1
|
Thomas BB, Rajendran Nair DS, Rahimian M, Hassan AK, Tran TL, Seiler MJ. Animal models for the evaluation of retinal stem cell therapies. Prog Retin Eye Res 2025; 106:101356. [PMID: 40239758 DOI: 10.1016/j.preteyeres.2025.101356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 04/18/2025]
Abstract
Retinal degeneration (RD) diseases leading to severe vision loss can affect photoreceptors (PRs) that are responsible for phototransduction, or retinal pigmented epithelium (RPE) providing support for PRs. Human pluripotent stem cell (hPSC)-based therapies are a potential approach for restoration of retinal structure in patients with currently incurable RD diseases. Currently, there are two targeted hPSC therapeutics: PR rescue and PR replacement. PR rescue involves the transplantation of RPE or other neural progenitors into the subretinal space to slow down or prevent further RD. RPE transplantation plays a critical role in preserving photoreceptors by providing trophic support and maintaining retinal integrity, particularly in diseases like age-related macular degeneration (AMD). Advances in RPE transplantation methods, such as polarized monolayer cultures and scaffold-based approaches, have shown promise in enhancing graft survival and integration. However, limitations include inconsistent integration, variable neurotrophic factor secretion, and immune rejection risks in non-autologous transplants. In PR replacement, stem cell-derived photoreceptor-like cells or photoreceptor progenitors (PRP) obtained are transplanted into the eye. While PRPs are commonly obtained from retinal organoids (ROs), alternative sources, such as early differentiation stages or direct differentiation protocols, are also utilized to enhance the efficiency and scalability of PRP generation. Challenges include achieving proper integration, forming outer segments, rosette formation, and avoiding immune rejection or tumorigenicity. Various animal models that simulate human RD diseases are being used for establishing surgical feasibility, graft survival and visual functional recovery but fail to replicate clinical immune challenges. Rodent models lack macula-like structures and have limited reliability in detecting subtle functional changes, while larger animal models pose ethical, logistical, and financial challenges. Immunocompromised models have been developed for minimizing xenograft issues. Visual functional testing for efficacy includes optokinetic testing (OKN), electroretinography (ERG), and electrophysiological recordings from the retina and brain. These tests often fail to capture the complexity of human visual recovery, highlighting the need for advanced models and improved functional testing techniques. This review aims to aggregate current knowledge about approaches to stem cell transplantation, requirements of animal models chosen for validating vision benefits of transplantation studies, advantages of using specific disease models and their limitations. While promising strides have been made, addressing these limitations remains essential for translating stem cell-based therapies into clinical success.
Collapse
Affiliation(s)
- Biju B Thomas
- Department of Ophthalmology, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, United States; USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, CA, United States
| | - Deepthi S Rajendran Nair
- Department of Ophthalmology, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, United States
| | - Mana Rahimian
- Department of Ophthalmology, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, United States
| | - Amr K Hassan
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, Irvine CA, United States
| | - Thuy-Linh Tran
- Stem Cell Research Center, University of California, Irvine, Irvine, CA, United States
| | - Magdalene J Seiler
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, Irvine CA, United States; Stem Cell Research Center, University of California, Irvine, Irvine, CA, United States; Department of Physical Medicine and Rehabilitation, University of California, Irvine, Irvine, CA, United States; Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, United States; Center for Translational Vision Research, University of California, Irvine, Irvine, CA, United States.
| |
Collapse
|
2
|
Wong WM, Mahroo OA. Monogenic Retinal Diseases Associated With Genes Encoding Phototransduction Proteins: A Review. Clin Exp Ophthalmol 2025; 53:260-280. [PMID: 40013354 PMCID: PMC11962696 DOI: 10.1111/ceo.14511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/29/2025] [Accepted: 02/17/2025] [Indexed: 02/28/2025]
Abstract
Phototransduction, the process by which captured photons elicit electrical changes in retinal rod and cone cells, represents the first neuronal step in vision and involves interactions between several highly specialised proteins. Pathogenic variants in genes encoding many of these proteins can give rise to significant vision impairment, accounting for a substantial portion of inherited retinal disease. Such genes include RHO, OPN1LW, OPN1MW, GNAT1, GNAT2, GNB3, PDE6A, PDE6B, PDE6G, PDE6C, PDE6H, CNGA1, CNGB1, CNGA3, CNGB3, GRK1, SAG, ARR3, RGS9, RGS9BP, GUCY2D, GUCA1A and SLC24A1. Many of these conditions have distinct mechanisms and clinical features. They follow several modes of inheritance (including in one case digenic, or tri-allelic, inheritance). Some conditions also entail myopia. Rod and cone phototransduction will be outlined, followed by the discussion of diseases associated with these genes. Some phenotypic features will be highlighted as well as their prevalence in a large genotyped inherited retinal disease cohort.
Collapse
Affiliation(s)
- Wendy M. Wong
- Institute of Ophthalmology, University College LondonLondonUK
- NIHR Biomedical Research Centre at Moorfields Eye Hospital and the UCL Institute of OphthalmologyLondonUK
- Centre for Innovation & Precision Eye Health, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Ophthalmology, National University HospitalNational University Health SystemSingaporeSingapore
| | - Omar A. Mahroo
- Institute of Ophthalmology, University College LondonLondonUK
- NIHR Biomedical Research Centre at Moorfields Eye Hospital and the UCL Institute of OphthalmologyLondonUK
- Section of Ophthalmology, King's College LondonSt Thomas' Hospital CampusLondonUK
- Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
- Department of Translational OphthalmologyWills Eye HospitalPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
3
|
Corsi F, Galante A, Maggi MA, Mazziotti R, Bisti S, Piano I, Gargini C. The efficacy of Saffron Repron® in counteracting the progression of retinitis pigmentosa: Neuroprotection and resilience. Asia Pac J Ophthalmol (Phila) 2025:100192. [PMID: 40122468 DOI: 10.1016/j.apjo.2025.100192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/13/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025] Open
Abstract
The mutations observed in the various forms of retinitis pigmentosa (RP) affect genes coding for rod-specific proteins and direct the progression of different forms of dystrophy toward total blindness. The present investigation aims to explore the protective effects of Saffron Repron®, in a mouse model of RP. Saffron was administered orally to pregnant females and weaned pups for 120 days. At different time points (P30, P60, P90, and P120), visual function, retinal function, cone lifespan, morphology, gene expression, and protein level were analyzed. The results indicate that chronic saffron treatment effectively slows the progression of long-term damage caused by genetic mutations in both the morphology and function of retinal neurons. Cellular mechanisms responsible for this action appear complex and, probably, due to coordinated and synergistic activities by its chemical components. Here we provide evidence that saffron is able to modulate the epigenetic pathway involved in neuroinflammation. Biochemical and molecular measures suggest that early saffron treatment may induce a form of adaptation known as acquired resilience.
Collapse
Affiliation(s)
- Francesca Corsi
- University of Pisa, Department of Pharmacy, Italy; National Institute of Biostructure and Biosystem (INBB), Italy
| | | | - Maria Anna Maggi
- Hortus Novus srl, via Campo Sportivo 2, Italy; Department of Physical and Chemical Sciences, University of L'Aquila, Italy
| | - Raffaele Mazziotti
- Department of Neuroscience, Psychology, Pharmacology, and Child Health, University of Florence, Italy; Institute of Neuroscience, National Research Council (CNR), Italy
| | - Silvia Bisti
- National Institute of Biostructure and Biosystem (INBB), Italy
| | - Ilaria Piano
- University of Pisa, Department of Pharmacy, Italy; National Institute of Biostructure and Biosystem (INBB), Italy.
| | - Claudia Gargini
- University of Pisa, Department of Pharmacy, Italy; National Institute of Biostructure and Biosystem (INBB), Italy
| |
Collapse
|
4
|
Singh A, Ratnapriya R. Integration of multiomic data identifies core-module of inherited-retinal diseases. Hum Mol Genet 2025; 34:454-465. [PMID: 39797395 DOI: 10.1093/hmg/ddaf001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/20/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025] Open
Abstract
Human diseases with similar phenotypes can be interconnected through shared biological pathways, genes, or molecular mechanisms. Inherited retinal diseases (IRDs) cause photoreceptor dysfunction due to mutations in approximately 300 genes, affecting visual transduction, photoreceptor morphogenesis, and transcription factors, suggesting common pathobiological mechanisms. This study examined the functional relationship between known IRDs genes by integrating binding sites and gene expression data from the key photoreceptor transcription factors (TFs), Crx and Nrl. We show that the targets of these TFs were enriched in IRDs causal genes. Co-expression network analysis revealed that IRD-centric networks were disrupted when Crx and Nrl were knocked out. Finally, we identified a highly connected core module comprising 14 IRD and 39 target genes, of which 29 were dysregulated in the rod photoreceptors of the four IRD mouse models. These findings offer a network-based interpretation of IRDs, aiding in the identification of common mechanisms, prioritizing genes for novel disease gene identification, and informing the development of gene-agnostic therapies for IRDs.
Collapse
Affiliation(s)
- Ajeet Singh
- Department of Ophthalmology, Baylor College of Medicine, 6565 Fannin St, NC205, Houston, TX 77030 United States
| | - Rinki Ratnapriya
- Department of Ophthalmology, Baylor College of Medicine, 6565 Fannin St, NC205, Houston, TX 77030 United States
- Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, 6565 Fannin St, NC205, Houston, TX 77030 United States
| |
Collapse
|
5
|
Dey PN, Singh N, Zelinger L, Batz Z, Nellissery J, White Carreiro ND, Qian H, Li T, Fariss RN, Dong L, Swaroop A. Loss of paired immunoglobin-like type 2 receptor B gene associated with age-related macular degeneration impairs photoreceptor function in mouse retina. Hum Mol Genet 2025; 34:64-76. [PMID: 39532089 PMCID: PMC12034095 DOI: 10.1093/hmg/ddae161] [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: 08/10/2024] [Revised: 10/10/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Genome-wide association studies have uncovered mostly non-coding variants at over 60 genetic loci linked to susceptibility for age-related macular degeneration (AMD). To ascertain the causal gene at the PILRB/PILRA locus, we used a CRISPR strategy to produce germline deletions in the mouse paired immunoglobin-like type 2 receptor (Pilr) genes that encode highly related activating (PILRB) and inhibitory (PILRA) receptors. We show that a combined loss of Pilrb1 and Pilrb2, but not Pilra, leads to an early but relatively stationary defect as the electroretinography (ERG) amplitudes of Pilrb1/2-/- mice exhibit a marked reduction as early as postnatal day 15 and do not show additional significant decrease at 3 and 12-months. No alterations are evident in Müller glia, microglia, bipolar, amacrine and horizontal cells based on immunohistochemistry using cell-type specific markers. PILRB immunostaining is specifically detected at the proximal part of photoreceptor outer segment. Reduced expression of select calcium-regulated phototransduction and synapse-associated proteins, including GCAP1 and 2, PDE6b, AIPL1, PSD95, and CTBP1 indicates dysregulation of calcium homeostasis as a possible mechanism of retinal phenotype in Pilrb1/2-/- mice. Our studies suggest a novel function of PILRB in retinal photoreceptors and an association of PILRB, but not PILRA, with AMD pathogenesis.
Collapse
Affiliation(s)
- Partha Narayan Dey
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| | - Nivedita Singh
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| | - Lina Zelinger
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| | - Zachary Batz
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| | - Jacob Nellissery
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| | - Noor D White Carreiro
- Biological Imaging Core, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| | - Haohua Qian
- Visual Function Core Facility, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| | - Tiansen Li
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| | - Robert N Fariss
- Biological Imaging Core, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| | - Lijin Dong
- Genetic Engineering Core, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| | - Anand Swaroop
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| |
Collapse
|
6
|
Valdés-Sánchez L, Moshtaghion SM, Caballano-Infantes E, Peñalver P, Rodríguez-Ruiz R, González-Alfonso JL, Plou FJ, Desmet T, Morales JC, Díaz-Corrales FJ. Synthesis and Evaluation of Glucosyl-, Acyl- and Silyl- Resveratrol Derivatives as Retinoprotective Agents: Piceid Octanoate Notably Delays Photoreceptor Degeneration in a Retinitis Pigmentosa Mouse Model. Pharmaceuticals (Basel) 2024; 17:1482. [PMID: 39598393 PMCID: PMC11597447 DOI: 10.3390/ph17111482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Retinitis pigmentosa (RP), the leading cause of inherited blindness in adults, is marked by the progressive degeneration of rod photoreceptors in the retina. While gene therapy has shown promise in treating RP in patients with specific mutations, no effective therapies currently exist for the majority of patients with diverse genetic backgrounds. Additionally, no intervention can yet prevent or delay photoreceptor loss across the broader RP patient population. Resveratrol (RES), a naturally occurring polyphenol, has shown cytoprotective effects in various neurodegenerative disease models; however, its therapeutic potential is limited by low bioavailability. METHODS In this study, we synthesized novel RES derivatives and assessed their retinoprotective effects in a murine model of RP (rd10 mice). RESULTS Among these derivatives, piceid octanoate (PIC-OCT) significantly delayed photoreceptor degeneration in the RP model, demonstrating superior efficacy compared to RES. CONCLUSIONS PIC-OCT shows strong potential as a leading candidate for developing new therapeutic strategies for RP.
