1
|
Hensman J, van Schooneveld MJ, Diederen RMH, Plomp AS, de Muijnck C, Ten Brink JB, Florijn RJ, de Baere E, van Genderen MM, Boon CJF. Pseudocolobomatous autosomal dominant atrophic maculopathy (PADAM). Ophthalmic Genet 2025:1-6. [PMID: 40415333 DOI: 10.1080/13816810.2025.2492045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/05/2025] [Accepted: 04/07/2025] [Indexed: 05/27/2025]
Abstract
PURPOSE To describe a family with a previously unreported maculopathy across three generations. METHODS This retrospective chart study describes three patients from three generations of a non-consanguineous Dutch family, with a distinctive maculopathy. All three patients underwent extensive ophthalmic examinations and multimodal imaging including best-corrected visual acuity, fundus photography, spectral-domain optical coherence tomography, fundus autofluorescence imaging, and full-field electroretinography (ffERG). Genetic analyses included next-generation sequencing, whole-exome sequencing, and single nucleotide polymorphism arrays. RESULTS Three affected family members had a history of low visual acuity and congenital nystagmus, in combination with sharply demarcated areas of chorioretinal atrophy in the macula, which developed from early childhood. The two adult patients who underwent ffERG had cone and rod responses within normal limits, suggesting a central condition with a normally functioning retina extending beyond the lesions. No (likely) pathogenic variants in the known disease associated genes were found through extensive genetic analysis. CONCLUSION Pseudocolobomatous autosomal dominant atrophic maculopathy (PADAM) is a striking hereditary maculopathy that leads to progressive central vision loss. Future studies may provide additional insights into the genetic basis and underlying mechanisms of this remarkable clinical picture.
Collapse
Affiliation(s)
- Jonathan Hensman
- Department of Ophthalmology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Mary J van Schooneveld
- Department of Ophthalmology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Roselie M H Diederen
- Department of Ophthalmology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Astrid S Plomp
- Department of Human Genetics, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
| | - Cansu de Muijnck
- Diagnostic Centre for Complex Visual Disorders, Zeist, The Netherlands
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jacoline B Ten Brink
- Department of Human Genetics, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Ralph J Florijn
- Department of Human Genetics, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Elfride de Baere
- Center for Medical Genetics Ghent, Ghent University Hospital & Ghent, University, Ghent, Belgium
| | - Maria M van Genderen
- Diagnostic Centre for Complex Visual Disorders, Zeist, The Netherlands
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Camiel J F Boon
- Department of Ophthalmology, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
2
|
Gómez-Escribano AP, García-García G, Pérez-Santamarina E, Aller-Mañas E, Vázquez-Manrique RP, Millán-Salvador JM. Innovative therapies for inherited retinal dystrophies: navigating DNA, RNA, and protein approaches. EBioMedicine 2025; 116:105751. [PMID: 40367639 DOI: 10.1016/j.ebiom.2025.105751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/30/2025] [Accepted: 04/25/2025] [Indexed: 05/16/2025] Open
Abstract
Gene therapy has become a promising treatment for inherited retinal dystrophies (IRDs), but understanding the genetic mechanisms involved is essential for its success. Various approaches, such as gene augmentation and DNA/RNA-based therapies, have shown effectiveness in some clinical trials. However, gene augmentation is not effective for some dominant mutations and genome editing produces off-target effects. Here, we review safer and viable alternatives that are being evaluated in preclinical models and clinical trials to address this challenge. We propose a novel perspective based on protein-targeting therapies, which although promising, remain unexplored. We suggest that frameshift variants, which produce novel epitopes, may allow for the development of mutant protein targeting agents for selective protein degradation. This approach could be useful for dominant variants, where gene replacement is ineffective. By examining these approaches, we aim to guide more targeted and effective gene therapies for IRDs, offering potential treatments where current methods fall short.
Collapse
Affiliation(s)
- Ana Pilar Gómez-Escribano
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain; Joint Unit for Rare Diseases IIS La Fe-CIPF, Valencia, Spain.
| | - Gema García-García
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain; Joint Unit for Rare Diseases IIS La Fe-CIPF, Valencia, Spain
| | | | - Elena Aller-Mañas
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain; Joint Unit for Rare Diseases IIS La Fe-CIPF, Valencia, Spain
| | - Rafael Pascual Vázquez-Manrique
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain; Joint Unit for Rare Diseases IIS La Fe-CIPF, Valencia, Spain
| | - José María Millán-Salvador
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain; Joint Unit for Rare Diseases IIS La Fe-CIPF, Valencia, Spain.
| |
Collapse
|
3
|
Spagnuolo V, Piergentili M, Passerini I, Murro V, Mucciolo DP, Giorgio D, Maccari M, Pelo E, Biagini I, Giansanti F, Virgili G, Sodi A. Retinal Dystrophies Associated with Mutations in the RP1 Gene: Genotype-Phenotype Correlations. Curr Issues Mol Biol 2025; 47:212. [PMID: 40136466 PMCID: PMC11941481 DOI: 10.3390/cimb47030212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/26/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND We evaluated the genetic and phenotypic features of a cohort of 10 Italian patients affected by Retinitis Pigmentosa (RP) associated with RP1 sequence variants. METHODS A retrospective, cross-sectional genotype-phenotype correlation study was conducted on a cohort of ten Italian patients (four males and six females) seen at Careggi University Hospital between 2012 and 2024, all affected by RP carrying pathogenic variants in the RP1 gene. A comprehensive ophthalmic assessment and pedigree analysis were performed, focusing on the onset of disease symptoms, the patient's age at first diagnosis, follow-up duration, and the presence of comorbidities. RESULTS Our cohort included ten Italian patients with a mean age of 59 (range of 32-79 years). The median age when symptoms first presented was 43 years (range of 2-74), with a mean follow-up period of 9.3 ± 2.6 years. The main symptoms at presentation were hemeralopia and visual field constriction. Fundus examination revealed a classic RP phenotype. Fundus autofluorescence (FAF), optical coherence tomography (OCT), Electroretinogram (ERG), and visual field testing confirmed the typical features of classic retinitis pigmentosa in most cases. CONCLUSIONS This single-center cohort of Italian patients provides insights into the clinical and genetic characteristics of RP1-associated RP. By comprehensively identifying genetic variations and their associated clinical manifestations, therapeutic interventions targeting specific genetic abnormalities can be better tailored. This approach holds promise for improving the prognosis and quality of life for individuals with RP1-associated RP.
Collapse
Affiliation(s)
- Vito Spagnuolo
- Eye Clinic, Department of Neuromuscular Diseases and Sense Organs, Careggi University Hospital, 50134 Florence, Italy; (M.P.); (V.M.); (D.P.M.); (D.G.); (I.B.); (F.G.); (G.V.)
- Department of Neuroscience, Psychology, Pharmacology and Pediatrics (Neurofarba), University of Florence, 50134 Florence, Italy
| | - Marco Piergentili
- Eye Clinic, Department of Neuromuscular Diseases and Sense Organs, Careggi University Hospital, 50134 Florence, Italy; (M.P.); (V.M.); (D.P.M.); (D.G.); (I.B.); (F.G.); (G.V.)
- Department of Neuroscience, Psychology, Pharmacology and Pediatrics (Neurofarba), University of Florence, 50134 Florence, Italy
| | - Ilaria Passerini
- Department of Genetic Diagnostics, Careggi University Hospital, 50134 Florence, Italy; (I.P.); (E.P.)
| | - Vittoria Murro
- Eye Clinic, Department of Neuromuscular Diseases and Sense Organs, Careggi University Hospital, 50134 Florence, Italy; (M.P.); (V.M.); (D.P.M.); (D.G.); (I.B.); (F.G.); (G.V.)
- Department of Neuroscience, Psychology, Pharmacology and Pediatrics (Neurofarba), University of Florence, 50134 Florence, Italy
| | - Dario Pasquale Mucciolo
- Eye Clinic, Department of Neuromuscular Diseases and Sense Organs, Careggi University Hospital, 50134 Florence, Italy; (M.P.); (V.M.); (D.P.M.); (D.G.); (I.B.); (F.G.); (G.V.)
- Department of Neuroscience, Psychology, Pharmacology and Pediatrics (Neurofarba), University of Florence, 50134 Florence, Italy
| | - Dario Giorgio
- Eye Clinic, Department of Neuromuscular Diseases and Sense Organs, Careggi University Hospital, 50134 Florence, Italy; (M.P.); (V.M.); (D.P.M.); (D.G.); (I.B.); (F.G.); (G.V.)
- Department of Neuroscience, Psychology, Pharmacology and Pediatrics (Neurofarba), University of Florence, 50134 Florence, Italy
- Department of Ophthalmology, Livorno Hospital, 57124 Livorno, Italy
| | - Martina Maccari
- Eye Clinic, Clinical Trial Center, Careggi University Hospital, 50134 Florence, Italy;
| | - Elisabetta Pelo
- Department of Genetic Diagnostics, Careggi University Hospital, 50134 Florence, Italy; (I.P.); (E.P.)
| | - Ilaria Biagini
- Eye Clinic, Department of Neuromuscular Diseases and Sense Organs, Careggi University Hospital, 50134 Florence, Italy; (M.P.); (V.M.); (D.P.M.); (D.G.); (I.B.); (F.G.); (G.V.)
