1
|
Zaluski J, Bassetto M, Kiser PD, Tochtrop GP. Advances and therapeutic opportunities in visual cycle modulation. Prog Retin Eye Res 2025; 106:101360. [PMID: 40280538 DOI: 10.1016/j.preteyeres.2025.101360] [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: 02/14/2025] [Revised: 04/19/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
The visual cycle is a metabolic pathway that enables continuous vision by regenerating the 11-cis-retinal chromophore for photoreceptors opsins. Although integral to normal visual function, the flux of retinoids through this cycle can contribute to a range of retinal pathologies, including Stargardt disease, age-related macular degeneration, and diabetic retinopathy. In such conditions, intermediates and byproducts of the visual cycle, such as bisretinoid components of lipofuscin, can accumulate, concomitant with cellular damage and eventual photoreceptor loss. This has inspired efforts to modulate the visual cycle, aiming to slow or prevent the formation of these toxic intermediates and thus preserve retinal structure and function. Over the past two decades, multiple strategies to modulate the visual cycle have emerged. These include both intrinsic approaches, targeting key enzymes, retinoid-binding proteins, or receptors within the pigment epithelium or photoreceptors (e.g., RPE65, CRBP1, and rhodopsin inhibitors/antagonists) and extrinsic strategies that indirectly alter retinoid availability within the retina (e.g., RBP4 antagonists). Many of these agents have shown promise in animal models of visual cycle-associated retinal diseases, reducing pathological changes, and improving retinal survival. Several have advanced into clinical studies, although none are currently FDA-approved. Challenges remain in optimizing drug specificity and duration of action while minimizing side effects such as nyctalopia. In this review, we comprehensively examine current and emerging visual cycle modulators, discuss their medicinal chemistry, mechanisms of action, efficacy in preclinical and clinical studies, and highlight future opportunities for drug discovery aimed at safely and effectively preserving vision through modulation of this biochemical pathway.
Collapse
Affiliation(s)
- Jordan Zaluski
- Department of Chemistry, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Marco Bassetto
- Department of Physiology and Biophysics, School of Medicine, University of California- Irvine, Irvine, CA, 92697, USA; Department of Ophthalmology, Gavin Herbert Eye Institute, Center for Translational Vision Research, School of Medicine, University of California- Irvine, Irvine, CA, 92697, USA; Research Service, VA Long Beach Healthcare System, Long Beach, CA, 90822, USA
| | - Philip D Kiser
- Department of Physiology and Biophysics, School of Medicine, University of California- Irvine, Irvine, CA, 92697, USA; Department of Ophthalmology, Gavin Herbert Eye Institute, Center for Translational Vision Research, School of Medicine, University of California- Irvine, Irvine, CA, 92697, USA; Research Service, VA Long Beach Healthcare System, Long Beach, CA, 90822, USA; Department of Clinical Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University of California - Irvine, Irvine, CA, 92697, USA.
| | - Gregory P Tochtrop
- Department of Chemistry, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
2
|
Arrigo A, Cremona O, Aragona E, Casoni F, Consalez G, Dogru RM, Hauck SM, Antropoli A, Bianco L, Parodi MB, Bandello F, Grosche A. Müller cells trophism and pathology as the next therapeutic targets for retinal diseases. Prog Retin Eye Res 2025; 106:101357. [PMID: 40254246 DOI: 10.1016/j.preteyeres.2025.101357] [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: 02/20/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/22/2025]
Abstract
Müller cells are a crucial retinal cell type involved in multiple regulatory processes and functions that are essential for retinal health and functionality. Acting as structural and functional support for retinal neurons and photoreceptors, Müller cells produce growth factors, regulate ion and fluid homeostasis, and facilitate neuronal signaling. They play a pivotal role in retinal morphogenesis and cell differentiation, significantly contributing to macular development. Due to their radial morphology and unique cytoskeletal organization, Müller cells act as optical fibers, efficiently channeling photons directly to the photoreceptors. In response to retinal damage, Müller cells undergo specific gene expression and functional changes that serve as a first line of defense for neurons, but can also lead to unwarranted cell dysfunction, contributing to cell death and neurodegeneration. In some species, Müller cells can reactivate their developmental program, promoting retinal regeneration and plasticity-a remarkable ability that holds promising therapeutic potential if harnessed in mammals. The crucial and multifaceted roles of Müller cells-that we propose to collectively call "Müller cells trophism"-highlight the necessity of maintaining their functionality. Dysfunction of Müller cells, termed "Müller cells pathology," has been associated with a plethora of retinal diseases, including age-related macular degeneration, diabetic retinopathy, vitreomacular disorders, macular telangiectasia, and inherited retinal dystrophies. In this review, we outline how even subtle disruptions in Müller cells trophism can drive the pathological cascade of Müller cells pathology, emphasizing the need for targeted therapies to preserve retinal health and prevent disease progression.
Collapse
Affiliation(s)
- Alessandro Arrigo
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; Eye Repair Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Ottavio Cremona
- Vita-Salute San Raffaele University, Milan, Italy; Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Emanuela Aragona
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Filippo Casoni
- Vita-Salute San Raffaele University, Milan, Italy; Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giacomo Consalez
- Vita-Salute San Raffaele University, Milan, Italy; Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rüya Merve Dogru
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, 80939, Germany
| | - Alessio Antropoli
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Bianco
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Francesco Bandello
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Antje Grosche
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany.
| |
Collapse
|
3
|
Yang B, Yang K, Chen Y, Li Q, Chen J, Li S, Wu Y. Exposure of A2E to blue light promotes ferroptosis in the retinal pigment epithelium. Cell Mol Biol Lett 2025; 30:22. [PMID: 39984833 PMCID: PMC11846388 DOI: 10.1186/s11658-025-00700-2] [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: 09/12/2024] [Accepted: 02/06/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Age-dependent accumulation of lipofuscin in the retinal pigment epithelium (RPE) is closely related to the etiology of autosomal recessive Stargardt's disease (STGD1) and dry age-related macular degeneration (AMD). N-retinylidene-N-retinylethanolamine (A2E) is a leading component of RPE lipofuscin that is highly susceptible to blue light. Ferroptosis is an iron-dependent form of non-apoptotic cell death characterized by the accumulation of lipid peroxides to a lethal level, which plays an important role in retinal diseases. However, it remains unknown whether A2E functions as a physiological trigger for eliciting blue light-induced ferroptosis of RPE cells. METHODS A2E-loaded RPE cells and Abca4-/-Rdh8-/- mice were exposed to blue light, respectively. Western blotting, immunofluorescence staining, reactive oxygen species (ROS) staining, intracellular iron staining, lipid peroxidation staining, fundus imaging, optical coherence tomography (OCT), hematoxylin-eosin (HE) staining, and electroretinography (ERG) were utilized to elucidate the role of blue light in A2E induced ferroptosis in the RPE and its potential mechanisms. RESULTS Exposure of A2E to blue light promoted ferroptotic cell death in RPE cells by elevating ferrous ion (Fe2+) levels and inhibiting the solute carrier family 7 membrane 11 (SLC7A11)-glutathione (GSH)-glutathione peroxidase 4 (GPX4) axis. GPX4 inactivation and ROS generated by Fe2+ overload and GSH depletion precipitated lipid peroxidation and subsequent ferroptosis in A2E-containing RPE cells upon exposure to blue light. In addition to GSH supplement, repressing either Fe2+ by deferiprone (DFP) or lipid peroxidation with ferrostatin-1 (Fer-1) significantly protected RPE cells against ferroptosis caused by blue light illumination of A2E. Abca4-/-Rdh8-/- mice featured by an accelerated deposition of A2E in the RPE is an animal model for STGD1 and dry AMD. It was observed that ferroptosis was indeed present in the RPE of Abca4-/-Rdh8-/- mice following exposure to blue light. Notably, alleviating ferroptosis by intraperitoneally injected Fer-1 effectively rescued retinal function and ameliorated RPE/photoreceptor degeneration in blue light-exposed Abca4-/-Rdh8-/- mice. CONCLUSIONS Our results suggest the importance of blue light in A2E-mediated ferroptosis in the RPE, and deeply broaden the understanding of mechanisms underlying RPE atrophy arising from lipofuscin accumulation in STGD1 and dry AMD.
Collapse
Affiliation(s)
- Bo Yang
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Kunhuan Yang
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Yuling Chen
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Qingjian Li
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Jingmeng Chen
- School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
- Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, Guangdong, China
| | - Shiying Li
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China.
| | - Yalin Wu
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China.
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China.
- Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, Guangdong, China.
| |
Collapse
|
4
|
Bassil FL, Colijn JM, Thiadens AAHJ, Biarnés M. Progression Rate of Macular Retinal Pigment Epithelium Atrophy in Geographic Atrophy and Selected Inherited Retinal Dystrophies. A Systematic Review and Meta-Analysis. Am J Ophthalmol 2025; 269:30-48. [PMID: 39153684 DOI: 10.1016/j.ajo.2024.07.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/11/2024] [Accepted: 07/29/2024] [Indexed: 08/19/2024]
Abstract
PURPOSE To compare the macular retinal pigment epithelium (RPE) atrophy progression rate of selected degenerative and macular inherited retinal diseases (IRD). DESIGN Systematic review and meta-analysis. METHODS The protocol was registered on the PROSPERO database. Medline, Embase, Web of Science, Cochrane Central Register of Controlled Trials, and Google Scholar were searched up to September 15, 2023 for articles reporting the RPE atrophy growth rate in treatment-naïve eyes with geographic atrophy (GA), Stargardt disease (STGD1), Best disease, pseudoxanthoma elasticum (PXE), central areolar choroidal dystrophy (CACD), or pattern dystrophies with no previous or current macular neovascularization and a minimum follow-up time of 12 months. Meta-analyses determined mean RPE atrophy growth rates per disease, imaging modality (fundus autofluorescence [FAF], optical coherence tomography [OCT], or color fundus photography [CFP]) and metric (mm2/y or mm/y). The Newcastle-Ottawa scale and the Cochrane Risk-of-Bias tool assessed the risk of bias, and funnel plots were used to evaluate small-study effects. RESULTS From 4354 publications, 85 were included for meta-analysis: 69 studies (7815 eyes) on GA, 15 (1367 eyes) on STGD1, and one on both. Two studies on PXE were only eligible for review. No studies for other diseases met our eligibility criteria. The overall mean RPE atrophy growth rate for GA using FAF was 1.65 mm2/y (95% confidence interval [CI], 1.49-1.81) and 0.35 mm/y (95% CI, 0.28-0.41); using OCT, it was 1.46 mm2/y (95% CI, 1.28-1.64) and 0.34 mm/y (95% CI, 0.28-0.40); and on CFP it was 1.76 mm2/y (95% CI, 1.56-1.97) and 0.30 mm/y (95% CI, 0.28-0.31). For STGD1, using FAF it was 1.0 mm2/y (95% CI, 0.77-1.23) and 0.20 mm/y (95% CI, 0.17-0.23); on OCT, it was 0.80 mm2/y (95% CI, 0.72-0.88). No studies on STGD1 reported the growth rate with other imaging modalities or metrics. Growth rates in GA were faster than in STGD1 (p < .05). A larger baseline area of atrophy was generally associated with faster growth rates. CONCLUSIONS The RPE atrophy growth rate in GA is faster than in STGD1 but with great variation between studies and imaging modalities. Limited information was available for other macular IRD, suggesting further research is needed.
Collapse
Affiliation(s)
- Fabiana L Bassil
- Department of Ophthalmology, Erasmus Medical Center (F.L.B., J.M.C., A.A.H.J.T.), Rotterdam, the Netherlands
| | - Johanna M Colijn
- Department of Ophthalmology, Erasmus Medical Center (F.L.B., J.M.C., A.A.H.J.T.), Rotterdam, the Netherlands
| | - Alberta A H J Thiadens
- Department of Ophthalmology, Erasmus Medical Center (F.L.B., J.M.C., A.A.H.J.T.), Rotterdam, the Netherlands
| | - Marc Biarnés
- Oftalmologia Mèdica i Quirúrgica (OMIQ) Research (M.B.), Barcelona, Spain.
| |
Collapse
|
5
|
Thanos DF, Saiti A, Giannopoulos-Dimitriou A, Kontouli-Pertesi N, Gorgoulis VG, Anagnostopoulos AK. Global Proteomic Profiling to Unravel Lipofuscin's Protein Repertoire. Methods Mol Biol 2025; 2906:215-227. [PMID: 40082358 DOI: 10.1007/978-1-0716-4426-3_12] [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: 03/16/2025]
Abstract
Lipofuscin, a yellow-brown complex mainly found in cellular waste, accumulates in cells as a byproduct of cellular degradation processes and is commonly associated with aging and oxidative stress. Lipofuscin's molecular "signature" comprises a diverse repertoire of oxidized proteins, lipids, and metals, yet the detailed protein composition of this unique complex remains undetermined. Toward this end, high-throughput proteomic analyses of lipofuscin are pivotal for elucidating its global protein content, the protein interactome, as well as biological mechanisms related to senescence-associated lipofuscin accumulation. Existing proteomic profiling approaches of lipofuscin primarily rely on "in-gel" digestion methods, often leading to incomplete protein digestion, suboptimal peptide recovery, and other analytical pitfalls leading to "loss" of significant molecular information. In this chapter, we introduce a global proteomic profiling methodology for lipofuscin preparations derived from cell cultures, based on FASP (Filter-Aided Sample Preparation) method coupled with nano-liquid chromatography-tandem mass spectrometry (nano-LC/MS-MS). This methodology aims to deliver robust characterization of the lipofuscin proteome, elucidate its complex interactions, and introduce a superior alternative to traditional "in-gel" digestion technique.
Collapse
Affiliation(s)
- Dimitris-Foivos Thanos
- Department of Histology and Embryology, Faculty of Medicine, National Kapodistrian University of Athens, Athens, Greece
| | - Aikaterini Saiti
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Alexandros Giannopoulos-Dimitriou
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nefeli Kontouli-Pertesi
- Department of Biotechnology, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Vassilis G Gorgoulis
- Department of Histology and Embryology, Faculty of Medicine, National Kapodistrian University of Athens, Athens, Greece
- Department of Biotechnology, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
- Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, UK
| | - Athanasios K Anagnostopoulos
- Department of Biotechnology, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
- Oncology Unit, Third Department of Internal Medicine, "Sotiria" Hospital, Faculty of Medicine, National Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
6
|
Rahman B, Anderson DMG, Chen C, Liu J, Migas LG, Van de Plas R, Schey KL, Kono M, Fan J, Koutalos Y. Sphingolipid Levels and Processing of the Retinyl Chromophore in the Retina of a Mouse Model of Niemann-Pick Disease. Invest Ophthalmol Vis Sci 2024; 65:24. [PMID: 39661357 PMCID: PMC11640910 DOI: 10.1167/iovs.65.14.24] [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: 07/18/2024] [Accepted: 11/21/2024] [Indexed: 12/12/2024] Open
Abstract
Purpose Mutations in the gene that encodes the enzyme acid sphingomyelinase (ASMase) are associated with Niemann-Pick disease, a lysosomal storage disorder. Mice that lack ASMase (ASMase-/-) exhibit age-related retinal degeneration and large increases in accumulation of lipofuscin in the retinal pigment epithelium (RPE). We examined which lipid species accumulate in the retina and the RPE of ASMase-/- mice and whether the retinal degeneration is associated with impaired photoreceptor metabolism and retinyl chromophore processing. Methods NADPH availability and all-trans retinol formation after rhodopsin bleaching were measured in isolated single rod photoreceptors with fluorescence imaging; sphingolipid levels in retinas and RPEs were measured with LC/MS; relative abundances of different lipid species in different retinal layers were measured with MALDI imaging mass spectrometry. Results There was no detectable difference in the kinetics of all-trans retinol formation or the NADPH-generating capacity between ASMase-/- and wild-type mice. Sphingomyelin levels were much higher in the retinas and RPEs of ASMase-/- animals compared to wild type, but there were no significant differences for ceramides. There was a large increase in the abundance of bis(monoacylglycero)phosphates (BMPs) in ASMase-/- mice, indicative of lysosomal dysfunction, but no substantial changes were detected for the bis-retinoid A2E. Conclusions Lysosomal dysfunction and retinal degeneration in ASMase-/- mice are not associated with defects in rod photoreceptor metabolism that affect all-trans retinol formation and availability of NADPH. Lysosomal dysfunction in ASMase-/- mice is not associated with bis-retinoid A2E accumulation.
