1
|
Bernardo-Colón A, Bighinati A, Parween S, Debnath S, Piano I, Adani E, Corsi F, Gargini C, Vergara N, Marigo V, Patricia Becerra S. H105A peptide eye drops promote photoreceptor survival in murine and human models of retinal degeneration. COMMUNICATIONS MEDICINE 2025; 5:81. [PMID: 40118996 PMCID: PMC11928584 DOI: 10.1038/s43856-025-00789-8] [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: 06/26/2024] [Accepted: 02/27/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND Photoreceptor death leads to inherited blinding retinal diseases, such as retinitis pigmentosa (RP). As disease progression often outpaces therapeutic advances, developing effective treatments is urgent. This study evaluates the efficacy of small peptides derived from pigment epithelium-derived factor (PEDF), which are known to restrict common cell death pathways associated with retinal diseases. METHODS We tested chemically synthesized peptides (17-mer and H105A) with affinity for the PEDF receptor, PEDF-R, delivered as eye drops to two RP mouse models: rd10 (phosphodiesterase 6b mutation) and RhoP23H/+ (rhodopsin P23H mutation). Additionally, we engineered AAV-H105A vectors for intravitreal delivery in RhoP23H/+ mice. To assess peptide effects in human tissue, we used retinal organoids exposed to cigarette smoke extract, a model of oxidative stress. Photoreceptor survival, morphology and function were evaluated. RESULTS Here we show that peptides 17-mer and H105A delivered via eye drops successfully reach the retina, promote photoreceptor survival, and improve retinal function in both RP mouse models. Intravitreal delivery of a AAV-H105A vector delays photoreceptor degeneration in RhoP23H/+ mice up to six months. In human retinal organoids, peptide H105A specifically prevents photoreceptor death induced by oxidative stress, a contributing factor to RP progression. CONCLUSIONS PEDF peptide-based eye drops offer a promising, minimally invasive therapy to prevent photoreceptor degeneration in retinal disorders, with a favorable safety profile.
Collapse
Grants
- Z01 EY000306 Intramural NIH HHS
- Intramural Research Program of the National Eye Institute, National Institutes of Health, United States of America (Project #EY000306, SPB); the Prevention of Blindness Society (SPB); Fondazione Telethon (Project #GGP19113, VM), the National Center for “Gene Therapy and Drugs based on RNA Technology” cod. Progetto CN00000041 and “Health Extended Alliance for Innovative Therapies, Advanced Lab-research, and Integrated Approaches of Precision Medicine - HEAL ITALIA” tematica 6 “Innovative diagnostics and therapies in precision medicine” cod. Progetto PE0000019 PIANO NAZIONALE DI RIPRESA E RESILIENZA (PNRR) – MISSIONE 4 “Istruzione Ricerca” COMPONENTE 2, “Dalla ricerca all’impresa” INVESTIMENTO 1.4, “Potenziamento strutture di ricerca e creazione di "campioni nazionali di R&S” su alcune Key enabling technologies”, finanziato dall’Unione europea – NextGenerationEU (VM and AB); The CellSight Development Fund (NV); and a Challenge Grant to the Department of Ophthalmology at the University of Colorado from Research to Prevent Blindness (NV).
Collapse
Affiliation(s)
- Alexandra Bernardo-Colón
- Section of Protein Structure and Function, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andrea Bighinati
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy
| | - Shama Parween
- CellSight Ocular Stem Cell and Regeneration Program, Sue Anschutz-Rodgers Eye Center, University of Colorado Anschutz Medical Campus; Aurora, Colorado, USA
| | - Subrata Debnath
- Section of Protein Structure and Function, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ilaria Piano
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy
| | - Elisa Adani
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy
| | - Francesca Corsi
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy
| | - Claudia Gargini
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy
| | - Natalia Vergara
- CellSight Ocular Stem Cell and Regeneration Program, Sue Anschutz-Rodgers Eye Center, University of Colorado Anschutz Medical Campus; Aurora, Colorado, USA
- Gates Center for Regenerative Medicine, Linda Crnic Institute for Down Syndrome and University of Colorado Alzheimer's and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Valeria Marigo
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy.
| | - S Patricia Becerra
- Section of Protein Structure and Function, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
2
|
Wijnholds J. miR-429 RNA therapy as generic strategy to protect against photoreceptor loss. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102477. [PMID: 40027881 PMCID: PMC11872468 DOI: 10.1016/j.omtn.2025.102477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Affiliation(s)
- Jan Wijnholds
- Department of Ophthalmology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, the Netherlands
- Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
| |
Collapse
|
3
|
Azam M, Jastrzebska B. Mechanisms of Rhodopsin-Related Inherited Retinal Degeneration and Pharmacological Treatment Strategies. Cells 2025; 14:49. [PMID: 39791750 PMCID: PMC11720364 DOI: 10.3390/cells14010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/18/2024] [Accepted: 12/21/2024] [Indexed: 01/12/2025] Open
Abstract
Retinitis pigmentosa (RP) is a hereditary disease characterized by progressive vision loss ultimately leading to blindness. This condition is initiated by mutations in genes expressed in retinal cells, resulting in the degeneration of rod photoreceptors, which is subsequently followed by the loss of cone photoreceptors. Mutations in various genes expressed in the retina are associated with RP. Among them, mutations in the rhodopsin gene (RHO) are the most common cause of this condition. Due to the involvement of numerous genes and multiple mutations in a single gene, RP is a highly heterogeneous disease making the development of effective treatments particularly challenging. The progression of this disease involves complex cellular responses to restore cellular homeostasis, including the unfolded protein response (UPR) signaling, autophagy, and various cell death pathways. These mechanisms, however, often fail to prevent photoreceptor cell degradation and instead contribute to cell death under certain conditions. Current research focuses on the pharmacological modulation of the components of these pathways and the direct stabilization of mutated receptors as potential treatment strategies. Despite these efforts, the intricate interplay between these mechanisms and the diverse causative mutations involved has hindered the development of effective treatments. Advancing our understanding of the interactions between photoreceptor cell death mechanisms and the specific genetic mutations driving RP is critical to accelerate the discovery and development of therapeutic strategies for this currently incurable disease.