- Department of Neuroscience, Psychology, Pharmacology and Pediatrics (Neurofarba), University of Florence, 50134 Florence, Italy
| | - Fabrizio Giansanti
- Eye Clinic, Department of Neuromuscular Diseases and Sense Organs, Careggi University Hospital, 50134 Florence, Italy; (M.P.); (V.M.); (D.P.M.); (D.G.); (I.B.); (F.G.); (G.V.)
- Department of Neuroscience, Psychology, Pharmacology and Pediatrics (Neurofarba), University of Florence, 50134 Florence, Italy
| | - Gianni Virgili
- Eye Clinic, Department of Neuromuscular Diseases and Sense Organs, Careggi University Hospital, 50134 Florence, Italy; (M.P.); (V.M.); (D.P.M.); (D.G.); (I.B.); (F.G.); (G.V.)
- Department of Neuroscience, Psychology, Pharmacology and Pediatrics (Neurofarba), University of Florence, 50134 Florence, Italy
- IRCCS—Fondazione Bietti, 00184 Rome, Italy
| | - Andrea Sodi
- Eye Clinic, Department of Neuromuscular Diseases and Sense Organs, Careggi University Hospital, 50134 Florence, Italy; (M.P.); (V.M.); (D.P.M.); (D.G.); (I.B.); (F.G.); (G.V.)
- Department of Neuroscience, Psychology, Pharmacology and Pediatrics (Neurofarba), University of Florence, 50134 Florence, Italy
| |
Collapse
|
4
|
D'Esposito F, Gagliano C, Avitabile A, Gagliano G, Musa M, Capobianco M, Visalli F, Dammino E, Zeppieri M, Cordeiro MF. Exploring Molecular Pathways in Refractive Errors Associated with Inherited Retinal Dystrophies. FRONT BIOSCI-LANDMRK 2025; 30:25584. [PMID: 40018922 DOI: 10.31083/fbl25584] [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: 07/08/2024] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 03/01/2025]
Abstract
The term inherited retinal dystrophies (IRDs) refers to a diverse range of conditions characterized by retinal dysfunction, and mostly deterioration, leading to a gradual decay of the visual function and eventually to total vision loss. IRDs have a global impact on about 1 in every 3000 to 4000 individuals. However, the prevalence statistics might differ significantly depending on the exact type of dystrophy and the demographic being examined. The cellular pathophysiology and genetic foundation of IRDs have been extensively studied, however, knowledge regarding associated refractive errors remain limited. This review aims to clarify the cellular and molecular processes that underlie refractive errors in IRDs. We did a thorough search of the current literature (Pubmed, accession Feb 2024), selecting works describing phenotypic differences among genes-related to IRDs, particularly in relation to refractive errors. First, we summarize the wide range of IRDs and their genetic causes, describing the genes and biological pathways connected to the etiology of the disease. We then explore the complex relationship between refractive errors and retinal dysfunction, including how the impairment of the vision-related mechanisms in the retina can affect ocular biometry and optical characteristics. New data about the involvement of aberrant signaling pathways, photoreceptor degeneration, and dysfunctional retinal pigment epithelium (RPE) in the development of refractive errors in IRDs have been examined. We also discuss the therapeutic implications of refractive defects in individuals with IRD, including possible approaches to treating visual impairments. In addition, we address the value of using cutting-edge imaging methods and animal models to examine refractive errors linked to IRDs and suggest future lines of inquiry for identifying new targets for treatment. In summary, this study presents an integrated understanding of the cellular and molecular mechanisms underlying refractive errors in IRDs. It illuminates the intricacies of ocular phenotypes in these conditions and offers a tool for understanding mechanisms underlying isolated refractive errors, besides the IRD-related forms.
Collapse
Affiliation(s)
- Fabiana D'Esposito
- Imperial College Ophthalmic Research Group (ICORG) Unit, Imperial College, NW15QH London, UK
- Department of Neurosciences, Reproductive Sciences and Dentistry, University of Naples Federico II, 80131 Napoli, Italy
| | - Caterina Gagliano
- Department of Medicine and Surgery, University of Enna "Kore", Piazza dell'Università, 94100 Enna, Italy
- Mediterranean Foundation "G.B. Morgagni", 95125 Catania, Italy
| | | | | | - Mutali Musa
- Department of Optometry, University of Benin, 300238 Benin City, Edo State, Nigeria
| | | | | | - Edoardo Dammino
- Mediterranean Foundation "G.B. Morgagni", 95125 Catania, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, 33100 Udine, Italy
| | | |
Collapse
|
5
|
Zheng Y, Stormo GD, Chen S. Aberrant homeodomain-DNA cooperative dimerization underlies distinct developmental defects in two dominant CRX retinopathy models. Genome Res 2025; 35:242-256. [PMID: 39715683 PMCID: PMC11874979 DOI: 10.1101/gr.279340.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 12/10/2024] [Indexed: 12/25/2024]
Abstract
Paired-class homeodomain (HD) transcription factors (TFs) play essential roles in vertebrate development, and their mutations are linked to human diseases. One unique feature of a paired-class HD is cooperative dimerization on specific palindrome DNA sequences. Yet, the functional significance of HD cooperative dimerization in animal development and its dysregulation in diseases remains elusive. Using the retinal TF cone-rod homeobox (CRX) as a model, we have studied how blindness-causing mutations in the paired HD, p.E80A and p.K88N, alter CRX's cooperative dimerization, leading to gene misexpression and photoreceptor developmental deficits in dominant manners. CRXE80A maintains binding at monomeric WT CRX motifs but is deficient in cooperative binding at dimeric motifs. CRXE80A's cooperativity defect impacts the exponential increase of photoreceptor gene expression in terminal differentiation and produces immature, nonfunctional photoreceptors in the Crx E80A retinas. CRXK88N is highly cooperative and localizes to ectopic genomic sites with strong enrichment of dimeric HD motifs. CRXK88N's altered biochemical properties disrupt CRX's ability to direct dynamic chromatin remodeling during development to activate photoreceptor differentiation programs and silence progenitor programs. Our study provides in vitro and in vivo molecular evidence that paired-class HD cooperative dimerization regulates neuronal development and that dysregulation of cooperative binding contributes to severe dominant blinding retinopathies.
Collapse
Affiliation(s)
- Yiqiao Zheng
- Molecular Genetics and Genomics Graduate Program, Division of Biology & Biomedical Sciences, Washington University in St. Louis, St. Louis, Missouri 63110, USA
- Department of Ophthalmology and Visual Sciences, Washington University in St Louis, Saint Louis, Missouri 63110, USA
| | - Gary D Stormo
- Department of Genetics, Washington University in St Louis, Saint Louis, Missouri 63110, USA;
| | - Shiming Chen
- Department of Ophthalmology and Visual Sciences, Washington University in St Louis, Saint Louis, Missouri 63110, USA;
- Department of Developmental Biology, Washington University in St Louis, Saint Louis, Missouri 63110, USA
| |
Collapse
|
6
|
Szabó V, Varsányi B, Barboni M, Takács Á, Knézy K, Molnár MJ, Nagy ZZ, György B, Rivolta C. Insights into eye genetics and recent advances in ocular gene therapy. Mol Cell Probes 2025; 79:102008. [PMID: 39805344 DOI: 10.1016/j.mcp.2025.102008] [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: 12/18/2024] [Revised: 01/04/2025] [Accepted: 01/05/2025] [Indexed: 01/16/2025]
Abstract
The rapid advancements in the field of genetics have significantly propelled the development of gene therapies, paving the way for innovative treatments of various hereditary disorders. This review focuses on the genetics of ophthalmologic conditions, highlighting the currently approved ophthalmic gene therapy and exploring emerging therapeutic strategies under development. Inherited retinal dystrophies represent a heterogeneous group of genetic disorders that manifest across a broad spectrum from infancy to late middle age. Key clinical features include nyctalopia (night blindness), constriction of the visual field, impairments in color perception, reduced central visual acuity, and rapid eye movements. Recent technological advancements, such as multimodal imaging, psychophysical assessments, and electrophysiological testing, have greatly enhanced our ability to understand disease progression and establish genotype-phenotype correlations. Additionally, the integration of molecular diagnostics into clinical practice is revolutionizing patient stratification and the design of targeted interventions, underscoring the transformative potential of personalized medicine in ophthalmology. The review also covers the challenges and opportunities in developing gene therapies for other ophthalmic conditions, such as age-related macular degeneration and optic neuropathies. We discuss the viral and non-viral vector systems used in ocular gene therapy, highlighting their advantages and limitations. Additionally, we explore the potential of emerging technologies like CRISPR/Cas9 in treating genetic eye diseases. We briefly address the regulatory landscape, concerns, challenges, and future directions of gene therapy in ophthalmology. We emphasize the need for long-term safety and efficacy data as these innovative treatments move from bench to bedside.