Collapse
Affiliation(s)
- Bushra Rahman
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - David M. G. Anderson
- Mass Spectrometry Research Center and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, Unites States
| | - Chunhe Chen
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Jian Liu
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Lukasz G. Migas
- Delft Center for Systems and Control (DCSC), Delft University of Technology, Delft, Netherlands
| | - Raf Van de Plas
- Delft Center for Systems and Control (DCSC), Delft University of Technology, Delft, Netherlands
| | - Kevin L. Schey
- Mass Spectrometry Research Center and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, Unites States
| | - Masahiro Kono
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Jie Fan
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Yiannis Koutalos
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| |
Collapse
|
7
|
Hsiao YT, Huang HM, Chen TC, Lo J, Chen YJ, Kuo HK, Lee JJ. Parafoveal Microperimetric Retinal Sensitivity as a Key Parameter Associated with Vision Loss in Retinitis Pigmentosa. Diagnostics (Basel) 2024; 14:2691. [PMID: 39682599 DOI: 10.3390/diagnostics14232691] [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: 09/15/2024] [Revised: 10/13/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Although optical coherence tomography (OCT) is useful in determining outer retinal architecture, it may be suboptimal when monitoring subtle changes in retinitis pigmentosa (RP) patients. The aim of this study is to investigate precise microperimetric parameters for disease severity identification in RP patients. Methods: A cross-sectional and retrospective study. Thirty-nine eyes of 39 RP patients were included. Associations between logMAR visual acuity (VA), spectral-domain OCT, fundus autofluorescence imaging (FAF), and various microperimetric measures were evaluated. Microperimetric test locations were grouped into "foveal", parafoveal "inner ring", and perifoveal "outer ring". Independent variables were analyzed based on logistic regression, then assessed using area under the receiver operating characteristic curve (AUROC). Results: Among all microperimetric measures, linear regression analysis indicated that mean retinal sensitivity and deep scotoma count at the parafoveal inner ring were the principal parameters associated with decreased VA. The AUROC was highest for deep scotoma count at the inner ring at a value of 0.829, with the cut-off point at 3.5. A visual function index was then established according to the number of parafoveal deep scotoma points, in order of mild (0 points), moderate (1-3 points), and severe (4 or more points). Our microperimetric visual function index also correlated significantly to logMAR VA and previously established FAF patterns. Conclusions: Our study discovered deep scotoma count at the parafoveal inner ring to be a key microperimetric parameter in evaluating vision loss in RP patients. Those with four or more deep scotoma points at the parafoveal inner ring are more likely to have functional low vision.
Collapse
Affiliation(s)
- Yu-Ting Hsiao
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83340, Taiwan
| | - Hsiu-Mei Huang
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83340, Taiwan
| | - Ta-Ching Chen
- Department of Ophthalmology, National Taiwan University Hospital, Taipei 10041, Taiwan
- Center of Frontier Medicine, National Taiwan University Hospital, Taipei 10041, Taiwan
| | - Jung Lo
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83340, Taiwan
| | - Yung-Jen Chen
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83340, Taiwan
| | - Hsi-Kung Kuo
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83340, Taiwan
| | - Jong-Jer Lee
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83340, Taiwan
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83340, Taiwan
| |
Collapse
|
8
|
Abdouh M, Chen Y, Goyeneche A, Burnier MN. Blue Light-Induced Mitochondrial Oxidative Damage Underlay Retinal Pigment Epithelial Cell Apoptosis. Int J Mol Sci 2024; 25:12619. [PMID: 39684332 DOI: 10.3390/ijms252312619] [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: 10/20/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Reactive oxygen species (ROS) play a pivotal role in apoptosis. We reported that Blue Light (BL) induced oxidative stress in human retinal pigment epithelial (RPE) cells in vitro and increased drusen deposition and RPE cell apoptosis in human eyes. Here, we investigated the mechanisms underlying BL-induced damage to RPE cells. Cells were exposed to BL with or without the antioxidant N-acetylcysteine. Cells were analyzed for levels of ROS, proliferation, viability, and mitochondria membrane potential (ΔΨM) fluctuation. We performed proteomic analyses to search for differentially expressed proteins. ROS levels increased following RPE cell exposure to BL. While ROS production did not affect RPE cell proliferation, it was accompanied by decreased ΔΨM and increased cell apoptosis due to the caspase cascade activation in a ROS-dependent manner. Proteomic analyses revealed that BL decreased the levels of ROS detoxifying enzymes in exposed cells. We conclude that BL-induced oxidative stress is cytotoxic to RPE cells. These findings bring new insights into the involvement of BL on RPE cell damage and its role in the progression of age-related macular degeneration. The use of antioxidants is an avenue to block or delay BL-mediated RPE cell apoptosis to counteract the disease progression.
Collapse
Affiliation(s)
- Mohamed Abdouh
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- The MUHC-McGill University Ocular Pathology & Translational Research Laboratory, McGill University, Montreal, QC H4A 3J1, Canada
| | - Yunxi Chen
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Alicia Goyeneche
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- The MUHC-McGill University Ocular Pathology & Translational Research Laboratory, McGill University, Montreal, QC H4A 3J1, Canada
| | - Miguel N Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- The MUHC-McGill University Ocular Pathology & Translational Research Laboratory, McGill University, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
9
|
Choi JA, Seo BR, Koh JY, Yoon YH. Protective effect of zinc against A2E-induced toxicity in ARPE-19 cells: Possible involvement of lysosomal acidification. Heliyon 2024; 10:e39100. [PMID: 39524844 PMCID: PMC11550603 DOI: 10.1016/j.heliyon.2024.e39100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/07/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
A key pathogenic mechanism of dry age-related macular degeneration (AMD) is lysosomal dysfunction in retinal pigment epithelium (RPE) cells, which results in the accumulation of lipofuscins such as A2E (N-retinylidene-N-retinylethanolamine) that further compromises lysosomal function. This vicious cycle leads to cell death and poor visual acuity. Here, we established an in vitro model of AMD by treating a human RPE cell line (ARPE-19) with A2E and examined whether raising zinc levels confers protective effects against lysosomal dysfunction and cytotoxicity. MTT assay showed that A2E induced apoptosis in ARPE-19 cells. pHrodo™ Red fluorescence staining showed that lysosomal pH increased in A2E-treated ARPE-19 cells. Treatment with a zinc ionophore (clioquinol) reduced A2E accumulation, restored lysosomal pH to the acidic range, and reduced A2E-induced cell death, all of which were reversed by the addition of a zinc chelator (TPEN). Consistent with the in vitro results, subretinal injections of A2E in mouse eyes resulted in the death of RPE cells as well as lysosomal dysfunction, all of which were reversed by co-treatment with clioquinol. Our results suggest that restoring the levels of intracellular zinc, especially in lysosomes, would be helpful in mitigating A2E-induced cytotoxic changes including lysosomal dysfunction in RPE cells in the pathogenesis of AMD.
Collapse
Affiliation(s)
- Jeong A. Choi
- Neural Injury Research Center, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Bo-Ra Seo
- Neural Injury Research Center, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jae-Young Koh
- Neural Injury Research Center, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Young Hee Yoon
- Department of Ophthalmology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
10
|
Guo Y, Chen S, Guan W, Xu N, Zhu L, Du W, Liu Z, Fong HKW, Huang L, Zhao M. Retinal G-protein-coupled receptor deletion exacerbates AMD-like changes via the PINK1-parkin pathway under oxidative stress. FASEB J 2024; 38:e70135. [PMID: 39467145 PMCID: PMC11580724 DOI: 10.1096/fj.202401160rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 09/18/2024] [Accepted: 10/15/2024] [Indexed: 10/30/2024]
Abstract
The intake of high dietary fat has been correlated with the progression of age-related macular degeneration (AMD), affecting the function of the retinal pigment epithelium through oxidative stress. A high-fat diet (HFD) can lead to lipid metabolism disorders, excessive production of circulating free fatty acids, and systemic inflammation by aggravating the degree of oxidative stress. Deletion of the retinal G-protein-coupled receptor (RGR-d) has been identified in drusen. In this study, we investigated how the RGR-d exacerbates AMD-like changes under oxidative stress, both in vivo and in vitro. Fundus atrophy became evident, at 12 months old, particularly in the RGR-d + HFD group, and fluorescence angiography revealed narrower retinal vessels and a reduced perfusion area in the peripheral retina. Although rod electroretinography revealed decreasing trends in the a- and b-wave amplitudes in the RGR-d + HFD group at 12 months, the changes were not statistically significant. Mice in the RGR-d + HFD group showed a significantly thinner and more fragile retinal morphology than those in the WT + HFD group, with disordered and discontinuous pigment distribution in the RGR-d + HFD mice. Transmission electron microscopy revealed a thickened Bruch's membrane along the choriocapillaris endothelial cell wall in the RGR-d + HFD mice, and the outer nuclear layer structure appeared disorganized, with reduced nuclear density. Kyoto Encyclopedia of Genes and Genomes pathway analysis indicated significantly lower levels of 25(OH)-vitamin D3 metabolites in the RGR-d + HFD group. Under oxidative stress, RGR-d localized to the mitochondria and reduced the levels of the PINK1-parkin pathway. RGR-d mice fed an HFD were used as a new animal model of dry AMD. Under high-fat-induced oxidative stress, RGR-d accumulated in the mitochondria, disrupting normal mitophagy and causing cellular damage, thus exacerbating AMD-like changes both in vivo and in vitro.
Collapse
Affiliation(s)
- Yue Guo
- Department of OphthalmologyPeking University People's HospitalBeijingChina
- Eye Diseases and Optometry InstituteBeijingChina
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid DiseasesBeijingChina
- College of OptometryPeking University Health Science CenterBeijingChina
| | - Sitong Chen
- Department of OphthalmologyPeking University People's HospitalBeijingChina
- Eye Diseases and Optometry InstituteBeijingChina
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid DiseasesBeijingChina
- College of OptometryPeking University Health Science CenterBeijingChina
| | - Wenxue Guan
- Department of OphthalmologyPeking University People's HospitalBeijingChina
- Eye Diseases and Optometry InstituteBeijingChina
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid DiseasesBeijingChina
- College of OptometryPeking University Health Science CenterBeijingChina
| | - Ningda Xu
- Department of OphthalmologyPeking University People's HospitalBeijingChina
- Eye Diseases and Optometry InstituteBeijingChina
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid DiseasesBeijingChina
- College of OptometryPeking University Health Science CenterBeijingChina
| | - Li Zhu
- Department of OphthalmologyPeking University People's HospitalBeijingChina
- Eye Diseases and Optometry InstituteBeijingChina
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid DiseasesBeijingChina
- College of OptometryPeking University Health Science CenterBeijingChina
| | - Wei Du
- Department of OphthalmologyPeking University People's HospitalBeijingChina
- Eye Diseases and Optometry InstituteBeijingChina
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid DiseasesBeijingChina
- College of OptometryPeking University Health Science CenterBeijingChina
| | - Zhiming Liu
- Department of OphthalmologyPeking University People's HospitalBeijingChina
- Eye Diseases and Optometry InstituteBeijingChina
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid DiseasesBeijingChina
- College of OptometryPeking University Health Science CenterBeijingChina
| | - Henry K. W. Fong
- Department of Ophthalmology, USC Roski Eye InstituteKeck School of Medicine of University of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of Molecular Microbiology and ImmunologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Lvzhen Huang
- Department of OphthalmologyPeking University People's HospitalBeijingChina
- Eye Diseases and Optometry InstituteBeijingChina
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid DiseasesBeijingChina
- College of OptometryPeking University Health Science CenterBeijingChina
| | - Mingwei Zhao
- Department of OphthalmologyPeking University People's HospitalBeijingChina
- Eye Diseases and Optometry InstituteBeijingChina
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid DiseasesBeijingChina
- College of OptometryPeking University Health Science CenterBeijingChina
| |
Collapse
|
11
|
Campbell JM, Gosnell M, Agha A, Handley S, Knab A, Anwer AG, Bhargava A, Goldys EM. Label-Free Assessment of Key Biological Autofluorophores: Material Characteristics and Opportunities for Clinical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403761. [PMID: 38775184 DOI: 10.1002/adma.202403761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/04/2024] [Indexed: 06/13/2024]
Abstract
Autofluorophores are endogenous fluorescent compounds that naturally occur in the intra and extracellular spaces of all tissues and organs. Most have vital biological functions - like the metabolic cofactors NAD(P)H and FAD+, as well as the structural protein collagen. Others are considered to be waste products - like lipofuscin and advanced glycation end products - which accumulate with age and are associated with cellular dysfunction. Due to their natural fluorescence, these materials have great utility for enabling non-invasive, label-free assays with direct ties to biological function. Numerous technologies, with different advantages and drawbacks, are applied to their assessment, including fluorescence lifetime imaging microscopy, hyperspectral microscopy, and flow cytometry. Here, the applications of label-free autofluorophore assessment are reviewed for clinical and health-research applications, with specific attention to biomaterials, disease detection, surgical guidance, treatment monitoring, and tissue assessment - fields that greatly benefit from non-invasive methodologies capable of continuous, in vivo characterization.
