1
|
Dufour VL, Aguirre GD. Canine models of inherited retinal diseases: from neglect to well-recognized translational value. Mamm Genome 2024:10.1007/s00335-024-10091-y. [PMID: 39739008 DOI: 10.1007/s00335-024-10091-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/05/2024] [Indexed: 01/02/2025]
Abstract
Large animal models of inherited retinal diseases, particularly dogs, have been extensively used over the past decades to study disease natural history and evaluate therapeutic interventions. Our group of investigators at the University of Pennsylvania, School of Veterinary Medicine, has played a pivotal role in characterizing several of these animal models, documenting the natural history of their diseases, developing gene therapies, and conducting proof-of-concept studies. Additionally, we have assessed the potential toxicity of these therapies for human clinical trials, contributing to the regulatory approval of voretigene neparvovec-rzyl (Luxturna®) by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for the treatment of patients with confirmed biallelic mutation-associated retinal dystrophy. In this review, we aim to summarize the clinical features of a subset of these diseases and reflect on the challenges encountered in integrating canine models into the translational pipeline.
Collapse
Affiliation(s)
- Valérie L Dufour
- Division of Experimental Retinal Therapies, Department of Clinical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Gustavo D Aguirre
- Division of Experimental Retinal Therapies, Department of Clinical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Anderson G, Borooah S, Megaw R, Bagnaninchi P, Weller R, McLeod A, Dhillon B. UVR and RPE - The Good, the Bad and the degenerate Macula. Prog Retin Eye Res 2024; 100:101233. [PMID: 38135244 DOI: 10.1016/j.preteyeres.2023.101233] [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: 01/22/2023] [Revised: 12/15/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023]
Abstract
Ultraviolet Radiation (UVR) has a well-established causative influence within the aetiology of conditions of the skin and the anterior segment of the eye. However, a grounded assessment of the role of UVR within conditions of the retina has been hampered by a historical lack of quantitative, and spectrally resolved, assessment of how UVR impacts upon the retina in terms congruent with contemporary theories of ageing. In this review, we sought to summarise the key findings of research investigating the connection between UVR exposure in retinal cytopathology while identifying necessary avenues for future research which can deliver a deeper understanding of UVR's place within the retinal risk landscape.
Collapse
Affiliation(s)
- Graham Anderson
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh BioQuarter, EH16 4UU, UK
| | - Shyamanga Borooah
- Viterbi Family Department of Ophthalmology, Shiley Eye Institute, UC San Diego, CA, 92093-0946, USA
| | - Roly Megaw
- Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, EH4 2XU, UK; Department of Clinical Ophthalmology, National Health Service Scotland, Edinburgh, EH3 9HA, UK
| | - Pierre Bagnaninchi
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh BioQuarter, EH16 4UU, UK; Robert O Curle Eyelab, Instute for Regeneration and Repair, Edinburgh BioQuarter, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Richard Weller
- Centre for Inflammation Research, University of Edinburgh, Edinburgh BioQuarter, EH16 4TJ, UK
| | - Andrew McLeod
- School of GeoSciences, University of Edinburgh, Crew Building, King's Buildings, EH9 3FF, UK
| | - Baljean Dhillon
- Department of Clinical Ophthalmology, National Health Service Scotland, Edinburgh, EH3 9HA, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh BioQuarter, EH16 4SB, UK; Robert O Curle Eyelab, Instute for Regeneration and Repair, Edinburgh BioQuarter, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK.
| |
Collapse
|
3
|
Petersen-Jones SM, Komáromy AM. Canine and Feline Models of Inherited Retinal Diseases. Cold Spring Harb Perspect Med 2024; 14:a041286. [PMID: 37217283 PMCID: PMC10835616 DOI: 10.1101/cshperspect.a041286] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Naturally occurring inherited retinal diseases (IRDs) in cats and dogs provide a rich source of potential models for human IRDs. In many cases, the phenotypes between the species with mutations of the homologous genes are very similar. Both cats and dogs have a high-acuity retinal region, the area centralis, an equivalent to the human macula, with tightly packed photoreceptors and higher cone density. This and the similarity in globe size to that of humans means these large animal models provide information not obtainable from rodent models. The established cat and dog models include those for Leber congenital amaurosis, retinitis pigmentosa (including recessive, dominant, and X-linked forms), achromatopsia, Best disease, congenital stationary night blindness and other synaptic dysfunctions, RDH5-associated retinopathy, and Stargardt disease. Several of these models have proven to be important in the development of translational therapies such as gene-augmentation therapies. Advances have been made in editing the canine genome, which necessitated overcoming challenges presented by the specifics of canine reproduction. Feline genome editing presents fewer challenges. We can anticipate the generation of specific cat and dog IRD models by genome editing in the future.
Collapse
Affiliation(s)
- Simon M Petersen-Jones
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan 48824, USA
| | - András M Komáromy
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan 48824, USA
| |
Collapse
|
4
|
Taskin HO, Wivel J, Aguirre GD, Beltran WA, Aguirre GK. Cone-Driven, Geniculocortical Responses in Canine Models of Outer Retinal Disease. Transl Vis Sci Technol 2024; 13:18. [PMID: 38241039 PMCID: PMC10807495 DOI: 10.1167/tvst.13.1.18] [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: 09/25/2023] [Accepted: 12/20/2023] [Indexed: 01/23/2024] Open
Abstract
Purpose Canine models of inherited retinal degeneration are used for proof of concept of emerging gene and cell-based therapies that aim to produce functional restoration of cone-mediated vision. We examined functional magnetic resonance imaging (MRI) measures of the postretinal response to cone-directed stimulation in wild-type (WT) dogs, and in three different retinal disease models. Methods Temporal spectral modulation of a uniform field of light around a photopic background was used to target the canine L/M (hereafter "L") and S cones and rods. Stimuli were designed to separately target the postreceptoral luminance (L+S) and chrominance (L-S) pathways, the rods, and all photoreceptors jointly (light flux). These stimuli were presented to WT, and mutant PDE6B-RCD1, RPGR-XLPRA2, and NPHP5-CRD2 dogs during pupillometry and functional MRI (fMRI). Results Pupil responses in WT dogs to light flux, L+S, and rod-directed stimuli were consistent with responses being driven by cone signals alone. For WT animals, both luminance and chromatic (L-S) stimuli evoked fMRI responses in the lateral geniculate nucleus or visual cortex; RCD1 animals with predominant rod loss had similar responses. Responses to cone-directed stimulation were reduced in XLPRA2 and absent in CRD2. NPHP5 gene augmentation restored the cortical response to luminance stimulation in a CRD2 animal. Conclusions Cone-directed stimulation during fMRI can be used to measure the integrity of luminance and chrominance responses in the dog visual system. The NPHP5-CRD2 model is appealing for studies of recovered cone function. Translational Relevance fMRI assessment of cone-driven cortical response provides a tool to translate cell/gene therapies for vision restoration.