Collapse
Affiliation(s)
- Maria Azam
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| | - Beata Jastrzebska
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
- Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| |
Collapse
|
4
|
Carleton M, Oesch NW. Bridging the gap of vision restoration. Front Cell Neurosci 2024; 18:1502473. [PMID: 39640234 PMCID: PMC11617155 DOI: 10.3389/fncel.2024.1502473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024] Open
Abstract
Retinitis pigmentosa (RP) and Age-Related Macular Degeneration (AMD) are similar in that both result in photoreceptor degeneration leading to permanent progressive vision loss. This affords the possibility of implementing vision restoration techniques, where light signaling is restored to spared retinal circuitry to recreate vision. There are far more AMD patients (Wong et al., 2014), yet more resources have been put towards researching and developing vision restoration strategies for RP despite it rarity, because of the tractability of RP disease models. The hope is that these therapies will extend to the AMD population, however, many questions remain about how the implementation of prosthetic or optogenetic vision restoration technologies will translate between RP and AMD patients. In this review, we discuss the difference and similarities of RP and AMD with a focus on aspects expected to impact vision restoration strategies, and we identify key gaps in knowledge needed to further improve vision restoration technologies for a broad patient population.
Collapse
Affiliation(s)
- Maya Carleton
- Department of Psychology, University of California San Diego, La Jolla, CA, United States
| | - Nicholas W. Oesch
- Department of Psychology, University of California San Diego, La Jolla, CA, United States
- Department of Ophthalmology, University of California San Diego, La Jolla, CA, United States
- Neuroscience Graduate Program, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
5
|
Wan H, Chen H, Liu J, Yang B, Zhang Y, Bai Y, Chen X, Wang J, Liu T, Zhang Y, Hua Q. PARP1 inhibition prevents oxidative stress in age-related hearing loss via PAR-Ca 2+-AIF axis in cochlear strial marginal cells. Free Radic Biol Med 2024; 220:222-235. [PMID: 38735540 DOI: 10.1016/j.freeradbiomed.2024.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/02/2024] [Accepted: 05/09/2024] [Indexed: 05/14/2024]
Abstract
Studies have highlighted oxidative damage in the inner ear as a critical pathological basis for sensorineural hearing loss, especially the presbycusis. Poly(ADP-ribose) polymerase-1 (PARP1) activation responds to oxidative stress-induced DNA damage with pro-repair and pro-death effects resembling two sides of the same coin. PARP1-related cell death, known as parthanatos, whose underlying mechanisms are attractive research hotspots but remain to be clarified. In this study, we observed that aged rats showed stria vascularis degeneration and oxidative damage, and PARP1-dependent cell death was prominent in age-related cochlear disorganization and dysfunction. Based on oxidative stress model of primary cultured stria marginal cells (MCs), we revealed that upregulated PARP1 and PAR (Poly(ADP-ribose)) polymers are responsible for MCs oxidative death with high mitochondrial permeability transition pore (mPTP) opening and mitochondrial membrane potential (MMP) collapse, while inhibition of PARP1 ameliorated the adverse outcomes. Importantly, the PARylation of apoptosis-inducing factor (AIF) is essential for its conformational change and translocation, which subsequently causes DNA break and cell death. Concretely, the interaction of PAR and truncated AIF (tAIF) is the mainstream in the parthanatos pathway. We also found that the effects of AIF cleavage and release were achieved through calpain activity and mPTP opening, both of which could be regulated by PARP1 via mediation of mitochondria Ca2+ concentration. In conclusion, the PAR-Ca2+-tAIF signaling pathway in parthanatos contributes to the oxidative stress damage observed in MCs. Targeting PAR-Ca2+-tAIF might be a potential therapeutic strategy for the early intervention of presbycusis and other oxidative stress-associated sensorineural deafness.
Collapse
Affiliation(s)
- Huanzhi Wan
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China; Research Institute of Otolaryngology-Head and Neck Surgery, Wuhan University, Wuhan, 430060, Hubei Province, China; The First Clinical School of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Huidong Chen
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China; Research Institute of Otolaryngology-Head and Neck Surgery, Wuhan University, Wuhan, 430060, Hubei Province, China; The First Clinical School of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Jingchun Liu
- The First Clinical School of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Bingqian Yang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China; Research Institute of Otolaryngology-Head and Neck Surgery, Wuhan University, Wuhan, 430060, Hubei Province, China; The First Clinical School of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Yunlong Zhang
- Research Institute of Otolaryngology-Head and Neck Surgery, Wuhan University, Wuhan, 430060, Hubei Province, China; The First Clinical School of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Yutong Bai
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China; Research Institute of Otolaryngology-Head and Neck Surgery, Wuhan University, Wuhan, 430060, Hubei Province, China; The First Clinical School of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Xiaoying Chen
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China; Research Institute of Otolaryngology-Head and Neck Surgery, Wuhan University, Wuhan, 430060, Hubei Province, China; The First Clinical School of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Jie Wang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China; Research Institute of Otolaryngology-Head and Neck Surgery, Wuhan University, Wuhan, 430060, Hubei Province, China; The First Clinical School of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Tianyi Liu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China; Research Institute of Otolaryngology-Head and Neck Surgery, Wuhan University, Wuhan, 430060, Hubei Province, China.
| | - Yuanyuan Zhang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China; Research Institute of Otolaryngology-Head and Neck Surgery, Wuhan University, Wuhan, 430060, Hubei Province, China.
| | - Qingquan Hua
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China; Research Institute of Otolaryngology-Head and Neck Surgery, Wuhan University, Wuhan, 430060, Hubei Province, China.
| |
Collapse
|
6
|
Bernardo-Colón A, Bighinati A, Parween S, Debnath S, Piano I, Adani E, Corsi F, Gargini C, Vergara N, Marigo V, Becerra SP. H105A peptide eye drops promote photoreceptor survival in murine and human models of retinal degeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.602890. [PMID: 39109177 PMCID: PMC11302621 DOI: 10.1101/2024.07.10.602890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Photoreceptor death causes blinding inheritable retinal diseases, such as retinitis pigmentosa (RP). As disease progression often outpaces therapeutic advances, finding effective treatments is urgent. This study focuses on developing a targeted approach by evaluating the efficacy of small peptides derived from pigment epithelium-derived factor (PEDF), known to restrict common cell death pathways associated with retinal diseases. Peptides with affinity for the PEDF receptor, PEDF-R, (17-mer and H105A) delivered via eye drops reached the retina, efficiently promoted photoreceptor survival, and improved retinal function in RP mouse models based on both the rd10 mutation and the rhodopsin P23H mutation. Additionally, intravitreal delivery of AAV-H105A vectors delayed photoreceptor degeneration in the latter RP mouse model. Furthermore, peptide H105A specifically prevented photoreceptor death induced by oxidative stress, a contributing factor to RP progression, in human retinal organoids. This promising approach for peptide eye drop delivery holds significant potential as a therapeutic for preventing photoreceptor death in retinal disorders, offering a high safety profile, low invasiveness and multiple delivery options.