Collapse
Affiliation(s)
- Jared M Campbell
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2033, Australia
| | | | - Adnan Agha
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2033, Australia
| | - Shannon Handley
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2033, Australia
| | - Aline Knab
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2033, Australia
| | - Ayad G Anwer
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2033, Australia
| | - Akanksha Bhargava
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2033, Australia
| | - Ewa M Goldys
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2033, Australia
| |
Collapse
|
12
|
Finzi A, Ottoboni S, Cellini M, Corcioni B, Gaudiano C, Fontana L. Color Doppler Imaging, Endothelin-1, Corneal Biomechanics and Scleral Rigidity in Asymmetric Age-Related Macular Degeneration. Clin Ophthalmol 2024; 18:2583-2591. [PMID: 39281979 PMCID: PMC11401527 DOI: 10.2147/opth.s479225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/02/2024] [Indexed: 09/18/2024] Open
Abstract
Purpose Age-related macular degeneration (AMD) presents a multifaceted etiopathogenesis involving ischemic, inflammatory, and genetic components. This study investigates the correlation between ocular hemodynamics, scleral rigidity (SR), and plasma endothelin-1 (ET1) levels in treatment-naive patients with asymmetrical AMD. Patients and Methods This study included 20 treatment-naive patients (12 females and 8 males) with an average age of 76.4 ± 3.7 years, who presented with AMD with neovascular membrane formation (nAMD) in one eye, and intermediate grade 2 AMD (iAMD) in the other eye. The control group consisted of 20 healthy subjects (13 females and 7 males) with a mean age of 74.7 ± 3.9 years. All patients and healthy controls underwent color Doppler imaging (i) of the ophthalmic artery (OA), short posterior ciliary arteries (SPCAs), and central retinal artery (CRA); Plasma ET-1 levels were measured for all patients and healthy subjects. Corneal biomechanics were assessed using an Ocular Response Analyzer and two indices were obtained: corneal hysteresis (CH) and corneal resistance factor (CRF). Results Results showed reduced blood flow velocities and increased resistance indices in AMD eyes, particularly affecting the short posterior ciliary arteries. According to mechanical theory, ARMD eyes exhibited elevated scleral rigidity and corneal resistance factor compared to controls, with a notable rise in SR in neovascular AMD (nAMD) eyes. As per the chronic subacute inflammation theory, plasma ET-1 levels were significantly higher in AMD patients, correlating with abnormal SPCAs blood flow and increased resistance indices. Conclusion Findings suggest a multifactorial etiology of AMD involving an increase of ET-1 plasma levels with biomechanic damages of corneal and scleral tissue in nAMD.
Collapse
Affiliation(s)
- Alessandro Finzi
- Ophthalmology Unit, Department of Experimental, Diagnostic and Specialty Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Simone Ottoboni
- Ophthalmology Unit, Department of Experimental, Diagnostic and Specialty Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Mauro Cellini
- Ophthalmology Unit, Department of Experimental, Diagnostic and Specialty Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Beniamino Corcioni
- Department of Radiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Caterina Gaudiano
- Department of Radiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Luigi Fontana
- Ophthalmology Unit, Department of Experimental, Diagnostic and Specialty Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
13
|
Basu B, Karwatka M, China B, McKibbin M, Khan K, Inglehearn CF, Ladbury JE, Johnson CA. Glycogen myophosphorylase loss causes increased dependence on glucose in iPSC-derived retinal pigment epithelium. J Biol Chem 2024; 300:107569. [PMID: 39009342 PMCID: PMC11342771 DOI: 10.1016/j.jbc.2024.107569] [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: 01/20/2024] [Revised: 06/07/2024] [Accepted: 06/27/2024] [Indexed: 07/17/2024] Open
Abstract
Loss of glycogen myophosphorylase (PYGM) expression results in an inability to break down muscle glycogen, leading to McArdle disease-an autosomal recessive metabolic disorder characterized by exercise intolerance and muscle cramps. While previously considered relatively benign, this condition has recently been associated with pattern dystrophy in the retina, accompanied by variable sight impairment, secondary to retinal pigment epithelial (RPE) cell involvement. However, the pathomechanism of this condition remains unclear. In this study, we generated a PYGM-null induced pluripotent stem cell line and differentiated it into mature RPE to examine structural and functional defects, along with metabolite release into apical and basal media. Mutant RPE exhibited normal photoreceptor outer segment phagocytosis but displayed elevated glycogen levels, reduced transepithelial resistance, and increased cytokine secretion across the epithelial layer compared to isogenic WT controls. Additionally, decreased expression of the visual cycle component, RDH11, encoding 11-cis-retinol dehydrogenase, was observed in PYGM-null RPE. While glycolytic flux and oxidative phosphorylation levels in PYGM-null RPE were near normal, the basal oxygen consumption rate was increased. Oxygen consumption rate in response to physiological levels of lactate was significantly greater in WT than PYGM-null RPE. Inefficient lactate utilization by mutant RPE resulted in higher glucose dependence and increased glucose uptake from the apical medium in the presence of lactate, suggesting a reduced capacity to spare glucose for photoreceptor use. Metabolic tracing confirmed slower 13C-lactate utilization by PYGM-null RPE. These findings have key implications for retinal health since they likely underlie the vision impairment in individuals with McArdle disease.
Collapse
Affiliation(s)
- Basudha Basu
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Magdalena Karwatka
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Becky China
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Martin McKibbin
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK; Department of Ophthalmology, St James's University Hospital, Leeds, UK
| | - Kamron Khan
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Chris F Inglehearn
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - John E Ladbury
- School of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - Colin A Johnson
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK.
| |
Collapse
|
14
|
Kwon YS, Munsoor J, Kaufmann M, Zheng M, Smirnov AI, Han Z. Polydopamine Nanoparticles as Mimicking RPE Melanin for the Protection of Retinal Cells Against Blue Light-Induced Phototoxicity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400230. [PMID: 38816934 PMCID: PMC11304300 DOI: 10.1002/advs.202400230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/15/2024] [Indexed: 06/01/2024]
Abstract
Exposure of the eyes to blue light can induce the overproduction of reactive oxygen species (ROS) in the retina and retinal pigment epithelium (RPE) cells, potentially leading to pathological damage of age-related macular degeneration (AMD). While the melanin in RPE cells absorbs blue light and prevents ROS accumulation, the loss and dysfunction of RPE melanin due to age-related changes may contribute to photooxidation toxicity. Herein, a novel approach utilizing a polydopamine-replenishing strategy via a single-dose intravitreal (IVT) injection is presented to protect retinal cells against blue light-induced phototoxicity. To investigate the effects of overexposure to blue light on retinal cells, a blue light exposure Nrf2-deficient mouse model is created, which is susceptible to light-induced retinal lesions. After blue light irradiation, retina degeneration and an overproduction of ROS are observed. The polydopamine-replenishing strategy demonstrated effectiveness in maintaining retinal structural integrity and preventing retina degeneration by reducing ROS production in retinal cells and limiting the phototoxicity of blue light exposure. These findings highlight the potential of polydopamine as a simple and effective replenishment for providing photoprotection against high-energy blue light exposure.
Collapse
Affiliation(s)
- Yong-Su Kwon
- Department of Ophthalmology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Julie Munsoor
- Department of Ophthalmology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Mary Kaufmann
- Department of Ophthalmology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Min Zheng
- Department of Ophthalmology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Alex I Smirnov
- Department of Chemistry, North Carolina State University, Raleigh, NC, 27695, USA
| | - Zongchao Han
- Department of Ophthalmology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
15
|
Gregory-Evans K, Kolawole OU, Molday RS, Gregory-Evans CY. Novel Variants in ABCA4-Related Retinopathies with Structural Re-Assessment of Variants of Uncertain Significance. Ophthalmologica 2024; 247:231-240. [PMID: 39043154 DOI: 10.1159/000540361] [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: 02/07/2024] [Accepted: 07/10/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION Conclusive molecular genetic diagnoses in inherited retinal diseases remains a major challenge due to the large number of variants of uncertain significance (VUS) identified in genetic testing. Here, we determined the genotypic and phenotypic spectrum of ABCA4 gene variants in a cohort of Canadian inherited retinal dystrophy subjects. METHODS This retrospective study evaluated 64 subjects with an inherited retinal dystrophy diagnosis with variants in the ABCA4 gene. Pathogenicity of variants was assessed by comparison to genetic databases and in silico modelling. ABCA4 variants classified as VUS were further evaluated using a cryo-electron structural model of the ABCA4 protein to predict impact on protein function and were also assessed for evolutionary conservation. RESULTS Conclusive disease-causing biallelic ABCA4 variants were detected in 52 subjects with either Stargardt's disease, cone-rod dystrophy, macular dystrophy, or pattern dystrophy. A further 14 variants were novel comprising 1 nonsense, 1 frameshift, 3 splicing, and 9 missense variants. Based on in silico modelling, protein modelling and evolutionary conservation from human to zebrafish, we re-classified 5 of these as pathogenic and a further 3 as likely pathogenic. We also added to the ABCA4 phenotypic spectrum seen with four known pathogenic variants (c.2161-2A>G; Leu296Cysfs*4; Arg1640Gln; and Pro1380Leu). CONCLUSIONS This study expands the genotypic and phenotypic spectrum of ABCA4 disease-associated variants. By panel-based genetic testing, we identified 14 novel ABCA4 variants of which 8 were determined to be disease-causing or likely disease-causing. These methodologies could circumvent somewhat the need for labour intensive in vitro and in vivo assessments of novel ABCA4 variants.
Collapse
Affiliation(s)
- Kevin Gregory-Evans
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Olubayo U Kolawole
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Robert S Molday
- Department Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Cheryl Y Gregory-Evans
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
16
|
Ong J, Zarnegar A, Selvam A, Driban M, Chhablani J. The Complement System as a Therapeutic Target in Retinal Disease. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:945. [PMID: 38929562 PMCID: PMC11205777 DOI: 10.3390/medicina60060945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 05/16/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024]
Abstract
The complement cascade is a vital system in the human body's defense against pathogens. During the natural aging process, it has been observed that this system is imperative for ensuring the integrity and homeostasis of the retina. While this system is critical for proper host defense and retinal integrity, it has also been found that dysregulation of this system may lead to certain retinal pathologies, including geographic atrophy and diabetic retinopathy. Targeting components of the complement system for retinal diseases has been an area of interest, and in vivo, ex vivo, and clinical trials have been conducted in this area. Following clinical trials, medications targeting the complement system for retinal disease have also become available. In this manuscript, we discuss the pathophysiology of complement dysfunction in the retina and specific pathologies. We then describe the results of cellular, animal, and clinical studies targeting the complement system for retinal diseases. We then provide an overview of complement inhibitors that have been approved by the Food and Drug Administration (FDA) for geographic atrophy. The complement system in retinal diseases continues to serve as an emerging therapeutic target, and further research in this field will provide additional insights into the mechanisms and considerations for treatment of retinal pathologies.
Collapse
Affiliation(s)
- Joshua Ong
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, MI 48105, USA
| | - Arman Zarnegar
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Amrish Selvam
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Matthew Driban
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jay Chhablani
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
17
|
Dontsov A, Ostrovsky M. Retinal Pigment Epithelium Pigment Granules: Norms, Age Relations and Pathology. Int J Mol Sci 2024; 25:3609. [PMID: 38612421 PMCID: PMC11011557 DOI: 10.3390/ijms25073609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
The retinal pigment epithelium (RPE), which ensures the normal functioning of the neural retina, is a pigmented single-cell layer that separates the retina from the Bruch's membrane and the choroid. There are three main types of pigment granules in the RPE cells of the human eye: lipofuscin granules (LG) containing the fluorescent "age pigment" lipofuscin, melanoprotein granules (melanosomes, melanolysosomes) containing the screening pigment melanin and complex melanolipofuscin granules (MLG) containing both types of pigments simultaneously-melanin and lipofuscin. This review examines the functional role of pigment granules in the aging process and in the development of oxidative stress and associated pathologies in RPE cells. The focus is on the process of light-induced oxidative degradation of pigment granules caused by reactive oxygen species. The reasons leading to increased oxidative stress in RPE cells as a result of the oxidative degradation of pigment granules are considered. A mechanism is proposed to explain the phenomenon of age-related decline in melanin content in RPE cells. The essence of the mechanism is that when the lipofuscin part of the melanolipofuscin granule is exposed to light, reactive oxygen species are formed, which destroy the melanin part. As more melanolipofuscin granules are formed with age and the development of degenerative diseases, the melanin in pigmented epithelial cells ultimately disappears.
Collapse
Affiliation(s)
| | - Mikhail Ostrovsky
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow 119334, Russia;
| |
Collapse
|
18
|
Fujinami K, Waheed N, Laich Y, Yang P, Fujinami-Yokokawa Y, Higgins JJ, Lu JT, Curtiss D, Clary C, Michaelides M. Stargardt macular dystrophy and therapeutic approaches. Br J Ophthalmol 2024; 108:495-505. [PMID: 37940365 PMCID: PMC10958310 DOI: 10.1136/bjo-2022-323071] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 10/06/2023] [Indexed: 11/10/2023]
Abstract
Stargardt macular dystrophy (Stargardt disease; STGD1; OMIM 248200) is the most prevalent inherited macular dystrophy. STGD1 is an autosomal recessive disorder caused by multiple pathogenic sequence variants in the large ABCA4 gene (OMIM 601691). Major advances in understanding both the clinical and molecular features, as well as the underlying pathophysiology, have culminated in many completed, ongoing and planned human clinical trials of novel therapies.The aims of this concise review are to describe (1) the detailed phenotypic and genotypic characteristics of the disease, multimodal imaging findings, natural history of the disease, and pathogenesis, (2) the multiple avenues of research and therapeutic intervention, including pharmacological, cellular therapies and diverse types of genetic therapies that have either been investigated or are under investigation and (3) the exciting novel therapeutic approaches on the translational horizon that aim to treat STGD1 by replacing the entire 6.8 kb ABCA4 open reading frame.
Collapse
Affiliation(s)
- Kaoru Fujinami
- Laboratory of Visual Physiology, Division of Vision Research, National Institute of Sensory Organs, NHO Tokyo Medical Center, Meguro-ku, Tokyo, Japan
- Institute of Ophthalmology, University College London, London, UK
- Moorfields Eye Hospital NHS Foundation Trust, London, UK
| | - Nadia Waheed
- Department of Ophthalmology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Yannik Laich
- Moorfields Eye Hospital NHS Foundation Trust, London, UK
- Eye Center, Medical Center, University of Freiburg Faculty of Medicine, Freiburg, Germany
| | - Paul Yang
- Oregon Health and Science University Casey Eye Institute, Portland, Oregon, USA
| | - Yu Fujinami-Yokokawa
- Laboratory of Visual Physiology, Division of Vision Research, National Institute of Sensory Organs, NHO Tokyo Medical Center, Meguro-ku, Tokyo, Japan
- Institute of Ophthalmology, University College London, London, UK
- Department of Health Policy and Management, Keio University School of Medicine Graduate School of Medicine, Shinjuku-ku, Tokyo, Japan
| | | | - Jonathan T Lu
- SalioGen Therapeutics Inc, Lexington, Massachusetts, USA
| | - Darin Curtiss
- Applied Genetic Technologies Corporation, Alachua, Florida, USA
| | - Cathryn Clary
- SalioGen Therapeutics Inc, Lexington, Massachusetts, USA
| | - Michel Michaelides
- Institute of Ophthalmology, University College London, London, UK
- Moorfields Eye Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
19
|
Rzhanova LA, Markitantova YV, Aleksandrova MA. Recent Achievements in the Heterogeneity of Mammalian and Human Retinal Pigment Epithelium: In Search of a Stem Cell. Cells 2024; 13:281. [PMID: 38334673 PMCID: PMC10854871 DOI: 10.3390/cells13030281] [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: 11/30/2023] [Revised: 01/23/2024] [Accepted: 02/01/2024] [Indexed: 02/10/2024] Open
Abstract
Retinal pigment epithelium (RPE) cells are important fundamentally for the development and function of the retina. In this regard, the study of the morphological and molecular properties of RPE cells, as well as their regenerative capabilities, is of particular importance for biomedicine. However, these studies are complicated by the fact that, despite the external morphological similarity of RPE cells, the RPE is a population of heterogeneous cells, the molecular genetic properties of which have begun to be revealed by sequencing methods only in recent years. This review carries out an analysis of the data from morphological and molecular genetic studies of the heterogeneity of RPE cells in mammals and humans, which reveals the individual differences in the subpopulations of RPE cells and the possible specificity of their functions. Particular attention is paid to discussing the properties of "stemness," proliferation, and plasticity in the RPE, which may be useful for uncovering the mechanisms of retinal diseases associated with pathologies of the RPE and finding new ways of treating them.