Collapse
Affiliation(s)
- Viktória Szabó
- Semmelweis University, Department of Ophthalmology, Mária Str. 39, Budapest, 1085, Hungary.
| | - Balázs Varsányi
- Semmelweis University, Department of Ophthalmology, Mária Str. 39, Budapest, 1085, Hungary; Ganglion Medical Center, Váradi Str. 10/A, Pécs, 7621, Hungary.
| | - Mirella Barboni
- Semmelweis University, Department of Ophthalmology, Mária Str. 39, Budapest, 1085, Hungary; Institute of Molecular and Clinical Ophthalmology Basel, Mittlere Strasse 91, Basel, CH-4031, Switzerland.
| | - Ágnes Takács
- Semmelweis University, Department of Ophthalmology, Mária Str. 39, Budapest, 1085, Hungary.
| | - Krisztina Knézy
- Semmelweis University, Department of Ophthalmology, Mária Str. 39, Budapest, 1085, Hungary.
| | - Mária Judit Molnár
- Semmelweis University, Institute of Genomic Medicine and Rare Disorders, Gyulai Pál Str. 2, Budapest, 1085, Hungary.
| | - Zoltán Zsolt Nagy
- Semmelweis University, Department of Ophthalmology, Mária Str. 39, Budapest, 1085, Hungary.
| | - Bence György
- Institute of Molecular and Clinical Ophthalmology Basel, Mittlere Strasse 91, Basel, CH-4031, Switzerland; Department of Ophthalmology, University of Basel, Mittlere Strasse 91, Basel, CH-4031, Switzerland.
| | - Carlo Rivolta
- Institute of Molecular and Clinical Ophthalmology Basel, Mittlere Strasse 91, Basel, CH-4031, Switzerland.
| |
Collapse
|
7
|
Lynn J, Huang SJ, Trigler GK, Kingsley R, Coussa RG, Bennett LD. Expanding the Mutation Spectrum for Inherited Retinal Diseases. Genes (Basel) 2024; 16:32. [PMID: 39858579 PMCID: PMC11764958 DOI: 10.3390/genes16010032] [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: 11/18/2024] [Revised: 12/22/2024] [Accepted: 12/26/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Inherited retinal diseases (IRDs) represent a diverse group of genetic disorders characterized by degeneration of the retina, leading to visual impairment and blindness. IRDs are heterogeneous, sharing common clinical features that can be difficult to diagnose without knowing the genetic basis of the disease. To improve diagnostic accuracy and advance understanding of disease mechanisms, genetic testing was performed for 103 unrelated patients with an IRD at a single clinical site between 30 August 2022 and 5 February 2024. METHODS Informed consent was obtained before buccal samples were collected for panel-based sequencing at BluePrint Genetics (BpG), sponsored by the Foundation Fighting Blindness MyRetina Tracker program. A retina specialist performed standard visit assessments, including visual acuity (Snellen chart), slit lamp examination, fundus photography (Optos®, Dunfermline, UK), and spectral-domain optical coherence tomography (SD-OCT; Zeiss). RESULTS From 103 patients, genetic findings were reported for 70 individuals. Among these included 20 novel variants. CONCLUSIONS These results clarify and confirm clinical diagnoses, aid in counseling patients on prognosis and family planning, and guide treatment options. This study not only holds promise for affected individuals but also expands the mutation spectrum to guide understanding of IRD.
Collapse
Affiliation(s)
- Jacob Lynn
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73114, USA;
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73114, USA; (S.J.H.); (G.K.T.)
| | - Samuel J. Huang
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73114, USA; (S.J.H.); (G.K.T.)
- Dean McGee Eye Institute, Oklahoma City, OK 73140, USA; (R.K.); (R.G.C.)
| | - Grace K. Trigler
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73114, USA; (S.J.H.); (G.K.T.)
| | - Ronald Kingsley
- Dean McGee Eye Institute, Oklahoma City, OK 73140, USA; (R.K.); (R.G.C.)
| | - Razek G. Coussa
- Dean McGee Eye Institute, Oklahoma City, OK 73140, USA; (R.K.); (R.G.C.)
| | - Lea D. Bennett
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73114, USA;
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73114, USA; (S.J.H.); (G.K.T.)
- Dean McGee Eye Institute, Oklahoma City, OK 73140, USA; (R.K.); (R.G.C.)
| |
Collapse
|
8
|
García-Bohórquez B, Barberán-Martínez P, Aller E, Jaijo T, Mínguez P, Rodilla C, Fernández-Caballero L, Blanco-Kelly F, Ayuso C, Sanchis-Juan A, Broekman S, de Vrieze E, van Wijk E, García-García G, Millán JM. Exploring non-coding variants and evaluation of antisense oligonucleotides for splicing redirection in Usher syndrome. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102374. [PMID: 39629117 PMCID: PMC11612772 DOI: 10.1016/j.omtn.2024.102374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/24/2024] [Indexed: 12/06/2024]
Abstract
Exploring non-coding regions is increasingly gaining importance in the diagnosis of inherited retinal dystrophies. Deep-intronic variants causing aberrant splicing have been identified, prompting the development of antisense oligonucleotides (ASOs) to modulate splicing. We performed a screening of five previously described USH2A deep-intronic variants among USH2A monoallelic patients with Usher syndrome (USH) or isolated retinitis pigmentosa. Sequencing of entire USH2A or USH genes was then conducted in unresolved or newly monoallelic cases. The splicing impact of identified variants was assessed using minigene assays, and ASOs were designed to correct splicing. The screening allowed to diagnose 30.95% of the studied patients. The sequencing of USH genes revealed 16 new variants predicted to affect splicing, with four confirmed to affect splicing through minigene assays. Two of them were unreported deep-intronic variants and predicted to include a pseudoexon in the pre-mRNA, and the other two could alter a regulatory cis-element. ASOs designed for three USH2A deep-intronic variants successfully redirected splicing in vitro. Our study demonstrates the improvement in genetic characterization of IRDs when analyzing non-coding regions, highlighting that deep-intronic variants significantly contribute to USH2A pathogenicity. Furthermore, successful splicing modulation through ASOs highlights their therapeutic potential for patients carrying deep-intronic variants.
Collapse
Affiliation(s)
- Belén García-Bohórquez
- Molecular, Cellular and Genomics Biomedicine, Health Research Institute La Fe, 46026 Valencia, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Joint Unit CIPF-IIS La Fe Molecular, Cellular and Genomic Biomedicine, 46026 Valencia, Spain
| | - Pilar Barberán-Martínez
- Molecular, Cellular and Genomics Biomedicine, Health Research Institute La Fe, 46026 Valencia, Spain
- Joint Unit CIPF-IIS La Fe Molecular, Cellular and Genomic Biomedicine, 46026 Valencia, Spain
| | - Elena Aller
- Molecular, Cellular and Genomics Biomedicine, Health Research Institute La Fe, 46026 Valencia, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Joint Unit CIPF-IIS La Fe Molecular, Cellular and Genomic Biomedicine, 46026 Valencia, Spain
- University and Polytechnic La Fe Hospital of Valencia, 46026 Valencia, Spain
| | - Teresa Jaijo
- Molecular, Cellular and Genomics Biomedicine, Health Research Institute La Fe, 46026 Valencia, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Joint Unit CIPF-IIS La Fe Molecular, Cellular and Genomic Biomedicine, 46026 Valencia, Spain
- University and Polytechnic La Fe Hospital of Valencia, 46026 Valencia, Spain
| | - Pablo Mínguez
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain
- Bioinformatics Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain
| | - Cristina Rodilla
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain
| | - Lidia Fernández-Caballero
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain
| | - Fiona Blanco-Kelly
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain
| | - Carmen Ayuso
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain
| | - Alba Sanchis-Juan
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sanne Broekman
- Department of Otorhinolaryngology, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Erik de Vrieze
- Department of Otorhinolaryngology, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Erwin van Wijk
- Department of Otorhinolaryngology, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Gema García-García
- Molecular, Cellular and Genomics Biomedicine, Health Research Institute La Fe, 46026 Valencia, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Joint Unit CIPF-IIS La Fe Molecular, Cellular and Genomic Biomedicine, 46026 Valencia, Spain
| | - José M. Millán
- Molecular, Cellular and Genomics Biomedicine, Health Research Institute La Fe, 46026 Valencia, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Joint Unit CIPF-IIS La Fe Molecular, Cellular and Genomic Biomedicine, 46026 Valencia, Spain
- University and Polytechnic La Fe Hospital of Valencia, 46026 Valencia, Spain
| |
Collapse
|
9
|
Moseley J, Leest T, Larsson K, Magrelli A, Stoyanova-Beninska V. Inherited retinal dystrophies and orphan designations in the European Union. Eur J Ophthalmol 2024; 34:1631-1641. [PMID: 38500388 PMCID: PMC11542323 DOI: 10.1177/11206721241236214] [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: 10/02/2023] [Accepted: 02/13/2024] [Indexed: 03/20/2024]
Abstract
Inherited Retinal Dystrophies (IRD) are diverse rare diseases that affect the retina and lead to visual impairment or blindness. Research in this field is ongoing, with over 60 EU orphan medicinal products designated in this therapeutic area by the Committee for Orphan Medicinal Products (COMP) at the European Medicines Agency (EMA). Up to now, COMP has used traditional disease terms, like retinitis pigmentosa, for orphan designation regardless of the product's mechanism of action. The COMP reviewed the designation approach for IRDs taking into account all previous Orphan Designations (OD) experience in IRDs, the most relevant up to date scientific literature and input from patients and clinical experts. Following the review, the COMP decided that there should be three options available for orphan designation concerning the condition: i) an amended set of OD groups for therapies that might be used in a broad spectrum of conditions, ii) a gene-specific designation for targeted therapies, and iii) an occasional term for products that do not fit in the above two categories. The change in the approach to orphan designation in IRDs caters for different scenarios to allow an optimum approach for future OD applications including the option of a gene-specific designation. By applying this new approach, the COMP increases the regulatory clarity, efficiency, and predictability for sponsors, aligns EU regulatory tools with the latest scientific and medical developments in the field of IRDs, and ensures that all potentially treatable patients will be included in the scope of an OD.
