1
|
Nair PP, Keskar MP, Borghare PT, Dzoagbe HY, Kumar T. The New Era of Therapeutic Strategies for the Management of Retinitis Pigmentosa: A Narrative Review of the Pathomolecular Mechanism for Gene Therapies. Cureus 2024; 16:e66814. [PMID: 39280562 PMCID: PMC11393205 DOI: 10.7759/cureus.66814] [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: 07/25/2024] [Accepted: 08/11/2024] [Indexed: 09/18/2024] Open
Abstract
Retinitis pigmentosa, or RP, is a group of inherited retinal degenerations involving progressive loss of photoreceptor cells- rods and cones- ultimately causing severe vision loss and blindness. RP, although a very common ailment, continues to be an incurable disease with little to be done medically. However, with the breakthroughs in gene therapy and stem cell transplantation in recent years, a new door has been opened to the treatment of RP. This narrative review summarizes the pathomolecular mechanisms of RP, focusing on the genetic and molecular abnormalities that lead to the process of retinal degeneration. In this section, we talk about the current theories of how RP develops, gene mutations, oxidative stress, and inflammation. We also delve into new therapeutic approaches such as gene therapy, stem cell transplantation and genome surgery, which are designed to either replace or repair the damaged photoreceptors to restore vision and ultimately enhance the life of the RP patient. Another topic covered is the obstacles and research frontiers of these revolutionary treatments. This article is intended to give a complete overview of the molecular processes of RP and the promising treatment strategies that could change the way this devastating disease is treated.
Collapse
Affiliation(s)
- Praveena P Nair
- Otolaryngology, Mandsaur Institute of Ayurved Education and Research, Bhunyakhedi, IND
- Otolaryngology, Parul Institute of Ayurved, Parul University, Limda, IND
| | - Manjiri P Keskar
- Otolaryngology, Parul institute of Ayurved, Parul University, Limda, IND
| | - Pramod T Borghare
- Otolaryngology, Mahatma Gandhi Ayurved College Hospital and Research, Wardha, IND
| | - Hellen Y Dzoagbe
- Anatomy, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Tanish Kumar
- Medicine, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
2
|
Becker S, L'Ecuyer Z, Jones BW, Zouache MA, McDonnell FS, Vinberg F. Modeling complex age-related eye disease. Prog Retin Eye Res 2024; 100:101247. [PMID: 38365085 PMCID: PMC11268458 DOI: 10.1016/j.preteyeres.2024.101247] [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: 08/15/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/18/2024]
Abstract
Modeling complex eye diseases like age-related macular degeneration (AMD) and glaucoma poses significant challenges, since these conditions depend highly on age-related changes that occur over several decades, with many contributing factors remaining unknown. Although both diseases exhibit a relatively high heritability of >50%, a large proportion of individuals carrying AMD- or glaucoma-associated genetic risk variants will never develop these diseases. Furthermore, several environmental and lifestyle factors contribute to and modulate the pathogenesis and progression of AMD and glaucoma. Several strategies replicate the impact of genetic risk variants, pathobiological pathways and environmental and lifestyle factors in AMD and glaucoma in mice and other species. In this review we will primarily discuss the most commonly available mouse models, which have and will likely continue to improve our understanding of the pathobiology of age-related eye diseases. Uncertainties persist whether small animal models can truly recapitulate disease progression and vision loss in patients, raising doubts regarding their usefulness when testing novel gene or drug therapies. We will elaborate on concerns that relate to shorter lifespan, body size and allometries, lack of macula and a true lamina cribrosa, as well as absence and sequence disparities of certain genes and differences in their chromosomal location in mice. Since biological, rather than chronological, age likely predisposes an organism for both glaucoma and AMD, more rapidly aging organisms like small rodents may open up possibilities that will make research of these diseases more timely and financially feasible. On the other hand, due to the above-mentioned anatomical and physiological features, as well as pharmacokinetic and -dynamic differences small animal models are not ideal to study the natural progression of vision loss or the efficacy and safety of novel therapies. In this context, we will also discuss the advantages and pitfalls of alternative models that include larger species, such as non-human primates and rabbits, patient-derived retinal organoids, and human organ donor eyes.
Collapse
Affiliation(s)
- Silke Becker
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Zia L'Ecuyer
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Bryan W Jones
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Moussa A Zouache
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Fiona S McDonnell
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Frans Vinberg
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
3
|
Lewandowski D, Gao F, Imanishi S, Tworak A, Bassetto M, Dong Z, Pinto AFM, Tabaka M, Kiser PD, Imanishi Y, Skowronska-Krawczyk D, Palczewski K. Restoring retinal polyunsaturated fatty acid balance and retina function by targeting ceramide in AdipoR1-deficient mice. J Biol Chem 2024; 300:107291. [PMID: 38636661 PMCID: PMC11107370 DOI: 10.1016/j.jbc.2024.107291] [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: 02/23/2024] [Revised: 03/30/2024] [Accepted: 04/09/2024] [Indexed: 04/20/2024] Open
Abstract
Mutations in the adiponectin receptor 1 gene (AdipoR1) lead to retinitis pigmentosa and are associated with age-related macular degeneration. This study explores the effects of AdipoR1 gene deficiency in mice, revealing a striking decline in ω3 polyunsaturated fatty acids (PUFA), an increase in ω6 fatty acids, and elevated ceramides in the retina. The AdipoR1 deficiency impairs peroxisome proliferator-activated receptor α signaling, which is crucial for FA metabolism, particularly affecting proteins associated with FA transport and oxidation in the retina and retinal pigmented epithelium. Our lipidomic and proteomic analyses indicate changes that could affect membrane composition and viscosity through altered ω3 PUFA transport and synthesis, suggesting a potential influence of AdipoR1 on these properties. Furthermore, we noted a reduction in the Bardet-Biedl syndrome proteins, which are crucial for forming and maintaining photoreceptor outer segments that are PUFA-enriched ciliary structures. Diminution in Bardet-Biedl syndrome-proteins content combined with our electron microscopic observations raises the possibility that AdipoR1 deficiency might impair ciliary function. Treatment with inhibitors of ceramide synthesis led to substantial elevation of ω3 LC-PUFAs, alleviating photoreceptor degeneration and improving retinal function. These results serve as the proof of concept for a ceramide-targeted strategy to treat retinopathies linked to PUFA deficiency, including age-related macular degeneration.
Collapse
Affiliation(s)
- Dominik Lewandowski
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, California, USA.
| | - Fangyuan Gao
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, California, USA
| | - Sanae Imanishi
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Aleksander Tworak
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, California, USA
| | - Marco Bassetto
- Department of Physiology and Biophysics, University of California, Irvine, California, USA
| | - Zhiqian Dong
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, California, USA
| | - Antonio F M Pinto
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Marcin Tabaka
- International Centre for Translational Eye Research, Warsaw, Poland; Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Philip D Kiser
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, California, USA; Department of Physiology and Biophysics, University of California, Irvine, California, USA; Department of Clinical Pharmacy Practice, University of California, Irvine, California, USA; Research Service, Veterans Affairs Long Beach Healthcare System, Long Beach, California, USA
| | - Yoshikazu Imanishi
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Dorota Skowronska-Krawczyk
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, California, USA; Department of Physiology and Biophysics, University of California, Irvine, California, USA
| | - Krzysztof Palczewski
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, California, USA; Department of Physiology and Biophysics, University of California, Irvine, California, USA; Department of Chemistry, and Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA.
| |
Collapse
|
4
|
Sun X, Liang C, Chen Y, Cui T, Han J, Dai M, Zhang Y, Zhou Q, Li W. Knockout and Replacement Gene Surgery to Treat Rhodopsin-Mediated Autosomal Dominant Retinitis Pigmentosa. Hum Gene Ther 2024; 35:151-162. [PMID: 38368562 DOI: 10.1089/hum.2023.201] [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] [Indexed: 02/19/2024] Open
Abstract
Mutations in the rhodopsin (RHO) gene are the predominant causes of autosomal dominant retinitis pigmentosa (adRP). Given the diverse gain-of-function mutations, therapeutic strategies targeting specific sequences face significant challenges. Here, we provide a universal approach to conquer this problem: we have devised a CRISPR-Cas12i-based, mutation-independent gene knockout and replacement compound therapy carried by a dual AAV2/8 system. In this study, we successfully delayed the progression of retinal degeneration in the classic mouse disease model RhoP23H, and also RhoP347S, a new native mouse mutation model we developed. Our research expands the horizon of potential options for future treatments of RHO-mediated adRP.
Collapse
Affiliation(s)
- Xuehan Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China; and
| | - Chen Liang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China; and
| | - Yangcan Chen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Tongtong Cui
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Jiabao Han
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China; and
| | - Moyu Dai
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China; and
| | - Ying Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China; and
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China; and
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| |
Collapse
|
5
|
Wang B, Arbuckle RK, Davoli KA, Clinger OD, Brown R, Sahel JA, Chen Y, Pi S. Compensation of inner retina to early-stage photoreceptor degeneration in a Rho P23H/+ mouse model of retinitis pigmentosa. Exp Eye Res 2024; 240:109826. [PMID: 38340947 PMCID: PMC10940204 DOI: 10.1016/j.exer.2024.109826] [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: 12/01/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/12/2024]
Abstract
Retinitis pigmentosa (RP) is an inherited retinal disorder characterized by the degeneration of photoreceptors. RhoP23H/+ mice, which carry a Pro23His mutation in the RHODOPSIN (Rho) gene, are one of the most studied animal models for RP. However, except for the photoreceptors, other retinal neural cells have not been fully investigated in this model. Here, we record the temporal changes of the retina by optical coherence tomography (OCT) imaging of the RhoP23H/+ mice, from early to mid-phase of retinal degeneration. Based on thickness analysis, we identified a natural retinal thickness adaption in wild-type mice during early adulthood and observed morphological compensation of the inner retina layer to photoreceptor degeneration in the RhoP23H/+ mice, primarily on the inner nuclear layer (INL). RhoP23H/+ mice findings were further validated via: histology showing the negative correlation of INL and ONL thicknesses; as well as electroretinogram (ERG) showing an increased b-wave to a-wave ratio. These results unravel the sequential morphologic events in this model and suggest a better understanding of retinal degeneration of RP for future studies.
Collapse
Affiliation(s)
- Bingjie Wang
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; UPMC Vision Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Riley K Arbuckle
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; UPMC Vision Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, 15213, USA
| | - Katherine A Davoli
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; UPMC Vision Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Owen D Clinger
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; UPMC Vision Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Richard Brown
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; UPMC Vision Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - José-Alain Sahel
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; UPMC Vision Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Yuanyuan Chen
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; UPMC Vision Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Shaohua Pi
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, 15213, USA; UPMC Vision Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
6
|
Vasudevan S, Senapati S, Pendergast M, Park PSH. Aggregation of rhodopsin mutants in mouse models of autosomal dominant retinitis pigmentosa. Nat Commun 2024; 15:1451. [PMID: 38365903 PMCID: PMC10873427 DOI: 10.1038/s41467-024-45748-4] [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: 04/07/2023] [Accepted: 02/01/2024] [Indexed: 02/18/2024] Open
Abstract
Mutations in rhodopsin can cause it to misfold and lead to retinal degeneration. A distinguishing feature of these mutants in vitro is that they mislocalize and aggregate. It is unclear whether or not these features contribute to retinal degeneration observed in vivo. The effect of P23H and G188R misfolding mutations were examined in a heterologous expression system and knockin mouse models, including a mouse model generated here expressing the G188R rhodopsin mutant. In vitro characterizations demonstrate that both mutants aggregate, with the G188R mutant exhibiting a more severe aggregation profile compared to the P23H mutant. The potential for rhodopsin mutants to aggregate in vivo was assessed by PROTEOSTAT, a dye that labels aggregated proteins. Both mutants mislocalize in photoreceptor cells and PROTEOSTAT staining was detected surrounding the nuclei of photoreceptor cells. The G188R mutant promotes a more severe retinal degeneration phenotype and greater PROTEOSTAT staining compared to that promoted by the P23H mutant. Here, we show that the level of PROTEOSTAT positive cells mirrors the progression and level of photoreceptor cell death, which suggests a potential role for rhodopsin aggregation in retinal degeneration.