Collapse
Affiliation(s)
| | - Yuliya V. Markitantova
- Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia; (L.A.R.); (M.A.A.)
| | | |
Collapse
|
20
|
Françon A, Behar-Cohen F, Torriglia A. The blue light hazard and its use on the evaluation of photochemical risk for domestic lighting. An in vivo study. ENVIRONMENT INTERNATIONAL 2024; 184:108471. [PMID: 38335626 DOI: 10.1016/j.envint.2024.108471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 01/16/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Nowadays artificial light highly increases human exposure to light leading to circadian rhythm and sleep perturbations. Moreover, excessive exposure of ocular structures to photons can induce irreversible retinal damage. Meta-analyses showed that sunlight exposure influences the age of onset and the progression of Age-related macular degeneration (AMD), the leading cause of blindness in people over fifty-year old. Currently, the blue-light hazard (BLH) curve is used in the evaluation of the phototoxicity of a light source for domestic lighting regulations. OBJECTIVES Here, we analyze the phototoxicity threshold in rats and investigate the role played by the light spectrum, assessing the relevance of the use of the BLH-weighting to define phototoxicity. METHODS We exposed albino rats to increasing doses of blue and white light, or to lights of different colors to evaluate the impact of each component of the white light spectrum on phototoxicity. Cellular mechanisms of cell death and cellular stress induced by light were analyzed. RESULTS Our results show that the phototoxicity threshold currently accepted for rats is overestimated by a factor of 50 when considering blue light and by a factor of 550 concerning white light. This is the result of the toxicity induced by green light that increases white light toxicity by promoting an inflammatory response. The content of green in white light induces 8 fold more invasion of macrophages in the retina than the content of blue light. Moreover, the use of BLH-weighting does not evaluate the amount of red radiations contained in white light that mitigates damage by inhibiting the nuclear translocation of L-DNase II and reducing by 33% the number of TUNEL-positive cells. DISCUSSION These findings question the current methods to determine the phototoxicity of a light source and show the necessity to take into account the entire emission spectrum. As current human phototoxicity thresholds were estimated with the same methods used for rats, our results suggest that they might need to be reconsidered.
Collapse
Affiliation(s)
- Anaïs Françon
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Université Paris Cité, Sorbonne Université. Team: Physiopathology of Ocular Diseases: Therapeutic Innovations. 15, rue de l'école de Médecine, 75006 Paris, France
| | - Francine Behar-Cohen
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Université Paris Cité, Sorbonne Université. Team: Physiopathology of Ocular Diseases: Therapeutic Innovations. 15, rue de l'école de Médecine, 75006 Paris, France; Assistance Publique, Hôpitaux de Paris, Hôpital Cochin, Ophtalmopole, 27, rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - Alicia Torriglia
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Université Paris Cité, Sorbonne Université. Team: Physiopathology of Ocular Diseases: Therapeutic Innovations. 15, rue de l'école de Médecine, 75006 Paris, France.
| |
Collapse
|
21
|
Romano F, Cozzi E, Boon CJF, Staurenghi G, Salvetti AP. MULTIMODAL RETINAL IMAGING REVEALS NEW PATHOGENIC INSIGHTS IN CENTRAL AREOLAR CHOROIDAL DYSTROPHY: A CASE SERIES. Retin Cases Brief Rep 2024; 18:32-38. [PMID: 36731070 DOI: 10.1097/icb.0000000000001325] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
PURPOSE To describe novel imaging findings in a family affected by central areolar choroidal dystrophy. METHODS Case series with multimodal retinal imaging assessment. RESULTS A 19-year-old asymptomatic woman was referred for bilateral macular defects of the retinal pigment epithelium. Blue-light autofluorescence of her right eye revealed a speckled pattern in the macular area with a ring of decreased autofluorescence using near-infrared autofluorescence. Multimodal assessment of her left eye disclosed a single parafoveal spot of decreased pigmentation that was clearly visible as hyperautofluorescent using blue-light autofluorescence and as hypoautofluorescent using near-infrared autofluorescence. Optical coherence tomography angiography revealed several tiny areas of flow voids in correspondence of the retinal pigment epithelium alterations of both eyes. Three family members were recently diagnosed with presumed age-related macular degeneration and demonstrated well-demarcated areas of retinal pigment epithelium atrophy surrounded by yellowish deposits and a hypopigmented halo. Next-generation genetic analysis for inherited macular dystrophies was performed on the index case and the affected family members and revealed a p.Arg172Gln missense mutation in PRPH2 gene, leading to the diagnosis of central areolar choroidal dystrophy. CONCLUSION Multimodal imaging can reveal new pathogenic insights in central areolar choroidal dystrophy. Of notice, near-infrared autofluorescence and optical coherence tomography angiography are able to detect retinal pigment epithelium hypopigmentation and choriocapillaris rarefaction, respectively, since the earliest stages of the disease.
Collapse
Affiliation(s)
- Francesco Romano
- Eye Clinic, Department of Biomedical and Clinical Sciences, Luigi Sacco Hospital, University of Milan, Milan, Italy
| | - Elisa Cozzi
- Eye Clinic, Department of Biomedical and Clinical Sciences, Luigi Sacco Hospital, University of Milan, Milan, Italy
| | - Camiel J F Boon
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands; and
- Department of Ophthalmology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Giovanni Staurenghi
- Eye Clinic, Department of Biomedical and Clinical Sciences, Luigi Sacco Hospital, University of Milan, Milan, Italy
| | - Anna Paola Salvetti
- Eye Clinic, Department of Biomedical and Clinical Sciences, Luigi Sacco Hospital, University of Milan, Milan, Italy
| |
Collapse
|
22
|
Różanowska MB. Lipofuscin, Its Origin, Properties, and Contribution to Retinal Fluorescence as a Potential Biomarker of Oxidative Damage to the Retina. Antioxidants (Basel) 2023; 12:2111. [PMID: 38136230 PMCID: PMC10740933 DOI: 10.3390/antiox12122111] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/05/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Lipofuscin accumulates with age as intracellular fluorescent granules originating from incomplete lysosomal digestion of phagocytosed and autophagocytosed material. The purpose of this review is to provide an update on the current understanding of the role of oxidative stress and/or lysosomal dysfunction in lipofuscin accumulation and its consequences, particularly for retinal pigment epithelium (RPE). Next, the fluorescence of lipofuscin, spectral changes induced by oxidation, and its contribution to retinal fluorescence are discussed. This is followed by reviewing recent developments in fluorescence imaging of the retina and the current evidence on the prognostic value of retinal fluorescence for the progression of age-related macular degeneration (AMD), the major blinding disease affecting elderly people in developed countries. The evidence of lipofuscin oxidation in vivo and the evidence of increased oxidative damage in AMD retina ex vivo lead to the conclusion that imaging of spectral characteristics of lipofuscin fluorescence may serve as a useful biomarker of oxidative damage, which can be helpful in assessing the efficacy of potential antioxidant therapies in retinal degenerations associated with accumulation of lipofuscin and increased oxidative stress. Finally, amendments to currently used fluorescence imaging instruments are suggested to be more sensitive and specific for imaging spectral characteristics of lipofuscin fluorescence.
Collapse
Affiliation(s)
- Małgorzata B. Różanowska
- School of Optometry and Vision Sciences, College of Biomedical and Life Sciences, Cardiff University, Maindy Road, Cardiff CF24 4HQ, Wales, UK;
- Cardiff Institute for Tissue Engineering and Repair (CITER), Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, Wales, UK
| |
Collapse
|
23
|
Neiteler A, Palakkan AA, Gallagher KM, Ross JA. Oxidative stress and docosahexaenoic acid injury lead to increased necroptosis and ferroptosis in retinal pigment epithelium. Sci Rep 2023; 13:21143. [PMID: 38036571 PMCID: PMC10689458 DOI: 10.1038/s41598-023-47721-5] [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: 05/01/2023] [Accepted: 11/17/2023] [Indexed: 12/02/2023] Open
Abstract
Age-related macular degeneration (AMD) is a complex disease caused by different genetic and environmental risk factors leading to loss of cells in the central part of the retina. Oxidative stress appears to be an important environmental risk factor that contributes to both the initiation and progression of AMD. Retinal pigment epithelium (RPE) plays an important role in regulating oxidative stress in the retina and is one of the main retinal cell types affected in AMD. A main function of RPE is to phagocytose photoreceptor outer segments (POS) which are rich in the polyunsaturated fatty acid (PUFA) docosahexaenoic acid (DHA), making this cell type potentially more susceptible to oxidative stress-induced lipid peroxidation which can lead to cell death. RPE is known to undergo necrotic cell death in response to oxidative stress. The aim of this study was to determine if DHA in POS can increase oxidative damage to RPE. It was found that RPE undergo increased lipid peroxidation and decreased cell viability when stressed with hydrogen peroxide in combination with DHA or POS. H2O2-induced oxidative stress was found to cause both ferroptosis and necroptosis. However, the ferroptosis regulator acyl-CoA synthetase long-chain family member 4 (ACSL4) was found to be downregulated in RPE exposed to H2O2 and this effect was exacerbated when the RPE cells were simultaneously treated with DHA. Together, these results show a response of RPE when stressed which will likely be overwhelmed under disease conditions such as AMD resulting in cell death.
Collapse
Affiliation(s)
- Almar Neiteler
- Tissue Injury and Repair Group, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK.
| | - Anwar A Palakkan
- Immunology and Stem Cell Biology, Aravind Medical Research Foundation, Anna Nagar, Madurai, 625020, India
| | - Kevin M Gallagher
- Tissue Injury and Repair Group, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - James A Ross
- Tissue Injury and Repair Group, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| |
Collapse
|
24
|
Chan-Ling T, Hu P, Li Calzi S, Warner J, Uddin N, DuPont M, Neuringer M, Kievit P, Renner L, Stoddard J, Ryals R, Boulton ME, McGill T, Grant MB. Glial, Neuronal, Vascular, Retinal Pigment Epithelium, and Inflammatory Cell Damage in a New Western Diet-Induced Primate Model of Diabetic Retinopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1789-1808. [PMID: 36965774 PMCID: PMC10616715 DOI: 10.1016/j.ajpath.2023.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/31/2023] [Accepted: 02/16/2023] [Indexed: 03/27/2023]
Abstract
This study investigated retinal changes in a Western diet (WD)-induced nonhuman primate model of type 2 diabetes. Rhesus nonhuman primates, aged 15 to 17 years, were fed a high-fat diet (n = 7) for >5 years reflective of the traditional WD. Age-matched controls (n = 6) were fed a standard laboratory primate diet. Retinal fundus photography, optical coherence tomography, autofluorescence imaging, and fluorescein angiography were performed before euthanasia. To assess diabetic retinopathy (DR), eyes were examined using trypsin digests, lipofuscin autofluorescence, and multimarker immunofluorescence on cross-sections and whole mounts. Retinal imaging showed venous engorgement and tortuosity, aneurysms, macular exudates, dot and blot hemorrhages, and a marked increase in fundus autofluorescence. Post-mortem changes included the following: decreased CD31 blood vessel density (P < 0.05); increased acellular capillaries (P < 0.05); increased density of ionized calcium-binding adaptor molecule expressing amoeboid microglia/macrophage; loss of regular distribution in stratum and spacing typical of ramified microglia; and increased immunoreactivity of aquaporin 4 and glial fibrillary acidic protein (P < 0.05). However, rhodopsin immunoreactivity (P < 0.05) in rods and neuronal nuclei antibody-positive neuronal density of 50% (P < 0.05) were decreased. This is the first report of a primate model of DR solely induced by a WD that replicates key features of human DR.
Collapse
Affiliation(s)
- Tailoi Chan-Ling
- Department of Anatomy, Faculty of Medicine and Health, Bosch Institute, University of Sydney, Camperdown, New South Wales, Australia.
| | - Ping Hu
- Department of Anatomy, Faculty of Medicine and Health, Bosch Institute, University of Sydney, Camperdown, New South Wales, Australia; Department of Ophthalmology and Visual Sciences, University of Alabama, Birmingham, Alabama
| | - Sergio Li Calzi
- Department of Ophthalmology and Visual Sciences, University of Alabama, Birmingham, Alabama
| | - Jeff Warner
- Department of Ophthalmology and Visual Sciences, University of Alabama, Birmingham, Alabama
| | - Nasir Uddin
- Department of Anatomy, Faculty of Medicine and Health, Bosch Institute, University of Sydney, Camperdown, New South Wales, Australia; Faculty of Science and Technology, Centre for Research in Therapeutic Solutions, University of Canberra, Bruce, Australian Capital Territory, Australia
| | - Mariana DuPont
- Department of Ophthalmology and Visual Sciences, University of Alabama, Birmingham, Alabama
| | - Martha Neuringer
- Department of Neuroscience, Oregon Health and Science University, Beaverton, Oregon
| | - Paul Kievit
- Division of Cardiometabolic Health, Oregon Health and Science University, Beaverton, Oregon
| | - Lauren Renner
- Department of Neuroscience, Oregon Health and Science University, Beaverton, Oregon
| | - Jonathan Stoddard
- Integrated Pathology Core, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon
| | - Renee Ryals
- Department of Ophthalmology, Oregon Health and Science University, Beaverton, Oregon
| | - Michael E Boulton
- Department of Ophthalmology and Visual Sciences, University of Alabama, Birmingham, Alabama
| | - Trevor McGill
- Department of Neuroscience, Oregon Health and Science University, Beaverton, Oregon
| | - Maria B Grant
- Department of Ophthalmology and Visual Sciences, University of Alabama, Birmingham, Alabama.
| |
Collapse
|
25
|
Plau J, Morgan CE, Fedorov Y, Banerjee S, Adams DJ, Blaner WS, Yu EW, Golczak M. Discovery of Nonretinoid Inhibitors of CRBP1: Structural and Dynamic Insights for Ligand-Binding Mechanisms. ACS Chem Biol 2023; 18:2309-2323. [PMID: 37713257 PMCID: PMC10591915 DOI: 10.1021/acschembio.3c00402] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/01/2023] [Indexed: 09/16/2023]
Abstract
The dysregulation of retinoid metabolism has been linked to prevalent ocular diseases including age-related macular degeneration and Stargardt disease. Modulating retinoid metabolism through pharmacological approaches holds promise for the treatment of these eye diseases. Cellular retinol-binding protein 1 (CRBP1) is the primary transporter of all-trans-retinol (atROL) in the eye, and its inhibition has recently been shown to protect mouse retinas from light-induced retinal damage. In this report, we employed high-throughput screening to identify new chemical scaffolds for competitive, nonretinoid inhibitors of CRBP1. To understand the mechanisms of interaction between CRBP1 and these inhibitors, we solved high-resolution X-ray crystal structures of the protein in complex with six selected compounds. By combining protein crystallography with hydrogen/deuterium exchange mass spectrometry, we quantified the conformational changes in CRBP1 caused by different inhibitors and correlated their magnitude with apparent binding affinities. Furthermore, using molecular dynamic simulations, we provided evidence for the functional significance of the "closed" conformation of CRBP1 in retaining ligands within the binding pocket. Collectively, our study outlines the molecular foundations for understanding the mechanism of high-affinity interactions between small molecules and CRBPs, offering a framework for the rational design of improved inhibitors for this class of lipid-binding proteins.