Collapse
Affiliation(s)
- Lourdes Valdés-Sánchez
- Department of Integrative Pathophysiology and Therapies, Andalusian Molecular Biology and Regenerative Medicine Centre (CABIMER), Junta de Andalucía, CSIC, Universidad de Sevilla, Universidad Pablo de Olavide, Avda. Américo Vespucio 24, 41092 Seville, Seville, Spain; (L.V.-S.); (S.M.M.); (E.C.-I.)
| | - Seyed Mohamadmehdi Moshtaghion
- Department of Integrative Pathophysiology and Therapies, Andalusian Molecular Biology and Regenerative Medicine Centre (CABIMER), Junta de Andalucía, CSIC, Universidad de Sevilla, Universidad Pablo de Olavide, Avda. Américo Vespucio 24, 41092 Seville, Seville, Spain; (L.V.-S.); (S.M.M.); (E.C.-I.)
| | - Estefanía Caballano-Infantes
- Department of Integrative Pathophysiology and Therapies, Andalusian Molecular Biology and Regenerative Medicine Centre (CABIMER), Junta de Andalucía, CSIC, Universidad de Sevilla, Universidad Pablo de Olavide, Avda. Américo Vespucio 24, 41092 Seville, Seville, Spain; (L.V.-S.); (S.M.M.); (E.C.-I.)
| | - Pablo Peñalver
- Department of Biochemistry and Molecular Pharmacology, Instituto de Parasitología y Biomedicina López Neyra, CSIC, PTS Granada, Avda. del Conocimiento, 17, 18016 Armilla, Granada, Spain; (P.P.); (R.R.-R.)
| | - Rosario Rodríguez-Ruiz
- Department of Biochemistry and Molecular Pharmacology, Instituto de Parasitología y Biomedicina López Neyra, CSIC, PTS Granada, Avda. del Conocimiento, 17, 18016 Armilla, Granada, Spain; (P.P.); (R.R.-R.)
| | - José Luis González-Alfonso
- Instituto de Catálisis y Petroleoquímica, CSIC, Marie Curie 2, 28049 Madrid, Madrid, Spain; (J.L.G.-A.) (F.J.P.)
| | - Francisco José Plou
- Instituto de Catálisis y Petroleoquímica, CSIC, Marie Curie 2, 28049 Madrid, Madrid, Spain; (J.L.G.-A.) (F.J.P.)
| | - Tom Desmet
- Centre for Synthetic Biology, Department of Biotechnology, Ghent University, Coupure Links 653, 9000 Ghent, Belgium;
| | - Juan C. Morales
- Department of Biochemistry and Molecular Pharmacology, Instituto de Parasitología y Biomedicina López Neyra, CSIC, PTS Granada, Avda. del Conocimiento, 17, 18016 Armilla, Granada, Spain; (P.P.); (R.R.-R.)
| | - Francisco J. Díaz-Corrales
- Department of Integrative Pathophysiology and Therapies, Andalusian Molecular Biology and Regenerative Medicine Centre (CABIMER), Junta de Andalucía, CSIC, Universidad de Sevilla, Universidad Pablo de Olavide, Avda. Américo Vespucio 24, 41092 Seville, Seville, Spain; (L.V.-S.); (S.M.M.); (E.C.-I.)
| |
Collapse
|
7
|
Montaser AB, Gao F, Peters D, Vainionpää K, Zhibin N, Skowronska-Krawczyk D, Figeys D, Palczewski K, Leinonen H. Retinal Proteome Profiling of Inherited Retinal Degeneration Across Three Different Mouse Models Suggests Common Drug Targets in Retinitis Pigmentosa. Mol Cell Proteomics 2024; 23:100855. [PMID: 39389360 PMCID: PMC11602984 DOI: 10.1016/j.mcpro.2024.100855] [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: 06/14/2024] [Revised: 09/14/2024] [Accepted: 10/06/2024] [Indexed: 10/12/2024] Open
Abstract
Inherited retinal degenerations (IRDs) are a leading cause of blindness among the population of young people in the developed world. Approximately half of IRDs initially manifest as gradual loss of night vision and visual fields, characteristic of retinitis pigmentosa (RP). Due to challenges in genetic testing, and the large heterogeneity of mutations underlying RP, targeted gene therapies are an impractical largescale solution in the foreseeable future. For this reason, identifying key pathophysiological pathways in IRDs that could be targets for mutation-agnostic and disease-modifying therapies (DMTs) is warranted. In this study, we investigated the retinal proteome of three distinct IRD mouse models, in comparison to sex- and age-matched wild-type mice. Specifically, we used the Pde6βRd10 (rd10) and RhoP23H/WT (P23H) mouse models of autosomal recessive and autosomal dominant RP, respectively, as well as the Rpe65-/- mouse model of Leber's congenital amaurosis type 2 (LCA2). The mice were housed at two distinct institutions and analyzed using LC-MS in three separate facilities/instruments following data-dependent and data-independent acquisition modes. This cross-institutional and multi-methodological approach signifies the reliability and reproducibility of the results. The large-scale profiling of the retinal proteome, coupled with in vivo electroretinography recordings, provided us with a reliable basis for comparing the disease phenotypes and severity. Despite evident inflammation, cellular stress, and downscaled phototransduction observed consistently across all three models, the underlying pathologies of RP and LCA2 displayed many differences, sharing only four general KEGG pathways. The opposite is true for the two RP models in which we identify remarkable convergence in proteomic phenotype even though the mechanism of primary rod death in rd10 and P23H mice is different. Our data highlights the cAMP and cGMP second-messenger signaling pathways as potential targets for therapeutic intervention. The proteomic data is curated and made publicly available, facilitating the discovery of universal therapeutic targets for RP.
Collapse
Affiliation(s)
- Ahmed B Montaser
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland.
| | - Fangyuan Gao
- Center for Translational Vision Research, Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, California, USA; Department of Physiology and Biophysics, University of California, Irvine, California, USA
| | - Danielle Peters
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Katri Vainionpää
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Ning Zhibin
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Dorota Skowronska-Krawczyk
- Center for Translational Vision Research, Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, California, USA; Department of Physiology and Biophysics, University of California, Irvine, California, USA
| | - Daniel Figeys
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Krzysztof Palczewski
- Center for Translational Vision Research, Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, California, USA; Department of Physiology and Biophysics, University of California, Irvine, California, USA; Department of Chemistry, University of California, Irvine, California, USA; Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
| | - Henri Leinonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
8
|
Leinonen H, Zhang J, Occelli LM, Seemab U, Choi EH, L P Marinho LF, Querubin J, Kolesnikov AV, Galinska A, Kordecka K, Hoang T, Lewandowski D, Lee TT, Einstein EE, Einstein DE, Dong Z, Kiser PD, Blackshaw S, Kefalov VJ, Tabaka M, Foik A, Petersen-Jones SM, Palczewski K. A combination treatment based on drug repurposing demonstrates mutation-agnostic efficacy in pre-clinical retinopathy models. Nat Commun 2024; 15:5943. [PMID: 39009597 PMCID: PMC11251169 DOI: 10.1038/s41467-024-50033-5] [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: 08/11/2023] [Accepted: 06/21/2024] [Indexed: 07/17/2024] Open
Abstract
Inherited retinopathies are devastating diseases that in most cases lack treatment options. Disease-modifying therapies that mitigate pathophysiology regardless of the underlying genetic lesion are desirable due to the diversity of mutations found in such diseases. We tested a systems pharmacology-based strategy that suppresses intracellular cAMP and Ca2+ activity via G protein-coupled receptor (GPCR) modulation using tamsulosin, metoprolol, and bromocriptine coadministration. The treatment improves cone photoreceptor function and slows degeneration in Pde6βrd10 and RhoP23H/WT retinitis pigmentosa mice. Cone degeneration is modestly mitigated after a 7-month-long drug infusion in PDE6A-/- dogs. The treatment also improves rod pathway function in an Rpe65-/- mouse model of Leber congenital amaurosis but does not protect from cone degeneration. RNA-sequencing analyses indicate improved metabolic function in drug-treated Rpe65-/- and rd10 mice. Our data show that catecholaminergic GPCR drug combinations that modify second messenger levels via multiple receptor actions provide a potential disease-modifying therapy against retinal degeneration.
Collapse
Affiliation(s)
- Henri Leinonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, 70211, Kuopio, Finland.
| | - Jianye Zhang
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA
| | - Laurence M Occelli
- Small Animal Clinical Sciences, Michigan State University, East Lansing, MI, 48824, USA
| | - Umair Seemab
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, 70211, Kuopio, Finland
| | - Elliot H Choi
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA
| | | | - Janice Querubin
- Small Animal Clinical Sciences, Michigan State University, East Lansing, MI, 48824, USA
| | - Alexander V Kolesnikov
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA
| | - Anna Galinska
- International Centre for Translational Eye Research, Warsaw, Poland
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Kordecka
- International Centre for Translational Eye Research, Warsaw, Poland
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Thanh Hoang
- Department of Ophthalmology, Department of Cell & Developmental Biology, Ann Arbor, MI, 48105, USA
| | - Dominik Lewandowski
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA
| | - Timothy T Lee
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA
| | - Elliott E Einstein
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA
| | - David E Einstein
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA
| | - Zhiqian Dong
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA
| | - Philip D Kiser
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA
- Department of Physiology and Biophysics, School of Medicine, University of California - Irvine, Irvine, CA, 92697, USA
- Department of Clinical Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University of California - Irvine, Irvine, CA, 92697, USA
- Research Service, VA Long Beach Healthcare System, Long Beach, California, 90822, USA
| | - Seth Blackshaw
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Vladimir J Kefalov
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA
- Department of Physiology and Biophysics, School of Medicine, University of California - Irvine, Irvine, CA, 92697, USA
| | - Marcin Tabaka
- International Centre for Translational Eye Research, Warsaw, Poland
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Andrzej Foik
- International Centre for Translational Eye Research, Warsaw, Poland
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | | | - Krzysztof Palczewski
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA.
- Department of Physiology and Biophysics, School of Medicine, University of California - Irvine, Irvine, CA, 92697, USA.
- Department of Chemistry, University of California-Irvine, Irvine, CA, 92697, USA.
- Department of Molecular Biology and Biochemistry, University of California-Irvine, Irvine, CA, 92697, USA.
- Gavin Herbert Eye Institute-Center for Translational Vision Research, Department of Ophthalmology, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
9
|
Szabó K, Dékány B, Énzsöly A, Hajdú RI, Laurik-Feuerstein LK, Szabó A, Radovits T, Mátyás C, Oláh A, Kovács KA, Szél Á, Somfai GM, Lukáts Á. Possible retinotoxicity of long-term vardenafil treatment. Exp Eye Res 2024; 243:109890. [PMID: 38615833 DOI: 10.1016/j.exer.2024.109890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 03/10/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
Phosphodiesterase (PDE) inhibitors - such as vardenafil - are used primarily for treating erectile dysfunction via increasing cyclic guanosine monophosphate (cGMP) levels. Recent studies have also demonstrated their significant cardioprotective effects in several diseases, including diabetes, upon long-term, continuous application. However, PDE inhibitors are not specific for PDE5 and also inhibit the retinal isoform. A sustained rise in cGMP in photoreceptors is known to be toxic; therefore, we hypothesized that long-term vardenafil treatment might result in retinotoxicity. The hypothesis was tested in a clinically relevant animal model of type 2 diabetes mellitus. Histological experiments were performed on lean and diabetic Zucker Diabetic Fatty rats. Half of the animals were treated with vardenafil for six months, and the retinal effects were evaluated. Vardenafil treatment alleviated rod outer segment degeneration but decreased rod numbers in some positions and induced changes in the interphotoreceptor matrix, even in control animals. Vardenafil treatment decreased total retinal thickness in the control and diabetic groups and reduced the number of nuclei in the outer nuclear layer. Müller cell activation was detectable even in the vardenafil-treated control animals, and vardenafil did not improve gliosis in the diabetic group. Vardenafil-treated animals showed complex retinal alterations with improvements in some parameters while deterioration in others. Our results point towards the retinotoxicity of vardenafil, even without diabetes, which raises doubts about the retinal safety of long-term continuous vardenafil administration. This effect needs to be considered when approving PDE inhibitors for alternative indications.
Collapse
Affiliation(s)
- Klaudia Szabó
- Institute of Education and Psychology at Szombathely, Faculty of Education and Psychology, ELTE Eötvös Loránd University, Szombathely, Hungary; Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Bulcsú Dékány
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Anna Énzsöly
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary; Department of Ophthalmology, Semmelweis University, Budapest, Hungary
| | - Rozina Ida Hajdú
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary; Department of Ophthalmology, Semmelweis University, Budapest, Hungary; Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Arnold Szabó
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Tamás Radovits
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Csaba Mátyás
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Attila Oláh
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Krisztián András Kovács
- Institute of Translational Medicine, Translational Retina Research Group, Semmelweis University, Budapest, Hungary
| | - Ágoston Szél
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Gábor Márk Somfai
- Department of Ophthalmology, Semmelweis University, Budapest, Hungary; Spross Research Institute, Zurich, Switzerland; Department of Ophthalmology, Stadtspital Zurich, Zurich, Switzerland
| | - Ákos Lukáts
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary; Institute of Translational Medicine, Translational Retina Research Group, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
10
|
Pfeffer ME, DiFrancesco ML, Marchesi A, Galluzzi F, Moschetta M, Rossini A, Francia S, Franz CM, Fok Y, Valotteau C, Paternò GM, Redondo Morata L, Vacca F, Mattiello S, Magni A, Maragliano L, Beverina L, Mattioli G, Lanzani G, Baldelli P, Colombo E, Benfenati F. Nanoactuator for Neuronal Optoporation. ACS NANO 2024; 18:12427-12452. [PMID: 38687909 DOI: 10.1021/acsnano.4c01672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Light-driven modulation of neuronal activity at high spatial-temporal resolution is becoming of high interest in neuroscience. In addition to optogenetics, nongenetic membrane-targeted nanomachines that alter the electrical state of the neuronal membranes are in demand. Here, we engineered and characterized a photoswitchable conjugated compound (BV-1) that spontaneously partitions into the neuronal membrane and undergoes a charge transfer upon light stimulation. The activity of primary neurons is not affected in the dark, whereas millisecond light pulses of cyan light induce a progressive decrease in membrane resistance and an increase in inward current matched to a progressive depolarization and action potential firing. We found that illumination of BV-1 induces oxidation of membrane phospholipids, which is necessary for the electrophysiological effects and is associated with decreased membrane tension and increased membrane fluidity. Time-resolved atomic force microscopy and molecular dynamics simulations performed on planar lipid bilayers revealed that the underlying mechanism is a light-driven formation of pore-like structures across the plasma membrane. Such a phenomenon decreases membrane resistance and increases permeability to monovalent cations, namely, Na+, mimicking the effects of antifungal polyenes. The same effect on membrane resistance was also observed in nonexcitable cells. When sustained light stimulations are applied, neuronal swelling and death occur. The light-controlled pore-forming properties of BV-1 allow performing "on-demand" light-induced membrane poration to rapidly shift from cell-attached to perforated whole-cell patch-clamp configuration. Administration of BV-1 to ex vivo retinal explants or in vivo primary visual cortex elicited neuronal firing in response to short trains of light stimuli, followed by activity silencing upon prolonged light stimulations. BV-1 represents a versatile molecular nanomachine whose properties can be exploited to induce either photostimulation or space-specific cell death, depending on the pattern and duration of light stimulation.
