1
|
Zhang Q, Sun J, Liu Z, Wang H, Zhou H, Liu W, Jia H, Li N, Li T, Wang F, Sun X. Clinical and Molecular Characterization of AIPL1-Associated Leber Congenital Amaurosis/Early-Onset Severe Retinal Dystrophy. Am J Ophthalmol 2024; 266:235-247. [PMID: 38880373 DOI: 10.1016/j.ajo.2024.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 06/18/2024]
Abstract
PURPOSE This study aimed to characterize the clinical features, genetic findings, and genotype-phenotype correlations of patients with Leber congenital amaurosis (LCA) or early-onset severe retinal dystrophy (EOSRD) harboring biallelic AIPL1 pathogenic variants. DESIGN Retrospective case series. METHODS This study consecutively enrolled 51 patients from 47 families with a clinical diagnosis of LCA/EOSRD harboring disease-causing variants in the AIPL1 gene, from October 2021 to September 2023. Molecular genetic findings, medical history, and ophthalmic evaluation including visual acuity (VA), multimodal retinal imaging, and electrophysiologic assessment were reviewed. RESULTS Of the 51 patients (32 with LCA and 19 with EOSRD), 27 (53%) were females, and age at last review ranged from 0.5 to 58.4 years. We identified 28 disease-causing AIPL1 variants, with 18 being novel. In patients with EOSRD, the mean (range) VA was 1.3 (0.7-2.7) logMAR and 1.3 (0.5-2.3) logMAR for right and left eyes respectively, with an average annual decline of 0.03 logMAR (R2 = 0.7547, P < .01). For patients with LCA, the VA ranged from light perception to counting fingers. Optical coherence tomography imaging demonstrated preservation of foveal ellipsoid zone in the 5 youngest EOSRD patients and 9 LCA children. Electroretinography showed severe cone-rod patterns in 78.6% (11/14) of patients with EOSRD, while classical extinguished pattern was documented in all patients with LCA available for the examination. The most common mutation was the nonsense variants of c.421C>T, with an allele frequency of 53.9%. All patients with EOSRD carried at least one missense mutation, of whom 13 identified with c.152A>G and 5 with c.572T>C. Twenty-six patients with LCA harbored two null AIPL1 variants, while 18 were homozygous for c.421C>T and 6 were heterozygous for c.421C>T with another loss-of-function variant. CONCLUSIONS This study reveals distinct clinical features and variation spectrum between AIPL1-associated LCA and EOSRD. Patients harboring at least one nonnull mutation, especially c.152A>G and c.572T>C, were significantly more likely to have a milder EOSRD phenotype than those with two null mutations. Residual foveal outer retinal structure observed in the youngest proportion of patients suggests an early window for gene augmentation therapy.
Collapse
Affiliation(s)
- Quan Zhang
- From the Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (QZ, JS, ZL, HW, HZ, WL, HJ, NL, TL, and XS), Shanghai, China; National Clinical Research Center for Ophthalmic Diseases (QZ, JS, ZL, HW, WL, HJ, NL, TL, and XS), Shanghai, China
| | - Junran Sun
- From the Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (QZ, JS, ZL, HW, HZ, WL, HJ, NL, TL, and XS), Shanghai, China; National Clinical Research Center for Ophthalmic Diseases (QZ, JS, ZL, HW, WL, HJ, NL, TL, and XS), Shanghai, China
| | - Zishi Liu
- From the Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (QZ, JS, ZL, HW, HZ, WL, HJ, NL, TL, and XS), Shanghai, China; National Clinical Research Center for Ophthalmic Diseases (QZ, JS, ZL, HW, WL, HJ, NL, TL, and XS), Shanghai, China
| | - Hong Wang
- From the Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (QZ, JS, ZL, HW, HZ, WL, HJ, NL, TL, and XS), Shanghai, China; National Clinical Research Center for Ophthalmic Diseases (QZ, JS, ZL, HW, WL, HJ, NL, TL, and XS), Shanghai, China
| | - Hao Zhou
- From the Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (QZ, JS, ZL, HW, HZ, WL, HJ, NL, TL, and XS), Shanghai, China
| | - Wenjia Liu
- From the Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (QZ, JS, ZL, HW, HZ, WL, HJ, NL, TL, and XS), Shanghai, China; National Clinical Research Center for Ophthalmic Diseases (QZ, JS, ZL, HW, WL, HJ, NL, TL, and XS), Shanghai, China
| | - Huixun Jia
- From the Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (QZ, JS, ZL, HW, HZ, WL, HJ, NL, TL, and XS), Shanghai, China; National Clinical Research Center for Ophthalmic Diseases (QZ, JS, ZL, HW, WL, HJ, NL, TL, and XS), Shanghai, China; Shanghai Engineering Center for Visual Science and Photomedicine (HJ, FW, and XS), Shanghai, China
| | - Ningdong Li
- From the Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (QZ, JS, ZL, HW, HZ, WL, HJ, NL, TL, and XS), Shanghai, China; National Clinical Research Center for Ophthalmic Diseases (QZ, JS, ZL, HW, WL, HJ, NL, TL, and XS), Shanghai, China
| | - Tong Li
- From the Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (QZ, JS, ZL, HW, HZ, WL, HJ, NL, TL, and XS), Shanghai, China; National Clinical Research Center for Ophthalmic Diseases (QZ, JS, ZL, HW, WL, HJ, NL, TL, and XS), Shanghai, China.
| | - Fenghua Wang
- Shanghai Engineering Center for Visual Science and Photomedicine (HJ, FW, and XS), Shanghai, China; Innostellar Biotherapeutics Co., Ltd (FW), Shanghai, China
| | - Xiaodong Sun
- From the Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (QZ, JS, ZL, HW, HZ, WL, HJ, NL, TL, and XS), Shanghai, China; National Clinical Research Center for Ophthalmic Diseases (QZ, JS, ZL, HW, WL, HJ, NL, TL, and XS), Shanghai, China; Shanghai Engineering Center for Visual Science and Photomedicine (HJ, FW, and XS), Shanghai, China; Shanghai Key Laboratory of Fundus Diseases (XS), Shanghai, China.
| |
Collapse
|
2
|
Danish E, Alhashem A, Naaman N, Almhmoudi F, Mushiba A, Almatrafi A, Raggam A, Bukhari R. Mutations in the ciliary transport gene IFT140 cause syndromic congenital retinal dystrophy. J AAPOS 2024; 28:104007. [PMID: 39304031 DOI: 10.1016/j.jaapos.2024.104007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 09/22/2024]
Abstract
Early-onset, severe retinal dystrophy can be isolated or syndromic, presenting as part of an underlying systemic disease. Mainzer-Saldino syndrome, a rare systemic ciliopathy characterized by skeletal and renal disease, is caused by recessive mutations in the intraflagellar transport 140 chlamydomonas homologue (IFT140) gene. We present a series of 13 cases of early-onset retinal dysfunction with confirmed IFT140 mutations from 8 unrelated Saudi families belonging to 3 well-known tribes. All carried the same homozygous missense IFT140 mutation (c.1990G>A; p.Glu664Lys) except for a single family, which included 4 affected subjects, 3 of whom were aborted fetuses, with compound heterozygous pathogenic IFT140 variants (c.1525-1G>A and c.1990G>A; p.Glu664Lys). Severe retinal dystrophy was present in all living subjects, phenotypically apparent as hyperopia, nystagmus, nyctalopia, poor vision and nonrecordable full-field electroretinography. All affected individuals had skeletal abnormalities, and neurological abnormalities were common, but there was no evidence of chronic renal failure.
Collapse
Affiliation(s)
- Enam Danish
- Department of Ophthalmology, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
| | - Amal Alhashem
- Department of Pediatric, Division of Genetic and Metabolic Medicine, Prince Sultan Military Medical City, Riyadh, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Nada Naaman
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia; King Abdullah International Medical Research Center, Jeddah, Saudi Arabia; Department of Ophthalmology, Ministry of National Guard - Health Affairs, Jeddah, Saudi Arabia.
| | - Faeeqah Almhmoudi
- Department of Ophthalmology, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
| | - Aziza Mushiba
- Section of Medical Genetics, Children's Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | | | - Abdulaziz Raggam
- Department of Medicine, King Abdulaziz Medical City, Jeddah, Saudi Arabia
| | - Raghad Bukhari
- College of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
3
|
Parameswarappa DC, Bagga DK, Upadhyaya A, Balasubramanian J, Pochaboina V, Muthineni V, Jalali S, Kannabiran C. RPE65 mutations in Leber congenital amaurosis, early-onset severe retinal dystrophy, and retinitis pigmentosa from a tertiary eye care center in India. Ophthalmic Genet 2024; 45:303-312. [PMID: 38323530 DOI: 10.1080/13816810.2024.2309559] [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: 06/22/2023] [Revised: 11/10/2023] [Accepted: 01/19/2024] [Indexed: 02/08/2024]
Abstract
INTRODUCTION Mutations in the retinal pigment epithelial 65 kilodalton protein (RPE65) gene are associated with various inherited retinal diseases (IRDs), including Leber congenital amaurosis (LCA), early-onset severe retinal dystrophy (EOSRD), and retinitis pigmentosa (RP). We screened for mutations in RPE65 in a series of Indian patients with these IRDs to determine the frequency/types of mutations and to describe the associated phenotypes. MATERIALS AND METHODS Diagnosis of LCA, EOSRD, and RP was made by standard and pre-defined criteria. Patients were evaluated by clinical, retinal imaging, and electrophysiological parameters. Genomic DNA from patients and available family members were used for identifying mutations by direct Sanger sequencing of the RPE65 gene or targeted NGS gene panel for IRDs covering 260+ genes. Variations detected were tested in healthy control populations and for co-segregation with the disease in available family members. RESULTS Mutations were found in eight patients, out of 220 total cases screened, all homozygous for the respective mutant alleles. Seven patients had mutations leading to premature termination codons and one patient had a missense change. The onset of visual loss ranged from birth to <2 years of life. At presentation, RPE mottling in the background retina was present in all cases with macular involvement in five cases with or without vascular attenuation and optic disc pallor. CONCLUSION RPE65 mutations in this series were found in 3.6% of cases associated with severe, early-onset disease, with consistent RPE mottling and variable manifestations with regard to the extent of disc pallor, arteriolar attenuation, and appearance of the macula.
Collapse
Affiliation(s)
- Deepika C Parameswarappa
- Srimati Kanuri Santhamma Center for Vitreoretinal Diseases, Anant Bajaj Retina Institute, Kallam Anji Reddy Campus, L V Prasad Eye Institute, Hyderabad, India
| | - Deepak Kumar Bagga
- Meera and L B Deshpande Centre for Sight Enhancement, Institute for Vision Rehabilitation, L V Prasad Eye Institute, Hyderabad, India
| | - Abhishek Upadhyaya
- Srimati Kanuri Santhamma Center for Vitreoretinal Diseases, Anant Bajaj Retina Institute, Kallam Anji Reddy Campus, L V Prasad Eye Institute, Hyderabad, India
| | | | - Venkatesh Pochaboina
- Department of Clinical Genetics and Genetic Counseling, L V Prasad Eye Institute, Hyderabad, India
| | - Vani Muthineni
- Kallam Anji Reddy Molecular Genetics Laboratory, L V Prasad Eye Institute, Hyderabad, India
| | - Subhadra Jalali
- Srimati Kanuri Santhamma Center for Vitreoretinal Diseases, Anant Bajaj Retina Institute, Kallam Anji Reddy Campus, L V Prasad Eye Institute, Hyderabad, India
| | - Chitra Kannabiran
- Kallam Anji Reddy Molecular Genetics Laboratory, L V Prasad Eye Institute, Hyderabad, India
| |
Collapse
|
4
|
Daich Varela M, Dixit M, Kalitzeos A, Michaelides M. Adaptive Optics Retinal Imaging in RDH12-Associated Early Onset Severe Retinal Dystrophy. Invest Ophthalmol Vis Sci 2024; 65:9. [PMID: 38466282 PMCID: PMC10929749 DOI: 10.1167/iovs.65.3.9] [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: 10/11/2023] [Accepted: 12/03/2023] [Indexed: 03/12/2024] Open
Abstract
Purpose RDH12 is among the most common genes found in individuals with early-onset severe retinal (EOSRD). Adaptive optics scanning light ophthalmoscopy (AOSLO) enables resolution of individual rod and cone photoreceptors in the retina. This study presents the first AOSLO imaging of individuals with RDH12-associated EOSRD. Methods Case series of patients who attended Moorfields Eye Hospital (London, UK). Spectral-domain optical coherence tomography, near-infrared reflectance (NIR), and blue autofluorescence imaging were analyzed. En face image sequences of photoreceptors were recorded using either of two AOSLO modalities. Cross-sectional analysis was undertaken for seven patients and longitudinal analysis for one patient. Results Nine eyes from eight patients are presented in this case series. The mean age at the time of the assessment was 11.2 ± 6.5 years of age (range 7-29). A subfoveal continuous ellipsoid zone (EZ) line was present in eight eyes. Posterior pole AOSLO revealed patches of cone mosaics. Average cone densities at regions of interest 0.5° to the fovea ranged from 12,620 to 23,660 cells/mm2, whereas intercell spacing ranged from 7.0 to 9.7 µm. Conclusions This study demonstrates that AOSLO can provide useful high-quality images in patients with EOSRD, even during childhood, with nystagmus, and early macular atrophy. Cones at the posterior pole can appear as scattered islands or, possibly later in life, as a single subfoveal conglomerate. Detailed image analysis suggests that retinal pigment epithelial stress and dysfunction may be the initial step toward degeneration, with NIR being a useful tool to assess retinal well-being in RDH12-associated EOSRD.