Collapse
Affiliation(s)
- Huseyin O. Taskin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jacqueline Wivel
- Division of Experimental Retinal Therapies, Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gustavo D. Aguirre
- Division of Experimental Retinal Therapies, Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - William A. Beltran
- Division of Experimental Retinal Therapies, Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Geoffrey K. Aguirre
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
5
|
Taskin HO, Wivel J, Aguirre GD, Beltran WA, Aguirre GK. Cone-driven, geniculo-cortical responses in canine models of outer retinal disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.13.571523. [PMID: 38168165 PMCID: PMC10760074 DOI: 10.1101/2023.12.13.571523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Purpose Canine models of inherited retinal degeneration are used for proof-of-concept of emerging gene and cell-based therapies that aim to produce functional restoration of cone-mediated vision. We examined functional MRI measures of the post-retinal response to cone-directed stimulation in wild type (WT) dogs, and in three different retinal disease models. Methods Temporal spectral modulation of a uniform field of light around a photopic background was used to target the canine L/M (hereafter "L") and S cones and rods. Stimuli were designed to separately target the post-receptoral luminance (L+S) and chrominance (L-S) pathways, the rods, and all photoreceptors jointly (light flux). These stimuli were presented to WT, and mutant PDE6B-RCD1, RPGR-XLPRA2, and NPHP5-CRD2 dogs during pupillometry and fMRI. Results Pupil responses in WT dogs to light flux, L+S, and rod-directed stimuli were consistent with responses being driven by cone signals alone. For WT animals, both luminance and chromatic (L-S) stimuli evoked fMRI responses in the lateral geniculate nucleus (LGN) or visual cortex; RCD1 animals with predominant rod loss had similar responses. Responses to cone-directed stimulation were reduced in XLPRA2 and absent in CRD2. NPHP5 gene augmentation restored the cortical response to luminance stimulation in a CRD2 animal. Conclusions Cone-directed stimulation during fMRI can be used to measure the integrity of luminance and chrominance responses in the dog visual system. The NPHP5-CRD2 model is appealing for studies of recovered cone function. Translational Relevance fMRI assessment of cone driven cortical response provides a tool to translate cell/gene therapies for vision restoration.
Collapse
Affiliation(s)
- Huseyin O. Taskin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Jacqueline Wivel
- Division of Experimental Retinal Therapies, Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Gustavo D. Aguirre
- Division of Experimental Retinal Therapies, Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - William A. Beltran
- Division of Experimental Retinal Therapies, Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Geoffrey K. Aguirre
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
6
|
Leinonen H, Fu Z, Bull E. Neural and Müller glial adaptation of the retina to photoreceptor degeneration. Neural Regen Res 2023; 18:701-707. [DOI: 10.4103/1673-5374.354511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
7
|
Ripolles-Garcia A, Chen Y, Sato Y, Gray A, Ying GS, Aguirre GD, Beltran WA. Retinal Vascular Plexuses Are Unequally Affected in Canine Inherited Retinal Degenerations. Invest Ophthalmol Vis Sci 2022; 63:22. [PMID: 36378130 PMCID: PMC9672900 DOI: 10.1167/iovs.63.12.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Purpose To characterize the progression of vascular changes that occur in each retinal plexus, in three canine models of inherited retinal degeneration. Methods In this retrospective cohort study, we examined the retinal imaging records of 44 dogs from a research colony that had undergone optical coherence tomography angiography (OCTA) imaging. Animals enrolled included crd2/NPHP5 and xlpra2/RPGR mutant dogs imaged at different stages of photoreceptor loss, as well as RHOT4R/+ dogs after acute light-induced rod degeneration. Also included were normal controls imaged at similar ages. OCT angiograms of the superficial vascular plexus combined with the intermediate capillary plexus (SVP + ICP), and the deep capillary plexus (DCP) were analyzed using the AngioTool software to calculate vessel density and other vascular parameters. Results A reduction in vessel density was seen over time in both the SVP + ICP and DCP in all mutant dogs but was more pronounced in the DCP. Scans were subclassified based on outer nuclear layer (ONL) thinning compared to age-matched normal controls. When ONL loss was 0% to 50%, vessel density in the DCP was significantly lower than in age-matched controls. In all cases, when ONL loss exceeded 87.5%, vessel density in the SVP + ICP was significantly reduced as well. In the acute light-induced rod degeneration model, the vascular regression changes were observed mainly in the DCP. Conclusions Vessel density reduction in dogs undergoing retinal degeneration is first detected by OCTA in the DCP, and only at later stages in the SVP + ICP.
Collapse
Affiliation(s)
- Ana Ripolles-Garcia
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Yineng Chen
- Department of Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Yu Sato
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Alexa Gray
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Gui-Shuang Ying
- Department of Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Gustavo D. Aguirre
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - William A. Beltran
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| |
Collapse
|
8
|
Mischi E, Soukup P, Harman CD, Oikawa K, Kowalska ME, Hartnack S, McLellan GJ, Komáromy AM, Pot SA. Outer retinal thickness and visibility of the choriocapillaris in four distinct retinal regions imaged with spectral domain optical coherence tomography in dogs and cats. Vet Ophthalmol 2022; 25 Suppl 1:122-135. [PMID: 35611616 PMCID: PMC9246961 DOI: 10.1111/vop.12989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 04/04/2022] [Accepted: 04/04/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE To evaluate the outer retinal band thickness and choriocapillaris (CC) visibility in four distinct retinal regions in dogs and cats imaged with spectral domain optical coherence tomography (SD-OCT). To attempt delineation of a fovea-like region in canine and feline SD-OCT scans, aided by the identification of outer retinal thickness differences between retinal regions. METHODS Spectralis® HRA + OCT SD-OCT scans from healthy, anesthetized dogs (n = 10) and cats (n = 12) were analyzed. Scanlines on which the CC was identifiable were counted and CC visibility was scored. Outer nuclear layer (ONL) thickness and the distances from external limiting membrane (ELM) to retinal pigment epithelium/Bruch's membrane complex (RPE/BM) and ELM to CC were measured in the area centralis (AC), a visually identified fovea-like region, and in regions superior and inferior to the optic nerve head (ONH). Measurements were analyzed using a multilevel regression. RESULTS The CC was visible in over 90% of scanlines from dogs and cats. The ONL was consistently thinnest in the fovea-like region. The outer retina (ELM-RPE and ELM-CC) was thickest within the AC compared with superior and inferior to the ONH in dogs and cats (p < .001 for all comparisons). CONCLUSIONS The CC appears a valid, albeit less than ideal outer retinal boundary marker in tapetal species. The AC can be objectively differentiated from the surrounding retina on SD-OCT images of dogs and cats; a fovea-like region was identified in dogs and its presence was suggested in cats. These findings allow targeted imaging and image evaluation of these regions of retinal specialization.