Collapse
Affiliation(s)
- Sreelakshmi Vasudevan
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Subhadip Senapati
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
- Prayoga Institute of Education Research, Bengaluru, KA, 560116, India
| | - Maryanne Pendergast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Paul S-H Park
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
7
|
Scott BM, Chen SK, Van Nynatten A, Liu J, Schott RK, Heon E, Peisajovich SG, Chang BSW. Scaling up Functional Analyses of the G Protein-Coupled Receptor Rhodopsin. J Mol Evol 2024; 92:61-71. [PMID: 38324225 DOI: 10.1007/s00239-024-10154-3] [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: 10/17/2023] [Accepted: 12/22/2023] [Indexed: 02/08/2024]
Abstract
Eukaryotic cells use G protein-coupled receptors (GPCRs) to convert external stimuli into internal signals to elicit cellular responses. However, how mutations in GPCR-coding genes affect GPCR activation and downstream signaling pathways remain poorly understood. Approaches such as deep mutational scanning show promise in investigations of GPCRs, but a high-throughput method to measure rhodopsin activation has yet to be achieved. Here, we scale up a fluorescent reporter assay in budding yeast that we engineered to study rhodopsin's light-activated signal transduction. Using this approach, we measured the mutational effects of over 1200 individual human rhodopsin mutants, generated by low-frequency random mutagenesis of the GPCR rhodopsin (RHO) gene. Analysis of the data in the context of rhodopsin's three-dimensional structure reveals that transmembrane helices are generally less tolerant to mutations compared to flanking helices that face the lipid bilayer, which suggest that mutational tolerance is contingent on both the local environment surrounding specific residues and the specific position of these residues in the protein structure. Comparison of functional scores from our screen to clinically identified rhodopsin disease variants found many pathogenic mutants to be loss of function. Lastly, functional scores from our assay were consistent with a complex counterion mechanism involved in ligand-binding and rhodopsin activation. Our results demonstrate that deep mutational scanning is possible for rhodopsin activation and can be an effective method for revealing properties of mutational tolerance that may be generalizable to other transmembrane proteins.
Collapse
Affiliation(s)
- Benjamin M Scott
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Steven K Chen
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | | | - Jing Liu
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Ryan K Schott
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, ON, Canada
- Department of Biology and Centre for Vision Research, York University, Toronto, ON, Canada
- Department of Vertebrate Zoology, National Museum of Natural History, Smithsonian Institution, Washington, DC, USA
| | - Elise Heon
- Department of Ophthalmology, Hospital for Sick Children, Toronto, ON, Canada
| | - Sergio G Peisajovich
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Belinda S W Chang
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada.
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, ON, Canada.
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
8
|
Sechrest ER, Ma X, Cahill ME, Barbera RJ, Wang Y, Deng WT. Structural and functional rescue of cones carrying the most common cone opsin C203R missense mutation. JCI Insight 2024; 9:e172834. [PMID: 38060327 PMCID: PMC10906232 DOI: 10.1172/jci.insight.172834] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 12/05/2023] [Indexed: 01/24/2024] Open
Abstract
An arginine to cysteine substitution at amino acid position 203 (C203R) is the most common missense mutation in human cone opsin. Linked to color blindness and blue cone monochromacy (BCM), C203 is involved in a crucial disulfide bond required for proper folding. It has previously been postulated that expression of mutant C203R cone opsin exerts a toxic effect on cone photoreceptors, similar to some well-characterized missense mutations in rhodopsin that lead to protein misfolding. In this study, we generated and characterized a BCM mouse model carrying the equivalent C203R mutation (Opn1mwC198R Opn1sw-/-) to investigate the disease mechanism and develop a gene therapy approach for this disorder. Untreated Opn1mwC198R Opn1sw-/- cones phenocopied affected cones in human patients with the equivalent mutation, exhibiting shortened or absent cone outer segments and loss of function. We determined that gene augmentation targeting cones specifically yielded robust rescue of cone function and structure when Opn1mwC198R Opn1sw-/- mice were treated at early ages. Importantly, treated cones displayed elaborated outer segments and replenished expression of crucial cone phototransduction proteins. Interestingly, we were unable to detect OPN1MWC198R mutant opsin at any age. We believe this is the first proof-of-concept study exploring the efficacy of gene therapy in BCM associated with a C203R mutation.
Collapse
Affiliation(s)
- Emily R. Sechrest
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, USA
| | - Xiaojie Ma
- Department of Ophthalmology, University of Florida, Gainesville, Florida, USA
| | - Marion E. Cahill
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, USA
- Department of Biology and
| | - Robert J. Barbera
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, USA
| | - Yixiao Wang
- Department of Ophthalmology, University of Florida, Gainesville, Florida, USA
| | - Wen-Tao Deng
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, USA
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
9
|
Katada Y, Yoshida K, Serizawa N, Lee D, Kobayashi K, Negishi K, Okano H, Kandori H, Tsubota K, Kurihara T. Highly sensitive visual restoration and protection via ectopic expression of chimeric rhodopsin in mice. iScience 2023; 26:107716. [PMID: 37720108 PMCID: PMC10504486 DOI: 10.1016/j.isci.2023.107716] [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: 05/10/2023] [Revised: 07/22/2023] [Accepted: 08/22/2023] [Indexed: 09/19/2023] Open
Abstract
Photoreception requires amplification by mammalian rhodopsin through G protein activation, which requires a visual cycle. To achieve this in retinal gene therapy, we incorporated human rhodopsin cytoplasmic loops into Gloeobacter rhodopsin, thereby generating Gloeobacter and human chimeric rhodopsin (GHCR). In a murine model of inherited retinal degeneration, we induced retinal GHCR expression by intravitreal injection of a recombinant adeno-associated virus vector. Retinal explant and visual thalamus electrophysiological recordings, behavioral tests, and histological analysis showed that GHCR restored dim-environment vision and prevented the progression of retinal degeneration. Thus, GHCR may be a potent clinical tool for the treatment of retinal disorders.
Collapse
Affiliation(s)
- Yusaku Katada
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kazuho Yoshida
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Aichi 466-0061, Japan
| | - Naho Serizawa
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Nutritional Sciences, Toyo University, Kita-ku, Tokyo 115-8650, Japan
| | - Deokho Lee
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hideki Kandori
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Aichi 466-0061, Japan
| | - Kazuo Tsubota
- Tsubota Laboratory, Inc., Shinjuku-ku, Tokyo 160-0016, Japan
| | - Toshihide Kurihara
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
10
|
Du W, Li J, Tang X, Yu W, Zhao M. CRISPR/SaCas9-based gene editing rescues photoreceptor degeneration throughout a rhodopsin-associated autosomal dominant retinitis pigmentosa mouse model. Exp Biol Med (Maywood) 2023; 248:1818-1828. [PMID: 37837380 PMCID: PMC10792415 DOI: 10.1177/15353702231199069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/12/2023] [Indexed: 10/16/2023] Open
Abstract
Rhodopsin (Rho) gene mutation was considered the highest prevalent mutation in autosomal dominant retinitis pigmentosa (ADRP); however, effective therapeutics for ADRP have not been developed. The process of gene editing via the clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9 system offers the potentiality to provide cures for dominantly inherited disorders. Herein, we generated a CRISPR/SaCas9-mediated gene reduction system to inactivate the Rho mutant, while replacing normal rhodopsin in a rhodopsin mutation mouse model. When Rho-P23H knock-in mice were administered a subretinal injection of the "reduction and replacement" system, the expression of mutant rhodopsin was reduced, and retinal function was improved. Therefore, we concluded that CRISPR/SaCas9-based "reduction and replacement" gene therapy could provide structural and functional benefits for Rho mutant ADRP, as well as new directions for future clinical research on the treatment of such gain-of-function genetic diseases.
Collapse
Affiliation(s)
- Wei Du
- Department of Ophthalmology and Clinical Centre of Optometry, Peking University People’s Hospital, Beijing 100044, China
- Eye Diseases and Optometry Institute, Peking University People’s Hospital, Beijing 100044, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing 100044, China
- College of Optometry, Peking University Health Science Center, Beijing 100044, China
| | - Jiarui Li
- Department of Ophthalmology and Clinical Centre of Optometry, Peking University People’s Hospital, Beijing 100044, China
- Eye Diseases and Optometry Institute, Peking University People’s Hospital, Beijing 100044, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing 100044, China
- College of Optometry, Peking University Health Science Center, Beijing 100044, China
| | - Xin Tang
- Department of Ophthalmology and Clinical Centre of Optometry, Peking University People’s Hospital, Beijing 100044, China
- Eye Diseases and Optometry Institute, Peking University People’s Hospital, Beijing 100044, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing 100044, China
- College of Optometry, Peking University Health Science Center, Beijing 100044, China
| | - Wenzhen Yu
- Department of Ophthalmology and Clinical Centre of Optometry, Peking University People’s Hospital, Beijing 100044, China
- Eye Diseases and Optometry Institute, Peking University People’s Hospital, Beijing 100044, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing 100044, China
- College of Optometry, Peking University Health Science Center, Beijing 100044, China
| | - Mingwei Zhao
- Department of Ophthalmology and Clinical Centre of Optometry, Peking University People’s Hospital, Beijing 100044, China
- Eye Diseases and Optometry Institute, Peking University People’s Hospital, Beijing 100044, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing 100044, China
- College of Optometry, Peking University Health Science Center, Beijing 100044, China
| |
Collapse
|
11
|
Chakrabarty K, Nayak D, Debnath J, Das D, Shetty R, Ghosh A. Retinal organoids in disease modeling and drug discovery: Opportunities and challenges. Surv Ophthalmol 2023:S0039-6257(23)00127-3. [PMID: 37778668 DOI: 10.1016/j.survophthal.2023.09.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 09/25/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
Diseases leading to retinal cell loss can cause severe visual impairment and blindness. The lack of effective therapies to address retinal cell loss and the absence of intrinsic regeneration in the human retina leads to an irreversible pathological condition. Progress in recent years in the generation of human three-dimensional retinal organoids from pluripotent stem cells makes it possible to recreate the cytoarchitecture and associated cell-cell interactions of the human retina in remarkable detail. These human three-dimensional retinal organoid systems made of distinct retinal cell types and possessing contextual physiological responses allow the study of human retina development and retinal disease pathology in a way animal model and two-dimensional cell cultures were unable to achieve. We describe the derivation of retinal organoids from human pluripotent stem cells and their application for modeling retinal disease pathologies, while outlining the opportunities and challenges for its application in academia and industry.
Collapse
Affiliation(s)
- Koushik Chakrabarty
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, Karnataka, India.
| | - Divyani Nayak
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, Karnataka, India
| | - Jayasree Debnath
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, Karnataka, India
| | - Debashish Das
- Stem Cell Research Lab, GROW Lab, Narayana Nethralaya Foundation, Narayana Nethralaya, Bangalore, Karnataka, India
| | - Rohit Shetty
- Department of Cornea and Refractive Surgery, Narayana Nethralaya Eye Hospital, Bangalore, Karnataka, India
| | - Arkasubhra Ghosh
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, Karnataka, India
| |
Collapse
|
12
|
Daich Varela M, Georgiadis A, Michaelides M. Genetic treatment for autosomal dominant inherited retinal dystrophies: approaches, challenges and targeted genotypes. Br J Ophthalmol 2023; 107:1223-1230. [PMID: 36038193 DOI: 10.1136/bjo-2022-321903] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/01/2022] [Indexed: 11/04/2022]
Abstract
Inherited retinal diseases (IRDs) have been in the front line of gene therapy development for the last decade, providing a useful platform to test novel therapeutic approaches. More than 40 clinical trials have been completed or are ongoing, tackling autosomal recessive and X-linked conditions, mostly through adeno-associated viral vector delivery of a normal copy of the disease-causing gene. However, only recently has autosomal dominant (ad) disease been targeted, with the commencement of a trial for rhodopsin (RHO)-associated retinitis pigmentosa (RP), implementing antisense oligonucleotide (AON) therapy, with promising preliminary results (NCT04123626).Autosomal dominant RP represents 15%-25% of all RP, with RHO accounting for 20%-30% of these cases. Autosomal dominant macular and cone-rod dystrophies (MD/CORD) correspond to approximately 7.5% of all IRDs, and approximately 35% of all MD/CORD cases, with the main causative gene being BEST1 Autosomal dominant IRDs are not only less frequent than recessive, but also tend to be less severe and have later onset; for example, an individual with RHO-adRP would typically become severely visually impaired at an age 2-3 times older than in X-linked RPGR-RP.Gain-of-function and dominant negative aetiologies are frequently seen in the prevalent adRP genes RHO, RP1 and PRPF31 among others, which would not be effectively addressed by gene supplementation alone and need creative, novel approaches. Zinc fingers, RNA interference, AON, translational read-through therapy, and gene editing by clustered regularly interspaced short palindromic repeats/Cas are some of the strategies that are currently under investigation and will be discussed here.