Collapse
Affiliation(s)
- Marlene E Pfeffer
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, 16132 Genova, Italy
| | | | - Arin Marchesi
- WPI Nano Life Science Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Via Tronto 10/a, 60126 Torrette di Ancona, Italy
| | - Filippo Galluzzi
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy
- The Open University Affiliated Research Centre at Istituto Italiano di Tecnologia (ARC@IIT), Via Morego 30, 16163 Genova, Italy
| | - Matteo Moschetta
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Via Raffaele Rubattino 81, 20134 Milano, Italy
| | - Andrea Rossini
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Via Raffaele Rubattino 81, 20134 Milano, Italy
| | - Simona Francia
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Clemens M Franz
- WPI Nano Life Science Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Yulia Fok
- Aix-Marseille University, INSERM, DyNaMo, Turing Centre for Living Systems, 163 Avenue de Luminy, 13288 Marseille Cedex 09, France
| | - Claire Valotteau
- Aix-Marseille University, INSERM, DyNaMo, Turing Centre for Living Systems, 163 Avenue de Luminy, 13288 Marseille Cedex 09, France
| | - Giuseppe Maria Paternò
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Via Raffaele Rubattino 81, 20134 Milano, Italy
- Department of Physics, Politecnico di Milano, Piazza Leonardo Da Vinci, 32, 20133 Milan, Italy
| | - Lorena Redondo Morata
- Aix-Marseille University, INSERM, DyNaMo, Turing Centre for Living Systems, 163 Avenue de Luminy, 13288 Marseille Cedex 09, France
| | - Francesca Vacca
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Sara Mattiello
- Department of Material Science, Bicocca University, Via Roberto Cozzi 55, 20126 Milano, Italy
| | - Arianna Magni
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Via Raffaele Rubattino 81, 20134 Milano, Italy
- Department of Physics, Politecnico di Milano, Piazza Leonardo Da Vinci, 32, 20133 Milan, Italy
| | - Luca Maragliano
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Luca Beverina
- Department of Material Science, Bicocca University, Via Roberto Cozzi 55, 20126 Milano, Italy
| | - Giuseppe Mattioli
- Istituto di Struttura della Materia, Consiglio Nazionale delle Ricerche (CNR-ISM), Via Salaria km 29.300, 00015 Monterotondo (RM), Italy
| | - Guglielmo Lanzani
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Via Raffaele Rubattino 81, 20134 Milano, Italy
- Department of Physics, Politecnico di Milano, Piazza Leonardo Da Vinci, 32, 20133 Milan, Italy
| | - Pietro Baldelli
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Elisabetta Colombo
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| |
Collapse
|
11
|
Han R, Gaurav A, Mai CW, Gautam V, Gabriel Akyirem A. Phosphodiesterase Inhibitors of Natural Origin. THE NATURAL PRODUCTS JOURNAL 2024; 14. [DOI: 10.2174/0122103155251390230927064442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 08/03/2023] [Accepted: 08/21/2023] [Indexed: 01/09/2025]
Abstract
Abstract:
Phosphodiesterases (PDEs) function to hydrolyze intracellular cyclic adenosine monophosphate
(cAMP) and cyclic guanosine monophosphate (cGMP), regulating a variety of intracellular
signal transduction and physiological activities. PDEs can be divided into 11 families
(PDE1~11) and the diversity and complex expression of PDE family genes suggest that different
subtypes may have different mechanisms. PDEs are involved in various disease pathologies such
as inflammation, asthma, depression, and erectile dysfunction and are thus targets of interest for
several drug discovery campaigns. Natural products have always been an important source of bioactive
compounds for drug discovery, over the years several natural compounds have shown potential
as inhibitors of PDEs. In this article, phosphodiesterase inhibitors of natural origin have been
reviewed with emphasis on their chemistry and biological activities.
Collapse
Affiliation(s)
- Rui Han
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, UCSI University, Taman Connaught,
Cheras Kuala Lumpur, 56000, Malaysia
| | - Anand Gaurav
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, UCSI University, Taman Connaught,
Cheras Kuala Lumpur, 56000, Malaysia
- Department of Pharmaceutical Sciences, School of Health Sciences and
Technology, UPES, Dehradun, 248007, Uttarakhand, India
- Faculty of Health Sciences, Villa College, QI Campus,
Rahdhebai Hingun, Male', 20373, Republic of Maldives
| | - Chun-Wai Mai
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, UCSI University, Taman Connaught,
Cheras Kuala Lumpur, 56000, Malaysia
| | - Vertika Gautam
- Institute of Pharmaceutical Research, GLA University,
Mathura, 281406, Uttar Pradesh, India
| | - Akowuah Gabriel Akyirem
- School of Pharmacy, Monash University Malaysia Jalan Lagoon Selatan,
47500 Bandar Sunway, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
12
|
Kolesnikov AV, Murphy DP, Corbo JC, Kefalov VJ. Germline knockout of Nr2e3 protects photoreceptors in three distinct mouse models of retinal degeneration. Proc Natl Acad Sci U S A 2024; 121:e2316118121. [PMID: 38442152 PMCID: PMC10945761 DOI: 10.1073/pnas.2316118121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 01/17/2024] [Indexed: 03/07/2024] Open
Abstract
Retinitis pigmentosa (RP) is a common form of retinal dystrophy that can be caused by mutations in any one of dozens of rod photoreceptor genes. The genetic heterogeneity of RP represents a significant challenge for the development of effective therapies. Here, we present evidence for a potential gene-independent therapeutic strategy based on targeting Nr2e3, a transcription factor required for the normal differentiation of rod photoreceptors. Nr2e3 knockout results in hybrid rod photoreceptors that express the full complement of rod genes, but also a subset of cone genes. We show that germline deletion of Nr2e3 potently protects rods in three mechanistically diverse mouse models of retinal degeneration caused by bright-light exposure (light damage), structural deficiency (rhodopsin-deficient Rho-/- mice), or abnormal phototransduction (phosphodiesterase-deficient rd10 mice). Nr2e3 knockout confers strong neuroprotective effects on rods without adverse effects on their gene expression, structure, or function. Furthermore, in all three degeneration models, prolongation of rod survival by Nr2e3 knockout leads to lasting preservation of cone morphology and function. These findings raise the possibility that upregulation of one or more cone genes in Nr2e3-deficient rods may be responsible for the neuroprotective effects we observe.
Collapse
Affiliation(s)
- Alexander V. Kolesnikov
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, CA92697
| | - Daniel P. Murphy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO63110
| | - Joseph C. Corbo
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO63110
| | - Vladimir J. Kefalov
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, CA92697
| |
Collapse
|
13
|
Yalaz C, Bridges E, Alham NK, Zois CE, Chen J, Bensaad K, Miar A, Pires E, Muschel RJ, McCullagh JSO, Harris AL. Cone photoreceptor phosphodiesterase PDE6H inhibition regulates cancer cell growth and metabolism, replicating the dark retina response. Cancer Metab 2024; 12:5. [PMID: 38350962 PMCID: PMC10863171 DOI: 10.1186/s40170-023-00326-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/24/2023] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND PDE6H encodes PDE6γ', the inhibitory subunit of the cGMP-specific phosphodiesterase 6 in cone photoreceptors. Inhibition of PDE6, which has been widely studied for its role in light transduction, increases cGMP levels. The purpose of this study is to characterise the role of PDE6H in cancer cell growth. METHODS From an siRNA screen for 487 genes involved in metabolism, PDE6H was identified as a controller of cell cycle progression in HCT116 cells. Role of PDE6H in cancer cell growth and metabolism was studied through the effects of its depletion on levels of cell cycle controllers, mTOR effectors, metabolite levels, and metabolic energy assays. Effect of PDE6H deletion on tumour growth was also studied in a xenograft model. RESULTS PDE6H knockout resulted in an increase of intracellular cGMP levels, as well as changes to the levels of nucleotides and key energy metabolism intermediates. PDE6H knockdown induced G1 cell cycle arrest and cell death and reduced mTORC1 signalling in cancer cell lines. Both knockdown and knockout of PDE6H resulted in the suppression of mitochondrial function. HCT116 xenografts revealed that PDE6H deletion, as well as treatment with the PDE5/6 inhibitor sildenafil, slowed down tumour growth and improved survival, while sildenafil treatment did not have an additive effect on slowing the growth of PDE6γ'-deficient tumours. CONCLUSIONS Our results indicate that the changes in cGMP and purine pools, as well as mitochondrial function which is observed upon PDE6γ' depletion, are independent of the PKG pathway. We show that in HCT116, PDE6H deletion replicates many effects of the dark retina response and identify PDE6H as a new target in preventing cancer cell proliferation and tumour growth.
Collapse
Affiliation(s)
- Ceren Yalaz
- Molecular Oncology Laboratories, Department of Medical Oncology, John Radcliffe Hospital, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK.
| | - Esther Bridges
- Molecular Oncology Laboratories, Department of Medical Oncology, John Radcliffe Hospital, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Nasullah K Alham
- Department of Engineering Science, Institute of Biomedical Engineering (IBME), University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Christos E Zois
- Molecular Oncology Laboratories, Department of Medical Oncology, John Radcliffe Hospital, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Jianzhou Chen
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Karim Bensaad
- Molecular Oncology Laboratories, Department of Medical Oncology, John Radcliffe Hospital, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Ana Miar
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Elisabete Pires
- Department of Chemistry, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Ruth J Muschel
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - James S O McCullagh
- Department of Chemistry, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Adrian L Harris
- Molecular Oncology Laboratories, Department of Medical Oncology, John Radcliffe Hospital, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| |
Collapse
|
14
|
Bighinati A, Adani E, Stanzani A, D’Alessandro S, Marigo V. Molecular mechanisms underlying inherited photoreceptor degeneration as targets for therapeutic intervention. Front Cell Neurosci 2024; 18:1343544. [PMID: 38370034 PMCID: PMC10869517 DOI: 10.3389/fncel.2024.1343544] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 01/16/2024] [Indexed: 02/20/2024] Open
Abstract
Retinitis pigmentosa (RP) is a form of retinal degeneration characterized by primary degeneration of rod photoreceptors followed by a secondary cone loss that leads to vision impairment and finally blindness. This is a rare disease with mutations in several genes and high genetic heterogeneity. A challenging effort has been the characterization of the molecular mechanisms underlying photoreceptor cell death during the progression of the disease. Some of the cell death pathways have been identified and comprise stress events found in several neurodegenerative diseases such as oxidative stress, inflammation, calcium imbalance and endoplasmic reticulum stress. Other cell death mechanisms appear more relevant to photoreceptor cells, such as high levels of cGMP and metabolic changes. Here we review some of the cell death pathways characterized in the RP mutant retina and discuss preclinical studies of therapeutic approaches targeting the molecular outcomes that lead to photoreceptor cell demise.
Collapse
Affiliation(s)
- Andrea Bighinati
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisa Adani
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Agnese Stanzani
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sara D’Alessandro
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Valeria Marigo
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Center for Neuroscience and Neurotechnology, Modena, Italy
| |
Collapse
|
15
|
Ding J, Kim TH, Ma G, Yao X. Intrinsic signal optoretinography of dark adaptation abnormality due to rod photoreceptor degeneration. Exp Biol Med (Maywood) 2024; 249:10024. [PMID: 38463390 PMCID: PMC10911128 DOI: 10.3389/ebm.2024.10024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/04/2024] [Indexed: 03/12/2024] Open
Abstract
This research aims to investigate the potential of using intrinsic optical signal (IOS) optoretinography (ORG) to objectively detect dark adaptation (DA) abnormalities related to rod photoreceptor degeneration. Functional optical coherence tomography (OCT) was employed in both wild-type (WT) and retinal degeneration 10 (rd10) mice to conduct this assessment. Dynamic OCT measurements captured the changes in retinal thickness and reflectance from light-to-dark transition. Comparative analysis revealed significant IOS alterations within the outer retina. Specifically, a reduction in thickness from external limiting membrane (ELM) peak to retinal pigment epithelium (RPE) peak was observed (WT: 1.13 ± 0.69 µm, 30 min DA; rd10: 2.64 ± 0.86 µm, 30 min DA), as well as a decrease in the intensity of the inner segment ellipsoid zone (EZ) in 30 min DA compared to light adaptation (LA). The reduction of relative EZ intensity was notable in rd10 after 5 min DA and in WT after 15 min DA, with a distinguishable difference between rd10 and WT after 10 min DA. Furthermore, our findings indicated a significant decrease in the relative intensity of the hypo-reflective band between EZ and RPE in rd10 retinas during DA, which primarily corresponds to the outer segment (OS) region. In conclusion, the observed DA-IOS abnormalities, including changes in ELM-RPE thickness, EZ, and OS intensity, hold promise as differentiators between WT and rd10 mice before noticeable morphological abnormalities occur. These findings suggest the potential of this non-invasive imaging technique for the early detection of dysfunction in retinal photoreceptors.
Collapse
Affiliation(s)
- Jie Ding
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, United States
| | - Tae-Hoon Kim
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, United States
| | - Guangying Ma
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, United States
| | - Xincheng Yao
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, United States
- Department of Ophthalmology and Visual Sciences, University of Illinois Chicago, Chicago, IL, United States
| |
Collapse
|
16
|
Chirinskaite AV, Rotov AY, Ermolaeva ME, Tkachenko LA, Vaganova AN, Danilov LG, Fedoseeva KN, Kostin NA, Sopova JV, Firsov ML, Leonova EI. Does Background Matter? A Comparative Characterization of Mouse Models of Autosomal Retinitis Pigmentosa rd1 and Pde6b-KO. Int J Mol Sci 2023; 24:17180. [PMID: 38139011 PMCID: PMC10742838 DOI: 10.3390/ijms242417180] [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: 10/23/2023] [Revised: 12/02/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Many retinal degenerative diseases result in vision impairment or permanent blindness due to photoreceptor loss or dysfunction. It has been observed that Pde6brd1 mice (rd1), which carry a spontaneous nonsense mutation in the pde6b gene, have a strong phenotypic similarity to patients suffering from autosomal recessive retinitis pigmentosa. In this study, we present a novel mouse model of retinitis pigmentosa generated through pde6b gene knockout using CRISPR/Cas9 technology. We compare this Pde6b-KO mouse model to the rd1 mouse model to gain insights into the progression of retinal degeneration. The functional assessment of the mouse retina and the tracking of degeneration dynamics were performed using electrophysiological methods, while retinal morphology was analyzed through histology techniques. Interestingly, the Pde6b-KO mouse model demonstrated a higher amplitude of photoresponse than the rd1 model of the same age. At postnatal day 12, the thickness of the photoreceptor layer in both mouse models did not significantly differ from that of control animals; however, by day 15, a substantial reduction was observed. Notably, the decline in the number of photoreceptors in the rd1 model occurred at a significantly faster rate. These findings suggest that the C3H background may play a significant role in the early stages of retinal degeneration.
