost- and time-effective strategy for the routine genetic diagnosis of retinal dystrophies.
Collapse
|
36
|
Ramkumar HL, Gudiseva HV, Kishaba KT, Suk JJ, Verma R, Tadimeti K, Thorson JA, Ayyagari R. A Report on Molecular Diagnostic Testing for Inherited Retinal Dystrophies by Targeted Genetic Analyses. Genet Test Mol Biomarkers 2016; 21:66-73. [PMID: 28005406 DOI: 10.1089/gtmb.2016.0251] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
AIM To test the utility of targeted sequencing as a method of clinical molecular testing in patients diagnosed with inherited retinal degeneration (IRD). METHODS After genetic counseling, peripheral blood was drawn from 188 probands and 36 carriers of IRD. Single gene testing was performed on each patient in a Clinical Laboratory Improvement Amendment (CLIA) certified laboratory. DNA was isolated, and all exons in the gene of interest were analyzed along with 20 base pairs of flanking intronic sequence. Genetic testing was most often performed on ABCA4, CTRP5, ELOV4, BEST1, CRB1, and PRPH2. Pathogenicity of novel sequence changes was predicted by PolyPhen2 and sorting intolerant from tolerant (SIFT). RESULTS Of the 225 genetic tests performed, 150 were for recessive IRD, and 75 were for dominant IRD. A positive molecular diagnosis was made in 70 (59%) of probands with recessive IRD and 19 (26%) probands with dominant IRD. Analysis confirmed 12 (34%) of individuals as carriers of familial mutations associated with IRD. Thirty-two novel variants were identified; among these, 17 sequence changes in four genes were predicted to be possibly or probably damaging including: ABCA4 (14), BEST1 (2), PRPH2 (1), and TIMP3 (1). CONCLUSIONS Targeted analysis of clinically suspected genes in 225 subjects resulted in a positive molecular diagnosis in 26% of patients with dominant IRD and 59% of patients with recessive IRD. Novel damaging mutations were identified in four genes. Single gene screening is not an ideal method for diagnostic testing given the phenotypic and genetic heterogeneity among IRD cases. High-throughput sequencing of all genes associated with retinal degeneration may be more efficient for molecular diagnosis.
Collapse
Affiliation(s)
- Hema L Ramkumar
- 1 Shiley Eye Institute, Jacobs Retina Center, University of California , San Diego, La Jolla, California
| | - Harini V Gudiseva
- 1 Shiley Eye Institute, Jacobs Retina Center, University of California , San Diego, La Jolla, California
| | - Kameron T Kishaba
- 1 Shiley Eye Institute, Jacobs Retina Center, University of California , San Diego, La Jolla, California
| | - John J Suk
- 1 Shiley Eye Institute, Jacobs Retina Center, University of California , San Diego, La Jolla, California
| | - Rohan Verma
- 1 Shiley Eye Institute, Jacobs Retina Center, University of California , San Diego, La Jolla, California
| | - Keerti Tadimeti
- 1 Shiley Eye Institute, Jacobs Retina Center, University of California , San Diego, La Jolla, California
| | - John A Thorson
- 2 Department of Pathology, University of California , San Diego, La Jolla, California
| | - Radha Ayyagari
- 1 Shiley Eye Institute, Jacobs Retina Center, University of California , San Diego, La Jolla, California
| |
Collapse
|
37
|
Arno G, Agrawal SA, Eblimit A, Bellingham J, Xu M, Wang F, Chakarova C, Parfitt DA, Lane A, Burgoyne T, Hull S, Carss KJ, Fiorentino A, Hayes MJ, Munro PM, Nicols R, Pontikos N, Holder GE, Asomugha C, Raymond FL, Moore AT, Plagnol V, Michaelides M, Hardcastle AJ, Li Y, Cukras C, Webster AR, Cheetham ME, Chen R. Mutations in REEP6 Cause Autosomal-Recessive Retinitis Pigmentosa. Am J Hum Genet 2016; 99:1305-1315. [PMID: 27889058 PMCID: PMC5142109 DOI: 10.1016/j.ajhg.2016.10.008] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 10/13/2016] [Indexed: 12/15/2022] Open
Abstract
Retinitis pigmentosa (RP) is the most frequent form of inherited retinal dystrophy. RP is genetically heterogeneous and the genes identified to date encode proteins involved in a wide range of functional pathways, including photoreceptor development, phototransduction, the retinoid cycle, cilia, and outer segment development. Here we report the identification of biallelic mutations in Receptor Expression Enhancer Protein 6 (REEP6) in seven individuals with autosomal-recessive RP from five unrelated families. REEP6 is a member of the REEP/Yop1 family of proteins that influence the structure of the endoplasmic reticulum but is relatively unstudied. The six variants identified include three frameshift variants, two missense variants, and a genomic rearrangement that disrupts exon 1. Human 3D organoid optic cups were used to investigate REEP6 expression and confirmed the expression of a retina-specific isoform REEP6.1, which is specifically affected by one of the frameshift mutations. Expression of the two missense variants (c.383C>T [p.Pro128Leu] and c.404T>C [p.Leu135Pro]) and the REEP6.1 frameshift mutant in cultured cells suggest that these changes destabilize the protein. Furthermore, CRISPR-Cas9-mediated gene editing was used to produce Reep6 knock-in mice with the p.Leu135Pro RP-associated variant identified in one RP-affected individual. The homozygous knock-in mice mimic the clinical phenotypes of RP, including progressive photoreceptor degeneration and dysfunction of the rod photoreceptors. Therefore, our study implicates REEP6 in retinal homeostasis and highlights a pathway previously uncharacterized in retinal dystrophy.
Collapse
Affiliation(s)
- Gavin Arno
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK; Moorfields Eye Hospital, London EC1V 2PD, UK
| | - Smriti A Agrawal
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030-3411, USA; Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030-3411, USA
| | - Aiden Eblimit
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030-3411, USA; Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030-3411, USA
| | - James Bellingham
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Mingchu Xu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030-3411, USA; Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030-3411, USA
| | - Feng Wang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030-3411, USA; Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030-3411, USA
| | | | - David A Parfitt
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Amelia Lane
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Thomas Burgoyne
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Sarah Hull
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK; Moorfields Eye Hospital, London EC1V 2PD, UK
| | - Keren J Carss
- NIHR BioResource - Rare Diseases, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; Department of Haematology, University of Cambridge, NHS Blood and Transplant Centre, Cambridge CB2 0PT, UK
| | | | - Matthew J Hayes
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Peter M Munro
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Ralph Nicols
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030-3411, USA
| | - Nikolas Pontikos
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Graham E Holder
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK; Moorfields Eye Hospital, London EC1V 2PD, UK
| | - Chinwe Asomugha
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030-3411, USA
| | - F Lucy Raymond
- NIHR BioResource - Rare Diseases, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK
| | - Anthony T Moore
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK; Moorfields Eye Hospital, London EC1V 2PD, UK; Ophthalmology Department, UCSF School of Medicine, Koret Vision Center, San Francisco, CA 94133-0644, USA
| | - Vincent Plagnol
- UCL Genetics Institute, University College London, London WC1E 6BT, UK
| | - Michel Michaelides
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK; Moorfields Eye Hospital, London EC1V 2PD, UK
| | | | - Yumei Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030-3411, USA; Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030-3411, USA
| | | | - Andrew R Webster
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK; Moorfields Eye Hospital, London EC1V 2PD, UK
| | | | - Rui Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030-3411, USA; Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030-3411, USA.
| |
Collapse
|