Collapse
Affiliation(s)
- Malena Daich Varela
- Moorfields Eye Hospital, London, United Kingdom
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Mira Dixit
- Moorfields Eye Hospital, London, United Kingdom
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Angelos Kalitzeos
- Moorfields Eye Hospital, London, United Kingdom
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Michel Michaelides
- Moorfields Eye Hospital, London, United Kingdom
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| |
Collapse
|
5
|
Hristodorov D, Lohoff T, Luneborg N, Mulder GJ, Clark SJ. Investing in vision: Innovation in retinal therapeutics and the influence on venture capital investment. Prog Retin Eye Res 2024; 99:101243. [PMID: 38218527 DOI: 10.1016/j.preteyeres.2024.101243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/06/2024] [Accepted: 01/08/2024] [Indexed: 01/15/2024]
Abstract
Since the groundbreaking approval of the first anti-VEGF therapy in 2004, the retinal therapeutics field has undergone a remarkable transformation, witnessing a surge in novel, disease-modifying therapeutics for a broad spectrum of retinal diseases, extending beyond exudative VEGF-driven conditions. The surge in scientific advancement and the pressing, unmet, medical need have captured the attention of venture capital investors, who have collectively invested close to $10 billion in research and development of new retinal therapeutics between 2004 and 2023. Notably, the field of exudative diseases has gradually shifted away from trying to outcompete anti-VEGF therapeutics towards lowering the overall treatment burden by reducing injection frequency. Simultaneously, a new era has emerged in the non-exudative field, targeting prevalent conditions like dry AMD and rare indications such as Retinitis pigmentosa. This has led to promising drug candidates in development, culminating in the landmark approval of Luxturna for a rare form of Retinitis pigmentosa. The validation of new mechanisms, such as the complement pathway in dry AMD has paved the way for the approvals of Syvovre (Apellis) and Izervay (Iveric/Astellas), marking the first two therapies for this condition. In this comprehensive review, we share our view on the cumulative lessons from the past two decades in developing retinal therapeutics, covering both positive achievements and challenges. We also contextualize the investments, strategic partnering deals, and acquisitions of biotech companies, pharmaceutical companies venture capital investors in retinal therapeutics, respectively. Finally, we provide an outlook and potentially a forward-looking roadmap on novel retinal therapeutics, highlighting the emergence of potential new intervention strategies, such as cell-based therapies, gene editing, and combination therapies. We conclude that upcoming developments have the potential to further stimulate venture capital investments, which ultimately could facilitate the development and delivery of new therapies to patients in need.
Collapse
Affiliation(s)
| | | | | | | | - Simon J Clark
- Institute for Ophthalmic Research, Department for Ophthalmology, University Medical Center, Eberhard Karls University of Tübingen, Tübingen, Germany; University Eye Clinic, University Hospital Tübingen, Tübingen, Germany; Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, UK
| |
Collapse
|
6
|
Sather R, Ihinger J, Simmons M, Lobo GP, Montezuma SR. The Clinical Findings, Pathogenic Variants, and Gene Therapy Qualifications Found in a Leber Congenital Amaurosis Phenotypic Spectrum Patient Cohort. Int J Mol Sci 2024; 25:1253. [PMID: 38279252 PMCID: PMC10816538 DOI: 10.3390/ijms25021253] [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: 12/15/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
This retrospective study examines the clinical characteristics and underlying genetic variants that exist in a Leber congenital amaurosis (LCA) patient cohort evaluated at the inherited retinal disease (IRD) clinic at the University of Minnesota (UMN)/M Health System. Our LCA cohort consisted of 33 non-syndromic patients and one patient with Joubert syndrome. We report their relevant history, clinical findings, and genetic testing results. We monitored disease presentation utilizing ocular coherence tomography (OCT) and fundus autofluorescence (FAF). Electroretinogram testing (ERG) was performed in patients when clinically indicated. Next-generation sequencing (NGS) and genetic counseling was offered to all evaluated patients. Advanced photoreceptor loss was noted in 85.7% of the subjects. All patients who underwent FAF had findings of either a ring of macular hypo/hyper AF or peripheral hypo-AF. All patients had abnormal ERG findings. A diagnostic genetic test result was identified in 74.2% of the patients via NGS single-gene testing or panel testing. Two patients in our cohort qualified for Luxturna® and both received treatment at the time of this study. These data will help IRD specialists to understand the genetic variants and clinical presentations that characterize our patient population in the Midwest region of the United States.
Collapse
Affiliation(s)
| | | | | | | | - Sandra R. Montezuma
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (R.S.III); (J.I.); (G.P.L.)
| |
Collapse
|
7
|
Hakim A, Guido B, Narsineni L, Chen DW, Foldvari M. Gene therapy strategies for glaucoma from IOP reduction to retinal neuroprotection: progress towards non-viral systems. Adv Drug Deliv Rev 2023; 196:114781. [PMID: 36940751 DOI: 10.1016/j.addr.2023.114781] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/25/2023] [Accepted: 03/15/2023] [Indexed: 03/23/2023]
Abstract
Glaucoma is the result of the gradual death of retinal ganglion cells (RGCs) whose axons form the optic nerve. Elevated intraocular pressure (IOP) is a major risk factors thatcontributes to RGC apoptosis and axonal loss at the lamina cribrosa, resulting in progressive reduction and eventual anterograde-retrograde transport blockade of neurotrophic factors. Current glaucoma management mainly focuses on pharmacological or surgical lowering of IOP, to manage the only modifiable risk factor. Although IOP reduction delays disease progression, it does not address previous and ongoing optic nerve degeneration. Gene therapy is a promising direction to control or modify genes involved in the pathophysiology of glaucoma. Both viral and non-viral gene therapy delivery systems are emerging as promising alternatives or add-on therapies to traditional treatments for improving IOP control and provide neuroprotection. The specific spotlight on non-viral gene delivery systems shows further progress towards improving the safety of gene therapy and implementing neuroprotection by targeting specific tissues and cells in the eye and specifically in the retina.
Collapse
Affiliation(s)
- Antoine Hakim
- School of Pharmacy, University of Waterloo, 200 University Avenue West, Waterloo, ON, Canada N2L 3G1
| | - Benjamin Guido
- School of Pharmacy, University of Waterloo, 200 University Avenue West, Waterloo, ON, Canada N2L 3G1
| | - Lokesh Narsineni
- School of Pharmacy, University of Waterloo, 200 University Avenue West, Waterloo, ON, Canada N2L 3G1
| | - Ding-Wen Chen
- School of Pharmacy, University of Waterloo, 200 University Avenue West, Waterloo, ON, Canada N2L 3G1
| | - Marianna Foldvari
- School of Pharmacy, University of Waterloo, 200 University Avenue West, Waterloo, ON, Canada N2L 3G1; Waterloo Institute of Nanotechnology and Center for Bioengineering and Biotechnology University of Waterloo, 200 University Avenue West, Waterloo, ON, Canada N2L 3G1.
| |
Collapse
|
8
|
Duda T, Sharma RK. Multilimbed membrane guanylate cyclase signaling system, evolutionary ladder. Front Mol Neurosci 2023; 15:1022771. [PMID: 36683846 PMCID: PMC9849996 DOI: 10.3389/fnmol.2022.1022771] [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: 08/18/2022] [Accepted: 11/14/2022] [Indexed: 01/07/2023] Open
Abstract
One monumental discovery in the field of cell biology is the establishment of the membrane guanylate cyclase signal transduction system. Decoding its fundamental, molecular, biochemical, and genetic features revolutionized the processes of developing therapies for diseases of endocrinology, cardio-vasculature, and sensory neurons; lastly, it has started to leave its imprints with the atmospheric carbon dioxide. The membrane guanylate cyclase does so via its multi-limbed structure. The inter-netted limbs throughout the central, sympathetic, and parasympathetic systems perform these functions. They generate their common second messenger, cyclic GMP to affect the physiology. This review describes an historical account of their sequential evolutionary development, their structural components and their mechanisms of interaction. The foundational principles were laid down by the discovery of its first limb, the ACTH modulated signaling pathway (the companion monograph). It challenged two general existing dogmas at the time. First, there was the question of the existence of a membrane guanylate cyclase independent from a soluble form that was heme-regulated. Second, the sole known cyclic AMP three-component-transduction system was modulated by GTP-binding proteins, so there was the question of whether a one-component transduction system could exclusively modulate cyclic GMP in response to the polypeptide hormone, ACTH. The present review moves past the first question and narrates the evolution and complexity of the cyclic GMP signaling pathway. Besides ACTH, there are at least five additional limbs. Each embodies a unique modular design to perform a specific physiological function; exemplified by ATP binding and phosphorylation, Ca2+-sensor proteins that either increase or decrease cyclic GMP synthesis, co-expression of antithetical Ca2+ sensors, GCAP1 and S100B, and modulation by atmospheric carbon dioxide and temperature. The complexity provided by these various manners of operation enables membrane guanylate cyclase to conduct diverse functions, exemplified by the control over cardiovasculature, sensory neurons and, endocrine systems.
Collapse
|
9
|
Jacobson SG, Cideciyan AV, Ho AC, Roman AJ, Wu V, Garafalo AV, Sumaroka A, Krishnan AK, Swider M, Mascio AA, Kay CN, Yoon D, Fujita KP, Boye SL, Peshenko IV, Dizhoor AM, Boye SE. Night vision restored in days after decades of congenital blindness. iScience 2022; 25:105274. [PMID: 36274938 PMCID: PMC9579015 DOI: 10.1016/j.isci.2022.105274] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/29/2022] [Accepted: 09/30/2022] [Indexed: 11/29/2022] Open
Abstract
Signaling of vision to the brain starts with the retinal phototransduction cascade which converts visible light from the environment into chemical changes. Vision impairment results when mutations inactivate proteins of the phototransduction cascade. A severe monogenically inherited blindness, Leber congenital amaurosis (LCA), is caused by mutations in the GUCY2D gene, leading to a molecular defect in the production of cyclic GMP, the second messenger of phototransduction. We studied two patients with GUCY2D-LCA who were undergoing gene augmentation therapy. Both patients had large deficits in rod photoreceptor-based night vision before intervention. Within days of therapy, rod vision in both patients changed dramatically; improvements in visual function and functional vision in these hyper-responding patients reached more than 3 log10 units (1000-fold), nearing healthy rod vision. Quick activation of the complex molecular pathways from retinal photoreceptor to visual cortex and behavior is thus possible in patients even after being disabled and dormant for decades.
Collapse
Affiliation(s)
- Samuel G. Jacobson
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Artur V. Cideciyan
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Allen C. Ho
- Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Alejandro J. Roman
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vivian Wu
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexandra V. Garafalo
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexander Sumaroka
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Arun K. Krishnan
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Malgorzata Swider
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Abraham A. Mascio
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Dan Yoon
- Atsena Therapeutics, Inc., Durham, NC 27709, USA
| | | | - Sanford L. Boye
- Department of Pediatrics, Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32601, USA
| | - Igor V. Peshenko
- Pennsylvania College of Optometry, Salus University, Elkins Park, PA 19027, USA
| | | | - Shannon E. Boye
- Department of Pediatrics, Division of Cellular and Molecular Therapy, University of Florida College of Medicine, Gainesville, FL 32611, USA
| |
Collapse
|
10
|
OCT guided micro-focal ERG system with multiple stimulation wavelengths for characterization of ocular health. Sci Rep 2022; 12:4009. [PMID: 35256656 PMCID: PMC8901822 DOI: 10.1038/s41598-022-07622-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 01/31/2022] [Indexed: 11/25/2022] Open
Abstract
Inherited retinal disorders and dry age-related macular degeneration are characterized by the degeneration and death of different types of photoreceptors at different rate and locations. Advancement of new therapeutic interventions such as optogenetics gene therapy and cell replacement therapies are dependent on electrophysiological measurements at cellular resolution. Here, we report the development of an optical coherence tomography (OCT) guided micro-focal multi-color laser stimulation and electroretinogram (ERG) platform for highly localized monitoring of retina function. Functional evaluation of wild type and transgenic pigs affected by retinal degeneration was carried out using OCT guided micro-focal ERG (μfERG) with selected stimulation wavelengths for S, M and L cones as well as rod photoreceptors. In wild type pigs, μfERG allowed functional recording from rods and each type of cone photoreceptor cells separately. Furthermore, functional deficits in P23H transgenic pigs consistent with their retinal degeneration phenotype were observed, including decrease in the S and M cone function and lack of rod photoreceptor function. OCT guided μfERG based monitoring of physiological function will enable characterization of animal models of retinal degenerative diseases and evaluation of therapeutic interventions at the cellular level.
Collapse
|
11
|
Genome editing in large animal models. Mol Ther 2021; 29:3140-3152. [PMID: 34601132 DOI: 10.1016/j.ymthe.2021.09.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/26/2021] [Accepted: 09/26/2021] [Indexed: 12/21/2022] Open
Abstract
Although genome editing technologies have the potential to revolutionize the way we treat human diseases, barriers to successful clinical implementation remain. Increasingly, preclinical large animal models are being used to overcome these barriers. In particular, the immunogenicity and long-term safety of novel gene editing therapeutics must be evaluated rigorously. However, short-lived small animal models, such as mice and rats, cannot address secondary pathologies that may arise years after a gene editing treatment. Likewise, immunodeficient mouse models by definition lack the ability to quantify the host immune response to a novel transgene or gene-edited locus. Large animal models, including dogs, pigs, and non-human primates (NHPs), bear greater resemblance to human anatomy, immunology, and lifespan and can be studied over longer timescales with clinical dosing regimens that are more relevant to humans. These models allow for larger scale and repeated blood and tissue sampling, enabling greater depth of study and focus on rare cellular subsets. Here, we review current progress in the development and evaluation of novel genome editing therapies in large animal models, focusing on applications in human immunodeficiency virus 1 (HIV-1) infection, cancer, and genetic diseases including hemoglobinopathies, Duchenne muscular dystrophy (DMD), hypercholesterolemia, and inherited retinal diseases.