Collapse
Affiliation(s)
- Elisa Mischi
- Ophthalmology Section, Equine Department, Vetsuisse FacultyUniversity of ZurichZurichSwitzerland
| | - Petr Soukup
- Ophthalmology Section, Equine Department, Vetsuisse FacultyUniversity of ZurichZurichSwitzerland
| | - Christine D. Harman
- Department of Small Animal Clinical Sciences, College of Veterinary MedicineMichigan State UniversityEast LansingMichiganUSA
| | - Kazuya Oikawa
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Malwina E. Kowalska
- Ophthalmology Section, Equine Department, Vetsuisse FacultyUniversity of ZurichZurichSwitzerland
- Section of Epidemiology, Vetsuisse FacultyUniversity of ZurichZurichSwitzerland
| | - Sonja Hartnack
- Section of Epidemiology, Vetsuisse FacultyUniversity of ZurichZurichSwitzerland
| | - Gillian J. McLellan
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - András M. Komáromy
- Department of Small Animal Clinical Sciences, College of Veterinary MedicineMichigan State UniversityEast LansingMichiganUSA
| | - Simon A. Pot
- Ophthalmology Section, Equine Department, Vetsuisse FacultyUniversity of ZurichZurichSwitzerland
| |
Collapse
|
9
|
Graham KL, Johnson PJ, Barry EF, Pérez Orrico M, Soligo DJ, Lawlor M, White A. Diffusion tensor imaging of the visual pathway in dogs with primary angle-closure glaucoma. Vet Ophthalmol 2020; 24 Suppl 1:63-74. [PMID: 32990378 DOI: 10.1111/vop.12824] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/09/2020] [Accepted: 08/14/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To describe measurements of in vivo structures of the visual pathway beyond the retina and optic nerve head associated with canine primary angle-closure glaucoma (PACG). METHODS A prospective pilot study was conducted using magnetic resonance diffusion tensor imaging (DTI) to obtain quantitative measures of the optic nerve, chiasm, tract, and lateral geniculate nucleus (LGN) in dogs with and without PACG. 3-Tesla DTI was performed on six affected dogs and five breed, age- and sex-matched controls. DTI indices of the optic nerve, optic chiasm, optic tracts, and LGN were compared between normal, unilateral PACG, and bilateral PACG groups. Intra-class correlation coefficient (ICC) was calculated to assess intra-observer reliability. RESULTS Quantitative measurements of the optic nerve, optic tract, optic chiasm, and LGN were obtained in all dogs. There was a trend for reduced fractional anisotropy (FA) associated with disease for all structures assessed. Compared to the same structure in normal dogs, FA, and radial diffusivity (RD) of the optic nerve was consistently higher in the unaffected eye in dogs with unilateral PACG. Intra-observer reliability was excellent for measurements of the optic nerve (ICC: 0.92), good for measurements of the optic tract (ICC: 0.89) and acceptable for measures of the optic chiasm (ICC: 0.71) and lateral geniculate nuclei (ICC: 0.76). CONCLUSION Diffusivity and anisotropy measures provide a quantifiable means to evaluate the visual pathway in dogs. DTI has potential to provide in vivo measures of axonal and myelin injury and transsynaptic degeneration in canine PACG.
Collapse
Affiliation(s)
- Kathleen L Graham
- Clinical Ophthalmology and Eye Health, University of Sydney, Sydney, NSW, Australia
| | | | - Erica F Barry
- Cornell College of Veterinary Medicine, Ithaca, NY, USA
| | | | | | - Mitchell Lawlor
- Clinical Ophthalmology and Eye Health, University of Sydney, Sydney, NSW, Australia
| | - Andrew White
- Clinical Ophthalmology and Eye Health, University of Sydney, Sydney, NSW, Australia.,Westmead Institute for Medical Research, Westmead, NSW, Australia
| |
Collapse
|
10
|
Response to Comment on: Sector retinitis pigmentosa caused by mutations of the RHO gene. Eye (Lond) 2020; 34:1476. [PMID: 31659285 PMCID: PMC7470851 DOI: 10.1038/s41433-019-0649-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 09/24/2019] [Indexed: 11/13/2022] Open
|
11
|
Ramachandra Rao S, Skelton LA, Wu F, Onysk A, Spolnik G, Danikiewicz W, Butler MC, Stacks DA, Surmacz L, Mu X, Swiezewska E, Pittler SJ, Fliesler SJ. Retinal Degeneration Caused by Rod-Specific Dhdds Ablation Occurs without Concomitant Inhibition of Protein N-Glycosylation. iScience 2020; 23:101198. [PMID: 32526701 PMCID: PMC7287266 DOI: 10.1016/j.isci.2020.101198] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/10/2020] [Accepted: 05/20/2020] [Indexed: 12/16/2022] Open
Abstract
Dehydrodolichyl diphosphate synthase (DHDDS) catalyzes the committed step in dolichol synthesis. Recessive mutations in DHDDS cause retinitis pigmentosa (RP59), resulting in blindness. We hypothesized that rod photoreceptor-specific ablation of Dhdds would cause retinal degeneration due to diminished dolichol-dependent protein N-glycosylation. Dhddsflx/flx mice were crossed with rod-specific Cre recombinase-expressing (Rho-iCre75) mice to generate rod-specific Dhdds knockout mice (Dhddsflx/flx iCre+). In vivo morphological and electrophysiological evaluation of Dhddsflx/flx iCre+ retinas revealed mild retinal dysfunction at postnatal (PN) 4 weeks, compared with age-matched controls; however, rapid photoreceptor degeneration ensued, resulting in almost complete loss of rods and cones by PN 6 weeks. Retina dolichol levels were markedly decreased by PN 4 weeks in Dhddsflx/flx iCre+ mice, relative to controls; despite this, N-glycosylation of retinal proteins, including opsin (the dominant rod-specific glycoprotein), persisted in Dhddsflx/flx iCre+ mice. These findings challenge the conventional mechanistic view of RP59 as a congenital disorder of glycosylation. Deletion of Dhdds in rod cells caused rapid retinal degeneration in mice Retinal dolichol levels markedly decreased before onset of degeneration Protein N-glycosylation was uncompromised despite Dhdds deletion Degeneration also involved gliosis, microglial activation, and phagoptosis
Collapse
Affiliation(s)
- Sriganesh Ramachandra Rao