Collapse
Affiliation(s)
- Malena Daich Varela
- Moorfields Eye Hospital, London, UK
- UCL Institute of Ophthalmology, University College London, London, UK
| | | | - Michel Michaelides
- Moorfields Eye Hospital, London, UK
- UCL Institute of Ophthalmology, University College London, London, UK
| |
Collapse
|
13
|
Network biology analysis of P23H rhodopsin interactome identifies protein and mRNA quality control mechanisms. Sci Rep 2022; 12:17405. [PMID: 36258031 PMCID: PMC9579138 DOI: 10.1038/s41598-022-22316-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/12/2022] [Indexed: 02/05/2023] Open
Abstract
Rhodopsin is essential for phototransduction, and many rhodopsin mutations cause heritable retinal degenerations. The P23H rhodopsin variant generates a misfolded rhodopsin protein that photoreceptors quickly target for degradation by mechanisms that are incompletely understood. To gain insight into how P23H rhodopsin is removed from rods, we used mass spectrometry to identify protein interaction partners of P23H rhodopsin immunopurified from RhoP23H/P23H mice and compared them with protein interaction partners of wild-type rhodopsin from Rho+/+ mice. We identified 286 proteins associated with P23H rhodopsin and 276 proteins associated with wild-type rhodopsin. 113 proteins were shared between wild-type and mutant rhodopsin protein interactomes. In the P23H rhodopsin protein interactome, we saw loss of phototransduction, retinal cycle, and rhodopsin protein trafficking proteins but gain of ubiquitin-related proteins when compared with the wild-type rhodopsin protein interactome. In the P23H rhodopsin protein interactome, we saw enrichment of gene ontology terms related to ER-associated protein degradation, ER stress, and translation. Protein-protein interaction network analysis revealed that translational and ribosomal quality control proteins were significant regulators in the P23H rhodopsin protein interactome. The protein partners identified in our study may provide new insights into how photoreceptors recognize and clear mutant rhodopsin, offering possible novel targets involved in retinal degeneration pathogenesis.
Collapse
|
14
|
Yefimova MG. Myelinosome organelles in pathological retinas: ubiquitous presence and dual role in ocular proteostasis maintenance. Neural Regen Res 2022; 18:1009-1016. [PMID: 36254982 PMCID: PMC9827766 DOI: 10.4103/1673-5374.355753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The timely and efficient elimination of aberrant proteins and damaged organelles, formed in response to various genetic and environmental stressors, is a vital need for all cells of the body. Recent lines of evidence point out several non-classical strategies employed by ocular tissues to cope with aberrant constituents generated in the retina and in the retinal pigmented epithelium cells exposed to various stressors. Along with conventional strategies relying upon the intracellular degradation of aberrant constituents through ubiquitin-proteasome and/or lysosome-dependent autophagy proteolysis, two non-conventional mechanisms also contribute to proteostasis maintenance in ocular tissues. An exosome-mediated clearing and a myelinosome-driven secretion mechanism do not require intracellular degradation but provide the export of aberrant constituents and "waste proteins" outside of the cells. The current review is centered on the non-degradative myelinosome-driven secretion mechanism, which operates in the retina of transgenic Huntington's disease R6/1 model mice. Myelinosome-driven secretion is supported by rare organelles myelinosomes that are detected not only in degenerative Huntington's disease R6/1 retina but also in various pathological states of the retina and of the retinal pigmented epithelium. The intra-retinal traffic and inter-cellular exchange of myelinosomes was discussed in the context of a dual role of the myelinosome-driven secretion mechanism for proteostasis maintenance in different ocular compartments. Special focus was made on the interplay between degradative and non-degradative strategies in ocular pathophysiology, to delineate potential therapeutic approaches to counteract several vision diseases.
Collapse
Affiliation(s)
- Marina G. Yefimova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St-Petersburg, Russia,Laboratoire STIM CNRS ERL 7003, Université de Poitiers, Poitiers, France,Correspondence to: Marina G. Yefimova, .
| |
Collapse
|
15
|
Ortega JT, McKee AG, Roushar FJ, Penn WD, Schlebach JP, Jastrzebska B. Chromenone derivatives as novel pharmacological chaperones for retinitis pigmentosa-linked rod opsin mutants. Hum Mol Genet 2022; 31:3439-3457. [PMID: 35642742 PMCID: PMC9558842 DOI: 10.1093/hmg/ddac125] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 11/14/2022] Open
Abstract
The correct expression of folded, functional rhodopsin (Rho) is critical for visual perception. However, this seven-transmembrane helical G protein-coupled receptor is prone to mutations with pathological consequences of retinal degeneration in retinitis pigmentosa (RP) due to Rho misfolding. Pharmacological chaperones that stabilize the inherited Rho variants by assisting their folding and membrane targeting could slow the progression of RP. In this study, we employed virtual screening of synthetic compounds with a natural product scaffold in conjunction with in vitro and in vivo evaluations to discover a novel chromenone-containing small molecule with favorable pharmacological properties that stabilize rod opsin. This compound reversibly binds to unliganded bovine rod opsin with an EC50 value comparable to the 9-cis-retinal chromophore analog and partially rescued membrane trafficking of multiple RP-related rod opsin variants in vitro. Importantly, this novel ligand of rod opsin was effective in vivo in murine models, protecting photoreceptors from deterioration caused by either bright light or genetic insult. Together, our current study suggests potential broad therapeutic implications of the new chromenone-containing non-retinoid small molecule against retinal diseases associated with photoreceptor degeneration.
Collapse
Affiliation(s)
- Joseph T Ortega
- Department of Pharmacology and Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Andrew G McKee
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, IN 47405-7102, USA
| | - Francis J Roushar
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, IN 47405-7102, USA
| | - Wesley D Penn
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, IN 47405-7102, USA
| | - Jonathan P Schlebach
- To whom correspondence should be addressed at: Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 441064965, USA. Tel: +1 2163685683; Fax: +1 2163681300; (Beata Jastrzebska); Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, IN 47405-7102, USA. Tel: +1 812-855-6779; Fax: +1 812-855-8300; (Jonathan P. Schlebach)
| | - Beata Jastrzebska
- To whom correspondence should be addressed at: Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 441064965, USA. Tel: +1 2163685683; Fax: +1 2163681300; (Beata Jastrzebska); Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, IN 47405-7102, USA. Tel: +1 812-855-6779; Fax: +1 812-855-8300; (Jonathan P. Schlebach)
| |
Collapse
|
16
|
Zhao X, Tebbe L, Naash MI, Al-Ubaidi MR. The Neuroprotective Role of Retbindin, a Metabolic Regulator in the Neural Retina. Front Pharmacol 2022; 13:919667. [PMID: 35873559 PMCID: PMC9298789 DOI: 10.3389/fphar.2022.919667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/10/2022] [Indexed: 11/13/2022] Open
Abstract
Dysregulation of retinal metabolism is emerging as one of the major reasons for many inherited retinal diseases (IRDs), a leading cause of blindness worldwide. Thus, the identification of a common regulator that can preserve or revert the metabolic ecosystem to homeostasis is a key step in developing a treatment for different forms of IRDs. Riboflavin (RF) and its derivatives (flavins), flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD), are essential cofactors for a wide range of cellular metabolic processes; hence, they are particularly critical in highly metabolically active tissues such as the retina. Patients with RF deficiency (ariboflavinosis) often display poor photosensitivity resulting in impaired low-light vision. We have identified a novel retina-specific RF binding protein called retbindin (Rtbdn), which plays a key role in retaining flavin levels in the neural retina. This role is mediated by its specific localization at the interface between the neural retina and retinal pigment epithelium (RPE), which is essential for metabolite and nutrient exchange. As a consequence of this vital function, Rtbdn's role in flavin utilization and metabolism in retinal degeneration is discussed. The principal findings are that Rtbdn helps maintain high levels of retinal flavins, and its ablation leads to an early-onset retinal metabolic dysregulation, followed by progressive degeneration of rod and cone photoreceptors. Lack of Rtbdn reduces flavin levels, forcing the neural retina to repurpose glucose to reduce the production of free radicals during ATP production. This leads to metabolic breakdown followed by retinal degeneration. Assessment of the role of Rtbdn in several preclinical retinal disease models revealed upregulation of its levels by several folds prior to and during the degenerative process. Ablation of Rtbdn in these models accelerated the rate of retinal degeneration. In agreement with these in vivo studies, we have also demonstrated that Rtbdn protects immortalized cone photoreceptor cells (661W cells) from light damage in vitro. This indicates that Rtbdn plays a neuroprotective role during retinal degeneration. Herein, we discussed the specific function of Rtbdn and its neuroprotective role in retinal metabolic homeostasis and its role in maintaining retinal health.
Collapse
Affiliation(s)
| | | | - Muna I. Naash
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Muayyad R. Al-Ubaidi
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| |
Collapse
|
17
|
Robichaux MA, Nguyen V, Chan F, Kailasam L, He F, Wilson JH, Wensel TG. Subcellular localization of mutant P23H rhodopsin in an RFP fusion knock-in mouse model of retinitis pigmentosa. Dis Model Mech 2022; 15:274688. [PMID: 35275162 PMCID: PMC9092655 DOI: 10.1242/dmm.049336] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 02/28/2022] [Indexed: 12/15/2022] Open
Abstract
The P23H mutation in rhodopsin (Rho), the rod visual pigment, is the most common allele associated with autosomal-dominant retinitis pigmentosa (adRP). The fate of misfolded mutant Rho in rod photoreceptors has yet to be elucidated. We generated a new mouse model, in which the P23H-Rho mutant allele is fused to the fluorescent protein Tag-RFP-T (P23HhRhoRFP). In heterozygotes, outer segments formed, and wild-type (WT) rhodopsin was properly localized, but mutant P23H-Rho protein was mislocalized in the inner segments. Heterozygotes exhibited slowly progressing retinal degeneration. Mislocalized P23HhRhoRFP was contained in greatly expanded endoplasmic reticulum (ER) membranes. Quantification of mRNA for markers of ER stress and the unfolded protein response revealed little or no increases. mRNA levels for both the mutant human rhodopsin allele and the WT mouse rhodopsin were reduced, but protein levels revealed selective degradation of the mutant protein. These results suggest that the mutant rods undergo an adaptative process that prolongs survival despite unfolded protein accumulation in the ER. The P23H-Rho-RFP mouse may represent a useful tool for the future study of the pathology and treatment of P23H-Rho and adRP. This article has an associated First Person interview with the first author of the paper. Summary: A mouse line with a knock-in of the human rhodopsin gene altered to contain the P23H mutation and a red fluorescent protein fusion provides a new model for autosomal-dominant retinitis pigmentosa.
Collapse
Affiliation(s)
- Michael A Robichaux
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA.,Departments of Ophthalmology and Biochemistry, West Virginia University, 108 Biomedical Road, Morgantown, WV 26506, USA
| | - Vy Nguyen
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Fung Chan
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Lavanya Kailasam
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Feng He
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - John H Wilson
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Theodore G Wensel
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
18
|
Vats A, Xi Y, Feng B, Clinger OD, St Leger AJ, Liu X, Ghosh A, Dermond CD, Lathrop KL, Tochtrop GP, Picaud S, Chen Y. Non-retinoid chaperones improve rhodopsin homeostasis in a mouse model of retinitis pigmentosa. JCI Insight 2022; 7:153717. [PMID: 35472194 PMCID: PMC9220944 DOI: 10.1172/jci.insight.153717] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 04/20/2022] [Indexed: 11/17/2022] Open
Abstract
Rhodopsin-associated (RHO-associated) retinitis pigmentosa (RP) is a progressive retinal disease that currently has no cure. RHO protein misfolding leads to disturbed proteostasis and the death of rod photoreceptors, resulting in decreased vision. We previously identified nonretinoid chaperones of RHO, including YC-001 and F5257-0462, by small-molecule high-throughput screening. Here, we profile the chaperone activities of these molecules toward the cell-surface level of 27 RP-causing human RHO mutants in NIH3T3 cells. Furthermore, using retinal explant culture, we show that YC-001 improves retinal proteostasis by supporting RHO homeostasis in RhoP23H/+ mouse retinae, which results in thicker outer nuclear layers (ONL), indicating delayed photoreceptor degeneration. Interestingly, YC-001 ameliorated retinal immune responses and reduced the number of microglia/macrophages in the RhoP23H/+ retinal explants. Similarly, F5257-0462 also protects photoreceptors in RhoP23H/+ retinal explants. In vivo, intravitreal injection of YC-001 or F5257-0462 microparticles in PBS shows that F5257-0462 has a higher efficacy in preserving photoreceptor function and delaying photoreceptor death in RhoP23H/+ mice. Collectively, we provide proof of principle that nonretinoid chaperones are promising drug candidates in treating RHO-associated RP.