Collapse
Affiliation(s)
- Jacqueline Plau
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Christopher E. Morgan
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
- Department
of Chemistry, Thiel College, Greenville, Pennsylvania 16125, United States
| | - Yuriy Fedorov
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Surajit Banerjee
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14850, United States
- Northeastern
Collaborative Access Team, Argonne National
Laboratory, Argonne, Illinois 60439, United States
| | - Drew J. Adams
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - William S. Blaner
- Department
of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York 10032, United States
| | - Edward W. Yu
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Marcin Golczak
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| |
Collapse
|
26
|
Kaltak M, Corradi Z, Collin RWJ, Swildens J, Cremers FPM. Stargardt disease-associated missense and synonymous ABCA4 variants result in aberrant splicing. Hum Mol Genet 2023; 32:3078-3089. [PMID: 37555651 PMCID: PMC10586196 DOI: 10.1093/hmg/ddad129] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/03/2023] [Accepted: 08/03/2023] [Indexed: 08/10/2023] Open
Abstract
Missense variants in ABCA4 constitute ~50% of causal variants in Stargardt disease (STGD1). Their pathogenicity is attributed to their direct effect on protein function, whilst their potential impact on pre-mRNA splicing disruption remains poorly understood. Interestingly, synonymous ABCA4 variants have previously been classified as 'severe' variants based on in silico analyses. Here, we systemically investigated the role of synonymous and missense variants in ABCA4 splicing by combining computational predictions and experimental assays. To identify variants of interest, we used SpliceAI to ascribe defective splice predictions on a dataset of 5579 biallelic STGD1 probands. We selected those variants with predicted delta scores for acceptor/donor gain > 0.20, and no previous reports on their effect on splicing. Fifteen ABCA4 variants were selected, 4 of which were predicted to create a new splice acceptor site and 11 to create a new splice donor site. In addition, three variants of interest with delta scores < 0.20 were included. The variants were introduced in wild-type midigenes that contained 4-12 kb of ABCA4 genomic sequence, which were subsequently expressed in HEK293T cells. By using RT-PCR and Sanger sequencing, we identified splice aberrations for 16 of 18 analyzed variants. SpliceAI correctly predicted the outcomes for 15 out of 18 variants, illustrating its reliability in predicting the impact of coding ABCA4 variants on splicing. Our findings highlight a causal role for coding ABCA4 variants in splicing aberrations, improving the severity assessment of missense and synonymous ABCA4 variants, and guiding to new treatment strategies for STGD1.
Collapse
Affiliation(s)
- Melita Kaltak
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, 6525 GA, The Netherlands
- R&D Department, ProQR Therapeutics, Leiden, 2333 CK, The Netherlands
| | - Zelia Corradi
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, 6525 GA, The Netherlands
| | - Rob W J Collin
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, 6525 GA, The Netherlands
| | - Jim Swildens
- R&D Department, ProQR Therapeutics, Leiden, 2333 CK, The Netherlands
| | - Frans P M Cremers
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, 6525 GA, The Netherlands
| |
Collapse
|
27
|
Corradi Z, Khan M, Hitti-Malin R, Mishra K, Whelan L, Cornelis SS, Hoyng CB, Kämpjärvi K, Klaver CCW, Liskova P, Stöhr H, Weber BHF, Banfi S, Farrar GJ, Sharon D, Zernant J, Allikmets R, Dhaenens CM, Cremers FPM. Targeted sequencing and in vitro splice assays shed light on ABCA4-associated retinopathies missing heritability. HGG ADVANCES 2023; 4:100237. [PMID: 37705246 PMCID: PMC10534262 DOI: 10.1016/j.xhgg.2023.100237] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/15/2023] Open
Abstract
The ABCA4 gene is the most frequently mutated Mendelian retinopathy-associated gene. Biallelic variants lead to a variety of phenotypes, however, for thousands of cases the underlying variants remain unknown. Here, we aim to shed further light on the missing heritability of ABCA4-associated retinopathy by analyzing a large cohort of macular dystrophy probands. A total of 858 probands were collected from 26 centers, of whom 722 carried no or one pathogenic ABCA4 variant, while 136 cases carried two ABCA4 alleles, one of which was a frequent mild variant, suggesting that deep-intronic variants (DIVs) or other cis-modifiers might have been missed. After single molecule molecular inversion probes (smMIPs)-based sequencing of the complete 128-kb ABCA4 locus, the effect of putative splice variants was assessed in vitro by midigene splice assays in HEK293T cells. The breakpoints of copy number variants (CNVs) were determined by junction PCR and Sanger sequencing. ABCA4 sequence analysis solved 207 of 520 (39.8%) naive or unsolved cases and 70 of 202 (34.7%) monoallelic cases, while additional causal variants were identified in 54 of 136 (39.7%) probands carrying two variants. Seven novel DIVs and six novel non-canonical splice site variants were detected in a total of 35 alleles and characterized, including the c.6283-321C>G variant leading to a complex splicing defect. Additionally, four novel CNVs were identified and characterized in five alleles. These results confirm that smMIPs-based sequencing of the complete ABCA4 gene provides a cost-effective method to genetically solve retinopathy cases and that several rare structural and splice altering defects remain undiscovered in Stargardt disease cases.
Collapse
Affiliation(s)
- Zelia Corradi
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Mubeen Khan
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands; Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
| | - Rebekkah Hitti-Malin
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ketan Mishra
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Laura Whelan
- The School of Genetics & Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Stéphanie S Cornelis
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Carel B Hoyng
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Caroline C W Klaver
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands; Institute of Molecular & Clinical Ophthalmology, Basel, Switzerland
| | - Petra Liskova
- Research Unit for Rare Diseases, Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic; Department of Ophthalmology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Heidi Stöhr
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
| | - Bernhard H F Weber
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany; Institute of Clinical Human Genetics, University Hospital Regensburg, Regensburg, Germany
| | - Sandro Banfi
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli," Naples and Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - G Jane Farrar
- The School of Genetics & Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Dror Sharon
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jana Zernant
- Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Rando Allikmets
- Department of Ophthalmology, Columbia University, New York, NY, USA; Department of Pathology & Cell Biology, Columbia University, New York, NY, USA
| | - Claire-Marie Dhaenens
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands; University Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, 59000 Lille, France
| | - Frans P M Cremers
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
28
|
Shome I, Thathapudi NC, Aramati BMR, Kowtharapu BS, Jangamreddy JR. Stages, pathogenesis, clinical management and advancements in therapies of age-related macular degeneration. Int Ophthalmol 2023; 43:3891-3909. [PMID: 37347455 DOI: 10.1007/s10792-023-02767-2] [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: 02/16/2022] [Accepted: 06/08/2023] [Indexed: 06/23/2023]
Abstract
Age-related macular degeneration (AMD) is a retinal degenerative disorder prevalent in the elderly population, which leads to the loss of central vision. The disease progression can be managed, if not prevented, either by blocking neovascularization ("wet" form of AMD) or by preserving retinal pigment epithelium and photoreceptor cells ("dry" form of AMD). Although current therapeutic modalities are moderately successful in delaying the progression and management of the disease, advances over the past years in regenerative medicine using iPSC, embryonic stem cells, advanced materials (including nanomaterials) and organ bio-printing show great prospects in restoring vision and efficient management of either forms of AMD. This review focuses on the molecular mechanism of the disease, model systems (both cellular and animal) used in studying AMD, the list of various regenerative therapies and the current treatments available. The article also highlights on the recent clinical trials using regenerative therapies and management of the disease.
Collapse
Affiliation(s)
- Ishita Shome
- UR Advanced Therapeutics Private Limited, ASPIRE-BioNest, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, 500046, India
| | - Neethi C Thathapudi
- Centre de Recherche Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
- Department of Ophthalmology and Institute of Biomedical Engineering, Université de Montréal, Montréal, QC, Canada
| | - Bindu Madhav Reddy Aramati
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, 500046, India
| | - Bhavani S Kowtharapu
- UR Advanced Therapeutics Private Limited, ASPIRE-BioNest, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, 500046, India
| | - Jaganmohan R Jangamreddy
- UR Advanced Therapeutics Private Limited, ASPIRE-BioNest, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, 500046, India.
| |
Collapse
|
29
|
Lee JR, Jeong KW. N-retinylidene- N-retinylethanolamine degradation in human retinal pigment epithelial cells via memantine- and ifenprodil-mediated autophagy. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2023; 27:449-456. [PMID: 37641807 PMCID: PMC10466070 DOI: 10.4196/kjpp.2023.27.5.449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 08/31/2023]
Abstract
N-methyl-D-aspartate (NMDA) receptors are ionic glutamine receptors involved in brain development and functions such as learning and memory formation. NMDA receptor inhibition is associated with autophagy activation. In this study, we investigated whether the NMDA receptor antagonists, memantine and ifenprodil, induce autophagy in human retinal pigment epithelial cells (ARPE-19) to remove Nretinylidene- N-retinylethanolamine (A2E), an intracellular lipofuscin component. Fluorometric analysis using labeled A2E (A2E-BDP) and confocal microscopic examination revealed that low concentrations of NMDA receptor antagonists, which did not induce cytotoxicity, significantly reduced A2E accumulation in ARPE-19 cells. In addition, memantine and ifenprodil activated autophagy in ARPE-19 cells as measured by microtubule-associated protein 1A/1B-light chain3-II formation and phosphorylated p62 protein levels. Further, to understand the correlation between memantine- and ifenprodil-mediated A2E degradation and autophagy, autophagy-related 5 (ATG5) was depleted using RNA interference. Memantine and ifenprodil failed to degrade A2E in ARPE-19 cells lacking ATG5. Taken together, our study indicates that the NMDA receptor antagonists, memantine and ifenprodil, can remove A2E accumulated in cells via autophagy activation in ARPE-19 cells.
Collapse
Affiliation(s)
- Jae Rim Lee
- College of Pharmacy, Gachon Research Institute of Pharmaceutical Sciences, Gachon University, Incheon 21936, Korea
| | - Kwang Won Jeong
- College of Pharmacy, Gachon Research Institute of Pharmaceutical Sciences, Gachon University, Incheon 21936, Korea
| |
Collapse
|
30
|
Farnoodian M, Bose D, Barone F, Nelson LM, Boyle M, Jun B, Do K, Gordon W, Guerin MAK, Perera R, Ji JX, Cogliati T, Sharma R, Brooks BP, Bazan NG, Bharti K. Retina and RPE lipid profile changes linked with ABCA4 associated Stargardt's maculopathy. Pharmacol Ther 2023; 249:108482. [PMID: 37385300 PMCID: PMC10530239 DOI: 10.1016/j.pharmthera.2023.108482] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/20/2023] [Accepted: 06/26/2023] [Indexed: 07/01/2023]
Abstract
Stargardt maculopathy, caused predominantly by mutations in the ABCA4 gene, is characterized by an accumulation of non-degradable visual pigment derivative, lipofuscin, in the retinal pigment epithelium (RPE) - resulting in RPE atrophy. RPE is a monolayer tissue located adjacent to retinal photoreceptors and regulates their health and functioning; RPE atrophy triggers photoreceptor cell death and vision loss in Stargardt patients. Previously, ABCA4 mutations in photoreceptors were thought to be the major contributor to lipid homeostasis defects in the eye. Recently, we demonstrated that ABCA4 loss of function in the RPE leads to cell-autonomous lipid homeostasis defects. Our work underscores that an incomplete understanding of lipid metabolism and lipid-mediated signaling in the retina and RPE are potential causes for lacking treatments for this disease. Here we report altered lipidomic in mouse and human Stargardt models. This work provides the basis for therapeutics that aim to restore lipid homeostasis in the retina and the RPE.
Collapse
Affiliation(s)
- Mitra Farnoodian
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institute of Health, Bethesda, MD, USA
| | - Devika Bose
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institute of Health, Bethesda, MD, USA
| | - Francesca Barone
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institute of Health, Bethesda, MD, USA
| | - Luke Mathew Nelson
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institute of Health, Bethesda, MD, USA
| | - Marisa Boyle
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institute of Health, Bethesda, MD, USA
| | - Bokkyoo Jun
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, USA
| | - Khanh Do
- Faculty of Medicine, Phenikaa University, Hanoi, Viet Nam
| | - William Gordon
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, USA
| | - Marie-Audrey Kautzmann Guerin
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, USA
| | - Rasangi Perera
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, USA
| | - Jeff X Ji
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, USA
| | - Tiziana Cogliati
- Division of Aging Biology, National Institute on Aging, National Institute of Health, Bethesda, MD, USA
| | - Ruchi Sharma
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institute of Health, Bethesda, MD, USA
| | - Brian P Brooks
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institute of Health, Bethesda, MD, USA
| | - Nicolas G Bazan
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, USA
| | - Kapil Bharti
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institute of Health, Bethesda, MD, USA.
| |
Collapse
|
31
|
Abstract
Endogenous photosensitizers play a critical role in both beneficial and harmful light-induced transformations in biological systems. Understanding their mode of action is essential for advancing fields such as photomedicine, photoredox catalysis, environmental science, and the development of sun care products. This review offers a comprehensive analysis of endogenous photosensitizers in human skin, investigating the connections between their electronic excitation and the subsequent activation or damage of organic biomolecules. We gather the physicochemical and photochemical properties of key endogenous photosensitizers and examine the relationships between their chemical reactivity, location within the skin, and the primary biochemical events following solar radiation exposure, along with their influence on skin physiology and pathology. An important take-home message of this review is that photosensitization allows visible light and UV-A radiation to have large effects on skin. The analysis presented here unveils potential causes for the continuous increase in global skin cancer cases and emphasizes the limitations of current sun protection approaches.