Collapse
Affiliation(s)
- Alexandra Bernardo-Colón
- Section of Protein Structure and Function, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health; Bethesda, MD, USA
| | - Andrea Bighinati
- Department of Life Sciences, University of Modena and Reggio Emilia; 41125 Modena, Italy
| | - Shama Parween
- CellSight Ocular Stem Cell and Regeneration Program, Sue Anschutz-Rodgers Eye Center, University of Colorado Anschutz Medical Campus; Aurora, Colorado, USA
| | - Subrata Debnath
- Section of Protein Structure and Function, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health; Bethesda, MD, USA
| | - Ilaria Piano
- Department of Pharmacy, University of Pisa; 56126 Pisa, Italy
| | - Elisa Adani
- Department of Life Sciences, University of Modena and Reggio Emilia; 41125 Modena, Italy
| | - Francesca Corsi
- Department of Pharmacy, University of Pisa; 56126 Pisa, Italy
| | - Claudia Gargini
- Department of Pharmacy, University of Pisa; 56126 Pisa, Italy
| | - Natalia Vergara
- CellSight Ocular Stem Cell and Regeneration Program, Sue Anschutz-Rodgers Eye Center, University of Colorado Anschutz Medical Campus; Aurora, Colorado, USA
- Gates Center for Regenerative Medicine, Linda Crnic Institute for Down Syndrome and University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus; Aurora, Colorado, USA
| | - Valeria Marigo
- Department of Life Sciences, University of Modena and Reggio Emilia; 41125 Modena, Italy
| | - S. Patricia Becerra
- Section of Protein Structure and Function, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health; Bethesda, MD, USA
| |
Collapse
|
7
|
Vasudevan S, Senapati S, Pendergast M, Park PSH. Aggregation of rhodopsin mutants in mouse models of autosomal dominant retinitis pigmentosa. Nat Commun 2024; 15:1451. [PMID: 38365903 PMCID: PMC10873427 DOI: 10.1038/s41467-024-45748-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 02/01/2024] [Indexed: 02/18/2024] Open
Abstract
Mutations in rhodopsin can cause it to misfold and lead to retinal degeneration. A distinguishing feature of these mutants in vitro is that they mislocalize and aggregate. It is unclear whether or not these features contribute to retinal degeneration observed in vivo. The effect of P23H and G188R misfolding mutations were examined in a heterologous expression system and knockin mouse models, including a mouse model generated here expressing the G188R rhodopsin mutant. In vitro characterizations demonstrate that both mutants aggregate, with the G188R mutant exhibiting a more severe aggregation profile compared to the P23H mutant. The potential for rhodopsin mutants to aggregate in vivo was assessed by PROTEOSTAT, a dye that labels aggregated proteins. Both mutants mislocalize in photoreceptor cells and PROTEOSTAT staining was detected surrounding the nuclei of photoreceptor cells. The G188R mutant promotes a more severe retinal degeneration phenotype and greater PROTEOSTAT staining compared to that promoted by the P23H mutant. Here, we show that the level of PROTEOSTAT positive cells mirrors the progression and level of photoreceptor cell death, which suggests a potential role for rhodopsin aggregation in retinal degeneration.
Collapse
Affiliation(s)
- Sreelakshmi Vasudevan
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Subhadip Senapati
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
- Prayoga Institute of Education Research, Bengaluru, KA, 560116, India
| | - Maryanne Pendergast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Paul S-H Park
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
8
|
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
|
9
|
Zhang SX, Wang JJ, Starr CR, Lee EJ, Park KS, Zhylkibayev A, Medina A, Lin JH, Gorbatyuk M. The endoplasmic reticulum: Homeostasis and crosstalk in retinal health and disease. Prog Retin Eye Res 2024; 98:101231. [PMID: 38092262 PMCID: PMC11056313 DOI: 10.1016/j.preteyeres.2023.101231] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/19/2023]
Abstract
The endoplasmic reticulum (ER) is the largest intracellular organelle carrying out a broad range of important cellular functions including protein biosynthesis, folding, and trafficking, lipid and sterol biosynthesis, carbohydrate metabolism, and calcium storage and gated release. In addition, the ER makes close contact with multiple intracellular organelles such as mitochondria and the plasma membrane to actively regulate the biogenesis, remodeling, and function of these organelles. Therefore, maintaining a homeostatic and functional ER is critical for the survival and function of cells. This vital process is implemented through well-orchestrated signaling pathways of the unfolded protein response (UPR). The UPR is activated when misfolded or unfolded proteins accumulate in the ER, a condition known as ER stress, and functions to restore ER homeostasis thus promoting cell survival. However, prolonged activation or dysregulation of the UPR can lead to cell death and other detrimental events such as inflammation and oxidative stress; these processes are implicated in the pathogenesis of many human diseases including retinal disorders. In this review manuscript, we discuss the unique features of the ER and ER stress signaling in the retina and retinal neurons and describe recent advances in the research to uncover the role of ER stress signaling in neurodegenerative retinal diseases including age-related macular degeneration, inherited retinal degeneration, achromatopsia and cone diseases, and diabetic retinopathy. In some chapters, we highlight the complex interactions between the ER and other intracellular organelles focusing on mitochondria and illustrate how ER stress signaling regulates common cellular stress pathways such as autophagy. We also touch upon the integrated stress response in retinal degeneration and diabetic retinopathy. Finally, we provide an update on the current development of pharmacological agents targeting the UPR response and discuss some unresolved questions and knowledge gaps to be addressed by future research.
