1
|
Rinaldi M, Pezone A, Quadrini GI, Abbadessa G, Laezza MP, Passaro ML, Porcellini A, Costagliola C. Targeting shared pathways in tauopathies and age-related macular degeneration: implications for novel therapies. Front Aging Neurosci 2024; 16:1371745. [PMID: 38633983 PMCID: PMC11021713 DOI: 10.3389/fnagi.2024.1371745] [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: 01/16/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
The intricate parallels in structure and function between the human retina and the central nervous system designate the retina as a prospective avenue for understanding brain-related processes. This review extensively explores the shared physiopathological mechanisms connecting age-related macular degeneration (AMD) and proteinopathies, with a specific focus on tauopathies. The pivotal involvement of oxidative stress and cellular senescence emerges as key drivers of pathogenesis in both conditions. Uncovering these shared elements not only has the potential to enhance our understanding of intricate neurodegenerative diseases but also sets the stage for pioneering therapeutic approaches in AMD.
Collapse
Affiliation(s)
- Michele Rinaldi
- Department of Neurosciences, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - Antonio Pezone
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Gaia Italia Quadrini
- Department of Neurosciences, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - Gianmarco Abbadessa
- Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Maria Paola Laezza
- Department of Neurosciences, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| | - Maria Laura Passaro
- Department of Neurosciences, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| | | | - Ciro Costagliola
- Department of Neurosciences, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| |
Collapse
|
2
|
Miller RD, Ratnayaka JA. Impaired lysosomes in the retinal pigment epithelium play a central role in the degeneration of the neuroretina. Neural Regen Res 2023; 18:2697-2698. [PMID: 37449628 DOI: 10.4103/1673-5374.373713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Affiliation(s)
- Rebecca D Miller
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - J Arjuna Ratnayaka
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
3
|
García-Bermúdez MY, Vohra R, Freude K, van Wijngaarden P, Martin K, Thomsen MS, Aldana BI, Kolko M. Potential Retinal Biomarkers in Alzheimer's Disease. Int J Mol Sci 2023; 24:15834. [PMID: 37958816 PMCID: PMC10649108 DOI: 10.3390/ijms242115834] [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: 09/01/2023] [Revised: 10/18/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Alzheimer's disease (AD) represents a major diagnostic challenge, as early detection is crucial for effective intervention. This review examines the diagnostic challenges facing current AD evaluations and explores the emerging field of retinal alterations as early indicators. Recognizing the potential of the retina as a noninvasive window to the brain, we emphasize the importance of identifying retinal biomarkers in the early stages of AD. However, the examination of AD is not without its challenges, as the similarities shared with other retinal diseases introduce complexity in the search for AD-specific markers. In this review, we address the relevance of using the retina for the early diagnosis of AD and the complex challenges associated with the search for AD-specific retinal biomarkers. We provide a comprehensive overview of the current landscape and highlight avenues for progress in AD diagnosis by retinal examination.
Collapse
Affiliation(s)
| | - Rupali Vohra
- Eye Translational Research Unit, Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark
- Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, 2600 Glostrup, Denmark
| | - Kristine Freude
- Group of Stem Cell Models and Embryology, Department of Veterinary and Animal Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark
| | - Peter van Wijngaarden
- Center for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Keith Martin
- Center for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Maj Schneider Thomsen
- Neurobiology Research and Drug Delivery, Department of Health, Science and Technology, Aalborg University, 9220 Aalborg, Denmark
| | - Blanca Irene Aldana
- Neurometabolism Research Group, Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Miriam Kolko
- Eye Translational Research Unit, Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark
- Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, 2600 Glostrup, Denmark
| |
Collapse
|
4
|
Li X, Ma B, Zhang W, Song Z, Zhang X, Liao M, Li X, Zhao X, Du M, Yu J, He S, Yan H. The essential role of N6-methyladenosine RNA methylation in complex eye diseases. Genes Dis 2023; 10:505-520. [PMID: 37223523 PMCID: PMC10201676 DOI: 10.1016/j.gendis.2022.05.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/29/2022] [Accepted: 05/08/2022] [Indexed: 11/20/2022] Open
Abstract
There are many complex eye diseases which are the leading causes of blindness, however, the pathogenesis of the complex eye diseases is not fully understood, especially the underlying molecular mechanisms of N6-methyladenosine (m6A) RNA methylation in the eye diseases have not been extensive clarified. Our review summarizes the latest advances in the studies of m6A modification in the pathogenesis of the complex eye diseases, including cornea disease, cataract, diabetic retinopathy, age-related macular degeneration, proliferative vitreoretinopathy, Graves' disease, uveal melanoma, retinoblastoma, and traumatic optic neuropathy. We further discuss the possibility of developing m6A modification signatures as biomarkers for the diagnosis of the eye diseases, as well as potential therapeutic approaches.
Collapse
Affiliation(s)
- Xiaohua Li
- Henan Provincial People’s Hospital, Henan Eye Hospital, Henan Eye Institute, Henan Key Laboratory of Ophthalmology and Visual Science, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, Henan 450003, China
| | - Binyun Ma
- Department of Medicine/Hematology, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Wenfang Zhang
- Department of Ophthalmology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Zongming Song
- Henan Provincial People’s Hospital, Henan Eye Hospital, Henan Eye Institute, Henan Key Laboratory of Ophthalmology and Visual Science, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, Henan 450003, China
| | - Xiaodan Zhang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Laboratory of Molecular Ophthalmology, Tianjin Medical University. Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300052, China
| | - Mengyu Liao
- Department of Ophthalmology, Tianjin Medical University General Hospital, Laboratory of Molecular Ophthalmology, Tianjin Medical University. Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300052, China
| | - Xue Li
- Henan Provincial People’s Hospital, Henan Eye Hospital, Henan Eye Institute, Henan Key Laboratory of Ophthalmology and Visual Science, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, Henan 450003, China
| | - Xueru Zhao
- Henan Provincial People’s Hospital, Henan Eye Hospital, Henan Eye Institute, Henan Key Laboratory of Ophthalmology and Visual Science, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, Henan 450003, China
| | - Mei Du
- Department of Ophthalmology, Tianjin Medical University General Hospital, Laboratory of Molecular Ophthalmology, Tianjin Medical University. Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300052, China
| | - Jinguo Yu
- Department of Ophthalmology, Tianjin Medical University General Hospital, Laboratory of Molecular Ophthalmology, Tianjin Medical University. Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300052, China
| | - Shikun He
- Henan Provincial People’s Hospital, Henan Eye Hospital, Henan Eye Institute, Henan Key Laboratory of Ophthalmology and Visual Science, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, Henan 450003, China
- Department of Ophthalmology, Tianjin Medical University General Hospital, Laboratory of Molecular Ophthalmology, Tianjin Medical University. Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300052, China
- Department of Pathology and Ophthalmology, USC Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Hua Yan
- Department of Ophthalmology, Tianjin Medical University General Hospital, Laboratory of Molecular Ophthalmology, Tianjin Medical University. Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300052, China
| |
Collapse
|
5
|
Targeting immunoproteasome in neurodegeneration: A glance to the future. Pharmacol Ther 2023; 241:108329. [PMID: 36526014 DOI: 10.1016/j.pharmthera.2022.108329] [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: 10/03/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
The immunoproteasome is a specialized form of proteasome equipped with modified catalytic subunits that was initially discovered to play a pivotal role in MHC class I antigen processing and immune system modulation. However, over the last years, this proteolytic complex has been uncovered to serve additional functions unrelated to antigen presentation. Accordingly, it has been proposed that immunoproteasome synergizes with canonical proteasome in different cell types of the nervous system, regulating neurotransmission, metabolic pathways and adaptation of the cells to redox or inflammatory insults. Hence, studying the alterations of immunoproteasome expression and activity is gaining research interest to define the dynamics of neuroinflammation as well as the early and late molecular events that are likely involved in the pathogenesis of a variety of neurological disorders. Furthermore, these novel functions foster the perspective of immunoproteasome as a potential therapeutic target for neurodegeneration. In this review, we provide a brain and retina-wide overview, trying to correlate present knowledge on structure-function relationships of immunoproteasome with the variety of observed neuro-modulatory functions.
Collapse
|
6
|
Kurzawa‐Akanbi M, Whitfield P, Burté F, Bertelli PM, Pathak V, Doherty M, Hilgen B, Gliaudelytė L, Platt M, Queen R, Coxhead J, Porter A, Öberg M, Fabrikova D, Davey T, Beh CS, Georgiou M, Collin J, Boczonadi V, Härtlova A, Taggart M, Al‐Aama J, Korolchuk VI, Morris CM, Guduric‐Fuchs J, Steel DH, Medina RJ, Armstrong L, Lako M. Retinal pigment epithelium extracellular vesicles are potent inducers of age-related macular degeneration disease phenotype in the outer retina. J Extracell Vesicles 2022; 11:e12295. [PMID: 36544284 PMCID: PMC9772497 DOI: 10.1002/jev2.12295] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 11/18/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of blindness. Vision loss is caused by the retinal pigment epithelium (RPE) and photoreceptors atrophy and/or retinal and choroidal angiogenesis. Here we use AMD patient-specific RPE cells with the Complement Factor H Y402H high-risk polymorphism to perform a comprehensive analysis of extracellular vesicles (EVs), their cargo and role in disease pathology. We show that AMD RPE is characterised by enhanced polarised EV secretion. Multi-omics analyses demonstrate that AMD RPE EVs carry RNA, proteins and lipids, which mediate key AMD features including oxidative stress, cytoskeletal dysfunction, angiogenesis and drusen accumulation. Moreover, AMD RPE EVs induce amyloid fibril formation, revealing their role in drusen formation. We demonstrate that exposure of control RPE to AMD RPE apical EVs leads to the acquisition of AMD features such as stress vacuoles, cytoskeletal destabilization and abnormalities in the morphology of the nucleus. Retinal organoid treatment with apical AMD RPE EVs leads to disrupted neuroepithelium and the appearance of cytoprotective alpha B crystallin immunopositive cells, with some co-expressing retinal progenitor cell markers Pax6/Vsx2, suggesting injury-induced regenerative pathways activation. These findings indicate that AMD RPE EVs are potent inducers of AMD phenotype in the neighbouring RPE and retinal cells.
