1
|
He Y, Lu J, Du Y, Zhao L, Gong L, Wu P, Shu Q, Peng H, Wang X. Investigation of PANoptosis pathway in age-related macular degeneration triggered by Aβ1-40. Sci Rep 2025; 15:13514. [PMID: 40251333 PMCID: PMC12008305 DOI: 10.1038/s41598-025-98174-x] [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: 01/15/2025] [Accepted: 04/09/2025] [Indexed: 04/20/2025] Open
Abstract
Our study aimed to identify PANoptosis in Aβ1-40-induced AMD, both in vivo and in vitro, and to determine if AIM2-PANoptosome mediates this process. We used transcriptomics to explore the signaling pathways and target genes linked to PANoptosis within a mouse model of AMD triggered by Aβ1-40. Optical coherence tomography (OCT), hematoxylin and eosin (H&E) staining, and electroretinography (ERG) were employed to assess retinal damage in terms of morphology and function. Morphological changes in ARPE-19 cells were observed using optical microscopy and scanning electron microscopy. Enzyme-linked immunosorbent assay (ELISA) was used to detect the levels of cytokines in cell supernatants, mouse orbital serum, and human plasma to evaluate the severity of inflammation. CO-immunoprecipitation(CoIP) and molecular docking were performed to assess the impact and expression of proteins associated with the AIM2-PANoptosome. Quantitative polymerase chain reaction (qPCR), Western blot (WB), immunofluorescence, and apoptosis detection kits were used to evaluate the expression levels of genes and proteins related to PANoptosis-like cell death. Our results showed that the Aβ1-40-induced AMD model had increased expression of apoptosis, necroptosis, and pyroptosis pathways, and AIM2-PANoptosome components. CoIP and docking confirmed increased AIM2, ZBP1, and PYRIN levels under Aβ1-40 treatment. WB and immunofluorescence showed upregulation of PANoptosis-related proteins. Inhibitors reduced Aβ-induced protein expression. ELISA showed increased inflammatory cytokines. Apoptosis assays and microscopy revealed Aβ1-40-induced ARPE-19 cell loss and morphological changes. In conclusion, the Aβ1-40-induced AMD model displayed PANoptosis-like cell death, offering insights into disease pathogenesis.
Collapse
Affiliation(s)
- Yuxia He
- Department of Ophthalmology, Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Guiyang Aier Eye Hospital, Guiyang, Guizhou Province, China
| | - Jing Lu
- Department of Ophthalmology, Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yong Du
- Department of Ophthalmology, Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Long Zhao
- Department of Ophthalmology, Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lili Gong
- Guiyang Aier Eye Hospital, Guiyang, Guizhou Province, China
| | - Ping Wu
- Department of Ophthalmology, Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Qinxin Shu
- Department of Ophthalmology, Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hui Peng
- Department of Ophthalmology, Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Xing Wang
- Department of Ophthalmology, Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
2
|
Lizama BN, Keeling E, Cho E, Malagise EM, Knezovich N, Waybright L, Watto E, Look G, Di Caro V, Caggiano AO, Ratnayaka JA, Hamby ME. Sigma-2 receptor modulator CT1812 alters key pathways and rescues retinal pigment epithelium (RPE) functional deficits associated with dry age-related macular degeneration (AMD). Sci Rep 2025; 15:4256. [PMID: 39929889 PMCID: PMC11810999 DOI: 10.1038/s41598-025-87921-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 01/22/2025] [Indexed: 02/13/2025] Open
Abstract
Trafficking defects in retinal pigmented epithelial (RPE) cells contribute to RPE atrophy, a hallmark of geographic atrophy (GA) in dry age-related macular degeneration (AMD). Dry AMD pathogenesis is multifactorial, including amyloid-β (Aβ) accumulation and oxidative stress-common features of Alzheimer's disease (AD). The Sigma-2 receptor (S2R) regulates lipid and protein trafficking, and S2R modulators reverse trafficking deficits in neurodegeneration in vitro models. Given overlapping mechanisms contributing to AD and AMD, S2R modulator effects on RPE function were investigated. The S2R modulator CT1812 is in clinical trials for AD, dementia with Lewy bodies, and GA. Leveraging AD trials testing CT1812, unbiased analyses of patient biofluid proteomes revealed that proteins altered by CT1812 associated with GA and macular degeneration disease ontologies and overlapped with proteins altered in dry AMD. Differential expression analysis of RPE transcripts from APP-Swedish/London mutant transgenic mice, a model featuring Aβ accumulation, revealed reversal of autophagy/trafficking transcripts in S2R modulator-treated animals versus vehicle toward healthy control levels. Photoreceptor outer segment (POS) trafficking in human RPE cells showed deficits in response to Aβ1-42 or hydrogen peroxide compared to vehicle. S2R modulators normalized stressor-induced POS trafficking deficits, resembling healthy control. Taken together, S2R modulation may provide a novel therapeutic strategy for dry AMD.
Collapse
Affiliation(s)
| | - Eloise Keeling
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton, SO16 6YD, UK
| | - Eunah Cho
- Cognition Therapeutics Inc., Pittsburgh, PA, USA
| | - Evi M Malagise
- Cognition Therapeutics Inc., Pittsburgh, PA, USA
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | | | | | - Emily Watto
- Cognition Therapeutics Inc., Pittsburgh, PA, USA
| | - Gary Look
- Cognition Therapeutics Inc., Pittsburgh, PA, USA
| | | | | | - J Arjuna Ratnayaka
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton, SO16 6YD, UK
| | - Mary E Hamby
- Cognition Therapeutics Inc., Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Qarawani A, Naaman E, Ben‐Zvi Elimelech R, Harel M, Sigal‐Dror S, Ben‐Zur T, Ziv T, Offen D, Zayit‐Soudry S. Mesenchymal stem cell-derived exosomes mitigate amyloid β-induced retinal toxicity: Insights from rat model and cellular studies. JOURNAL OF EXTRACELLULAR BIOLOGY 2025; 4:e70024. [PMID: 39844926 PMCID: PMC11752158 DOI: 10.1002/jex2.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 10/21/2024] [Accepted: 11/14/2024] [Indexed: 01/24/2025]
Abstract
Amyloid β (Aβ) has emerged as a pathophysiological driver in age-related macular degeneration (AMD), emphasizing its significance in the aetiology of this prevalent sight-threatening condition. The multifaceted nature of AMD pathophysiology, presumably involving diverse retinal cascades, corresponds with the complexity of Aβ-induced retinopathy. Therefore, targeting a broad array of pathogenic processes holds promise for therapeutic intervention in AMD-associated retinal pathology. This study investigates the potential of exosomes derived from adipose tissue mesenchymal stem cells (AT-MSC-Exosomes) in alleviating Aβ-induced retinotoxicity. Through intravitreal injections in wild-type rats and RPE-like cell culture experiments, we examined the protective effects of AT-MSC-Exosomes against Aβ42 retinotoxicity. Our findings reveal that pre-treatment with AT-MSC-Exosomes enabled nearly-intact retinal function in vivo and maintained retinal cell viability in vitro, evidenced by longitudinal electroretinography (ERG) and XTT proliferation assays, respectively. Fluorescent labelling demonstrated increased migration of AT-MSC-Exosomes towards retinal cells under conditions of amyloid-related toxicity. Proteomic analysis indicated a decrease in the retinal levels of heat-shock proteins activated by pathogenic Aβ fibrils following AT-MSC-Exosome treatment. Similarly, immunostaining highlighted the modulation of α-crystallin expression in retinal astrocytes by AT-MSC-Exosomes. These results suggest the potential therapeutic relevance of AT-MSC-Exosomes in Aβ-related retinal pathology, offering a promising avenue for future AMD treatment strategies.