Collapse
Affiliation(s)
- Sarah X Zhang
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States.
| | - Josh J Wang
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Christopher R Starr
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Eun-Jin Lee
- Department of Ophthalmology and Byers Eye Institute, Stanford University, Stanford, CA, United States; VA Palo Alto Healthcare System, Palo Alto, CA, United States; Department of Pathology, Stanford University, Stanford, CA, United States
| | - Karen Sophia Park
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Assylbek Zhylkibayev
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andy Medina
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Jonathan H Lin
- Department of Ophthalmology and Byers Eye Institute, Stanford University, Stanford, CA, United States; VA Palo Alto Healthcare System, Palo Alto, CA, United States; Department of Pathology, Stanford University, Stanford, CA, United States
| | - Marina Gorbatyuk
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
10
|
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
|
11
|
Zhen F, Zou T, Wang T, Zhou Y, Dong S, Zhang H. Rhodopsin-associated retinal dystrophy: Disease mechanisms and therapeutic strategies. Front Neurosci 2023; 17:1132179. [PMID: 37077319 PMCID: PMC10106759 DOI: 10.3389/fnins.2023.1132179] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 03/13/2023] [Indexed: 04/05/2023] Open
Abstract
Rhodopsin is a light-sensitive G protein-coupled receptor that initiates the phototransduction cascade in rod photoreceptors. Mutations in the rhodopsin-encoding gene RHO are the leading cause of autosomal dominant retinitis pigmentosa (ADRP). To date, more than 200 mutations have been identified in RHO. The high allelic heterogeneity of RHO mutations suggests complicated pathogenic mechanisms. Here, we discuss representative RHO mutations as examples to briefly summarize the mechanisms underlying rhodopsin-related retinal dystrophy, which include but are not limited to endoplasmic reticulum stress and calcium ion dysregulation resulting from protein misfolding, mistrafficking, and malfunction. Based on recent advances in our understanding of disease mechanisms, various treatment methods, including adaptation, whole-eye electrical stimulation, and small molecular compounds, have been developed. Additionally, innovative therapeutic treatment strategies, such as antisense oligonucleotide therapy, gene therapy, optogenetic therapy, and stem cell therapy, have achieved promising outcomes in preclinical disease models of rhodopsin mutations. Successful translation of these treatment strategies may effectively ameliorate, prevent or rescue vision loss related to rhodopsin mutations.
Collapse
Affiliation(s)
- Fangyuan Zhen
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Provincial Ophthalmic Hospital, Zhengzhou, China
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Tongdan Zou
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ting Wang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yongwei Zhou
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Provincial Ophthalmic Hospital, Zhengzhou, China
| | - Shuqian Dong
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Provincial Ophthalmic Hospital, Zhengzhou, China
- *Correspondence: Shuqian Dong, ; Houbin Zhang,
| | - Houbin Zhang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Research Unit for Blindness Prevention, Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
- *Correspondence: Shuqian Dong, ; Houbin Zhang,
| |
Collapse
|
12
|
GADD34 Ablation Exacerbates Retinal Degeneration in P23H RHO Mice. Int J Mol Sci 2022; 23:ijms232213748. [PMID: 36430227 PMCID: PMC9697375 DOI: 10.3390/ijms232213748] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
The UPR is sustainably activated in degenerating retinas, leading to translational inhibition via p-eIF2α. Recent findings have demonstrated that ablation of growth arrest and DNA damage-inducible protein 34 (GADD34), a protein phosphatase 1 regulatory subunit permitting translational machinery operation through p-eIF2α elevation, does not impact the rate of translation in fast-degenerating rd16 mice. The current study aimed to validate whether P23H RHO mice degenerating at a slower pace manifest translational attenuation and whether GADD34 ablation impacts the rate of retinal degeneration via further suppression of retinal protein synthesis and apoptotic cell death. For this study, mice were examined with ERG and histological analyses. The molecular assessment was conducted in the naïve and LPS-challenged mice using Western blot and qRT-PCR analyses. Thus, this study demonstrates that the P23H RHO retinas manifest translational attenuation. However, GADD34 ablation resulted in a more prominent p-eIF2a increase without impacting the translation rate. GADD34 deficiency also led to a reduction in scotopic ERG amplitudes and an increased number of TUNEL-positive cells. Molecular analysis revealed that GADD34 deficiency reduces the expression of p-STAT3 and Il-6 while increasing the expression of Tnfa. Overall, the data indicate that GADD34 plays a multifunctional role. Under chronic UPR activation, GADD34 acts as a feedback player, dephosphorylating p-eIF2a, although this role does not seem to be critical. Additionally, GADD34 controls cytokine expression and STAT3 activation. Perhaps these molecular events are particularly important in controlling the pace of retinal degeneration.
Collapse
|
13
|
Robichaux MA, Nguyen V, Chan F, Kailasam L, He F, Wilson JH, Wensel TG. Subcellular localization of mutant P23H rhodopsin in an RFP fusion knock-in mouse model of retinitis pigmentosa. Dis Model Mech 2022; 15:274688. [PMID: 35275162 PMCID: PMC9092655 DOI: 10.1242/dmm.049336] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 02/28/2022] [Indexed: 12/15/2022] Open
Abstract
The P23H mutation in rhodopsin (Rho), the rod visual pigment, is the most common allele associated with autosomal-dominant retinitis pigmentosa (adRP). The fate of misfolded mutant Rho in rod photoreceptors has yet to be elucidated. We generated a new mouse model, in which the P23H-Rho mutant allele is fused to the fluorescent protein Tag-RFP-T (P23HhRhoRFP). In heterozygotes, outer segments formed, and wild-type (WT) rhodopsin was properly localized, but mutant P23H-Rho protein was mislocalized in the inner segments. Heterozygotes exhibited slowly progressing retinal degeneration. Mislocalized P23HhRhoRFP was contained in greatly expanded endoplasmic reticulum (ER) membranes. Quantification of mRNA for markers of ER stress and the unfolded protein response revealed little or no increases. mRNA levels for both the mutant human rhodopsin allele and the WT mouse rhodopsin were reduced, but protein levels revealed selective degradation of the mutant protein. These results suggest that the mutant rods undergo an adaptative process that prolongs survival despite unfolded protein accumulation in the ER. The P23H-Rho-RFP mouse may represent a useful tool for the future study of the pathology and treatment of P23H-Rho and adRP. This article has an associated First Person interview with the first author of the paper. Summary: A mouse line with a knock-in of the human rhodopsin gene altered to contain the P23H mutation and a red fluorescent protein fusion provides a new model for autosomal-dominant retinitis pigmentosa.