Collapse
Affiliation(s)
- Marzena Kurzawa‐Akanbi
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Phillip Whitfield
- Glasgow Polyomics and Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Florence Burté
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Pietro Maria Bertelli
- The Welcome‐Wolfson Institute for Experimental MedicineQueen's University BelfastBelfastUK
| | - Varun Pathak
- The Welcome‐Wolfson Institute for Experimental MedicineQueen's University BelfastBelfastUK
| | - Mary Doherty
- Lipidomics Research FacilityUniversity of the Highlands and IslandsInvernessUK
| | - Birthe Hilgen
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Lina Gliaudelytė
- Translational and Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUK
| | | | - Rachel Queen
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Jonathan Coxhead
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Andrew Porter
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Maria Öberg
- Institute of Biomedicine, Department of Microbiology and Immunology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Wallenberg Center for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
| | - Daniela Fabrikova
- Institute of Biomedicine, Department of Microbiology and Immunology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Wallenberg Center for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
| | - Tracey Davey
- Electron Microscopy Research ServicesNewcastle UniversityNewcastle upon TyneUK
| | - Chia Shyan Beh
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Maria Georgiou
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Joseph Collin
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Veronika Boczonadi
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Anetta Härtlova
- Institute of Biomedicine, Department of Microbiology and Immunology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Wallenberg Center for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- The Institute of Medical Microbiology and HygieneUniversity Medical Center Freiburg (Universitätklinikum Freiburg)FreiburgGermany
| | - Michael Taggart
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Jumana Al‐Aama
- Faculty of MedicineKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Viktor I Korolchuk
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Christopher M Morris
- Translational and Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Jasenka Guduric‐Fuchs
- The Welcome‐Wolfson Institute for Experimental MedicineQueen's University BelfastBelfastUK
| | - David H Steel
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Reinhold J Medina
- The Welcome‐Wolfson Institute for Experimental MedicineQueen's University BelfastBelfastUK
| | - Lyle Armstrong
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Majlinda Lako
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| |
Collapse
|
7
|
Lynn SA, Soubigou F, Dewing JM, Smith A, Ballingall J, Sass T, Nica I, Watkins C, Gupta B, Almuhtaseb H, Lash SC, Yuen HM, Cree A, Newman TA, Lotery AJ, Ratnayaka JA. An Exploratory Study Provides Insights into MMP9 and Aβ Levels in the Vitreous and Blood across Different Ages and in a Subset of AMD Patients. Int J Mol Sci 2022; 23:14603. [PMID: 36498929 PMCID: PMC9736887 DOI: 10.3390/ijms232314603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/15/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
Matrix metalloproteinase-9 (MMP9) and total amyloid-beta (Aβ) are prospective biomarkers of ocular ageing and retinopathy. These were quantified by ELISA in the vitreous and blood from controls (n = 55) and in a subset of age-related macular degeneration (AMD) patients (n = 12) for insights and possible additional links between the ocular and systemic compartments. Vitreous MMP9 levels in control and AMD groups were 932.5 ± 240.9 pg/mL and 813.7 ± 157.6 pg/mL, whilst serum levels were 2228 ± 193 pg/mL and 2386.8 ± 449.4 pg/mL, respectively. Vitreous Aβ in control and AMD groups were 1173.5 ± 117.1 pg/mL and 1275.6 ± 332.9 pg/mL, whilst plasma Aβ were 574.3 ± 104.8 pg/mL and 542.2 ± 139.9 pg/mL, respectively. MMP9 and Aβ showed variable levels across the lifecourse, indicating no correlation to each other or with age nor AMD status, though the smaller AMD cohort was a limiting factor. Aβ and MMP9 levels in the vitreous and blood were unrelated to mean arterial pressure. Smoking, another modifiable risk, showed no association with vitreous Aβ. However, smoking may be linked with vitreous (p = 0.004) and serum (p = 0.005) MMP9 levels in control and AMD groups, though this did not reach our elevated (p = 0.001) significance. A bioinformatics analysis revealed promising MMP9 and APP/Aβ partners for further scrutiny, many of which are already linked with retinopathy.
Collapse
Affiliation(s)
- Savannah A. Lynn
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton SO16 6YD, UK
| | - Flavie Soubigou
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton SO16 6YD, UK
| | - Jennifer M. Dewing
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton SO16 6YD, UK
| | - Amanda Smith
- Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Joanna Ballingall
- Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Thea Sass
- Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Isabela Nica
- Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Catrin Watkins
- Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Bhaskar Gupta
- Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Hussein Almuhtaseb
- Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Stephen C. Lash
- Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Ho Ming Yuen
- Primary Care, Population Sciences and Medical Education, Faculty of Medicine, University of Southampton, MP 801, Tremona Road, Southampton SO16 6YD, UK
| | - Angela Cree
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton SO16 6YD, UK
| | - Tracey A. Newman
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton SO16 6YD, UK
| | - Andrew J. Lotery
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton SO16 6YD, UK
- Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - J. Arjuna Ratnayaka
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton SO16 6YD, UK
| |
Collapse
|
8
|
Elsayed MEAA, Kaukonen M, Kiraly P, Kapetanovic JC, MacLaren RE. Potential CRISPR Base Editing Therapeutic Options in a Sorsby Fundus Dystrophy Patient. Genes (Basel) 2022; 13:2103. [PMID: 36421778 PMCID: PMC9690532 DOI: 10.3390/genes13112103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 07/30/2023] Open
Abstract
TIMP3 mutations are associated with early-onset macular choroidal neovascularisation for which no treatment currently exists. CRISPR base editing, with its ability to irreversibly correct point mutations by chemical modification of nucleobases at DNA level, may be a therapeutic option. We report a bioinformatic analysis of potential therapeutic options in a patient presenting with Sorsby fundus dystrophy. Genetic testing in a 35-year-old gentleman with bilateral macular choroidal neovascularisation revealed the patient to be heterozygous for a TIMP3 variant c.610A>T, p.(Ser204Cys). Using a glycosylase base editor (GBE), another DNA-edit could be introduced that would revert the variant back to wild-type on amino acid level. Alternatively, the mutated residue could be changed to another amino acid that would be better tolerated, and for that, an available 'NG'-PAM site was found to be available for the SpCas9-based adenine base editor (ABE) that would introduce p.(Ser204Arg). In silico analyses predicted this variant to be non-pathogenic; however, a bystander edit, p.Ile205Thr, would be introduced. This case report highlights the importance of considering genetic testing in young patients with choroidal neovascularisation, particularly within the context of a strong family history of presumed wet age-related macular degeneration, and describes potential therapeutic options.
Collapse
Affiliation(s)
| | - Maria Kaukonen
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX3 9DU, UK
| | - Peter Kiraly
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Jasmina Cehajic Kapetanovic
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX3 9DU, UK
| | - Robert E. MacLaren
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
9
|
Blasiak J, Sobczuk P, Pawlowska E, Kaarniranta K. Interplay between aging and other factors of the pathogenesis of age-related macular degeneration. Ageing Res Rev 2022; 81:101735. [PMID: 36113764 DOI: 10.1016/j.arr.2022.101735] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/03/2022] [Accepted: 09/12/2022] [Indexed: 01/31/2023]
Abstract
Age-related macular degeneration (AMD) is a complex eye disease with the retina as the target tissue and aging as per definition the most serious risk factor. However, the retina contains over 60 kinds of cells that form different structures, including the neuroretina and retinal pigment epithelium (RPE) which can age at different rates. Other established or putative AMD risk factors can differentially affect the neuroretina and RPE and can differently interplay with aging of these structures. The occurrence of β-amyloid plaques and increased levels of cholesterol in AMD retinas suggest that AMD may be a syndrome of accelerated brain aging. Therefore, the question about the real meaning of age in AMD is justified. In this review we present and update information on how aging may interplay with some aspects of AMD pathogenesis, such as oxidative stress, amyloid beta formation, circadian rhythm, metabolic aging and cellular senescence. Also, we show how this interplay can be specific for photoreceptors, microglia cells and RPE cells as well as in Bruch's membrane and the choroid. Therefore, the process of aging may differentially affect different retinal structures. As an accurate quantification of biological aging is important for risk stratification and early intervention for age-related diseases, the determination how photoreceptors, microglial and RPE cells age in AMD may be helpful for a precise diagnosis and treatment of this largely untreatable disease.