Collapse
Affiliation(s)
- Jane Moseley
- European Medicines Agency, Amsterdam, The Netherlands
| | - Tim Leest
- Committee for Orphan Medicinal Products at the European Medicines Agency, Amsterdam, The Netherlands
- Federal Agency for Medicines and Health Products, Brussels, Belgium
| | | | - Armando Magrelli
- Committee for Orphan Medicinal Products at the European Medicines Agency, Amsterdam, The Netherlands
- National Center for Drug Research and Evaluation- Istituto Superiore di Sanità, Rome, Italy
| | - Violeta Stoyanova-Beninska
- Committee for Orphan Medicinal Products at the European Medicines Agency, Amsterdam, The Netherlands
- Medicines Evaluation Board (MEB), Utrecht, The Netherlands
| |
Collapse
|
10
|
Del Pozo-Valero M, Almoallem B, Dueñas Rey A, Mahieu Q, Van Heetvelde M, Jeddawi L, Bauwens M, De Baere E. Autozygome-guided exome-first study in a consanguineous cohort with early-onset retinal disease uncovers an isolated RIMS2 phenotype and a retina-enriched RIMS2 isoform. Clin Genet 2024; 106:127-139. [PMID: 38468396 DOI: 10.1111/cge.14517] [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: 12/20/2023] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 03/13/2024]
Abstract
Leber congenital amaurosis (LCA) and early-onset retinal degeneration (EORD) are inherited retinal diseases (IRD) characterized by early-onset vision impairment. Herein, we studied 15 Saudi families by whole exome sequencing (WES) and run-of-homozygosity (ROH) detection via AutoMap in 12/15 consanguineous families. This revealed (likely) pathogenic variants in 11/15 families (73%). A potential founder variant was found in RPGRIP1. Homozygous pathogenic variants were identified in known IRD genes (ATF6, CRB1, CABP4, RDH12, RIMS2, RPGRIP1, SPATA7). We established genotype-driven clinical reclassifications for ATF6, CABP4, and RIMS2. Specifically, we observed isolated IRD in the individual with the novel RIMS2 variant, and we found a retina-enriched RIMS2 isoform conserved but not annotated in mouse. The latter illustrates potential different phenotypic consequences of pathogenic variants depending on the particular tissue/cell-type specific isoforms they affect. Lastly, a compound heterozygous genotype in GUCY2D in one non-consanguineous family was demonstrated, and homozygous variants in novel candidate genes ATG2B and RUFY3 were found in the two remaining consanguineous families. Reporting these genes will allow to validate them in other IRD cohorts. Finally, the missing heritability of the two unsolved IRD cases may be attributed to variants in non-coding regions or structural variants that remained undetected, warranting future WGS studies.
Collapse
Affiliation(s)
- Marta Del Pozo-Valero
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Basamat Almoallem
- Department of Ophthalmology, College of Medicine, King Saud University (KSU), Riyadh, Saudi Arabia
- Department of Ophthalmology, King Saud University Medical City (KSUMC), Riyadh, Saudi Arabia
| | - Alfredo Dueñas Rey
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Quinten Mahieu
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Mattias Van Heetvelde
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Laila Jeddawi
- Pediatric Ophthalmology Division, Dr. Sulaiman AL Habib Medical Group, AL Khobar, Saudi Arabia
| | - Miriam Bauwens
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Elfride De Baere
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
11
|
Ibrahim M, Jaffal L, Assi A, Helou C, El Shamieh S. ABCA4-related retinopathies in Lebanon. Heliyon 2024; 10:e30304. [PMID: 38694055 PMCID: PMC11061736 DOI: 10.1016/j.heliyon.2024.e30304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024] Open
Abstract
Variants in ATP-binding cassette transporter type A4 (ABCA4) have been linked to several forms of inherited retinal diseases (IRDs) besides the classically defined Stargardt disease (STGD), known collectively as ABCA4 retinopathies. ABCA4 is a sizable locus harboring 50 exons; thus, its analysis has revealed over 2,400 variants described, of which more than 2,000 are causal. Due to the clinical and genetic heterogeneity, diagnosing ABCA4 retinopathies is challenging. To date, no ABCA4-related retinopathy has been detected in Lebanon. Using next-generation sequencing, we analyzed our IRDs' cohort retrospectively (61 families) and identified five with ABCA4-related retinopathies, making it a relatively abundant cause of IRDs (about 8 %). Three families were diagnosed with rod-cone dystrophy (RCD), two with STGD, and one with cone-rod dystrophy (CRD). In conclusion, our study showed the presence of ABCA4 variants with a high degree of heterogeneity in Lebanon.
Collapse
Affiliation(s)
- Mariam Ibrahim
- Molecular Testing Laboratory, Department of Medical Laboratory Technology, Faculty of Health Sciences, Beirut Arab University, Beirut, Lebanon
- Rammal Hassan Rammal Research Laboratory, PhyToxE Research Group, Faculty of Sciences, Lebanese University, Nabatieh, Lebanon
| | - Lama Jaffal
- Department of Biological and Chemical Sciences, School of Arts and Sciences, Lebanese International University, Beirut, Lebanon
| | | | - Charles Helou
- Retinal Service, Beirut Eye & ENT Specialist Hospital, Beirut, Lebanon
| | - Said El Shamieh
- Molecular Testing Laboratory, Department of Medical Laboratory Technology, Faculty of Health Sciences, Beirut Arab University, Beirut, Lebanon
| |
Collapse
|
12
|
Zheng Y, Stormo GD, Chen S. Aberrant homeodomain-DNA cooperative dimerization underlies distinct developmental defects in two dominant CRX retinopathy models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584677. [PMID: 38559186 PMCID: PMC10979960 DOI: 10.1101/2024.03.12.584677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Paired-class homeodomain transcription factors (HD TFs) play essential roles in vertebrate development, and their mutations are linked to human diseases. One unique feature of paired-class HD is cooperative dimerization on specific palindrome DNA sequences. Yet, the functional significance of HD cooperative dimerization in animal development and its dysregulation in diseases remain elusive. Using the retinal TF Cone-rod Homeobox (CRX) as a model, we have studied how blindness-causing mutations in the paired HD, p.E80A and p.K88N, alter CRX's cooperative dimerization, lead to gene misexpression and photoreceptor developmental deficits in dominant manners. CRXE80A maintains binding at monomeric WT CRX motifs but is deficient in cooperative binding at dimeric motifs. CRXE80A's cooperativity defect impacts the exponential increase of photoreceptor gene expression in terminal differentiation and produces immature, non-functional photoreceptors in the CrxE80A retinas. CRXK88N is highly cooperative and localizes to ectopic genomic sites with strong enrichment of dimeric HD motifs. CRXK88N's altered biochemical properties disrupt CRX's ability to direct dynamic chromatin remodeling during development to activate photoreceptor differentiation programs and silence progenitor programs. Our study here provides in vitro and in vivo molecular evidence that paired-class HD cooperative dimerization regulates neuronal development and dysregulation of cooperative binding contributes to severe dominant blinding retinopathies.