Collapse
Affiliation(s)
- Abhishek Vats
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Yibo Xi
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Bing Feng
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Owen D Clinger
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Anthony J St Leger
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Xujie Liu
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Archisha Ghosh
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Chase D Dermond
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Kira L Lathrop
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Gregory P Tochtrop
- Department of Chemistry, Case Western Reserve University, Cleveland, United States of America
| | - Serge Picaud
- Institut de la Vision, Sorbonne Université, Paris, France
| | - Yuanyuan Chen
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| |
Collapse
|
19
|
Kajtna J, Tsang SH, Koch SF. Late-stage rescue of visually guided behavior in the context of a significantly remodeled retinitis pigmentosa mouse model. Cell Mol Life Sci 2022; 79:148. [PMID: 35195763 PMCID: PMC8866266 DOI: 10.1007/s00018-022-04161-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/13/2022] [Accepted: 01/19/2022] [Indexed: 11/25/2022]
Abstract
Patients with progressive neurodegenerative disorder retinitis pigmentosa (RP) are diagnosed in the midst of ongoing retinal degeneration and remodeling. Here, we used a Pde6b-deficient RP gene therapy mouse model to test whether treatment at late disease stages can halt photoreceptor degeneration and degradative remodeling, while sustaining constructive remodeling and restoring function. We demonstrated that when fewer than 13% of rods remain, our genetic rescue halts photoreceptor degeneration, electroretinography (ERG) functional decline and inner retinal remodeling. In addition, in a water maze test, the performance of mice treated at 16 weeks of age or earlier was indistinguishable from wild type. In contrast, no efficacy was apparent in mice treated at 24 weeks of age, suggesting the photoreceptors had reached a point of no return. Further, remodeling in the retinal pigment epithelium (RPE) and retinal vasculature was not halted at 16 or 24 weeks of age, although there appeared to be some slowing of blood vessel degradation. These data suggest a novel working model in which restoration of clinically significant visual function requires only modest threshold numbers of resilient photoreceptors, halting of destructive remodeling and sustained constructive remodeling. These novel findings define the potential and limitations of RP treatment and suggest possible nonphotoreceptor targets for gene therapy optimization.
Collapse
Affiliation(s)
- Jacqueline Kajtna
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
- Physiological Genomics, BioMedical Center, Ludwig-Maximilians-Universität München, Planegg/Martinsried, Germany
| | - Stephen H Tsang
- Jonas Children's Vision Care, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, 10032, USA
| | - Susanne F Koch
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany.
- Physiological Genomics, BioMedical Center, Ludwig-Maximilians-Universität München, Planegg/Martinsried, Germany.
| |
Collapse
|
20
|
Yu Y, Li L, Lin S, Hu J. Update of application of olfactory ensheathing cells and stem cells/exosomes in the treatment of retinal disorders. Stem Cell Res Ther 2022; 13:11. [PMID: 35012635 PMCID: PMC8751324 DOI: 10.1186/s13287-021-02685-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 12/07/2021] [Indexed: 11/10/2022] Open
Abstract
Age-related macular degeneration, diabetic retinopathy, retinitis pigmentosa and other retinal disorders are the main causes of visual impairment worldwide. In the past, these retinal diseases, especially dry age-related macular degeneration, proliferative diabetic retinopathy and retinitis pigmentosa, were treated with traditional surgery and drugs. However, the effect was moderate. In recent years, researchers have used embryonic stem cells, induced pluripotent stem cells, mesenchymal stem cells, olfactory ensheathing cells and other stem cells to conduct experiments and found that stem cells can inhibit inflammation, regulate immune response, secrete neurotrophic factors, and differentiate into retinal cells to replace and promote restoration of the damaged parts. These stem cells have the potential to treat retinal diseases. Whether it is in animal experiments or clinical trials, the increase in the number of retinal cells, maintenance of function and improvement of visual function all reflect the advanced of stem cells to treat retinal diseases, but its risk preserves the donor's hidden pathogenic genes, immune rejection and tumorigenicity. With the development of exosomes study, researchers have discovered that exosomes come from a wide range of sources and can be secreted by almost all types of cells. Using exosomes with stem cell to treat retinal diseases is more effective than using stem cells alone. This review article summarizes the recent advances in the application of olfactory ensheathing cells and stem cells/exosomes in the treatment of retinal disorders.
Collapse
Affiliation(s)
- Yang Yu
- Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Engineering Research Center of Assistive Technology for Visual Impairment, Fujian Province University, Quanzhou, 362000, Fujian Province, China
| | - Licheng Li
- Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Engineering Research Center of Assistive Technology for Visual Impairment, Fujian Province University, Quanzhou, 362000, Fujian Province, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian Province, China. .,Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia.
| | - Jianmin Hu
- Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Engineering Research Center of Assistive Technology for Visual Impairment, Fujian Province University, Quanzhou, 362000, Fujian Province, China. .,The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, 350004, Fujian Province, China.
| |
Collapse
|
21
|
Ortega JT, Jastrzebska B. Rhodopsin as a Molecular Target to Mitigate Retinitis Pigmentosa. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1371:61-77. [PMID: 34962636 DOI: 10.1007/5584_2021_682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Retinitis pigmentosa (RP) is a group of hereditary degenerative diseases affecting 1 of 4000 people worldwide and being the most prevalent cause of visual handicap among working populations in developed countries. These disorders are mainly related to the abnormalities in the rod G protein-coupled receptor (GPCR), rhodopsin reflected in the dysregulated membrane trafficking, stability and phototransduction processes that lead to progressive loss of retina function and eventually blindness. Currently, there is no cure for RP, and the therapeutic options are limited. Targeting rhodopsin with small molecule chaperones to improve the folding and stability of the mutant receptor is one of the most promising pharmacological approaches to alleviate the pathology of RP. This review provides an update on the current knowledge regarding small molecule compounds that have been evaluated as rhodopsin modulators to be considered as leads for the development of novel therapies for RP.
Collapse
Affiliation(s)
- Joseph T Ortega
- Department of Pharmacology, School of Medicine, Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Beata Jastrzebska
- Department of Pharmacology, School of Medicine, Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
22
|
Liu X, Jia R, Meng X, Li Y, Yang L. Retinal degeneration in humanized mice expressing mutant rhodopsin under the control of the endogenous murine promoter. Exp Eye Res 2021; 215:108893. [PMID: 34919893 DOI: 10.1016/j.exer.2021.108893] [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: 04/12/2021] [Revised: 10/31/2021] [Accepted: 12/08/2021] [Indexed: 11/04/2022]
Abstract
RHO is one of the most common genetic causes of autosomal dominant retinitis Pigmentosa (adRP) and there is no effective therapy for this disease. While rapidly developed CRISPR/Cas9 gene editing technology presents a promising therapeutic strategy to treat adRP. A large number of studies for treating adRP using CRISPR/Cas9 have been performed based on transgenic mouse models which are affected with adRP caused by mutant mouse rhodopsin allele, the counterpart of human rhodopsin. Recently, some RHO humanized mouse models like T17M, P23H are generated, which permit testing of the therapeutic effect of CRISPR/Cas9 in preclinical in vivo systems, without putting humans at risk. While available humanized mouse models are few compared to the number of known RHO mutations, but it is time-consuming and costly to build humanized mice for each mutation. We wonder whether a humanized mouse model having several mutations simultaneously can be developed, although which rarely occurs in patients, to investigate the therapeutic effect of CRISPR/Cas9 for RHO-mediated adRP in preclinical in vivo systems. Homology directed repair strategy combing with CRISPR/Cas9 was employed to introduce human RHO genomic fragment containing the replacement of mouse exon1(mE1) after the start codon to mE5 before the stop codon and all introns by the human counterparts. The human rhodopsin could express under the control of the endogenous murine promoter both transcriptionally and translationally in vivo. Human rhodopsin in humanized mouse lines (without mutation) could replace murine rhodopsin morphologically and functionally. While human rhodopsin containing T17M, G51D, G114R, R135W and P171R mutations simultaneously in mutant humanized (Mut-Rhowt/hum and Mut-Rhohum/hum) mouse lines caused retinal degeneration. Mut-Rhohum/hum mice suffered from severe retinal degeneration with defective formation of rod outer segment, leaving nonrecordable electroretinogram (ERG) at 3 months. Mut- Rhowt/hum mice had a slower rate of photoreceptors loss. In 7-month-old Mut- Rhowt/hum mice, statistically reduced scotopic ERG responses were visible compared with age-matched WT mice, but the shortened outer segment and thinner outer nuclear layer could be observed from 3 months. From 7 months to 9 months, significantly abnormal scotopic ERG responses were visible and photoreceptors loss were also obvious in 9-month-old Mut-Rhowt/hum mice. In 12-month-old Mut- Rhowt/hum mice, statistically reduced scotopic and photopic ERG responses and retinal degeneration throughout the retina were visible. Because scotopic responses were more affected than photopic responses in mutant humanized mice, demonstrating that rods dysfunction was more severe than cones dysfunction and deteriorated earlier, the pattern of retinal degeneration caused by mutant human rhodopsin was a typical rod-cone decay. Immunocytochemistry in cells indicated human rhodopsin proteins with 5 mutations aggregated in the cytoplasm and were also retained in the endoplasmic reticulum. The mutant human rhodopsin also accumulated in rod inner segments and cellular bodies in vivo. In conclusion, our humanized models provide excellent opportunities to study the human rhodopsin expression patterns. Our mutant humanized heterozygotes can provide opportunities to explore gene editing therapies via CRISPR/Cas9 for these five mutations in preclinical studies, it is time-saving and cost-effective.
Collapse
Affiliation(s)
- Xiaozhen Liu
- Department of Ophthalmology, Peking University Third Hospital, Beijing, 100191, China; Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, 100191, China
| | - Ruixuan Jia
- Department of Ophthalmology, Peking University Third Hospital, Beijing, 100191, China; Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, 100191, China
| | - Xiang Meng
- Department of Ophthalmology, Peking University Third Hospital, Beijing, 100191, China; Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, 100191, China
| | - Ying Li
- Department of Ophthalmology, Peking University Third Hospital, Beijing, 100191, China; Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, 100191, China
| | - Liping Yang
- Department of Ophthalmology, Peking University Third Hospital, Beijing, 100191, China; Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
23
|
Felline A, Schiroli D, Comitato A, Marigo V, Fanelli F. Structure network-based landscape of rhodopsin misfolding by mutations and algorithmic prediction of small chaperone action. Comput Struct Biotechnol J 2021; 19:6020-6038. [PMID: 34849206 PMCID: PMC8605067 DOI: 10.1016/j.csbj.2021.10.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/09/2021] [Accepted: 10/31/2021] [Indexed: 11/28/2022] Open
Abstract
Failure of a protein to achieve its functional structural state and normal cellular location contributes to the etiology and pathology of heritable human conformational diseases. The autosomal dominant form of retinitis pigmentosa (adRP) is an incurable blindness largely linked to mutations of the membrane protein rod opsin. While the mechanisms underlying the noxious effects of the mutated protein are not completely understood, a common feature is the functional protein conformational loss. Here, the wild type and 39 adRP rod opsin mutants were subjected to mechanical unfolding simulations coupled to the graph theory-based protein structure network analysis. A robust computational model was inferred and in vitro validated in its ability to predict endoplasmic reticulum retention of adRP mutants, a feature linked to the mutation-caused misfolding. The structure-based approach could also infer the structural determinants of small chaperone action on misfolded protein mutants with therapeutic implications. The approach is exportable to conformational diseases linked to missense mutations in any membrane protein.
Collapse
Affiliation(s)
- Angelo Felline
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi 103, 41125 Modena, Italy
| | - Davide Schiroli
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi 287, 41125 Modena, Italy
| | - Antonella Comitato
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi 287, 41125 Modena, Italy
| | - Valeria Marigo
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi 287, 41125 Modena, Italy.,Center for Neuroscience and Neurotechnology, Italy
| | - Francesca Fanelli
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi 103, 41125 Modena, Italy.,Center for Neuroscience and Neurotechnology, Italy
| |
Collapse
|
24
|
Pharmacological Inhibition of the VCP/Proteasome Axis Rescues Photoreceptor Degeneration in RHO P23H Rat Retinal Explants. Biomolecules 2021; 11:biom11101528. [PMID: 34680161 PMCID: PMC8534135 DOI: 10.3390/biom11101528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/02/2021] [Accepted: 10/13/2021] [Indexed: 12/13/2022] Open
Abstract
Rhodopsin (RHO) misfolding mutations are a common cause of the blinding disease autosomal dominant retinitis pigmentosa (adRP). The most prevalent mutation, RHOP23H, results in its misfolding and retention in the endoplasmic reticulum (ER). Under homeostatic conditions, misfolded proteins are selectively identified, retained at the ER, and cleared via ER-associated degradation (ERAD). Overload of these degradation processes for a prolonged period leads to imbalanced proteostasis and may eventually result in cell death. ERAD of misfolded proteins, such as RHOP23H, includes the subsequent steps of protein recognition, targeting for ERAD, retrotranslocation, and proteasomal degradation. In the present study, we investigated and compared pharmacological modulation of ERAD at these four different major steps. We show that inhibition of the VCP/proteasome activity favors cell survival and suppresses P23H-mediated retinal degeneration in RHOP23H rat retinal explants. We suggest targeting this activity as a therapeutic approach for patients with currently untreatable adRP.