- Department of Ophthalmology/Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14209, USA; Research Service, VA Western NY Healthcare System, Buffalo, NY 142015, USA; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14203, USA
| | - Lara A Skelton
- Department of Ophthalmology/Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14209, USA; Research Service, VA Western NY Healthcare System, Buffalo, NY 142015, USA; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14203, USA
| | - Fuguo Wu
- Department of Ophthalmology/Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14209, USA; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14203, USA; New York State Center of Excellence in Bioinformatics and Life Sciences, State University of New York- University at Buffalo, Buffalo, NY 14203, USA
| | - Agnieszka Onysk
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Grzegorz Spolnik
- Institute of Organic Chemistry, Polish Academy of Sciences, Warsaw 02106, Poland
| | - Witold Danikiewicz
- Institute of Organic Chemistry, Polish Academy of Sciences, Warsaw 02106, Poland
| | - Mark C Butler
- Department of Ophthalmology/Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14209, USA; Research Service, VA Western NY Healthcare System, Buffalo, NY 142015, USA
| | - Delores A Stacks
- Department of Optometry and Vision Science, Vision Science Research Center, School of Optometry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Liliana Surmacz
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Xiuqian Mu
- Department of Ophthalmology/Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14209, USA; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14203, USA; New York State Center of Excellence in Bioinformatics and Life Sciences, State University of New York- University at Buffalo, Buffalo, NY 14203, USA
| | - Ewa Swiezewska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Steven J Pittler
- Department of Optometry and Vision Science, Vision Science Research Center, School of Optometry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Steven J Fliesler
- Department of Ophthalmology/Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14209, USA; Research Service, VA Western NY Healthcare System, Buffalo, NY 142015, USA; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14203, USA.
| |
Collapse
|
12
|
Talib M, Boon CJF. Retinal Dystrophies and the Road to Treatment: Clinical Requirements and Considerations. Asia Pac J Ophthalmol (Phila) 2020; 9:159-179. [PMID: 32511120 PMCID: PMC7299224 DOI: 10.1097/apo.0000000000000290] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/01/2020] [Indexed: 12/15/2022] Open
Abstract
: Retinal dystrophies (RDs) comprise relatively rare but devastating causes of progressive vision loss. They represent a spectrum of diseases with marked genetic and clinical heterogeneity. Mutations in the same gene may lead to different diagnoses, for example, retinitis pigmentosa or cone dystrophy. Conversely, mutations in different genes may lead to the same phenotype. The age at symptom onset, and the rate and characteristics of peripheral and central vision decline, may vary widely per disease group and even within families. For most RD cases, no effective treatment is currently available. However, preclinical studies and phase I/II/III gene therapy trials are ongoing for several RD subtypes, and recently the first retinal gene therapy has been approved by the US Food and Drug Administration for RPE65-associated RDs: voretigene neparvovec-rzyl (Luxturna). With the rapid advances in gene therapy studies, insight into the phenotypic spectrum and long-term disease course is crucial information for several RD types. The vast clinical heterogeneity presents another important challenge in the evaluation of potential efficacy in future treatment trials, and in establishing treatment candidacy criteria. This perspective describes these challenges, providing detailed clinical descriptions of several forms of RD that are caused by genes of interest for ongoing and future gene or cell-based therapy trials. Several ongoing and future treatment options will be described.
Collapse
Affiliation(s)
- Mays Talib
- Department of Ophthalmology, Leiden, The Netherlands
| | - Camiel J F Boon
- Department of Ophthalmology, Leiden, The Netherlands
- Department of Ophthalmology, Amsterdam UMC, Academic Medical Center, University of Amsterdam. Amsterdam, The Netherlands
| |
Collapse
|
13
|
Winkler PA, Occelli LM, Petersen-Jones SM. Large Animal Models of Inherited Retinal Degenerations: A Review. Cells 2020; 9:cells9040882. [PMID: 32260251 PMCID: PMC7226744 DOI: 10.3390/cells9040882] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
Studies utilizing large animal models of inherited retinal degeneration (IRD) have proven important in not only the development of translational therapeutic approaches, but also in improving our understanding of disease mechanisms. The dog is the predominant species utilized because spontaneous IRD is common in the canine pet population. Cats are also a source of spontaneous IRDs. Other large animal models with spontaneous IRDs include sheep, horses and non-human primates (NHP). The pig has also proven valuable due to the ease in which transgenic animals can be generated and work is ongoing to produce engineered models of other large animal species including NHP. These large animal models offer important advantages over the widely used laboratory rodent models. The globe size and dimensions more closely parallel those of humans and, most importantly, they have a retinal region of high cone density and denser photoreceptor packing for high acuity vision. Laboratory rodents lack such a retinal region and, as macular disease is a critical cause for vision loss in humans, having a comparable retinal region in model species is particularly important. This review will discuss several large animal models which have been used to study disease mechanisms relevant for the equivalent human IRD.