Collapse
Affiliation(s)
- Angelina V. Chirinskaite
- Center of Transgenesis and Genome Editing, St. Petersburg State University, Universitetskaja Emb., 7/9, 199034 St. Petersburg, Russia (J.V.S.)
| | - Alexander Yu. Rotov
- Laboratory of Evolution of Sense Organs, Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Thorez Ave., 44, 194223 St. Petersburg, Russia (M.L.F.)
| | - Mariia E. Ermolaeva
- Laboratory of Evolution of Sense Organs, Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Thorez Ave., 44, 194223 St. Petersburg, Russia (M.L.F.)
| | - Lyubov A. Tkachenko
- Department of Cytology and Histology, St. Petersburg State University, Universitetskaja Emb., 7/9, 199034 St. Petersburg, Russia
| | - Anastasia N. Vaganova
- Institute of Translational Biomedicine, St. Petersburg State University, Universitetskaja Emb., 7/9, 199034 St. Petersburg, Russia
| | - Lavrentii G. Danilov
- Department of Genetics and biotechnology, St. Petersburg State University, Universitetskaja Emb., 7/9, 199034 St. Petersburg, Russia
| | - Ksenia N. Fedoseeva
- Resource Center “Molecular and Cell Technologies”, St. Petersburg State University, Universitetskaja Emb., 7/9, 199034 St. Petersburg, Russia
| | - Nicolay A. Kostin
- Resource Center “Molecular and Cell Technologies”, St. Petersburg State University, Universitetskaja Emb., 7/9, 199034 St. Petersburg, Russia
| | - Julia V. Sopova
- Center of Transgenesis and Genome Editing, St. Petersburg State University, Universitetskaja Emb., 7/9, 199034 St. Petersburg, Russia (J.V.S.)
- Laboratory of Amyloid Biology, St. Petersburg State University, Universitetskaja Emb., 7/9, 199034 St. Petersburg, Russia
| | - Michael L. Firsov
- Laboratory of Evolution of Sense Organs, Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Thorez Ave., 44, 194223 St. Petersburg, Russia (M.L.F.)
| | - Elena I. Leonova
- Center of Transgenesis and Genome Editing, St. Petersburg State University, Universitetskaja Emb., 7/9, 199034 St. Petersburg, Russia (J.V.S.)
| |
Collapse
|
17
|
Xia CH, Liu H, Li M, Zhang H, Xing X, Gong X. Identification and Characterization of Retinitis Pigmentosa in a Novel Mouse Model Caused by PDE6B-T592I. Biomedicines 2023; 11:3173. [PMID: 38137394 PMCID: PMC10740990 DOI: 10.3390/biomedicines11123173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
The cGMP-phosphodiesterase 6 beta subunit (PDE6B) is an essential component in the phototransduction pathway for light responses in photoreceptor cells. PDE6B gene mutations cause the death of rod photoreceptors, named as hereditary retinitis pigmentosa (RP) in humans and retinal degeneration (RD) in rodents. Here, we report a new RD model, identified from a phenotypic screen of N-ethyl-N-nitrosourea (ENU)-induced mutant mice, which displays retinal degeneration caused by a point mutation in the Pde6b gene that results in PDE6B-T592I mutant protein. The homozygous mutant mice show an extensive loss of rod photoreceptors at the age of 3 weeks; unexpectedly, the loss of rod photoreceptors can be partly rescued by dark rearing. Thus, this RD mutant model displays a light-dependent loss of rod photoreceptors. Both western blot and immunostaining results show very low level of mutant PDE6B-T592I protein in the retina. Structure modeling suggests that the T592I mutation probably affects the function and stability of PDE6B protein by changing intramolecular interactions. We further demonstrate that the expression of wild-type PDE6B delivered by subretinally injected adeno-associated virus (rAAV) prevents photoreceptor cell death in this RD model in vivo. The PDE6B-T592I mutant is, therefore, a valuable RD model for evaluating rAAV-mediated treatment and for investigating the molecular mechanism of light-dependent rod photoreceptor cell death that is related to impaired PDE6B function.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaohua Gong
- Herbert Wertheim School of Optometry and Vision Science Program, University of California, Berkeley, CA 94720, USA; (C.-H.X.); (H.L.); (M.L.); (H.Z.); (X.X.)
| |
Collapse
|
18
|
Sukkar B, Oktay L, Sahaboglu A, Moayedi A, Zenouri S, Al-Maghout T, Cantó A, Miranda M, Durdagi S, Hosseinzadeh Z. Inhibition of altered Orai1 channels in Müller cells protects photoreceptors in retinal degeneration. Glia 2023; 71:2511-2526. [PMID: 37533369 DOI: 10.1002/glia.24429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 08/04/2023]
Abstract
The expressions of ion channels by Müller glial cells (MGCs) may change in response to various retinal pathophysiological conditions. There remains a gap in our understanding of MGCs' responses to photoreceptor degeneration towards finding therapies. The study explores how an inhibition of store-operated Ca2+ entry (SOCE) and its major component, Orai1 channel, in MGCs protects photoreceptors from degeneration. The study revealed increased Orai1 expression in the MGCs of retinal degeneration 10 (rd10) mice. Enhanced expression of oxidative stress markers was confirmed as a crucial pathological mechanism in rd10 retina. Inducing oxidative stress in rat MGCs resulted in increasing SOCE and Ca2+ release-activated Ca2+ (CRAC) currents. SOCE inhibition by 2-Aminoethoxydiphenyl borate (2-APB) protected photoreceptors in degenerated retinas. Finally, molecular simulations proved the structural and dynamical features of 2-APB to the target structure Orai1. Our results provide new insights into the physiology of MGCs regarding retinal degeneration and shed a light on SOCE and Orai1 as new therapeutic targets.
Collapse
Affiliation(s)
- Basma Sukkar
- Paul Flechsig Institute, Centre of Neuropathology and Brain Research, University of Leipzig, Leipzig, Germany
| | - Lalehan Oktay
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey
| | - Ayse Sahaboglu
- Institute for Ophthalmic Research, Centre for Ophthalmology, Eberhard Karls University, Tübingen, Germany
| | - Aylin Moayedi
- Paul Flechsig Institute, Centre of Neuropathology and Brain Research, University of Leipzig, Leipzig, Germany
| | - Shima Zenouri
- Paul Flechsig Institute, Centre of Neuropathology and Brain Research, University of Leipzig, Leipzig, Germany
| | - Tamer Al-Maghout
- Department of Cardiology and Vascular Medicine and Physiology, University of Tübingen, Tübingen, Germany
| | - Antolin Cantó
- Departamento Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - María Miranda
- Departamento Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Serdar Durdagi
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey
- Molecular Therapy Laboratory, School of Pharmacy, Bahcesehir University, Istanbul, Turkey
| | - Zohreh Hosseinzadeh
- Paul Flechsig Institute, Centre of Neuropathology and Brain Research, University of Leipzig, Leipzig, Germany
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| |
Collapse
|
19
|
Kramer RH. Suppressing Retinal Remodeling to Mitigate Vision Loss in Photoreceptor Degenerative Disorders. Annu Rev Vis Sci 2023; 9:131-153. [PMID: 37713276 DOI: 10.1146/annurev-vision-112122-020957] [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: 09/17/2023]
Abstract
Rod and cone photoreceptors degenerate in retinitis pigmentosa and age-related macular degeneration, robbing the visual system of light-triggered signals necessary for sight. However, changes in the retina do not stop with the photoreceptors. A stereotypical set of morphological and physiological changes, known as remodeling, occur in downstream retinal neurons. Some aspects of remodeling are homeostatic, with structural or functional changes compensating for partial loss of visual inputs. However, other aspects are nonhomeostatic, corrupting retinal information processing to obscure vision mediated naturally by surviving photoreceptors or artificially by vision-restoration technologies. In this review, I consider the mechanism of remodeling and its consequences for residual and restored visual function; discuss the role of retinoic acid, a critical molecular trigger of detrimental remodeling; and discuss strategies for suppressing retinoic acid biosynthesis or signaling as therapeutic possibilities for mitigating vision loss.
Collapse
Affiliation(s)
- Richard H Kramer
- Department of Molecular and Cell Biology, University of California, Berkeley, USA;
| |
Collapse
|
20
|
Li S, Ma H, Yang F, Ding X. cGMP Signaling in Photoreceptor Degeneration. Int J Mol Sci 2023; 24:11200. [PMID: 37446378 PMCID: PMC10342299 DOI: 10.3390/ijms241311200] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
Photoreceptors in the retina are highly specialized neurons with photosensitive molecules in the outer segment that transform light into chemical and electrical signals, and these signals are ultimately relayed to the visual cortex in the brain to form vision. Photoreceptors are composed of rods and cones. Rods are responsible for dim light vision, whereas cones are responsible for bright light, color vision, and visual acuity. Photoreceptors undergo progressive degeneration over time in many hereditary and age-related retinal diseases. Despite the remarkable heterogeneity of disease-causing genes, environmental factors, and pathogenesis, the progressive death of rod and cone photoreceptors ultimately leads to loss of vision/blindness. There are currently no treatments available for retinal degeneration. Cyclic guanosine 3', 5'-monophosphate (cGMP) plays a pivotal role in phototransduction. cGMP governs the cyclic nucleotide-gated (CNG) channels on the plasma membrane of the photoreceptor outer segments, thereby regulating membrane potential and signal transmission. By gating the CNG channels, cGMP regulates cellular Ca2+ homeostasis and signal transduction. As a second messenger, cGMP activates the cGMP-dependent protein kinase G (PKG), which regulates numerous targets/cellular events. The dysregulation of cGMP signaling is observed in varieties of photoreceptor/retinal degenerative diseases. Abnormally elevated cGMP signaling interferes with various cellular events, which ultimately leads to photoreceptor degeneration. In line with this, strategies to reduce cellular cGMP signaling result in photoreceptor protection in mouse models of retinal degeneration. The potential mechanisms underlying cGMP signaling-induced photoreceptor degeneration involve the activation of PKG and impaired Ca2+ homeostasis/Ca2+ overload, resulting from overactivation of the CNG channels, as well as the subsequent activation of the downstream cellular stress/death pathways. Thus, targeting the cellular cGMP/PKG signaling and the Ca2+-regulating pathways represents a significant strategy for photoreceptor protection in retinal degenerative diseases.
Collapse
Affiliation(s)
| | | | | | - Xiqin Ding
- Department of Cell Biology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (S.L.); (H.M.); (F.Y.)
| |
Collapse
|
21
|
Ellis EM, Paniagua AE, Scalabrino ML, Thapa M, Rathinavelu J, Jiao Y, Williams DS, Field GD, Fain GL, Sampath AP. Cones and cone pathways remain functional in advanced retinal degeneration. Curr Biol 2023; 33:1513-1522.e4. [PMID: 36977418 PMCID: PMC10133175 DOI: 10.1016/j.cub.2023.03.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/14/2022] [Accepted: 03/03/2023] [Indexed: 03/29/2023]
Abstract
Most defects causing retinal degeneration in retinitis pigmentosa (RP) are rod-specific mutations, but the subsequent degeneration of cones, which produces loss of daylight vision and high-acuity perception, is the most debilitating feature of the disease. To understand better why cones degenerate and how cone vision might be restored, we have made the first single-cell recordings of light responses from degenerating cones and retinal interneurons after most rods have died and cones have lost their outer-segment disk membranes and synaptic pedicles. We show that degenerating cones have functional cyclic-nucleotide-gated channels and can continue to give light responses, apparently produced by opsin localized either to small areas of organized membrane near the ciliary axoneme or distributed throughout the inner segment. Light responses of second-order horizontal and bipolar cells are less sensitive but otherwise resemble those of normal retina. Furthermore, retinal output as reflected in responses of ganglion cells is less sensitive but maintains spatiotemporal receptive fields at cone-mediated light levels. Together, these findings show that cones and their retinal pathways can remain functional even as degeneration is progressing, an encouraging result for future research aimed at enhancing the light sensitivity of residual cones to restore vision in patients with genetically inherited retinal degeneration.
Collapse
Affiliation(s)
- Erika M Ellis
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095-7000, USA
| | - Antonio E Paniagua
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095-7000, USA
| | - Miranda L Scalabrino
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095-7000, USA; Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Mishek Thapa
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095-7000, USA; Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jay Rathinavelu
- Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Yuekan Jiao
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095-7000, USA
| | - David S Williams
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095-7000, USA.
| | - Greg D Field
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095-7000, USA; Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Gordon L Fain
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095-7000, USA.
| | - Alapakkam P Sampath
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095-7000, USA.
| |
Collapse
|
22
|
Yamoah A, Tripathi P, Guo H, Scheve L, Walter P, Johnen S, Müller F, Weis J, Goswami A. Early Alterations of RNA Binding Protein (RBP) Homeostasis and ER Stress-Mediated Autophagy Contributes to Progressive Retinal Degeneration in the rd10 Mouse Model of Retinitis Pigmentosa (RP). Cells 2023; 12:cells12071094. [PMID: 37048167 PMCID: PMC10092976 DOI: 10.3390/cells12071094] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 04/14/2023] Open
Abstract
The retinal degeneration 10 (rd10) mouse model is widely used to study retinitis pigmentosa (RP) pathomechanisms. It offers a rather unique opportunity to study trans-neuronal degeneration because the cell populations in question are separated anatomically and the mutated Pde6b gene is selectively expressed in rod photoreceptors. We hypothesized that RNA binding protein (RBP) aggregation and abnormal autophagy might serve as early pathogenic events, damaging non-photoreceptor retinal cell types that are not primarily targeted by the Pde6b gene defect. We used a combination of immunohistochemistry (DAB, immunofluorescence), electron microscopy (EM), subcellular fractionation, and Western blot analysis on the retinal preparations obtained from both rd10 and wild-type mice. We found early, robust increases in levels of the protective endoplasmic reticulum (ER) calcium (Ca2+) buffering chaperone Sigma receptor 1 (SigR1) together with other ER-Ca2+ buffering proteins in both photoreceptors and non-photoreceptor neuronal cells before any noticeable photoreceptor degeneration. In line with this, we found markedly altered expression of the autophagy proteins p62 and LC3, together with abnormal ER widening and large autophagic vacuoles as detected by EM. Interestingly, these changes were accompanied by early, prominent cytoplasmic and nuclear aggregation of the key RBPs including pTDP-43 and FET family RBPs and stress granule formation. We conclude that progressive neurodegeneration in the rd10 mouse retina is associated with early disturbances of proteostasis and autophagy, along with abnormal cytoplasmic RBP aggregation.