Collapse
|
25
|
Massengill MT, Lewin AS. Gene Therapy for Rhodopsin-associated Autosomal Dominant Retinitis Pigmentosa. Int Ophthalmol Clin 2021; 61:79-96. [PMID: 34584046 PMCID: PMC8478325 DOI: 10.1097/iio.0000000000000383] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
26
|
Lee EJ, Chan P, Chea L, Kim K, Kaufman RJ, Lin JH. ATF6 is required for efficient rhodopsin clearance and retinal homeostasis in the P23H rho retinitis pigmentosa mouse model. Sci Rep 2021; 11:16356. [PMID: 34381136 PMCID: PMC8357971 DOI: 10.1038/s41598-021-95895-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/26/2021] [Indexed: 12/21/2022] Open
Abstract
Retinitis Pigmentosa (RP) is a blinding disease that arises from loss of rods and subsequently cones. The P23H rhodopsin knock-in (P23H-KI) mouse develops retinal degeneration that mirrors RP phenotype in patients carrying the orthologous variant. Previously, we found that the P23H rhodopsin protein was degraded in P23H-KI retinas, and the Unfolded Protein Response (UPR) promoted P23H rhodopsin degradation in heterologous cells in vitro. Here, we investigated the role of a UPR regulator gene, activating transcription factor 6 (Atf6), in rhodopsin protein homeostasis in heterozygous P23H rhodopsin (Rho+/P23H) mice. Significantly increased rhodopsin protein levels were found in Atf6-/-Rho+/P23H retinas compared to Atf6+/-Rho+/P23H retinas at early ages (~ P12), while rhodopsin mRNA levels were not different. The IRE1 pathway of the UPR was hyper-activated in young Atf6-/-Rho+/P23H retinas, and photoreceptor layer thickness was unchanged at this early age in Rho+/P23H mice lacking Atf6. By contrast, older Atf6-/-Rho+/P23H mice developed significantly increased retinal degeneration in comparison to Atf6+/-Rho+/P23H mice in all retinal layers, accompanied by reduced rhodopsin protein levels. Our findings demonstrate that Atf6 is required for efficient clearance of rhodopsin protein in rod photoreceptors expressing P23H rhodopsin, and that loss of Atf6 ultimately accelerates retinal degeneration in P23H-KI mice.
Collapse
Affiliation(s)
- Eun-Jin Lee
- Department of Ophthalmology, Stanford University, Palo Alto, CA, USA.,Department of Pathology, Stanford University, Palo Alto, CA, USA.,VA Palo Alto Healthcare System, Palo Alto, CA, USA.,USC ROSKI Eye Institute and Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Priscilla Chan
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Leon Chea
- Department of Ophthalmology, Stanford University, Palo Alto, CA, USA.,Department of Pathology, Stanford University, Palo Alto, CA, USA.,VA Palo Alto Healthcare System, Palo Alto, CA, USA
| | - Kyle Kim
- Department of Ophthalmology, Stanford University, Palo Alto, CA, USA.,Department of Pathology, Stanford University, Palo Alto, CA, USA.,VA Palo Alto Healthcare System, Palo Alto, CA, USA
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Jonathan H Lin
- Department of Ophthalmology, Stanford University, Palo Alto, CA, USA. .,Department of Pathology, Stanford University, Palo Alto, CA, USA. .,VA Palo Alto Healthcare System, Palo Alto, CA, USA. .,School of Medicine, Stanford University, 300 Pasteur Dr. L235, Palo Alto, CA, 94305, USA.
| |
Collapse
|
27
|
Abstract
Inherited retinal diseases (IRDs) are an important cause of blindness worldwide. Over 270 genes have been associated with IRD. Genetic testing can determine the cause of the clinical disease in the majority of patients. However, at least 25-50% of patients with clinical diagnosis of IRD remain unsolved even after whole genome sequencing. Animal models of IRD can be useful for expanding the set of established IRD genes, to gain biological understanding of the function of these genes in the retina, and to test advanced therapeutics prior to human clinical trials. In this chapter some small and large animal models of IRD are discussed including some of the advantages and limitations of each for various forms of retinopathy.
Collapse
|
28
|
Li S, Datta S, Brabbit E, Love Z, Woytowicz V, Flattery K, Capri J, Yao K, Wu S, Imboden M, Upadhyay A, Arumugham R, Thoreson WB, DeAngelis MM, Haider NB. Nr2e3 is a genetic modifier that rescues retinal degeneration and promotes homeostasis in multiple models of retinitis pigmentosa. Gene Ther 2021; 28:223-241. [PMID: 32123325 PMCID: PMC7483267 DOI: 10.1038/s41434-020-0134-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 02/05/2020] [Accepted: 02/18/2020] [Indexed: 12/21/2022]
Abstract
Recent advances in viral vector engineering, as well as an increased understanding of the cellular and molecular mechanism of retinal diseases, have led to the development of novel gene therapy approaches. Furthermore, ease of accessibility and ocular immune privilege makes the retina an ideal target for gene therapies. In this study, the nuclear hormone receptor gene Nr2e3 was evaluated for efficacy as broad-spectrum therapy to attenuate early to intermediate stages of retinal degeneration in five unique mouse models of retinitis pigmentosa (RP). RP is a group of heterogenic inherited retinal diseases associated with over 150 gene mutations, affecting over 1.5 million individuals worldwide. RP varies in age of onset, severity, and rate of progression. In addition, ~40% of RP patients cannot be genetically diagnosed, confounding the ability to develop personalized RP therapies. Remarkably, Nr2e3 administered therapy resulted in reduced retinal degeneration as observed by increase in photoreceptor cells, improved electroretinogram, and a dramatic molecular reset of key transcription factors and associated gene networks. These therapeutic effects improved retinal homeostasis in diseased tissue. Results of this study provide evidence that Nr2e3 can serve as a broad-spectrum therapy to treat multiple forms of RP.
Collapse
Affiliation(s)
- Sujun Li
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Shyamtanu Datta
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Emily Brabbit
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Zoe Love
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Victoria Woytowicz
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Kyle Flattery
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Jessica Capri
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Katie Yao
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Siqi Wu
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | | | | | | | - Wallace B Thoreson
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Neena B Haider
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
29
|
Temporal Contrast Sensitivity Increases despite Photoreceptor Degeneration in a Mouse Model of Retinitis Pigmentosa. eNeuro 2021; 8:ENEURO.0020-21.2021. [PMID: 33509952 PMCID: PMC8059883 DOI: 10.1523/eneuro.0020-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 01/18/2021] [Indexed: 11/21/2022] Open
Abstract
The detection of temporal variations in amplitude of light intensity, or temporal contrast sensitivity (TCS), depends on the kinetics of rod photoresponse recovery. Uncharacteristically fast rod recovery kinetics are facets of both human patients and transgenic animal models with a P23H rhodopsin mutation, a prevalent cause of retinitis pigmentosa (RP). Here, we show that mice with this mutation (RhoP23H/+) exhibit an age-dependent and illumination-dependent enhancement in TCS compared with controls. At retinal illumination levels producing ≥1000 R*/rod/s or more, postnatal day 30 (P30) RhoP23H/+ mice exhibit a 1.2-fold to 2-fold increase in retinal and optomotor TCS relative to controls in response to flicker frequencies of 3, 6, and 12 Hz despite significant photoreceptor degeneration and loss of flash electroretinogram (ERG) b-wave amplitude. Surprisingly, the TCS of RhoP23H/+ mice further increases as degeneration advances. Enhanced TCS is also observed in a second model (rhodopsin heterozygous mice, Rho+/-) with fast rod recovery kinetics and no apparent retinal degeneration. In both mouse models, enhanced TCS is explained quantitatively by a comprehensive model that includes photoresponse recovery kinetics, density and collecting area of degenerating rods. Measurement of TCS may be a non-invasive early diagnostic tool indicative of rod dysfunction in some forms of retinal degenerative disease.
Collapse
|
30
|
Crane R, Conley SM, Al-Ubaidi MR, Naash MI. Gene Therapy to the Retina and the Cochlea. Front Neurosci 2021; 15:652215. [PMID: 33815052 PMCID: PMC8010260 DOI: 10.3389/fnins.2021.652215] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/22/2021] [Indexed: 12/20/2022] Open
Abstract
Vision and hearing disorders comprise the most common sensory disorders found in people. Many forms of vision and hearing loss are inherited and current treatments only provide patients with temporary or partial relief. As a result, developing genetic therapies for any of the several hundred known causative genes underlying inherited retinal and cochlear disorders has been of great interest. Recent exciting advances in gene therapy have shown promise for the clinical treatment of inherited retinal diseases, and while clinical gene therapies for cochlear disease are not yet available, research in the last several years has resulted in significant advancement in preclinical development for gene delivery to the cochlea. Furthermore, the development of somatic targeted genome editing using CRISPR/Cas9 has brought new possibilities for the treatment of dominant or gain-of-function disease. Here we discuss the current state of gene therapy for inherited diseases of the retina and cochlea with an eye toward areas that still need additional development.
Collapse
Affiliation(s)
- Ryan Crane
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Shannon M. Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Oklahoma Center for Neurosciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Muayyad R. Al-Ubaidi
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
- College of Optometry, University of Houston, Houston, TX, United States
- Depatment of Biology and Biochemistry, University of Houston, Houston, TX, United States
| | - Muna I. Naash
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
- College of Optometry, University of Houston, Houston, TX, United States
- Depatment of Biology and Biochemistry, University of Houston, Houston, TX, United States
| |
Collapse
|
31
|
Li S, Hu Y, Li Y, Hu M, Wang W, Ma Y, Cai Y, Wei M, Yao Y, Wang Y, Dong K, Gu Y, Zhao H, Bao J, Qiu Z, Zhang M, Hu X, Xue T. Generation of nonhuman primate retinitis pigmentosa model by in situ knockout of RHO in rhesus macaque retina. Sci Bull (Beijing) 2021; 66:374-385. [PMID: 36654417 DOI: 10.1016/j.scib.2020.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/11/2020] [Accepted: 07/15/2020] [Indexed: 01/20/2023]
Abstract
Retinitis pigmentosa (RP) is a form of inherited retinal degenerative diseases that ultimately involves the macula, which is present in primates but not in the rodents. Therefore, creating nonhuman primate (NHP) models of RP is of critical importance to study its mechanism of pathogenesis and to evaluate potential therapeutic options in the future. Here we applied adeno-associated virus (AAV)-delivered CRISPR/SaCas9 technology to knockout the RHO gene in the retinae of the adult rhesus macaque (Macaca mulatta) to investigate the hypothesis whether non-germline mutation of the RHO gene is sufficient to recapitulate RP. Through a series of studies, we were able to demonstrate successful somatic editing of the RHO gene and reduced RHO protein expression. More importantly, the mutant macaque retinae displayed clinical RP phenotypes, including photoreceptor degeneration, retinal thinning, abnormal rod subcellular structures, and reduced photoresponse. Therefore, we suggest somatic editing of the RHO gene is able to phenocopy RP, and the reduced time span in generating NHP mutant accelerates RP research and expands the utility of NHP model for human disease study.
Collapse
Affiliation(s)
- Shouzhen Li
- Hefei National Laboratory for Physical Sciences at the Microscale, Eye Center at The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Yingzhou Hu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650107, China
| | - Yunqin Li
- Second People's Hospital of Yunnan Province, Yunnan Eye Institute, Key Laboratory of Yunnan Province for the Prevention and Treatment of Ophthalmology, Kunming 650223, China
| | - Min Hu
- Second People's Hospital of Yunnan Province, Yunnan Eye Institute, Key Laboratory of Yunnan Province for the Prevention and Treatment of Ophthalmology, Kunming 650223, China
| | - Wenchao Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Yuqian Ma
- Hefei National Laboratory for Physical Sciences at the Microscale, Eye Center at The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Yuan Cai
- Hefei National Laboratory for Physical Sciences at the Microscale, Eye Center at The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Min Wei
- Hefei National Laboratory for Physical Sciences at the Microscale, Eye Center at The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Yichuan Yao
- Hefei National Laboratory for Physical Sciences at the Microscale, Eye Center at The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Yun Wang
- Kunming Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Kai Dong
- Hefei National Laboratory for Physical Sciences at the Microscale, Eye Center at The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Yonghao Gu
- Hefei National Laboratory for Physical Sciences at the Microscale, Eye Center at The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Huan Zhao
- Department of Biological and Environmental Engineering, Hefei University, Hefei 230601, China
| | - Jin Bao
- Hefei National Laboratory for Physical Sciences at the Microscale, Eye Center at The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Zilong Qiu
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mei Zhang
- Hefei National Laboratory for Physical Sciences at the Microscale, Eye Center at The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei 230026, China.
| | - Xintian Hu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650107, China.
| | - Tian Xue
- Hefei National Laboratory for Physical Sciences at the Microscale, Eye Center at The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
32
|
Sen M, Bassetto M, Poulhes F, Zelphati O, Ueffing M, Arango-Gonzalez B. Efficient Ocular Delivery of VCP siRNA via Reverse Magnetofection in RHO P23H Rodent Retina Explants. Pharmaceutics 2021; 13:pharmaceutics13020225. [PMID: 33562020 PMCID: PMC7914601 DOI: 10.3390/pharmaceutics13020225] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/23/2021] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
The use of synthetic RNA for research purposes as well as RNA-based therapy and vaccination has gained increasing importance. Given the anatomical seclusion of the eye, small interfering RNA (siRNA)-induced gene silencing bears great potential for targeted reduction of pathological gene expression that may allow rational treatment of chronic eye diseases in the future. However, there is yet an unmet need for techniques providing safe and efficient siRNA delivery to the retina. We used magnetic nanoparticles (MNPs) and magnetic force (Reverse Magnetofection) to deliver siRNA/MNP complexes into retinal explant tissue, targeting valosin-containing protein (VCP) previously established as a potential therapeutic target for autosomal dominant retinitis pigmentosa (adRP). Safe and efficient delivery of VCP siRNA was achieved into all retinal cell layers of retinal explants from the RHO P23H rat, a rodent model for adRP. No toxicity or microglial activation was observed. VCP silencing led to a significant decrease of retinal degeneration. Reverse Magnetofection thus offers an effective method to deliver siRNA into retinal tissue. Used in combination with retinal organotypic explants, it can provide an efficient and reliable preclinical test platform of RNA-based therapy approaches for ocular diseases.