Collapse
Affiliation(s)
- Michael A Robichaux
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA.,Departments of Ophthalmology and Biochemistry, West Virginia University, 108 Biomedical Road, Morgantown, WV 26506, USA
| | - Vy Nguyen
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Fung Chan
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Lavanya Kailasam
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Feng He
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - John H Wilson
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Theodore G Wensel
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
14
|
Effects of Epigenetic Modification of PGC-1α by a Chemical Chaperon on Mitochondria Biogenesis and Visual Function in Retinitis Pigmentosa. Cells 2022; 11:cells11091497. [PMID: 35563803 PMCID: PMC9099608 DOI: 10.3390/cells11091497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/13/2022] [Accepted: 04/27/2022] [Indexed: 11/17/2022] Open
Abstract
Retinitis pigmentosa (RP) is a hereditary blinding disease characterized by gradual photoreceptor death, which lacks a definitive treatment. Here, we demonstrated the effect of 4-phenylbutyric acid (PBA), a chemical chaperon that can suppress endoplasmic reticulum (ER) stress, in P23H mutant rhodopsin knock-in RP models. In the RP models, constant PBA treatment led to the retention of a greater number of photoreceptors, preserving the inner segment (IS), a mitochondrial- and ER-rich part of the photoreceptors. Electroretinography showed that PBA treatment preserved photoreceptor function. At the early point, ER-associated degradation markers, xbp1s, vcp, and derl1, mitochondrial kinetic-related markers, fis1, lc3, and mfn1 and mfn2, as well as key mitochondrial regulators, pgc-1α and tfam, were upregulated in the retina of the models treated with PBA. In vitro analyses showed that PBA upregulated pgc-1α and tfam transcription, leading to an increase in the mitochondrial membrane potential, cytochrome c oxidase activity, and ATP levels. Histone acetylation of the PGC-1α promoter was increased by PBA, indicating that PBA affected the mitochondrial condition through epigenetic changes. Our findings constituted proof of concept for the treatment of ER stress-related RP using PBA and revealed PBA’s neuroprotective effects, paving the way for its future clinical application.
Collapse
|
15
|
Belhadj S, Hermann NS, Zhu Y, Christensen G, Strasser T, Paquet-Durand F. Visualizing Cell Death in Live Retina: Using Calpain Activity Detection as a Biomarker for Retinal Degeneration. Int J Mol Sci 2022; 23:ijms23073892. [PMID: 35409251 PMCID: PMC8999672 DOI: 10.3390/ijms23073892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 01/27/2023] Open
Abstract
Calpains are a family of calcium-activated proteases involved in numerous disorders. Notably, previous studies have shown that calpain activity was substantially increased in various models for inherited retinal degeneration (RD). In the present study, we tested the capacity of the calpain-specific substrate t-BOC-Leu-Met-CMAC to detect calpain activity in living retina, in organotypic retinal explant cultures derived from wild-type mice, as well as from rd1 and RhoP23H/+ RD-mutant mice. Test conditions were refined until the calpain substrate readily detected large numbers of cells in the photoreceptor layer of RD retina but not in wild-type retina. At the same time, the calpain substrate was not obviously toxic to photoreceptor cells. Comparison of calpain activity with immunostaining for activated calpain-2 furthermore suggested that individual calpain isoforms may be active in distinct temporal stages of photoreceptor cell death. Notably, calpain-2 activity may be a relatively short-lived event, occurring only towards the end of the cell-death process. Finally, our results support the development of calpain activity detection as a novel in vivo biomarker for RD suitable for combination with non-invasive imaging techniques.
Collapse
Affiliation(s)
- Soumaya Belhadj
- Cell Death Mechanisms Group, Institute for Ophthalmic Research, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany; (S.B.); (Y.Z.); (G.C.)
- Graduate Training Center of Neuroscience, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany;
| | - Nina Sofia Hermann
- Graduate Training Center of Neuroscience, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany;
| | - Yu Zhu
- Cell Death Mechanisms Group, Institute for Ophthalmic Research, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany; (S.B.); (Y.Z.); (G.C.)
- Graduate Training Center of Neuroscience, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany;
| | - Gustav Christensen
- Cell Death Mechanisms Group, Institute for Ophthalmic Research, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany; (S.B.); (Y.Z.); (G.C.)
- Graduate Training Center of Neuroscience, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany;
| | - Torsten Strasser
- Applied Vision Research Group, Institute for Ophthalmic Research, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany;
- University Eye Hospital Tübingen, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany
| | - François Paquet-Durand
- Cell Death Mechanisms Group, Institute for Ophthalmic Research, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany; (S.B.); (Y.Z.); (G.C.)
- Correspondence:
| |
Collapse
|
16
|
McLaughlin T, Medina A, Perkins J, Yera M, Wang JJ, Zhang SX. Cellular stress signaling and the unfolded protein response in retinal degeneration: mechanisms and therapeutic implications. Mol Neurodegener 2022; 17:25. [PMID: 35346303 PMCID: PMC8962104 DOI: 10.1186/s13024-022-00528-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 03/04/2022] [Indexed: 12/22/2022] Open
Abstract
Background The retina, as part of the central nervous system (CNS) with limited capacity for self-reparation and regeneration in mammals, is under cumulative environmental stress due to high-energy demands and rapid protein turnover. These stressors disrupt the cellular protein and metabolic homeostasis, which, if not alleviated, can lead to dysfunction and cell death of retinal neurons. One primary cellular stress response is the highly conserved unfolded protein response (UPR). The UPR acts through three main signaling pathways in an attempt to restore the protein homeostasis in the endoplasmic reticulum (ER) by various means, including but not limited to, reducing protein translation, increasing protein-folding capacity, and promoting misfolded protein degradation. Moreover, recent work has identified a novel function of the UPR in regulation of cellular metabolism and mitochondrial function, disturbance of which contributes to neuronal degeneration and dysfunction. The role of the UPR in retinal neurons during aging and under disease conditions in age-related macular degeneration (AMD), retinitis pigmentosa (RP), glaucoma, and diabetic retinopathy (DR) has been explored over the past two decades. Each of the disease conditions and their corresponding animal models provide distinct challenges and unique opportunities to gain a better understanding of the role of the UPR in the maintenance of retinal health and function. Method We performed an extensive literature search on PubMed and Google Scholar using the following keywords: unfolded protein response, metabolism, ER stress, retinal degeneration, aging, age-related macular degeneration, retinitis pigmentosa, glaucoma, diabetic retinopathy. Results and conclusion We summarize recent advances in understanding cellular stress response, in particular the UPR, in retinal diseases, highlighting the potential roles of UPR pathways in regulation of cellular metabolism and mitochondrial function in retinal neurons. Further, we provide perspective on the promise and challenges for targeting the UPR pathways as a new therapeutic approach in age- and disease-related retinal degeneration.