Collapse
Affiliation(s)
- Amanda Qarawani
- Ruth and Bruce Rappaport Faculty of MedicineTechnion Israel Institute of TechnologyHaifaIsrael
- Clinical Research InstituteRambam Health Care CampusHaifaIsrael
| | - Efrat Naaman
- Ruth and Bruce Rappaport Faculty of MedicineTechnion Israel Institute of TechnologyHaifaIsrael
- Department of OphthalmologyRambam Health Care CampusHaifaIsrael
| | - Rony Ben‐Zvi Elimelech
- Ruth and Bruce Rappaport Faculty of MedicineTechnion Israel Institute of TechnologyHaifaIsrael
- Clinical Research InstituteRambam Health Care CampusHaifaIsrael
| | - Michal Harel
- Ruth and Bruce Rappaport Faculty of MedicineTechnion Israel Institute of TechnologyHaifaIsrael
| | - Shahaf Sigal‐Dror
- Ruth and Bruce Rappaport Faculty of MedicineTechnion Israel Institute of TechnologyHaifaIsrael
- Clinical Research InstituteRambam Health Care CampusHaifaIsrael
| | - Tali Ben‐Zur
- Department of Human Genetics and Biochemistry, School of Medicine, Felsenstein Medical Research CenterTel Aviv UniversityTel AvivIsrael
| | - Tamar Ziv
- The Smoler Proteomics CenterTechnion Israel Institute of TechnologyHaifaIsrael
| | - Daniel Offen
- Department of Human Genetics and Biochemistry, School of Medicine, Felsenstein Medical Research CenterTel Aviv UniversityTel AvivIsrael
| | - Shiri Zayit‐Soudry
- Clinical Research InstituteRambam Health Care CampusHaifaIsrael
- Department of Ophthalmology, Rabin Medical CenterTel Aviv UniversityTel AvivIsrael
| |
Collapse
|
4
|
Hyttinen JMT, Koskela A, Blasiak J, Kaarniranta K. Autophagy in drusen biogenesis secondary to age-related macular degeneration. Acta Ophthalmol 2024; 102:759-772. [PMID: 39087629 DOI: 10.1111/aos.16744] [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: 03/25/2024] [Accepted: 07/11/2024] [Indexed: 08/02/2024]
Abstract
Age-related macular degeneration (AMD) is an emerging cause of blindness in aged people worldwide. One of the key signs of AMD is the degeneration of the retinal pigment epithelium (RPE), which is indispensable for the maintenance of the adjacent photoreceptors. Because of impaired energy metabolism resulting from constant light exposure, hypoxia, and oxidative stress, accumulation of drusen in AMD-affected eyes is observed. Drusen contain damaged cellular proteins, lipoprotein particles, lipids and carbohydrates and they are related to impaired protein clearance, inflammation, and extracellular matrix modification. When autophagy, a major cellular proteostasis pathway, is impaired, the accumulations of intracellular lipofuscin and extracellular drusen are detected. As these aggregates grow over time, they finally cause the disorganisation and destruction of the RPE and photoreceptors leading to visual loss. In this review, the role of autophagy in drusen biogenesis is discussed since impairment in removing cellular waste in RPE cells plays a key role in AMD progression. In the future, means which improve intracellular clearance might be of use in AMD therapy to slow the progression of drusen formation.
Collapse
Affiliation(s)
- Juha M T Hyttinen
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Ali Koskela
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Janusz Blasiak
- Faculty of Medicine, Collegium Medicum, Mazovian Academy in Plock, Plock, Poland
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Molecular Genetics, University of Lodz, Lodz, Poland
- Department of Ophthalmology, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
5
|
Kim SY, Cheon J. Senescence-associated microvascular endothelial dysfunction: A focus on the blood-brain and blood-retinal barriers. Ageing Res Rev 2024; 100:102446. [PMID: 39111407 DOI: 10.1016/j.arr.2024.102446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/05/2024] [Accepted: 08/02/2024] [Indexed: 08/17/2024]
Abstract
The blood-brain barrier (BBB) and blood-retinal barrier (BRB) constitute critical physiochemical interfaces, precisely orchestrating the bidirectional communication between the brain/retina and blood. Increased permeability or leakage of these barriers has been demonstrably linked to age-related vascular and parenchymal damage. While it has been suggested that the gradual aging process may coincide with disruptions in these barriers, this phenomenon is significantly exacerbated in individuals with age-related neurodegenerative disorders (ARND). This review focuses on the microvascular endothelium, a key constituent of BBB and BRB, highlighting the impact of endothelial senescence on barrier dysfunction and exploring recent discoveries regarding core pathways implicated in its breakdown. Subsequently, we address the "vascular senescence hypothesis" for ARND, with a particular emphasis on Alzheimer's disease and age-related macular degeneration, centered on endothelial senescence. Finally, we discuss potential senotherapeutic strategies targeting barrier dysfunction.
Collapse
Affiliation(s)
- Sung Young Kim
- Department of Biochemistry, Konkuk University School of Medicine, Republic of Korea; Research Institute of Medical Science, Konkuk University, Republic of Korea; IBST, Konkuk University, Republic of Korea.
| | - Jaejoung Cheon
- Department of Biochemistry, Konkuk University School of Medicine, Republic of Korea
| |
Collapse
|
6
|
Abyadeh M, Gupta V, Paulo JA, Mahmoudabad AG, Shadfar S, Mirshahvaladi S, Gupta V, Nguyen CT, Finkelstein DI, You Y, Haynes PA, Salekdeh GH, Graham SL, Mirzaei M. Amyloid-beta and tau protein beyond Alzheimer's disease. Neural Regen Res 2024; 19:1262-1276. [PMID: 37905874 PMCID: PMC11467936 DOI: 10.4103/1673-5374.386406] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/08/2023] [Accepted: 09/07/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT The aggregation of amyloid-beta peptide and tau protein dysregulation are implicated to play key roles in Alzheimer's disease pathogenesis and are considered the main pathological hallmarks of this devastating disease. Physiologically, these two proteins are produced and expressed within the normal human body. However, under pathological conditions, abnormal expression, post-translational modifications, conformational changes, and truncation can make these proteins prone to aggregation, triggering specific disease-related cascades. Recent studies have indicated associations between aberrant behavior of amyloid-beta and tau proteins and various neurological diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, as well as retinal neurodegenerative diseases like Glaucoma and age-related macular degeneration. Additionally, these proteins have been linked to cardiovascular disease, cancer, traumatic brain injury, and diabetes, which are all leading causes of morbidity and mortality. In this comprehensive review, we provide an overview of the connections between amyloid-beta and tau proteins and a spectrum of disorders.
Collapse
Affiliation(s)
| | - Vivek Gupta
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | | - Sina Shadfar
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Shahab Mirshahvaladi
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Veer Gupta
- School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Christine T.O. Nguyen
- Department of Optometry and Vision Sciences, School of Health Sciences, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - David I. Finkelstein
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Yuyi You
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Paul A. Haynes
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Ghasem H. Salekdeh
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Stuart L. Graham
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| |
Collapse
|
7
|
Qarawani A, Naaman E, Ben-Zvi Elimelech R, Harel M, Itzkovich C, Safuri S, Dahan N, Henkin J, Zayit-Soudry S. PEDF-derived peptide protects against Amyloid-β toxicity in vitro and prevents retinal dysfunction in rats. Exp Eye Res 2024; 242:109861. [PMID: 38522635 DOI: 10.1016/j.exer.2024.109861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 02/29/2024] [Accepted: 03/11/2024] [Indexed: 03/26/2024]
Abstract
Amyloid-beta (Aβ), a family of aggregation-prone and neurotoxic peptides, has been implicated in the pathophysiology of age-related macular degeneration (AMD). We have previously shown that oligomeric and fibrillar species of Aβ42 exerted retinal toxicity in rats, but while the consequences of exposure to amyloid were related to intracellular effects, the mechanism of Aβ42 internalization in the retina is not well characterized. In the brain, the 67 kDa laminin receptor (67LR) participates in Aβ-related neuronal cell death. A short peptide derived from pigment epithelium-derived factor (PEDF), formerly designated PEDF-335, was found to mitigate experimental models of ischemic retinopathy via targeting of 67LR. In the present study, we hypothesized that 67LR mediates the uptake of pathogenic Aβ42 assemblies in the retina, and that targeting of this receptor by PEDF-335 may limit the internalization of Aβ, thereby ameliorating its retinotoxicity. To test this assumption ARPE-19 cells in culture were incubated with PEDF-335 before treatment with fibrillar or oligomeric structures of Aβ42. Immunostaining confirmed that PEDF-335 treatment substantially prevented amyloid internalization into ARPE-19 cells and maintained their viability in the presence of toxic oligomeric and fibrillar Aβ42 entities in vitro. FRET competition assay was performed and confirmed the binding of PEDF-335 to 67LR in RPE-like cells. Wild-type rats were treated with intravitreal PEDF-335 in the experimental eye 2 days prior to administration of retinotoxic Aβ42 oligomers or fibrils to both eyes. Retinal function was assessed by electroretinography through 6 weeks post injection. The ERG responses in rats treated with oligomeric or fibrillar Aβ42 assemblies were near-normal in eyes previously treated with intravitreal PEDF-335, whereas those measured in the control eyes treated with injection of the Aβ42 assemblies alone showed pathologic attenuation of the retinal function through 6 weeks. The retinal presence of 67LR was determined ex vivo by immunostaining and western blotting. Retinal staining demonstrated the constitutional expression of 67LR mainly in the retinal nuclear layers. In the presence of Aβ42, the levels of 67LR were increased, although its retinal distribution remained largely unaltered. In contrast, no apparent differences in the retinal expression level of 67LR were noted following exposure to PEDF-335 alone, and its pattern of localization in the retina remained similarly concentrated primarily in the inner and outer nuclear layers. In summary, we found that PEDF-335 confers protection against Aβ42-mediated retinal toxicity, with significant effects noted in cells as well as in vivo in rats. The effects of PEDF-335 in the retina are potentially mediated via binding to 67LR and by at least partial inhibition of Aβ42 internalization. These results suggest that PEDF-335 may merit further consideration in the development of targeted inhibition of amyloid-related toxicity in the retina. More broadly, our observations provide evidence on the importance of extracellular versus intracellular Aβ42 in the retina and suggest concepts on the molecular mechanism of Aβ retinal pathogenicity.