Collapse
Affiliation(s)
- Yiqiao Zheng
- Molecular Genetics and Genomics Graduate Program, Division of Biological and Biomedical Sciences, Washington University in St Louis, Saint Louis, Missouri, 63110, USA
- Department of Ophthalmology and Visual Sciences, Washington University in St Louis, Saint Louis, Missouri, 63110, USA
| | - Gary D. Stormo
- Department of Genetics, Washington University in St Louis, Saint Louis, Missouri, 63110, USA
| | - Shiming Chen
- Department of Ophthalmology and Visual Sciences, Washington University in St Louis, Saint Louis, Missouri, 63110, USA
- Department of Developmental Biology, Washington University in St Louis, Saint Louis, Missouri, 63110, USA
| |
Collapse
|
13
|
Esteve-Garcia A, Cobos E, Sau C, Padró-Miquel A, Català-Mora J, Barberán-Martínez P, Millán JM, García-García G, Aguilera C. Deciphering complexity: TULP1 variants linked to an atypical retinal dystrophy phenotype. Front Genet 2024; 15:1352063. [PMID: 38450199 PMCID: PMC10915255 DOI: 10.3389/fgene.2024.1352063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 01/29/2024] [Indexed: 03/08/2024] Open
Abstract
Introduction: TULP1 exemplifies the remarkable clinical and genetic heterogeneity observed in inherited retinal dystrophies. Our research describes the clinical and molecular characteristics of a patient manifesting an atypical retinal dystrophy pattern, marked by the identification of both a previously unreported and a rarely encountered TULP1 variant. Methods: Whole-exome sequencing was performed to identify potential causative variants. The pathogenicity of the identified TULP1 variants was evaluated through in silico predictors and a minigene splice assay, specifically designed to assess the effect of the unreported TULP1 variant. Results: We identified two TULP1 gene variants in a patient exhibiting unusual and symmetrical alterations in both retinas, characterized by an increase in autofluorescence along the distribution of retinal vessels. These variants included a known rare missense variant, c.1376T>C, and a novel splice site variant, c.822G>T. For the latter variant (c.822G>T), we conducted a minigene splice assay that demonstrated the incorporation of a premature stop codon. This finding suggests a likely activation of the nonsense-mediated mRNA decay mechanism, ultimately resulting in the absence of protein production from this allele. Segregation analysis confirmed that these variants were in trans. Discussion: Our data support that individuals with biallelic TULP1 variants may present with a unique pattern of macular degeneration and periarteriolar vascular pigmentation. This study highlights the importance of further clinical and molecular characterization of TULP1 variants to elucidate genotype-phenotype correlations in the context of inherited retinal dystrophies.
Collapse
Affiliation(s)
- Anna Esteve-Garcia
- Department of Clinical Genetics, Bellvitge University Hospital, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Estefania Cobos
- Department of Ophthalmology, Bellvitge University Hospital, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Cristina Sau
- Department of Clinical Genetics, Bellvitge University Hospital, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ariadna Padró-Miquel
- Genetics Laboratory, Bellvitge University Hospital, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Jaume Català-Mora
- Department of Ophthalmology, SJD Barcelona Children’s Hospital, Barcelona, Spain
| | - Pilar Barberán-Martínez
- Molecular, Cellular, and Genomic Biomedicine Group, Valencia, Spain
- Joint Unit CIPF-IIS La Fe Molecular, Cellular and Genomic Biomedicine, Valencia, Spain
| | - José M. Millán
- Molecular, Cellular, and Genomic Biomedicine Group, Valencia, Spain
- Joint Unit CIPF-IIS La Fe Molecular, Cellular and Genomic Biomedicine, Valencia, Spain
- Center for Rare Diseases (CIBERER), Madrid, Spain
- University and Polytechnic La Fe Hospital of Valencia, Valencia, Spain
| | - Gema García-García
- Molecular, Cellular, and Genomic Biomedicine Group, Valencia, Spain
- Joint Unit CIPF-IIS La Fe Molecular, Cellular and Genomic Biomedicine, Valencia, Spain
- Center for Rare Diseases (CIBERER), Madrid, Spain
| | - Cinthia Aguilera
- Genetics Laboratory, Bellvitge University Hospital, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
14
|
Kiraly P, Cottriall CL, Taylor LJ, Jolly JK, Cehajic-Kapetanovic J, Yusuf IH, Martinez-Fernandez de la Camara C, Shanks M, Downes SM, MacLaren RE, Fischer MD. Outcomes and Adverse Effects of Voretigene Neparvovec Treatment for Biallelic RPE65-Mediated Inherited Retinal Dystrophies in a Cohort of Patients from a Single Center. Biomolecules 2023; 13:1484. [PMID: 37892166 PMCID: PMC10605275 DOI: 10.3390/biom13101484] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/27/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
Our study evaluated the morphological and functional outcomes, and the side effects, of voretigene neparvovec (VN) gene therapy for RPE65-mediated inherited retinal dystrophies (IRDs) in 12 eyes (six patients) at the Oxford Eye Hospital with a mean follow-up duration of 8.2 (range 1-12) months. All patients reported a subjective vision improvement 1 month after gene therapy. Best-corrected visual acuity (BCVA) remained stable (baseline: 1.28 (±0.71) vs. last follow-up: 1.46 (±0.60); p = 0.25). Average white Full-Field Stimulus Testing (FST) showed a trend towards improvement (baseline: -4.41 (±10.62) dB vs. last follow-up: -11.98 (±13.83) dB; p = 0.18). No changes in central retinal thickness or macular volume were observed. The side effects included mild intraocular inflammation (two eyes) and cataracts (four eyes). Retinal atrophy occurred in 10 eyes (eight mild, two severe) but did not impact FST measurements during the follow-up period. Increased intraocular pressure (IOP) was noted in three patients (six eyes); four eyes (two patients) required glaucoma surgery. The overall safety and effectiveness of VN treatment in our cohort align with previous VN clinical trials, except for the higher occurrence of retinal atrophy and increased IOP in our cohort. This suggests that raised IOP and retinal atrophy may be more common than previously reported.
Collapse
Affiliation(s)
- Peter Kiraly
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK; (P.K.); (J.C.-K.); (I.H.Y.); (C.M.-F.d.l.C.); (S.M.D.); (R.E.M.)
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford OX3 9DU, UK;
| | - Charles L. Cottriall
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK; (P.K.); (J.C.-K.); (I.H.Y.); (C.M.-F.d.l.C.); (S.M.D.); (R.E.M.)
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford OX3 9DU, UK;
| | - Laura J. Taylor
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK; (P.K.); (J.C.-K.); (I.H.Y.); (C.M.-F.d.l.C.); (S.M.D.); (R.E.M.)
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford OX3 9DU, UK;
| | - Jasleen K. Jolly
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford OX3 9DU, UK;
- Vision and Eye Research Institute, Anglia Ruskin University, Cambridge CB1 1PT, UK
| | - Jasmina Cehajic-Kapetanovic
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK; (P.K.); (J.C.-K.); (I.H.Y.); (C.M.-F.d.l.C.); (S.M.D.); (R.E.M.)
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford OX3 9DU, UK;
| | - Imran H. Yusuf
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK; (P.K.); (J.C.-K.); (I.H.Y.); (C.M.-F.d.l.C.); (S.M.D.); (R.E.M.)
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford OX3 9DU, UK;
| | - Cristina Martinez-Fernandez de la Camara
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK; (P.K.); (J.C.-K.); (I.H.Y.); (C.M.-F.d.l.C.); (S.M.D.); (R.E.M.)
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford OX3 9DU, UK;
| | - Morag Shanks
- Oxford Regional Genetics Laboratories, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK;
| | - Susan M. Downes
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK; (P.K.); (J.C.-K.); (I.H.Y.); (C.M.-F.d.l.C.); (S.M.D.); (R.E.M.)
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford OX3 9DU, UK;
| | - Robert E. MacLaren
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK; (P.K.); (J.C.-K.); (I.H.Y.); (C.M.-F.d.l.C.); (S.M.D.); (R.E.M.)
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford OX3 9DU, UK;
| | - M. Dominik Fischer
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK; (P.K.); (J.C.-K.); (I.H.Y.); (C.M.-F.d.l.C.); (S.M.D.); (R.E.M.)
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford OX3 9DU, UK;
- Centre for Ophthalmology, University Hospital Tubingen, 72076 Tubingen, Germany
| |
Collapse
|
15
|
Kramer AC, Carthage J, Berry Y, Gurdziel K, Cook TA, Thummel R. A comparative analysis of gene and protein expression in chronic and acute models of photoreceptor degeneration in adult zebrafish. Front Cell Dev Biol 2023; 11:1233269. [PMID: 37745292 PMCID: PMC10512720 DOI: 10.3389/fcell.2023.1233269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
Background: Adult zebrafish are capable of photoreceptor (PR) regeneration following acute phototoxic lesion (AL). We developed a chronic low light (CLL) exposure model that more accurately reflects chronic PR degeneration observed in many human retinal diseases. Methods: Here, we characterize the morphological and transcriptomic changes associated with acute and chronic models of PR degeneration at 8 time-points over a 28-day window using immunohistochemistry and 3'mRNA-seq. Results: We first observed a differential sensitivity of rod and cone PRs to CLL. Next, we found no evidence for Müller glia (MG) gliosis or regenerative cell-cycle re-entry in the CLL model, which is in contrast to the robust gliosis and proliferative response from resident MG in the AL model. Differential responses of microglia between the models was also observed. Transcriptomic comparisons between the models revealed gene-specific networks of PR regeneration and degeneration, including genes that are activated under conditions of chronic PR stress. Finally, we showed that CLL is at least partially reversible, allowing for rod and cone outer segment outgrowth and replacement of rod cell nuclei via an apparent upregulation of the existing rod neurogenesis mechanism. Discussion: Collectively, these data provide a direct comparison of the morphological and transcriptomic PR degeneration and regeneration models in zebrafish.