Collapse
Affiliation(s)
- Janusz Blasiak
- Department of Molecular Genetics, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland.
| | - Piotr Sobczuk
- Emergency Medicine and Disaster Medicine Department, Medical University of Lodz, Pomorska 251, 92-209 Lodz, Poland; Department of Orthopaedics and Traumatology, Polish Mothers' Memorial Hospital - Research Institute, Rzgowska 281, 93-338 Lodz, Poland
| | - Elzbieta Pawlowska
- Department of Pediatric Dentistry, Medical University of Lodz, Pomorska 251, 92-216 Lodz, Poland
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland; Department of Ophthalmology, Kuopio University Hospital, KYS, P.O. Box 100, FI-70029 Finland
| |
Collapse
|
10
|
Gonzalez VH, Berger B, Goldberg R, Gordon CM, Khurana RN, Angeles R, Shams N. Safety and Tolerability of Intravitreal Carotuximab (DE-122) in Patients With Persistent Exudative Age-Related Macular Degeneration: A Phase I Study. Transl Vis Sci Technol 2021; 10:27. [PMID: 34935908 PMCID: PMC8711001 DOI: 10.1167/tvst.10.14.27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Carotuximab (DE-122) is a novel endoglin antibody that exhibits potent anti-angiogenic activity. The aim of this study was to evaluate the safety and tolerability of a single intravitreal injection of four ascending doses of carotuximab in patients with persistent exudative age-related macular degeneration (AMD). Methods In an open-label, dose-escalating, sequential cohort study, patients with persistent exudative AMD were assigned to an intravitreal injection of carotuximab 0.5 mg, 1.0 mg, 2.0 mg, or 4.0 mg (n = 3 per group). Safety and change in central subfield thickness (CST), as measured by spectral domain–optical coherence tomography, were assessed from baseline until day 90. Rescue therapy with an anti-vascular endothelial growth factor medication was allowed on days 8, 30, and 60. Results Seven patients (58%) experienced at least one adverse event (AE), including five patients (41.7%) who experienced one or more AEs in the study eye and two patients (16.7%) who experienced one or more non-ocular AEs. Posterior eye deposits were reported in one patient 2 days after receiving 1.0 mg, but they resolved spontaneously by day 43. A >50-µm reduction in CST on two consecutive visits was observed in four patients (33%), including one patient in each dose cohort. Conclusions In this study, carotuximab was generally well tolerated, with no serious AEs reported, when administered as a single intravitreal injection to patients with persistent exudative AMD. Translational Relevance Further characterization of the safety and efficacy of carotuximab will be needed to determine what role it may have in the treatment of exudative AMD.
Collapse
Affiliation(s)
| | | | | | | | - Rahul N Khurana
- Northern California Retina Vitreous Associates, Mountain View, CA, USA.,Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | | | | |
Collapse
|
11
|
Hu ML, Quinn J, Xue K. Interactions between Apolipoprotein E Metabolism and Retinal Inflammation in Age-Related Macular Degeneration. Life (Basel) 2021; 11:life11070635. [PMID: 34210002 PMCID: PMC8305051 DOI: 10.3390/life11070635] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/24/2021] [Accepted: 06/27/2021] [Indexed: 02/07/2023] Open
Abstract
Age-related macular degeneration (AMD) is a multifactorial retinal disorder that is a major global cause of severe visual impairment. The development of an effective therapy to treat geographic atrophy, the predominant form of AMD, remains elusive due to the incomplete understanding of its pathogenesis. Central to AMD diagnosis and pathology are the hallmark lipid and proteinaceous deposits, drusen and reticular pseudodrusen, that accumulate in the subretinal pigment epithelium and subretinal spaces, respectively. Age-related changes and environmental stressors, such as smoking and a high-fat diet, are believed to interact with the many genetic risk variants that have been identified in several major biochemical pathways, including lipoprotein metabolism and the complement system. The APOE gene, encoding apolipoprotein E (APOE), is a major genetic risk factor for AMD, with the APOE2 allele conferring increased risk and APOE4 conferring reduced risk, in comparison to the wildtype APOE3. Paradoxically, APOE4 is the main genetic risk factor in Alzheimer’s disease, a disease with features of neuroinflammation and amyloid-beta deposition in common with AMD. The potential interactions of APOE with the complement system and amyloid-beta are discussed here to shed light on their roles in AMD pathogenesis, including in drusen biogenesis, immune cell activation and recruitment, and retinal inflammation.
Collapse
Affiliation(s)
- Monica L. Hu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia;
| | - Joel Quinn
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK;
| | - Kanmin Xue
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK;
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
- Correspondence:
| |
Collapse
|
12
|
Cytoprotective Potential of Fucoxanthin in Oxidative Stress-Induced Age-Related Macular Degeneration and Retinal Pigment Epithelial Cell Senescence In Vivo and In Vitro. Mar Drugs 2021; 19:md19020114. [PMID: 33670685 PMCID: PMC7923087 DOI: 10.3390/md19020114] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/10/2021] [Accepted: 02/15/2021] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress is identified as a major inducer of retinal pigment epithelium (RPE) cell dysregulation and is associated with age-related macular degeneration (AMD). The protection of RPE disorders plays an essential role in the pathological progress of retinal degeneration diseases. The pharmacological functions of fucoxanthin, a characteristic carotenoid, including anti-inflammatory and antioxidant properties, may ameliorate an outstanding bioactivity against premature senescence and cellular dysfunction. This study demonstrates that fucoxanthin protects RPE cells from oxidative stress-induced premature senescence and decreased photoreceptor cell loss in a sodium iodate-induced AMD animal model. Similarly, oxidative stress induced by hydrogen peroxide, nuclear phosphorylated histone (γH2AX) deposition and premature senescence-associated β-galactosidase staining were inhibited by fucoxanthin pretreatment in a human RPE cell line, ARPE-19 cells. Results reveal that fucoxanthin treatment significantly inhibited reactive oxygen species (ROS) generation, reduced malondialdehyde (MDA) concentrations and increased the mitochondrial metabolic rate in oxidative stress-induced RPE cell damage. Moreover, atrophy of apical microvilli was inhibited in cells treated with fucoxanthin after oxidative stress. During aging, the RPE undergoes well-characterized pathological changes, including amyloid beta (Aβ) deposition, beta-site amyloid precursor protein-cleaving enzyme 1 (BACE1) expression and tight junction disruption, which were also reduced in fucoxanthin-treated groups by immunofluorescence. Altogether, pretreatment with fucoxanthin may protect against premature senescence and cellular dysfunction in retinal cells by oxidative stress in experimental AMD animal and human RPE cell models.
Collapse
|
13
|
Lynn SA, Johnston DA, Scott JA, Munday R, Desai RS, Keeling E, Weaterton R, Simpson A, Davis D, Freeman T, Chatelet DS, Page A, Cree AJ, Lee H, Newman TA, Lotery AJ, Ratnayaka JA. Oligomeric Aβ 1-42 Induces an AMD-Like Phenotype and Accumulates in Lysosomes to Impair RPE Function. Cells 2021; 10:413. [PMID: 33671133 PMCID: PMC7922851 DOI: 10.3390/cells10020413] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 02/04/2021] [Accepted: 02/11/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease-associated amyloid beta (Aβ) proteins accumulate in the outer retina with increasing age and in eyes of age-related macular degeneration (AMD) patients. To study Aβ-induced retinopathy, wild-type mice were injected with nanomolar human oligomeric Aβ1-42, which recapitulate the Aβ burden reported in human donor eyes. In vitro studies investigated the cellular effects of Aβ in endothelial and retinal pigment epithelial (RPE) cells. Results show subretinal Aβ-induced focal AMD-like pathology within 2 weeks. Aβ exposure caused endothelial cell migration, and morphological and barrier alterations to the RPE. Aβ co-localized to late-endocytic compartments of RPE cells, which persisted despite attempts to clear it through upregulation of lysosomal cathepsin B, revealing a novel mechanism of lysosomal impairment in retinal degeneration. The rapid upregulation of cathepsin B was out of step with the prolonged accumulation of Aβ within lysosomes, and contrasted with enzymatic responses to internalized photoreceptor outer segments (POS). Furthermore, RPE cells exposed to Aβ were identified as deficient in cargo-carrying lysosomes at time points that are critical to POS degradation. These findings imply that Aβ accumulation within late-endocytic compartments, as well as lysosomal deficiency, impairs RPE function over time, contributing to visual defects seen in aging and AMD eyes.