Collapse
Affiliation(s)
- Amanda Qarawani
- Clinical Research Institute, Rambam Health Care Campus, Haifa, Israel; Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Efrat Naaman
- Clinical Research Institute, Rambam Health Care Campus, Haifa, Israel; Department of Ophthalmology, Rambam Health Care Campus, Haifa, Israel
| | - Rony Ben-Zvi Elimelech
- Clinical Research Institute, Rambam Health Care Campus, Haifa, Israel; Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Michal Harel
- Clinical Research Institute, Rambam Health Care Campus, Haifa, Israel; Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Chen Itzkovich
- Clinical Research Institute, Rambam Health Care Campus, Haifa, Israel; Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Shadi Safuri
- Clinical Research Institute, Rambam Health Care Campus, Haifa, Israel; Department of Ophthalmology, Rambam Health Care Campus, Haifa, Israel
| | - Nitsan Dahan
- Life Sciences and Engineering (LS&E) Infrastructure Center, Technion-Israel Institute of Technology, Haifa, Israel
| | - Jack Henkin
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, United States
| | - Shiri Zayit-Soudry
- Clinical Research Institute, Rambam Health Care Campus, Haifa, Israel; Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel; Department of Ophthalmology, Rambam Health Care Campus, Haifa, Israel.
| |
Collapse
|
8
|
Ban N, Shinojima A, Negishi K, Kurihara T. Drusen in AMD from the Perspective of Cholesterol Metabolism and Hypoxic Response. J Clin Med 2024; 13:2608. [PMID: 38731137 PMCID: PMC11084323 DOI: 10.3390/jcm13092608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/15/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Drusen are one of the most characteristic pathologies of precursor lesion of age-related macular degeneration (AMD). Drusen comprise a yellowish white substance that accumulates typically under the retinal pigment epithelium (RPE), and their constituents are lipids, complement, amyloid, crystallin, and others. In the past, many researchers have focused on drusen and tried to elucidate the pathophysiology of AMD because they believed that disease progression from early AMD to advanced AMD might be based on drusen or drusen might cause AMD. In fact, it is well established that drusen are the hallmark of precursor lesion of AMD and a major risk factor for AMD progression mainly based on their size and number. However, the existence of advanced AMD without drusen has long been recognized. For example, polypoidal choroidal vasculopathy (PCV), which comprises the majority of AMD cases in Asians, often lacks drusen. Thus, there is the possibility that drusen might be no more than a biomarker of AMD and not a cause of AMD. Now is the time to reconsider the relationship between AMD and drusen. In this review, we focus on early AMD pathogenesis based on basic research from the perspective of cholesterol metabolism and hypoxic response in the retina, and we discuss the role of drusen.
Collapse
Affiliation(s)
- Norimitsu Ban
- Laboratory of Aging and Retinal Biology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan; (A.S.); (K.N.)
| | - Ari Shinojima
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan; (A.S.); (K.N.)
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan; (A.S.); (K.N.)
| | - Toshihide Kurihara
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan; (A.S.); (K.N.)
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| |
Collapse
|
9
|
Latifi-Navid H, Barzegar Behrooz A, Jamehdor S, Davari M, Latifinavid M, Zolfaghari N, Piroozmand S, Taghizadeh S, Bourbour M, Shemshaki G, Latifi-Navid S, Arab SS, Soheili ZS, Ahmadieh H, Sheibani N. Construction of an Exudative Age-Related Macular Degeneration Diagnostic and Therapeutic Molecular Network Using Multi-Layer Network Analysis, a Fuzzy Logic Model, and Deep Learning Techniques: Are Retinal and Brain Neurodegenerative Disorders Related? Pharmaceuticals (Basel) 2023; 16:1555. [PMID: 38004422 PMCID: PMC10674956 DOI: 10.3390/ph16111555] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Neovascular age-related macular degeneration (nAMD) is a leading cause of irreversible visual impairment in the elderly. The current management of nAMD is limited and involves regular intravitreal administration of anti-vascular endothelial growth factor (anti-VEGF). However, the effectiveness of these treatments is limited by overlapping and compensatory pathways leading to unresponsiveness to anti-VEGF treatments in a significant portion of nAMD patients. Therefore, a system view of pathways involved in pathophysiology of nAMD will have significant clinical value. The aim of this study was to identify proteins, miRNAs, long non-coding RNAs (lncRNAs), various metabolites, and single-nucleotide polymorphisms (SNPs) with a significant role in the pathogenesis of nAMD. To accomplish this goal, we conducted a multi-layer network analysis, which identified 30 key genes, six miRNAs, and four lncRNAs. We also found three key metabolites that are common with AMD, Alzheimer's disease (AD) and schizophrenia. Moreover, we identified nine key SNPs and their related genes that are common among AMD, AD, schizophrenia, multiple sclerosis (MS), and Parkinson's disease (PD). Thus, our findings suggest that there exists a connection between nAMD and the aforementioned neurodegenerative disorders. In addition, our study also demonstrates the effectiveness of using artificial intelligence, specifically the LSTM network, a fuzzy logic model, and genetic algorithms, to identify important metabolites in complex metabolic pathways to open new avenues for the design and/or repurposing of drugs for nAMD treatment.
Collapse
Affiliation(s)
- Hamid Latifi-Navid
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran 1497716316, Iran; (H.L.-N.); (M.D.); (N.Z.); (S.P.); (S.T.); (Z.-S.S.)
- Departments of Ophthalmology and Visual Sciences and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Amir Barzegar Behrooz
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3T 2N2, Canada;
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran 1416634793, Iran
| | - Saleh Jamehdor
- Department of Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan 6517838636, Iran;
| | - Maliheh Davari
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran 1497716316, Iran; (H.L.-N.); (M.D.); (N.Z.); (S.P.); (S.T.); (Z.-S.S.)
| | - Masoud Latifinavid
- Department of Mechatronic Engineering, University of Turkish Aeronautical Association, 06790 Ankara, Turkey;
| | - Narges Zolfaghari
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran 1497716316, Iran; (H.L.-N.); (M.D.); (N.Z.); (S.P.); (S.T.); (Z.-S.S.)
| | - Somayeh Piroozmand
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran 1497716316, Iran; (H.L.-N.); (M.D.); (N.Z.); (S.P.); (S.T.); (Z.-S.S.)
| | - Sepideh Taghizadeh
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran 1497716316, Iran; (H.L.-N.); (M.D.); (N.Z.); (S.P.); (S.T.); (Z.-S.S.)
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Mahsa Bourbour
- Department of Biotechnology, Alzahra University, Tehran 1993893973, Iran;
| | - Golnaz Shemshaki
- Department of Studies in Zoology, University of Mysore, Manasagangothri, Mysore 570005, India;
| | - Saeid Latifi-Navid
- Department of Biology, Faculty of Sciences, University of Mohaghegh Ardabili, Ardabil 5619911367, Iran;
| | - Seyed Shahriar Arab
- Biophysics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 1411713116, Iran;
| | - Zahra-Soheila Soheili
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran 1497716316, Iran; (H.L.-N.); (M.D.); (N.Z.); (S.P.); (S.T.); (Z.-S.S.)
| | - Hamid Ahmadieh
- Ophthalmic Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran 1666673111, Iran;
| | - Nader Sheibani
- Departments of Ophthalmology and Visual Sciences and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
10
|
Hu Y, Chen J, Wang Y, Sun J, Huang P, Feng J, Liu T, Sun X. Fat mass and obesity-associated protein alleviates Aβ 1-40 induced retinal pigment epithelial cells degeneration via PKA/CREB signaling pathway. Cell Biol Int 2023; 47:584-597. [PMID: 36378581 DOI: 10.1002/cbin.11959] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 10/19/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022]
Abstract
Amyloid-β (Aβ) is thought to be a critical pathologic factor of retinal pigment epithelium (RPE) degeneration in age-related macular degeneration (AMD). Aβ induces inflammatory responses in RPE cells and recent studies demonstrate the N6-methyladenosine (m6A) regulatory role in RPE cell inflammation. m6A is a reversible epigenetic posttranslational modification, but its relationship with Aβ-induced RPE degeneration is yet to be thoroughly investigated. The present study explored the role and mechanism of m6A in Aβ-induced RPE degeneration model. This model was induced via intravitreally injecting oligomeric Aβ and the morphology of its retina was analyzed. One of m6A demethylases, the fat mass and obesity-associated (FTO) gene expression, was assessed. An m6A-messenger RNA (mRNA) epitranscriptomic microarray was employed for further bioinformatic analyses. It was confirmed that Aβ induced FTO upregulation within the RPE. Hypopigmentation alterations and structural disorganization were observed in Aβ-treated eyes, and inhibition of FTO exacerbated retinal degeneration and RPE impairment. Moreover, the m6A-mRNA epitranscriptomic microarray suggested that protein kinase A (PKA) was a target of FTO, and the PKA/cyclic AMP-responsive element binding (CREB) signaling pathway was involved in Aβ-induced RPE degeneration. m6A-RNA binding protein immunoprecipitation confirmed that FTO demethylated PKA within the RPE cells of Aβ-treated eyes. Altered expression of PKA and its downstream targets (CREB and brain-derived neurotrophic factor) was confirmed by quantitative reverse-transcription polymerase chain reaction and Western blot analyses. Hence, this study's findings shed light on FTO-mediated m6A modification in Aβ-induced RPE degeneration and indicate potential therapeutic targets for AMD.