Collapse
Affiliation(s)
- Alfred Yamoah
- Institute of Neuropathology, University Hospital RWTH Aachen, 52074 Aachen, Germany
- EURON-European Graduate School of Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Priyanka Tripathi
- Institute of Neuropathology, University Hospital RWTH Aachen, 52074 Aachen, Germany
- EURON-European Graduate School of Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Haihong Guo
- Institute of Neuropathology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Leonie Scheve
- Institute of Neuropathology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Peter Walter
- Department of Ophthalmology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Sandra Johnen
- Department of Ophthalmology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Frank Müller
- Institute of Biological Information Processing, Molecular and Cellular Physiology, IBI-1, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
| | - Joachim Weis
- Institute of Neuropathology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Anand Goswami
- Institute of Neuropathology, University Hospital RWTH Aachen, 52074 Aachen, Germany
- Department of Neurology, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA
- Department of Neurology, Eleanor and Lou Gehrig ALS Center, Columbia University, New York, NY 10032, USA
| |
Collapse
|
23
|
Rasmussen M, Tolone A, Paquet-Durand F, Welinder C, Schwede F, Ekström P. The photoreceptor protective cGMP-analog Rp-8-Br-PET-cGMPS interacts with cGMP-interactors PKGI, PDE1, PDE6, and PKAI in the degenerating mouse retina. J Comp Neurol 2023; 531:935-951. [PMID: 36989379 DOI: 10.1002/cne.25475] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/12/2022] [Accepted: 03/06/2023] [Indexed: 03/31/2023]
Abstract
The inherited eye disease retinitis pigmentosa (RP) causes the loss of photoreceptors by a still unknown cell death mechanism. During this degeneration, cyclic guanosine-3',5'-monophosphate (cGMP) levels become elevated, leading to over-activation of the cGMP-binding protein cGMP-dependent protein kinase (PKG). cGMP analogs selectively modified to have inhibitory actions on PKG have aided in impeding photoreceptor death, and one such cGMP analog is Rp-8-Br-PET-cGMPS. However, cGMP analogs have previously been shown to interact with numerous targets, so to better understand the therapeutic action of Rp-8-Br-PET-cGMPS, it is necessary to elucidate its target-selectivity and hence what potential cellular mechanism(s) it may affect within the photoreceptors. Here, we, therefore, applied affinity chromatography together with mass spectrometry to isolate and identify Rp-8-Br-PET-cGMPS interactors from retinas derived from three different murine RP models (i.e., rd1, rd2, and rd10 mice). Our findings revealed that Rp-8-Br-PET-cGMPS bound seven known cGMP-binding proteins, including PKG1β, PDE1β, PDE1c, PDE6α, and PKA1α. Furthermore, an additional 28 proteins were found to be associated with Rp-8-Br-PET-cGMPS. This latter group included MAPK1/3, which is known to connect with cGMP/PKG in other systems. However, in organotypic retinal cultures, Rp-8-Br-PET-cGMPS had no effect on photoreceptor MAPK1/3 expression or activity. To summarize, Rp-8-Br-PET-cGMPS is more target specific compared to regular cGMP.
Collapse
Affiliation(s)
- Michel Rasmussen
- Faculty of Medicine, Department of Clinical Sciences Lund, Lund University, Ophthalmology, Lund, Sweden
| | - Arianna Tolone
- Insitute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | | | - Charlotte Welinder
- Faculty of Medicine, Department of Clinical Sciences Lund, Mass Spectrometry, Lund University, Lund, Sweden
| | - Frank Schwede
- BIOLOG Life Science Institute GmbH & Co. KG, Bremen, Germany
| | - Per Ekström
- Faculty of Medicine, Department of Clinical Sciences Lund, Lund University, Ophthalmology, Lund, Sweden
| |
Collapse
|
24
|
Tay HG, Andre H, Chrysostomou V, Adusumalli S, Guo J, Ren X, Tan WS, Tor JE, Moreno-Moral A, Plastino F, Bartuma H, Cai Z, Tun SBB, Barathi VA, Siew Wei GT, Grenci G, Chong LY, Holmgren A, Kvanta A, Crowston JG, Petretto E, Tryggvason K. Photoreceptor laminin drives differentiation of human pluripotent stem cells to photoreceptor progenitors that partially restore retina function. Mol Ther 2023; 31:825-846. [PMID: 36638800 PMCID: PMC10014235 DOI: 10.1016/j.ymthe.2022.12.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 07/12/2022] [Accepted: 12/21/2022] [Indexed: 01/14/2023] Open
Abstract
Blindness caused by advanced stages of inherited retinal diseases and age-related macular degeneration are characterized by photoreceptor loss. Cell therapy involving replacement with functional photoreceptor-like cells generated from human pluripotent stem cells holds great promise. Here, we generated a human recombinant retina-specific laminin isoform, LN523, and demonstrated the role in promoting the differentiation of human embryonic stem cells into photoreceptor progenitors. This chemically defined and xenogen-free method enables reproducible production of photoreceptor progenitors within 32 days. We observed that the transplantation into rd10 mice were able to protect the host photoreceptor outer nuclear layer (ONL) up to 2 weeks post transplantation as measured by full-field electroretinogram. At 4 weeks post transplantation, the engrafted cells were found to survive, mature, and associate with the host's rod bipolar cells. Visual behavioral assessment using the water maze swimming test demonstrated visual improvement in the cell-transplanted rodents. At 20 weeks post transplantation, the maturing engrafted cells were able to replace the loss of host ONL by extensive association with host bipolar cells and synapses. Post-transplanted rabbit model also provided congruent evidence for synaptic connectivity with the degenerated host retina. The results may pave the way for the development of stem cell-based therapeutics for retina degeneration.
Collapse
Affiliation(s)
- Hwee Goon Tay
- Centre for Vision Research, Duke-NUS Medical School, Singapore; Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore.
| | - Helder Andre
- Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Vicki Chrysostomou
- Centre for Vision Research, Duke-NUS Medical School, Singapore; Academic Clinical Program, Duke-NUS Medical School, Singapore
| | | | - Jing Guo
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Xiaoyuan Ren
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Wei Sheng Tan
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Jia En Tor
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Aida Moreno-Moral
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Flavia Plastino
- Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Hammurabi Bartuma
- Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Zuhua Cai
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Sai Bo Bo Tun
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Veluchamy Amutha Barathi
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Gavin Tan Siew Wei
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Gianluca Grenci
- Mechanobiology Institute (MBI) and Department of Biomedical Engineering, NUS, Singapore
| | - Li Yen Chong
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Arne Holmgren
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Anders Kvanta
- Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Jonathan Guy Crowston
- Centre for Vision Research, Duke-NUS Medical School, Singapore; Academic Clinical Program, Duke-NUS Medical School, Singapore; Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Enrico Petretto
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Karl Tryggvason
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore; Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden; Division of Nephrology, Department of Medicine, Duke University, Durham, NC, USA.
| |
Collapse
|
25
|
Yang X, Xu Z, Hu S, Shen J. Perspectives of PDE inhibitor on treating idiopathic pulmonary fibrosis. Front Pharmacol 2023; 14:1111393. [PMID: 36865908 PMCID: PMC9973527 DOI: 10.3389/fphar.2023.1111393] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/03/2023] [Indexed: 02/16/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive interstitial lung disease (ILD) without an identifiable cause. If not treated after diagnosis, the average life expectancy is 3-5 years. Currently approved drugs for the treatment of IPF are Pirfenidone and Nintedanib, as antifibrotic drugs, which can reduce the decline rate of forced vital capacity (FVC) and reduce the risk of acute exacerbation of IPF. However these drugs can not relieve the symptoms associated with IPF, nor improve the overall survival rate of IPF patients. We need to develop new, safe and effective drugs to treat pulmonary fibrosis. Previous studies have shown that cyclic nucleotides participate in the pathway and play an essential role in the process of pulmonary fibrosis. Phosphodiesterase (PDEs) is involved in cyclic nucleotide metabolism, so PDE inhibitors are candidates for pulmonary fibrosis. This paper reviews the research progress of PDE inhibitors related to pulmonary fibrosis, so as to provide ideas for the development of anti-pulmonary fibrosis drugs.
Collapse
Affiliation(s)
- Xudan Yang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, China
| | | | - Songhua Hu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, China
| | - Juan Shen
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, China
| |
Collapse
|
26
|
Kumari A, Borooah S. The Role of Microglia in Inherited Retinal Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:197-205. [PMID: 37440034 DOI: 10.1007/978-3-031-27681-1_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Inherited retinal diseases (IRDs) are a leading cause of irreversible visual loss in the developed world. The primary driver of pathology in IRDs is pathogenic genetic variant. However, there is increasing evidence, from recent studies, for a role of the immune system in disease mechanism, particularly retinal microglia. Microglia are the primary immune cells in the retina and actively contribute to disease pathogenesis when activated locally by phagocytosing photoreceptors, inducing inflammation and signaling infiltration of circulating monocytes. In this article, we discuss the evidence for the contribution of retinal microglia to IRD pathogenesis reported so far using mice model.
Collapse
Affiliation(s)
- Asha Kumari
- Jacobs Retina Center, Shiley Eye Institute, University of California San Diego, La Jolla, CA, USA
| | - Shyamanga Borooah
- Jacobs Retina Center, Shiley Eye Institute, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
27
|
Das A, Imanishi Y. Drug Discovery Strategies for Inherited Retinal Degenerations. BIOLOGY 2022; 11:1338. [PMID: 36138817 PMCID: PMC9495580 DOI: 10.3390/biology11091338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/31/2022] [Accepted: 09/07/2022] [Indexed: 12/03/2022]
Abstract
Inherited retinal degeneration is a group of blinding disorders afflicting more than 1 in 4000 worldwide. These disorders frequently cause the death of photoreceptor cells or retinal ganglion cells. In a subset of these disorders, photoreceptor cell death is a secondary consequence of retinal pigment epithelial cell dysfunction or degeneration. This manuscript reviews current efforts in identifying targets and developing small molecule-based therapies for these devastating neuronal degenerations, for which no cures exist. Photoreceptors and retinal ganglion cells are metabolically demanding owing to their unique structures and functional properties. Modulations of metabolic pathways, which are disrupted in most inherited retinal degenerations, serve as promising therapeutic strategies. In monogenic disorders, great insights were previously obtained regarding targets associated with the defective pathways, including phototransduction, visual cycle, and mitophagy. In addition to these target-based drug discoveries, we will discuss how phenotypic screening can be harnessed to discover beneficial molecules without prior knowledge of their mechanisms of action. Because of major anatomical and biological differences, it has frequently been challenging to model human inherited retinal degeneration conditions using small animals such as rodents. Recent advances in stem cell-based techniques are opening new avenues to obtain pure populations of human retinal ganglion cells and retinal organoids with photoreceptor cells. We will discuss concurrent ideas of utilizing stem-cell-based disease models for drug discovery and preclinical development.
Collapse
Affiliation(s)
- Arupratan Das
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yoshikazu Imanishi
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
28
|
Berkowitz BA, Podolsky RH, Childers KL, Burgoyne T, De Rossi G, Qian H, Roberts R, Katz R, Waseem R, Goodman C. Functional Changes Within the Rod Inner Segment Ellipsoid in Wildtype Mice: An Optical Coherence Tomography and Electron Microscopy Study. Invest Ophthalmol Vis Sci 2022; 63:8. [PMID: 35816042 PMCID: PMC9284466 DOI: 10.1167/iovs.63.8.8] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 06/19/2022] [Indexed: 12/12/2022] Open
Abstract
Purpose To test the hypothesis that changing energy needs alter mitochondria distribution within the rod inner segment ellipsoid. Methods In mice with relatively smaller (C57BL/6J [B6J]) or greater (129S6/ev [S6]) retina mitochondria maximum reserve capacity, the profile shape of the rod inner segment ellipsoid zone (ISez) was measured with optical coherence tomography (OCT) under higher (dark) or lower (light) energy demand conditions. ISez profile shape was characterized using an unbiased ellipse descriptor (minor/major aspect ratio). Other bioenergy indexes evaluated include the external limiting membrane-retinal pigment epithelium (ELM-RPE) thickness and the magnitude of the signal intensity of a hyporeflective band located between the photoreceptor tips and apical RPE. The spatial distribution of rod ellipsoid mitochondria were also examined with electron microscopy. Results In B6J mice, darkness produced a greater ISez aspect ratio, thinner ELM-RPE, and a smaller hyporeflective band intensity than in light. In S6 mice, dark and light ISez aspect ratio values were not different and were greater than in light-adapted B6J mice; dark-adapted S6 mice showed smaller ELM-RPE thinning versus light, and negligible hyporeflective band intensity in the light. In B6J mice, mitochondria number in light increased in the distal inner segment ellipsoid and decreased proximally. In S6 mice, mitochondria number in the inner segment ellipsoid were not different between light and dark, and were greater than in B6J mice. Conclusions These data raise the possibility that rod mitochondria activity in mice can be noninvasively evaluated based on the ISez profile shape, a new OCT index that complements OCT energy biomarkers measured outside of the ISez region.
Collapse
Affiliation(s)
- Bruce A Berkowitz
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Robert H Podolsky
- Biostatistics and Study Methodology, Children's National Hospital, Silver Spring, Maryland, United States
| | - Karen Lins Childers
- Beaumont Research Institute, Beaumont Health, Royal Oak, Michigan, Unites States
| | - Tom Burgoyne
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Giulia De Rossi
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Haohua Qian
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland, Unites States
| | - Robin Roberts
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Ryan Katz
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Rida Waseem
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Cole Goodman
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| |
Collapse
|
29
|
Karademir D, Todorova V, Ebner LJA, Samardzija M, Grimm C. Single-cell RNA sequencing of the retina in a model of retinitis pigmentosa reveals early responses to degeneration in rods and cones. BMC Biol 2022; 20:86. [PMID: 35413909 PMCID: PMC9006580 DOI: 10.1186/s12915-022-01280-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 03/12/2022] [Indexed: 11/18/2022] Open
Abstract
Background In inherited retinal disorders such as retinitis pigmentosa (RP), rod photoreceptor-specific mutations cause primary rod degeneration that is followed by secondary cone death and loss of high-acuity vision. Mechanistic studies of retinal degeneration are challenging because of retinal heterogeneity. Moreover, the detection of early cone responses to rod death is especially difficult due to the paucity of cones in the retina. To resolve heterogeneity in the degenerating retina and investigate events in both types of photoreceptors during primary rod degeneration, we utilized droplet-based single-cell RNA sequencing in an RP mouse model, rd10. Results Using trajectory analysis, we defined two consecutive phases of rod degeneration at P21, characterized by the early transient upregulation of Egr1 and the later induction of Cebpd. EGR1 was the transcription factor most significantly associated with the promoters of differentially regulated genes in Egr1-positive rods in silico. Silencing Egr1 affected the expression levels of two of these genes in vitro. Degenerating rods exhibited changes associated with metabolism, neuroprotection, and modifications to synapses and microtubules. Egr1 was also the most strongly upregulated transcript in cones. Its upregulation in cones accompanied potential early respiratory dysfunction and changes in signaling pathways. The expression pattern of EGR1 in the retina was dynamic during degeneration, with a transient increase of EGR1 immunoreactivity in both rods and cones during the early stages of their degenerative processes. Conclusion Our results identify early and late changes in degenerating rd10 rod photoreceptors and reveal early responses to rod degeneration in cones not expressing the disease-causing mutation, pointing to mechanisms relevant for secondary cone degeneration. In addition, our data implicate EGR1 as a potential key regulator of early degenerative events in rods and cones, providing a potential broad target for modulating photoreceptor degeneration. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01280-9.