Collapse
Affiliation(s)
- Ashley C. Kramer
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Justin Carthage
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Yasmeen Berry
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Katherine Gurdziel
- Genomic Sciences Core, Wayne State University, Detroit, MI, United States
| | - Tiffany A. Cook
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, United States
| | - Ryan Thummel
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
16
|
Jones MK, Orozco LD, Qin H, Truong T, Caplazi P, Elstrott J, Modrusan Z, Chaney SY, Jeanne M. Integration of human stem cell-derived in vitro systems and mouse preclinical models identifies complex pathophysiologic mechanisms in retinal dystrophy. Front Cell Dev Biol 2023; 11:1252547. [PMID: 37691820 PMCID: PMC10483287 DOI: 10.3389/fcell.2023.1252547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/14/2023] [Indexed: 09/12/2023] Open
Abstract
Rare DRAM2 coding variants cause retinal dystrophy with early macular involvement via unknown mechanisms. We found that DRAM2 is ubiquitously expressed in the human eye and expression changes were observed in eyes with more common maculopathy such as Age-related Macular Degeneration (AMD). To gain insights into pathogenicity of DRAM2-related retinopathy, we used a combination of in vitro and in vivo models. We found that DRAM2 loss in human pluripotent stem cell (hPSC)-derived retinal organoids caused the presence of additional mesenchymal cells. Interestingly, Dram2 loss in mice also caused increased proliferation of cells from the choroid in vitro and exacerbated choroidal neovascular lesions in vivo. Furthermore, we observed that DRAM2 loss in human retinal pigment epithelial (RPE) cells resulted in increased susceptibility to stress-induced cell death in vitro and that Dram2 loss in mice caused age-related photoreceptor degeneration. This highlights the complexity of DRAM2 function, as its loss in choroidal cells provided a proliferative advantage, whereas its loss in post-mitotic cells, such as photoreceptor and RPE cells, increased degeneration susceptibility. Different models such as human pluripotent stem cell-derived systems and mice can be leveraged to study and model human retinal dystrophies; however, cell type and species-specific expression must be taken into account when selecting relevant systems.
Collapse
Affiliation(s)
- Melissa K. Jones
- Department of Neuroscience, Genentech Inc., South San Francisco, CA, United States
- Product Development Clinical Science Ophthalmology, Genentech Inc., South San Francisco, CA, United States
| | - Luz D. Orozco
- Department of Bioinformatics, Genentech Inc., South San Francisco, CA, United States
| | - Han Qin
- Department of Neuroscience, Genentech Inc., South San Francisco, CA, United States
| | - Tom Truong
- Department of Translational Immunology, Genentech Inc., South San Francisco, CA, United States
| | - Patrick Caplazi
- Department of Research Pathology, Genentech Inc., South San Francisco, CA, United States
| | - Justin Elstrott
- Department of Translational Imaging, Genentech Inc., South San Francisco, CA, United States
| | - Zora Modrusan
- Department of Microchemistry, Proteomics, Lipidomics and Next-Generation Sequencing, Genentech Inc., South San Francisco, CA, United States
| | - Shawnta Y. Chaney
- Department of Translational Immunology, Genentech Inc., South San Francisco, CA, United States
| | - Marion Jeanne
- Department of Neuroscience, Genentech Inc., South San Francisco, CA, United States
| |
Collapse
|
17
|
Jaffal L, Joumaa H, Noureldine J, Banjak M, Ibrahim M, Mrad Z, Salami A, Shamieh SE. The genetic landscape of inherited retinal dystrophies in Arabs. BMC Med Genomics 2023; 16:89. [PMID: 37127645 PMCID: PMC10150479 DOI: 10.1186/s12920-023-01518-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 04/13/2023] [Indexed: 05/03/2023] Open
Abstract
Inherited retinal dystrophies (IRDs) are a major cause of vision loss. Altogether are highly heterogeneous genotypically and phenotypically, exhibiting substantial differences worldwide. To shed more light on these conditions, we investigated the genetic and phenotypic landscape of IRDs in the Arabs globally and per country.We analyzed 1,621 affected individuals from 16 Arabic countries reported in 198 articles. At the phenotypic level, rod-cone dystrophy (RCD) and Usher syndrome were the most prevalent conditions among non-syndromic and syndromic IRDs. At the gene level, TULP1, ABCA4, RP1, CRB1, MYO7A, RPE65, KCNV2, and IMPG2 were the most mutated genes. Interestingly, all except CRB1 were highly prevalent because they harbored founder mutations, implying that consanguinity is a major determinant in Arab countries. Of note, ~ 93% of the investigated individuals carried homozygous mutations. The country analysis for the IRDs conditions and their associated genotypes revealed that whereas Leber Congenital Amaurosis, RCD, and USHER syndrome were widely distributed, bestrophinopathies and non-syndromic hearing loss were restricted to specific countries (till now).This study could be a starting point for initiating suitable health policies towards IRDs in the Arab world. The high degree of homozygosity urges the need for genetic counsellors to provide personalized information and support the affected individuals.
Collapse
Affiliation(s)
- Lama Jaffal
- Department of Biological and Chemical Sciences, School of Arts and Sciences, Lebanese International University, Beirut, Lebanon
| | - Hawraa Joumaa
- Rammal Hassan Rammal Research Laboratory, PhyToxE Research Group, Faculty of Sciences, Lebanese University, Nabatieh, Lebanon
| | - Jinane Noureldine
- Rammal Hassan Rammal Research Laboratory, PhyToxE Research Group, Faculty of Sciences, Lebanese University, Nabatieh, Lebanon
| | - Malak Banjak
- Rammal Hassan Rammal Research Laboratory, PhyToxE Research Group, Faculty of Sciences, Lebanese University, Nabatieh, Lebanon
| | - Mariam Ibrahim
- Rammal Hassan Rammal Research Laboratory, PhyToxE Research Group, Faculty of Sciences, Lebanese University, Nabatieh, Lebanon
| | - Zamzam Mrad
- Rammal Hassan Rammal Research Laboratory, PhyToxE Research Group, Faculty of Sciences, Lebanese University, Nabatieh, Lebanon
| | - Ali Salami
- Department of Mathematics, Faculty of Sciences, Lebanese University, Nabatieh, Lebanon
| | - Said El Shamieh
- Molecular Testing Laboratory, Department of Medical Laboratory Technology, Faculty of Health Sciences, Beirut Arab University, Beirut, Lebanon.
| |
Collapse
|
18
|
Lee BJH, Tham YC, Tan TE, Bylstra Y, Lim WK, Jain K, Chan CM, Mathur R, Cheung CMG, Fenner BJ. Characterizing the genotypic spectrum of retinitis pigmentosa in East Asian populations: a systematic review. Ophthalmic Genet 2023; 44:109-118. [PMID: 36856324 DOI: 10.1080/13816810.2023.2182329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
BACKGROUND Ongoing trials for retinitis pigmentosa (RP) are genotype-specific, with most trials conducted on European cohorts. Due to genetic differences across diverse ancestries and populations, these therapies may not be efficacious in East Asians. MATERIALS AND METHODS A literature search was conducted from 1966 to September 2022 for cohort studies on East Asian populations reporting on non-syndromic RP genotypes and variants. Population-weighted prevalence was used to determine the genotypes and individual variants across the entire cohort. The carrier prevalence of common variants was compared against those in Europe. RESULTS A total of 12 articles describing 2,932 clinically diagnosed East Asian RP probands were included. We identified 876 variants across 54 genes. The most common genotypes included USH2A, EYS, RPGR, ABCA4, PRPF31, RHO, RP1, RP2, PDE6B and SNRNP200, with USH2A as the most common (17.1%). Overall, 60.5% of probands with clinically relevant variants were found to have one of the genotypes above, with 543/876 (62.0%) of the variants occurring in these genes. The most frequently reported variant was USH2A missense variant c.2802T>G/p.C934W (4.9%). Carrier prevalence of these variants was significantly different (p < 0.0001) than in Europe. CONCLUSIONS USH2A was the most commonly affected RP gene in this East Asian cohort, although sub-population analysis revealed distinct genotype prevalence patterns. While the genotypes are similar between East Asia and European cohorts, variants are specific to East Asia. The identification of several prevalent variants in USH2A and EYS provides an opportunity for the development of therapeutics that are relevant for East Asia patients.