Collapse
Affiliation(s)
- Yifan Hu
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), National Clinical Research Center for Ophthalmic Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,Department of Ophthalmology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, and Beijing Ophthalmology & Visual Sciences Key Lab, Beijing, China
| | - Jieqiong Chen
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), National Clinical Research Center for Ophthalmic Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Yuwei Wang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), National Clinical Research Center for Ophthalmic Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,Department of Ophthalmology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junran Sun
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), National Clinical Research Center for Ophthalmic Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Peirong Huang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), National Clinical Research Center for Ophthalmic Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Jingyang Feng
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), National Clinical Research Center for Ophthalmic Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Te Liu
- Central Laboratory, Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), National Clinical Research Center for Ophthalmic Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,Department of Ophthalmology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Tsai HR, Lo RY, Liang KH, Chen TL, Huang HK, Wang JH, Lee YC. Risk of Subsequent Dementia or Alzheimer Disease Among Patients With Age-Related Macular Degeneration: A Systematic Review and Meta-analysis. Am J Ophthalmol 2023; 247:161-169. [PMID: 36375591 DOI: 10.1016/j.ajo.2022.11.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022]
Abstract
PURPOSE Alzheimer disease (AD), a common form of dementia, shares several clinical and pathologic features with age-related macular degeneration (AMD). Epidemiologic reports on the association of AMD with subsequent dementia or AD are inconsistent. DESIGN Systematic review and meta-analysis. METHODS The Meta-analysis of Observational Studies in Epidemiology reporting guidelines were applied. The Newcastle-Ottawa Scale was used to evaluate the risk of bias in the included cohort studies that examined the association of AMD with subsequent dementia or AD. We estimated the pooled hazard ratios (HRs) of dementia or AD using random effects model meta-analysis and subgroup analysis on different follow-up periods, AMD subtype, gender, age, study design, and methods to ascertain dementia or AD. RESULTS A total of 8 223 581 participants were included in 8 studies published during 2000-2021. The meta-analysis showed that AMD was significantly associated with subsequent dementia (pooled HR 1.22, 95% CI 1.01-1.47) or AD (pooled HR 1.21, 95% CI 1.03-1.43). Our secondary analysis revealed that the association was more noticeable in dry AMD than wet AMD. CONCLUSIONS Patients with AMD have higher risks of developing dementia or AD, and therefore identifying related comorbidities and retinal biomarkers is much warranted for older adults with AMD in ophthalmologic practice.
Collapse
Affiliation(s)
- Hou-Ren Tsai
- From the Department of Ophthalmology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation (H.-R.T., Y.-C.L.), Hualien
| | - Raymond Y Lo
- Division of Cognitive/Geriatric Neurology, Department of Neurology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation and Tzu Chi University (R.Y.L.), Hualien; Institute of Medical Sciences, Tzu Chi University (R.Y.L.), Hualien
| | - Kai-Hsiang Liang
- Department of Medical Education, Medical Administration Office, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City
| | - Tai-Li Chen
- Center for Aging and Health, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation (T.-L.C.), Hualien; Department of Dermatology, Taipei Veterans General Hospital (T.-L.C.), Taipei
| | - Huei-Kai Huang
- Department of Family medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation (H.-K.H.), Hualien; Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation (H.-K.H., J.-H.W.), Hualien
| | - Jen-Hung Wang
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation (H.-K.H., J.-H.W.), Hualien
| | - Yuan-Chieh Lee
- From the Department of Ophthalmology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation (H.-R.T., Y.-C.L.), Hualien; Department of Ophthalmology and Visual Science, Tzu Chi University (Y.-C.L.), Hualien.
| |
Collapse
|
12
|
Arrigo A, Aragona E, Bandello F. The Role of Inflammation in Age-Related Macular Degeneration: Updates and Possible Therapeutic Approaches. Asia Pac J Ophthalmol (Phila) 2023; 12:158-167. [PMID: 36650098 DOI: 10.1097/apo.0000000000000570] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/22/2022] [Indexed: 01/19/2023] Open
Abstract
Age-related macular degeneration (AMD) is a common retinal disease characterized by complex pathogenesis and extremely heterogeneous characteristics. Both in "dry" and "wet" AMD forms, the inflammation has a central role to promote the degenerative process and to stimulate the onset of complications. AMD is characterized by several proinflammatory stimuli, cells and mediators involved, and metabolic pathways. Nowadays, inflammatory biomarkers may be unveiled and analyzed by means of several techniques, including laboratory approaches, histology, immunohistochemistry, and noninvasive multimodal retinal imaging. These methodologies allowed to perform remarkable steps forward for understanding the role of inflammation in AMD pathogenesis, also offering new opportunities to optimize the diagnostic workup of the patients and to develop new treatments. The main goal of the present paper is to provide an updated scenario of the current knowledge regarding the role of inflammation in "dry" and "wet" AMD and to discuss new possible therapeutic strategies.
Collapse
Affiliation(s)
- Alessandro Arrigo
- Department of Ophthalmology, IRCCS San Raffaele Scientific Institute, University Vita-Salute San Raffaele, Milan, Italy
| | | | | |
Collapse
|
13
|
Park KW, Joo JY, Kim ST. Comparison of brain-derived neurotrophic factor among subtypes of exudative age-related macular degeneration. Eur J Ophthalmol 2023; 33:408-414. [PMID: 35505604 DOI: 10.1177/11206721221099488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE To compare the levels of brain-derived neurotrophic factor (BDNF) in serum and aqueous humor (AH) in eyes with typical neovascular age-related macular degeneration (tAMD), polypoidal choroidal vasculopathy (PCV), and retinal angiomatous proliferation (RAP). METHODS This prospective study included 20 patients with tAMD, 20 patients with PCV, 20 patients with RAP, and 20 healthy controls. BDNF levels in the serum and AH were assessed using enzyme-linked immunosorbent assay. RESULTS Serum and AH BDNF levels were significantly lower in the age-related macular degeneration groups (tAMD, PCV, and RAP) than in the control group (p < 0.05). There was no significant difference in the mean BDNF levels in the serum and AH among the different nAMD subtypes (p = 0.538). CONCLUSIONS We confirmed that serum and AH BDNF levels were independent of the nAMD subtype.
Collapse
Affiliation(s)
- Keon Woo Park
- Department of Ophthalmology, School of Medicine, Chosun University, Gwang-ju, Republic of Korea
| | - Jung Yeon Joo
- Department of Pediatrics, 92203Ewha Womans University College of Medicine, Seoul, Republic of Korea
| | - Seong Taeck Kim
- Department of Ophthalmology, School of Medicine, Chosun University, Gwang-ju, Republic of Korea
| |
Collapse
|
14
|
Seyed Hosseini Fin N, Georgevsky D, Sukkar MB, Golzan SM. RAGE and its ligand amyloid beta promote retinal ganglion cell loss following ischemia-reperfusion injury. Front Cell Neurosci 2023; 17:1156084. [PMID: 37124398 PMCID: PMC10130520 DOI: 10.3389/fncel.2023.1156084] [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: 02/01/2023] [Accepted: 03/28/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction Glaucoma is a progressive neurodegenerative disease associated with age. Accumulation of amyloid-beta (Aß) proteins in the ganglion cell layer (GCL) and subsequent retinal ganglion cell (RGC) loss is an established pathological hallmark of the disease. The mechanism through which Aß provokes RGC loss remains unclear. The receptor for the advanced glycation end product (RAGE), and its ligand Aß, have been shown to mediate neuronal loss via internalizing Aß within the neurons. In this study, we investigated whether the RAGE-Aß axis plays a role in RGC loss in experimental glaucoma. Methods Retinal ischemia was induced by an acute elevation of intraocular pressure in RAGE-/- and wild-type (WT) control mice. In a subset of animals, oligomeric Aß was injected directly into the vitreous of both strains. RGC loss was assessed using histology and biochemical assays. Baseline and terminal positive scotopic threshold (pSTR) were also recorded. Results Retinal ischemia resulted in 1.9-fold higher RGC loss in WT mice compared to RAGE-/- mice (36 ± 3% p < 0.0001 vs. 19 ± 2%, p = 0.004). Intravitreal injection of oligomeric Aß resulted in 2.3-fold greater RGC loss in WT mice compared to RAGE-/- mice, 7-days post-injection (55 ± 4% p = 0.008 vs. 24 ± 2%, p = 0.02). We also found a significant decline in the positive scotopic threshold response (pSTR) amplitude of WT mice compared to RAGE-/- (36 ± 3% vs. 16 ± 6%). Discussion RAGE-/- mice are protected against RGC loss following retinal ischemia. Intravitreal injection of oligomeric Aß accelerated RGC loss in WT mice but not RAGE-/-. A co-localization of RAGE and Aß, suggests that RAGE-Aß binding may contribute to RGC loss.