Collapse
Affiliation(s)
- Duygu Karademir
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Zurich, Switzerland. .,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.
| | - Vyara Todorova
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| | - Lynn J A Ebner
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Marijana Samardzija
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Christian Grimm
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
30
|
Weh E, Scott K, Wubben TJ, Besirli CG. Dark-reared rd10 mice experience rapid photoreceptor degeneration with short exposure to room-light during in vivo retinal imaging. Exp Eye Res 2022; 215:108913. [PMID: 34965404 PMCID: PMC8923962 DOI: 10.1016/j.exer.2021.108913] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 02/03/2023]
Abstract
Inherited retinal diseases (IRDs) are a collection of rare genetic conditions, which can lead to complete blindness. A large number of causative genes have been identified for IRDs and while some success has been achieved with gene therapies, they are limited in scope to each individual gene and/or the specific mutation harbored by each patient with an IRD. Multiple studies are underway to elucidate common underlying mechanisms contributing to photoreceptor (PR) loss and to design gene-agnostic, pan-disease therapeutics. The rd10 mouse, which recapitulates slow degeneration of PRs, is an in vivo IRD model used commonly by vision researchers. Light deprivation by rearing animals in complete darkness significantly delays PR death in rd10 mice, subsequently increasing the time window for in vivo studies investigating neuroprotective strategies. Longitudinal in vivo retinal imaging following the same rd10 mice over time is a potential solution for reducing the number of animals required to complete a study. We describe a previously unreported phenotype in the dark-reared rd10 model that is characterized by dramatic PR degeneration following brief exposure to low-intensity light. This exquisite light sensitivity precludes the use of longitudinal studies employing in vivo imaging or other functional assessment requiring room light in rd10 mice and highlights the importance of closely following animal models of IRD to determine any deviations from the expected degeneration curve during routine experimentation.
Collapse
Affiliation(s)
| | | | | | - Cagri G. Besirli
- Corresponding Author, please direct all correspondence to: Cagri Besirli, 1000 Wall St., Ann Arbor, MI 48105, 734-232-8404,
| |
Collapse
|
31
|
Shughoury A, Ciulla TA, Bakall B, Pennesi ME, Kiss S, Cunningham ET. Genes and Gene Therapy in Inherited Retinal Disease. Int Ophthalmol Clin 2021; 61:3-45. [PMID: 34584043 DOI: 10.1097/iio.0000000000000377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
32
|
Yang C, Georgiou M, Atkinson R, Collin J, Al-Aama J, Nagaraja-Grellscheid S, Johnson C, Ali R, Armstrong L, Mozaffari-Jovin S, Lako M. Pre-mRNA Processing Factors and Retinitis Pigmentosa: RNA Splicing and Beyond. Front Cell Dev Biol 2021; 9:700276. [PMID: 34395430 PMCID: PMC8355544 DOI: 10.3389/fcell.2021.700276] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 07/09/2021] [Indexed: 12/20/2022] Open
Abstract
Retinitis pigmentosa (RP) is the most common inherited retinal disease characterized by progressive degeneration of photoreceptors and/or retinal pigment epithelium that eventually results in blindness. Mutations in pre-mRNA processing factors (PRPF3, 4, 6, 8, 31, SNRNP200, and RP9) have been linked to 15–20% of autosomal dominant RP (adRP) cases. Current evidence indicates that PRPF mutations cause retinal specific global spliceosome dysregulation, leading to mis-splicing of numerous genes that are involved in a variety of retina-specific functions and/or general biological processes, including phototransduction, retinol metabolism, photoreceptor disk morphogenesis, retinal cell polarity, ciliogenesis, cytoskeleton and tight junction organization, waste disposal, inflammation, and apoptosis. Importantly, additional PRPF functions beyond RNA splicing have been documented recently, suggesting a more complex mechanism underlying PRPF-RPs driven disease pathogenesis. The current review focuses on the key RP-PRPF genes, depicting the current understanding of their roles in RNA splicing, impact of their mutations on retinal cell’s transcriptome and phenome, discussed in the context of model species including yeast, zebrafish, and mice. Importantly, information on PRPF functions beyond RNA splicing are discussed, aiming at a holistic investigation of PRPF-RP pathogenesis. Finally, work performed in human patient-specific lab models and developing gene and cell-based replacement therapies for the treatment of PRPF-RPs are thoroughly discussed to allow the reader to get a deeper understanding of the disease mechanisms, which we believe will facilitate the establishment of novel and better therapeutic strategies for PRPF-RP patients.
Collapse
Affiliation(s)
- Chunbo Yang
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Maria Georgiou
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Robert Atkinson
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Joseph Collin
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Jumana Al-Aama
- Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Colin Johnson
- Leeds Institute of Molecular Medicine, University of Leeds, Leeds, United Kingdom
| | - Robin Ali
- King's College London, London, United Kingdom
| | - Lyle Armstrong
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Sina Mozaffari-Jovin
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Cellular Biochemistry, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Majlinda Lako
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
33
|
Inhibition of Epigenetic Modifiers LSD1 and HDAC1 Blocks Rod Photoreceptor Death in Mouse Models of Retinitis Pigmentosa. J Neurosci 2021; 41:6775-6792. [PMID: 34193554 DOI: 10.1523/jneurosci.3102-20.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 06/11/2021] [Accepted: 06/18/2021] [Indexed: 11/21/2022] Open
Abstract
Epigenetic modifiers are increasingly being investigated as potential therapeutics to modify and overcome disease phenotypes. Diseases of the nervous system present a particular problem as neurons are postmitotic and demonstrate relatively stable gene expression patterns and chromatin organization. We have explored the ability of epigenetic modifiers to prevent degeneration of rod photoreceptors in a mouse model of retinitis pigmentosa (RP), using rd10 mice of both sexes. The histone modification eraser enzymes lysine demethylase 1 (LSD1) and histone deacetylase 1 (HDAC1) are known to have dramatic effects on the development of rod photoreceptors. In the RP mouse model, inhibitors of these enzymes blocked rod degeneration, preserved vision, and affected the expression of multiple genes including maintenance of rod-specific transcripts and downregulation of those involved in inflammation, gliosis, and cell death. The neuroprotective activity of LSD1 inhibitors includes two pathways. First, through targeting histone modifications, they increase accessibility of chromatin and upregulate neuroprotective genes, such as from the Wnt pathway. We propose that this process is going in rod photoreceptors. Second, through nonhistone targets, they inhibit transcription of inflammatory genes and inflammation. This process is going in microglia, and lack of inflammation keeps rod photoreceptors alive.SIGNIFICANCE STATEMENT Retinal degenerations are a leading cause of vision loss. RP is genetically very heterogeneous, and the multiple pathways leading to cell death are one reason for the slow progress in identifying suitable treatments for patients. Here we demonstrate that inhibition of LSD1and HDAC1 in a mouse model of RP leads to preservation of rod photoreceptors and visual function, retaining of expression of rod-specific genes, and with decreased inflammation, cell death, and Müller cell gliosis. We propose that these epigenetic inhibitors cause more open and accessible chromatin, allowing expression of neuroprotective genes. A second mechanism that allows rod photoreceptor survival is suppression of inflammation by epigenetic inhibitors in microglia. Manipulation of epigenetic modifiers is a new strategy to fight neurodegeneration in RP.
Collapse
|
34
|
Photoreceptor Phosphodiesterase (PDE6): Structure, Regulatory Mechanisms, and Implications for Treatment of Retinal Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1371:33-59. [PMID: 34170501 DOI: 10.1007/5584_2021_649] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The photoreceptor phosphodiesterase (PDE6) is a member of large family of Class I phosphodiesterases responsible for hydrolyzing the second messengers cAMP and cGMP. PDE6 consists of two catalytic subunits and two inhibitory subunits that form a tetrameric protein. PDE6 is a peripheral membrane protein that is localized to the signal-transducing compartment of rod and cone photoreceptors. As the central effector enzyme of the G-protein coupled visual transduction pathway, activation of PDE6 catalysis causes a rapid decrease in cGMP levels that results in closure of cGMP-gated ion channels in the photoreceptor plasma membrane. Because of its importance in the phototransduction pathway, mutations in PDE6 genes result in various retinal diseases that currently lack therapeutic treatment strategies due to inadequate knowledge of the structure, function, and regulation of this enzyme. This review focuses on recent progress in understanding the structure of the regulatory and catalytic domains of the PDE6 holoenzyme, the central role of the multi-functional inhibitory γ-subunit, the mechanism of activation by the heterotrimeric G protein, transducin, and future directions for pharmacological interventions to treat retinal degenerative diseases arising from mutations in the PDE6 genes.
Collapse
|
35
|
Ruiz-Pastor MJ, Kutsyr O, Lax P, Cuenca N. Decrease in DHA and other fatty acids correlates with photoreceptor degeneration in retinitis pigmentosa. Exp Eye Res 2021; 209:108667. [PMID: 34119484 DOI: 10.1016/j.exer.2021.108667] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/21/2021] [Accepted: 06/07/2021] [Indexed: 10/21/2022]
Abstract
Fatty acids, and especially docosahexaenoic acid (DHA), are essential for photoreceptor cell integrity and are involved in the phototransduction cascade. In this study, we analyzed the changes in the fatty acid profile in the retina of the rd10 mouse, model of retinitis pigmentosa, in order to identify potential risk factors for retinal degeneration and possible therapeutic approaches. Fatty acids from C57BL/6J and rd10 mouse retinas were extracted with Folch's method and analyzed by gas chromatography/mass spectrometry. Changes in retinal morphology were evaluated by immunohistochemistry. The rd10 mouse retina showed a decreased number of photoreceptor rows and alterations in photoreceptor morphology compared to C57BL/6J mice. The total amount of fatty acids dropped by 29.4% in the dystrophic retinas compared to C57BL/6J retinas. A positive correlation was found between the retinal content of specific fatty acids and the number of photoreceptor rows. We found that the amount of several short-chain and long-chain saturated fatty acids, as well as monounsaturated fatty acids, decreased in the retina of rd10 mice. Moreover, the content of the n-6 polyunsaturated fatty acid arachidonic acid and the n-3 polyunsaturated DHA decreased markedly in the dystrophic retina. The fall of DHA was more pronounced, hence the n-6/n-3 ratio was significantly increased in the diseased retina. The content of specific fatty acids in the retina decreased with photoreceptor degeneration in retinitis pigmentosa mice, with a remarkable reduction in DHA and other saturated and unsaturated fatty acids. These fatty acids could be essential for photoreceptor cell viability, and they should be evaluated for the design of therapeutical strategies and nutritional supplements.
Collapse
Affiliation(s)
- María José Ruiz-Pastor
- Department of Physiology, Genetics, and Microbiology, University of Alicante, Alicante, Spain
| | - Oksana Kutsyr
- Department of Physiology, Genetics, and Microbiology, University of Alicante, Alicante, Spain
| | - Pedro Lax
- Department of Physiology, Genetics, and Microbiology, University of Alicante, Alicante, Spain.
| | - Nicolás Cuenca
- Department of Physiology, Genetics, and Microbiology, University of Alicante, Alicante, Spain.
| |
Collapse
|
36
|
Jiang K, Fairless E, Kanda A, Gotoh N, Cogliati T, Li T, Swaroop A. Divergent Effects of HSP70 Overexpression in Photoreceptors During Inherited Retinal Degeneration. Invest Ophthalmol Vis Sci 2021; 61:25. [PMID: 33107904 PMCID: PMC7594617 DOI: 10.1167/iovs.61.12.25] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Purpose Disruption of proteostasis is a key event in many neurodegenerative diseases. Heat shock proteins (HSPs) participate in multiple functions associated with intracellular transport and proteostasis. We evaluated the effect of augmented HSP70 expression in mutant photoreceptors of mouse retinal degeneration models to test the hypothesis that failure to sustain HSP70 expression contributes to photoreceptor cell death. Methods We examined HSP70 expression in retinas of wild-type and mutant mice by RNA and protein analysis. A transgenic mouse line, TgCrx-Hspa1a-Flag, was generated to express FLAG-tagged full-length HSP70 protein under control of a 2.3 kb mouse Crx promoter. This line was crossed to three distinct retinal degeneration mouse models. Retinal structure and function were evaluated by histology, immunohistochemistry, and electroretinography. Results In seven different mouse models of retinal degeneration, we detected transient elevation of endogenous HSP70 expression at early stages, followed by a dramatic reduction as cell death ensues, suggesting an initial adaptive response to cellular stress. Augmented expression of HSP70 in RHOT17M mice, in which mutant rhodopsin is misfolded, marginally improved photoreceptor survival, whereas elevated HSP70 led to more severe retinal degeneration in rd10 mutants that produce a partially functional PDE6B. In Rpgrip1−/− mice that display a ciliary defect, higher HSP70 had no impact on photoreceptor survival or function. Conclusions HSP70 overexpression has divergent effects in photoreceptors determined, at least in part, by the nature of the mutant protein each model carries. Additional investigations on HSP pathways and associated chaperone networks in photoreceptors are needed before designing therapeutic strategies targeting proteostasis.
Collapse
Affiliation(s)
- Ke Jiang
- Neurobiology, Neurodegeneration, and Repair Laboratory (NNRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Elizabeth Fairless
- Neurobiology, Neurodegeneration, and Repair Laboratory (NNRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Atsuhiro Kanda
- Neurobiology, Neurodegeneration, and Repair Laboratory (NNRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Norimoto Gotoh
- Neurobiology, Neurodegeneration, and Repair Laboratory (NNRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Tiziana Cogliati
- Neurobiology, Neurodegeneration, and Repair Laboratory (NNRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Tiansen Li
- Neurobiology, Neurodegeneration, and Repair Laboratory (NNRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Anand Swaroop
- Neurobiology, Neurodegeneration, and Repair Laboratory (NNRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
37
|
Berkowitz BA, Podolsky RH, Childers KL, Roche SL, Cotter TG, Graffice E, Harp L, Sinan K, Berri AM, Schneider M, Qian H, Gao S, Roberts R. Rod Photoreceptor Neuroprotection in Dark-Reared Pde6brd10 Mice. Invest Ophthalmol Vis Sci 2021; 61:14. [PMID: 33156341 PMCID: PMC7671864 DOI: 10.1167/iovs.61.13.14] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Purpose The purpose of this study was to test the hypothesis that anti-oxidant and / or anti-inflammation drugs that suppress rod death in cyclic light-reared Pde6brd10 mice are also effective in dark-reared Pde6brd10 mice. Methods In untreated dark-reared Pde6brd10 mice at post-natal (P) days 23 to 24, we measured the outer nuclear layer (ONL) thickness (histology) and dark-light thickness difference in external limiting membrane-retinal pigment epithelium (ELM-RPE) (optical coherence tomography [OCT]), retina layer oxidative stress (QUEnch-assiSTed [QUEST] magnetic resonance imaging [MRI]); and microglia/macrophage-driven inflammation (immunohistology). In dark-reared P50 Pde6brd10 mice, ONL thickness was measured (OCT) in groups given normal chow or chow admixed with methylene blue (MB) + Norgestrel (anti-oxidant, anti-inflammatory), or MB or Norgestrel separately. Results P24 Pde6brd10 mice showed no significant dark-light ELM-RPE response in superior and inferior retina consistent with high cGMP levels. Norgestrel did not significantly suppress the oxidative stress of Pde6brd10 mice that is only found in superior central outer retina of males at P23. Overt rod degeneration with microglia/macrophage activation was observed but only in the far peripheral superior retina in male and female P23 Pde6brd10 mice. Significant rod protection was measured in female P50 Pde6brd10 mice given 5 mg/kg/day MB + Norgestrel diet; no significant benefit was seen with MB chow or Norgestrel chow alone, nor in similarly treated male mice. Conclusions In early rod degeneration in dark-reared Pde6brd10 mice, little evidence is found in central retina for spatial associations among biomarkers of the PDE6B mutation, oxidative stress, and rod death; neuroprotection at P50 was limited to a combination of anti-oxidant/anti-inflammation treatment in a sex-specific manner.