Collapse
Affiliation(s)
- Todd McLaughlin
- Department of Ophthalmology and Ira G. Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 955 Main Street, Buffalo, NY, 14203, USA
| | - Andy Medina
- Department of Ophthalmology and Ira G. Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 955 Main Street, Buffalo, NY, 14203, USA
| | - Jacob Perkins
- Department of Ophthalmology and Ira G. Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 955 Main Street, Buffalo, NY, 14203, USA
| | - Maria Yera
- Department of Ophthalmology and Ira G. Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 955 Main Street, Buffalo, NY, 14203, USA.,Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Joshua J Wang
- Department of Ophthalmology and Ira G. Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 955 Main Street, Buffalo, NY, 14203, USA.,Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Sarah X Zhang
- Department of Ophthalmology and Ira G. Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 955 Main Street, Buffalo, NY, 14203, USA. .,Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA. .,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA.
| |
Collapse
|
17
|
Loss of αA or αB-Crystallin Accelerates Photoreceptor Cell Death in a Mouse Model of P23H Autosomal Dominant Retinitis Pigmentosa. Int J Mol Sci 2021; 23:ijms23010070. [PMID: 35008496 PMCID: PMC8744961 DOI: 10.3390/ijms23010070] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 02/08/2023] Open
Abstract
Inherited retinal degenerations (IRD) are a leading cause of visual impairment and can result from mutations in any one of a multitude of genes. Mutations in the light-sensing protein rhodopsin (RHO) is a leading cause of IRD with the most common of those being a missense mutation that results in substitution of proline-23 with histidine. This variant, also known as P23H-RHO, results in rhodopsin misfolding, initiation of endoplasmic reticulum stress, the unfolded protein response, and activation of cell death pathways. In this study, we investigate the effect of α-crystallins on photoreceptor survival in a mouse model of IRD secondary to P23H-RHO. We find that knockout of either αA- or αB-crystallin results in increased intraretinal inflammation, activation of apoptosis and necroptosis, and photoreceptor death. Our data suggest an important role for the ⍺-crystallins in regulating photoreceptor survival in the P23H-RHO mouse model of IRD.
Collapse
|
18
|
Ortega JT, Jastrzebska B. Neuroinflammation as a Therapeutic Target in Retinitis Pigmentosa and Quercetin as Its Potential Modulator. Pharmaceutics 2021; 13:pharmaceutics13111935. [PMID: 34834350 PMCID: PMC8623264 DOI: 10.3390/pharmaceutics13111935] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 12/25/2022] Open
Abstract
The retina is a multilayer neuronal tissue located in the back of the eye that transduces the environmental light into a neural impulse. Many eye diseases caused by endogenous or exogenous harm lead to retina degeneration with neuroinflammation being a major hallmark of these pathologies. One of the most prevalent retinopathies is retinitis pigmentosa (RP), a clinically and genetically heterogeneous hereditary disorder that causes a decline in vision and eventually blindness. Most RP cases are related to mutations in the rod visual receptor, rhodopsin. The mutant protein triggers inflammatory reactions resulting in the activation of microglia to clear degenerating photoreceptor cells. However, sustained insult caused by the abnormal genetic background exacerbates the inflammatory response and increases oxidative stress in the retina, leading to a decline in rod photoreceptors followed by cone photoreceptors. Thus, inhibition of inflammation in RP has received attention and has been explored as a potential therapeutic strategy. However, pharmacological modulation of the retinal inflammatory response in combination with rhodopsin small molecule chaperones would likely be a more advantageous therapeutic approach to combat RP. Flavonoids, which exhibit antioxidant and anti-inflammatory properties, and modulate the stability and folding of rod opsin, could be a valid option in developing treatment strategies against RP.
Collapse
|
19
|
Ryu J, Gulamhusein H, Oh JK, Chang JH, Chen J, Tsang SH. Nutrigenetic reprogramming of oxidative stress. Taiwan J Ophthalmol 2021; 11:207-215. [PMID: 34703735 PMCID: PMC8493979 DOI: 10.4103/tjo.tjo_4_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/17/2021] [Indexed: 12/30/2022] Open
Abstract
Retinal disorders such as retinitis pigmentosa, age-related retinal degeneration, oxygen-induced retinopathy, and ischemia-reperfusion injury cause debilitating and irreversible vision loss. While the exact mechanisms underlying these conditions remain unclear, there has been a growing body of evidence demonstrating the pathological contributions of oxidative stress across different cell types within the eye. Nuclear factor erythroid-2-related factor (Nrf2), a transcriptional activator of antioxidative genes, and its regulator Kelch-like ECH-associated protein 1 (Keap1) have emerged as promising therapeutic targets. The purpose of this review is to understand the protective role of the Nrf2-Keap1 pathway in different retinal tissues and shed light on the complex mechanisms underlying these processes. In the photoreceptors, we highlight that Nrf2 preserves their survival and function by maintaining oxidation homeostasis. In the retinal pigment epithelium, Nrf2 similarly plays a critical role in oxidative stabilization but also maintains mitochondrial motility and autophagy-related lipid metabolic processes. In endothelial cells, Nrf2 seems to promote proper vascularization and revascularization through concurrent activation of antioxidative and angiogenic factors as well as inhibition of inflammatory cytokines. Finally, Nrf2 protects retinal ganglion cells against apoptotic cell death. Importantly, we show that Nrf2-mediated protection of the various retinal tissues corresponds to a preservation of functional vision. Altogether, this review underscores the potential of the Nrf2-Keap1 pathway as a powerful tool against retinal degeneration. Key insights into this elegant oxidative defense mechanism may ultimately pave the path toward a universal therapy for various inherited and environmental retinal disorders.