Collapse
Affiliation(s)
- Nafiseh Seyed Hosseini Fin
- Vision Science Group, Graduate School of Health, University of Technology Sydney, Sydney, NSW, Australia
| | - Dana Georgevsky
- Vision Science Group, Graduate School of Health, University of Technology Sydney, Sydney, NSW, Australia
| | - Maria B. Sukkar
- Pharmacy Discipline, Graduate School of Health, University of Technology Sydney, Sydney, NSW, Australia
| | - S. Mojtaba Golzan
- Vision Science Group, Graduate School of Health, University of Technology Sydney, Sydney, NSW, Australia
- *Correspondence: S. Mojtaba Golzan,
| |
Collapse
|
15
|
Malek G, Campisi J, Kitazawa K, Webster C, Lakkaraju A, Skowronska-Krawczyk D. Does senescence play a role in age-related macular degeneration? Exp Eye Res 2022; 225:109254. [PMID: 36150544 PMCID: PMC10032649 DOI: 10.1016/j.exer.2022.109254] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 09/07/2022] [Accepted: 09/14/2022] [Indexed: 12/29/2022]
Abstract
Advanced age is the most established risk factor for developing age-related macular degeneration (AMD), one of the leading causes of visual impairment in the elderly, in Western and developed countries. Similarly, after middle age, there is an exponential increase in pathologic molecular and cellular events that can induce senescence, traditionally defined as an irreversible loss of the cells' ability to divide and most recently reported to also occur in select post-mitotic and terminally differentiated cells, such as neurons. Together these facts raise the question as to whether or not cellular senescence, may play a role in the development of AMD. A number of studies have reported the effect of ocular-relevant inducers of senescence using primarily in vitro models of poorly polarized, actively dividing retinal pigment epithelial (RPE) cell lines. However, in interpretating the data, the fidelity of these culture models to the RPE in vivo, must be considered. Fewer studies have explored the presence and/or impact of senescent cells in in vivo models that present with phenotypic features of AMD, leaving this an open field for further investigation. The goal of this review is to discuss current thoughts on the potential role of senescence in AMD development and progression, with consideration of the model systems used and their relevance to human disease.
Collapse
Affiliation(s)
- Goldis Malek
- Duke Eye Center, Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA; Department of Pathology, Duke University School of Medicine, Durham, NC, USA.
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA, USA; Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Koji Kitazawa
- Buck Institute for Research on Aging, Novato, CA, USA; Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Corey Webster
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Aparna Lakkaraju
- Departments of Ophthalmology and Anatomy, School of Medicine, University of California, San Francisco, CA, USA
| | - Dorota Skowronska-Krawczyk
- Department of Physiology and Biophysics, Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, University of California, Irvine, CA, USA
| |
Collapse
|
16
|
Askari S, Azizi F, Javadpour P, Karimi N, Ghasemi R. Endoplasmic reticulum stress as an underlying factor in leading causes of blindness and potential therapeutic effects of 4-phenylbutyric acid: from bench to bedside. EXPERT REVIEW OF OPHTHALMOLOGY 2022. [DOI: 10.1080/17469899.2022.2145945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Sahar Askari
- Neuroscience Research center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Azizi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Pegah Javadpour
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasser Karimi
- Eye and Skull Base Research Centers, The Five Senses Institute, Iran University of Medical Sciences, Tehran, Iran5Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rasoul Ghasemi
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
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: 32] [Impact Index Per Article: 10.7] [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
|
18
|
Cheung EYW, Chau ACM, Shea YF, Chiu PKC, Kwan JSK, Mak HKF. Level of Amyloid-β (Aβ) Binding Leading to Differential Effects on Resting State Functional Connectivity in Major Brain Networks. Biomedicines 2022; 10:biomedicines10092321. [PMID: 36140422 PMCID: PMC9496530 DOI: 10.3390/biomedicines10092321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/07/2022] [Accepted: 09/12/2022] [Indexed: 12/01/2022] Open
Abstract
Introduction: Amyloid-β protein (Aβ) is one of the biomarkers for Alzheimer’s disease (AD). The recent application of interhemispheric functional connectivity (IFC) in resting-state fMRI has been used as a non-invasive diagnostic tool for early dementia. In this study, we focused on the level of Aβ accumulated and its effects on the major functional networks, including default mode network (DMN), central executive network (CEN), salience network (SN), self-referential network (SRN) and sensory motor network (SMN). Methods: 58 participants (27 Hi Aβ (HiAmy) and 31 low Aβ (LowAmy)) and 25 healthy controls (HC) were recruited. [18F]flutemetamol PET/CT was performed for diseased groups, and MRI scanning was done for all participants. Voxel-by-voxel correlation analysis was done for both groups in all networks. Results: In HiAmy, IFC was reduced in all networks except SN. A negative correlation in DMN, CEN, SRN and SMN suggests high Aβ related to IFC reduction; However, a positive correlation in SN suggests high Aβ related to an increase in IFC. In LowAmy, IFC increased in CEN, SMN, SN and SRN. Positive correlation in all major brain networks. Conclusion: The level of Aβ accumulated demonstrated differential effects on IFC in various brain networks. As the treatment to reduce Aβ plaque deposition is available in the market, it may be an option for the HiAmy group to improve their IFC in major brain networks.
Collapse
Affiliation(s)
- Eva Y. W. Cheung
- Department of Diagnostic Radiology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong
- School of Medical Health and Sciences, Tung Wah College, 19/F, 31 Wylie Road, Ho Man Tin, Hong Kong
- Correspondence: (E.Y.W.C.); (H.K.F.M.)
| | - Anson C. M. Chau
- Medical Radiation Science, Allied Health and Human Performance Unit, University of South Australia, City East Campus, Bonython Jubilee Building, 1-26, Adelaide, SA 5001, Australia
| | - Yat-Fung Shea
- Division of Geriatrics, Department of Medicine, Queen Mary Hospital, Hong Kong
| | - Patrick K. C. Chiu
- Division of Geriatrics, Department of Medicine, Queen Mary Hospital, Hong Kong
| | - Joseph S. K. Kwan
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK
| | - Henry K. F. Mak
- Department of Diagnostic Radiology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong
- Alzheimer’s Disease Research Network, The University of Hong Kong, Hong Kong
- Correspondence: (E.Y.W.C.); (H.K.F.M.)
| |
Collapse
|
19
|
Sultan F, Parkin ET. The Amyloid Precursor Protein Plays Differential Roles in the UVA
Resistance and Proliferation of Human Retinal Pigment Epithelial Cells. Protein Pept Lett 2022; 29:313-327. [DOI: 10.2174/0929866529666220217124152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/08/2021] [Accepted: 12/13/2021] [Indexed: 11/22/2022]
Abstract
Background:
Age-related macular degeneration (AMD) can be characterised by
degeneration of retinal pigment epithelial (RPE) cells and the accumulation, in retinal drusen
deposits, of amyloid beta-peptides proteolytically derived, by secretases, from the amyloid precursor
protein (APP). Ultraviolet (UV) light exposure is a risk factor for the development of AMD.
Objectives:
In the current study, we investigated whether APP and/or its proteolysis are linked to the
UVA resistance or proliferation of ARPE-19 human RPE cells.
Methods:
Cell viability was determined, following UVA exposure, with prior small interfering
RNA-mediated APP depletion or secretase inhibitor treatments. APP levels/proteolysis were
analysed by immunoblotting. Cells were also grown in the presence/absence of secretase inhibitors
to assess their effects on longer-term culture growth. Finally, the effects of APP proteolytic
fragments on ARPE-19 cell proliferation were monitored following co-culture with human
embryonic kidney cells stably over-expressing these fragments.
Results:
Endogenous APP was depleted following UVA irradiation and β-secretase, but not α-
secretase, and the processing of the protein was reduced. Experimental APP depletion or γ-secretase
(but not α- or β-secretase) inhibition ablated the detrimental effect of UVA on cell viability. In
contrast, α-secretase, and possibly γ-secretase but not β-secretase activity, appeared to promote the
longer-term proliferation of ARPE-19 cells in the absence of UVA irradiation.
Conclusions:
There are clear but differential links between APP expression/proteolysis and the
proliferation and UVA resistance of ARPE-19 cells indicating that the protein should be
investigated further in relation to the identification of possible drug targets for the treatment of
AMD.