Collapse
|
12
|
Shughoury A, Ciulla TA, Bakall B, Pennesi ME, Kiss S, Cunningham ET. Genes and Gene Therapy in Inherited Retinal Disease. Int Ophthalmol Clin 2021; 61:3-45. [PMID: 34584043 DOI: 10.1097/iio.0000000000000377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
13
|
Rider AT, Henning GB, Stockman A. A reinterpretation of critical flicker-frequency (CFF) data reveals key details about light adaptation and normal and abnormal visual processing. Prog Retin Eye Res 2021; 87:101001. [PMID: 34506951 DOI: 10.1016/j.preteyeres.2021.101001] [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] [Received: 06/02/2021] [Revised: 08/25/2021] [Accepted: 08/30/2021] [Indexed: 10/20/2022]
Abstract
Our ability to see flicker has an upper frequency limit above which flicker is invisible, known as the "critical flicker frequency" (CFF), that typically grows with light intensity (I). The relation between CFF and I, the focus of nearly 200 years of research, is roughly logarithmic, i.e., CFF ∝ log(I)-a relation called the Ferry-Porter law. However, why this law should occur, and how it relates to the underlying physiology, have never been adequately explained. Over the past two decades we have measured CFF in normal observers and in patients with retinal gene defects. Here, we reanalyse and model our data and historical CFF data. Remarkably, CFF-versus-I functions measured under a wide range of conditions in patients and in normal observers all have broadly similar shapes when plotted in double-logarithmic coordinates, i.e., log (CFF)-versus-log(I). Thus, the entire dataset can be characterised by horizontal and vertical logarithmic shifts of a fixed-shape template. Shape invariance can be predicted by a simple model of visual processing built from a sequence of low-pass filters, subtractive feedforward stages and gain adjustment (Rider, Henning & Stockman, 2019). It depends primarily on the numbers of visual processing stages that approach their power-law region at a given intensity and a frequency-independent gain reduction at higher light levels. Counter-intuitively, the CFF-versus-I relation depends primarily on the gain of the visual response rather than its speed-a conclusion that changes our understanding and interpretation of human flicker perception. The Ferry-Porter "law" is merely an approximation of the shape-invariant template.
Collapse
Affiliation(s)
- Andrew T Rider
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, England, UK
| | - G Bruce Henning
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, England, UK
| | - Andrew Stockman
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, England, UK.
| |
Collapse
|
14
|
Huang CH, Yang CM, Yang CH, Hou YC, Chen TC. Leber's Congenital Amaurosis: Current Concepts of Genotype-Phenotype Correlations. Genes (Basel) 2021; 12:genes12081261. [PMID: 34440435 PMCID: PMC8392113 DOI: 10.3390/genes12081261] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/15/2021] [Accepted: 08/18/2021] [Indexed: 12/26/2022] Open
Abstract
Leber’s congenital amaurosis (LCA), one of the most severe inherited retinal dystrophies, is typically associated with extremely early onset of visual loss, nystagmus, and amaurotic pupils, and is responsible for 20% of childhood blindness. With advances in molecular diagnostic technology, the knowledge about the genetic background of LCA has expanded widely, while disease-causing variants have been identified in 38 genes. Different pathogenetic mechanisms have been found among these varieties of genetic mutations, all of which result in the dysfunction or absence of their encoded proteins participating in the visual cycle. Hence, the clinical phenotypes also exhibit extensive heterogenicity, including the course of visual impairment, involvement of the macular area, alteration in retinal structure, and residual function of the diseased photoreceptor. By reviewing the clinical course, fundoscopic images, optical coherent tomography examination, and electroretinogram, genotype-phenotype correlations could be established for common genetic mutations in LCA, which would benefit the timing of the diagnosis and thus promote early intervention. Gene therapy is promising in the management of LCA, while several clinical trials are ongoing and preliminary success has been announced, focusing on RPE65 and other common disease-causing genes. This review provides an update on the genetics, clinical examination findings, and genotype-phenotype correlations in the most well-established causative genetic mutations of LCA.
Collapse
Affiliation(s)
- Chu-Hsuan Huang
- Department of Ophthalmology, Cathay General Hospital, Taipei 106, Taiwan; (C.-H.H.); (Y.-C.H.)
| | - Chung-May Yang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei 100, Taiwan; (C.-M.Y.); (C.-H.Y.)
- Department of Ophthalmology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chang-Hao Yang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei 100, Taiwan; (C.-M.Y.); (C.-H.Y.)
- Department of Ophthalmology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Yu-Chih Hou
- Department of Ophthalmology, Cathay General Hospital, Taipei 106, Taiwan; (C.-H.H.); (Y.-C.H.)
| | - Ta-Ching Chen
- Department of Ophthalmology, National Taiwan University Hospital, Taipei 100, Taiwan; (C.-M.Y.); (C.-H.Y.)
- Correspondence: ; Tel.: +886-2-23123456
| |
Collapse
|
15
|
Motta FL, Filippelli-Silva R, Kitajima JP, Batista DA, Wohler ES, Sobreira NL, Martin RP, Ferraz Sallum JM. Analysis of an NGS retinopathy panel detects chromosome 1 uniparental isodisomy in a patient with RPE65-related leber congenital amaurosis. Ophthalmic Genet 2021; 42:553-560. [PMID: 34157943 DOI: 10.1080/13816810.2021.1938136] [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] [Indexed: 10/21/2022]
Abstract
Purpose: This study aims to demonstrate the possibility of detecting segmental uniparental isodisomy (iUPD) using a next-generation sequencing gene panel by reporting a Leber congenital amaurosis (LCA) case caused by a homozygous pathogenic variant in RPE65 (c.1022 T > C:p.Leu341Ser) inherited exclusively from the proband's mother.Methods: Samples from the trio (proband, mother, and father) were sequenced with a next-generation sequencing (NGS) retinopathy gene panel (224 genes) and the VCF file containing all variants was used in order to determine single nucleotide variant (SNV) counts from each sample across all chromosomes.Results: Trio analysis showed that of 81 Chr1 inherited variants 41 were exclusively maternal, including 21 homozygous. The other 40 variants were common to both parents. On remaining autosomal chromosomes (Chr2-22) 645 inherited variants were found, 147 of them were exclusively maternal and 132 exclusively paternal. Based on these NGS data, it was possible to note that the proband's chromosomes 1 are more similar to his mother's chromosome 1 than his father's, suggesting the pathogenic homozygous variant found in this patient was inherited exclusively from the mother due to uniparental maternal isodisomy.Conclusions: This study presents a secondary analysis pipeline to identify responsible variants for a phenotype and the correct inheritance pattern, which is a critical step to the proper and accurate genetic counseling of all family members. In addition, this approach could be used to determine iUPD in different Mendelian disorders if the sequencing panel identifies variants spread throughout the genome.
Collapse
Affiliation(s)
- Fabiana Louise Motta
- Department of Ophthalmology, Universidade Federal de São Paulo, Sao Paulo, Brazil
| | | | | | - Denise A Batista
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Elizabeth S Wohler
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Nara L Sobreira
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Renan Paulo Martin
- Department of Biophysics, Universidade Federal de São Paulo, Sao Paulo, Brazil.,McKusick-Nathans Department of Genetic Medicine, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Juliana Maria Ferraz Sallum
- Department of Ophthalmology, Universidade Federal de São Paulo, Sao Paulo, Brazil.,Instituto de Genética Ocular, Sao Paulo, Brazil
| |
Collapse
|
16
|
Suppiej A, Ceccato C, Maritan V, Cermakova I, Colavito D, Leon A. Exome sequencing and electro-clinical features in pediatric patients with very early-onset retinal dystrophies: A cohort study. Eur J Paediatr Neurol 2021; 31:1-9. [PMID: 33529788 DOI: 10.1016/j.ejpn.2021.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/01/2021] [Accepted: 01/10/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND OBJECTIVE Inherited retinal dystrophies (IRDs) are a major cause of childhood blindness. Timely diagnosis requires a high level of clinical suspicion from both neurologists and ophthalmologists and is increasingly important given recent advancements in gene therapy. We focused our study on genotype-phenotype associations in very early-onset forms of retinal dystrophy, the least well characterized and most challenging diagnostic subgroup. METHODS From January 12, 2015 to March 31, 2017, we prospectively performed whole exome sequencing targeted on the phenotype of non-syndromic IRDs and phenotype characterization in a cohort of 68 children affected by very early-onset inherited retinal dystrophies, defined by the onset before five years of age. Phenotype parameters included age at onset, clinical presentation, ophthalmic evaluation, electrophysiological patterns and clinical course. RESULTS A genetically confirmed diagnosis was achieved in 50 out of 60 (83%) families. The median age at onset was 4 months (<6 m in 70%, < 2 y in 82% of the cases). Clinical presentation was associated with visual loss and nystagmus in the majority of patients. Three (CNGB3, CNGA3 and CACNA1F) out of 22 genes considered pathogenic in the cohort, accounted for 51% of all IRD's, all within the class of stationary IRDs. CONCLUSIONS This study reports on the largest cohort of very early-onset retinal dystrophies, including a description of electroretinography patterns. The electro-clinical phenotype coupled with genetic diagnosis provided additional clues for child neurologists dealing with low vision and nystagmus in infancy. A high level of clinical suspicion improves the diagnosis with important implications for the future of the affected child.
Collapse
Affiliation(s)
- A Suppiej
- Department of Medical Sciences, Paediatric Section, University of Ferrara, Italy; Paediatric University Hospital of Padova- Neuro-ophthalmology Program, Italy; Robert Hollman Foundation, Padova, Italy.
| | - C Ceccato
- Robert Hollman Foundation, Padova, Italy
| | - V Maritan
- Paediatric University Hospital of Padova- Neuro-ophthalmology Program, Italy; Ophthalmology Unit, Azienda ULSS 6 Euganea, Padova, Italy
| | | | - D Colavito
- Research & Innovation (R&I Genetics), Padova, Italy
| | - A Leon
- Research & Innovation (R&I Genetics), Padova, Italy
| |
Collapse
|
17
|
Stockman A, Henning GB, Rider AT. Clinical vision and molecular loss: Integrating visual psychophysics with molecular genetics reveals key details of normal and abnormal visual processing. Prog Retin Eye Res 2020; 83:100937. [PMID: 33388434 DOI: 10.1016/j.preteyeres.2020.100937] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 01/08/2023]
Abstract
Over the past two decades we have developed techniques and models to investigate the ways in which known molecular defects affect visual performance. Because molecular defects in retinal signalling invariably alter the speed of visual processing, our strategy has been to measure the resulting changes in flicker sensitivity. Flicker measurements provide not only straightforward clinical assessments of visual performance but also reveal fundamental details about the functioning of both abnormal and normal visual systems. Here, we bring together our past measurements of patients with pathogenic variants in the GNAT2, RGS9, GUCA1A, RPE65, OPA1, KCNV2 and NR2E3 genes and analyse the results using a standard model of visual processing. The model treats flicker sensitivity as the result of the actions of a sequence of simple processing steps, one or more of which is altered by the genetic defect. Our analyses show that most defects slow down the visual response directly, but some speed it up. Crucially, however, other steps in the processing sequence can make compensatory adjustments to offset the abnormality. For example, if the abnormal step slows down the visual response, another step is likely to speed up or attenuate the response to rebalance system performance. Such compensatory adjustments are probably made by steps in the sequence that usually adapt to changing light levels. Our techniques and modelling also allow us to tease apart stationary and progressive effects, and the localised molecular losses help us to unravel and characterise individual steps in the normal and abnormal processing sequences.
Collapse
Affiliation(s)
- Andrew Stockman
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, England, UK.
| | - G Bruce Henning
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, England, UK
| | - Andrew T Rider
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, England, UK
| |
Collapse
|
18
|
Padhy SK, Takkar B, Narayanan R, Venkatesh P, Jalali S. Voretigene Neparvovec and Gene Therapy for Leber's Congenital Amaurosis: Review of Evidence to Date. APPLICATION OF CLINICAL GENETICS 2020; 13:179-208. [PMID: 33268999 PMCID: PMC7701157 DOI: 10.2147/tacg.s230720] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/06/2020] [Indexed: 12/13/2022]
Abstract
Gene therapy has now evolved as the upcoming modality for management of many disorders, both inheritable and non-inheritable. Knowledge of genetics pertaining to a disease has therefore become paramount for physicians across most specialities. Inheritable retinal dystrophies (IRDs) are notorious for progressive and relentless vision loss, frequently culminating in complete blindness in both eyes. Leber’s congenital amaurosis (LCA) is a typical example of an IRD that manifests very early in childhood. Research in gene therapy has led to the development and approval of voretigene neparvovec (VN) for use in patients of LCA with a deficient biallelic RPE65 gene. The procedure involves delivery of a recombinant virus vector that carries the RPE65 gene in the subretinal space. This comprehensive review reports the evidence thus far in support of gene therapy for LCA. We explore and compare the various gene targets including but not limited to RPE65, and discuss the choice of vector and method for ocular delivery. The review details the evolution of gene therapy with VN in a phased manner, concluding with the challenges that lie ahead for its translation for use in communities that differ much both genetically and economically.
Collapse
Affiliation(s)
- Srikanta Kumar Padhy
- Vitreoretina and Uveitis Services, L V Prasad Eye Institute, Mithu Tulsi Chanrai Campus, Bhubaneswar, India
| | - Brijesh Takkar
- Srimati Kanuri Santhamma Center for Vitreoretinal Diseases, Kallam Anji Reddy Campus, L.V. Prasad Eye Institute, Hyderabad, India.,Center of Excellence for Rare Eye Diseases, Kallam Anji Reddy Campus, L V Prasad Eye Institute, Hyderabad, India
| | - Raja Narayanan
- Srimati Kanuri Santhamma Center for Vitreoretinal Diseases, Kallam Anji Reddy Campus, L.V. Prasad Eye Institute, Hyderabad, India
| | - Pradeep Venkatesh
- Dr RP Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Subhadra Jalali
- Srimati Kanuri Santhamma Center for Vitreoretinal Diseases, Kallam Anji Reddy Campus, L.V. Prasad Eye Institute, Hyderabad, India.,Jasti V. Ramanamma Childrens' Eye Care Centre, Kallam Anji Reddy Campus, L V Prasad Eye Institute, Hyderabad, India
| |
Collapse
|
19
|
Gao J, Hussain RM, Weng CY. Voretigene Neparvovec in Retinal Diseases: A Review of the Current Clinical Evidence. Clin Ophthalmol 2020; 14:3855-3869. [PMID: 33223822 PMCID: PMC7671481 DOI: 10.2147/opth.s231804] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 10/21/2020] [Indexed: 12/26/2022] Open
Abstract
Subretinal gene therapy trials began with the discovery of RPE65 variants and their association with Leber congenital amaurosis. The RPE65 protein is critical for the normal functioning of the visual phototransduction cascade. RPE65 gene knockout animal models were developed and showed similar diseased phenotypes to their human counterparts. Proof of concept studies were carried out in these animal models using subretinal RPE65 gene replacement therapy, resulting in improvements in various visual function markers including electroretinograms, pupillary light responses, and object avoidance behaviors. Positive results in animal models led to Phase 1 human studies using adeno-associated viral vectors. Results in these initial human studies also showed positive impact on visual function and acceptable safety. A landmark Phase 3 study was then conducted by Spark Therapeutics using a dose of 1.5 x1011 vector genomes after dose-escalation studies confirmed its efficacy and safety. Multi-luminance mobility testing was used to measure the primary efficacy endpoint due to its excellent reliability in detecting the progression of inherited retinal diseases. After the study met its primary endpoint, the Food and Drug Administration approved voretigene neparvovec (Luxturna®) for use in RPE65-associated inherited retinal diseases.