Collapse
Affiliation(s)
- Savannah A. Lynn
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP 806, Tremona Road, Southampton SO16 6YD, UK; (S.A.L.); (J.A.S.); (R.M.); (R.S.D.); (E.K.); (R.W.); (A.S.); (D.D.); (T.F.); (A.J.C.); (H.L.); (T.A.N.); (A.J.L.)
| | - David A. Johnston
- Biomedical Imaging Unit, University of Southampton, MP12, Tremona Road, Southampton SO16 6YD, UK; (D.A.J.); (D.S.C.); (A.P.)
| | - Jenny A. Scott
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP 806, Tremona Road, Southampton SO16 6YD, UK; (S.A.L.); (J.A.S.); (R.M.); (R.S.D.); (E.K.); (R.W.); (A.S.); (D.D.); (T.F.); (A.J.C.); (H.L.); (T.A.N.); (A.J.L.)
| | - Rosie Munday
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP 806, Tremona Road, Southampton SO16 6YD, UK; (S.A.L.); (J.A.S.); (R.M.); (R.S.D.); (E.K.); (R.W.); (A.S.); (D.D.); (T.F.); (A.J.C.); (H.L.); (T.A.N.); (A.J.L.)
| | - Roshni S. Desai
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP 806, Tremona Road, Southampton SO16 6YD, UK; (S.A.L.); (J.A.S.); (R.M.); (R.S.D.); (E.K.); (R.W.); (A.S.); (D.D.); (T.F.); (A.J.C.); (H.L.); (T.A.N.); (A.J.L.)
| | - Eloise Keeling
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP 806, Tremona Road, Southampton SO16 6YD, UK; (S.A.L.); (J.A.S.); (R.M.); (R.S.D.); (E.K.); (R.W.); (A.S.); (D.D.); (T.F.); (A.J.C.); (H.L.); (T.A.N.); (A.J.L.)
| | - Ruaridh Weaterton
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP 806, Tremona Road, Southampton SO16 6YD, UK; (S.A.L.); (J.A.S.); (R.M.); (R.S.D.); (E.K.); (R.W.); (A.S.); (D.D.); (T.F.); (A.J.C.); (H.L.); (T.A.N.); (A.J.L.)
| | - Alexander Simpson
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP 806, Tremona Road, Southampton SO16 6YD, UK; (S.A.L.); (J.A.S.); (R.M.); (R.S.D.); (E.K.); (R.W.); (A.S.); (D.D.); (T.F.); (A.J.C.); (H.L.); (T.A.N.); (A.J.L.)
| | - Dillon Davis
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP 806, Tremona Road, Southampton SO16 6YD, UK; (S.A.L.); (J.A.S.); (R.M.); (R.S.D.); (E.K.); (R.W.); (A.S.); (D.D.); (T.F.); (A.J.C.); (H.L.); (T.A.N.); (A.J.L.)
| | - Thomas Freeman
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP 806, Tremona Road, Southampton SO16 6YD, UK; (S.A.L.); (J.A.S.); (R.M.); (R.S.D.); (E.K.); (R.W.); (A.S.); (D.D.); (T.F.); (A.J.C.); (H.L.); (T.A.N.); (A.J.L.)
| | - David S. Chatelet
- Biomedical Imaging Unit, University of Southampton, MP12, Tremona Road, Southampton SO16 6YD, UK; (D.A.J.); (D.S.C.); (A.P.)
| | - Anton Page
- Biomedical Imaging Unit, University of Southampton, MP12, Tremona Road, Southampton SO16 6YD, UK; (D.A.J.); (D.S.C.); (A.P.)
| | - Angela J. Cree
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP 806, Tremona Road, Southampton SO16 6YD, UK; (S.A.L.); (J.A.S.); (R.M.); (R.S.D.); (E.K.); (R.W.); (A.S.); (D.D.); (T.F.); (A.J.C.); (H.L.); (T.A.N.); (A.J.L.)
| | - Helena Lee
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP 806, Tremona Road, Southampton SO16 6YD, UK; (S.A.L.); (J.A.S.); (R.M.); (R.S.D.); (E.K.); (R.W.); (A.S.); (D.D.); (T.F.); (A.J.C.); (H.L.); (T.A.N.); (A.J.L.)
- Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Tracey A. Newman
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP 806, Tremona Road, Southampton SO16 6YD, UK; (S.A.L.); (J.A.S.); (R.M.); (R.S.D.); (E.K.); (R.W.); (A.S.); (D.D.); (T.F.); (A.J.C.); (H.L.); (T.A.N.); (A.J.L.)
| | - Andrew J. Lotery
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP 806, Tremona Road, Southampton SO16 6YD, UK; (S.A.L.); (J.A.S.); (R.M.); (R.S.D.); (E.K.); (R.W.); (A.S.); (D.D.); (T.F.); (A.J.C.); (H.L.); (T.A.N.); (A.J.L.)
- Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - J. Arjuna Ratnayaka
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP 806, Tremona Road, Southampton SO16 6YD, UK; (S.A.L.); (J.A.S.); (R.M.); (R.S.D.); (E.K.); (R.W.); (A.S.); (D.D.); (T.F.); (A.J.C.); (H.L.); (T.A.N.); (A.J.L.)
| |
Collapse
|
14
|
Zhu J, Su T, Wang M, Li M, Liu L, Wang F. Highly Expressed Amyloid Beta-42 Of Aqueous Humor In Patients With Neovascular Macular Degeneration. Semin Ophthalmol 2021; 36:9-13. [PMID: 33587673 DOI: 10.1080/08820538.2021.1883679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Background: Age-related macular degeneration (AMD) is a type of macular degeneration disease, and amyloid beta (aβ) is the main component of vitreous warts in AMD patients. Neovascular AMD (nAMD) is the most serious type of AMD, but its pathogenesis remains unclear. The aim of this study was to detect the expression of aβ42 in the aqueous humor of nAMD patients and to evaluate whether aβ42 expression of aqueous humor is correlated with cognitive function in these patients.Methods: A total of 70 patients were enrolled in this study, including 50 nAMD patients (nAMD group) and 20 patients with cataract (control group). The cognitive function of the patients was assessed using the Mini-Mental State Examination and Montreal Cognitive Assessment Scale, and based on their scores, 50 patients with nAMD were divided into two subgroups: the p-nAMD group (18 nAMD patients with normal cognition) and the ci-nAMD group (32 nAMD patients with cognitive impairment). An immunofluorescence microsphere probe technique was used to detect the aβ42 expression of aqueous humor in all patients. Pearson correlation analysis was used.Results: The aβ42 expression of aqueous humor was significantly higher in the nAMD group (124.56 ± 41.93 pg/mL) as compared with the control group (82.94 ± 33.75 pg/mL; P < .01). There was no significant difference in aβ42 expression of aqueous humor between the p-nAMD group (136.42 ± 51.68 pg/mL) and ci-nAMD group (117.90 ± 34.46 pg/mL; P = .14).Conclusion: In nAMD patients, aβ42 was highly expressed in the aqueous humor but was not correlated with cognitive function.
Collapse
Affiliation(s)
- Juming Zhu
- Department of Ophthalmology, the Fourth Affiliated Hospital of Nantong University, Yancheng No.1 People's Hospital, Yancheng, Jiangsu Province, China
| | - Tu Su
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Affiliate of Tongji University, School of Medicine, Shanghai, China
| | - Minli Wang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Affiliate of Tongji University, School of Medicine, Shanghai, China
| | - Min Li
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Affiliate of Tongji University, School of Medicine, Shanghai, China
| | - Lin Liu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Affiliate of Tongji University, School of Medicine, Shanghai, China
| | - Fang Wang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Affiliate of Tongji University, School of Medicine, Shanghai, China
| |
Collapse
|
15
|
Su Y, Yi Y, Li L, Chen C. circRNA-miRNA-mRNA network in age-related macular degeneration: From construction to identification. Exp Eye Res 2020; 203:108427. [PMID: 33383027 DOI: 10.1016/j.exer.2020.108427] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 02/09/2023]
Abstract
The aim of the present study was to investigate the pathogenesis of age-related macular degeneration (AMD) by constructing a regulatory circRNA-miRNA-mRNA network. By adjusting the P value to <0.05 and the absolute log value of fold change to >0.25, 2920 and 1057 differentially expressed mRNAs were identified from GSE50195 and GSE29801, respectively. Based on a literature review, Starbase database analysis, and RNA hybrid assays, we obtained 77 miRNA-mRNA and 331 circRNA-miRNA pairs. After combining these pairs, we constructed a circRNA-miRNA-mRNA network possessing 303 circRNA nodes, 4 miRNA nodes, 51 mRNA nodes, and 408 edges. By utilizing protein-protein network analysis, the MCODE algorithm, and the highest degree of circRNA node, we identified the regulatory axis of hsa_circRNA7329/hsa-miR-9/SCD. Hsa_circRNA7329 may regulate SCD through hsa-miR-9 to promote macrophage-mediated inflammation and pathologic angiogenesis, which lead to AMD development. However, the underlying details require further investigation.
Collapse
Affiliation(s)
- Yu Su
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, Hubei province, 430060, PR China
| | - Yuexiong Yi
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei province, 430071, PR China
| | - Lu Li
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, Hubei province, 430060, PR China
| | - Changzheng Chen
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, Hubei province, 430060, PR China.
| |
Collapse
|
16
|
Soundara Pandi SP, Ratnayaka JA, Lotery AJ, Teeling JL. Progress in developing rodent models of age-related macular degeneration (AMD). Exp Eye Res 2020; 203:108404. [PMID: 33340497 DOI: 10.1016/j.exer.2020.108404] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 12/25/2022]
Abstract
Age-related macular degeneration (AMD) is the leading cause of irreversible central vision loss, typically affecting individuals from mid-life onwards. Its multifactorial aetiology and the lack of any effective treatments has spurred the development of animal models as research and drug discovery tools. Several rodent models have been developed which recapitulate key features of AMD and provide insights into its underlying pathology. These have contributed to making significant progress in understanding the disease and the identification of novel therapeutic targets. However, a major caveat with existing models is that they do not demonstrate the full disease spectrum. In this review, we outline advances in rodent AMD models from the last decade. These models feature various hallmarks associated with AMD, including oxidative stress, hypoxia, immune dysregulation, genetic mutations and environmental risk factors. The review summarises the methods by which each model was created, its pathological characteristics as well as its relation to the disease in humans.