Collapse
Affiliation(s)
- Fatima Sultan
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, United
Kingdom
| | - Edward T. Parkin
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, United
Kingdom
| |
Collapse
|
20
|
Kim SY, Qian H. Comparison between sodium iodate and lipid peroxide murine models of age-related macular degeneration for drug evaluation-a narrative review. ANNALS OF EYE SCIENCE 2022; 7:8. [PMID: 37622161 PMCID: PMC10448775 DOI: 10.21037/aes-21-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
Objective In this review, non-transgenic models of age-related macular degeneration (AMD) are discussed, with focuses on murine retinal degeneration induced by sodium iodate and lipid peroxide (HpODE) as preclinical study platforms. Background AMD is the most common cause of vision loss in a world with an increasingly aging population. The major phenotypes of early and intermediate AMD are increased drusen and autofluorescence, Müller glia activation, infiltrated subretinal microglia and inward moving retinal pigment epithelium cells. Intermediate AMD may progress to advanced AMD, characterized by geography atrophy and/or choroidal neovascularization. Various transgenic and non-transgenic animal models related to retinal degeneration have been generated to investigate AMD pathogenesis and pathobiology, and have been widely used as potential therapeutic evaluation platforms. Methods Two retinal degeneration murine models induced by sodium iodate and HpODE are described. Distinct pathological features and procedures of these two models are compared. In addition, practical protocol and material preparation and assessment methods are elaborated. Conclusion Retina degeneration induced by sodium iodate and HpODE in mouse eye resembles many clinical aspects of human AMD and complimentary to the existent other animal models. However, standardization of procedure and assessment protocols is needed for preclinical studies. Further studies of HpODE on different routes, doses and species will be valuable for the future extensive use. Despite many merits of murine studies, differences between murine and human should be always considered.
Collapse
Affiliation(s)
- Soo-Young Kim
- Department of Pharmaceutics, Department of Biology, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Center for Nanomedicine, Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University of Medicine, Baltimore, MD, 21287, USA
| | - Haohua Qian
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
21
|
Flores‐Bellver M, Mighty J, Aparicio‐Domingo S, Li KV, Shi C, Zhou J, Cobb H, McGrath P, Michelis G, Lenhart P, Bilousova G, Heissel S, Rudy MJ, Coughlan C, Goodspeed AE, Becerra SP, Redenti S, Canto‐Soler MV. Extracellular vesicles released by human retinal pigment epithelium mediate increased polarised secretion of drusen proteins in response to AMD stressors. J Extracell Vesicles 2021; 10:e12165. [PMID: 34750957 PMCID: PMC8575963 DOI: 10.1002/jev2.12165] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 10/06/2021] [Accepted: 10/25/2021] [Indexed: 12/22/2022] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of blindness worldwide. Drusen are key contributors to the etiology of AMD and the ability to modulate drusen biogenesis could lead to therapeutic strategies to slow or halt AMD progression. The mechanisms underlying drusen biogenesis, however, remain mostly unknown. Here we demonstrate that under homeostatic conditions extracellular vesicles (EVs) secreted by retinal pigment epithelium (RPE) cells are enriched in proteins associated with mechanisms involved in AMD pathophysiology, including oxidative stress, immune response, inflammation, complement system and drusen composition. Furthermore, we provide first evidence that drusen-associated proteins are released as cargo of extracellular vesicles secreted by RPE cells in a polarised apical:basal mode. Notably, drusen-associated proteins exhibited distinctive directional secretion modes in homeostatic conditions and, differential modulation of this directional secretion in response to AMD stressors. These observations underpin the existence of a finely-tuned mechanism regulating directional apical:basal sorting and secretion of drusen-associated proteins via EVs, and its modulation in response to mechanisms involved in AMD pathophysiology. Collectively, our results strongly support an active role of RPE-derived EVs as a key source of drusen proteins and important contributors to drusen development and growth.
Collapse
Affiliation(s)
- Miguel Flores‐Bellver
- CellSight Ocular Stem Cell and Regeneration ProgramDepartment of OphthalmologySue Anschutz‐Rodgers Eye CenterUniversity of Colorado, School of MedicineAuroraColoradoUSA
| | - Jason Mighty
- Lehman CollegeBronxNew YorkUSA
- Biology Doctoral ProgramThe Graduate School and University CenterCity University of New YorkNew YorkNew YorkUSA
| | - Silvia Aparicio‐Domingo
- CellSight Ocular Stem Cell and Regeneration ProgramDepartment of OphthalmologySue Anschutz‐Rodgers Eye CenterUniversity of Colorado, School of MedicineAuroraColoradoUSA
| | - Kang V. Li
- CellSight Ocular Stem Cell and Regeneration ProgramDepartment of OphthalmologySue Anschutz‐Rodgers Eye CenterUniversity of Colorado, School of MedicineAuroraColoradoUSA
| | - Cui Shi
- Lehman CollegeBronxNew YorkUSA
- Biology Doctoral ProgramThe Graduate School and University CenterCity University of New YorkNew YorkNew YorkUSA
| | | | - Hannah Cobb
- CellSight Ocular Stem Cell and Regeneration ProgramDepartment of OphthalmologySue Anschutz‐Rodgers Eye CenterUniversity of Colorado, School of MedicineAuroraColoradoUSA
| | - Patrick McGrath
- Department of DermatologyUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - German Michelis
- Section of Protein Structure and FunctionNEINIHBethesdaMarylandUSA
| | - Patricia Lenhart
- CellSight Ocular Stem Cell and Regeneration ProgramDepartment of OphthalmologySue Anschutz‐Rodgers Eye CenterUniversity of Colorado, School of MedicineAuroraColoradoUSA
| | - Ganna Bilousova
- Department of DermatologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- Charles C. Gates Center for Regenerative MedicineUniversity of Colorado School of MedicineAuroraColoradoUSA
- Linda Crnic Institute for Down SyndromeUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Søren Heissel
- Proteomics Resource CenterThe Rockefeller UniversityNew YorkNew YorkUSA
| | - Michael J. Rudy
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Christina Coughlan
- University of Colorado Alzheimer's and Cognition CenterDepartment of NeurologyLinda Crnic Institute for Down SyndromeUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Andrew E. Goodspeed
- Department of PharmacologyUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
- University of Colorado Cancer CenterUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | | | - Stephen Redenti
- Lehman CollegeBronxNew YorkUSA
- Biology Doctoral ProgramThe Graduate School and University CenterCity University of New YorkNew YorkNew YorkUSA
- Biochemistry Doctoral ProgramThe Graduate SchoolCity University of New YorkNew YorkNew YorkUSA
| | - M. Valeria Canto‐Soler
- CellSight Ocular Stem Cell and Regeneration ProgramDepartment of OphthalmologySue Anschutz‐Rodgers Eye CenterUniversity of Colorado, School of MedicineAuroraColoradoUSA
- Charles C. Gates Center for Regenerative MedicineUniversity of Colorado School of MedicineAuroraColoradoUSA
| |
Collapse
|
22
|
Amyloid-Beta Peptides and Activated Astroglia Impairs Proliferation of Nerve Growth Factor Releasing Cells In Vitro: Implication for Encapsulated Cell Biodelivery-Mediated AD Therapy. Cells 2021; 10:cells10112834. [PMID: 34831056 PMCID: PMC8616486 DOI: 10.3390/cells10112834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/29/2021] [Accepted: 10/14/2021] [Indexed: 01/13/2023] Open
Abstract
Alzheimer’s disease (AD) treatment is constrained due to the inability of peripherally administered therapeutic molecules to cross the blood–brain barrier. Encapsulated cell biodelivery (ECB) devices, a tissue-targeted approach for local drug release, was previously optimized for human mature nerve growth factor (hmNGF) delivery in AD patients but was found to have reduced hmNGF release over time. To understand the reason behind reduced ECB efficacy, we exposed hmNGF-releasing cells (NGC0211) in vitro to human cerebrospinal fluid (CSF) obtained from Subjective Cognitive Impairment (SCI), Lewy Body Dementia (LBD), and AD patients. Subsequently, we exposed NGC0211 cells directly to AD-related factors like amyloid-β peptides (Aβ40/42) or activated astrocyte-conditioned medium (Aβ40/42/IL-1β/TNFα-treated) and evaluated biochemical stress markers, cell death indicators, cell proliferation marker (Ki67), and hmNGF release. We found that all patients’ CSF significantly reduced hmNGF release from NGC0211 cells in vitro. Aβ40/42, inflammatory molecules, and activated astrocytes significantly affected NGC0211 cell proliferation without altering hmNGF release or other parameters important for essential functions of the NGC0211 cells. Long-term constant cell proliferation within the ECB device is critically important to maintain a steady cell population needed for stable mNGF release. These data show hampered proliferation of NGC0211 cells, which may lead to a decline of the NGC0211 cell population in ECBs, thereby reducing hmNGF release. Our study highlights the need for future studies to strengthen ECB-mediated long-term drug delivery approaches.