Collapse
Affiliation(s)
- Jie Gao
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | | | - Christina Y Weng
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
20
|
Feng X, Wei T, Sun J, Luo Y, Huo Y, Yu P, Chen J, Wei X, Qi M, Ye Y. The pathogenicity of novel GUCY2D mutations in Leber congenital amaurosis 1 assessed by HPLC-MS/MS. PLoS One 2020; 15:e0231115. [PMID: 32255808 PMCID: PMC7138296 DOI: 10.1371/journal.pone.0231115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/16/2020] [Indexed: 12/04/2022] Open
Abstract
Leber congenital amaurosis (LCA) is a group of severe congenital retinal diseases. Variants in the guanylate cyclase 2D gene (GUCY2D), which encodes guanylate cyclase 1 (ROS-GC1), are associated with LCA1 and account for 6%–21% of all LCA cases. In this study, one family with LCA1 was recruited from China. A combination of next generation sequencing and Sanger sequencing was used to screen for disease-causing mutations. We found three novel mutations (c.139delC, p.Ala49Profs*36; c.835G>A, p.Asp279Asn and c.2783G>A, p.Gly928Glu) in the GUCY2D gene. Proband III-2 carries mutations c.139delC and c.2783G>A, which are inherited from the heterozygous mutation carriers, II-2 (c.139delC) and II-3 (c.2783G>A) that possess c.139delC and c.2783G>A. Additionally, II-8 carries heterozygous mutation c.835G>A. Sanger sequencing was used to confirm the presence of the three novel mutations in other family members. Mutation c.139delC results in a truncated protein. Mutations c.835G>A and c.2783G>A significantly reduce the catalytic activity of ROS-GC1. Our findings highlight the gene variants range of LCA. Moreover, HPLC-coupled tandem mass spectrometry (HPLC-MS/MS) was used to analyze the concentration of 3',5'-cyclic guanosine monophosphate (cGMP), suggesting that HPLC-MS/MS is an effective alternative method to evaluate the catalytic activity of wild-type and mutant ROS-GC1.
Collapse
Affiliation(s)
- Xue Feng
- Department of Cell Biology and Medical Genetics, School of Medicine Zhejiang University, Hangzhou, Zhejiang, China
| | - Tianying Wei
- Department of Cell Biology and Medical Genetics, School of Medicine Zhejiang University, Hangzhou, Zhejiang, China
| | - Junhui Sun
- Department of Cell Biology and Medical Genetics, School of Medicine Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuqin Luo
- Department of Reproductive Genetics, Women’s Hospital, School of Medicine Zhejiang University, Hangzhou, Zhejiang, China
| | - Yanan Huo
- Department of Eye Center, The Second Affiliated Hospital of School of Medicine Zhejiang University, Hangzhou, Zhejiang, China
| | - Ping Yu
- Department of Cell Biology and Medical Genetics, School of Medicine Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiao Chen
- Department of Cell Biology and Medical Genetics, School of Medicine Zhejiang University, Hangzhou, Zhejiang, China
| | | | - Ming Qi
- Department of Cell Biology and Medical Genetics, School of Medicine Zhejiang University, Hangzhou, Zhejiang, China
- * E-mail: (YY); (MQ)
| | - Yinghui Ye
- Department of Reproductive Endocrinology, Key Laboratory of Reproductive Genetics, Ministry of Education, and Women’s Reproductive Health Laboratory of Zhejiang Province, Women’s Hospital School of Medicine Zhejiang University, Hangzhou, Zhejiang, China
- * E-mail: (YY); (MQ)
| |
Collapse
|
21
|
Gorbatyuk MS, Starr CR, Gorbatyuk OS. Endoplasmic reticulum stress: New insights into the pathogenesis and treatment of retinal degenerative diseases. Prog Retin Eye Res 2020; 79:100860. [PMID: 32272207 DOI: 10.1016/j.preteyeres.2020.100860] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 03/08/2020] [Accepted: 03/17/2020] [Indexed: 12/13/2022]
Abstract
Physiological equilibrium in the retina depends on coordinated work between rod and cone photoreceptors and can be compromised by the expression of mutant proteins leading to inherited retinal degeneration (IRD). IRD is a diverse group of retinal dystrophies with multifaceted molecular mechanisms that are not fully understood. In this review, we focus on the contribution of chronically activated unfolded protein response (UPR) to inherited retinal pathogenesis, placing special emphasis on studies employing genetically modified animal models. As constitutively active UPR in degenerating retinas may activate pro-apoptotic programs associated with oxidative stress, pro-inflammatory signaling, dysfunctional autophagy, free cytosolic Ca2+ overload, and altered protein synthesis rate in the retina, we focus on the regulatory mechanisms of translational attenuation and approaches to overcoming translational attenuation in degenerating retinas. We also discuss current research on the role of the UPR mediator PERK and its downstream targets in degenerating retinas and highlight the therapeutic benefits of reprogramming PERK signaling in preclinical animal models of IRD. Finally, we describe pharmacological approaches targeting UPR in ocular diseases and consider their potential applications to IRD.
Collapse
Affiliation(s)
- Marina S Gorbatyuk
- The University of Alabama at Birmingham, Department of Optometry and Vision Science, School of Optometry, USA.
| | - Christopher R Starr
- The University of Alabama at Birmingham, Department of Optometry and Vision Science, School of Optometry, USA
| | - Oleg S Gorbatyuk
- The University of Alabama at Birmingham, Department of Optometry and Vision Science, School of Optometry, USA
| |
Collapse
|
22
|
Dikkaya F, Seyhan S, Erdur SK, Şentürk F, Aras C. Optical coherence tomography and fundus autofluorescence imaging in an infant with RD3-related leber congenital amaurosis. Ophthalmic Genet 2020; 41:79-82. [PMID: 32083505 DOI: 10.1080/13816810.2020.1731837] [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] [Indexed: 10/25/2022]
Abstract
Background: Leber congenital amaurosis (LCA) is both genetically and phenotypically heterogeneous group of retinal disorder. Mutations in retinal degeneration 3 (RD3) have been reported as an infrequent cause of LCA which account for less than 1% of all known LCA cases. This case report provides Optical Coherence Tomography (OCT) and Fundus Autofluorescence (FAF) findings of an infant with LCA related to a mutation in RD3.Materials and Methods: Single retrospective case report.Results: TruSight One Expanded Sequencing Panel was applied to the patient on the Illumina NextSeq. Homozygous pathogenic variant (c.112 C > T, p.Arg38Ter) was detected in the RD3 gene. Well-demarcated central foveal atrophy was noted in the infrared imaging. FAF imaging showed perifoveal hyperautofluorescent ring and irregular hyperautofluorescence outside the vascular arcade. An arrest in foveal development and loss of outer retinal structure including outer nuclear layer, external limiting membrane, ellipsoid zone and interdigitation zone at the fovea were detected in the OCT imaging.Conclusion: This study indicates that RD3-related LCA has a very severe phenotype with foveal development arrest and very early loss of all photoreceptor layer and external limiting membrane at the fovea.
Collapse
Affiliation(s)
- Funda Dikkaya
- Department of Ophthalmology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Serhat Seyhan
- Department of Medical Genetic, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Sevil Karaman Erdur
- Department of Ophthalmology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Fevzi Şentürk
- Department of Ophthalmology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Cengiz Aras
- Department of Ophthalmology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
23
|
Barny I, Perrault I, Michel C, Soussan M, Goudin N, Rio M, Thomas S, Attié-Bitach T, Hamel C, Dollfus H, Kaplan J, Rozet JM, Gerard X. Basal exon skipping and nonsense-associated altered splicing allows bypassing complete CEP290 loss-of-function in individuals with unusually mild retinal disease. Hum Mol Genet 2019; 27:2689-2702. [PMID: 29771326 DOI: 10.1093/hmg/ddy179] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/02/2018] [Accepted: 05/06/2018] [Indexed: 12/18/2022] Open
Abstract
CEP290 mutations cause a spectrum of ciliopathies from Leber congenital amaurosis type 10 (LCA10) to embryo-lethal Meckel syndrome (MKS). Using panel-based molecular diagnosis testing for inherited retinal diseases, we identified two individuals with some preserved vision despite biallelism for presumably truncating CEP290 mutations. The first one carried a homozygous 1 base pair deletion in Exon 17, introducing a premature termination codon (PTC) in Exon 18 (c.1666del; p.Ile556Phefs*17). mRNA analysis revealed a basal exon skipping (BES) of Exon 18, providing mutant cells with the ability to escape protein truncation, while disrupting the reading frame in controls. The second individual harbored compound heterozygous nonsense mutations in Exon 8 (c.508A>T, p.Lys170*) and Exon 32 (c.4090G>T, p.Glu1364*), respectively. Some CEP290 lacking Exon 8 were detected in mutant fibroblasts but not in controls whereas some skipping of Exon 32 occurred in both lines, but with higher amplitude in the mutant. Considering that the deletion of either exon maintains the reading frame in either line, skipping in mutant cells likely involves nonsense-associated altered splicing alone (Exon 8), or with BES (Exon 32). Skipping of PTC-containing exons in mutant cells allowed production of CEP290 isoforms with preserved ability to assemble into a high molecular weight complex and to interact efficiently with proteins important for cilia formation and intraflagellar trafficking. In contrast, studying LCA10 and MKS fibroblasts we show moderate to severe cilia alterations, providing support for a correlation between disease severity and the ability of cells to express shortened, yet functional, CEP290 isoforms.
Collapse
Affiliation(s)
- Iris Barny
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetics Diseases
| | - Isabelle Perrault
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetics Diseases
| | - Christel Michel
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetics Diseases
| | - Mickael Soussan
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetics Diseases
| | - Nicolas Goudin
- Cell Imaging Core Facility of the Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Imagine and Paris Descartes University, Paris, France
| | - Marlène Rio
- Department of Genetics, IHU Necker-Enfants Malades, University Paris Descartes, Paris, France
| | - Sophie Thomas
- Laboratory of Embryology and Genetics of Human Malformation, INSERM UMR1163, Institute of Genetics Diseases, Imagine and Paris Descartes University, Paris, France
| | - Tania Attié-Bitach
- Laboratory of Embryology and Genetics of Human Malformation, INSERM UMR1163, Institute of Genetics Diseases, Imagine and Paris Descartes University, Paris, France
| | - Christian Hamel
- Centre de Référence des Affections Sensorielles Génétiques, Institut des Neurosciences de Montpellier, CHU-Saint Eloi Montpellier, Montpellier, France
| | - Hélène Dollfus
- Centre de Référence pour les Affections Génétiques Ophtalmologiques CARGO, CHRU Strasbourg, INSERM 1112, Université de Strasbourg, Strasbourg, France
| | - Josseline Kaplan
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetics Diseases
| | - Jean-Michel Rozet
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetics Diseases
| | - Xavier Gerard
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetics Diseases
| |
Collapse
|
24
|
Leber congenital amaurosis: Current genetic basis, scope for genetic testing and personalized medicine. Exp Eye Res 2019; 189:107834. [PMID: 31639339 DOI: 10.1016/j.exer.2019.107834] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 10/06/2019] [Accepted: 10/10/2019] [Indexed: 02/07/2023]
Abstract
Retinal dystrophies are one of the leading causes of pediatric congenital blindness. Leber's congenital amaurosis (LCA) encompasses one of the most severe forms of inherited retinal dystrophy responsible for early-onset childhood blindness in infancy. These are clinically characterized by nystagmus, amaurotic pupil response and markedly reduced or in most instances completely absent full-field electroretinogram. LCA exhibits immense genetic heterogeneity. With advances in next-generation genetic technologies, tremendous progress has been achieved over the last two decades in discovering genes and genetic defects leading to retinal dystrophies. Currently, 28 genes have been implicated in the pathogenesis of LCA and with initial reports of success in management with targeted gene therapy the disease has attracted a lot of research attention in the recent time. The review provides an update on genetic basis of LCA, scope for genetic testing and pharmacogenetic medicine in diagnosis and treatment of these diseases.
Collapse
|
25
|
Duda T, Pertzev A, Ravichandran S, Sharma RK. Ca 2+-Sensor Neurocalcin δ and Hormone ANF Modulate ANF-RGC Activity by Diverse Pathways: Role of the Signaling Helix Domain. Front Mol Neurosci 2018; 11:430. [PMID: 30546296 PMCID: PMC6278801 DOI: 10.3389/fnmol.2018.00430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/05/2018] [Indexed: 11/24/2022] Open
Abstract
Prototype member of the membrane guanylate cyclase family, ANF-RGC (Atrial Natriuretic Factor Receptor Guanylate Cyclase), is the physiological signal transducer of two most hypotensive hormones ANF and BNP, and of the intracellular free Ca2+. Both the hormonal and the Ca2+-modulated signals operate through a common second messenger, cyclic GMP; yet, their operational modes are divergent. The hormonal pathways originate at the extracellular domain of the guanylate cyclase; and through a cascade of structural changes in its successive domains activate the C-terminal catalytic domain (CCD). In contrast, the Ca2+ signal operating via its sensor, myristoylated neurocalcin δ both originates and is translated directly at the CCD. Through a detailed sequential deletion and expression analyses, the present study examines the role of the signaling helix domain (SHD) in these two transduction pathways. SHD is a conserved 35-amino acid helical region of the guanylate cyclase, composed of five heptads, each meant to tune and transmit the hormonal signals to the CCD for their translation and generation of cyclic GMP. Its structure is homo-dimeric and the molecular docking analyses point out to the possibility of antiparallel arrangement of the helices. Contrary to the hormonal signaling, SHD has no role in regulation of the Ca2+- modulated pathway. The findings establish and define in molecular terms the presence of two distinct non-overlapping transduction modes of ANF-RGC, and for the first time demonstrate how differently they operate, and, yet generate cyclic GMP utilizing common CCD machinery.