Collapse
|
14
|
Dufour VL, Cideciyan AV, Ye GJ, Song C, Timmers A, Habecker PL, Pan W, Weinstein NM, Swider M, Durham AC, Ying GS, Robinson PM, Jacobson SG, Knop DR, Chulay JD, Shearman MS, Aguirre GD, Beltran WA. Toxicity and Efficacy Evaluation of an Adeno-Associated Virus Vector Expressing Codon-Optimized RPGR Delivered by Subretinal Injection in a Canine Model of X-linked Retinitis Pigmentosa. Hum Gene Ther 2020; 31:253-267. [PMID: 31910043 DOI: 10.1089/hum.2019.297] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Applied Genetic Technologies Corporation (AGTC) is developing a recombinant adeno-associated virus (rAAV) vector AGTC-501, also designated rAAV2tYF-GRK1-hRPGRco, to treat X-linked retinitis pigmentosa (XLRP) in patients with mutations in the retinitis pigmentosa GTPase regulator (RPGR) gene. The vector contains a codon-optimized human RPGR cDNA (hRPGRco) driven by a photoreceptor-specific promoter (G protein-coupled receptor kinase 1 [GRK1]), and is packaged in an AAV2 capsid variant with three surface tyrosine residues changed to phenylalanine (AAV2tYF). We conducted a toxicity and efficacy study of this vector administered by subretinal injection in the naturally occurring RPGR mutant (X-linked progressive retinal atrophy 2 [XLPRA2]) dog model. Sixteen RPGR mutant dogs divided into four groups of three to five animals each received either a subretinal injection of 0.07 mL of AGTC-501 at low (1.2 × 1011 vector genome [vg]/mL), mid (6 × 1011 vg/mL), or high dose (3 × 1012 vg/mL), or of vehicle control in the right eye at early-stage disease. The left eye remained untreated. Subretinal injections were well tolerated and were not associated with systemic toxicity. Electroretinography, in vivo retinal imaging, and histological analysis showed rescue of photoreceptor function and structure in the absence of ocular toxicity in the low- and mid-dose treatment groups when compared with the vehicle-treated group. The high-dose group showed evidence of both photoreceptor rescue and posterior segment toxicity. These results support the use of AGTC-501 in clinical studies with patients affected with XLRP caused by RPGR mutations and define the no-observed-adverse-effect level at 6 × 1011 vg/mL.
Collapse
Affiliation(s)
- Valérie L Dufour
- Division of Experimental Retinal Therapies, Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Artur V Cideciyan
- Department of Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Guo-Jie Ye
- Applied Genetic Technologies Corporation, Alachua, Florida
| | - Chunjuan Song
- Applied Genetic Technologies Corporation, Alachua, Florida
| | - Adrian Timmers
- Applied Genetic Technologies Corporation, Alachua, Florida
| | - Perry L Habecker
- Department of Pathobiology, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, Pennsylvania
| | - Wei Pan
- Department of Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nicole M Weinstein
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Malgorzata Swider
- Department of Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Amy C Durham
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gui-Shuang Ying
- Department of Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Samuel G Jacobson
- Department of Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David R Knop
- Applied Genetic Technologies Corporation, Alachua, Florida
| | | | | | - Gustavo D Aguirre
- Division of Experimental Retinal Therapies, Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - William A Beltran
- Division of Experimental Retinal Therapies, Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
15
|
Orlans HO, Merrill J, Barnard AR, Charbel Issa P, Peirson SN, MacLaren RE. Filtration of Short-Wavelength Light Provides Therapeutic Benefit in Retinitis Pigmentosa Caused by a Common Rhodopsin Mutation. Invest Ophthalmol Vis Sci 2019; 60:2733-2742. [PMID: 31247114 DOI: 10.1167/iovs.19-26964] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The role of light exposure in accelerating retinitis pigmentosa (RP) remains controversial. Faster degeneration has however been observed in the inferior than superior retina in several forms ("sector" RP), including those caused by the rhodopsin P23H mutation, suggesting a modifying role of incident light exposure in such cases. Rearing of equivalent animal models in complete darkness has been shown to slow the degeneration. Here we investigate the use of red filters as a potential treatment strategy, with the hypothesis that minimizing retinal exposure to light <600 nm to which rods are maximally sensitive may provide therapeutic benefit. Methods Knockin mice heterozygous for the P23H dominant rhodopsin mutation (RhoP23H/+) housed in red-tinted plastic cages were divided at weaning into either untinted or red-tinted cages. Subsequently, photoreceptor layer (PRL) thickness was measured by spectral-domain ocular coherence tomography, retinal function quantified by ERG, and cone morphology determined by immunohistochemical analysis (IHC) of retinal flatmounts. Results Mice remaining in red-tinted cages had a significantly greater PRL thickness than those housed in untinted cages at all time points. Red housing also led to a highly significant rescue of retinal function as determined by both dark- and light-adapted ERG responses. IHC further revealed a dramatic benefit on cone morphology and number in the red- as compared with the clear-housed group. Conclusions Limitation of short-wavelength light exposure significantly slows degeneration in the RhoP23H/+ mouse model. Red filters may represent a cost-effective and low-risk treatment for patients with rod-cone dystrophy in whom a sectoral phenotype is noted.
Collapse
Affiliation(s)
- Harry O Orlans
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford, United Kingdom.,Western Eye Hospital, London, United Kingdom
| | - Jonathon Merrill
- Biomedical Services, University of Oxford, Oxford, United Kingdom
| | - Alun R Barnard
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford, United Kingdom
| | - Peter Charbel Issa
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford, United Kingdom.,Oxford Eye Hospital, Oxford, United Kingdom
| | - Stuart N Peirson
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford, United Kingdom
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford, United Kingdom.,Oxford Eye Hospital, Oxford, United Kingdom
| |
Collapse
|
16
|
Orlans HO, MacLaren RE. Comment on: 'Sector retinitis pigmentosa caused by mutations of the RHO gene'. Eye (Lond) 2019; 34:1477-1478. [PMID: 31659286 DOI: 10.1038/s41433-019-0648-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 09/24/2019] [Indexed: 12/20/2022] Open
Affiliation(s)
- Harry O Orlans
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences & NIHR Oxford, Oxford, UK. .,Moorfields Eye Hospital NHS Foundation Trust, London, UK.