Collapse
Affiliation(s)
- Bruce A Berkowitz
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Robert H Podolsky
- Beaumont Research Institute, Beaumont Health, Royal Oak, Michigan, United States
| | - Karen Lins Childers
- Beaumont Research Institute, Beaumont Health, Royal Oak, Michigan, United States
| | - Sarah L Roche
- Cell Development and Disease Laboratory, Biochemistry Department, Biosciences Institute, University College Cork, Cork, Ireland
| | - Thomas G Cotter
- Cell Development and Disease Laboratory, Biochemistry Department, Biosciences Institute, University College Cork, Cork, Ireland
| | - Emma Graffice
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Lamis Harp
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Kenan Sinan
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Ali M Berri
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Michael Schneider
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Haohua Qian
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Shasha Gao
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Robin Roberts
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| |
Collapse
|
38
|
Das S, Chen Y, Yan J, Christensen G, Belhadj S, Tolone A, Paquet-Durand F. The role of cGMP-signalling and calcium-signalling in photoreceptor cell death: perspectives for therapy development. Pflugers Arch 2021; 473:1411-1421. [PMID: 33864120 PMCID: PMC8370896 DOI: 10.1007/s00424-021-02556-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/04/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022]
Abstract
The second messengers, cGMP and Ca2+, have both been implicated in retinal degeneration; however, it is still unclear which of the two is most relevant for photoreceptor cell death. This problem is exacerbated by the close connections and crosstalk between cGMP-signalling and calcium (Ca2+)-signalling in photoreceptors. In this review, we summarize key aspects of cGMP-signalling and Ca2+-signalling relevant for hereditary photoreceptor degeneration. The topics covered include cGMP-signalling targets, the role of Ca2+ permeable channels, relation to energy metabolism, calpain-type proteases, and how the related metabolic processes may trigger and execute photoreceptor cell death. A focus is then put on cGMP-dependent mechanisms and how exceedingly high photoreceptor cGMP levels set in motion cascades of Ca2+-dependent and independent processes that eventually bring about photoreceptor cell death. Finally, an outlook is given into mutation-independent therapeutic approaches that exploit specific features of cGMP-signalling. Such approaches might be combined with suitable drug delivery systems for translation into clinical applications.
Collapse
Affiliation(s)
- Soumyaparna Das
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| | - Yiyi Chen
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| | - Jie Yan
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| | - Gustav Christensen
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| | - Soumaya Belhadj
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| | - Arianna Tolone
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| | - François Paquet-Durand
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany.
| |
Collapse
|
39
|
Kutsyr O, Maestre-Carballa L, Lluesma-Gomez M, Martinez-Garcia M, Cuenca N, Lax P. Retinitis pigmentosa is associated with shifts in the gut microbiome. Sci Rep 2021; 11:6692. [PMID: 33758301 PMCID: PMC7988170 DOI: 10.1038/s41598-021-86052-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/10/2021] [Indexed: 12/14/2022] Open
Abstract
The gut microbiome is known to influence the pathogenesis and progression of neurodegenerative diseases. However, there has been relatively little focus upon the implications of the gut microbiome in retinal diseases such as retinitis pigmentosa (RP). Here, we investigated changes in gut microbiome composition linked to RP, by assessing both retinal degeneration and gut microbiome in the rd10 mouse model of RP as compared to control C57BL/6J mice. In rd10 mice, retinal responsiveness to flashlight stimuli and visual acuity were deteriorated with respect to observed in age-matched control mice. This functional decline in dystrophic animals was accompanied by photoreceptor loss, morphologic anomalies in photoreceptor cells and retinal reactive gliosis. Furthermore, 16S rRNA gene amplicon sequencing data showed a microbial gut dysbiosis with differences in alpha and beta diversity at the genera, species and amplicon sequence variants (ASV) levels between dystrophic and control mice. Remarkably, four fairly common ASV in healthy gut microbiome belonging to Rikenella spp., Muribaculaceace spp., Prevotellaceae UCG-001 spp., and Bacilli spp. were absent in the gut microbiome of retinal disease mice, while Bacteroides caecimuris was significantly enriched in mice with RP. The results indicate that retinal degenerative changes in RP are linked to relevant gut microbiome changes. The findings suggest that microbiome shifting could be considered as potential biomarker and therapeutic target for retinal degenerative diseases.
Collapse
Affiliation(s)
- Oksana Kutsyr
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Lucía Maestre-Carballa
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Mónica Lluesma-Gomez
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Manuel Martinez-Garcia
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain.
| | - Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
- Institute Ramón Margalef, University of Alicante, Alicante, Spain
| | - Pedro Lax
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain.
| |
Collapse
|
40
|
Sildenafil-evoked photoreceptor oxidative stress in vivo is unrelated to impaired visual performance in mice. PLoS One 2021; 16:e0245161. [PMID: 33661941 PMCID: PMC7932139 DOI: 10.1371/journal.pone.0245161] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 12/22/2020] [Indexed: 12/22/2022] Open
Abstract
Purpose The phosphodiesterase inhibitor sildenafil is a promising treatment for neurodegenerative disease, but it can cause oxidative stress in photoreceptors ex vivo and degrade visual performance in humans. Here, we test the hypotheses that in wildtype mice sildenafil causes i) wide-spread photoreceptor oxidative stress in vivo that is linked with ii) impaired vision. Methods In dark or light-adapted C57BL/6 mice ± sildenafil treatment, the presence of oxidative stress was evaluated in retina laminae in vivo by QUEnch-assiSTed (QUEST) magnetic resonance imaging, in the subretinal space in vivo by QUEST optical coherence tomography, and in freshly excised retina by a dichlorofluorescein assay. Visual performance indices were also evaluated by QUEST optokinetic tracking. Results In light-adapted mice, 1 hr post-sildenafil administration, oxidative stress was most evident in the superior peripheral outer retina on both in vivo and ex vivo examinations; little evidence was noted for central retina oxidative stress in vivo and ex vivo. In dark-adapted mice 1 hr after sildenafil, no evidence for outer retina oxidative stress was found in vivo. Evidence for sildenafil-induced central retina rod cGMP accumulation was suggested as a panretinally thinner, dark-like subretinal space thickness in light-adapted mice at 1 hr but not 5 hr post-sildenafil. Cone-based visual performance was impaired by 5 hr post-sildenafil and not corrected with anti-oxidants; vision was normal at 1 hr and 24 hr post-sildenafil. Conclusions The sildenafil-induced spatiotemporal pattern of oxidative stress in photoreceptors dominated by rods was unrelated to impairment of cone-based visual performance in wildtype mice.
Collapse
|
41
|
Yang P, Lockard R, Titus H, Hiblar J, Weller K, Wafai D, Weleber RG, Duvoisin RM, Morgans CW, Pennesi ME. Suppression of cGMP-Dependent Photoreceptor Cytotoxicity With Mycophenolate Is Neuroprotective in Murine Models of Retinitis Pigmentosa. Invest Ophthalmol Vis Sci 2021; 61:25. [PMID: 32785677 PMCID: PMC7441375 DOI: 10.1167/iovs.61.10.25] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Purpose To determine the effect of mycophenolate mofetil (MMF) on retinal degeneration on two mouse models of retinitis pigmentosa. Methods Intraperitoneal injections of MMF were administered daily in rd10 and c57 mice starting at postoperative day 12 (P12) and rd1 mice starting at P8. The effect of MMF was assessed with optical coherence tomography, immunohistochemistry, electroretinography, and OptoMotry. Whole retinal cyclic guanosine monophosphate (cGMP) and mycophenolic acid levels were quantified with mass spectrometry. Photoreceptor cGMP cytotoxicity was evaluated with cell counts of cGMP immunostaining. Results MMF treatment significantly delays the onset of retinal degeneration and cGMP-dependent photoreceptor cytotoxicity in rd10 and rd1 mice, albeit a more modest effect in the latter. In rd10 mice, treatment with MMF showed robust preservation of the photoreceptors up to P22 with associated suppression of cGMP immunostaining and microglial activation; The neuroprotective effect diminished after P22, but outer retinal thickness was still significantly thicker by P35 and OptoMotry response was significantly better up to P60. Whereas cGMP immunostaining of the photoreceptors were present in rd10 and rd1 mice, hyperphysiological whole retinal cGMP levels were observed only in rd1 mice. Conclusions Early treatment with MMF confers potent neuroprotection in two animal models of RP by suppressing the cGMP-dependent common pathway for photoreceptor cell death. The neuroprotective effect of MMF on cGMP-dependent cytotoxicity occurs independently of the presence of hyperphysiological whole retinal cGMP levels. Thus our data suggest that MMF may be an important new class of neuroprotective agent that could be useful in the treatment of patients with RP.
Collapse
Affiliation(s)
- Paul Yang
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Rachel Lockard
- School of Medicine, Oregon Health & Science University, Portland, Oregon, United States
| | - Hope Titus
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Jordan Hiblar
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Kyle Weller
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Dahlia Wafai
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Richard G Weleber
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Robert M Duvoisin
- Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon, United States
| | - Catherine W Morgans
- Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon, United States
| | - Mark E Pennesi
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| |
Collapse
|
42
|
Kutsyr O, Sánchez-Sáez X, Martínez-Gil N, de Juan E, Lax P, Maneu V, Cuenca N. Gradual Increase in Environmental Light Intensity Induces Oxidative Stress and Inflammation and Accelerates Retinal Neurodegeneration. Invest Ophthalmol Vis Sci 2021; 61:1. [PMID: 32744596 PMCID: PMC7441298 DOI: 10.1167/iovs.61.10.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose Retinitis pigmentosa (RP) is a blinding neurodegenerative disease of the retina that can be affected by many factors. The present study aimed to analyze the effect of different environmental light intensities in rd10 mice retina. Methods C57BL/6J and rd10 mice were bred and housed under three different environmental light intensities: scotopic (5 lux), mesopic (50 lux), and photopic (300 lux). Visual function was studied using electroretinography and optomotor testing. The structural and morphological integrity of the retinas was evaluated by optical coherence tomography imaging and immunohistochemistry. Additionally, inflammatory processes and oxidative stress markers were analyzed by flow cytometry and western blotting. Results When the environmental light intensity was higher, retinal function decreased in rd10 mice and was accompanied by light-dependent photoreceptor loss, followed by morphological alterations, and synaptic connectivity loss. Moreover, light-dependent retinal degeneration was accompanied by an increased number of inflammatory cells, which became more activated and phagocytic, and by an exacerbated reactive gliosis. Furthermore, light-dependent increment in oxidative stress markers in rd10 mice retina pointed to a possible mechanism for light-induced photoreceptor degeneration. Conclusions An increase in rd10 mice housing light intensity accelerates retinal degeneration, activating cell death, oxidative stress pathways, and inflammatory cells. Lighting intensity is a key factor in the progression of retinal degeneration, and standardized lighting conditions are advisable for proper analysis and interpretation of experimental results from RP animal models, and specifically from rd10 mice. Also, it can be hypothesized that light protection could be an option to slow down retinal degeneration in some cases of RP.
Collapse
|
43
|
Ryals RC, Huang SJ, Wafai D, Bernert C, Steele W, Six M, Bonthala S, Titus H, Yang P, Gillingham M, Pennesi ME. A Ketogenic & Low-Protein Diet Slows Retinal Degeneration in rd10 Mice. Transl Vis Sci Technol 2020; 9:18. [PMID: 33117609 PMCID: PMC7571290 DOI: 10.1167/tvst.9.11.18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/23/2020] [Indexed: 01/01/2023] Open
Abstract
Purpose Treatments that delay retinal cell death regardless of genetic causation are needed for inherited retinal degeneration (IRD) patients. The ketogenic diet is a high-fat, low-carbohydrate diet, used to treat epilepsy, and has beneficial effects for neurodegenerative diseases. This study aimed to determine whether the ketogenic diet could slow retinal degeneration. Methods Early weaned, rd10 and wild-type (WT) mice were placed on either standard chow, a ketogenic diet, or a ketogenic & low-protein diet. From postnatal day (PD) 23 to PD50, weight and blood β-hydroxybutyrate levels were recorded. Retinal thickness, retinal function, and visual performance were measured via optical coherence tomography, electroretinography (ERG), and optokinetic tracking (OKT). At PD40, serum albumin, rhodopsin protein, and phototransduction gene expression were measured. Results Both ketogenic diets elicited a systemic induction of ketosis. However, rd10 mice on the ketogenic & low-protein diet had significant increases in photoreceptor thickness, ERG amplitudes, and OKT thresholds, whereas rd10 mice on the ketogenic diet showed no photoreceptor preservation. In both rd10 and WT mice, the ketogenic & low-protein diet was associated with abnormal weight gain and decreases in serum albumin levels, 27% and 56%, respectively. In WT mice, the ketogenic & low-protein diet was also associated with an ∼20% to 30% reduction in ERG amplitudes. Conclusions The ketogenic & low-protein diet slowed retinal degeneration in a clinically relevant IRD model. In WT mice, the ketogenic & low-protein diet was associated with a decrease in phototransduction and serum albumin, which could serve as a protective mechanism in the rd10 model. Although ketosis was associated with protection, its role remains unclear. Translational Relevance Neuroprotective mechanisms associated with the ketogenic & low-protein diet have potential to slow retinal degeneration.