Collapse
Affiliation(s)
- Erick L Bastos
- Department of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, 05508-000 São Paulo, São Paulo, Brazil
| | - Frank H Quina
- Department of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, 05508-000 São Paulo, São Paulo, Brazil
- Department of Chemical Engineering, Polytechnic School, University of São Paulo, 05508-000 São Paulo, São Paulo, Brazil
| | - Maurício S Baptista
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, 05508-000 São Paulo, São Paulo, Brazil
| |
Collapse
|
32
|
Wada I, Mori K, Sreekumar PG, Ji R, Spee C, Hong E, Ishikawa K, Sonoda KH, Kannan R. Characterization and contribution of RPE senescence to Age-related macular degeneration in Tnfrsf10 knock out mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.04.552052. [PMID: 37577701 PMCID: PMC10418235 DOI: 10.1101/2023.08.04.552052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Background Retinal pigment epithelial cells (RPE) play vital role in the pathogenesis of age-related macular degeneration (AMD). Our laboratory has shown that RPE cellular senescence contributed to the pathophysiology of experimental AMD, and SASP members are involved in this process. Recently, we presented confirmatory evidence to earlier GWAS studies that dysregulation of tumor necrosis factor receptor superfamily 10A (TNFRSF10A) dysregulation leads to AMD development and is linked to RPE dysfunction. This study aims to investigate the contribution of RPE senescence to AMD pathophysiology using TNFRSF10A silenced human RPE (hRPE) cells and Tnfrsf10 KO mice. Methods Sub-confluent primary hRPE cells and TNFRSF10A silenced hRPE were exposed to stress-induced premature senescence with H2O2 (500 μM, 48h), and senescence-associated markers (βgal, p16, and p21) were analyzed by RT-PCR and WB analysis. The effect of H2O2-induced senescence in non-silenced and silenced hRPE on OXPHOS and glycolysis was determined using Seahorse XF96 analyzer. Male C57BL/6J Tnfrsf10 KO ( Tnfrsf10 -/- ) mice were used to study the regulation of senescence by TNFRSF10A in vivo . Expression of p16 and p21 in control and KO mice of varying ages were determined by RT-PCR, WB, and immunostaining analysis. Results The senescence-associated p16 and p21 showed a significant ( p < 0.01) upregulation with H2O2 induction at the gene (1.8- and 3-fold) and protein (3.2- and 4-fold) levels in hRPE cells. The protein expression of p16 and p21 was further significantly increased by co-treatment with siRNA ( p < 0.05 vs. H2O2). Mitochondrial oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) (pmol/min/total DNA) increased with senescence induction by H2O2 for 48h in control RPE, and knockdown of TNFRSF10A caused a further increase in OCR and ECAR. In addition, co-treatment with PKC activator significantly improved all parameters. Similarly, in vivo studies showed upregulation of p16 and p21 by RT-PCR, WB, and immunostaining analysis in RPE/choroid of Tnfrsf10 KO mice. When subjected to examination across distinct age groups, namely young (1-3 months), middle (6-9 months), and old (12-15 months) mice, a discernible age-related elevation in the expression of p16 and p21 was observed. Conclusions Our findings suggest that TNRSF10A is a regulator of regulates in RPE senescence. Further work on elucidating pathways of senescence will facilitate the development of new therapeutic targets for AMD.
Collapse
|
33
|
Whelan L, Dockery A, Stephenson KAJ, Zhu J, Kopčić E, Post IJM, Khan M, Corradi Z, Wynne N, O' Byrne JJ, Duignan E, Silvestri G, Roosing S, Cremers FPM, Keegan DJ, Kenna PF, Farrar GJ. Detailed analysis of an enriched deep intronic ABCA4 variant in Irish Stargardt disease patients. Sci Rep 2023; 13:9380. [PMID: 37296172 PMCID: PMC10256698 DOI: 10.1038/s41598-023-35889-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Over 15% of probands in a large cohort of more than 1500 inherited retinal degeneration patients present with a clinical diagnosis of Stargardt disease (STGD1), a recessive form of macular dystrophy caused by biallelic variants in the ABCA4 gene. Participants were clinically examined and underwent either target capture sequencing of the exons and some pathogenic intronic regions of ABCA4, sequencing of the entire ABCA4 gene or whole genome sequencing. ABCA4 c.4539 + 2028C > T, p.[= ,Arg1514Leufs*36] is a pathogenic deep intronic variant that results in a retina-specific 345-nucleotide pseudoexon inclusion. Through analysis of the Irish STGD1 cohort, 25 individuals across 18 pedigrees harbour ABCA4 c.4539 + 2028C > T and another pathogenic variant. This includes, to the best of our knowledge, the only two homozygous patients identified to date. This provides important evidence of variant pathogenicity for this deep intronic variant, highlighting the value of homozygotes for variant interpretation. 15 other heterozygous incidents of this variant in patients have been reported globally, indicating significant enrichment in the Irish population. We provide detailed genetic and clinical characterization of these patients, illustrating that ABCA4 c.4539 + 2028C > T is a variant of mild to intermediate severity. These results have important implications for unresolved STGD1 patients globally with approximately 10% of the population in some western countries claiming Irish heritage. This study exemplifies that detection and characterization of founder variants is a diagnostic imperative.
Collapse
Affiliation(s)
- Laura Whelan
- The School of Genetics and Microbiology, Trinity College Dublin, Dublin 2, Ireland.
| | - Adrian Dockery
- The School of Genetics and Microbiology, Trinity College Dublin, Dublin 2, Ireland
- Next Generation Sequencing Laboratory, Pathology Department, The Mater Misericordiae University Hospital, Dublin 7, Ireland
| | - Kirk A J Stephenson
- Department of Ophthalmology, Royal Victoria Eye and Ear Hospital, Dublin 2, Ireland
- Mater Clinical Ophthalmic Genetics Unit, The Mater Misericordiae University Hospital, Dublin 7, Ireland
| | - Julia Zhu
- Mater Clinical Ophthalmic Genetics Unit, The Mater Misericordiae University Hospital, Dublin 7, Ireland
| | - Ella Kopčić
- The School of Genetics and Microbiology, Trinity College Dublin, Dublin 2, Ireland
| | - Iris J M Post
- The School of Genetics and Microbiology, Trinity College Dublin, Dublin 2, Ireland
| | - Mubeen Khan
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
- International Max Planck Research School for Language Sciences, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
| | - Zelia Corradi
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Academic Alliance Genetics, Radboud University Medical Center, Nijmegen, and Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Niamh Wynne
- Department of Ophthalmology, Royal Victoria Eye and Ear Hospital, Dublin 2, Ireland
| | - James J O' Byrne
- Mater Clinical Ophthalmic Genetics Unit, The Mater Misericordiae University Hospital, Dublin 7, Ireland
- International Max Planck Research School for Language Sciences, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
- National Centre for Inherited Metabolic Disorders, The Mater Misericordiae University Hospital, Dublin 7, Ireland
- School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Emma Duignan
- Department of Ophthalmology, Royal Victoria Eye and Ear Hospital, Dublin 2, Ireland
| | - Giuliana Silvestri
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, UK
- School of Medicine, University College Dublin, Dublin 4, Ireland
- Department of Ophthalmology, The Royal Victoria Hospital, Belfast, Northern Ireland, UK
| | - Susanne Roosing
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Academic Alliance Genetics, Radboud University Medical Center, Nijmegen, and Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Frans P M Cremers
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Academic Alliance Genetics, Radboud University Medical Center, Nijmegen, and Maastricht University Medical Center+, Maastricht, The Netherlands
| | - David J Keegan
- Mater Clinical Ophthalmic Genetics Unit, The Mater Misericordiae University Hospital, Dublin 7, Ireland
| | - Paul F Kenna
- The School of Genetics and Microbiology, Trinity College Dublin, Dublin 2, Ireland
- Department of Ophthalmology, Royal Victoria Eye and Ear Hospital, Dublin 2, Ireland
| | - G Jane Farrar
- The School of Genetics and Microbiology, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
34
|
Huang X, Zhang L, Fu Y, Zhang M, Yang Q, Peng J. Rethinking the potential and necessity of drug delivery systems in neovascular age-related macular degeneration therapy. Front Bioeng Biotechnol 2023; 11:1199922. [PMID: 37288355 PMCID: PMC10242387 DOI: 10.3389/fbioe.2023.1199922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/09/2023] [Indexed: 06/09/2023] Open
Abstract
Age-related macular degeneration (AMD) is the predominant threat to human vision and ultimately results in blindness. With the increase in the aging population, it has become a more crucial issue to human health. AMD is a multifactorial disease with the unique feature of uncontrollable angiogenesis during initiation and progression. Although increasing evidence indicates that AMD is largely hereditary, the predominant efficient treatment is antiangiogenesis, which mainly involves VEGF and HIF-α as therapeutic targets. The repeated administration of this treatment over the long term, generally through intravitreal injection, has called for the introduction of long-term drug delivery systems, which are expected to be achieved by biomaterials. However, the clinical results of the port delivery system indicate that the optimization of medical devices toward prolonging the activities of therapeutic biologics in AMD therapy seems more promising. These results indicate that we should rethink the possibility and potential of biomaterials as drug delivery systems in achieving long-term, sustained inhibition of angiogenesis in AMD therapy. In this review, the etiology, categorization, risk factors, pathogenesis, and current clinical treatments of AMD are briefly introduced. Next, the development status of long-term drug delivery systems is discussed, and the drawbacks and shortages of these systems are emphasized. By comprehensively considering the pathological aspect and the recent application of drug delivery systems in AMD therapy, we hope to find a better solution for the further development of long-term therapeutic strategies for AMD.
Collapse
Affiliation(s)
- Xi Huang
- Department of Ophthalmology, Research Laboratory of Macular Disease, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Zhang
- Department of Ophthalmology, Research Laboratory of Macular Disease, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanyan Fu
- Department of Ophthalmology, Research Laboratory of Macular Disease, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Meixia Zhang
- Department of Ophthalmology, Research Laboratory of Macular Disease, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qian Yang
- Center of Scientific Research, Chengdu Medical College, Chengdu, Sichuan, China
| | - Jinrong Peng
- Department of Ophthalmology, Research Laboratory of Macular Disease, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
35
|
Chang YJ, Jenny L, Li YS, Cui X, Kong Y, Li Y, Sparrow J, Tsang S. CRISPR editing demonstrates rs10490924 raised oxidative stress in iPSC-derived retinal cells from patients with ARMS2/HTRA1-related AMD. Proc Natl Acad Sci U S A 2023; 120:e2215005120. [PMID: 37126685 PMCID: PMC10175836 DOI: 10.1073/pnas.2215005120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 02/07/2023] [Indexed: 05/03/2023] Open
Abstract
Genome-wide association studies (GWAS) have identified genetic risk loci for age-related macular degeneration (AMD) on the chromosome 10q26 (Chr10) locus and are tightly linked: the A69S (G>T) rs10490924 single-nucleotide variant (SNV) and the AATAA-rich insertion-deletion (indel, del443/ins54), which are found in the age-related maculopathy susceptibility 2 (ARMS2) gene, and the G512A (G>A) rs11200638 SNV, which is found in the high-temperature requirement A serine peptidase 1 (HTRA1) promoter. The fourth variant is Y402H complement factor H (CFH), which directs CFH signaling. CRISPR manipulation of retinal pigment epithelium (RPE) cells may allow one to isolate the effects of the individual SNV and thus identify SNV-specific effects on cell phenotype. Clustered regularly interspaced short palindromic repeats (CRISPR) editing demonstrates that rs10490924 raised oxidative stress in induced pluripotent stem cell (iPSC)-derived retinal cells from patients with AMD. Sodium phenylbutyrate preferentially reverses the cell death caused by ARMS2 rs10490924 but not HTRA1 rs11200638. This study serves as a proof of concept for the use of patient-specific iPSCs for functional annotation of tightly linked GWAS to study the etiology of a late-onset disease phenotype. More importantly, we demonstrate that antioxidant administration may be useful for reducing reactive oxidative stress in AMD, a prevalent late-onset neurodegenerative disorder.
Collapse
Affiliation(s)
- Ya-Ju Chang
- Jonas Children’s Vision Care, Department of Ophthalmology, Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY10032
| | - Laura A. Jenny
- Jonas Children’s Vision Care, Department of Ophthalmology, Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY10032
| | - Yong-Shi Li
- Jonas Children’s Vision Care, Department of Ophthalmology, Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY10032
| | - Xuan Cui
- Jonas Children’s Vision Care, Department of Ophthalmology, Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY10032
| | - Yang Kong
- Jonas Children’s Vision Care, Department of Ophthalmology, Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY10032
| | - Yao Li
- Jonas Children’s Vision Care, Department of Ophthalmology, Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY10032
| | - Janet R. Sparrow
- Jonas Children’s Vision Care, Department of Ophthalmology, Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY10032
- Department of Ophthalmology, Columbia University, New York, NY10032
- Department of Biomedical Engineering, Columbia University, New York, NY10032
- Department of Pathology and Cell Biology, Columbia University, New York, NY10032
| | - Stephen H. Tsang
- Jonas Children’s Vision Care, Department of Ophthalmology, Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY10032
- Department of Ophthalmology, Columbia University, New York, NY10032
- Department of Biomedical Engineering, Columbia University, New York, NY10032
- Department of Pathology and Cell Biology, Columbia University, New York, NY10032
- Institute of Human Nutrition, and Columbia Stem Cell Initiative, Columbia University, New York, NY10032
| |
Collapse
|
36
|
Cellular and Molecular Mechanisms of Pathogenesis Underlying Inherited Retinal Dystrophies. Biomolecules 2023; 13:biom13020271. [PMID: 36830640 PMCID: PMC9953031 DOI: 10.3390/biom13020271] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Inherited retinal dystrophies (IRDs) are congenital retinal degenerative diseases that have various inheritance patterns, including dominant, recessive, X-linked, and mitochondrial. These diseases are most often the result of defects in rod and/or cone photoreceptor and retinal pigment epithelium function, development, or both. The genes associated with these diseases, when mutated, produce altered protein products that have downstream effects in pathways critical to vision, including phototransduction, the visual cycle, photoreceptor development, cellular respiration, and retinal homeostasis. The aim of this manuscript is to provide a comprehensive review of the underlying molecular mechanisms of pathogenesis of IRDs by delving into many of the genes associated with IRD development, their protein products, and the pathways interrupted by genetic mutation.
Collapse
|
37
|
TRANSPLANTATION OF SUBRETINAL STEM CELL-DERIVED RETINAL PIGMENT EPITHELIUM FOR STARGARDT DISEASE: A Phase I Clinical Trial. Retina 2023; 43:263-274. [PMID: 36223778 DOI: 10.1097/iae.0000000000003655] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
PURPOSE To assess the safety of injecting human embryonic stem cell retinal pigment epithelial cell dose to treat Stargardt disease. METHODS In this prospective, Phase I clinical trial, human embryonic stem cell retinal pigment epithelial cells in suspension were injected into the subretinal space in eyes with the worse best-corrected visual acuity (BCVA). After vitrectomy/posterior hyaloid removal, a partial retinal detachment was created and the human embryonic stem cell retinal pigment epithelial cells were administered. Phacoemulsification with intraocular lens implantation was performed in eyes with lens opacity. All procedures were optical coherence tomography-guided. The 12-month follow-up included retinal imaging, optical coherence tomography, visual field/electrophysiologic testing, and systemic evaluation. The main outcome was the absence of ocular/systemic inflammation or rejection, tumor formation, or toxicity during follow-up. RESULTS The mean baseline BCVAs in the phacoemulsification and no phacoemulsification groups were similar (1.950 ± 0.446 and 1.575 ± 0.303, respectively). One year postoperatively, treated eyes showed a nonsignificant increase in BCVA. No adverse effects occurred during follow-up. Intraoperative optical coherence tomography was important for guiding all procedures. CONCLUSION This surgical procedure was feasible and safe without cellular migration, rejection, inflammation, or development of ocular or systemic tumors during follow-up.