Collapse
Affiliation(s)
- Sudha Priya Soundara Pandi
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - J Arjuna Ratnayaka
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton, SO16 6YD, United Kingdom.
| | - Andrew J Lotery
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton, SO16 6YD, United Kingdom; Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton, SO16 6YD, United Kingdom.
| | - Jessica L Teeling
- Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, MP840, Tremona Road, Southampton, SO16 6YD, United Kingdom.
| |
Collapse
|
17
|
The retinal toxicity profile towards assemblies of Amyloid-β indicate the predominant pathophysiological activity of oligomeric species. Sci Rep 2020; 10:20954. [PMID: 33262378 PMCID: PMC7708452 DOI: 10.1038/s41598-020-77712-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 11/17/2020] [Indexed: 11/22/2022] Open
Abstract
Amyloid-β (Aβ), reported as a significant constituent of drusen, was implicated in the pathophysiology of age-related macular degeneration (AMD), yet the identity of the major pathogenic Aβ species in the retina has remained hitherto unclear. Here, we examined the in-vivo retinal impact of distinct supramolecular assemblies of Aβ. Fibrillar (Aβ40, Aβ42) and oligomeric (Aβ42) preparations showed clear biophysical hallmarks of amyloid assemblies. Measures of retinal structure and function were studied longitudinally following intravitreal administration of the various Aβ assemblies in rats. Electroretinography (ERG) delineated differential retinal neurotoxicity of Aβ species. Oligomeric Aβ42 inflicted the major toxic effect, exerting diminished ERG responses through 30 days post injection. A lesser degree of retinal dysfunction was noted following treatment with fibrillar Aβ42, whereas no retinal compromise was recorded in response to Aβ40 fibrils. The toxic effect of Aβ42 architectures was further reflected by retinal glial response. Fluorescence labelling of Aβ42 species was used to detect their accumulation into the retinal tissue. These results provide conceptual evidence of the differential toxicity of particular Aβ species in-vivo, and promote the mechanistic understanding of their retinal pathogenicity. Stratifying the impact of pathological Aβ aggregation in the retina may merit further investigation to decipher the pathophysiological relevance of processes of molecular self-assembly in retinal disorders.
Collapse
|
18
|
Micera A, Bruno L, Cacciamani A, Rongioletti M, Squitti R. Alzheimer's Disease and Retinal Degeneration: A Glimpse at Essential Trace Metals in Ocular Fluids and Tissues. Curr Alzheimer Res 2020; 16:1073-1083. [PMID: 31642780 DOI: 10.2174/1567205016666191023114015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 09/28/2019] [Accepted: 10/21/2019] [Indexed: 01/19/2023]
Abstract
BACKGROUND Life expectancy is increasing all over the world, although neurodegenerative disorders might drastically affect the individual activity of aged people. Of those, Alzheimer's Disease (AD) is one of the most social-cost age-linked diseases of industrialized countries. To date, retinal diseases seem to be more common in the developing world and characterize principally aged people. Agerelated Macular Degeneration (AMD) is a late-onset, neurodegenerative retinal disease that shares several clinical and pathological features with AD, including stress stimuli such as oxidative stress, inflammation and amyloid formations. METHODS In both diseases, the detrimental intra/extra-cellular deposits have many similarities. Aging, hypercholesterolemia, hypertension, obesity, arteriosclerosis and smoking are risk factors to develop both diseases. Cellular aging routes have similar organelle and signaling patterns in retina and brain. The possibility to find out new research strategies represent a step forward to disclose potential treatment for both of them. Essential trace metals play critical roles in both physiological and pathological condition of retina, optic nerve and brain, by influencing metabolic processes chiefly upon complex multifactorial pathogenesis. CONCLUSION Hence, this review addresses current knowledge about some up-to-date investigated essential trace metals associated with AD and AMD. Changes in the levels of systemic and ocular fluid essential metals might reflect the early stages of AMD, possibly disclosing neurodegeneration pathways shared with AD, which might open to potential early detection.
Collapse
Affiliation(s)
- Alessandra Micera
- Research Laboratories in Ophthalmology, IRCCS - Fondazione Bietti, Rome, Italy
| | - Luca Bruno
- Research Laboratories in Ophthalmology, IRCCS - Fondazione Bietti, Rome, Italy
| | - Andrea Cacciamani
- Research Laboratories in Ophthalmology, IRCCS - Fondazione Bietti, Rome, Italy
| | - Mauro Rongioletti
- Department of Laboratory Medicine, Research and Development Division, San Giovanni Calibita, Fatebenefratelli Hospital, Isola Tiberina, Rome, Italy
| | - Rosanna Squitti
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, BS, Italy
| |
Collapse
|
19
|
Retinal Degeneration and Alzheimer's Disease: An Evolving Link. Int J Mol Sci 2020; 21:ijms21197290. [PMID: 33023198 PMCID: PMC7582766 DOI: 10.3390/ijms21197290] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/15/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
Age-related macular degeneration (AMD) and glaucoma are degenerative conditions of the retina and a significant cause of irreversible blindness in developed countries. Alzheimer’s disease (AD), the most common dementia of the elderly, is often associated with AMD and glaucoma. The cardinal features of AD include extracellular accumulation of amyloid β (Aβ) and intracellular deposits of hyper-phosphorylated tau (p-tau). Neuroinflammation and brain iron dyshomeostasis accompany Aβ and p-tau deposits and, together, lead to progressive neuronal death and dementia. The accumulation of Aβ and iron in drusen, the hallmark of AMD, and Aβ and p-tau in retinal ganglion cells (RGC), the main retinal cell type implicated in glaucoma, and accompanying inflammation suggest overlapping pathology. Visual abnormalities are prominent in AD and are believed to develop before cognitive decline. Some are caused by degeneration of the visual cortex, while others are due to RGC loss or AMD-associated retinal degeneration. Here, we review recent information on Aβ, p-tau, chronic inflammation, and iron dyshomeostasis as common pathogenic mechanisms linking the three degenerative conditions, and iron chelation as a common therapeutic option for these disorders. Additionally discussed is the role of prion protein, infamous for prion disorders, in Aβ-mediated toxicity and, paradoxically, in neuroprotection.
Collapse
|
20
|
Zhang L, Cui X, Han Y, Park KS, Gao X, Zhang X, Yuan Z, Hu Y, Hsu CW, Li X, Bassuk AG, Mahajan VB, Wang NK, Tsang SH. Hypoxic drive caused type 3 neovascularization in a preclinical model of exudative age-related macular degeneration. Hum Mol Genet 2020; 28:3475-3485. [PMID: 31518400 DOI: 10.1093/hmg/ddz159] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 03/29/2019] [Accepted: 07/01/2019] [Indexed: 01/27/2023] Open
Abstract
Hypoxia associated with the high metabolic demand of rods has been implicated in the pathology of age-related macular degeneration (AMD), the most common cause of adult blindness in the developed world. The majority of AMD-associated severe vision loss cases are due to exudative AMD, characterized by neovascularization. To further investigate the causes and histopathology of exudative AMD, we conditionally induced hypoxia in a novel preclinical AMD model (Pde6gcreERT2/+;Vhl-/-) by targeting Vhl and used multimodal imaging and immunohistochemistry to track the development of hypoxia-induced neovascularization. In addition to developing a preclinical model that phenocopies exudative AMD, our studies revealed that the photoreceptor hypoxic response initiates and drives type 3 neovascularization, mainly in the outer retina. Activation of the VHL-HIF1a-VEGF-EPO pathway in the adult retina led to long-term neovascularization, retinal hemorrhages and compromised retinal layers. Our novel preclinical model would accelerate the testing of therapies that use metabolomic approaches to ameliorate AMD.