Collapse
|
23
|
Nashine S. Potential Therapeutic Candidates for Age-Related Macular Degeneration (AMD). Cells 2021; 10:cells10092483. [PMID: 34572131 PMCID: PMC8464988 DOI: 10.3390/cells10092483] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/02/2021] [Accepted: 09/09/2021] [Indexed: 12/27/2022] Open
Abstract
Aging contributes to the risk of development of ocular diseases including, but not limited to, Age-related Macular Degeneration (AMD) that is a leading cause of blindness in the United States as well as worldwide. Retinal aging, that contributes to AMD pathogenesis, is characterized by accumulation of drusen deposits, alteration in the composition of Bruch’s membrane and extracellular matrix, vascular inflammation and dysregulation, mitochondrial dysfunction, and accumulation of reactive oxygen species (ROS), and subsequent retinal pigment epithelium (RPE) cell senescence. Since there are limited options available for the prophylaxis and treatment of AMD, new therapeutic interventions are constantly being looked into to identify new therapeutic targets for AMD. This review article discusses the potential candidates for AMD therapy and their known mechanisms of cytoprotection in AMD. These target therapeutic candidates include APE/REF-1, MRZ-99030, Ciliary NeuroTrophic Factor (CNTF), RAP1 GTPase, Celecoxib, and SS-31/Elamipretide.
Collapse
Affiliation(s)
- Sonali Nashine
- Department of Ophthalmology, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
24
|
Retinal Pigment Epithelium Expressed Toll-like Receptors and Their Potential Role in Age-Related Macular Degeneration. Int J Mol Sci 2021; 22:ijms22168387. [PMID: 34445096 PMCID: PMC8395065 DOI: 10.3390/ijms22168387] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/03/2021] [Accepted: 08/03/2021] [Indexed: 01/17/2023] Open
Abstract
(1) Background: Inflammation is a major pathomechanism in the development and progression of age-related macular degeneration (AMD). The retinal pigment epithelium (RPE) may contribute to retinal inflammation via activation of its Toll-like receptors (TLR). TLR are pattern recognition receptors that detect the pathogen- or danger-associated molecular pattern. The involvement of TLR activation in AMD is so far not understood. (2) Methods: We performed a systematic literature research, consulting the National Library of Medicine (PubMed). (3) Results: We identified 106 studies, of which 54 were included in this review. Based on these studies, the current status of TLR in AMD, the effects of TLR in RPE activation and of the interaction of TLR activated RPE with monocytic cells are given, and the potential of TLR activation in RPE as part of the AMD development is discussed. (4) Conclusion: The activation of TLR2, -3, and -4 induces a profound pro-inflammatory response in the RPE that may contribute to (long-term) inflammation by induction of pro-inflammatory cytokines, reducing RPE function and causing RPE cell degeneration, thereby potentially constantly providing new TLR ligands, which could perpetuate and, in the long run, exacerbate the inflammatory response, which may contribute to AMD development. Furthermore, the combined activation of RPE and microglia may exacerbate neurotoxic effects.
Collapse
|
25
|
Wang M, Su S, Jiang S, Sun X, Wang J. Role of amyloid β-peptide in the pathogenesis of age-related macular degeneration. BMJ Open Ophthalmol 2021; 6:e000774. [PMID: 34263061 PMCID: PMC8245440 DOI: 10.1136/bmjophth-2021-000774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/18/2021] [Indexed: 01/13/2023] Open
Abstract
Age-related macular degeneration (AMD) is the most common eye disease in elderly patients, which could lead to irreversible vision loss and blindness. Increasing evidence indicates that amyloid β-peptide (Aβ) might be associated with the pathogenesis of AMD. In this review, we would like to summarise the current findings in this field. The literature search was done from 1995 to Feb, 2021 with following keywords, ‘Amyloid β-peptide and age-related macular degeneration’, ‘Inflammation and age-related macular degeneration’, ‘Angiogenesis and age-related macular degeneration’, ‘Actin cytoskeleton and amyloid β-peptide’, ‘Mitochondrial dysfunction and amyloid β-peptide’, ‘Ribosomal dysregulation and amyloid β-peptide’ using search engines Pubmed, Google Scholar and Web of Science. Aβ congregates in subretinal drusen of patients with AMD and participates in the pathogenesis of AMD through enhancing inflammatory activity, inducing mitochondrial dysfunction, altering ribosomal function, regulating the lysosomal pathway, affecting RNA splicing, modulating angiogenesis and modifying cell structure in AMD. The methods targeting Aβ are shown to inhibit inflammatory signalling pathway and restore the function of retinal pigment epithelium cells and photoreceptor cells in the subretinal region. Targeting Aβ may provide a novel therapeutic strategy for AMD.
Collapse
Affiliation(s)
- Minwei Wang
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, USA
| | - Shiqi Su
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California, USA
| | - Shaoyun Jiang
- Stomatological Center, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Xinghuai Sun
- Fudan University Eye Ear Nose and Throat Hospital, Shanghai, China
| | - Jiantao Wang
- Shenzhen Eye Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
26
|
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: 27] [Impact Index Per Article: 6.8] [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
|
27
|
Wen LY, Wan L, Lai JN, Chen CS, Chen JJY, Wu MY, Hu KC, Chiu LT, Tien PT, Lin HJ. Increased risk of Alzheimer's disease among patients with age-related macular degeneration: A nationwide population-based study. PLoS One 2021; 16:e0250440. [PMID: 33961642 PMCID: PMC8104445 DOI: 10.1371/journal.pone.0250440] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/07/2021] [Indexed: 01/16/2023] Open
Abstract
OBJECTIVE This study aimed to investigate the risk of Alzheimer's disease among patients with age-related macular degeneration and its association with confounding comorbidities. METHOD This was a population-based, retrospective cohort study. By accessing data from the National Health Insurance Research Database of Taiwan, we identified 10,578 patients aged 50-100 years who were newly diagnosed with age-related macular degeneration between 2000 and 2012 and 10,578 non- age-related macular degeneration individuals. The comorbidities assessed were osteoporosis, diabetes, cirrhosis, cerebrovascular disease, chronic kidney disease, hypertension, hyperlipidemia, coronary artery disease, and chronic obstructive pulmonary disease. RESULTS Patients with age-related macular degeneration had a 1.23-fold increased risk of their condition advancing to Alzheimer's disease (aHR = 1.23, 95% CI = 1.04-1.46). The younger patients were diagnosed with age-related macular degeneration, the more likely patients got Alzheimer's disease (50-64 age group: aHR = 1.97, 95% CI = 1.04-3.73; 65-79 age group: aHR = 1.27, 95% CI = 1.02-1.58; 80-100 age group: aHR = 1.06, 95% CI = 0.78-1.45). In addition, there were significantly higher risks of Alzheimer's disease for patients with cirrhosis (aHR = 1.50, 95% CI = 1.09-2.06) in the age-related macular degeneration cohort than in the non-age-related macular degeneration cohort. CONCLUSION Patients with age-related macular degeneration may exhibit a higher risk of Alzheimer's disease than people without age-related macular degeneration.
Collapse
Affiliation(s)
- Li-Yen Wen
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Lei Wan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
- Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
| | - Jung-Nien Lai
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chih Sheng Chen
- Division of Chinese Medicine, Asia University Hospital, Taichung, Taiwan
| | - Jamie Jiin-Yi Chen
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Ophthalmology and Department of Molecular Genetics, China Medical University Hospital, Taichung, Taiwan
| | - Ming-Yen Wu
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Ophthalmology and Department of Molecular Genetics, China Medical University Hospital, Taichung, Taiwan
| | - Kai-Chieh Hu
- Management office for Health Data, China Medical University Hospital, Taichung, Taiwan
| | - Lu-Ting Chiu
- Management office for Health Data, China Medical University Hospital, Taichung, Taiwan
| | - Peng-Tai Tien
- Department of Ophthalmology and Department of Molecular Genetics, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Clinical Medical Science, College of Medicine, China Medical University, Taichung, Taiwan
| | - Hui-Ju Lin
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Ophthalmology and Department of Molecular Genetics, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
28
|
Thomas CN, Sim DA, Lee WH, Alfahad N, Dick AD, Denniston AK, Hill LJ. Emerging therapies and their delivery for treating age-related macular degeneration. Br J Pharmacol 2021; 179:1908-1937. [PMID: 33769566 DOI: 10.1111/bph.15459] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/11/2021] [Accepted: 03/14/2021] [Indexed: 12/13/2022] Open
Abstract
Age-related macular degeneration (AMD) is the most common cause of blindness in the Western world and is characterised in its latter stages by retinal cell death and neovascularisation and earlier stages with the loss of parainflammatory homeostasis. Patients with neovascular AMD (nAMD) are treated with frequent intraocular injections of anti-vascular endothelial growth factor (VEGF) therapies, which are not only unpopular with patients but carry risks of sight-threatening complications. A minority of patients are unresponsive with no alternative treatment available, and some patients who respond initially eventually develop a tolerance to treatment. New therapeutics with improved delivery methods and sustainability of clinical effects are required, in particular for non-neovascular AMD (90% of cases and no current approved treatments). There are age-related and disease-related changes that occur which can affect ocular drug delivery. Here, we review the latest emerging therapies for AMD, their delivery routes and implications for translating to clinical practice.