Collapse
|
38
|
Su N, Hansen U, Plagemann T, Gäher K, Leclaire MD, König J, Höhn A, Grune T, Uhlig CE, Eter N, Heiduschka P. Sub-Retinal Injection of Human Lipofuscin in the Mouse - A Model of "Dry" Age-Related Macular Degeneration? Aging Dis 2023; 14:184-203. [PMID: 36818570 PMCID: PMC9937713 DOI: 10.14336/ad.2022.0626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/26/2022] [Indexed: 11/18/2022] Open
Abstract
Lipofuscin (LF) accumulates during lifetime in the retinal pigment epithelium (RPE) and is thought to play a crucial role in intermediate and late age-related macular degeneration (AMD). In an attemt to simulate aged retina and to study response of retinal microglia and RPE cells to LF, we injected a suspension of LF into the subretinal space of adult mice. LF suspension was obtained from human donor eyes. Subretinal injection of PBS or sham injection served as a control. Eyes were inspected by autofluorescence and optical coherence tomography, by electroretinography and on histological and ultrastructural levels. Levels of cytokine mRNA were determined by quantitative PCR separately in the RPE/choroid complex and in the retina. After injection of LF, microglial cells migrated quickly into the subretinal space to close proximity to RPE cells and phagocytosed LF particles. Retinal function was affected only slightly by LF within the first two weeks. After longer time, RPE cells showed clear signs of melanin loss and degradation. Levels of mRNA of inflammatory cytokines increased sharply after injection of both PBS and LF and were higher in the RPE/choroid complex than in the retina and were slightly higher after LF injection. In conclusion, subretinal injection of LF causes an activation of microglial cells and their migration into subretinal space, enhanced expression of inflammatory cytokines and a gradual degradation of RPE cells. These features are found also in an aging retina, and subretinal injection of LF could be a model for intermediate and late AMD.
Collapse
Affiliation(s)
- Nan Su
- Research Laboratory, Department of Ophthalmology, University
Medical Center, Münster, Germany.
- Department of Ophthalmology, The First Affiliated Hospital of
Zhengzhou University, Zhengzhou, China.
| | - Uwe Hansen
- Institute of Musculoskeletal Medicine, Medical Faculty,
University of Münster, Münster, Germany.
| | - Tanja Plagemann
- Research Laboratory, Department of Ophthalmology, University
Medical Center, Münster, Germany.
| | - Karin Gäher
- Institute of Musculoskeletal Medicine, Medical Faculty,
University of Münster, Münster, Germany.
| | - M. Dominik Leclaire
- Research Laboratory, Department of Ophthalmology, University
Medical Center, Münster, Germany.
| | - Jeannette König
- German Institute of Human Nutrition,
Potsdam-Rehbrücke, Germany.
| | - Annika Höhn
- German Institute of Human Nutrition,
Potsdam-Rehbrücke, Germany.
| | - Tilman Grune
- German Institute of Human Nutrition,
Potsdam-Rehbrücke, Germany.
| | - Constantin E. Uhlig
- Cornea Bank Münster, Department of Ophthalmology,
University Medical Center, Münster, Germany.
| | - Nicole Eter
- Department of Ophthalmology, University Medical Center,
Münster, Germany.
| | - Peter Heiduschka
- Research Laboratory, Department of Ophthalmology, University
Medical Center, Münster, Germany.
| |
Collapse
|
39
|
Theruveethi N, Joshi MB, Jathanna JS, Valiathan M, Kabekkodu SP, Bhandarkar M, Thomas RH, Thangarajan R, Bhat SS, Surendran S. Effect of Light Emitting Diodes (LED) Exposure on Vitreous Metabolites-Rodent Study. Metabolites 2023; 13:81. [PMID: 36677006 PMCID: PMC9861686 DOI: 10.3390/metabo13010081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/20/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023] Open
Abstract
The exposure to blue and white Light emitting diodes (LED) light leads to damage in the visual system with short-term LED light exposure. Chronic exposure, adaptive responses to light, and self-protective mechanisms against LED light exposures need to be explored, and it would be essential to understand the repercussions of LED radiation on vitreous metabolites. A total of 24 male Wistar rats were used in this study, divided into four groups (n = 6 in each group). Three experimental groups of rats were exposed to either blue, white, or yellow LED light for 90 days (12:12 light-dark cycle routine) with uniform illumination (450−500 lux). Standard lab settings were used to maintain control rats. Vitreous fluids were subjected to untargeted metabolomics analysis using liquid chromatography-mass spectrometry (LC/MS). PLS-DA analysis indicated significant the separation of m metabolites among groups, suggesting that LED exposure induces metabolic reprogramming in the vitreous. Amino acids and their modifications showed significant alterations among groups which included D-alanine, D-serine (p < 0.05), lysine (p < 0.001), aspartate (p = 0.0068), glutathione (p = 0.0263), taurine (p = 0.007), and hypotaurine. In chronic light exposure, the self-protective or reworking system could be depleted, which may decrease the ability to compensate for the defending mechanism. This might fail to maintain the metabolomic structural integrity of the vitreous metabolites.
Collapse
Affiliation(s)
- Nagarajan Theruveethi
- Department of Optometry, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal 576104, India
| | - Manjunath B. Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Judith S. Jathanna
- Department of Optometry, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal 576104, India
| | - Manna Valiathan
- Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Manasa Bhandarkar
- Department of Optometry, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal 576104, India
| | - R. Huban Thomas
- Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Rajesh Thangarajan
- Department of Anatomy, International Medical School, Management and Science University (MSU), Shah Alam 40100, Malaysia
| | - Shailaja S. Bhat
- Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Sudarshan Surendran
- American University of Antigua College of Medicine, University Park, Jabberwock Beach Road, Coolidge P.O. Box 1451, Antigua and Barbuda
| |
Collapse
|
40
|
Dysli C, Dysli M, Wolf S, Zinkernagel M. Fluorescence lifetime distribution in phakic and pseudophakic healthy eyes. PLoS One 2023; 18:e0279158. [PMID: 36608033 PMCID: PMC9821472 DOI: 10.1371/journal.pone.0279158] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 11/14/2022] [Indexed: 01/07/2023] Open
Abstract
PURPOSE To investigate the influence of the lens status and to describe fundus autofluorescence lifetimes (FLT) in a large cohort of healthy eyes across a wide age range. MATERIALS AND METHODS FLT data were acquired from healthy phakic and pseudophakic eyes using fluorescence lifetime imaging ophthalmoscopy (FLIO). Retinal autofluorescence was excited with a 473 nm laser and emitted autofluorescence was detected in a short and a long spectral channel (SSC: 498-560 nm; LSC: 560-720 nm). RESULTS 141 healthy eyes from 141 participants (56 ± 18 years) were included. The shortest mean FLTs were measured within the macular center, followed by the temporal inner and outer ETDRS (Early Treatment Diabetic Retinopathy Study) grid segments, and the remaining areas of the inner and the outer ETDRS ring. In phakic participants (81%), mean, short and long FLTs correlated with the age (SSC: r2 = 0.54; LSC: r2 = 0.7; both p<0.0001) with an increase of about 33 ps in the SSC resp. 28 ps in the LSC per decade. In pseudophakic subjects (19%), mean FLTs only correlated with age in the long spectral channel (r2 = 0.44; p = 0.0002) but not in the short spectral channel (r2 = 0.066; p = 0.2). CONCLUSIONS Fundus autofluorescence lifetimes are age dependent. FLTs in the SSC are more susceptible to lens opacities but less dependent on age changes, whereas FLTs in the LSC are largely independent of the lens status but display a higher degree of age dependency. STUDY REGISTRY ClinicalTrials.gov NCT01981148.
Collapse
Affiliation(s)
- Chantal Dysli
- Department of Ophthalmology, Inselspital, Bern University Hospital and Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Muriel Dysli
- Department of Ophthalmology, Inselspital, Bern University Hospital and Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Sebastian Wolf
- Department of Ophthalmology, Inselspital, Bern University Hospital and Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Martin Zinkernagel
- Department of Ophthalmology, Inselspital, Bern University Hospital and Department of BioMedical Research, University of Bern, Bern, Switzerland
- * E-mail:
| |
Collapse
|
41
|
Kolesnikova M, Oh JK, Wang J, Lee W, Zernant J, Su PY, Kim AH, Jenny LA, Yang T, Allikmets R, Tsang SH. A pathogenic in-frame deletion-insertion variant in BEST1 phenocopies Stargardt disease. JCI Insight 2022; 7:e162687. [PMID: 36264634 PMCID: PMC9746905 DOI: 10.1172/jci.insight.162687] [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: 06/14/2022] [Accepted: 10/18/2022] [Indexed: 01/12/2023] Open
Abstract
Here, we describe affected members of a 2-generation family with a Stargardt disease-like phenotype caused by a 2-base pair deletion insertion, c.1014_1015delGAinsCT;p.(Trp338_Asn339delinsCysTyr), in BEST1. The variant was identified by whole-exome sequencing, and its pathogenicity was verified through chloride channel recording using WT and transfected mutant HEK293 cells. Clinical examination of both patients revealed similar phenotypes at 2 different disease stages that were attributable to differences in their age at presentation. Hyperautofluorescent flecks along the arcades were observed in the proband, while the affected mother exhibited more advanced retinal pigment epithelium (RPE) loss in the central macula. Full-field electroretinogram testing was unremarkable in the daughter; however, moderate attenuation of generalized cone function was detected in the mother. Results from electrooculogram testing in the daughter were consistent with widespread dysfunction of the RPE characteristic of Best disease. Whole-cell patch-clamp recordings revealed a statistically significant decrease in chloride conductance of the mutant compared with WT cells. This report on a mother and daughter with a BEST1 genotype that phenocopies Stargardt disease broadens the clinical spectrum of BEST1-associated retinopathy.
Collapse
Affiliation(s)
- Masha Kolesnikova
- Jonas Children’s Vision Care and Bernard and Shirlee Brown Glaucoma Laboratory, Columbia University, New York, New York, USA
- SUNY Downstate Health Sciences University, New York, New York, USA
| | | | | | - Winston Lee
- Department of Ophthalmology
- Department of Genetics and Development, and
| | | | | | - Angela H. Kim
- Jonas Children’s Vision Care and Bernard and Shirlee Brown Glaucoma Laboratory, Columbia University, New York, New York, USA
- SUNY Downstate Health Sciences University, New York, New York, USA
| | - Laura A. Jenny
- Jonas Children’s Vision Care and Bernard and Shirlee Brown Glaucoma Laboratory, Columbia University, New York, New York, USA
| | | | - Rando Allikmets
- Department of Ophthalmology
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Stephen H. Tsang
- Jonas Children’s Vision Care and Bernard and Shirlee Brown Glaucoma Laboratory, Columbia University, New York, New York, USA
- Department of Ophthalmology
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
- Institute of Human Nutrition, Columbia Stem Cell Initiative, New York, New York, USA
| |
Collapse
|
42
|
Harju N. Regulation of oxidative stress and inflammatory responses in human retinal pigment epithelial cells. Acta Ophthalmol 2022; 100 Suppl 273:3-59. [DOI: 10.1111/aos.15275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Niina Harju
- School of Pharmacy University of Eastern Finland Kuopio Finland
| |
Collapse
|
43
|
Intartaglia D, Giamundo G, Conte I. Autophagy in the retinal pigment epithelium: a new vision and future challenges. FEBS J 2022; 289:7199-7212. [PMID: 33993621 PMCID: PMC9786786 DOI: 10.1111/febs.16018] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/07/2021] [Accepted: 05/12/2021] [Indexed: 01/13/2023]
Abstract
The retinal pigment epithelium (RPE) is a highly specialized monolayer of polarized, pigmented epithelial cells that resides between the vessels of the choriocapillaris and the neural retina. The RPE is essential for the maintenance and survival of overlying light-sensitive photoreceptors, as it participates in the formation of the outer blood-retinal barrier, phagocytosis, degradation of photoreceptor outer segment (POS) tips, maintenance of the retinoid cycle, and protection against light and oxidative stress. Autophagy is an evolutionarily conserved 'self-eating' process, designed to maintain cellular homeostasis. The daily autophagy demands in the RPE require precise gene regulation for the digestion and recycling of intracellular and POS components in lysosomes in response to light and stress conditions. In this review, we discuss selective autophagy and focus on the recent advances in our understanding of the mechanism of cell clearance in the RPE for visual function. Understanding how this catabolic process is regulated by both transcriptional and post-transcriptional mechanisms in the RPE will promote the recognition of pathological pathways in genetic disease and shed light on potential therapeutic strategies to treat visual impairments in patients with retinal disorders associated with lysosomal dysfunction.
Collapse
Affiliation(s)
| | | | - Ivan Conte
- Telethon Institute of Genetics and MedicinePozzuoli (Naples)Italy,Department of BiologyUniversity of Naples Federico IINaplesItaly
| |
Collapse
|
44
|
Feldman T, Ostrovskiy D, Yakovleva M, Dontsov A, Borzenok S, Ostrovsky M. Lipofuscin-Mediated Photic Stress Induces a Dark Toxic Effect on ARPE-19 Cells. Int J Mol Sci 2022; 23:12234. [PMID: 36293088 PMCID: PMC9602730 DOI: 10.3390/ijms232012234] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 12/25/2023] Open
Abstract
Lipofuscin granules from retinal pigment epithelium (RPE) cells contain bisretinoid fluorophores, which are photosensitizers and are phototoxic to cells. In the presence of oxygen, bisretinoids are oxidized to form various products, containing aldehydes and ketones, which are also potentially cytotoxic. In a prior study, we identified that bisretinoid oxidation and degradation products have both hydrophilic and amphiphilic properties, allowing their diffusion through the lipofuscin granule membrane into the RPE cell cytoplasm, and are thiobarbituric acid (TBA)-active. The purpose of the present study was to determine if these products exhibit a toxic effect to the RPE cell also in the absence of light. The experiments were performed using the lipofuscin-fed ARPE-19 cell culture. The RPE cell viability analysis was performed with the use of flow cytofluorimetry and laser scanning confocal microscopy. The results obtained indicated that the cell viability of the lipofuscin-fed ARPE-19 sample was clearly reduced not immediately after visible light irradiation for 18 h, but after 4 days maintaining in the dark. Consequently, we could conclude that bisretinoid oxidation products have a damaging effect on the RPE cell in the dark and can be considered as an aggravating factor in age-related macular degeneration progression.