Collapse
Affiliation(s)
- Lijuan Zhang
- Shanxi Eye Hospital, affiliated with Shanxi Medical University. Fudong St. 100, Xinghualing, Taiyuan, Shanxi 030002, China
| | - Xuan Cui
- Tianjin Medical University Eye Hospital, Tianjin Medical University Eye Institute & Tianjin Medical University School of Optometry and Ophthalmology, Tianjin 300384, China, New York, NY10032, USA.,Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Herbert Irving Comprehensive Cancer Center, New York, NY 10032, USA.,Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY 10032, USA
| | - Yangjun Han
- Shanxi Cardiovascular Disease Hospital, Yifen street 18, Wanbailin, Taiyuan, Shanxi 030024, China
| | - Karen Sophia Park
- Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Herbert Irving Comprehensive Cancer Center, New York, NY 10032, USA.,Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY 10032, USA
| | - Xiaohong Gao
- Shanxi Eye Hospital, affiliated with Shanxi Medical University. Fudong St. 100, Xinghualing, Taiyuan, Shanxi 030002, China
| | - Ximei Zhang
- Shanxi Eye Hospital, affiliated with Shanxi Medical University. Fudong St. 100, Xinghualing, Taiyuan, Shanxi 030002, China
| | - Zhigang Yuan
- Shanxi Eye Hospital, affiliated with Shanxi Medical University. Fudong St. 100, Xinghualing, Taiyuan, Shanxi 030002, China
| | - Yong Hu
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Chun-Wei Hsu
- Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Herbert Irving Comprehensive Cancer Center, New York, NY 10032, USA.,Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY 10032, USA
| | - Xiaorong Li
- Tianjin Medical University Eye Hospital, Tianjin Medical University Eye Institute & Tianjin Medical University School of Optometry and Ophthalmology, Tianjin 300384, China, New York, NY10032, USA
| | | | - Vinit B Mahajan
- Byers Eye Institute, Omics Laboratory, Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94303, USA.,Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Nan-Kai Wang
- Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Herbert Irving Comprehensive Cancer Center, New York, NY 10032, USA.,Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY 10032, USA
| | - Stephen H Tsang
- Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Herbert Irving Comprehensive Cancer Center, New York, NY 10032, USA.,Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY 10032, USA.,Department of Pathology & Cell Biology, Stem Cell Initiative (CSCI), Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| |
Collapse
|
21
|
Ratnayaka JA, Lotery AJ. Challenges in studying geographic atrophy (GA) age-related macular degeneration: the potential of a new mouse model with GA-like features. Neural Regen Res 2020; 15:863-864. [PMID: 31719250 PMCID: PMC6990775 DOI: 10.4103/1673-5374.268972] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- J Arjuna Ratnayaka
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Andrew J Lotery
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton; Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
22
|
Dewing JM, Carare RO, Lotery AJ, Ratnayaka JA. The Diverse Roles of TIMP-3: Insights into Degenerative Diseases of the Senescent Retina and Brain. Cells 2019; 9:cells9010039. [PMID: 31877820 PMCID: PMC7017234 DOI: 10.3390/cells9010039] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022] Open
Abstract
Tissue inhibitor of metalloproteinase-3 (TIMP-3) is a component of the extracellular environment, where it mediates diverse processes including matrix regulation/turnover, inflammation and angiogenesis. Rare TIMP-3 risk alleles and mutations are directly linked with retinopathies such as age-related macular degeneration (AMD) and Sorsby fundus dystrophy, and potentially, through indirect mechanisms, with Alzheimer's disease. Insights into TIMP-3 activities may be gleaned from studying Sorsby-linked mutations. However, recent findings do not fully support the prevailing hypothesis that a gain of function through the dimerisation of mutated TIMP-3 is responsible for retinopathy. Findings from Alzheimer's patients suggest a hitherto poorly studied relationship between TIMP-3 and the Alzheimer's-linked amyloid-beta (A) proteins that warrant further scrutiny. This may also have implications for understanding AMD as aged/diseased retinae contain high levels of A. Findings from TIMP-3 knockout and mutant knock-in mice have not led to new treatments, particularly as the latter does not satisfactorily recapitulate the Sorsby phenotype. However, recent advances in stem cell and in vitro approaches offer novel insights into understanding TIMP-3 pathology in the retina-brain axis, which has so far not been collectively examined. We propose that TIMP-3 activities could extend beyond its hitherto supposed functions to cause age-related changes and disease in these organs.
Collapse
Affiliation(s)
- Jennifer M. Dewing
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton SO16 6YD, UK; (J.M.D.); (R.O.C.); (A.J.L.)
| | - Roxana O. Carare
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton SO16 6YD, UK; (J.M.D.); (R.O.C.); (A.J.L.)
| | - Andrew J. Lotery
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton SO16 6YD, UK; (J.M.D.); (R.O.C.); (A.J.L.)
- Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - J. Arjuna Ratnayaka
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton SO16 6YD, UK; (J.M.D.); (R.O.C.); (A.J.L.)
- Correspondence: ; Tel.: +44-238120-8183
| |
Collapse
|
23
|
A dual inhibitor of the proteasome catalytic subunits LMP2 and Y attenuates disease progression in mouse models of Alzheimer's disease. Sci Rep 2019; 9:18393. [PMID: 31804556 PMCID: PMC6895163 DOI: 10.1038/s41598-019-54846-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/15/2019] [Indexed: 12/18/2022] Open
Abstract
The immunoproteasome (iP) is a variant of the constitutive proteasome (cP) that is abundantly expressed in immune cells which can also be induced in somatic cells by cytokines such as TNF-α or IFN-γ. Accumulating evidence support that the iP is closely linked to multiple facets of inflammatory response, eventually leading to the development of several iP inhibitors as potential therapeutic agents for autoimmune diseases. Recent studies also found that the iP is upregulated in reactive glial cells surrounding amyloid β (Aβ) deposits in brains of Alzheimer’s disease (AD) patients, but the role it plays in the pathogenesis of AD remains unclear. In this study, we investigated the effects of several proteasome inhibitors on cognitive function in AD mouse models and found that YU102, a dual inhibitor of the iP catalytic subunit LMP2 and the cP catalytic subunit Y, ameliorates cognitive impairments in AD mouse models without affecting Aβ deposition. The data obtained from our investigation revealed that YU102 suppresses the secretion of inflammatory cytokines from microglial cells. Overall, this study indicates that there may exist a potential link between LMP2/Y and microglia-mediated neuroinflammation and that inhibition of these subunits may offer a new therapeutic strategy for AD.
Collapse
|
24
|
Elovanoids counteract oligomeric β-amyloid-induced gene expression and protect photoreceptors. Proc Natl Acad Sci U S A 2019; 116:24317-24325. [PMID: 31712409 PMCID: PMC6883841 DOI: 10.1073/pnas.1912959116] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
This study uncovers biosynthetic pathway insufficiencies of prohomeostatic/neuroprotective mediators neuroprotectin D1 and elovanoids in the retina during early pathogenesis in transgenic Alzheimer’s disease 5xFAD mouse. These changes correlate with photoreceptor cell functional impairments preceding their loss. Amyloid beta (Aβ) peptide accumulates in drusen in AMD. Thus, injecting oligomeric Aβ in wild-type mice behind the retina leads to photoreceptor cell degeneration and transcriptional disruptions including upregulation of a senescence program and of senescence-associated secretory phenotype (SASP). Similar changes take place in human retinal pigment epithelium cells in culture. Novel lipid mediators, the elovanoids, restore Aβ-peptide-induced gene expression changes and SASP secretome and, in turn, protect these cells. This study opens avenues of potential therapeutic exploration of elovanoids for AMD. The onset of neurodegenerative diseases activates inflammation that leads to progressive neuronal cell death and impairments in cognition (Alzheimer’s disease) and sight (age-related macular degeneration [AMD]). How neuroinflammation can be counteracted is not known. In AMD, amyloid β-peptide (Aβ) accumulates in subretinal drusen. In the 5xFAD retina, we found early functional deficiencies (ERG) without photoreceptor cell (PRC) death and identified early insufficiency in biosynthetic pathways of prohomeostatic/neuroprotective mediators neuroprotectin D1 (NPD1) and elovanoids (ELVs). To mimic an inflammatory milieu in wild-type mouse, we triggered retinal pigment epithelium (RPE) damage/PRC death by subretinally injected oligomeric β-amyloid (OAβ) and observed that ELVs administration counteracted their effects, protecting these cells. In addition, ELVs prevented OAβ-induced changes in gene expression engaged in senescence, inflammation, autophagy, extracellular matrix remodeling, and AMD. Moreover, as OAβ targets the RPE, we used primary human RPE cell cultures and demonstrated that OAβ caused cell damage, while ELVs protected and restored gene expression as in mouse. Our data show OAβ activates senescence as reflected by enhanced expression of p16INK4a, MMP1, p53, p21, p27, and Il-6, and of senescence-associated phenotype secretome, followed by RPE and PRC demise, and that ELVs 32 and 34 blunt these events and elicit protection. In addition, ELVs counteracted OAβ-induced expression of genes engaged in AMD, autophagy, and extracellular matrix remodeling. Overall, our data uncovered that ELVs downplay OAβ-senescence program induction and inflammatory transcriptional events and protect RPE cells and PRC, and therefore have potential as a possible therapeutic avenue for AMD.
Collapse
|
25
|
Bogolepova AN, Makhnovich EV, Zhuravleva AN. [Comorbidity of Alzheimer's disease and gegontophtalmic pathology]. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 119:17-22. [PMID: 31626214 DOI: 10.17116/jnevro201911909117] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
AIM To compare the frequency of age-related ophthalmic diseases in patients with Alzheimer's disease and vascular dementia. MATERIAL AND METHODS The study included 60 patients with severe cognitive impairment divided into two equal groups matched for sex and age. The first group included patients diagnosed with Alzheimer's disease. The second group consisted of people with a diagnosis of vascular dementia. All patients underwent clinical and neuropsychological as well as standard (visometry, refraction, measurement of intraocular pressure, biomicroscopy, ophthalmoscopy, computed perimetry) and complex (optic coherent tomography) ophthalmologic examinations. RESULTS AND CONCLUSION In patients with Alzheimer's disease, concomitant ophthalmologic pathology in the form of glaucoma was observed more often in 46,7% of cases, pseudoexfoliation syndrome in 20% of cases, age-related macular degeneration in 16,7% of cases, while in the vascular dementia group the majority (80%) of patients did not have concomitant ophthalmologic pathology. When analyzing the severity of cognitive impairment in patients with Alzheimer's disease and various concomitant ophthalmic pathology, it was shown that patients with Alzheimer's disease and glaucoma have more significant impairments compared to patients without concomitant ophthalmological pathology.