Collapse
Affiliation(s)
- Merve Sen
- Centre of Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany;
- Graduate Training Centre of Neuroscience, University of Tübingen, 72074 Tübingen, Germany
| | - Marco Bassetto
- OZ Biosciences, Parc Scientifique de Luminy, CEDEX 9, 13288 Marseille, France; (M.B.); (F.P.); (O.Z.)
| | - Florent Poulhes
- OZ Biosciences, Parc Scientifique de Luminy, CEDEX 9, 13288 Marseille, France; (M.B.); (F.P.); (O.Z.)
| | - Olivier Zelphati
- OZ Biosciences, Parc Scientifique de Luminy, CEDEX 9, 13288 Marseille, France; (M.B.); (F.P.); (O.Z.)
| | - Marius Ueffing
- Centre of Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany;
- Correspondence: (M.U.); (B.A.-G.)
| | - Blanca Arango-Gonzalez
- Centre of Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany;
- Correspondence: (M.U.); (B.A.-G.)
| |
Collapse
|
33
|
Xu J, Zhao H, Wang T. Suppression of retinal degeneration by two novel ERAD ubiquitin E3 ligases SORDD1/2 in Drosophila. PLoS Genet 2020; 16:e1009172. [PMID: 33137101 PMCID: PMC7660902 DOI: 10.1371/journal.pgen.1009172] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 11/12/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
Mutations in the gene rhodopsin are one of the major causes of autosomal dominant retinitis pigmentosa (adRP). Mutant forms of Rhodopsin frequently accumulate in the endoplasmic reticulum (ER), cause ER stress, and trigger photoreceptor cell degeneration. Here, we performed a genome-wide screen to identify suppressors of retinal degeneration in a Drosophila model of adRP, carrying a point mutation in the major rhodopsin, Rh1 (Rh1G69D). We identified two novel E3 ubiquitin ligases SORDD1 and SORDD2 that effectively suppressed Rh1G69D-induced photoreceptor dysfunction and retinal degeneration. SORDD1/2 promoted the ubiquitination and degradation of Rh1G69D through VCP (valosin containing protein) and independent of processes reliant on the HRD1 (HMG-CoA reductase degradation protein 1)/HRD3 complex. We further demonstrate that SORDD1/2 and HRD1 function in parallel and in a redundant fashion to maintain rhodopsin homeostasis and integrity of photoreceptor cells. These findings identify a new ER-associated protein degradation (ERAD) pathway and suggest that facilitating SORDD1/2 function may be a therapeutic strategy to treat adRP. Misfolded rhodopsins accumulated in endoplasmic reticulum (ER) could disrupt the homeostasis of the ER and cause ER stress. Chronic ER stress would finally lead to photoreceptor cell death and retinal degeneration. To diminish the stress and sustain homeostasis cells develop alternative strategies to clear the misfolded rhodopsins. Previous studies have suggested that ubiquitin E3 ligase HRD1 is involved in the degradation of misfolded rhodopsins. In this study, we define novel ubiquitin E3 ligase SORDD1/2 based on a genetic screen and demonstrate that SORDD1/2 promotes the degradation of misfolded rhodopsins through ER-associated degradation (ERAD) pathway. Furthermore, we demonstrate that SORDD1/2 function independently of HRD1 in misfolded rhodopsins degradation. We also show SORDD1/2 and HRD1 play redundant roles in rhodopsin homeostasis. Finally, we demonstrate that SORDD1 works well in a Drosophila disease model. Our studies identify a novel ERAD pathway that acts in parallel to HRD1, and suggest that SORDD1 is a good candidate therapeutic target.
Collapse
Affiliation(s)
- Jaiwei Xu
- College of Biological Sciences, China Agricultural University, China
- National Institute of Biological Sciences, China
| | - Haifang Zhao
- National Institute of Biological Sciences, China
| | - Tao Wang
- National Institute of Biological Sciences, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, China
- * E-mail:
| |
Collapse
|
34
|
Genc AM, Makia MS, Sinha T, Conley SM, Al-Ubaidi MR, Naash MI. Retbindin: A riboflavin Binding Protein, Is Critical for Photoreceptor Homeostasis and Survival in Models of Retinal Degeneration. Int J Mol Sci 2020; 21:ijms21218083. [PMID: 33138244 PMCID: PMC7662319 DOI: 10.3390/ijms21218083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/19/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022] Open
Abstract
The large number of inherited retinal disease genes (IRD), including the photopigment rhodopsin and the photoreceptor outer segment (OS) structural component peripherin 2 (PRPH2), has prompted interest in identifying common cellular mechanisms involved in degeneration. Although metabolic dysregulation has been shown to play an important role in the progression of the disease etiology, identifying a common regulator that can preserve the metabolic ecosystem is needed for future development of neuroprotective treatments. Here, we investigated whether retbindin (RTBDN), a rod-specific protein with riboflavin binding capability, and a regulator of riboflavin-derived cofactors flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD), is protective to the retina in different IRD models; one carrying the P23H mutation in rhodopsin (which causes retinitis pigmentosa) and one carrying the Y141C mutation in Prph2 (which causes a blended cone-rod dystrophy). RTBDN levels are significantly upregulated in both the rhodopsin (Rho)P23H/+ and Prph2Y141C/+ retinas. Rod and cone structural and functional degeneration worsened in models lacking RTBDN. In addition, removing Rtbdn worsened other phenotypes, such as fundus flecking. Retinal flavin levels were reduced in RhoP23H/+/Rtbdn−/− and Prph2Y141C/+/Rtbdn−/− retinas. Overall, these findings suggest that RTBDN may play a protective role during retinal degenerations that occur at varying rates and due to varying disease mechanisms.
Collapse
Affiliation(s)
- Ayse M. Genc
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA; (A.M.G.); (M.S.M.); (T.S.)
| | - Mustafa S. Makia
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA; (A.M.G.); (M.S.M.); (T.S.)
| | - Tirthankar Sinha
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA; (A.M.G.); (M.S.M.); (T.S.)
| | - Shannon M. Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Oklahoma Center for Neurosciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Muayyad R. Al-Ubaidi
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA; (A.M.G.); (M.S.M.); (T.S.)
- College of Optometry, University of Houston, Houston, TX 77204, USA
- Department of Biology and Biochemistry, University of Houston, TX 77204, USA
- Correspondence: (M.R.A.-U.); (M.I.N.); Tel.: +1-713-743-1651 (M.R.A.-U. & M.I.N.); Fax: +1-713-743-0226 (M.R.A.-U. & M.I.N.)
| | - Muna I. Naash
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA; (A.M.G.); (M.S.M.); (T.S.)
- College of Optometry, University of Houston, Houston, TX 77204, USA
- Department of Biology and Biochemistry, University of Houston, TX 77204, USA
- Correspondence: (M.R.A.-U.); (M.I.N.); Tel.: +1-713-743-1651 (M.R.A.-U. & M.I.N.); Fax: +1-713-743-0226 (M.R.A.-U. & M.I.N.)
| |
Collapse
|
35
|
Sechrest ER, Murphy J, Senapati S, Goldberg AFX, Park PSH, Kolandaivelu S. Loss of PRCD alters number and packaging density of rhodopsin in rod photoreceptor disc membranes. Sci Rep 2020; 10:17885. [PMID: 33087780 PMCID: PMC7577997 DOI: 10.1038/s41598-020-74628-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/28/2020] [Indexed: 02/06/2023] Open
Abstract
Progressive rod-cone degeneration (PRCD) is a small protein localized to photoreceptor outer segment (OS) disc membranes. Several mutations in PRCD are linked to retinitis pigmentosa (RP) in canines and humans, and while recent studies have established that PRCD is required for high fidelity disc morphogenesis, its precise role in this process remains a mystery. To better understand the part which PRCD plays in disease progression as well as its contribution to photoreceptor OS disc morphogenesis, we generated a Prcd-KO animal model using CRISPR/Cas9. Loss of PRCD from the retina results in reduced visual function accompanied by slow rod photoreceptor degeneration. We observed a significant decrease in rhodopsin levels in Prcd-KO retina prior to photoreceptor degeneration. Furthermore, ultrastructural analysis demonstrates that rod photoreceptors lacking PRCD display disoriented and dysmorphic OS disc membranes. Strikingly, atomic force microscopy reveals that many disc membranes in Prcd-KO rod photoreceptor neurons are irregular, containing fewer rhodopsin molecules and decreased rhodopsin packing density compared to wild-type discs. This study strongly suggests an important role for PRCD in regulation of rhodopsin incorporation and packaging density into disc membranes, a process which, when dysregulated, likely gives rise to the visual defects observed in patients with PRCD-associated RP.
Collapse
Affiliation(s)
- Emily R Sechrest
- Department of Pharmaceutical Sciences, One Medical Center Drive, West Virginia University, Morgantown, WV, 26506-9193, USA.,Department of Ophthalmology and Visual Sciences, Eye Institute, One Medical Center Drive, West Virginia University, Morgantown, WV, 26506-9193, USA
| | - Joseph Murphy
- Department of Ophthalmology and Visual Sciences, Eye Institute, One Medical Center Drive, West Virginia University, Morgantown, WV, 26506-9193, USA.,Department of Biochemistry, One Medical Center Drive, West Virginia University, Morgantown, WV, 26506-9193, USA
| | - Subhadip Senapati
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | | | - Paul S-H Park
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Saravanan Kolandaivelu
- Department of Ophthalmology and Visual Sciences, Eye Institute, One Medical Center Drive, West Virginia University, Morgantown, WV, 26506-9193, USA. .,Department of Biochemistry, One Medical Center Drive, West Virginia University, Morgantown, WV, 26506-9193, USA.
| |
Collapse
|
36
|
Leinonen H, Pham NC, Boyd T, Santoso J, Palczewski K, Vinberg F. Homeostatic plasticity in the retina is associated with maintenance of night vision during retinal degenerative disease. eLife 2020; 9:e59422. [PMID: 32960171 PMCID: PMC7529457 DOI: 10.7554/elife.59422] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/08/2020] [Indexed: 11/18/2022] Open
Abstract
Neuronal plasticity of the inner retina has been observed in response to photoreceptor degeneration. Typically, this phenomenon has been considered maladaptive and may preclude vision restoration in the blind. However, several recent studies utilizing triggered photoreceptor ablation have shown adaptive responses in bipolar cells expected to support normal vision. Whether such homeostatic plasticity occurs during progressive photoreceptor degenerative disease to help maintain normal visual behavior is unknown. We addressed this issue in an established mouse model of Retinitis Pigmentosa caused by the P23H mutation in rhodopsin. We show robust modulation of the retinal transcriptomic network, reminiscent of the neurodevelopmental state, and potentiation of rod - rod bipolar cell signaling following rod photoreceptor degeneration. Additionally, we found highly sensitive night vision in P23H mice even when more than half of the rod photoreceptors were lost. These results suggest retinal adaptation leading to persistent visual function during photoreceptor degenerative disease.