Collapse
Affiliation(s)
- Tatiana Feldman
- Department of Biology, Lomonosov Moscow State University, Leninskiye Gory 1, 119234 Moscow, Russia
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 4 Kosygin Street, 119334 Moscow, Russia
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia
| | - Dmitriy Ostrovskiy
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia
- Sv. Fyodorov Eye Microsurgery Complex, 59a Beskudnikovsky bld., 127486 Moscow, Russia
| | - Marina Yakovleva
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 4 Kosygin Street, 119334 Moscow, Russia
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia
| | - Alexander Dontsov
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 4 Kosygin Street, 119334 Moscow, Russia
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia
| | - Sergey Borzenok
- Sv. Fyodorov Eye Microsurgery Complex, 59a Beskudnikovsky bld., 127486 Moscow, Russia
| | - Mikhail Ostrovsky
- Department of Biology, Lomonosov Moscow State University, Leninskiye Gory 1, 119234 Moscow, Russia
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 4 Kosygin Street, 119334 Moscow, Russia
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia
| |
Collapse
|
45
|
Landrum JT, Mendez V, Cao Y, Gomez R, Neuringer M. Analysis of macular carotenoids in the developing macaque retina: The timeline of macular pigment development. Methods Enzymol 2022; 674:215-253. [PMID: 36008008 DOI: 10.1016/bs.mie.2022.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In the mature retina, the components of the macular pigment, lutein (L), R,R-zeaxanthin (RRZ), R,S-zeaxanthin (RSZ, meso-zeaxanthin) are most concentrated in the central macula. L and RRZ are of dietary origin but RSZ is produced in situ from L. The relative proportions of L and Z isomers vary across the retina with eccentricity in the adult retina. Early reports have shown that during development, the proportions of L and Z isomers undergo changes as the total pigment levels increase. The methods described here demonstrate the unique utility of chiral phase HPLC to measure the amounts of L, RRZ, and RSZ, discriminating between the two zeaxanthin stereoisomers. In three concentric retinal sections of macaque retinas chiral phase HPLC has been employed to document the developmental changes in the distribution of each L, RSZ, and RRZ during the period just prior to full term gestation through 19 months after birth. The net rate of accumulation of carotenoids within the central retina during the first 20 months is quasi-linear and fit by a linear regression. During development, the rate of transport of L (0.12 (±0.033)ngmm-2mo-1 (SE)) into the central 2mm of the retina is double that of RRZ (0.062 (±0.02)ngmm-2mo-1 (SE)). The rate of accumulation of RSZ (0.06 (±0.01)ngmm-2mo-1 (SE)) is comparable to that of RRZ. In the peripheral retina, the rates of accumulation of L and RRZ are not correlated with increasing age, whereas accumulation of RSZ does correlate with age. The changing proportions of L to Z isomers in the central retina during development are explained by the rates for carotenoid accumulation within the central retina. At birth, the macular pigment in the central retina is dominated by L and RRZ, 0.35±0.11 and 0.21±0.054ngmm-2. In the central retina, RSZ was rarely detected in the youngest tissues analyzed. It can be estimated to represent 6% of the total macular pigment (0.033±0.11ngmm-2) at birth based on extrapolation from measurements in the peripheral retina and the ratio of L/(RRZ+RSZ) is ≈1.5. At maturity, the concentrations for L, RRZ, and RSZ in the central macaque retina are estimated to be 1.7, 1.8 and 1.08ngmm-2, with L/(RRZ+RSZ) being 0.6.
Collapse
Affiliation(s)
- John T Landrum
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL, United States.
| | - Vanesa Mendez
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL, United States
| | - Yisi Cao
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL, United States
| | - Ramon Gomez
- Department of Statistics, Florida International University, Miami, FL, United States
| | - Martha Neuringer
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| |
Collapse
|
46
|
The Role of Medical Image Modalities and AI in the Early Detection, Diagnosis and Grading of Retinal Diseases: A Survey. Bioengineering (Basel) 2022; 9:bioengineering9080366. [PMID: 36004891 PMCID: PMC9405367 DOI: 10.3390/bioengineering9080366] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/28/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022] Open
Abstract
Traditional dilated ophthalmoscopy can reveal diseases, such as age-related macular degeneration (AMD), diabetic retinopathy (DR), diabetic macular edema (DME), retinal tear, epiretinal membrane, macular hole, retinal detachment, retinitis pigmentosa, retinal vein occlusion (RVO), and retinal artery occlusion (RAO). Among these diseases, AMD and DR are the major causes of progressive vision loss, while the latter is recognized as a world-wide epidemic. Advances in retinal imaging have improved the diagnosis and management of DR and AMD. In this review article, we focus on the variable imaging modalities for accurate diagnosis, early detection, and staging of both AMD and DR. In addition, the role of artificial intelligence (AI) in providing automated detection, diagnosis, and staging of these diseases will be surveyed. Furthermore, current works are summarized and discussed. Finally, projected future trends are outlined. The work done on this survey indicates the effective role of AI in the early detection, diagnosis, and staging of DR and/or AMD. In the future, more AI solutions will be presented that hold promise for clinical applications.
Collapse
|
47
|
Feldman TB, Dontsov AE, Yakovleva MA, Ostrovsky MA. Photobiology of lipofuscin granules in the retinal pigment epithelium cells of the eye: norm, pathology, age. Biophys Rev 2022; 14:1051-1065. [PMID: 36124271 PMCID: PMC9481861 DOI: 10.1007/s12551-022-00989-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/26/2022] [Indexed: 01/10/2023] Open
Abstract
Lipofuscin granules (LGs) are accumulated in the retinal pigment epithelium (RPE) cells. The progressive LG accumulation can somehow lead to pathology and accelerate the aging process. The review examines composition, spectral properties and photoactivity of LGs isolated from the human cadaver eyes. By use of atomic force microscopy and near-field microscopy, we have revealed the fluorescent heterogeneity of LGs. We have discovered the generation of reactive oxygen species by LGs, and found that LGs and melanolipofuscin granules are capable of photoinduced oxidation of lipids. It was shown that A2E, as the main fluorophore (bisretinoid) of LGs, is much less active as an oxidation photosensitizer than other fluorophores (bisretinoids) of LGs. Photooxidized products of bisretinoids pose a much greater danger to the cell than non-oxidized one. Our studies of the fluorescent properties of LGs and their fluorophores (bisretinoids) showed for the first time that their spectral characteristics change (shift to the short-wavelength region) in pathology and after exposure to ionizing radiation. By recording the fluorescence spectra and fluorescence decay kinetics of oxidized products of LG fluorophores, it is possible to improve the methods of early diagnosis of degenerative diseases. Lipofuscin ("aging pigment") is not an inert "slag". The photoactivity of LGs can pose a significant danger to the RPE cells. Fluorescence characteristics of LGs are a tool to detect early stages of degeneration in the retina and RPE.
Collapse
Affiliation(s)
- T. B. Feldman
- Department of Biology, Lomonosov Moscow State University, Moscow, Russia
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - A. E. Dontsov
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - M. A. Yakovleva
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - M. A. Ostrovsky
- Department of Biology, Lomonosov Moscow State University, Moscow, Russia
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
48
|
Tong Y, Zhang Z, Wang S. Role of Mitochondria in Retinal Pigment Epithelial Aging and Degeneration. FRONTIERS IN AGING 2022; 3:926627. [PMID: 35912040 PMCID: PMC9337215 DOI: 10.3389/fragi.2022.926627] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/21/2022] [Indexed: 12/17/2022]
Abstract
Retinal pigment epithelial (RPE) cells form a monolayer between the neuroretina and choroid. It has multiple important functions, including acting as outer blood-retina barrier, maintaining the function of neuroretina and photoreceptors, participating in the visual cycle and regulating retinal immune response. Due to high oxidative stress environment, RPE cells are vulnerable to dysfunction, cellular senescence, and cell death, which underlies RPE aging and age-related diseases, including age-related macular degeneration (AMD). Mitochondria are the powerhouse of cells and a major source of cellular reactive oxygen species (ROS) that contribute to mitochondrial DNA damage, cell death, senescence, and age-related diseases. Mitochondria also undergo dynamic changes including fission/fusion, biogenesis and mitophagy for quality control in response to stresses. The role of mitochondria, especially mitochondrial dynamics, in RPE aging and age-related diseases, is still unclear. In this review, we summarize the current understanding of mitochondrial function, biogenesis and especially dynamics such as morphological changes and mitophagy in RPE aging and age-related RPE diseases, as well as in the biological processes of RPE cellular senescence and cell death. We also discuss the current preclinical and clinical research efforts to prevent or treat RPE degeneration by restoring mitochondrial function and dynamics.
Collapse
Affiliation(s)
- Yao Tong
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
| | - Zunyi Zhang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
| | - Shusheng Wang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
- Department of Ophthalmology, Tulane University, New Orleans, LA, United States
- Tulane Personalized Health Institute, Tulane University, New Orleans, LA, United States
| |
Collapse
|
49
|
Burgos-Fernández FJ, Alterini T, Díaz-Doutón F, González L, Mateo C, Mestre C, Pujol J, Vilaseca M. Reflectance evaluation of eye fundus structures with a visible and near-infrared multispectral camera. BIOMEDICAL OPTICS EXPRESS 2022; 13:3504-3519. [PMID: 35781951 PMCID: PMC9208594 DOI: 10.1364/boe.457412] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/12/2022] [Accepted: 04/30/2022] [Indexed: 06/15/2023]
Abstract
We examined the spectral reflectance of fundus structures in the visible and near-infrared (400-1300 nm) range for contributing to the medical diagnosis of fundus diseases. Spectral images of healthy eye fundus and other ocular diseases were acquired using a novel multispectral fundus camera. Reflectance metrics were computed based on contrast to analyze the spectral features. Significant differences were observed among the structures in healthy and diseased eye fundus. Specifically, near-infrared analysis allows imaging of deeper layers, such as the choroid, which, to date, has not been retrieved using traditional color fundus cameras. Pathological structures, which were hardly observable in color fundus images owing to metamerism, were also revealed by the developed multispectral fundus camera.
Collapse
Affiliation(s)
- Francisco J. Burgos-Fernández
- Center for Sensors, Instruments and Systems Development, Universitat Politècnica de Catalunya, Rambla Sant Nebridi 10, Terrassa, 08222, Spain
| | - Tommaso Alterini
- Center for Sensors, Instruments and Systems Development, Universitat Politècnica de Catalunya, Rambla Sant Nebridi 10, Terrassa, 08222, Spain
| | - Fernando Díaz-Doutón
- Center for Sensors, Instruments and Systems Development, Universitat Politècnica de Catalunya, Rambla Sant Nebridi 10, Terrassa, 08222, Spain
| | - Laura González
- Instituto de Microcirugía Ocular (Miranza Group), Josep Maria Lladó Street 3, Barcelona, 08035, Spain
| | - Carlos Mateo
- Instituto de Microcirugía Ocular (Miranza Group), Josep Maria Lladó Street 3, Barcelona, 08035, Spain
| | - Clara Mestre
- Indiana University School of Optometry, 800 Atwater Ave, Bloomington, IN 47405, USA
| | - Jaume Pujol
- Center for Sensors, Instruments and Systems Development, Universitat Politècnica de Catalunya, Rambla Sant Nebridi 10, Terrassa, 08222, Spain
| | - Meritxell Vilaseca
- Center for Sensors, Instruments and Systems Development, Universitat Politècnica de Catalunya, Rambla Sant Nebridi 10, Terrassa, 08222, Spain
| |
Collapse
|
50
|
Xu Y, Li D, Su G, Cai S. The effect of A2E on lysosome membrane permeability during blue light-induced human RPEs apoptosis. BMC Ophthalmol 2022; 22:241. [PMID: 35641967 PMCID: PMC9158258 DOI: 10.1186/s12886-022-02464-1] [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: 11/05/2021] [Accepted: 05/24/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To investigate the effect of N-retinyl-N-retinylidene ethanolamine (A2E) on lysosome membrane permeability (LMP) during blue light-induced human retinal pigment epithelium cells (RPEs) apoptosis. METHODS By building an A2E and blue light irradiation inducing RPEs damage model, the CCK-8 assay was used to detect RPEs viability loaded with different concentrations of A2E after different culturing time to determine the optimum A2E loading concentration. And the RPEs fluorescence intensity changes were observed by fluorescence microscopy loaded with different concentration of A2E. The RPEs were divided into four groups randomly: control group, A2E-loaded group, blue light irradiation group, and A2E-loaded + blue light irradiation group. Annexin V-FITC/PI and TUNEL/DAPI methods were used to detect RPEs apoptotic rate. Laser scanning confocal microscopy (LSCM) was used to observe RPEs LMP changes stained by acridine orange (AO) method. RESULTS The CCK-8 result showed a downward trend in cells viability of RPEs loaded with increasing concentration of A2E and extending culturing time. The optimum A2E loading concentration was determined at 25 μmol/L. With increasing A2E loading concentrations, the intensity of fluorescence in RPEs decreased gradually. The RPEs apoptotic rate in blue light irradiation + A2E-loaded group was significantly higher than those in other three groups detected by Annexin V-FITC/PI method, which was similar to TUNEL/DAPI's result. After AO staining, cytoplasmic and nucleolar RNAs emits green fluorescence; lysosomes emit red fluorescence. Through the interference of A2E and blue light on RPEs, red fluorescent leakage from the lysosomes (means LMP increasing) can be observed. The mean red fluorescence intensity was chosen as the statistics indicator to estimate LMP change in RPEs cultured in vitro. Compared with the control group, the red fluorescence intensity decreased in A2E-loaded group, blue light irradiation group, and blue light irradiation + A2E-loaded group. Meanwhile, the mean red fluorescence intensity in blue light irradiation + A2E-loaded group was the lowest. CONCLUSIONS Both A2E-loaded and blue light irradiation could induce human RPEs apoptosis, and the two factors had a synergistic effect. In addition, both A2E and blue light can lead to LMP increasing, which indicated LMP change might be the upstream part in inducing mitochondrion-dependent apoptotic pathway. These data provided evidence that A2E as the most important auto-fluorescence substance in lipofuscin is an initiator of blue light-mediated damage of RPEs and participate in pathogenesis of retinal degenerative diseases in humans.
Collapse
Affiliation(s)
- Yan Xu
- Department of Ophthalmology, Affiliated Hospital of Zunyi Medical University, No.149 Dalian Road, Zunyi, 563000, Guizhou, China
- Department of Ophthalmology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo No.2 Hospital, Ningbo, Zhejiang, China
| | - Dan Li
- Department of Ophthalmology, Affiliated Hospital of Zunyi Medical University, No.149 Dalian Road, Zunyi, 563000, Guizhou, China
- Department of Ophthalmology, The People's Hospital of Longchang, Neijiang, Sichuan, China
| | - Gang Su
- Department of Ophthalmology, Affiliated Hospital of Zunyi Medical University, No.149 Dalian Road, Zunyi, 563000, Guizhou, China
- Guizhou Eye Hospital, Zunyi, Guizhou, China
- Guizhou Provincial Branch of National Eye Disease Clinical Medicine Research Center, Zunyi, Guizhou, China
- Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, China
| | - Shanjun Cai
- Department of Ophthalmology, Affiliated Hospital of Zunyi Medical University, No.149 Dalian Road, Zunyi, 563000, Guizhou, China.
- Guizhou Eye Hospital, Zunyi, Guizhou, China.
- Guizhou Provincial Branch of National Eye Disease Clinical Medicine Research Center, Zunyi, Guizhou, China.
- Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|