Collapse
Affiliation(s)
- A N Bogolepova
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - E V Makhnovich
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - A N Zhuravleva
- Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia
| |
Collapse
|
26
|
Grimaldi A, Pediconi N, Oieni F, Pizzarelli R, Rosito M, Giubettini M, Santini T, Limatola C, Ruocco G, Ragozzino D, Di Angelantonio S. Neuroinflammatory Processes, A1 Astrocyte Activation and Protein Aggregation in the Retina of Alzheimer's Disease Patients, Possible Biomarkers for Early Diagnosis. Front Neurosci 2019; 13:925. [PMID: 31551688 PMCID: PMC6737046 DOI: 10.3389/fnins.2019.00925] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/19/2019] [Indexed: 12/28/2022] Open
Abstract
Alzheimer’s disease (AD), a primary cause of dementia in the aging population, is characterized by extracellular amyloid-beta peptides aggregation, intracellular deposits of hyperphosphorylated tau, neurodegeneration and glial activation in the brain. It is commonly thought that the lack of early diagnostic criteria is among the main causes of pharmacological therapy and clinical trials failure; therefore, the actual challenge is to define new biomarkers and non-invasive technologies to measure neuropathological changes in vivo at pre-symptomatic stages. Recent evidences obtained from human samples and mouse models indicate the possibility to detect protein aggregates and other pathological features in the retina, paving the road for non-invasive rapid detection of AD biomarkers. Here, we report the presence of amyloid beta plaques, tau tangles, neurodegeneration and detrimental astrocyte and microglia activation according to a disease associated microglia phenotype (DAM). Thus, we propose the human retina as a useful site for the detection of cellular and molecular changes associated with Alzheimer’s disease.
Collapse
Affiliation(s)
- Alfonso Grimaldi
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, Rome, Italy
| | - Natalia Pediconi
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, Rome, Italy
| | - Francesca Oieni
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Rocco Pizzarelli
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, Rome, Italy
| | - Maria Rosito
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, Rome, Italy
| | | | - Tiziana Santini
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, Rome, Italy
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| | - Giancarlo Ruocco
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, Rome, Italy.,Department of Physics, Sapienza University, Rome, Italy
| | - Davide Ragozzino
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| | - Silvia Di Angelantonio
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, Rome, Italy.,Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| |
Collapse
|
27
|
Muraleva NA, Kozhevnikova OS, Fursova AZ, Kolosova NG. Suppression of AMD-Like Pathology by Mitochondria-Targeted Antioxidant SkQ1 Is Associated with a Decrease in the Accumulation of Amyloid β and in mTOR Activity. Antioxidants (Basel) 2019; 8:antiox8060177. [PMID: 31208023 PMCID: PMC6616484 DOI: 10.3390/antiox8060177] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/10/2019] [Accepted: 06/12/2019] [Indexed: 11/01/2022] Open
Abstract
Age-related macular degeneration (AMD) is a major cause of irreversible visual impairment and blindness in developed countries, and the molecular pathogenesis of AMD is poorly understood. Recent studies strongly indicate that amyloid β (Aβ) accumulation -found in the brain and a defining feature of Alzheimer's disease-also forms in the retina in both Alzheimer's disease and AMD. The reason why highly neurotoxic proteins of consistently aggregate in the aging retina, and to what extent they contribute to AMD, remains to be fully addressed. Nonetheless, the hypothesis that Aβ is a therapeutic target in AMD is debated. Here, we showed that long-term treatment with SkQ1 (250 nmol/[kg body weight] daily from the age of 1.5 to 22 months) suppressed the development of AMD-like pathology in senescence-accelerated OXYS rats by reducing the level of Aβ and suppressing the activity of mTOR in the retina. Inhibition of mTOR signaling activity, which plays key roles in aging and age-related diseases, can be considered a new mechanism of the prophylactic effect of SkQ1. It seems probable that dietary supplementation with mitochondria-targeted antioxidant SkQ1 can be a good prevention strategy to maintain eye health and possibly a treatment of AMD.
Collapse
Affiliation(s)
- Natalia A Muraleva
- Institute of Cytology and Genetics SB RAS, Pr. Lavrentyeva 10, Novosibirsk 630090, Russia.
| | - Oyuna S Kozhevnikova
- Institute of Cytology and Genetics SB RAS, Pr. Lavrentyeva 10, Novosibirsk 630090, Russia.
| | - Anzhela Z Fursova
- Institute of Cytology and Genetics SB RAS, Pr. Lavrentyeva 10, Novosibirsk 630090, Russia.
| | - Nataliya G Kolosova
- Institute of Cytology and Genetics SB RAS, Pr. Lavrentyeva 10, Novosibirsk 630090, Russia.
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry SB RAS, 9 Lavrentieva Avenue, Novosibirsk 630090, Russia.
| |
Collapse
|
28
|
Masuda N, Tsujinaka H, Hirai H, Yamashita M, Ueda T, Ogata N. Effects of concentration of amyloid β (Aβ) on viability of cultured retinal pigment epithelial cells. BMC Ophthalmol 2019; 19:70. [PMID: 30849957 PMCID: PMC6408759 DOI: 10.1186/s12886-019-1076-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 02/28/2019] [Indexed: 11/10/2022] Open
Abstract
Background Amyloid beta (Aβ) is a constituent of drusen that is a common sign of age-related macular degeneration (AMD). The purpose of this study was to investigate the effect of Aβ on human retinal pigment epithelial (RPE) cells in culture. Methods Cells from a human RPE cell line (ARPE-19) were exposed to 0 to 25 μM of Aβ 1–40 for 48 h, and the number of living cells was determined by WST-8 cleavage. Replicative DNA synthesis was measured by the incorporation of 5′-bromo-2′-deoxyuridine. The cell death pathway was investigated by the WST-8 cleavage assay after the addition of caspase-9 inhibitor, an anti-apoptotic factor. Real-time qRT-PCR was performed using Aβ-exposed cellular RNA to determine the level of vascular endothelial growth factor (VEGF)-A and pigment epithelium derived factor (PEDF). To determine the effect of receptor-for-advanced glycation end products (RAGE), the siRNA for RAGE was inserted into ARPE-19 treated with Aβ, and the levels of expression of VEGF-A and PEDF were determined. Results The number of living ARPE-19 cells was increased by exposure to 5 μM Aβ but was decreased by exposure to 25 μM of Aβ. Replicative DNA synthesis by ARPE-19 cells exposed to 25 μM of Aβ was significantly decreased indicating that 25 μM of Aβ inhibited cell proliferation. Real-time RT-PCR showed that the level of the mRNA of PEDF was increased by exposure to 5 μM Aβ, and the levels of the mRNAs of PEDF and VEGF-A were also increased by exposure to 25 μM Aβ. The addition of an inhibitor of caspase-9 blocked the decrease the number of ARPE-19 cells exposed to 25 μM Aβ. Exposure to si-RAGE attenuated the increase of VEGF-A and PEDF mRNA expression in ARPE-19 exposed to Aβ. Conclusions Exposure of ARPE-19 cells to low concentrations of Aβ increases the level of PEDF which then inhibits the apoptosis of ARPE-19 cells leading to RPE cell proliferation. Exposure to high concentrations of Aβ induces RPE cell death and enhances the expression of the mRNA of VEGF-A in RPE cells. The Aβ-RAGE pathway may lead to the expression VEGF-A and PEDF in RPE cells. These results suggest that Aβ is strongly related to the pathogenesis of choroidal neovascularization.
Collapse
Affiliation(s)
- Naonori Masuda
- Department of Ophthalmology, Nara Medical University, 840 Shijo-cho, Kashihara, 634-8522, Japan
| | - Hiroki Tsujinaka
- Department of Ophthalmology, Nara Medical University, 840 Shijo-cho, Kashihara, 634-8522, Japan
| | - Hiromasa Hirai
- Department of Ophthalmology, Nara Medical University, 840 Shijo-cho, Kashihara, 634-8522, Japan
| | - Mariko Yamashita
- Department of Ophthalmology, Nara Medical University, 840 Shijo-cho, Kashihara, 634-8522, Japan
| | - Tetsuo Ueda
- Department of Ophthalmology, Nara Medical University, 840 Shijo-cho, Kashihara, 634-8522, Japan
| | - Nahoko Ogata
- Department of Ophthalmology, Nara Medical University, 840 Shijo-cho, Kashihara, 634-8522, Japan.
| |
Collapse
|
29
|
Mohlin C, Delbro D, Kvanta A, Johansson K. Evaluation of Congo Red Staining in Degenerating Porcine Photoreceptors In Vitro: Protective Effects by Structural and Trophic Support. J Histochem Cytochem 2018; 66:631-641. [PMID: 29624116 PMCID: PMC6116089 DOI: 10.1369/0022155418768222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 03/07/2018] [Indexed: 11/22/2022] Open
Abstract
Congo red (CR) is a histological stain used for the detection of extracellular amyloids mediating various neurodegenerative diseases. Given that damaged photoreceptors appear to degenerate similarly to other nerve cells, CR staining was evaluated in experimentally injured porcine retina. CR staining appeared mostly as discrete cytosolic deposits with no obvious plaque formation during the investigated time period. Increases of CR labeling coincided temporally with the known accumulation of mislocalized opsins and increases of cell death. Coculture, either with human retinal pigment epithelium (ARPE) or human neural progenitor (ReN) cells, was accompanied by a significant reduction of CR labeling. Of particular interest was the reduction of CR labeling in cone photoreceptors, which are important for the perception of color and fine details and afflicted in age-related macular degeneration (AMD). Electron microscopy revealed inclusions in the inner segment, cell body, and occasionally synaptic terminals of photoreceptor cells in cultured specimens. Closer examinations indicated the presence of different types of inclusions resembling protein aggregates as well as inclusion bodies. The current results indicate that injury-related response resulted in accumulation of CR deposits in photoreceptor cells, and that trophic and/or structural support attenuated this response.