Collapse
Affiliation(s)
- Teresa Duda
- Research Divisions of Biochemistry and Molecular Biology, The Unit of Regulatory and Molecular Biology, Salus University, Elkins Park, PA, United States
| | - Alexandre Pertzev
- Research Divisions of Biochemistry and Molecular Biology, The Unit of Regulatory and Molecular Biology, Salus University, Elkins Park, PA, United States
| | - Sarangan Ravichandran
- Advanced Biomedical Computational Sciences Group, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Leidos Biomedical Research Inc., Fredrick, MD, United States
| | - Rameshwar K Sharma
- Research Divisions of Biochemistry and Molecular Biology, The Unit of Regulatory and Molecular Biology, Salus University, Elkins Park, PA, United States
| |
Collapse
|
26
|
Wimberg H, Lev D, Yosovich K, Namburi P, Banin E, Sharon D, Koch KW. Photoreceptor Guanylate Cyclase ( GUCY2D) Mutations Cause Retinal Dystrophies by Severe Malfunction of Ca 2+-Dependent Cyclic GMP Synthesis. Front Mol Neurosci 2018; 11:348. [PMID: 30319355 PMCID: PMC6167591 DOI: 10.3389/fnmol.2018.00348] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/06/2018] [Indexed: 12/12/2022] Open
Abstract
Over 100 mutations in GUCY2D that encodes the photoreceptor guanylate cyclase GC-E are known to cause two major diseases: autosomal recessive Leber congenital amaurosis (arLCA) or autosomal dominant cone-rod dystrophy (adCRD) with a poorly understood mechanism at the molecular level in most cases. Only few mutations were further characterized for their enzymatic and molecular properties. GC-E activity is under control of neuronal Ca2+-sensor proteins, which is often a possible route to dysfunction. We investigated five recently-identified GC-E mutants that have been reported in patients suffering from arLCA (one large family) and adCRD/maculopathy (four families). Microsatellite analysis revealed that one of the mutations, c.2538G > C (p.K846N), occurred de novo. To better understand the mechanism by which mutations that are located in different GC-E domains develop different phenotypes, we investigated the molecular consequences of these mutations by expressing wildtype and mutant GC-E variants in HEK293 cells. Analyzing their general enzymatic behavior, their regulation by Ca2+ sensor proteins and retinal degeneration protein 3 (RD3) dimerization domain mutants (p.E841K and p.K846N) showed a shift in Ca2+-sensitive regulation by guanylate cyclase-activating proteins (GCAPs). Mutations in the cyclase catalytic domain led to a loss of enzyme function in the mutant p.P873R, but not in p.V902L. Instead, the p.V902L mutation increased the guanylate cyclase activity more than 20-fold showing a high GCAP independent activity and leading to a constitutively active mutant. This is the first mutation to be described affecting the GC-E catalytic core in a complete opposite way.
Collapse
Affiliation(s)
- Hanna Wimberg
- Department of Neuroscience, Biochemistry Group, University of Oldenburg, Oldenburg, Germany
| | - Dorit Lev
- The Rina Mor Institute of Medical Genetics, Wolfson Medical Center, Holon, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Keren Yosovich
- The Rina Mor Institute of Medical Genetics, Wolfson Medical Center, Holon, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Prasanthi Namburi
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Eyal Banin
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Dror Sharon
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Karl-Wilhelm Koch
- Department of Neuroscience, Biochemistry Group, University of Oldenburg, Oldenburg, Germany
| |
Collapse
|
27
|
Xu CL, Park KS, Tsang SH. CRISPR/Cas9 genome surgery for retinal diseases. DRUG DISCOVERY TODAY. TECHNOLOGIES 2018; 28:23-32. [PMID: 30205877 DOI: 10.1016/j.ddtec.2018.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 04/17/2018] [Accepted: 05/29/2018] [Indexed: 12/25/2022]
Abstract
Retinal diseases that impair vision can impose heavy physical and emotional burdens on patients' lives. Currently, clustered regularly interspaced short palindromic repeats (CRISPR) is a prevalent gene-editing tool that can be harnessed to generate disease model organisms for specific retinal diseases, which are useful for elucidating pathophysiology and revealing important links between genetic mutations and phenotypic defects. These retinal disease models are fundamental for testing various therapies and are indispensible for potential future clinical trials. CRISPR-mediated procedures involving CRISPR-associated protein 9 (Cas9) may also be used to edit genome sequences and correct mutations. Thus, if used for future therapies, CRISPR/Cas9 genome surgery could eliminate the need for patients with retinal diseases to undergo repetitive procedures such as drug injections. In this review, we will provide an overview of CRISPR/Cas9, discuss the different types of Cas9, and compare Cas9 to other endonucleases. Furthermore, we will explore the many ways in which researchers are currently utilizing this versatile tool, as CRISPR/Cas9 may have far-reaching effects in the treatment of retinal diseases.
Collapse
Affiliation(s)
- Christine L Xu
- Edward S Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA; Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Karen Sophia Park
- Edward S Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA; Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Stephen H Tsang
- Edward S Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA; Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia University, New York, NY, USA; Department of Pathology & Cell Biology, Institute of Human Nutrition, Columbia Stem Cell Initiative, College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
28
|
Sheck L, Davies WIL, Moradi P, Robson AG, Kumaran N, Liasis AC, Webster AR, Moore AT, Michaelides M. Leber Congenital Amaurosis Associated with Mutations in CEP290, Clinical Phenotype, and Natural History in Preparation for Trials of Novel Therapies. Ophthalmology 2018; 125:894-903. [PMID: 29398085 PMCID: PMC5974693 DOI: 10.1016/j.ophtha.2017.12.013] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 12/01/2017] [Accepted: 12/07/2017] [Indexed: 11/21/2022] Open
Abstract
PURPOSE To investigate and describe in detail the demographics, functional and anatomic characteristics, and clinical course of Leber congenital amaurosis (LCA) associated with mutations in the CEP290 gene (LCA-CEP290) in a large cohort of adults and children. DESIGN Retrospective case series. PARTICIPANTS Patients with mutations in CEP290 identified at a single UK referral center. METHODS Review of case notes and results of retinal imaging (color fundus photography, fundus autofluorescence [FAF] imaging, OCT), electrophysiologic assessment, and molecular genetic testing. MAIN OUTCOME MEASURES Molecular genetic testing, clinical findings including visual acuity and retinal imaging, and electrophysiologic assessment. RESULTS Forty patients with LCA-CEP290 were identified. The deep intronic mutation c.2991+1655 A>G was the most common disease-causing variant (23/40 patients) identified in the compound heterozygous state in 20 patients (50%) and homozygous in 2 patients (5%). Visual acuity (VA) varied from 6/9 to no perception of light, and only 2 of 12 patients with longitudinal VA data showed deterioration in VA in their better-seeing eye over time. A normal fundus was found at diagnosis in younger patients (mean age, 1.9 years), with older patients showing white flecks (mean age, 5.9 years) or pigmentary retinopathy (mean age, 21.7 years). Eleven of 12 patients (92%) with OCT imaging had preservation of foveal architecture. Ten of 12 patients (83%) with FAF imaging had a perifoveal hyperautofluorescent ring. Having 2 nonsense CEP290 mutations was associated with worse final VA and the presence of nonocular features. CONCLUSIONS Detailed analysis of the clinical phenotype of LCA-CEP290 in a large cohort confirms that there is a window of opportunity in childhood for therapeutic intervention based on relative structural preservation in the central cone-rich retina in a significant proportion of patients, with the majority harboring the deep intronic variant potentially tractable to several planned gene editing approaches.
Collapse
Affiliation(s)
- Leo Sheck
- Moorfields Eye Hospital, London, United Kingdom
| | - Wayne I L Davies
- UWA Oceans Institute, University of Western Australia, Crawley, Australia; School of Biological Sciences, University of Western Australia, Crawley, Australia; Lions Eye Institute, University of Western Australia, Crawley, Australia
| | - Phillip Moradi
- Moorfields Eye Hospital, London, United Kingdom; University College London Institute of Ophthalmology, University College London, United Kingdom
| | - Anthony G Robson
- Moorfields Eye Hospital, London, United Kingdom; University College London Institute of Ophthalmology, University College London, United Kingdom
| | | | - Alki C Liasis
- Great Ormond Street Hospital, London, United Kingdom
| | - Andrew R Webster
- Moorfields Eye Hospital, London, United Kingdom; University College London Institute of Ophthalmology, University College London, United Kingdom
| | - Anthony T Moore
- Moorfields Eye Hospital, London, United Kingdom; University College London Institute of Ophthalmology, University College London, United Kingdom; Ophthalmology Department, School of Medicine, University of California San Francisco, San Francisco, California
| | - Michel Michaelides
- Moorfields Eye Hospital, London, United Kingdom; University College London Institute of Ophthalmology, University College London, United Kingdom.
| |
Collapse
|
29
|
DiCarlo JE, Mahajan VB, Tsang SH. Gene therapy and genome surgery in the retina. J Clin Invest 2018; 128:2177-2188. [PMID: 29856367 DOI: 10.1172/jci120429] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Precision medicine seeks to treat disease with molecular specificity. Advances in genome sequence analysis, gene delivery, and genome surgery have allowed clinician-scientists to treat genetic conditions at the level of their pathology. As a result, progress in treating retinal disease using genetic tools has advanced tremendously over the past several decades. Breakthroughs in gene delivery vectors, both viral and nonviral, have allowed the delivery of genetic payloads in preclinical models of retinal disorders and have paved the way for numerous successful clinical trials. Moreover, the adaptation of CRISPR-Cas systems for genome engineering have enabled the correction of both recessive and dominant pathogenic alleles, expanding the disease-modifying power of gene therapies. Here, we highlight the translational progress of gene therapy and genome editing of several retinal disorders, including RPE65-, CEP290-, and GUY2D-associated Leber congenital amaurosis, as well as choroideremia, achromatopsia, Mer tyrosine kinase- (MERTK-) and RPGR X-linked retinitis pigmentosa, Usher syndrome, neovascular age-related macular degeneration, X-linked retinoschisis, Stargardt disease, and Leber hereditary optic neuropathy.
Collapse
Affiliation(s)
- James E DiCarlo
- Jonas Children's Vision Care and Bernard and Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York, USA.,Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, New York, USA
| | - Vinit B Mahajan
- Omics Laboratory, Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, California, USA.,Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Stephen H Tsang
- Jonas Children's Vision Care and Bernard and Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York, USA.,Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, New York, USA
| |
Collapse
|
30
|
Duda T, Pertzev A, Sharma RK. CO 2/bicarbonate modulates cone photoreceptor ROS-GC1 and restores its CORD6-linked catalytic activity. Mol Cell Biochem 2018; 448:91-105. [PMID: 29427171 DOI: 10.1007/s11010-018-3317-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 02/02/2018] [Indexed: 01/12/2023]
Abstract
This study with recombinant reconstituted system mimicking the cellular conditions of the native cones documents that photoreceptor ROS-GC1 is modulated by gaseous CO2. Mechanistically, CO2 is sensed by carbonic anhydrase (CAII), generates bicarbonate that, in turn, directly targets the core catalytic domain of ROS-GC1, and activates it to increased synthesis of cyclic GMP. This, then, functions as a second messenger for the cone phototransduction. The study demonstrates that, in contrast to the Ca2+-modulated phototransduction, the CO2 pathway is Ca2+-independent, yet is linked with it and synergizes it. It, through R787C mutation in the third heptad of the signal helix domain of ROS-GC1, affects cone-rod dystrophy, CORD6. CORD6 is caused firstly by lowered basal and GCAP1-dependent ROS-GC1 activity and secondly, by a shift in Ca2+ sensitivity of the ROS-GC1/GCAP1 complex that remains active in darkness. Remarkably, the first but not the second defect disappears with bicarbonate thus explaining the basis for CORD6 pathological severity. Because cones, but not rods, express CAII, the excessive synthesis of cyclic GMP would be most acute in cones.
Collapse
Affiliation(s)
- Teresa Duda
- Research Divisions of Biochemistry and Molecular Biology, The Unit of Regulatory and Molecular Biology, Salus University, Elkins Park, PA, USA
| | - Alexander Pertzev
- Research Divisions of Biochemistry and Molecular Biology, The Unit of Regulatory and Molecular Biology, Salus University, Elkins Park, PA, USA
| | - Rameshwar K Sharma
- Research Divisions of Biochemistry and Molecular Biology, The Unit of Regulatory and Molecular Biology, Salus University, Elkins Park, PA, USA.
| |
Collapse
|
31
|
Luscan R, Mechaussier S, Paul A, Tian G, Gérard X, Defoort-Dellhemmes S, Loundon N, Audo I, Bonnin S, LeGargasson JF, Dumont J, Goudin N, Garfa-Traoré M, Bras M, Pouliet A, Bessières B, Boddaert N, Sahel JA, Lyonnet S, Kaplan J, Cowan NJ, Rozet JM, Marlin S, Perrault I. Mutations in TUBB4B Cause a Distinctive Sensorineural Disease. Am J Hum Genet 2017; 101:1006-1012. [PMID: 29198720 DOI: 10.1016/j.ajhg.2017.10.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 10/25/2017] [Indexed: 01/07/2023] Open
Abstract
Leber congenital amaurosis (LCA) is a neurodegenerative disease of photoreceptor cells that causes blindness within the first year of life. It occasionally occurs in syndromic metabolic diseases and plurisystemic ciliopathies. Using exome sequencing in a multiplex family and three simplex case subjects with an atypical association of LCA with early-onset hearing loss, we identified two heterozygous mutations affecting Arg391 in β-tubulin 4B isotype-encoding (TUBB4B). Inspection of the atomic structure of the microtubule (MT) protofilament reveals that the β-tubulin Arg391 residue contributes to a binding pocket that interacts with α-tubulin contained in the longitudinally adjacent αβ-heterodimer, consistent with a role in maintaining MT stability. Functional analysis in cultured cells overexpressing FLAG-tagged wild-type or mutant TUBB4B as well as in primary skin-derived fibroblasts showed that the mutant TUBB4B is able to fold, form αβ-heterodimers, and co-assemble into the endogenous MT lattice. However, the dynamics of growing MTs were consistently altered, showing that the mutations have a significant dampening impact on normal MT growth. Our findings provide a link between sensorineural disease and anomalies in MT behavior and describe a syndromic LCA unrelated to ciliary dysfunction.