Collapse
Affiliation(s)
- Renee C Ryals
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Samuel J Huang
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Dahlia Wafai
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Claire Bernert
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - William Steele
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Makayla Six
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Shasank Bonthala
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Hope Titus
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Paul Yang
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Melanie Gillingham
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - Mark E Pennesi
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
44
|
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: 36] [Impact Index Per Article: 7.2] [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
|
45
|
Kim TH, Wang B, Lu Y, Son T, Yao X. Functional optical coherence tomography enables in vivo optoretinography of photoreceptor dysfunction due to retinal degeneration. BIOMEDICAL OPTICS EXPRESS 2020; 11:5306-5320. [PMID: 33014616 PMCID: PMC7510876 DOI: 10.1364/boe.399334] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/20/2020] [Accepted: 08/20/2020] [Indexed: 05/16/2023]
Abstract
Stimulus-evoked intrinsic optical signal (IOS), which occurs almost immediately after the onset of retinal stimulus has been observed in retinal photoreceptors, promises to be a unique biomarker for objective optoretinography (ORG) of photoreceptor function. We report here the first-time in vivo ORG detection of photoreceptor dysfunction due to retinal degeneration. A custom-designed optical coherence tomography (OCT) was employed for longitudinal ORG monitoring of photoreceptor-IOS distortions in retinal degeneration mice. Depth-resolved OCT analysis confirmed the outer segment (OS) as the physical source of the photoreceptor-IOS. Comparative ERG measurement verified the phototransduction activation as the physiological correlator of the photoreceptor-IOS. Histological examination revealed disorganized OS discs, i.e. the pathological origin of the photoreceptor-IOS distortion.
Collapse
Affiliation(s)
- Tae-Hoon Kim
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Benquan Wang
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Yiming Lu
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Taeyoon Son
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Xincheng Yao
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
46
|
Reingruber J, Ingram NT, Griffis KG, Fain GL. A kinetic analysis of mouse rod and cone photoreceptor responses. J Physiol 2020; 598:3747-3763. [PMID: 32557629 PMCID: PMC7484371 DOI: 10.1113/jp279524] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 06/08/2020] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS Most vertebrate eyes have rods for dim-light vision and cones for brighter light and higher temporal sensitivity. Rods evolved from cone-like precursors through expression of different transduction genes or the same genes at different expression levels, but we do not know which molecular differences were most important. We approached this problem by analysing rod and cone responses with the same model but with different values for model parameters. We showed that, in addition to outer-segment volume, the most important differences between rods and cones are: (1) decreased transduction gain, reflecting smaller amplification in the G-protein cascade; (2) a faster rate of turnover of the second messenger cGMP in darkness; and (3) an accelerated rate of decay of the effector enzyme phosphodiesterase and perhaps also of activated visual pigment. We believe our analysis has identified the principal alterations during evolution responsible for the duplex retina. ABSTRACT Most vertebrates have rod and cone photoreceptors, which differ in their sensitivity and response kinetics. We know that rods evolved from cone-like precursors through the expression of different transduction genes or the same genes at different levels, but we do not know which molecular differences were most important. We have approached this problem in mouse retina by analysing the kinetic differences between rod flash responses and recent voltage-clamp recordings of cone flash responses, using a model incorporating the principal features of photoreceptor transduction. We apply a novel method of analysis using the log-transform of the current, and we ask which of the model's dynamic parameters need be changed to transform the flash response of a rod into that of a cone. The most important changes are a decrease in the gain of the response, reflecting a reduction in amplification of the transduction cascade; an increase in the rate of turnover of cGMP in darkness; and an increase in the rate of decay of activated phosphodiesterase, with perhaps also an increase in the rate of decay of light-activated visual pigment. Although we cannot exclude other differences, and in particular alterations in the Ca2+ economy of the photoreceptors, we believe that we have identified the kinetic parameters principally responsible for the differences in the flash responses of the two kinds of photoreceptors, which were likely during evolution to have resulted in the duplex retina.
Collapse
Affiliation(s)
- Jürgen Reingruber
- Institut de Biologie de l’École Normale Supérieure, 46 rue d’Ulm, 75005 Paris, France
| | - Norianne T. Ingram
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095–7239, USA
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, CA 90095–7000, USA
| | - Khris G. Griffis
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, CA 90095–7000, USA
| | - Gordon L. Fain
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095–7239, USA
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, CA 90095–7000, USA
| |
Collapse
|
47
|
Li H, Lian L, Liu B, Chen Y, Yang J, Jian S, Zhou J, Xu Y, Ma X, Qu J, Hou L. KIT ligand protects against both light-induced and genetic photoreceptor degeneration. eLife 2020; 9:51698. [PMID: 32242818 PMCID: PMC7170656 DOI: 10.7554/elife.51698] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 04/02/2020] [Indexed: 12/25/2022] Open
Abstract
Photoreceptor degeneration is a major cause of blindness and a considerable health burden during aging but effective therapeutic or preventive strategies have not so far become readily available. Here, we show in mouse models that signaling through the tyrosine kinase receptor KIT protects photoreceptor cells against both light-induced and inherited retinal degeneration. Upon light damage, photoreceptor cells upregulate Kit ligand (KITL) and activate KIT signaling, which in turn induces nuclear accumulation of the transcription factor NRF2 and stimulates the expression of the antioxidant gene Hmox1. Conversely, a viable Kit mutation promotes light-induced photoreceptor damage, which is reversed by experimental expression of Hmox1. Furthermore, overexpression of KITL from a viral AAV8 vector prevents photoreceptor cell death and partially restores retinal function after light damage or in genetic models of human retinitis pigmentosa. Hence, application of KITL may provide a novel therapeutic avenue for prevention or treatment of retinal degenerative diseases.
Collapse
Affiliation(s)
- Huirong Li
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Lili Lian
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Bo Liu
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yu Chen
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Jinglei Yang
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Shuhui Jian
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jiajia Zhou
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ying Xu
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Xiaoyin Ma
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Jia Qu
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Ling Hou
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
48
|
Gao ML, Lei XL, Han F, He KW, Jin SQ, Zhang YY, Jin ZB. Patient-Specific Retinal Organoids Recapitulate Disease Features of Late-Onset Retinitis Pigmentosa. Front Cell Dev Biol 2020; 8:128. [PMID: 32211407 PMCID: PMC7068133 DOI: 10.3389/fcell.2020.00128] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 02/13/2020] [Indexed: 12/18/2022] Open
Abstract
Although an increasing number of disease genes have been identified, the exact cellular mechanisms of retinitis pigmentosa (RP) remain largely unclear. Retinal organoids (ROs) derived from the induced pluripotent stem cells (iPSCs) of patients provide a potential but unvalidated platform for deciphering disease mechanisms and an advantageous tool for preclinical testing of new treatments. Notably, early-onset RP has been extensively recapitulated by patient-iPSC-derived ROs. However, it remains a challenge to model late-onset disease in a dish due to its chronicity, complexity, and instability. Here, we generated ROs from late-onset RP proband-derived iPSCs harboring a PDE6B mutation. Transcriptome analysis revealed a remarkably distinct gene expression profile in the patient ROs at differentiation day (D) 230. Changes in the expression genes regulating cGMP hydrolysis prompted the elevation of cGMP levels, which was verified by a cGMP enzyme-linked immunosorbent assay (ELISA) in patient ROs. Furthermore, significantly higher cGMP levels in patient ROs than in control ROs at D193 and D230 might lead to impaired formation of synaptic connections and the connecting cilium in photoreceptor cells. In this study, we established the first late-onset RP model with a consistent phenotype using an in vitro cell culture system and provided new insights into the PDE6B-related mechanism of RP.
Collapse
Affiliation(s)
- Mei-Ling Gao
- Laboratory of Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Center for International Research in Regenerative Medicine and Neurogenetics, National Clinical Research Center for Ophthalmology, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, China
| | - Xin-Lan Lei
- Laboratory of Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Center for International Research in Regenerative Medicine and Neurogenetics, National Clinical Research Center for Ophthalmology, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, China
| | - Fang Han
- Laboratory of Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Center for International Research in Regenerative Medicine and Neurogenetics, National Clinical Research Center for Ophthalmology, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, China
| | - Kai-Wen He
- Laboratory of Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Center for International Research in Regenerative Medicine and Neurogenetics, National Clinical Research Center for Ophthalmology, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, China
| | - Si-Qian Jin
- Laboratory of Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Center for International Research in Regenerative Medicine and Neurogenetics, National Clinical Research Center for Ophthalmology, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, China
| | - You-You Zhang
- Laboratory of Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Center for International Research in Regenerative Medicine and Neurogenetics, National Clinical Research Center for Ophthalmology, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, China
| | - Zi-Bing Jin
- Laboratory of Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Center for International Research in Regenerative Medicine and Neurogenetics, National Clinical Research Center for Ophthalmology, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, China
| |
Collapse
|
49
|
Zhu D, Xie M, Gademann F, Cao J, Wang P, Guo Y, Zhang L, Su T, Zhang J, Chen J. Protective effects of human iPS-derived retinal pigmented epithelial cells on retinal degenerative disease. Stem Cell Res Ther 2020; 11:98. [PMID: 32131893 PMCID: PMC7055119 DOI: 10.1186/s13287-020-01608-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/31/2020] [Accepted: 02/14/2020] [Indexed: 01/04/2023] Open
Abstract
Background Retinitis pigmentosa (RP) is an inherited retinal disease characterized by progressive loss of photoreceptor cells. This study aim at exploring the effect of retinal pigment epithelium (RPE) derived from human-induced pluripotent stem cell (hiPSC-RPE) on the retina of retinal degeneration 10 (rd10) mice, which are characterized with progressive photoreceptor death. Methods We generated RPE from hiPSCs by sequential supplementation with retinal-inducing factors and RPE specification signaling factors. The three-dimensional (3D) spheroid culture method was used to obtain optimal injectable hiPSC-RPE cells. Subretinal space transplantation was conducted to deliver hiPSC-RPE cells into the retina of rd10 mice. Neurotrophic factor secretion from transplanted hiPSC-RPE cells was detected by enzyme-linked immunosorbent assay (ELISA). Immunostaining, Western blotting, electroretinography (ERG), and visual behavior testing were performed to determine the effects of hiPSC-RPE on the retinal visual function in rd10 mice. Results Our data demonstrated that hiPSC-RPE cells exhibited classic RPE properties and phenotype after the sequential RPE induction from hiPSCs. hiPSC-RPE cells co-cultured with mouse retinal explants or retinal ganglion cells 5 (RGC5) exhibited decreased apoptosis. The viability and functional properties of hiPSC-RPE cells were enhanced by 3D spheroid culture. Transplanted hiPSC-derived RPE cells were identified by immunostaining with human nuclear antigen staining in the retina of rd10 14 days after subretinal space injection. The pigment epithelium-derived factor level was increased significantly. The expression of CD68, microglial activation marker, reduced after transplantation. The light avoidance behavior and ERG visual function in rd10 mice improved by the transplantation of hiPSC-RPE cells. Conclusion Our findings suggest that injectable hiPSC-RPE cells after 3D spheroid culture can rescue the structure and function of photoreceptors by sub-retinal transplantation, which lay the foundation for future clinical cell therapy to treat RP and other retinal degeneration diseases.
Collapse
Affiliation(s)
- Deliang Zhu
- Key Laboratory of Optoelectronic Information and Sensing Technologies of Guangdong Higher Educational Institutes, Jinan University, Guangzhou, China.,Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| | - Mengyuan Xie
- Key Laboratory of Optoelectronic Information and Sensing Technologies of Guangdong Higher Educational Institutes, Jinan University, Guangzhou, China
| | - Fabian Gademann
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| | - Jixing Cao
- Eye Institute, Medical College of Jinan University, Guangzhou, China
| | - Peiyuan Wang
- Eye Institute, Medical College of Jinan University, Guangzhou, China
| | - Yonglong Guo
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| | - Lan Zhang
- Eye Institute, Medical College of Jinan University, Guangzhou, China
| | - Ting Su
- Eye Institute, Medical College of Jinan University, Guangzhou, China
| | - Jun Zhang
- Key Laboratory of Optoelectronic Information and Sensing Technologies of Guangdong Higher Educational Institutes, Jinan University, Guangzhou, China.
| | - Jiansu Chen
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China. .,Eye Institute, Medical College of Jinan University, Guangzhou, China. .,Aier Eye Institute, Furong Middle Road, Changsha, China.
| |
Collapse
|
50
|
Sundar JC, Munezero D, Bryan-Haring C, Saravanan T, Jacques A, Ramamurthy V. Rhodopsin signaling mediates light-induced photoreceptor cell death in rd10 mice through a transducin-independent mechanism. Hum Mol Genet 2020; 29:394-406. [PMID: 31925423 PMCID: PMC7015845 DOI: 10.1093/hmg/ddz299] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/22/2019] [Accepted: 12/02/2019] [Indexed: 01/08/2023] Open
Abstract
Retinitis pigmentosa (RP) is a debilitating blinding disease affecting over 1.5 million people worldwide, but the mechanisms underlying this disease are not well understood. One of the common models used to study RP is the retinal degeneration-10 (rd10) mouse, which has a mutation in Phosphodiesterase-6b (Pde6b) that causes a phenotype mimicking the human disease. In rd10 mice, photoreceptor cell death occurs with exposure to normal light conditions, but as demonstrated in this study, rearing these mice in dark preserves their retinal function. We found that inactivating rhodopsin signaling protected photoreceptors from degeneration suggesting that the pathway activated by this G-protein-coupled receptor is causing light-induced photoreceptor cell death in rd10 mice. However, inhibition of transducin signaling did not prevent the loss of photoreceptors in rd10 mice reared under normal light conditions implying that the degeneration caused by rhodopsin signaling is not mediated through its canonical G-protein transducin. Inexplicably, loss of transducin in rd10 mice also led to photoreceptor cell death in darkness. Furthermore, we found that the rd10 mutation in Pde6b led to a reduction in the assembled PDE6αβγ2 complex, which was corroborated by our data showing mislocalization of the γ subunit. Based on our findings and previous studies, we propose a model where light activates a non-canonical pathway mediated by rhodopsin but independent of transducin that sensitizes cyclic nucleotide gated channels to cGMP and causes photoreceptor cell death. These results generate exciting possibilities for treatment of RP patients without affecting their vision or the canonical phototransduction cascade.
Collapse
Affiliation(s)
- Jesse C Sundar
- Departments of Biochemistry, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26505, USA
| | - Daniella Munezero
- Departments of Ophthalmology and Visual Sciences, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26505, USA
| | - Caitlyn Bryan-Haring
- Departments of Biochemistry, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26505, USA
| | - Thamaraiselvi Saravanan
- Departments of Ophthalmology and Visual Sciences, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26505, USA
| | - Angelica Jacques
- Departments of Ophthalmology and Visual Sciences, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26505, USA
| | - Visvanathan Ramamurthy
- Departments of Biochemistry, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26505, USA
- Departments of Ophthalmology and Visual Sciences, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26505, USA
- Departments of Neuroscience, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26505, USA
| |
Collapse
|