Collapse
Affiliation(s)
- Camilla Mohlin
- Department of Chemistry and Biomedicine,
Linnaeus University, Kalmar, Sweden
| | - Dick Delbro
- School of Medical Sciences, Örebro University,
Örebro, Sweden
| | - Anders Kvanta
- Department of Clinical Neuroscience, Section for
Ophthalmology and Vision, St. Erik Eye Hospital, Karolinska Institutet,
Stockholm, Sweden
| | - Kjell Johansson
- Department of Science, Kristianstad University,
Kristianstad, Sweden
| |
Collapse
|
30
|
Pogue AI, Lukiw WJ. Up-regulated Pro-inflammatory MicroRNAs (miRNAs) in Alzheimer's disease (AD) and Age-Related Macular Degeneration (AMD). Cell Mol Neurobiol 2018; 38:1021-1031. [PMID: 29302837 DOI: 10.1007/s10571-017-0572-3] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 12/21/2017] [Indexed: 11/25/2022]
Abstract
Alzheimer's disease (AD) of the brain neocortex and age-related macular degeneration (AMD) of the retina are two complex neurodegenerative disorders, which (i) involve the progressive dysregulation and deterioration of multiple neurobiological signaling pathways, (ii) exhibit the temporal accumulation of pro-inflammatory lesions including the amyloid beta (Aβ) peptide-containing senile plaques of AD and the drusen of AMD, and (iii) culminate in an insidious inflammatory neurodegeneration ending, respectively, in neural cell atrophy and death and progressive loss of cognition and central visual function. Recent independent research studies have indicated that AD and AMD share common, pathological signaling defects and disease mechanisms at the molecular genetic level. Using high-integrity total RNA samples pooled from AD brain and AMD retina, microfluidic hybridization miRNA arrays, and bioinformatics, the current study was undertaken to quantify microRNA (miRNA) speciation and complexity common to both AD and AMD. These small non-coding (sncRNAs) are known to post-transcriptionally regulate multiple neurobiological pathways and an abundance of research information has already been generated on the roles of these miRNAs in pathological situations involving inflammatory neuropathology and neural cell decline. Here, for the first time, we report the sequence and abundance of a septet of sncRNAs including miRNA-7, miRNA-9-1, miRNA-23a/miRNA-27a, miRNA-34a, miRNA-125b-1, miRNA-146a, and miRNA-155 that are significantly increased in abundance and common to both AD-affected superior temporal lobe neocortex (Brodmann A22) and the AMD-affected macular region of the retina. Bioinformatics, miRNA-mRNA complementarity, next-gen RNA sequencing, and feature alignment analysis further indicate that these 7 up-regulated miRNAs have the potential to interact with and down-regulate ~ 9460 target messenger RNAs (mRNAs; about 3.5% of the genome) involved in the synchronization of amyloid production and clearance, phagocytosis, innate-immune, pro-inflammatory, and neurotrophic signaling and/or synaptogenesis in diseased tissues.
Collapse
Affiliation(s)
| | - Walter J Lukiw
- Alchem Biotech Research, Toronto, ON, Canada.
- Neuroscience Center, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, 70112-2272, USA.
- Department of Neurology, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, 70112-2272, USA.
- Department of Ophthalmology, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, 70112-2272, USA.
- LSU Neuroscience Center, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite 904, New Orleans, LA, 70112-2272, USA.
| |
Collapse
|
31
|
Grimaldi A, Brighi C, Peruzzi G, Ragozzino D, Bonanni V, Limatola C, Ruocco G, Di Angelantonio S. Inflammation, neurodegeneration and protein aggregation in the retina as ocular biomarkers for Alzheimer's disease in the 3xTg-AD mouse model. Cell Death Dis 2018; 9:685. [PMID: 29880901 PMCID: PMC5992214 DOI: 10.1038/s41419-018-0740-5] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/18/2018] [Accepted: 05/22/2018] [Indexed: 12/03/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in the elderly. In the pathogenesis of AD a pivotal role is played by two neurotoxic proteins that aggregate and accumulate in the central nervous system: amyloid beta and hyper-phosphorylated tau. Accumulation of extracellular amyloid beta plaques and intracellular hyper-phosphorylated tau tangles, and consequent neuronal loss begins 10–15 years before any cognitive impairment. In addition to cognitive and behavioral deficits, sensorial abnormalities have been described in AD patients and in some AD transgenic mouse models. Retina can be considered a simple model of the brain, as some pathological changes and therapeutic strategies from the brain may be observed or applicable to the retina. Here we propose new retinal biomarkers that could anticipate the AD diagnosis and help the beginning and the follow-up of possible future treatments. We analyzed retinal tissue of triple-transgenic AD mouse model (3xTg-AD) for the presence of pathological hallmarks during disease progression. We found the presence of amyloid beta plaques, tau tangles, neurodegeneration, and astrogliosis in the retinal ganglion cell layer of 3xTg-AD mice, already at pre-symptomatic stage. Moreover, retinal microglia in pre-symptomatic mice showed a ramified, anti-inflammatory phenotype which, during disease progression, switches to a pro-inflammatory, less ramified one, becoming neurotoxic. We hypothesize retina as a window through which monitor AD-related neurodegeneration process.
Collapse
Affiliation(s)
- Alfonso Grimaldi
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, Rome, Italy
| | - Carlo Brighi
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, Rome, Italy
| | - Giovanna Peruzzi
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, Rome, Italy
| | - Davide Ragozzino
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| | | | - Cristina Limatola
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| | - Giancarlo Ruocco
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, Rome, Italy.,Department of Physics, Sapienza University, Rome, Italy
| | - Silvia Di Angelantonio
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, Rome, Italy. .,Department of Physiology and Pharmacology, Sapienza University, Rome, Italy.
| |
Collapse
|
32
|
Lei C, Lin R, Wang J, Tao L, Fu X, Qiu Y, Lei B. Amelioration of amyloid β-induced retinal inflammatory responses by a LXR agonist TO901317 is associated with inhibition of the NF-κB signaling and NLRP3 inflammasome. Neuroscience 2017; 360:48-60. [DOI: 10.1016/j.neuroscience.2017.07.053] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/19/2017] [Accepted: 07/20/2017] [Indexed: 02/08/2023]
|
33
|
Ex-vivo models of the Retinal Pigment Epithelium (RPE) in long-term culture faithfully recapitulate key structural and physiological features of native RPE. Tissue Cell 2017; 49:447-460. [PMID: 28669519 PMCID: PMC5545183 DOI: 10.1016/j.tice.2017.06.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 06/16/2017] [Accepted: 06/16/2017] [Indexed: 01/06/2023]
Abstract
Damage to the Retinal Pigment Epithelium (RPE) is a key feature of retinopathy. We describe 2 substrates which support RPE cultures for long-term studies. Substrates were; a polyester transwell membrane and a novel electrospun scaffold. Both support RPE cultures with structural and functional features of native RPE. Electrospun scaffolds may be better for studying some disease-linked RPE changes.
The Retinal Pigment Epithelium (RPE) forms the primary site of pathology in several blinding retinopathies. RPE cultures are being continuously refined so that dynamic disease processes in this important monolayer can be faithfully studied outside the eye over longer periods. The RPE substrate, which mimics the supportive Bruch’s membrane (BrM), plays a key role in determining how well in-vitro cultures recapitulate native RPE cells. Here, we evaluate how two different types of BrM substrates; (1) a commercially-available polyester transwell membrane, and (2) a novel electrospun scaffold developed in our laboratory, could support the generation of realistic RPE tissues in culture. Our findings reveal that both substrates were capable of supporting long-lasting RPE monolayers with structural and functional specialisations of in-situ RPE cells. These cultures were used to study autofluorescence and barrier formation, as well as activities such as outer-segment internalisation/trafficking and directional secretion of key proteins; the impairment of which underlies retinal disease. Hence, both substrates fulfilled important criteria for generating authentic in-vitro cultures and act as powerful tools to study RPE pathophysiology. However, RPE grown on electrospun scaffolds may be better suited to studying complex RPE-BrM interactions such as the formation of drusen-like deposits associated with early retinal disease.
Collapse
|