Collapse
Affiliation(s)
- Brian Juin Hsien Lee
- Department of Medical Retina, Singapore National Eye Centre, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Yih-Chung Tham
- Retina Research Group Singapore Eye Research Institute, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (EYE ACP), Duke-NUS Medical School, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Centre for Innovation & Precision Eye Health, Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore
| | - Tien-En Tan
- Department of Medical Retina, Singapore National Eye Centre, Singapore
- Retina Research Group Singapore Eye Research Institute, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (EYE ACP), Duke-NUS Medical School, Singapore
| | - Yasmin Bylstra
- SingHealth Duke-NUS Institute of Precision Medicine, Singapore Health Services, Singapore
| | - Weng Khong Lim
- SingHealth Duke-NUS Institute of Precision Medicine, Singapore Health Services, Singapore
| | - Kanika Jain
- POLARIS, Genome Institute of Singapore, Singapore
| | - Choi Mun Chan
- Department of Medical Retina, Singapore National Eye Centre, Singapore
- Retina Research Group Singapore Eye Research Institute, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (EYE ACP), Duke-NUS Medical School, Singapore
| | - Ranjana Mathur
- Department of Medical Retina, Singapore National Eye Centre, Singapore
- Retina Research Group Singapore Eye Research Institute, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (EYE ACP), Duke-NUS Medical School, Singapore
| | - Chui Ming Gemmy Cheung
- Department of Medical Retina, Singapore National Eye Centre, Singapore
- Retina Research Group Singapore Eye Research Institute, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (EYE ACP), Duke-NUS Medical School, Singapore
| | - Beau J Fenner
- Department of Medical Retina, Singapore National Eye Centre, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Retina Research Group Singapore Eye Research Institute, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (EYE ACP), Duke-NUS Medical School, Singapore
| |
Collapse
|
19
|
An Overview towards Zebrafish Larvae as a Model for Ocular Diseases. Int J Mol Sci 2023; 24:ijms24065387. [PMID: 36982479 PMCID: PMC10048880 DOI: 10.3390/ijms24065387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 03/14/2023] Open
Abstract
Despite the obvious morphological differences in the visual system, zebrafish share a similar architecture and components of the same embryonic origin as humans. The zebrafish retina has the same layered structure and cell types with similar metabolic and phototransduction support as humans, and is functional 72 h after fertilization, allowing tests of visual function to be performed. The zebrafish genomic database supports genetic mapping studies as well as gene editing, both of which are useful in the ophthalmological field. It is possible to model ocular disorders in zebrafish, as well as inherited retinal diseases or congenital or acquired malformations. Several approaches allow the evaluation of local pathological processes derived from systemic disorders, such as chemical exposure to produce retinal hypoxia or glucose exposure to produce hyperglycemia, mimicking retinopathy of prematurity or diabetic retinopathy, respectively. The pathogenesis of ocular infections, autoimmune diseases, or aging can also be assessed in zebrafish larvae, and the preserved cellular and molecular immune mechanisms can be assessed. Finally, the zebrafish model for the study of the pathologies of the visual system complements certain deficiencies in experimental models of mammals since the regeneration of the zebrafish retina is a valuable tool for the study of degenerative processes and the discovery of new drugs and therapies.
Collapse
|
20
|
Systematic analysis of CNGA3 splice variants identifies different mechanisms of aberrant splicing. Sci Rep 2023; 13:2896. [PMID: 36801918 PMCID: PMC9938885 DOI: 10.1038/s41598-023-29452-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 02/06/2023] [Indexed: 02/20/2023] Open
Abstract
Achromatopsia is an autosomal recessive cone photoreceptor disease that is frequently caused by pathogenic variants in the CNGA3 gene. Here, we present a systematic functional analysis of 20 CNGA3 splice site variants detected in our large cohort of achromatopsia patients and/or listed in common variant databases. All variants were analyzed by functional splice assays based on the pSPL3 exon trapping vector. We demonstrated that ten variants, both at canonical and non-canonical splice sites, induced aberrant splicing, including intronic nucleotide retention, exonic nucleotide deletion and exon skipping, resulting in 21 different aberrant transcripts. Of these, eleven were predicted to introduce a premature termination codon. The pathogenicity of all variants was assessed based on established guidelines for variant classification. Incorporation of the results of our functional analyses enabled re-classification of 75% of variants previously classified as variants of uncertain significance into either likely benign or likely pathogenic. Our study is the first in which a systematic characterization of putative CNGA3 splice variants has been performed. We demonstrated the utility of pSPL3 based minigene assays in the effective assessment of putative splice variants. Our findings improve the diagnosis of achromatopsia patients, who may thus benefit from future gene-based therapeutic strategies.
Collapse
|
21
|
Wu KY, Kulbay M, Toameh D, Xu AQ, Kalevar A, Tran SD. Retinitis Pigmentosa: Novel Therapeutic Targets and Drug Development. Pharmaceutics 2023; 15:685. [PMID: 36840007 PMCID: PMC9963330 DOI: 10.3390/pharmaceutics15020685] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/12/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023] Open
Abstract
Retinitis pigmentosa (RP) is a heterogeneous group of hereditary diseases characterized by progressive degeneration of retinal photoreceptors leading to progressive visual decline. It is the most common type of inherited retinal dystrophy and has a high burden on both patients and society. This condition causes gradual loss of vision, with its typical manifestations including nyctalopia, concentric visual field loss, and ultimately bilateral central vision loss. It is one of the leading causes of visual disability and blindness in people under 60 years old and affects over 1.5 million people worldwide. There is currently no curative treatment for people with RP, and only a small group of patients with confirmed RPE65 mutations are eligible to receive the only gene therapy on the market: voretigene neparvovec. The current therapeutic armamentarium is limited to retinoids, vitamin A supplements, protection from sunlight, visual aids, and medical and surgical interventions to treat ophthalmic comorbidities, which only aim to slow down the progression of the disease. Considering such a limited therapeutic landscape, there is an urgent need for developing new and individualized therapeutic modalities targeting retinal degeneration. Although the heterogeneity of gene mutations involved in RP makes its target treatment development difficult, recent fundamental studies showed promising progress in elucidation of the photoreceptor degeneration mechanism. The discovery of novel molecule therapeutics that can selectively target specific receptors or specific pathways will serve as a solid foundation for advanced drug development. This article is a review of recent progress in novel treatment of RP focusing on preclinical stage fundamental research on molecular targets, which will serve as a starting point for advanced drug development. We will review the alterations in the molecular pathways involved in the development of RP, mainly those regarding endoplasmic reticulum (ER) stress and apoptotic pathways, maintenance of the redox balance, and genomic stability. We will then discuss the therapeutic approaches under development, such as gene and cell therapy, as well as the recent literature identifying novel potential drug targets for RP.
Collapse
Affiliation(s)
- Kevin Y. Wu
- Division of Ophthalmology, Department of Surgery, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada
| | - Merve Kulbay
- Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Dana Toameh
- Faculty of Medicine, McGill University, Montreal, QC H3G 2M1, Canada
| | - An Qi Xu
- Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Ananda Kalevar
- Division of Ophthalmology, Department of Surgery, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada
| | - Simon D. Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| |
Collapse
|
22
|
Gopinath C, Rompicherla R, Mathias GP, Patil R, Poornachandra B, Vinekar A, Mochi TB, Braganza S, Shetty KB, Kumaramanickavel G, Ghosh A. Inherited retinal disorders: a genotype-phenotype correlation in an Indian cohort and the importance of genetic testing and genetic counselling. Graefes Arch Clin Exp Ophthalmol 2023:10.1007/s00417-022-05955-5. [PMID: 36648511 DOI: 10.1007/s00417-022-05955-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 10/10/2022] [Accepted: 12/21/2022] [Indexed: 01/18/2023] Open
Abstract
PURPOSE Recent advances in sequencing technologies have enabled radical and rapid progress in the genetic diagnosis of inherited retinal disorders (IRDs). Although the list of gene variations continues to grow, it lacks the genetic etiology of ethnic groups like South Asians. Differences in racial backgrounds and consanguinity add to genetic heterogeneity and phenotypic overlaps. METHODS This retrospective study includes documented data from the Gen-Eye clinic from years 2014 to 2019. Medical records and pedigrees of 591 IRD patients of Indian origin and genetic reports of 117 probands were reviewed. Genotype-phenotype correlations were performed to classify as correlating, non-correlating and unsolved cases. RESULTS Among the 591 patients, we observed a higher prevalence of clinically diagnosed retinitis pigmentosa (38.9%) followed by unspecified diagnoses (28.5%). Consanguinity was reported to be high (55.6%) in this cohort. Among the variants identified in 117 probands, 36.4% of variants were pathogenic, 19.2% were likely pathogenic, and 44.4% were of uncertain significance. Among the pathogenic and likely pathogenic variants, autosomal recessive inheritance showed higher prevalence. About 35% (41/117) of cases showed genotype-phenotype correlation. Within the correlating cases, retinitis pigmentosa and Stargardt disease were predominant. Novel variants identified in RP, Stargardt, and LCA are reported here. CONCLUSION This first-of-a-kind report on an Indian cohort contributes to existing knowledge and expansion of variant databases, presenting relevant and plausible novel variants. Phenotypic overlap and variability lead to a differential diagnosis and hence a clear genotype-phenotype correlation helps in precise clinical confirmation. The study also emphasizes the importance of genetic counselling and testing for personalized vision care in a tertiary eye hospital.