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences & NIHR Oxford, Oxford, UK.,Moorfields Eye Hospital NHS Foundation Trust, London, UK.,Oxford Eye Hospital, Oxford, UK
| |
Collapse
|
17
|
Lessons learned from quantitative fundus autofluorescence. Prog Retin Eye Res 2019; 74:100774. [PMID: 31472235 DOI: 10.1016/j.preteyeres.2019.100774] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/21/2019] [Accepted: 08/25/2019] [Indexed: 12/12/2022]
Abstract
Quantitative fundus autofluorescence (qAF) is an approach that is built on a confocal scanning laser platform and used to measure the intensity of the inherent autofluorescence of retina elicited by short-wavelength (488 nm) excitation. Being non-invasive, qAF does not interrupt tissue architecture, thus allowing for structural correlations. The spectral features, cellular origin and topographic distribution of the natural autofluorescence of the fundus indicate that it is emitted from retinaldehyde-adducts that form in photoreceptor cells and accumulate, under most conditions, in retinal pigment epithelial cells. The distributions and intensities of fundus autofluorescence deviate from normal in many retinal disorders and it is widely recognized that these changing patterns can aid in the diagnosis and monitoring of retinal disease. The standardized protocol employed by qAF involves the normalization of fundus grey levels to a fluorescent reference installed in the imaging instrument. Together with corrections for magnification and anterior media absorption, this approach facilitates comparisons with serial images and images acquired within groups of patients. Here we provide a comprehensive summary of the principles and practice of qAF and we highlight recent efforts to elucidate retinal disease processes by combining qAF with multi-modal imaging.
Collapse
|
18
|
Graham KL, McCowan CI, Caruso K, Billson FM, Whittaker CJG, White A. Optical coherence tomography of the retina, nerve fiber layer, and optic nerve head in dogs with glaucoma. Vet Ophthalmol 2019; 23:97-112. [PMID: 31297979 DOI: 10.1111/vop.12694] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 06/13/2019] [Accepted: 06/13/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To evaluate the retina and optic nerve head (ONH) in canine eyes predisposed to glaucoma using optical coherence tomography (OCT). ANIMALS Twenty-five eyes (24 dogs). METHODS Measures of peripapillary retinal, retinal nerve fiber layer (RNFL), and ganglion cell complex (GCC) thickness and ONH parameters were obtained in vivo by OCT of the unaffected eye in dogs diagnosed with unilateral primary glaucoma (predisposed; n = 12) and compared with measures of healthy control eyes (normal; n = 13). Repeatability and intrarater reliability were explored using intraclass correlation coefficients (ICC). RESULTS Compared to normal eyes, predisposed eyes had a thinner retina in the temporal (P = 0.005), inferior quadrants (P = 0.003), and decreased inner retinal thickness (superior: P = 0.003, temporal: P = 0.001, inferior: P < 0.001, nasal: P = 0.001). Predisposed eyes had a thinner RNFL compared to normal eyes (P = 0.005), and when analyzed in quadrants, it was thinner in the superior (P < 0.001), temporal (P = 0.034), and nasal quadrants (P = 0.001). Repeatability (ICC 0.763-0.835) and intrarater reliability (ICC 0.824-0.942) were good to excellent for measures of retinal thickness and adequate for RNFL measurements (ICC 0.701-0.798). Reliable measurements of optic disk area were obtained and were similar between groups (P = 0.597). Measurements of parameters relying on automated software detection (GCC, optic cup, optic rim) had inadequate repeatability and reliability. CONCLUSION Statistically significant differences in retinal and RNFL thicknesses were identified in normal and predisposed eyes. Reliable and consistent measurements of variables with manual adjustment of software detected parameters were obtained. Validation of OCT as a diagnostic tool for clinical assessment in canine glaucoma is warranted.
Collapse
Affiliation(s)
- Kathleen L Graham
- Clinical Ophthalmology and Eye Health, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Christina I McCowan
- University of Melbourne Veterinary Hospital, University of Melbourne, Melbourne, Victoria, Australia.,Department of Economic Development, Jobs, Transport and Resources, Melbourne, Victoria, Australia
| | - Kelly Caruso
- Eye Clinic for Animals, Crows Nest, New South Wales, Australia
| | - F Mark Billson
- Small Animal Specialist Hospital, North Ryde, New South Wales, Australia
| | | | - Andrew White
- Clinical Ophthalmology and Eye Health, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia.,Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| |
Collapse
|
19
|
Mitchell J, Balem F, Tirupula K, Man D, Dhiman HK, Yanamala N, Ollesch J, Planas-Iglesias J, Jennings BJ, Gerwert K, Iannaccone A, Klein-Seetharaman J. Comparison of the molecular properties of retinitis pigmentosa P23H and N15S amino acid replacements in rhodopsin. PLoS One 2019; 14:e0214639. [PMID: 31100078 PMCID: PMC6524802 DOI: 10.1371/journal.pone.0214639] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 03/19/2019] [Indexed: 12/16/2022] Open
Abstract
Mutations in the RHO gene encoding for the visual pigment protein, rhodopsin, are among the most common cause of autosomal dominant retinitis pigmentosa (ADRP). Previous studies of ADRP mutations in different domains of rhodopsin have indicated that changes that lead to more instability in rhodopsin structure are responsible for more severe disease in patients. Here, we further test this hypothesis by comparing side-by-side and therefore quantitatively two RHO mutations, N15S and P23H, both located in the N-terminal intradiscal domain. The in vitro biochemical properties of these two rhodopsin proteins, expressed in stably transfected tetracycline-inducible HEK293S cells, their UV-visible absorption, their Fourier transform infrared, circular dichroism and Metarhodopsin II fluorescence spectroscopy properties were characterized. As compared to the severely impaired P23H molecular function, N15S is only slightly defective in structure and stability. We propose that the molecular basis for these structural differences lies in the greater distance of the N15 residue as compared to P23 with respect to the predicted rhodopsin folding core. As described previously for WT rhodopsin, addition of the cytoplasmic allosteric modulator chlorin e6 stabilizes especially the P23H protein, suggesting that chlorin e6 may be generally beneficial in the rescue of those ADRP rhodopsin proteins whose stability is affected by amino acid replacement.