Collapse
Affiliation(s)
- Joseph Ryu
- Department of Ophthalmology, Jonas Children's Vision Care and the Bernard and Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Columbia University, New York, NY, USA
| | - Huzeifa Gulamhusein
- Department of Ophthalmology, Jonas Children's Vision Care and the Bernard and Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Columbia University, New York, NY, USA.,Department of Ophthalmology, Institute of Human Nutrition, Columbia University, New York, NY, USA.,Department of Ophthalmology, The University of Texas Rio Grande Valley School of Medicine, Edinburg, TX, USA
| | - Jin Kyun Oh
- Department of Ophthalmology, Jonas Children's Vision Care and the Bernard and Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Columbia University, New York, NY, USA.,Department of Ophthalmology, State University of New York at Downstate Medical Center, Brooklyn, USA
| | - Joseph H Chang
- Department of Ophthalmology, Jonas Children's Vision Care and the Bernard and Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Columbia University, New York, NY, USA.,Department of Ophthalmology, Institute of Human Nutrition, Columbia University, New York, NY, USA
| | - Jocelyn Chen
- Department of Ophthalmology, Jonas Children's Vision Care and the Bernard and Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Columbia University, New York, NY, USA.,Department of Ophthalmology, Columbia University, New York, USA
| | - Stephen H Tsang
- Department of Ophthalmology, Jonas Children's Vision Care and the Bernard and Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Columbia University, New York, NY, USA.,Department of Ophthalmology, Institute of Human Nutrition, Columbia University, New York, NY, USA.,Department of Pathology and Cell Biology, College of Physicians and Surgeons, Institute of Human Nutrition, Columbia University, NY, USA
| |
Collapse
|
20
|
New In Vitro Cellular Model for Molecular Studies of Retinitis Pigmentosa. Int J Mol Sci 2021; 22:ijms22126440. [PMID: 34208617 PMCID: PMC8235468 DOI: 10.3390/ijms22126440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/04/2021] [Accepted: 06/13/2021] [Indexed: 02/05/2023] Open
Abstract
Retinitis pigmentosa (RP) is an inherited form of retinal degeneration characterized by primary rod photoreceptor cell death followed by cone loss. Mutations in several genes linked to the disease cause increased levels of cyclic guanosine monophosphate (cGMP) and calcium ion influxes. The purpose of this project was to develop a new in vitro photoreceptor degeneration model for molecular studies of RP. 661W cells were genetically modified to stably express the neural retina leucine zipper (NRL) transcription factor. One clone (661W-A11) was selected based on the expression of Nrl target genes. 661W-A11 showed a significant increase in expression of rod-specific genes but not of cone-specific genes, compared with 661W cells. Zaprinast was used to inhibit phosphodiesterase 6 (PDE6) activity to mimic photoreceptor degeneration in vitro. The activation of cell death pathways resulting from PDE6 inhibition was confirmed by detection of decreased viability and increased intracellular cGMP and calcium, as well as activation of protein kinase G (PKG) and calpains. In this new in vitro system, we validated the effects of previously published neuroprotective drugs. The 661W-A11 cells may serve as a new model for molecular studies of RP and for high-throughput drug screening.
Collapse
|
21
|
Li J, Du W, Xu N, Tao T, Tang X, Huang L. RNA-seq analysis for exploring the pathogenesis of Retinitis pigmentosa in P23H knock-in mice. Ophthalmic Res 2021; 64:798-810. [PMID: 33971646 DOI: 10.1159/000515727] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 03/05/2021] [Indexed: 11/19/2022]
Affiliation(s)
- Jiarui Li
- Eye diseases and Optometry Institute, Department of Ophthalmology, Peking University People's Hospital, Beijing, China,
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China,
- College of Optometry, Peking University Health Science Center, Beijing, China,
| | - Wei Du
- Eye diseases and Optometry Institute, Department of Ophthalmology, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
| | - Ningda Xu
- Eye diseases and Optometry Institute, Department of Ophthalmology, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
| | - Tianchang Tao
- Eye diseases and Optometry Institute, Department of Ophthalmology, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
| | - Xin Tang
- Eye diseases and Optometry Institute, Department of Ophthalmology, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
| | - Lvzhen Huang
- Eye diseases and Optometry Institute, Department of Ophthalmology, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
| |
Collapse
|
22
|
2,3,5,6-Tetramethylpyrazine protects retinal photoreceptors against endoplasmic reticulum stress by modulating ATF4-mediated inhibition of PRP aggregation. J Mol Med (Berl) 2021; 99:383-402. [PMID: 33409554 DOI: 10.1007/s00109-020-02017-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 11/23/2020] [Accepted: 11/25/2020] [Indexed: 01/17/2023]
Abstract
Endoplasmic reticulum (ER) stress is a common threat to photoreceptors during the pathogenesis of chronic retinopathies and often results in irreversible visual impairment. 2,3,5,6-Tetramethylpyrazine (TMP), which possesses many beneficial pharmacological activities, is a potential drug that could be used to protect photoreceptors. In the present study, we found that the cellular growth rate of 661 W cells cultured under low glucose conditions was lower than that of control cells, while the G2/M phase of the cell cycle was longer. We further found that the mitochondrial membrane potential (ΔΨm) was lower and that ER stress factor expression was increased in 661 W cells cultured under low glucose conditions. TMP reversed these trends. Visual function and cell counts in the outer nuclear layer (ONL) were low and the TUNEL-positive rate in the ONL was high in a C3H mouse model of spontaneous retinal degeneration. Similarly, visual function was decreased, and the TUNEL-positive rate in the ONL was increased in fasted C57/BL6j mice compared with control mice. On the other hand, ER stress factor expression was found to be increased in the retinas of both mouse models, as shown by reverse transcription real-time PCR (RT-qPCR) and western blotting. TMP reversed the physiological and molecular biological variations observed in both mouse models, and ATF4 expression was enhanced again. Further investigation by using western blotting illustrated that the proportion of insoluble prion protein (PRP) versus soluble PRP was reduced both in vitro and in vivo. Taken together, these results suggest that TMP increased the functions of photoreceptors by alleviating ER stress in vitro and in vivo, and the intrinsic mechanism was the ATF4-mediated inhibition of PRP aggregation. TMP may potentially be used clinically as a therapeutic agent to attenuate the functional loss of photoreceptors during the pathogenesis of chronic retinopathies. KEY MESSAGES: • Already known: TMP is a beneficial drug mainly used in clinic to enhance organ functions, and the intrinsic mechanism is still worthy of exploring. • New in the study: We discovered that TMP ameliorated retinal photoreceptors function via ER stress alleviation, which was promoted by ATF4-mediated inhibition of PRP aggregation. • Application prospect: In prospective clinical practices, TMP may potentially be used in the clinic as a therapeutic agent to attenuate the photoreceptors functional reduction in chronic retinopathies.