Collapse
Affiliation(s)
- Henri Leinonen
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, IrvineIrvineUnited States
| | - Nguyen C Pham
- John A. Moran Eye Center, Department of Ophthalmology and Visual Sciences, University of UtahSalt Lake CityUnited States
| | - Taylor Boyd
- John A. Moran Eye Center, Department of Ophthalmology and Visual Sciences, University of UtahSalt Lake CityUnited States
| | - Johanes Santoso
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, IrvineIrvineUnited States
| | - Krzysztof Palczewski
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, IrvineIrvineUnited States
- Departments of Physiology and Biophysics, and Chemistry, University of California, IrvineIrvineUnited States
| | - Frans Vinberg
- John A. Moran Eye Center, Department of Ophthalmology and Visual Sciences, University of UtahSalt Lake CityUnited States
| |
Collapse
|
37
|
Aguilà M, Bellingham J, Athanasiou D, Bevilacqua D, Duran Y, Maswood R, Parfitt DA, Iwawaki T, Spyrou G, Smith AJ, Ali RR, Cheetham ME. AAV-mediated ERdj5 overexpression protects against P23H rhodopsin toxicity. Hum Mol Genet 2020; 29:1310-1318. [PMID: 32196553 PMCID: PMC7254845 DOI: 10.1093/hmg/ddaa049] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/17/2020] [Accepted: 03/17/2020] [Indexed: 02/06/2023] Open
Abstract
Rhodopsin misfolding caused by the P23H mutation is a major cause of autosomal dominant retinitis pigmentosa (adRP). To date, there are no effective treatments for adRP. The BiP co-chaperone and reductase ERdj5 (DNAJC10) is part of the endoplasmic reticulum (ER) quality control machinery, and previous studies have shown that overexpression of ERdj5 in vitro enhanced the degradation of P23H rhodopsin, whereas knockdown of ERdj5 increased P23H rhodopsin ER retention and aggregation. Here, we investigated the role of ERdj5 in photoreceptor homeostasis in vivo by using an Erdj5 knockout mouse crossed with the P23H knock-in mouse and by adeno-associated viral (AAV) vector-mediated gene augmentation of ERdj5 in P23H-3 rats. Electroretinogram (ERG) and optical coherence tomography of Erdj5-/- and P23H+/-:Erdj5-/- mice showed no effect of ERdj5 ablation on retinal function or photoreceptor survival. Rhodopsin levels and localization were similar to those of control animals at a range of time points. By contrast, when AAV2/8-ERdj5-HA was subretinally injected into P23H-3 rats, analysis of the full-field ERG suggested that overexpression of ERdj5 reduced visual function loss 10 weeks post-injection (PI). This correlated with a significant preservation of photoreceptor cells at 4 and 10 weeks PI. Assessment of the outer nuclear layer (ONL) morphology showed preserved ONL thickness and reduced rhodopsin retention in the ONL in the injected superior retina. Overall, these data suggest that manipulation of the ER quality control and ER-associated degradation factors to promote mutant protein degradation could be beneficial for the treatment of adRP caused by mutant rhodopsin.
Collapse
Affiliation(s)
| | | | | | | | - Yanai Duran
- UCL Institute of Ophthalmology, London EC1V 9EL, UK
| | - Ryea Maswood
- UCL Institute of Ophthalmology, London EC1V 9EL, UK
| | | | - Takao Iwawaki
- Division of Cell Medicine, Department of Life Science, Medical Research Institute, Kanazawa Medical University, Uchinada, 920-0293, Japan
| | - Giannis Spyrou
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, 581 83, Sweden
| | | | - Robin R Ali
- UCL Institute of Ophthalmology, London EC1V 9EL, UK
| | | |
Collapse
|
38
|
Lewis TR, Shores CR, Cady MA, Hao Y, Arshavsky VY, Burns ME. The F220C and F45L rhodopsin mutations identified in retinitis pigmentosa patients do not cause pathology in mice. Sci Rep 2020; 10:7538. [PMID: 32371886 PMCID: PMC7200662 DOI: 10.1038/s41598-020-64437-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/18/2020] [Indexed: 11/12/2022] Open
Abstract
Retinitis pigmentosa is a retinal degenerative disease that leads to blindness through photoreceptor loss. Rhodopsin is the most frequently mutated protein in this disease. While many rhodopsin mutations have well-understood consequences that lead to cell death, the disease association of several rhodopsin mutations identified in retinitis pigmentosa patients, including F220C and F45L, has been disputed. In this study, we generated two knockin mouse lines bearing each of these mutations. We did not observe any photoreceptor degeneration in either heterozygous or homozygous animals of either line. F220C mice exhibited minor disruptions of photoreceptor outer segment dimensions without any mislocalization of outer segment proteins, whereas photoreceptors of F45L mice were normal. Suction electrode recordings from individual photoreceptors of both mutant lines showed normal flash sensitivity and photoresponse kinetics. Taken together, these data suggest that neither the F220C nor F45L mutation has pathological consequences in mice and, therefore, may not be causative of retinitis pigmentosa in humans.
Collapse
Affiliation(s)
- Tylor R Lewis
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, 27710, United States
| | - Camilla R Shores
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA, 95616, United States
| | - Martha A Cady
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, 27710, United States
| | - Ying Hao
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, 27710, United States
| | - Vadim Y Arshavsky
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, 27710, United States.,Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710, United States
| | - Marie E Burns
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA, 95616, United States. .,Center for Neuroscience and Department of Ophthalmology & Vision Science, University of California, Davis, CA, 95616, United States.
| |
Collapse
|
39
|
Collin GB, Gogna N, Chang B, Damkham N, Pinkney J, Hyde LF, Stone L, Naggert JK, Nishina PM, Krebs MP. Mouse Models of Inherited Retinal Degeneration with Photoreceptor Cell Loss. Cells 2020; 9:cells9040931. [PMID: 32290105 PMCID: PMC7227028 DOI: 10.3390/cells9040931] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/05/2020] [Accepted: 04/07/2020] [Indexed: 12/12/2022] Open
Abstract
Inherited retinal degeneration (RD) leads to the impairment or loss of vision in millions of individuals worldwide, most frequently due to the loss of photoreceptor (PR) cells. Animal models, particularly the laboratory mouse, have been used to understand the pathogenic mechanisms that underlie PR cell loss and to explore therapies that may prevent, delay, or reverse RD. Here, we reviewed entries in the Mouse Genome Informatics and PubMed databases to compile a comprehensive list of monogenic mouse models in which PR cell loss is demonstrated. The progression of PR cell loss with postnatal age was documented in mutant alleles of genes grouped by biological function. As anticipated, a wide range in the onset and rate of cell loss was observed among the reported models. The analysis underscored relationships between RD genes and ciliary function, transcription-coupled DNA damage repair, and cellular chloride homeostasis. Comparing the mouse gene list to human RD genes identified in the RetNet database revealed that mouse models are available for 40% of the known human diseases, suggesting opportunities for future research. This work may provide insight into the molecular players and pathways through which PR degenerative disease occurs and may be useful for planning translational studies.
Collapse
Affiliation(s)
- Gayle B. Collin
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Navdeep Gogna
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Bo Chang
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Nattaya Damkham
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Jai Pinkney
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Lillian F. Hyde
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Lisa Stone
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Jürgen K. Naggert
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Patsy M. Nishina
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
- Correspondence: (P.M.N.); (M.P.K.); Tel.: +1-207-2886-383 (P.M.N.); +1-207-2886-000 (M.P.K.)
| | - Mark P. Krebs
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
- Correspondence: (P.M.N.); (M.P.K.); Tel.: +1-207-2886-383 (P.M.N.); +1-207-2886-000 (M.P.K.)
| |
Collapse
|
40
|
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
|
41
|
Liu X, Chen F, Chen Y, Lu H, Lu X, Peng X, Kaplan HJ, Dean DC, Gao L, Liu Y. Paracrine effects of intraocularly implanted cells on degenerating retinas in mice. Stem Cell Res Ther 2020; 11:142. [PMID: 32234075 PMCID: PMC7326149 DOI: 10.1186/s13287-020-01651-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/18/2020] [Accepted: 03/12/2020] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Retinal degeneration is a leading cause of blindness in the world; its etiology is complex and involves genetic defects and stress-associated aging. In addition to gene therapies for known genetically defective retinal degeneration, cellular therapies have been widely explored for restoring vision in both preclinical animal models and clinical trials. Stem cells of distinct tissue sources and their derived lineages have been tested for treating retinal degeneration; most of them were reported to be effective to some extent in restoring/improving deteriorated vision. Whether this visual improvement is due to a functional integration of grafted cells to substitute for lost retinal neurons in recipients or due to their neuroprotective and neurotrophic effects to retain recipient functional neurons, or both, is still under debate. METHODS We compared the results of subretinal transplantation of various somatic cell types, such as stem cells and differentiated cells, into RhoP23H/+ mice, a retinal degeneration model for human retinitis pigmentosa (RP) by evaluating their optokinetic response (OKR) and retinal histology. We identified some paracrine factors in the media that cultured cells secreted by western blotting (WB) and functionally evaluated the vascular endothelial growth factor Vegfa for its potential neurotrophic and neuroprotective effects on the neuroretina of model animals by intravitreal injection of VEGF antibody. RESULTS We found that live cells, regardless of whether they were stem cells or differentiated cell types, had a positive effect on improving degenerating retinas after subretinal transplantation; the efficacy depended on their survival duration in the host tissue. A few paracrine factors were identified in cell culture media; Vegfa was the most relevant neurotrophic and neuroprotective factor identified by our experiments to extend neuron survival duration in vivo. CONCLUSIONS Cellular therapy-produced benefits for remediating retinal degeneration are mostly, if not completely, due to a paracrine effect of implanted cells on the remaining host retinal neurons.
Collapse
Affiliation(s)
- Xiao Liu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, KY, 40202, USA
- Department of Ophthalmology, Second Xiangya Hospital of Central South University, Changsha, China
| | - Fenghua Chen
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, KY, 40202, USA
- Department of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yao Chen
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, KY, 40202, USA
- Department of Ophthalmology, Xiangya Hospital of Central South University, Changsha, China
| | - Huayi Lu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, KY, 40202, USA
- Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiaoqin Lu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, KY, 40202, USA
| | - Xiaoyan Peng
- Department of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Henry J Kaplan
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, KY, 40202, USA
| | - Douglas C Dean
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, KY, 40202, USA.
- James Graham Brown Cancer Center, Louisville, USA.
- Birth Defects Center, University of Louisville School of Medicine, Louisville, KY, USA.
| | - Ling Gao
- Department of Ophthalmology, Second Xiangya Hospital of Central South University, Changsha, China.
| | - Yongqing Liu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, KY, 40202, USA.
- James Graham Brown Cancer Center, Louisville, USA.
- Birth Defects Center, University of Louisville School of Medicine, Louisville, KY, USA.
| |
Collapse
|
42
|
Woods KN, Pfeffer J. Conformational perturbation, allosteric modulation of cellular signaling pathways, and disease in P23H rhodopsin. Sci Rep 2020; 10:2657. [PMID: 32060349 PMCID: PMC7021821 DOI: 10.1038/s41598-020-59583-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 01/30/2020] [Indexed: 02/07/2023] Open
Abstract
In this investigation we use THz spectroscopy and MD simulation to study the functional dynamics and conformational stability of P23H rhodopsin. The P23H mutation of rod opsin is the most common cause of human binding autosomal dominant retinitis pigmentosa (ADRP), but the precise mechanism by which this mutation leads to photoreceptor cell degeneration has not yet been elucidated. Our measurements confirm conformational instability in the global modes of the receptor and an active-state that uncouples the torsional dynamics of the retinal with protein functional modes, indicating inefficient signaling in P23H and a drastically altered mechanism of activation when contrasted with the wild-type receptor. Further, our MD simulations indicate that P23H rhodopsin is not functional as a monomer but rather, due to the instability of the mutant receptor, preferentially adopts a specific homodimerization motif. The preferred homodimer configuration induces structural changes in the receptor tertiary structure that reduces the affinity of the receptor for the retinal and significantly modifies the interactions of the Meta-II signaling state. We conjecture that the formation of the specific dimerization motif of P23H rhodopsin represents a cellular-wide signaling perturbation that is directly tied with the mechanism of P23H disease pathogenesis. Our results also support a direct role for rhodopsin P23H dimerization in photoreceptor rod death.