Collapse
Affiliation(s)
- Romain Luscan
- Laboratory of Embryology and Genetics of Human Malformation, INSERM UMR1163, Institute of Genetic Diseases, Imagine and Paris Descartes University, 75015 Paris, France
| | - Sabrina Mechaussier
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetic Diseases, Imagine and Paris Descartes University, 75015 Paris, France
| | - Antoine Paul
- Laboratory of Embryology and Genetics of Human Malformation, INSERM UMR1163, Institute of Genetic Diseases, Imagine and Paris Descartes University, 75015 Paris, France
| | - Guoling Tian
- Department of Biochemistry & Molecular Pharmacology, NYU Langone Medical Center, New York, NY 10016, USA
| | - Xavier Gérard
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetic Diseases, Imagine and Paris Descartes University, 75015 Paris, France
| | - Sabine Defoort-Dellhemmes
- Service d'Exploration de la Vision et Neuro-ophtalmologie, Pôle d'Imagerie et Explorations Fonctionnelles, CHRU de Lille, Hôpital Roger Salengro, 59000 Lille, France
| | - Natalie Loundon
- Pediatric ENT Department, Hôpital Necker-Enfants Malades, APHP and Paris Descartes University, 75015 Paris, France
| | - Isabelle Audo
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, 75012 Paris, France
| | - Sophie Bonnin
- Ophthalmology Department, Hôpital Lariboisière, APHP and Paris Diderot University, 75010 Paris, France
| | - Jean-François LeGargasson
- Visual Exploration Department, Hôpital Lariboisière, APHP, Paris, Diderot University, 75010 Paris, France
| | - Julien Dumont
- Cell Division and Reproduction, Institut Jacques Monod, CNRS, University Paris Diderot, 75013 Paris, France
| | - Nicolas Goudin
- Cell Imaging Core Facility of the Structure Fédérative de Recherche Necker INSERM US24/CNRS UMS3633 Imagine and Paris Descartes University, 75015 Paris, France
| | - Meriem Garfa-Traoré
- Cell Imaging Core Facility of the Structure Fédérative de Recherche Necker INSERM US24/CNRS UMS3633 Imagine and Paris Descartes University, 75015 Paris, France
| | - Marc Bras
- Bioinformatics Platform, Imagine and Paris Descartes University, 75015 Paris, France
| | - Aurore Pouliet
- Genomics Platform, Imagine and Paris Descartes University, 75015 Paris, France
| | - Bettina Bessières
- Unité d'Embryo-foetopathologie, Hôpital Necker-Enfants Malades, APHP and Paris Descartes University, 75015 Paris, France
| | - Nathalie Boddaert
- Department of Pediatric Radiology, Hôpital Necker-Enfants Malades, APHP, Paris, Descartes University, 75015 Paris, France
| | - José-Alain Sahel
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, 75012 Paris, France
| | - Stanislas Lyonnet
- Laboratory of Embryology and Genetics of Human Malformation, INSERM UMR1163, Institute of Genetic Diseases, Imagine and Paris Descartes University, 75015 Paris, France
| | - Josseline Kaplan
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetic Diseases, Imagine and Paris Descartes University, 75015 Paris, France
| | - Nicholas J Cowan
- Department of Biochemistry & Molecular Pharmacology, NYU Langone Medical Center, New York, NY 10016, USA
| | - Jean-Michel Rozet
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetic Diseases, Imagine and Paris Descartes University, 75015 Paris, France.
| | - Sandrine Marlin
- Laboratory of Embryology and Genetics of Human Malformation, INSERM UMR1163, Institute of Genetic Diseases, Imagine and Paris Descartes University, 75015 Paris, France; Centre de Référence des Surdités Génétiques, Genetic Department, Hôpital Necker-Enfants Malades, APHP and Paris Descartes University, 75015 Paris, France.
| | - Isabelle Perrault
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetic Diseases, Imagine and Paris Descartes University, 75015 Paris, France
| |
Collapse
|
32
|
Chung DC, McCague S, Yu ZF, Thill S, DiStefano-Pappas J, Bennett J, Cross D, Marshall K, Wellman J, High KA. Novel mobility test to assess functional vision in patients with inherited retinal dystrophies. Clin Exp Ophthalmol 2017; 46:247-259. [PMID: 28697537 DOI: 10.1111/ceo.13022] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 06/20/2017] [Accepted: 06/25/2017] [Indexed: 01/21/2023]
Abstract
IMPORTANCE This novel endpoint tracks functional vision changes in patients with inherited retinal dystrophies (IRDs) over time. BACKGROUND The aims of the study were to determine whether a multi-luminance mobility test (MLMT) can detect functional vision changes over time in subjects with IRDs and to assess natural history and potential effects of investigational agents. DESIGN This is a prospective, observational study. PARTICIPANTS Sixty-two subjects were enrolled. Sixty (29 normal sighted and 31 visually impaired) were eligible; 54 (28 visually impaired and 26 normal-sighted) completed all testing visits. METHODS Subjects navigated MLMT courses three times over 1 year. At each visit, subjects completed testing using individual eyes, and both eyes, at up to nine standardized, increasing luminance levels (range 1 to 400 lux). Accuracy and speed were evaluated and compared with visual acuity (VA), visual field (VF) and a visual function questionnaire. MAIN OUTCOME MEASURES Accuracy and speed of normal and visually impaired subjects on MLMT, and reliability and content validity of MLMT were the main outcome measures. RESULTS MLMT distinguished normal-sighted from visually impaired subjects. All control subjects passed all MLMT attempts at all tested light levels. Visually impaired subjects' performance varied widely; some declined over 1 year. Performance declined markedly below certain VA and VF thresholds. Concordance on performance on two baseline visits was high: correlations for accuracy were 94% and 98% for lowest common and highest common lux levels. CONCLUSIONS AND RELEVANCE MLMT differentiated visually impaired from control populations and, in visually impaired subjects, identified a range of performances; and tracked performance declines over time, consistent with these progressive conditions.
Collapse
Affiliation(s)
- Daniel C Chung
- Spark Therapeutics, Inc., Philadelphia, Pennsylvania, USA
| | - Sarah McCague
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Zi-Fan Yu
- Statistics Collaborative, Inc., Washington, District of Columbia, USA
| | - Satha Thill
- Statistics Collaborative, Inc., Washington, District of Columbia, USA
| | | | - Jean Bennett
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dominique Cross
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Kathleen Marshall
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
33
|
Kumaran N, Moore AT, Weleber RG, Michaelides M. Leber congenital amaurosis/early-onset severe retinal dystrophy: clinical features, molecular genetics and therapeutic interventions. Br J Ophthalmol 2017; 101:1147-1154. [PMID: 28689169 PMCID: PMC5574398 DOI: 10.1136/bjophthalmol-2016-309975] [Citation(s) in RCA: 195] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 04/26/2017] [Accepted: 04/30/2017] [Indexed: 12/29/2022]
Abstract
Leber congenital amaurosis (LCA) and early-onset severe retinal dystrophy (EOSRD) are both genetically and phenotypically heterogeneous, and characterised clinically by severe congenital/early infancy visual loss, nystagmus, amaurotic pupils and markedly reduced/absent full-field electroretinograms. The vast genetic heterogeneity of inherited retinal disease has been established over the last 10 - 20 years, with disease-causing variants identified in 25 genes to date associated with LCA/EOSRD, accounting for 70–80% of cases, with thereby more genes yet to be identified. There is now far greater understanding of the structural and functional associations seen in the various LCA/EOSRD genotypes. Subsequent development/characterisation of LCA/EOSRD animal models has shed light on the underlying pathogenesis and allowed the demonstration of successful rescue with gene replacement therapy and pharmacological intervention in multiple models. These advancements have culminated in more than 12 completed, ongoing and anticipated phase I/II and phase III gene therapy and pharmacological human clinical trials. This review describes the clinical and genetic characteristics of LCA/EOSRD and the differential diagnoses to be considered. We discuss in further detail the diagnostic clinical features, pathophysiology, animal models and human treatment studies and trials, in the more common genetic subtypes and/or those closest to intervention.
Collapse
Affiliation(s)
- Neruban Kumaran
- UCL Institute of Ophthalmology, University College London, London, UK.,Moorfields Eye Hospital NHS Foundation Trust, London, UK
| | - Anthony T Moore
- UCL Institute of Ophthalmology, University College London, London, UK.,Moorfields Eye Hospital NHS Foundation Trust, London, UK.,University of California San Francisco, San Francisco CA, California, USA
| | - Richard G Weleber
- Casey Eye Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Michel Michaelides
- UCL Institute of Ophthalmology, University College London, London, UK.,Moorfields Eye Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
34
|
Sharif W, Sharif Z. Leber's congenital amaurosis and the role of gene therapy in congenital retinal disorders. Int J Ophthalmol 2017; 10:480-484. [PMID: 28393043 DOI: 10.18240/ijo.2017.03.24] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 08/19/2016] [Indexed: 11/23/2022] Open
Abstract
Leber's congenital amaurosis (LCA) and recent gene therapy advancement for treating inherited retinopathies were extensive literature reviewed using MEDLINE, PubMed and EMBASE. Adeno-associated viral vectors were the most utilised vectors for ocular gene therapy. Cone photoreceptor cells might use an alternate pathway which was not reliant of the retinal pigment epithelium (RPE) derived retinoid isomerohydrolase (RPE65) to access the 11-cis retinal dehydechromophore. Research efforts dedicated on the progression of a gene-based therapy for the treatment of LCA2. Such gene therapy approaches were extremely successful in canine, porcine and rodent LCA2 models. The recombinant AAV2.hRPE65v2 adeno-associated vector contained the RPE65 cDNA and was replication deficient. Its in vitro injection in target cells induced RPE65 protein production. The gene therapy trials that were so far conducted for inherited retinopathies have generated promising results. Phase I clinical trials to cure LCA and choroideremia demonstrated that adeno-associated viral vectors containing RPE genes and photoreceptors respectively, could be successfully administered to inherited retinopathy patients. A phase III trial is presently ongoing and if successful, it will lead the way to additional gene therapy attempts to cure monogenic, inherited retinopathies.
Collapse
Affiliation(s)
- Walid Sharif
- Department of Ophthalmology Birmingham & Midland Eye Centre, City Hospital NHS Trust, Birmingham B18 7QH, UK
| | - Zuhair Sharif
- Institute of Ophthalmology, University College London 11-43 Bath St, London EC1V 9EL, UK
| |
Collapse
|
35
|
Sahu B, Maeda A. Retinol Dehydrogenases Regulate Vitamin A Metabolism for Visual Function. Nutrients 2016; 8:E746. [PMID: 27879662 PMCID: PMC5133129 DOI: 10.3390/nu8110746] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 11/13/2016] [Accepted: 11/16/2016] [Indexed: 02/07/2023] Open
Abstract
The visual system produces visual chromophore, 11-cis-retinal from dietary vitamin A, all-trans-retinol making this vitamin essential for retinal health and function. These metabolic events are mediated by a sequential biochemical process called the visual cycle. Retinol dehydrogenases (RDHs) are responsible for two reactions in the visual cycle performed in retinal pigmented epithelial (RPE) cells, photoreceptor cells and Müller cells in the retina. RDHs in the RPE function as 11-cis-RDHs, which oxidize 11-cis-retinol to 11-cis-retinal in vivo. RDHs in rod photoreceptor cells in the retina work as all-trans-RDHs, which reduce all-trans-retinal to all-trans-retinol. Dysfunction of RDHs can cause inherited retinal diseases in humans. To facilitate further understanding of human diseases, mouse models of RDHs-related diseases have been carefully examined and have revealed the physiological contribution of specific RDHs to visual cycle function and overall retinal health. Herein we describe the function of RDHs in the RPE and the retina, particularly in rod photoreceptor cells, their regulatory properties for retinoid homeostasis and future therapeutic strategy for treatment of retinal diseases.
Collapse
Affiliation(s)
- Bhubanananda Sahu
- Department of Ophthalmology and Visual Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4965, USA.
| | - Akiko Maeda
- Department of Ophthalmology and Visual Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4965, USA.
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106-4965, USA.
| |
Collapse
|
36
|
Abstract
Leber congenital amaurosis (LCA) is a group of severe inherited retinal dystrophies that lead to early childhood blindness. In the last decade, interest in LCA has increased as advances in genetics have been applied to better identify, classify, and treat LCA. To date, 23 LCA genes have been identified. Gene replacement in the RPE65 form of LCA represents a major advance in treatment, although limitations have been recognized. In this article, we review the clinical and genetic features of LCA and evaluate the evidence available for gene therapy in RPE65 disease.
Collapse
Affiliation(s)
- Maan Alkharashi
- a Boston Children's Hospital, Harvard Medical School , Boston , MA , USA.,b Department of Ophthalmology , King Saud University , Riyadh , Saudi Arabia
| | - Anne B Fulton
- b Department of Ophthalmology , King Saud University , Riyadh , Saudi Arabia
| |
Collapse
|
37
|
Wang SY, Zhang Q, Zhang X, Zhao PQ. Comprehensive analysis of genetic variations in strictly-defined Leber congenital amaurosis with whole-exome sequencing in Chinese. Int J Ophthalmol 2016; 9:1260-4. [PMID: 27672588 DOI: 10.18240/ijo.2016.09.04] [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: 02/21/2016] [Accepted: 03/11/2016] [Indexed: 11/23/2022] Open
Abstract
AIM To make a comprehensive analysis of the potential pathogenic genes related with Leber congenital amaurosis (LCA) in Chinese. METHODS LCA subjects and their families were retrospectively collected from 2013 to 2015. Firstly, whole-exome sequencing was performed in patients who had underwent gene mutation screening with nothing found, and then homozygous sites was selected, candidate sites were annotated, and pathogenic analysis was conducted using softwares including Sorting Tolerant from Intolerant (SIFT), Polyphen-2, Mutation assessor, Condel, and Functional Analysis through Hidden Markov Models (FATHMM). Furthermore, Gene Ontology function and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses of pathogenic genes were performed followed by co-segregation analysis using Fisher exact Test. Sanger sequencing was used to validate single-nucleotide variations (SNVs). Expanded verification was performed in the rest patients. RESULTS Totally 51 LCA families with 53 patients and 24 family members were recruited. A total of 104 SNVs (66 LCA-related genes and 15 co-segregated genes) were submitted for expand verification. The frequencies of homozygous mutation of KRT12 and CYP1A1 were simultaneously observed in 3 families. Enrichment analysis showed that the potential pathogenic genes were mainly enriched in functions related to cell adhesion, biological adhesion, retinoid metabolic process, and eye development biological adhesion. Additionally, WFS1 and STAU2 had the highest homozygous frequencies. CONCLUSION LCA is a highly heterogeneous disease. Mutations in KRT12, CYP1A1, WFS1, and STAU2 may be involved in the development of LCA.