Collapse
Affiliation(s)
- James Mitchell
- Division of Biomedical Sciences, Medical School, University of Warwick, Coventry, United Kingdom
| | - Fernanda Balem
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Kalyan Tirupula
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - David Man
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Harpreet Kaur Dhiman
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Naveena Yanamala
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Julian Ollesch
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Joan Planas-Iglesias
- Division of Biomedical Sciences, Medical School, University of Warwick, Coventry, United Kingdom
| | - Barbara J Jennings
- Retinal Degeneration & Ophthalmic Genetics Service & Lions Visual Function Diagnostic Lab, Hamilton Eye Institute, Dept. Ophthalmology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Klaus Gerwert
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Alessandro Iannaccone
- Retinal Degeneration & Ophthalmic Genetics Service & Lions Visual Function Diagnostic Lab, Hamilton Eye Institute, Dept. Ophthalmology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Judith Klein-Seetharaman
- Division of Biomedical Sciences, Medical School, University of Warwick, Coventry, United Kingdom
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
20
|
Natural models for retinitis pigmentosa: progressive retinal atrophy in dog breeds. Hum Genet 2019; 138:441-453. [PMID: 30904946 DOI: 10.1007/s00439-019-01999-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/14/2019] [Indexed: 01/24/2023]
Abstract
Retinitis pigmentosa (RP) is a heterogeneous group of inherited retinal disorders eventually leading to blindness with different ages of onset, progression and severity. Human RP, first characterized by the progressive degeneration of rod photoreceptor cells, shows high genetic heterogeneity with more than 90 genes identified. However, about one-third of patients have no known genetic causes. Interestingly, dogs are also severely affected by similar diseases, called progressive retinal atrophy (PRA). Indeed, RP and PRA have comparable clinical signs, physiopathology and outcomes, similar diagnosis methods and most often, orthologous genes are involved. The many different dog PRAs often segregate in specific breeds. Indeed, undesired alleles have been selected and amplified through drastic selection and excessive use of inbreeding. Out of the 400 breeds, nearly 100 have an inherited form of PRA, which are natural animal models that can be used to investigate the genetics, disease progression and therapies in dogs for the benefit of both dogs and humans. Recent knowledge on the canine genome and access to new genotyping and sequencing technologies now efficiently allows the identification of mutations involved in canine genetic diseases. To date, PRA genes identified in dog breeds correspond to the same genes in humans and represent relevant RP models, and new genes found in dogs represent good candidate for still unknown human RP. We present here a review of the main advantages of the dog models for human RP with the genes already identified and an X-linked PRA in the Border collie as a model for orphan X-linked RPs in human.
Collapse
|
21
|
Hardcastle AJ, Sieving PA, Sahel JA, Jacobson SG, Cideciyan AV, Flannery JG, Beltran WA, Aguirre GD. Translational Retinal Research and Therapies. Transl Vis Sci Technol 2018; 7:8. [PMID: 30225158 PMCID: PMC6138060 DOI: 10.1167/tvst.7.5.8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 04/13/2018] [Indexed: 01/01/2023] Open
Abstract
The following review summarizes the state of the art in representative aspects of gene therapy/translational medicine and evolves from a symposium held at the School of Veterinary Medicine, University of Pennsylvania on November 16, 2017 honoring Dr. Gustavo Aguirre, recipient of ARVO's 2017 Proctor Medal. Focusing on the retina, speakers highlighted current work on moving therapies for inherited retinal degenerative diseases from the laboratory bench to the clinic.
Collapse
Affiliation(s)
| | - Paul A Sieving
- Director, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - José-Alain Sahel
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Director of the UPMC Eye Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA and Director, Institut de la Vision, Sorbonne Université-Inserm-CNRS, Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Paris, France
| | - Samuel G Jacobson
- Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Artur V Cideciyan
- Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - John G Flannery
- Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - William A Beltran
- Department of Clinical Sciences and Advanced Medicine, Division of Experimental Retinal Therapies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gustavo D Aguirre
- Department of Clinical Sciences and Advanced Medicine, Division of Experimental Retinal Therapies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
22
|
Mutation-independent rhodopsin gene therapy by knockdown and replacement with a single AAV vector. Proc Natl Acad Sci U S A 2018; 115:E8547-E8556. [PMID: 30127005 DOI: 10.1073/pnas.1805055115] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Inherited retinal degenerations are caused by mutations in >250 genes that affect photoreceptor cells or the retinal pigment epithelium and result in vision loss. For autosomal recessive and X-linked retinal degenerations, significant progress has been achieved in the field of gene therapy as evidenced by the growing number of clinical trials and the recent commercialization of the first gene therapy for a form of congenital blindness. However, despite significant efforts to develop a treatment for the most common form of autosomal dominant retinitis pigmentosa (adRP) caused by >150 mutations in the rhodopsin (RHO) gene, translation to the clinic has stalled. Here, we identified a highly efficient shRNA that targets human (and canine) RHO in a mutation-independent manner. In a single adeno-associated viral (AAV) vector we combined this shRNA with a human RHO replacement cDNA made resistant to RNA interference and tested this construct in a naturally occurring canine model of RHO-adRP. Subretinal vector injections led to nearly complete suppression of endogenous canine RHO RNA, while the human RHO replacement cDNA resulted in up to 30% of normal RHO protein levels. Noninvasive retinal imaging showed photoreceptors in treated areas were completely protected from retinal degeneration. Histopathology confirmed retention of normal photoreceptor structure and RHO expression in rod outer segments. Long-term (>8 mo) follow-up by retinal imaging and electroretinography indicated stable structural and functional preservation. The efficacy of this gene therapy in a clinically relevant large-animal model paves the way for treating patients with RHO-adRP.
Collapse
|
23
|
Assessment of Safety and Functional Efficacy of Stem Cell-Based Therapeutic Approaches Using Retinal Degenerative Animal Models. Stem Cells Int 2017; 2017:9428176. [PMID: 28928775 PMCID: PMC5592015 DOI: 10.1155/2017/9428176] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Accepted: 06/19/2017] [Indexed: 02/06/2023] Open
Abstract
Dysfunction and death of retinal pigment epithelium (RPE) and or photoreceptors can lead to irreversible vision loss. The eye represents an ideal microenvironment for stem cell-based therapy. It is considered an “immune privileged” site, and the number of cells needed for therapy is relatively low for the area of focused vision (macula). Further, surgical placement of stem cell-derived grafts (RPE, retinal progenitors, and photoreceptor precursors) into the vitreous cavity or subretinal space has been well established. For preclinical tests, assessments of stem cell-derived graft survival and functionality are conducted in animal models by various noninvasive approaches and imaging modalities. In vivo experiments conducted in animal models based on replacing photoreceptors and/or RPE cells have shown survival and functionality of the transplanted cells, rescue of the host retina, and improvement of visual function. Based on the positive results obtained from these animal experiments, human clinical trials are being initiated. Despite such progress in stem cell research, ethical, regulatory, safety, and technical difficulties still remain a challenge for the transformation of this technique into a standard clinical approach. In this review, the current status of preclinical safety and efficacy studies for retinal cell replacement therapies conducted in animal models will be discussed.