Collapse
|
23
|
Newton F, Megaw R. Mechanisms of Photoreceptor Death in Retinitis Pigmentosa. Genes (Basel) 2020; 11:genes11101120. [PMID: 32987769 PMCID: PMC7598671 DOI: 10.3390/genes11101120] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/15/2020] [Accepted: 09/22/2020] [Indexed: 02/08/2023] Open
Abstract
Retinitis pigmentosa (RP) is the most common cause of inherited blindness and is characterised by the progressive loss of retinal photoreceptors. However, RP is a highly heterogeneous disease and, while much progress has been made in developing gene replacement and gene editing treatments for RP, it is also necessary to develop treatments that are applicable to all causative mutations. Further understanding of the mechanisms leading to photoreceptor death is essential for the development of these treatments. Recent work has therefore focused on the role of apoptotic and non-apoptotic cell death pathways in RP and the various mechanisms that trigger these pathways in degenerating photoreceptors. In particular, several recent studies have begun to elucidate the role of microglia and innate immune response in the progression of RP. Here, we discuss some of the recent progress in understanding mechanisms of rod and cone photoreceptor death in RP and summarise recent clinical trials targeting these pathways.
Collapse
Affiliation(s)
- Fay Newton
- MRC Human Genetics Unit, University of Edinburgh, South Bridge, Edinburgh EH8 9YL, UK;
- Correspondence:
| | - Roly Megaw
- MRC Human Genetics Unit, University of Edinburgh, South Bridge, Edinburgh EH8 9YL, UK;
- Princess Alexandra Eye Pavilion, NHS Lothian, Edinburgh EH3 9HA, UK
| |
Collapse
|
24
|
Collin GB, Gogna N, Chang B, Damkham N, Pinkney J, Hyde LF, Stone L, Naggert JK, Nishina PM, Krebs MP. Mouse Models of Inherited Retinal Degeneration with Photoreceptor Cell Loss. Cells 2020; 9:E931. [PMID: 32290105 PMCID: PMC7227028 DOI: 10.3390/cells9040931] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/05/2020] [Accepted: 04/07/2020] [Indexed: 12/12/2022] Open
Abstract
Inherited retinal degeneration (RD) leads to the impairment or loss of vision in millions of individuals worldwide, most frequently due to the loss of photoreceptor (PR) cells. Animal models, particularly the laboratory mouse, have been used to understand the pathogenic mechanisms that underlie PR cell loss and to explore therapies that may prevent, delay, or reverse RD. Here, we reviewed entries in the Mouse Genome Informatics and PubMed databases to compile a comprehensive list of monogenic mouse models in which PR cell loss is demonstrated. The progression of PR cell loss with postnatal age was documented in mutant alleles of genes grouped by biological function. As anticipated, a wide range in the onset and rate of cell loss was observed among the reported models. The analysis underscored relationships between RD genes and ciliary function, transcription-coupled DNA damage repair, and cellular chloride homeostasis. Comparing the mouse gene list to human RD genes identified in the RetNet database revealed that mouse models are available for 40% of the known human diseases, suggesting opportunities for future research. This work may provide insight into the molecular players and pathways through which PR degenerative disease occurs and may be useful for planning translational studies.
Collapse
Affiliation(s)
- Gayle B. Collin
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Navdeep Gogna
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Bo Chang
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Nattaya Damkham
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Jai Pinkney
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Lillian F. Hyde
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Lisa Stone
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Jürgen K. Naggert
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Patsy M. Nishina
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Mark P. Krebs
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| |
Collapse
|
25
|
Gorbatyuk MS, Starr CR, Gorbatyuk OS. Endoplasmic reticulum stress: New insights into the pathogenesis and treatment of retinal degenerative diseases. Prog Retin Eye Res 2020; 79:100860. [PMID: 32272207 DOI: 10.1016/j.preteyeres.2020.100860] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 03/08/2020] [Accepted: 03/17/2020] [Indexed: 12/13/2022]
Abstract
Physiological equilibrium in the retina depends on coordinated work between rod and cone photoreceptors and can be compromised by the expression of mutant proteins leading to inherited retinal degeneration (IRD). IRD is a diverse group of retinal dystrophies with multifaceted molecular mechanisms that are not fully understood. In this review, we focus on the contribution of chronically activated unfolded protein response (UPR) to inherited retinal pathogenesis, placing special emphasis on studies employing genetically modified animal models. As constitutively active UPR in degenerating retinas may activate pro-apoptotic programs associated with oxidative stress, pro-inflammatory signaling, dysfunctional autophagy, free cytosolic Ca2+ overload, and altered protein synthesis rate in the retina, we focus on the regulatory mechanisms of translational attenuation and approaches to overcoming translational attenuation in degenerating retinas. We also discuss current research on the role of the UPR mediator PERK and its downstream targets in degenerating retinas and highlight the therapeutic benefits of reprogramming PERK signaling in preclinical animal models of IRD. Finally, we describe pharmacological approaches targeting UPR in ocular diseases and consider their potential applications to IRD.
Collapse
Affiliation(s)
- Marina S Gorbatyuk
- The University of Alabama at Birmingham, Department of Optometry and Vision Science, School of Optometry, USA.
| | - Christopher R Starr
- The University of Alabama at Birmingham, Department of Optometry and Vision Science, School of Optometry, USA
| | - Oleg S Gorbatyuk
- The University of Alabama at Birmingham, Department of Optometry and Vision Science, School of Optometry, USA
| |
Collapse
|
26
|
Kutluer M, Huang L, Marigo V. Targeting molecular pathways for the treatment of inherited retinal degeneration. Neural Regen Res 2020; 15:1784-1791. [PMID: 32246618 PMCID: PMC7513962 DOI: 10.4103/1673-5374.280303] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Inherited retinal degeneration is a major cause of incurable blindness characterized by loss of retinal photoreceptor cells. Inherited retinal degeneration is characterized by high genetic and phenotypic heterogeneity with several genes mutated in patients affected by these genetic diseases. The high genetic heterogeneity of these diseases hampers the development of effective therapeutic interventions for the cure of a large cohort of patients. Common cell demise mechanisms can be envisioned as targets to treat patients regardless the specific mutation. One of these targets is the increase of intracellular calcium ions, that has been detected in several murine models of inherited retinal degeneration. Recently, neurotrophic factors that favor the efflux of calcium ions to concentrations below toxic levels have been identified as promising molecules that should be evaluated as new treatments for retinal degeneration. Here, we discuss therapeutic options for inherited retinal degeneration and we will focus on neuroprotective approaches, such as the neuroprotective activity of the Pigment epithelium-derived factor. The characterization of specific targets for neuroprotection opens new perspectives together with many questions that require deep analyses to take advantage of this knowledge and develop new therapeutic approaches. We believe that minimizing cell demise by neuroprotection may represent a promising treatment strategy for retinal degeneration.
Collapse
Affiliation(s)
- Meltem Kutluer
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Li Huang
- 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
| |
Collapse
|