Collapse
Affiliation(s)
- Shi-Yuan Wang
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Qi Zhang
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Xiang Zhang
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Pei-Quan Zhao
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
38
|
Sharma RK, Duda T, Makino CL. Integrative Signaling Networks of Membrane Guanylate Cyclases: Biochemistry and Physiology. Front Mol Neurosci 2016; 9:83. [PMID: 27695398 PMCID: PMC5023690 DOI: 10.3389/fnmol.2016.00083] [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] [Received: 07/13/2016] [Accepted: 08/29/2016] [Indexed: 12/24/2022] Open
Abstract
This monograph presents a historical perspective of cornerstone developments on the biochemistry and physiology of mammalian membrane guanylate cyclases (MGCs), highlighting contributions made by the authors and their collaborators. Upon resolution of early contentious studies, cyclic GMP emerged alongside cyclic AMP, as an important intracellular second messenger for hormonal signaling. However, the two signaling pathways differ in significant ways. In the cyclic AMP pathway, hormone binding to a G protein coupled receptor leads to stimulation or inhibition of an adenylate cyclase, whereas the cyclic GMP pathway dispenses with intermediaries; hormone binds to an MGC to affect its activity. Although the cyclic GMP pathway is direct, it is by no means simple. The modular design of the molecule incorporates regulation by ATP binding and phosphorylation. MGCs can form complexes with Ca2+-sensing subunits that either increase or decrease cyclic GMP synthesis, depending on subunit identity. In some systems, co-expression of two Ca2+ sensors, GCAP1 and S100B with ROS-GC1 confers bimodal signaling marked by increases in cyclic GMP synthesis when intracellular Ca2+ concentration rises or falls. Some MGCs monitor or are modulated by carbon dioxide via its conversion to bicarbonate. One MGC even functions as a thermosensor as well as a chemosensor; activity reaches a maximum with a mild drop in temperature. The complexity afforded by these multiple limbs of operation enables MGC networks to perform transductions traditionally reserved for G protein coupled receptors and Transient Receptor Potential (TRP) ion channels and to serve a diverse array of functions, including control over cardiac vasculature, smooth muscle relaxation, blood pressure regulation, cellular growth, sensory transductions, neural plasticity and memory.
Collapse
Affiliation(s)
- Rameshwar K Sharma
- The Unit of Regulatory and Molecular Biology, Research Divisions of Biochemistry and Molecular Biology, Salus University Elkins Park, PA, USA
| | - Teresa Duda
- The Unit of Regulatory and Molecular Biology, Research Divisions of Biochemistry and Molecular Biology, Salus University Elkins Park, PA, USA
| | - Clint L Makino
- Department of Physiology and Biophysics, Boston University School of Medicine Boston, MA, USA
| |
Collapse
|
39
|
Geisler A, Fechner H. MicroRNA-regulated viral vectors for gene therapy. World J Exp Med 2016; 6:37-54. [PMID: 27226955 PMCID: PMC4873559 DOI: 10.5493/wjem.v6.i2.37] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 03/02/2016] [Accepted: 03/17/2016] [Indexed: 02/06/2023] Open
Abstract
Safe and effective gene therapy approaches require targeted tissue-specific transfer of a therapeutic transgene. Besides traditional approaches, such as transcriptional and transductional targeting, microRNA-dependent post-transcriptional suppression of transgene expression has been emerging as powerful new technology to increase the specificity of vector-mediated transgene expression. MicroRNAs are small non-coding RNAs and often expressed in a tissue-, lineage-, activation- or differentiation-specific pattern. They typically regulate gene expression by binding to imperfectly complementary sequences in the 3' untranslated region (UTR) of the mRNA. To control exogenous transgene expression, tandem repeats of artificial microRNA target sites are usually incorporated into the 3' UTR of the transgene expression cassette, leading to subsequent degradation of transgene mRNA in cells expressing the corresponding microRNA. This targeting strategy, first shown for lentiviral vectors in antigen presenting cells, has now been used for tissue-specific expression of vector-encoded therapeutic transgenes, to reduce immune response against the transgene, to control virus tropism for oncolytic virotherapy, to increase safety of live attenuated virus vaccines and to identify and select cell subsets for pluripotent stem cell therapies, respectively. This review provides an introduction into the technical mechanism underlying microRNA-regulation, highlights new developments in this field and gives an overview of applications of microRNA-regulated viral vectors for cardiac, suicide gene cancer and hematopoietic stem cell therapy, as well as for treatment of neurological and eye diseases.
Collapse
|
40
|
Ashtari M, Zhang H, Cook PA, Cyckowski LL, Shindler KS, Marshall KA, Aravand P, Vossough A, Gee JC, Maguire AM, Baker CI, Bennett J. Plasticity of the human visual system after retinal gene therapy in patients with Leber's congenital amaurosis. Sci Transl Med 2016; 7:296ra110. [PMID: 26180100 DOI: 10.1126/scitranslmed.aaa8791] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Much of our knowledge of the mechanisms underlying plasticity in the visual cortex in response to visual impairment, vision restoration, and environmental interactions comes from animal studies. We evaluated human brain plasticity in a group of patients with Leber's congenital amaurosis (LCA), who regained vision through gene therapy. Using non-invasive multimodal neuroimaging methods, we demonstrated that reversing blindness with gene therapy promoted long-term structural plasticity in the visual pathways emanating from the treated retina of LCA patients. The data revealed improvements and normalization along the visual fibers corresponding to the site of retinal injection of the gene therapy vector carrying the therapeutic gene in the treated eye compared to the visual pathway for the untreated eye of LCA patients. After gene therapy, the primary visual pathways (for example, geniculostriate fibers) in the treated retina were similar to those of sighted control subjects, whereas the primary visual pathways of the untreated retina continued to deteriorate. Our results suggest that visual experience, enhanced by gene therapy, may be responsible for the reorganization and maturation of synaptic connectivity in the visual pathways of the treated eye in LCA patients. The interactions between the eye and the brain enabled improved and sustained long-term visual function in patients with LCA after gene therapy.
Collapse
Affiliation(s)
- Manzar Ashtari
- Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania Perelman School of Medicine, 309 Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA 19104, USA. F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19014, USA.
| | - Hui Zhang
- Department of Computer Science and Centre for Medical Image Computing, University College London, Gower Street, London WC1E 6BT, UK
| | - Philip A Cook
- Penn Image Computing and Science Laboratory, Department of Radiology, University of Pennsylvania, 3600 Market Street, Philadelphia, PA 19104, USA
| | - Laura L Cyckowski
- Department of Radiology, The Children's Hospital of Philadelphia, Philadelphia, PA 19014, USA
| | - Kenneth S Shindler
- Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania Perelman School of Medicine, 309 Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA 19104, USA. F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19014, USA
| | - Kathleen A Marshall
- Center for Cellular and Molecular Therapeutics at The Children's Hospital of Philadelphia, Colket Translational Research Building, 3501 Civic Center Boulevard, Philadelphia, PA 19014, USA
| | - Puya Aravand
- Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania Perelman School of Medicine, 309 Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA 19104, USA. F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19014, USA
| | - Arastoo Vossough
- Department of Radiology, The Children's Hospital of Philadelphia, Philadelphia, PA 19014, USA
| | - James C Gee
- Penn Image Computing and Science Laboratory, Department of Radiology, University of Pennsylvania, 3600 Market Street, Philadelphia, PA 19104, USA
| | - Albert M Maguire
- Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania Perelman School of Medicine, 309 Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA 19104, USA. F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19014, USA. Center for Cellular and Molecular Therapeutics at The Children's Hospital of Philadelphia, Colket Translational Research Building, 3501 Civic Center Boulevard, Philadelphia, PA 19014, USA
| | - Chris I Baker
- Laboratory of Brain and Cognition, National Institutes of Health, 10 Center Drive, MSC 1240, Bethesda, MD 20892, USA
| | - Jean Bennett
- Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania Perelman School of Medicine, 309 Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA 19104, USA. F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19014, USA. Center for Cellular and Molecular Therapeutics at The Children's Hospital of Philadelphia, Colket Translational Research Building, 3501 Civic Center Boulevard, Philadelphia, PA 19014, USA
| |
Collapse
|
41
|
Lyons LA, Creighton EK, Alhaddad H, Beale HC, Grahn RA, Rah H, Maggs DJ, Helps CR, Gandolfi B. Whole genome sequencing in cats, identifies new models for blindness in AIPL1 and somite segmentation in HES7. BMC Genomics 2016; 17:265. [PMID: 27030474 PMCID: PMC4815086 DOI: 10.1186/s12864-016-2595-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 03/16/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The reduced cost and improved efficiency of whole genome sequencing (WGS) is drastically improving the development of cats as biomedical models. Persian cats are models for Leber's congenital amaurosis (LCA), the most severe and earliest onset form of visual impairment in humans. Cats with innocuous breed-defining traits, such as a bobbed tail, can also be models for somite segmentation and vertebral column development. METHODS The first WGS in cats was conducted on a trio segregating for LCA and the bobbed tail abnormality. Variants were identified using FreeBayes and effects predicted using SnpEff. Variants within a known haplotype block for cat LCA and specific candidate genes for both phenotypes were prioritized by the predicted variant effect on the proteins and concordant segregation within the trio. The efficiency of WGS of a single trio of domestic cats was evaluated. RESULTS A stop gain was identified at position c.577C > T in cat AIPL1, a predicted p.Arg193*. A c.5A > G variant causing a p.V2A was identified in HES7. The variants segregated concordantly in a Persian - Japanese bobtail pedigree. Over 1700 cats from 40 different breeds and populations were genotyped for the AIPL1 variant, defining an allelic frequency in only Persian -related breeds of 1.15%. A sub-set of cats was genotyped for the HES7 variant, supporting the variant as private to the Japanese bobtail breed. Approximately 18 million SNPs were identified for application in cat research. The cat AIPL1 variant would have been considered a high priority variant for evaluation, regardless of a priori knowledge from previous genetic studies. CONCLUSIONS This study represents the first effort of the 99 Lives Cat Genome Sequencing Initiative to identify disease--causing variants in the domestic cat using WGS. The current cat reference assembly is efficient for gene and variant identification. However, as the feline variant database improves, development of cats as biomedical models for human disease will be more efficient, providing an alternative, large animal model for drug and gene therapy trials. Undiagnosed human patients with early-onset blindness should be screened for this AIPL1 variant. The HES7 variant should further calibrate the somite segmentation clock.
Collapse
Affiliation(s)
- Leslie A. Lyons
- />Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri - Columbia, E109 Vet Med Building, 1600 E. Rollins Street, Columbia, MO 65211 USA
| | - Erica K. Creighton
- />Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri - Columbia, E109 Vet Med Building, 1600 E. Rollins Street, Columbia, MO 65211 USA
| | - Hasan Alhaddad
- />College of Science, Kuwait University, Safat, 13060 Kuwait
| | | | - Robert A. Grahn
- />Veterinary Genetics Laboratory, School of Veterinary Medicine, University of California - Davis, Davis, CA 95616 USA
| | - HyungChul Rah
- />Graduate School of Health Science Business Convergence, College of Medicine, Chungbuk National University, Chongju, Chungbuk Province 28644 South Korea
| | - David J. Maggs
- />Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, Davis, CA 95616 USA
| | - Christopher R. Helps
- />Langford Veterinary Services, University of Bristol, Langford, Bristol, BS40 5DU UK
| | - Barbara Gandolfi
- />Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri - Columbia, E109 Vet Med Building, 1600 E. Rollins Street, Columbia, MO 65211 USA
| |
Collapse
|
42
|
Duda T, Pertzev A, Makino CL, Sharma RK. Bicarbonate and Ca(2+) Sensing Modulators Activate Photoreceptor ROS-GC1 Synergistically. Front Mol Neurosci 2016; 9:5. [PMID: 26858600 PMCID: PMC4729890 DOI: 10.3389/fnmol.2016.00005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 01/11/2016] [Indexed: 11/13/2022] Open
Abstract
Photoreceptor ROS-GC1, a prototype subfamily member of the membrane guanylate cyclase family, is a central component of phototransduction. It is a single transmembrane-spanning protein, composed of modular blocks. In rods, guanylate cyclase activating proteins (GCAPs) 1 and 2 bind to its juxtamembrane domain (JMD) and the C-terminal extension, respectively, to accelerate cyclic GMP synthesis when Ca(2+) levels are low. In cones, the additional expression of the Ca(2+)-dependent guanylate cyclase activating protein (CD-GCAP) S100B which binds to its C-terminal extension, supports acceleration of cyclic GMP synthesis at high Ca(2+) levels. Independent of Ca(2+), ROS-GC1 activity is also stimulated directly by bicarbonate binding to the core catalytic domain (CCD). Several enticing molecular features of this transduction system are revealed in the present study. In combination, bicarbonate and Ca(2+)-dependent modulators raised maximal ROS-GC activity to levels that exceeded the sum of their individual effects. The F(514)S mutation in ROS-GC1 that causes blindness in type 1 Leber's congenital amaurosis (LCA) severely reduced basal ROS-GC1 activity. GCAP2 and S100B Ca(2+) signaling modes remained functional, while the GCAP1-modulated mode was diminished. Bicarbonate nearly restored basal activity as well as GCAP2- and S100B-stimulated activities of the F(514)S mutant to normal levels but could not resurrect GCAP1 stimulation. We conclude that GCAP1 and GCAP2 forge distinct pathways through domain-specific modules of ROS-GC1 whereas the S100B and GCAP2 pathways may overlap. The synergistic interlinking of bicarbonate to GCAPs- and S100B-modulated pathways intensifies and tunes the dependence of cyclic GMP synthesis on intracellular Ca(2+). Our study challenges the recently proposed GCAP1 and GCAP2 "overlapping" phototransduction model (Peshenko et al., 2015b).