Collapse
|
24
|
Gargini C, Novelli E, Piano I, Biagioni M, Strettoi E. Pattern of retinal morphological and functional decay in a light-inducible, rhodopsin mutant mouse. Sci Rep 2017; 7:5730. [PMID: 28720880 PMCID: PMC5516022 DOI: 10.1038/s41598-017-06045-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/06/2017] [Indexed: 02/02/2023] Open
Abstract
Hallmarks of Retinitis Pigmentosa (RP), a family of genetic diseases, are a typical rod-cone-degeneration with initial night blindness and loss of peripheral vision, followed by decreased daylight sight and progressive visual acuity loss up to legal blindness. Great heterogeneity in nature and function of mutated genes, variety of mutations for each of them, variability in phenotypic appearance and transmission modality contribute to make RP a still incurable disease. Translational research relies on appropriate animal models mimicking the genetic and phenotypic diversity of the human pathology. Here, we provide a systematic, morphological and functional analysis of RhoTvrm4/Rho+ rhodopsin mutant mice, originally described in 2010 and portraying several features of common forms of autosomal dominant RP caused by gain-of-function mutations. These mice undergo photoreceptor degeneration only when exposed briefly to strong, white light and allow controlled timing of induction of rod and cone death, which therefore can be elicited in adult animals, as observed in human RP. The option to control severity and retinal extent of the phenotype by regulating intensity and duration of the inducing light opens possibilities to exploit this model for multiple experimental purposes. Altogether, the unique features of this mutant make it an excellent resource for retinal degeneration research.
Collapse
Affiliation(s)
| | | | - Ilaria Piano
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | | | | |
Collapse
|
25
|
Sudharsan R, Simone KM, Anderson NP, Aguirre GD, Beltran WA. Acute and Protracted Cell Death in Light-Induced Retinal Degeneration in the Canine Model of Rhodopsin Autosomal Dominant Retinitis Pigmentosa. Invest Ophthalmol Vis Sci 2017; 58:270-281. [PMID: 28114588 PMCID: PMC5464465 DOI: 10.1167/iovs.16-20749] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Purpose To characterize a light damage paradigm and establish structural and immunocytochemical measures of acute and protracted light-induced retinal degeneration in the rhodopsin (RHO) T4R dog model of RHO-autosomal dominant retinitis pigmentosa (ADRP). Methods Retinal light damage was induced in mutant dogs with a 1-minute exposure to various light intensities (0.1-1.0 mW/cm2) delivered with a Ganzfeld stimulator, or by fundus photography. Photoreceptor cell death was assessed by TUNEL assay, and alterations in retinal layers were examined by histology and immunohistochemistry 24 hours and 2 weeks after light exposure. Detailed topographic maps were made to document changes in the outer retinal layers of all four retinal quadrants 2 weeks post exposure. Results Twenty-four hours post light exposure, the severity of photoreceptor cell death was dose dependent. Immunohistochemical analysis revealed disruption of rod outer segments, focal loss of the RPE integrity, and an increase in expression of endothelin receptor B in Müller cells with the two highest doses of light and fundus photography. Two weeks after light exposure, persistence of photoreceptor death, thinning of the outer nuclear layer, and induction of Müller cell gliosis occurred with the highest doses of light. Conclusions We have characterized outcome measures of acute and continuing retinal degeneration in the RHO T4R dog following light exposure. These will be used to assess the molecular mechanisms of light-induced damage and rescue strategies in this large animal model of RHO-ADRP.
Collapse
Affiliation(s)
- Raghavi Sudharsan
- Section of Ophthalmology, Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Kristina M Simone
- Section of Ophthalmology, Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Nathan P Anderson
- Section of Ophthalmology, Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Gustavo D Aguirre
- Section of Ophthalmology, Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - William A Beltran
- Section of Ophthalmology, Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| |
Collapse
|
26
|
Aravand P, Ramachandran PS, Schpylchak I, Phelps NT, Nikonov S, Bennett J. Protocols for Visually Guided Navigation Assessment of Efficacy of Retina-Directed Cell or Gene Therapy in Canines. Front Neurosci 2017; 11:215. [PMID: 28491015 PMCID: PMC5405122 DOI: 10.3389/fnins.2017.00215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 03/30/2017] [Indexed: 11/13/2022] Open
Abstract
There has been marked progress in recent years in developing gene delivery approaches for the treatment of inherited blinding diseases. Many of the proof-of-concept studies have utilized rodent models of retinal degeneration. In those models, tests of visual function include a modified water maze swim test, optokinetic nystagmus, and light-dark activity assays. Test paradigms used in rodents can be difficult to replicate in large animals due to their size and awareness of non-visual aspects of the test system. Two types of visual behavior assays have been utilized in canines: an obstacle avoidance course and a forced choice Y maze. Given the progress in developing cell and gene therapies in large animals, such tests will become more and more valuable. This study provides guidelines for carrying out such tests and assesses the challenges and benefits associated with each test.
Collapse
Affiliation(s)
- Puya Aravand
- Department of Ophthalmology, Center for Advanced Retinal and Ocular Therapeutics, F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of MedicinePhiladelphia, PA, USA
| | - Pavitra S Ramachandran
- Department of Ophthalmology, Center for Advanced Retinal and Ocular Therapeutics, F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of MedicinePhiladelphia, PA, USA
| | - Ivan Schpylchak
- Department of Ophthalmology, Center for Advanced Retinal and Ocular Therapeutics, F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of MedicinePhiladelphia, PA, USA
| | - Nicholas T Phelps
- Department of Ophthalmology, Center for Advanced Retinal and Ocular Therapeutics, F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of MedicinePhiladelphia, PA, USA
| | - Sergei Nikonov
- Department of Ophthalmology, Center for Advanced Retinal and Ocular Therapeutics, F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of MedicinePhiladelphia, PA, USA
| | - Jean Bennett
- Department of Ophthalmology, Center for Advanced Retinal and Ocular Therapeutics, F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of MedicinePhiladelphia, PA, USA
| |
Collapse
|