Collapse
Affiliation(s)
- Kristina N Woods
- Lehrstuhl für BioMolekulare Optik, Ludwig-Maximilians-Universität, 80538, München, Germany.
| | - Jürgen Pfeffer
- Technical University of Munich, Bavarian School of Public Policy, 80333, München, Germany
| |
Collapse
|
43
|
Orlans HO, Barnard AR, MacLaren RE. Dynamic in vivo quantification of rod photoreceptor degeneration using fluorescent reporter mouse models of retinitis pigmentosa. Exp Eye Res 2020; 190:107895. [DOI: 10.1016/j.exer.2019.107895] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/25/2019] [Accepted: 12/04/2019] [Indexed: 02/01/2023]
|
44
|
Coussa RG, Basali D, Maeda A, DeBenedictis M, Traboulsi EI. Sector retinitis pigmentosa: Report of ten cases and a review of the literature. Mol Vis 2019; 25:869-889. [PMID: 31908405 PMCID: PMC6937219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 12/28/2019] [Indexed: 11/25/2022] Open
Abstract
PURPOSE To describe the genotypes and phenotypes of ten patients with sector retinitis pigmentosa (RP). We also review previously reported mutations associated with sector RP and provide a discussion of possible underlying pathophysiological mechanisms. METHODS Patients underwent detailed ophthalmologic examinations, fundus photography, fundus autofluorescence (FAF) imaging, spectral-domain optical coherence tomography (SD-OCT), as well as visual field and electroretinographic testing. All patients underwent genetic testing to identify the molecular etiology of their disease. RESULTS A total of ten patients were studied. Among these patients, nine had mutations in RHO (c.677T>C; p.Leu226Pro (novel), c.68C>A; p.Pro23His, c.808A>C; p.Ser270Arg, c.44A>G; p.Asn15Ser, and c.325G>A; p.Gly109Arg), and one patient had a mutation in RPGR (c.3092_3093delAG; p.Glu1031Glyfs*47). All patients with missense mutations in RHO had visual acuities (VAs) better than 20/30 and showed a retained foveal ellipsoid zone and overlying retinal structures. The patient with the c.3092_3093delAG deletion in RPGR had VA of 20/60 oculus dexter (OD) and 20/400 oculus sinister (OS), as well as significant foveal thinning and contour atrophy. All patients showed pigmentary changes, or marked atrophy along the inferior arcades, or both. This pattern of degeneration corresponded to hypo- and hyperFAF and superior visual defects. CONCLUSIONS Sector RP is an uncommon form of RP in which only one or two retinal quadrants display clinical pathological signs. The great majority of cases result from mutations in RHO. The present data confirmed previously reported phenotypic manifestations of sector RP. Inferior retinal quadrants are possibly more severely affected due to greater light exposure.
Collapse
Affiliation(s)
- Razek Georges Coussa
- Center for Genetic Eye Diseases, Cole Eye Institute, Cleveland Clinic, Cleveland, OH
| | - Diana Basali
- Center for Genetic Eye Diseases, Cole Eye Institute, Cleveland Clinic, Cleveland, OH
| | - Akiko Maeda
- Department of Ophthalmology & Visual Sciences, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Meghan DeBenedictis
- Center for Genetic Eye Diseases, Cole Eye Institute, Cleveland Clinic, Cleveland, OH
| | - Elias I. Traboulsi
- Center for Genetic Eye Diseases, Cole Eye Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
45
|
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: 13] [Impact Index Per Article: 2.6] [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
|
46
|
Optical Coherence Tomography of Animal Models of Retinitis Pigmentosa: From Animal Studies to Clinical Applications. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8276140. [PMID: 31781647 PMCID: PMC6875330 DOI: 10.1155/2019/8276140] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 09/18/2019] [Indexed: 11/17/2022]
Abstract
Purpose The aim of this study was to understand the relationship between the findings of spectral-domain optical coherence tomography (SD-OCT) of previously reported animal models of retinitis pigmentosa (RP) associated with known genetic mutations and their background structural and functional changes. Methods We reviewed previous publications reporting the SD-OCT findings of animal models of RP and summarized the characteristic findings of SD-OCT in nine different animal models (RCS -/- , RHO P23H, RHO S334ter, RHO -/- , Rpe65 -/- , rp12, Pde6β -/- (rd1 and rd10), and Arr1 -/- ) of human RP. Results Despite the various abnormal structural changes found in these different animal models, progressive thinning of the outer nuclear layer (ONL) and hyperreflective change in the inner and outer segment (IS-OS) layers of the photoreceptors were commonly observed on SD-OCT. In the rapidly progressive severe photoreceptor degeneration seen in rd10 and Arr1 -/- mice, the ONL appeared hyperreflective. Electroretinography revealed various degrees of disease severity in these animal models. Discussion and Conclusion: SD-OCT is sensitive enough to detect even mild changes in the photoreceptor OS. Conversely, SD-OCT cannot qualitatively differentiate the pathologic and functional differences in the photoreceptors associated with different genetic abnormalities, with the exception of the rapid progression of severe forms of photoreceptor degeneration. These findings can be of value to understand better the clinical findings and the heterogeneous degenerative processes in patients with RP.
Collapse
|
47
|
Comitato A, Schiroli D, Montanari M, Marigo V. Calpain Activation Is the Major Cause of Cell Death in Photoreceptors Expressing a Rhodopsin Misfolding Mutation. Mol Neurobiol 2019; 57:589-599. [PMID: 31401765 DOI: 10.1007/s12035-019-01723-5] [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: 04/26/2019] [Accepted: 07/26/2019] [Indexed: 10/26/2022]
Abstract
The majority of mutations in rhodopsin (RHO) cause misfolding of the protein and has been linked to degeneration of photoreceptor cells in the retina. A lot of attention has been set on targeting ER stress for the development of new therapies for inherited retinal degeneration caused by mutations in the RHO gene. Nevertheless, the cell death pathway activated by RHO misfolded protein is still debated. In this study, we analyzed the retina of the knock-in mouse expressing the P23H misfolded mutant RHO. We found persistent unfolded protein response (UPR) during degeneration. Interestingly, long-term stimulation of the PERK branch of ER stress had a protective effect by phosphorylating nuclear factor erythroid 2-related factor 2 (NRF2) transcription factor, associated with antioxidant responses. Otherwise, we provide evidence that increased intracellular calcium and activation of calpains strongly correlated with rod photoreceptor cell death. By blocking calpain activity, we significantly decreased the activation of caspase-7 and apoptosis-inducing factor (AIF), two cell death effectors, and cell demise, and effectively protected the retina from degeneration caused by the P23H dominant mutation in RHO.
Collapse
Affiliation(s)
- Antonella Comitato
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi, 287, 41125, Modena, Italy
| | - Davide Schiroli
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi, 287, 41125, Modena, Italy
| | - Monica Montanari
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi, 287, 41125, Modena, Italy
| | - Valeria Marigo
- Department of Life Sciences, University of Modena and Reggio Emilia, via Campi, 287, 41125, Modena, Italy.
| |
Collapse
|
48
|
Qiu Y, Yao J, Jia L, Thompson DA, Zacks DN. Shifting the balance of autophagy and proteasome activation reduces proteotoxic cell death: a novel therapeutic approach for restoring photoreceptor homeostasis. Cell Death Dis 2019; 10:547. [PMID: 31320609 PMCID: PMC6639359 DOI: 10.1038/s41419-019-1780-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/26/2019] [Accepted: 06/27/2019] [Indexed: 12/13/2022]
Abstract
The P23H variant of rhodopsin results in misfolding of the protein, and is a common cause of the blinding disease autosomal dominant retinitis pigmentosa (adRP). We have recently demonstrated that degeneration of photoreceptor cells in retinas of P23H mice is due to the endoplasmic reticulum stress (ERS)-induced activation of autophagy that leads to a secondary proteasome insufficiency and activation of cell death pathways. We propose that this increased level of autophagy flux relative to proteasome activity, which we term the A:P ratio, represents a marker of altered photoreceptor cell homeostasis, and that therapies aimed at normalizing this ratio will result in increased photoreceptor cell survival. To test this postulate, we treated P23H mice with a chemical chaperone (4-phenylbutyric acid) to improve rhodopsin folding, or with a selective phosphodiesterase-4 inhibitor (rolipram) to increase proteasome activity. P23H mice treated with either of these agents exhibited reduced ERS, decreased autophagy flux, increased proteasome activity, and decreased activation of cell death pathways. In addition, rates of retinal degeneration were decreased, and photoreceptor morphology and visual function were preserved. These findings support the conclusion that normalizing the A:P ratio, either by reducing the ERS-induced activation of autophagy, or by increasing proteasome activity, improves photoreceptor survival, and suggest a potential new therapeutic strategy for the treatment of adRP caused by protein folding defects.
Collapse
Affiliation(s)
- Yaoyan Qiu
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, MI, USA.,Department of Ophthalmology, Xiangya School of medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jingyu Yao
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, MI, USA
| | - Lin Jia
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, MI, USA
| | - Debra A Thompson
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, MI, USA.,Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, USA
| | - David N Zacks
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, MI, USA.
| |
Collapse
|
49
|
Ropelewski P, Imanishi Y. Disrupted Plasma Membrane Protein Homeostasis in a Xenopus Laevis Model of Retinitis Pigmentosa. J Neurosci 2019; 39:5581-5593. [PMID: 31061086 PMCID: PMC6616295 DOI: 10.1523/jneurosci.3025-18.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/23/2019] [Accepted: 04/26/2019] [Indexed: 12/17/2022] Open
Abstract
Rhodopsin mislocalization is frequently observed in retinitis pigmentosa (RP) patients. For example, class I mutant rhodopsin is deficient in the VxPx trafficking signal, mislocalizes to the plasma membrane (PM) of rod photoreceptor inner segments (ISs), and causes autosomal dominant RP. Mislocalized rhodopsin causes photoreceptor degeneration in a manner independent of light-activation. In this manuscript, we took advantage of Xenopus laevis models of both sexes expressing wild-type human rhodopsin or its class I Q344ter mutant fused to Dendra2 fluorescent protein to characterize a novel light-independent mechanism of photoreceptor degeneration caused by mislocalized rhodopsin. We found that rhodopsin mislocalized to the PM is actively internalized and transported to lysosomes where it is degraded. This degradation process results in the downregulation of a crucial component of the photoreceptor IS PM: the sodium-potassium ATPase α-subunit (NKAα). The downregulation of NKAα is not because of decreased NKAα mRNA, but due to cotransport of mislocalized rhodopsin and NKAα to lysosomes or autophagolysosomes. In a separate set of experiments, we found that class I mutant rhodopsin, which causes NKAα downregulation, also causes shortening and loss of rod outer segments (OSs); the symptoms frequently observed in the early stages of human RP. Likewise, pharmacological inhibition of NKAα led to shortening and loss of rod OSs. These combined studies suggest that mislocalized rhodopsin leads to photoreceptor dysfunction through disruption of the PM protein homeostasis and compromised NKAα function. This study unveiled a novel role of lysosome-mediated degradation in causing inherited disorders manifested by mislocalization of ciliary receptors.SIGNIFICANCE STATEMENT Retinal ciliopathy is the most common form of inherited blinding disorder frequently manifesting rhodopsin mislocalization. Our understanding of the relationships between rhodopsin mislocalization and photoreceptor dysfunction/degeneration has been far from complete. This study uncovers a hitherto uncharacterized consequence of rhodopsin mislocalization: the activation of the lysosomal pathway, which negatively regulates the amount of the sodium-potassium ATPase (NKAα) on the inner segment plasma membrane. On the plasma membrane, mislocalized rhodopsin extracts NKAα and sends it to lysosomes where they are co-degraded. Compromised NKAα function leads to shortening and loss of the photoreceptor outer segments as observed for various inherited blinding disorders. In summary, this study revealed a novel pathogenic mechanism applicable to various forms of blinding disorders caused by rhodopsin mislocalization.
Collapse
Affiliation(s)
- Philip Ropelewski
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106-4965
| | - Yoshikazu Imanishi
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106-4965
| |
Collapse
|
50
|
Bocchero U, Tam BM, Chiu CN, Torre V, Moritz OL. Electrophysiological Changes During Early Steps of Retinitis Pigmentosa. Invest Ophthalmol Vis Sci 2019; 60:933-943. [PMID: 30840038 DOI: 10.1167/iovs.18-25347] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The rhodopsin mutation P23H is responsible for a significant portion of autosomal-dominant retinitis pigmentosa, a disorder characterized by rod photoreceptor death. The mechanisms of toxicity remain unclear; previous studies implicate destabilization of P23H rhodopsin during light exposure, causing decreased endoplasmic reticulum (ER) exit and ER stress responses. Here, we probed phototransduction in Xenopus laevis rods expressing bovine P23H rhodopsin, in which retinal degeneration is inducible by light exposure, in order to examine early physiological changes that occur during retinal degeneration. Methods We recorded single-cell and whole-retina responses to light stimuli using electrophysiology. Moreover, we monitored morphologic changes in rods after different periods of light exposure. Results Initially, P23H rods had almost normal photoresponses, but following a brief light exposure varying from 4 to 32 photoisomerizations per disc, photoresponses became irreversibly prolonged. In intact retinas, rods began to shed OS fragments after a rod-saturating exposure of 12 minutes, corresponding to approximately 10 to 100 times more photoisomerizations. Conclusions Our results indicate that in P23H rods light-induced degeneration occurs in at least two stages, the first involving impairment of phototransduction and the second involving initiation of morphologic changes.
Collapse
Affiliation(s)
- Ulisse Bocchero
- Neuroscience Department, International School for Advanced Studies (SISSA), Trieste, Italy
| | - Beatrice M Tam
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Colette N Chiu
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Vincent Torre
- Neuroscience Department, International School for Advanced Studies (SISSA), Trieste, Italy
| | - Orson L Moritz
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|