Collapse
Affiliation(s)
- Teresa Duda
- Research Divisions of Biochemistry and Molecular Biology, The Unit of Regulatory and Molecular Biology, Salus University Elkins Park, PA, USA
| | - Alexandre Pertzev
- Research Divisions of Biochemistry and Molecular Biology, The Unit of Regulatory and Molecular Biology, Salus University Elkins Park, PA, USA
| | - Clint L Makino
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary and Harvard Medical School Boston, MA, USA
| | - Rameshwar K Sharma
- Research Divisions of Biochemistry and Molecular Biology, The Unit of Regulatory and Molecular Biology, Salus University Elkins Park, PA, USA
| |
Collapse
|
43
|
A Mini-review: Animal Models of GUCY2D Leber Congenital Amaurosis (LCA1). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 854:253-8. [PMID: 26427419 DOI: 10.1007/978-3-319-17121-0_34] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
GUCY2D encodes retinal guanylate cylase-1 (retGC1), a protein that plays a pivotal role in the recovery phase of phototransduction. Mutations in GUCY2D are associated with a leading cause of recessive Leber congenital amaurosis (LCA1). Patients present within the first year of life with aberrant or unrecordable electroretinogram (ERG), nystagmus and a relatively normal fundus. Aside from abnormalities in the outer segments of foveal cones and, in some patients, foveal cone loss, LCA1 patients retain normal retinal laminar architecture suggesting they may be good candidates for gene replacement therapy. Several animal models of LCA1, both naturally occurring and engineered, have been characterized and provide valuable tools for translational studies. This mini-review will summarize the phenotypes of these models and describe how each has been instrumental in proof of concept studies to develop a gene replacement therapy for GUCY2D-LCA1.
Collapse
|
44
|
Giacalone JC, Wiley LA, Burnight ER, Songstad AE, Mullins RF, Stone EM, Tucker BA. Concise Review: Patient-Specific Stem Cells to Interrogate Inherited Eye Disease. Stem Cells Transl Med 2015; 5:132-40. [PMID: 26683869 PMCID: PMC4729558 DOI: 10.5966/sctm.2015-0206] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 10/16/2015] [Indexed: 12/13/2022] Open
Abstract
Heritable diseases of the retina are major causes of blindness worldwide. The recent success of gene augmentation trials for the treatment of RPE65-associated Leber congenital amaurosis has underscored the need for model systems that accurately recapitulate disease. How induced pluripotent stem cell technology is being used to confirm the pathogenesis of novel genetic variants, interrogate the pathophysiology of disease, and accelerate the development of patient-centered treatments is discussed. Whether we are driving to work or spending time with loved ones, we depend on our sense of vision to interact with the world around us. Therefore, it is understandable why blindness for many is feared above death itself. Heritable diseases of the retina, such as glaucoma, age-related macular degeneration, and retinitis pigmentosa, are major causes of blindness worldwide. The recent success of gene augmentation trials for the treatment of RPE65-associated Leber congenital amaurosis has underscored the need for model systems that accurately recapitulate disease. With the advent of patient-specific induced pluripotent stem cells (iPSCs), researchers are now able to obtain disease-specific cell types that would otherwise be unavailable for molecular analysis. In the present review, we discuss how the iPSC technology is being used to confirm the pathogenesis of novel genetic variants, interrogate the pathophysiology of disease, and accelerate the development of patient-centered treatments. Significance Stem cell technology has created the opportunity to advance treatments for multiple forms of blindness. Researchers are now able to use a person’s cells to generate tissues found in the eye. This technology can be used to elucidate the genetic causes of disease and develop treatment strategies. In the present review, how stem cell technology is being used to interrogate the pathophysiology of eye disease and accelerate the development of patient-centered treatments is discussed.
Collapse
Affiliation(s)
- Joseph C Giacalone
- Stephen A. Wynn Institute for Vision Research, Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Luke A Wiley
- Stephen A. Wynn Institute for Vision Research, Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Erin R Burnight
- Stephen A. Wynn Institute for Vision Research, Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Allison E Songstad
- Stephen A. Wynn Institute for Vision Research, Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Robert F Mullins
- Stephen A. Wynn Institute for Vision Research, Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Edwin M Stone
- Stephen A. Wynn Institute for Vision Research, Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa, USA
| | - Budd A Tucker
- Stephen A. Wynn Institute for Vision Research, Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
45
|
Wang H, Wang X, Zou X, Xu S, Li H, Soens ZT, Wang K, Li Y, Dong F, Chen R, Sui R. Comprehensive Molecular Diagnosis of a Large Chinese Leber Congenital Amaurosis Cohort. Invest Ophthalmol Vis Sci 2015; 56:3642-55. [PMID: 26047050 DOI: 10.1167/iovs.14-15972] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Leber congenital amaurosis (LCA) is an inherited retinal disease that causes early-onset severe visual impairment. To evaluate the mutation spectrum in the Chinese population, we performed a mutation screen in 145 Chinese LCA families. METHODS First, we performed direct Sanger sequencing of 7 LCA disease genes in 81 LCA families. Next, we developed a capture panel that enriches the entire coding exons and splicing sites of 163 known retinal disease genes and other candidate retinal disease genes. The capture panel allowed us to quickly identify disease-causing mutations in a large number of genes at a relatively low cost. Thus, this method was applied to the 53 LCA families that were unsolved by direct Sanger sequencing of 7 LCA disease genes and an additional 64 LCA families. Systematic next-generation sequencing (NGS) data analysis, Sanger sequencing validation, and segregation analysis were used to identify pathogenic mutations. RESULTS Homozygous or compound heterozygous mutations were identified in 107 families, heterozygous autosomal dominant mutations were identified in 3 families and an X-linked mutation was found in 1 family, for a combined solving rate of 76.6%. In total, 136 novel pathogenic mutations were found in this study. In combination with two previous studies carried out in Chinese LCA patients, we concluded that the mutation spectrum in the Chinese population is distinct compared to that in the European population. After revisiting, we also refined the clinical diagnosis of 10 families based on their molecular diagnosis. CONCLUSIONS Our results highlight the importance of a molecular diagnosis as an integral part of the clinical diagnostic process.
Collapse
Affiliation(s)
- Hui Wang
- Institute of Developmental and Regenerative Biology Hangzhou Normal University, Jianggan, Hangzhou, Zhejiang, China
| | - Xia Wang
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States 3Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| | - Xuan Zou
- Department of Ophthalmology, Peking Union Medical College Hospital, Peking Union Medical College, Dongcheng, Beijing, China
| | - Shan Xu
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States
| | - Hui Li
- Department of Ophthalmology, Peking Union Medical College Hospital, Peking Union Medical College, Dongcheng, Beijing, China
| | - Zachry Tore Soens
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| | - Keqing Wang
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States 3Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| | - Yumei Li
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States 3Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| | - Fangtian Dong
- Department of Ophthalmology, Peking Union Medical College Hospital, Peking Union Medical College, Dongcheng, Beijing, China
| | - Rui Chen
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States 3Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States 5Structural and Computational Biology & Molecular Biophysics
| | - Ruifang Sui
- Department of Ophthalmology, Peking Union Medical College Hospital, Peking Union Medical College, Dongcheng, Beijing, China
| |
Collapse
|
46
|
Zhong H, Eblimit A, Moayedi Y, Boye SL, Chiodo VA, Chen Y, Li Y, Nichols RM, Hauswirth WW, Chen R, Mardon G. AAV8(Y733F)-mediated gene therapy in a Spata7 knockout mouse model of Leber congenital amaurosis and retinitis pigmentosa. Gene Ther 2015; 22:619-27. [PMID: 25965394 DOI: 10.1038/gt.2015.42] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/14/2015] [Accepted: 03/23/2015] [Indexed: 12/27/2022]
Abstract
Loss of SPATA7 function causes the pathogenesis of Leber congenital amaurosis and retinitis pigmentosa. Spata7 knockout mice mimic human SPATA7-related retinal disease with apparent photoreceptor degeneration observed as early as postnatal day 15 (P15). To test the efficacy of adeno-associated virus (AAV)-mediated gene therapy for rescue of photoreceptor survival and function in Spata7 mutant mice, we employed the AAV8(Y733F) vector carrying hGRK1-driven full-length FLAG-tagged Spata7 cDNA to target both rod and cone photoreceptors. Following subretinal injection of this vector, FLAG-tagged SPATA7 was found to colocalize with endogenous SPATA7 in wild-type mice. In Spata7 mutant mice initially treated at P15, we observed improvement of photoresponse, photoreceptor ultrastructure and significant alleviation of photoreceptor degeneration. Furthermore, we performed treatments at P28 and P56 and found that all treatments (P15-P56) can ameliorate rod and cone loss in the long term (1 year); however, none efficiently protect photoreceptors from degeneration by 86 weeks of age as only a small amount of treated photoreceptors can survive to this time. This study demonstrates long-term improvement of photoreceptor function by AAV8(Y733F)-introduced Spata7 expression in a mouse model as potential treatment of the human disease, but also suggests that treated mutant photoreceptors still undergo progressive degeneration.
Collapse
Affiliation(s)
- H Zhong
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - A Eblimit
- 1] HGSC, Baylor College of Medicine, Houston, TX, USA [2] Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Y Moayedi
- 1] Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA [2] Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - S L Boye
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - V A Chiodo
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Y Chen
- 1] HGSC, Baylor College of Medicine, Houston, TX, USA [2] Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Y Li
- 1] HGSC, Baylor College of Medicine, Houston, TX, USA [2] Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - R M Nichols
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - W W Hauswirth
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - R Chen
- 1] HGSC, Baylor College of Medicine, Houston, TX, USA [2] Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA [3] Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
| | - G Mardon
- 1] Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA [2] Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA [3] Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA [4] Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA [5] Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
47
|
Pierce EA, Bennett J. The Status of RPE65 Gene Therapy Trials: Safety and Efficacy. Cold Spring Harb Perspect Med 2015; 5:a017285. [PMID: 25635059 DOI: 10.1101/cshperspect.a017285] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Several groups have reported the results of clinical trials of gene augmentation therapy for Leber congenital amaurosis (LCA) because of mutations in the RPE65 gene. These studies have used subretinal injection of adeno-associated virus (AAV) vectors to deliver the human RPE65 cDNA to the retinal pigment epithelial (RPE) cells of the treated eyes. In all of the studies reported to date, this approach has been shown to be both safe and effective. The successful clinical trials of gene augmentation therapy for retinal degeneration caused by mutations in the RPE65 gene sets the stage for broad application of gene therapy to treat retinal degenerative disorders.
Collapse
Affiliation(s)
- Eric A Pierce
- Department of Ophthalmology, Ocular Genomics Institute, Massachusetts Eye and Ear Infirmary and Harvard Medical School, Boston, Massachusetts 02114
| | - Jean Bennett
- Department of Ophthalmology, Center for Advanced Retinal and Ophthalmic Therapeutics, F.M. Kirby Center for Molecular Ophthalmology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104
| |
Collapse
|
48
|
Rakoczy EP, Narfström K. Gene therapy for eye as regenerative medicine? Lessons from RPE65 gene therapy for Leber's Congenital Amaurosis. Int J Biochem Cell Biol 2014; 56:153-7. [PMID: 25286304 DOI: 10.1016/j.biocel.2014.09.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 09/08/2014] [Accepted: 09/25/2014] [Indexed: 11/15/2022]
Abstract
Recombinant virus mediated gene therapy of Leber's Congenital Amaurosis has provided a wide range of data on the utility of gene replacement therapy for recessive diseases. Studies to date demonstrate that gene therapy in the eye is safe and can result in long-term recovery of visual function, but they also highlight that further research is required to identify optimum intervention time-points, target populations and the compatibility of associate therapies. This article is part of a directed issue entitled: Regenerative Medicine: the challenge of translation.
Collapse
Affiliation(s)
- Elizabeth P Rakoczy
- Centre for Ophthalmology and Visual Sciences, The University of Western Australia, Crawley, 6009, Western Australia, Australia.
| | - Kristina Narfström
- College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
49
|
Abstract
Leber congenital amaurosis (LCA) is a clinically and genetically heterogeneous group of diseases that account for the most severe form of early-onset retinal dystrophy. Mutations in retinal guanylate cyclase-1 (GUCY2D) are associated with LCA1, a prevalent form. GUCY2D encodes guanylate cyclase-1 (GC1), a protein expressed in rod and cone photoreceptors that regulates cGMP and Ca(2+) levels within these cells. LCA1 patients present with severely impaired vision, reduced, or ablated electroretinogram and nystagmus. Despite a high degree of visual disturbance, LCA1 patients retain normal photoreceptor laminar architecture, except for foveal cone outer segment abnormalities and, in some patients, foveal cone loss. This article will summarize clinical characterization of patients and proof of concept gene replacement studies in several animal models of GC1 deficiency, both of which have laid the groundwork for clinical application of a gene therapy for treatment of LCA1.
Collapse
|
50
|
Boye SE. Insights gained from gene therapy in animal models of retGC1 deficiency. Front Mol Neurosci 2014; 7:43. [PMID: 24860425 PMCID: PMC4030156 DOI: 10.3389/fnmol.2014.00043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 04/26/2014] [Indexed: 12/29/2022] Open
Abstract
Vertebrate species possess two retinal guanylate cyclases (retGC1 and retGC2) and at least two guanylate cyclase activating proteins (GCAPs), GCAP1 and GCAP2. GCAPs function as Ca2+ sensors that regulate the activity of guanylate cyclases. Together, these proteins regulate cGMP and Ca2+ levels within the outer segments of rod and cone photoreceptors. Mutations in GUCY2D, the gene that encodes retGC1, are a leading cause of the most severe form of early onset retinal dystrophy, Leber congenital amaurosis (LCA1). These mutations, which reduce or abolish the ability of retGC1 to replenish cGMP in photoreceptors, are thought to lead to the biochemical equivalent of chronic light exposure in these cells. In spite of this, the majority of LCA1 patients retain normal photoreceptor laminar architecture aside from foveal cone outer segment abnormalities, suggesting they may be good candidates for gene replacement therapy. Work began in the 1980s to characterize multiple animal models of retGC1 deficiency. 34 years later, all models have been used in proof of concept gene replacement studies toward the goal of developing a therapy to treat GUCY2D-LCA1. Here we use the results of these studies as well as those of recent clinical studies to address specific questions relating to clinical application of a gene therapy for treatment of LCA1.
Collapse
Affiliation(s)
- Shannon E Boye
- Department of Ophthalmology, University of Florida Gainesville, FL, USA
| |
Collapse
|