Collapse
Affiliation(s)
- Chitra Gopinath
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, 560099, India
- Gen-Eye Clinic, Narayana Nethralaya, Bangalore, 560099, India
| | - Ramya Rompicherla
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, 560099, India
- Gen-Eye Clinic, Narayana Nethralaya, Bangalore, 560099, India
| | - Grace Priyaranjini Mathias
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, 560099, India
- Gen-Eye Clinic, Narayana Nethralaya, Bangalore, 560099, India
| | - Rajeshwari Patil
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, 560099, India
- Gen-Eye Clinic, Narayana Nethralaya, Bangalore, 560099, India
| | - B Poornachandra
- Vitreo-Retina Services, Narayana Nethralaya, Bangalore, 560010, India
| | - Anand Vinekar
- Vitreo-Retina Services, Narayana Nethralaya, Bangalore, 560010, India
| | | | - Sherine Braganza
- Vitreo-Retina Services, Narayana Nethralaya, Bangalore, 560010, India
| | - K Bhujang Shetty
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, 560099, India
- Gen-Eye Clinic, Narayana Nethralaya, Bangalore, 560099, India
- Vitreo-Retina Services, Narayana Nethralaya, Bangalore, 560010, India
| | - Govindasamy Kumaramanickavel
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, 560099, India
- Gen-Eye Clinic, Narayana Nethralaya, Bangalore, 560099, India
| | - Anuprita Ghosh
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, 560099, India.
- Gen-Eye Clinic, Narayana Nethralaya, Bangalore, 560099, India.
| |
Collapse
|
23
|
Hitti-Malin RJ, Dhaenens CM, Panneman DM, Corradi Z, Khan M, den Hollander AI, Farrar GJ, Gilissen C, Hoischen A, van de Vorst M, Bults F, Boonen EGM, Saunders P, Roosing S, Cremers FPM. Using single molecule Molecular Inversion Probes as a cost-effective, high-throughput sequencing approach to target all genes and loci associated with macular diseases. Hum Mutat 2022; 43:2234-2250. [PMID: 36259723 PMCID: PMC10092144 DOI: 10.1002/humu.24489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 09/06/2022] [Accepted: 10/17/2022] [Indexed: 01/25/2023]
Abstract
Macular degenerations (MDs) are a subgroup of retinal disorders characterized by central vision loss. Knowledge is still lacking on the extent of genetic and nongenetic factors influencing inherited MD (iMD) and age-related MD (AMD) expression. Single molecule Molecular Inversion Probes (smMIPs) have proven effective in sequencing the ABCA4 gene in patients with Stargardt disease to identify associated coding and noncoding variation, however many MD patients still remain genetically unexplained. We hypothesized that the missing heritability of MDs may be revealed by smMIPs-based sequencing of all MD-associated genes and risk factors. Using 17,394 smMIPs, we sequenced the coding regions of 105 iMD and AMD-associated genes and noncoding or regulatory loci, known pseudo-exons, and the mitochondrial genome in two test cohorts that were previously screened for variants in ABCA4. Following detailed sequencing analysis of 110 probands, a diagnostic yield of 38% was observed. This established an ''MD-smMIPs panel," enabling a genotype-first approach in a high-throughput and cost-effective manner, whilst achieving uniform and high coverage across targets. Further analysis will identify known and novel variants in MD-associated genes to offer an accurate clinical diagnosis to patients. Furthermore, this will reveal new genetic associations for MD and potential genetic overlaps between iMD and AMD.
Collapse
Affiliation(s)
- Rebekkah J Hitti-Malin
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Claire-Marie Dhaenens
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience & Cognition, Univ. Lille, Lille, France
| | - Daan M Panneman
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Zelia Corradi
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mubeen Khan
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anneke I den Hollander
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - G Jane Farrar
- The School of Genetics & Microbiology, The University of Dublin Trinity College, Dublin, Ireland
| | - Christian Gilissen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.,Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Alexander Hoischen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.,Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Internal Medicine, Radboud University Medical Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maartje van de Vorst
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Femke Bults
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Erica G M Boonen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | | - Susanne Roosing
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frans P M Cremers
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
24
|
Chen Y, Dong Y, Yan J, Wang L, Yu S, Jiao K, Paquet-Durand F. Single-Cell Transcriptomic Profiling in Inherited Retinal Degeneration Reveals Distinct Metabolic Pathways in Rod and Cone Photoreceptors. Int J Mol Sci 2022; 23:12170. [PMID: 36293024 PMCID: PMC9603353 DOI: 10.3390/ijms232012170] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/28/2022] [Accepted: 10/08/2022] [Indexed: 08/31/2023] Open
Abstract
The cellular mechanisms underlying hereditary photoreceptor degeneration are still poorly understood. The aim of this study was to systematically map the transcriptional changes that occur in the degenerating mouse retina at the single cell level. To this end, we employed single-cell RNA-sequencing (scRNA-seq) and retinal degeneration-1 (rd1) mice to profile the impact of the disease mutation on the diverse retinal cell types during early post-natal development. The transcriptome data allowed to annotate 43,979 individual cells grouped into 20 distinct clusters. We further characterized cluster-specific metabolic and biological changes in individual cell types. Our results highlight Ca2+-signaling as relevant to hereditary photoreceptor degeneration. Although metabolic reprogramming in retina, known as the 'Warburg effect', has been documented, further metabolic changes were noticed in rd1 mice. Such metabolic changes in rd1 mutation was likely regulated through mitogen-activated protein kinase (MAPK) pathway. By combining single-cell transcriptomes and immunofluorescence staining, our study revealed cell type-specific changes in gene expression, as well as interplay between Ca2+-induced cell death and metabolic pathways.
Collapse
Affiliation(s)
- Yiyi Chen
- Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany
- Graduate Training Centre of Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| | - Yujie Dong
- Yunnan Eye Institute & Key Laboratory of Yunnan Province, 650021 Kunming, China
| | - Jie Yan
- Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany
- Graduate Training Centre of Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| | - Lan Wang
- Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany
- Graduate Training Centre of Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| | - Shirley Yu
- Graduate Training Centre of Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| | - Kangwei Jiao
- Yunnan Eye Institute & Key Laboratory of Yunnan Province, 650021 Kunming, China
| | | |
Collapse
|
25
|
Krauss E, Macher J, Capasso J, Bernhardt B, Ali-KhanCatts Z, Levin A, Brandt R. Experiences of genetic testing among individuals with retinitis pigmentosa. Ophthalmic Genet 2022; 43:633-640. [PMID: 35796432 DOI: 10.1080/13816810.2022.2096243] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
BACKGROUND Retinitis pigmentosa (RP) is a genetically heterogeneous retinal dystrophy which results in progressive vision loss. There is scant literature on the experiences of genetic testing in patients with RP. MATERIALS AND METHODS Patients with a clinical diagnosis of RP who received genetic testing at the Wills Eye Ocular Genetics clinic between 2016 and 2020 were recruited. Telephone interviews were conducted using a semi-structured guide designed to elicit participant experiences with genetic testing. A thematic analysis was performed to describe patterns in participant responses. RESULTS Twelve patients participated. Seven participants identified as female and five as male, with ages ranging from 22 to 70. Ten patients had positive genetic test results, while two had negative genetic testing. Reported motivations for genetic testing included qualification for clinical trials (58% of total participants), determination of etiology or usal gene (50%), reproductive concerns (50%), and prognostic outlook (50%). Most participants (75%) expressed satisfaction about their decision to pursue genetic testing. Participants with both positive and negative genetic testing reported persistent uncertainty regarding their prognosis for visual decline (50%). Genetic confirmation of disease leads to initiation of safety and vision-protecting health behaviors (42%). CONCLUSION Patients with RP are generally satisfied with their testing experience, despite approaching testing with a wide range of motivations and expectations. Future research can leverage this methodology to identify targets for improvement in pre- and post-test education and counselling.
Collapse
Affiliation(s)
- Emily Krauss
- Duke Eye Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Jared Macher
- University of Rochester School of Medicine, Rochester, New York, USA
| | - Jenina Capasso
- Pediatric Ophthalmology and Ocular Genetics, Flaum Eye Institute, Rochester, New York, USA.,Pediatric Genetics, Golisano Children's Hospital, Rochester, New York, USA
| | - Barbara Bernhardt
- Human Genetics and Genetic Counseling, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Zohra Ali-KhanCatts
- Human Genetics and Genetic Counseling, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Alex Levin
- Pediatric Ophthalmology and Ocular Genetics, Flaum Eye Institute, Rochester, New York, USA.,Pediatric Genetics, Golisano Children's Hospital, Rochester, New York, USA
| | - Rachael Brandt
- Human Genetics and Genetic Counseling, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Lankenau Medical Center, Main Line Health Hospitals, Wynnewood, Pennsylvania, USA.,Lankenau Institute for Medical Research, Wynnewood, Pennsylvania, USA
| |
Collapse
|