Collapse
Affiliation(s)
- Chloe N Thomas
- School of Biomedical Sciences, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Dawn A Sim
- Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK.,National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital and University College London Institute of Ophthalmology, London, UK
| | - Wen Hwa Lee
- Action Against AMD, London, UK.,Affordable Medicines Programme, Oxford Martin School, University of Oxford, Oxford, UK
| | - Nada Alfahad
- School of Biomedical Sciences, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Andrew D Dick
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital and University College London Institute of Ophthalmology, London, UK.,Academic Unit of Ophthalmology, Bristol Medical School and School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Alastair K Denniston
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital and University College London Institute of Ophthalmology, London, UK.,Academic Unit of Ophthalmology, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.,Department of Ophthalmology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,Centre for Patient Reported Outcome Research, Institute of Applied Health Research, University of Birmingham, Birmingham, UK.,Birmingham Health Partners Centre for Regulatory Science and Innovation, University of Birmingham, Birmingham, UK.,Health Data Research UK, London, UK
| | - Lisa J Hill
- School of Biomedical Sciences, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
29
|
Elman-Shina K, Elman M, Efrati S. Inflammatory seborrheic keratosis resolution after hyperbaric oxygen therapy: Case presentation and pathophysiology review. Dermatol Reports 2021; 13:8871. [PMID: 33824706 PMCID: PMC8018260 DOI: 10.4081/dr.2021.8871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 10/02/2020] [Indexed: 11/23/2022] Open
Abstract
Seborrheic keratosis (SK) is a common epidermal tumor, consisting of a benign proliferation of immature keratinocytes. The natural history of SK is a slow progression over time and complete remission is not expected. The article presents the first case of a complete resolution of a large (2.5 cm diameter) SK lesion after hyperbaric oxygen therapy (HBOT). In addition to the case presentation, the pathophysiology of SK and the potential beneficial physiological effects of HBOT are reviewed and discussed.
Collapse
Affiliation(s)
- Karin Elman-Shina
- Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf Harofeh) Medical Center, Zerifin
| | | | - Shai Efrati
- Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf Harofeh) Medical Center, Zerifin.,Sackler School of Medicine, Tel- Aviv University, Tel-Aviv.,Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
30
|
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
|
31
|
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
|
32
|
Sun J, Chen J, Li T, Huang P, Li J, Shen M, Gao M, Sun Y, Liang J, Li X, Wang Y, Xiao Y, Shi X, Hu Y, Feng J, Jia H, Liu T, Sun X. ROS production and mitochondrial dysfunction driven by PU.1-regulated NOX4-p22 phox activation in Aβ-induced retinal pigment epithelial cell injury. Am J Cancer Res 2020; 10:11637-11655. [PMID: 33052238 PMCID: PMC7546003 DOI: 10.7150/thno.48064] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/08/2020] [Indexed: 12/17/2022] Open
Abstract
Rationale: Amyloid β (Aβ) deposition, an essential pathological process in age-related macular degeneration (AMD), causes retinal pigment epithelium (RPE) degeneration driven mostly by oxidative stress. However, despite intense investigations, the extent to which overoxidation contributes to Aβ-mediated RPE damage and its potential mechanism has not been fully elucidated. Methods: We performed tandem mass-tagged (TMT) mass spectrometry (MS) and bioinformatic analysis of the RPE-choroid complex in an Aβ1-40-induced mouse model of retinal degeneration to obtain a comprehensive proteomic profile. Key regulators in this model were confirmed by reactive oxygen species (ROS) detection, mitochondrial ROS assay, oxygen consumption rate (OCR) measurement, gene knockout experiment, chromatin immunoprecipitation (ChIP), and luciferase assay. Results: A total of 4243 proteins were identified, 1069 of which were significantly affected by Aβ1-40 and found to be enriched in oxidation-related pathways by bioinformatic analysis. Moreover, NADPH oxidases were identified as hub proteins in Aβ1-40-mediated oxidative stress, as evidenced by mitochondrial dysfunction and reactive oxygen species overproduction. By motif and binding site analyses, we found that the transcription factor PU.1/Spi1 acted as a master regulator of the activation of NADPH oxidases, especially the NOX4-p22phox complex. Also, PU.1 silencing impeded RPE oxidative stress and mitochondrial dysfunction and rescued the retinal structure and function. Conclusion: Our study suggests that PU.1 is a novel therapeutic target for AMD, and the regulation of PU.1 expression represents a potentially novel approach against excessive oxidative stress in Aβ-driven RPE injury.
Collapse
|
33
|
Intracellular amyloid-β disrupts tight junctions of the retinal pigment epithelium via NF-κB activation. Neurobiol Aging 2020; 95:115-122. [PMID: 32795848 DOI: 10.1016/j.neurobiolaging.2020.07.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/07/2020] [Accepted: 07/14/2020] [Indexed: 12/16/2022]
Abstract
Drusen are focal deposits between the retinal pigment epithelium (RPE) and Bruch's membrane in the retina of patients with age-related macular degeneration. Amyloid-β is one of the important components of drusen, which leads to local inflammation. Furthermore, intracellular amyloid-β disrupts tight junctions of the RPE. However, the intracellular mechanisms linking intracellular amyloid-β and tight-junction disruption are not clear. In this study, intracellular amyloid-β oligomers activated nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) p65, leading to the disorganization of tight junctions of the RPE in mice after subretinal injection of amyloid-β. Amyloid-β also triggered NF-κB activation in the RPE cells in confluent culture, which was inhibited by the suppression of the advanced glycosylation end product-specific receptor. NF-κB inhibition by an IκB kinase inhibitor prevented the suppression of expression of tight-junction proteins, zonula occuludens-1 and occludin in RPE cells. In addition, tight-junction complexes remained intact in the RPE of mice with NF-κB inhibition, although there were intracellular amyloid-β oligomers. These data suggested that NF-κB inhibition might be a therapeutic approach to prevent amyloid-β-mediated tight-junction disruption.
Collapse
|
34
|
Gemenetzi M, Lotery AJ. Epigenetics in age-related macular degeneration: new discoveries and future perspectives. Cell Mol Life Sci 2020; 77:807-818. [PMID: 31897542 PMCID: PMC7058675 DOI: 10.1007/s00018-019-03421-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 12/04/2019] [Accepted: 12/10/2019] [Indexed: 12/13/2022]
Abstract
The study of epigenetics has explained some of the 'missing heritability' of age-related macular degeneration (AMD). The epigenome also provides a substantial contribution to the organisation of the functional retina. There is emerging evidence of specific epigenetic mechanisms associated with AMD. This 'AMD epigenome' may offer the chance to develop novel AMD treatments.
Collapse
Affiliation(s)
- M Gemenetzi
- NIHR Biomedical Research Centre At Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, 162 City Road, London, EC1V 2PD, UK
| | - A J Lotery
- Clinical and Experimental Sciences, Faculty of Medicine, University Hospital Southampton, University of Southampton, South Lab and Path Block, Mailpoint 806, Level D, Southampton, SO16 6YD, UK.
| |
Collapse
|
35
|
Pfeiffer RL, Marc RE, Jones BW. Persistent remodeling and neurodegeneration in late-stage retinal degeneration. Prog Retin Eye Res 2020; 74:100771. [PMID: 31356876 PMCID: PMC6982593 DOI: 10.1016/j.preteyeres.2019.07.004] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/15/2019] [Accepted: 07/18/2019] [Indexed: 02/06/2023]
Abstract
Retinal remodeling is a progressive series of negative plasticity revisions that arise from retinal degeneration, and are seen in retinitis pigmentosa, age-related macular degeneration and other forms of retinal disease. These processes occur regardless of the precipitating event leading to degeneration. Retinal remodeling then culminates in a late-stage neurodegeneration that is indistinguishable from progressive central nervous system (CNS) proteinopathies. Following long-term deafferentation from photoreceptor cell death in humans, and long-lived animal models of retinal degeneration, most retinal neurons reprogram, then die. Glial cells reprogram into multiple anomalous metabolic phenotypes. At the same time, survivor neurons display degenerative inclusions that appear identical to progressive CNS neurodegenerative disease, and contain aberrant α-synuclein (α-syn) and phosphorylated α-syn. In addition, ultrastructural analysis indicates a novel potential mechanism for misfolded protein transfer that may explain how proteinopathies spread. While neurodegeneration poses a barrier to prospective retinal interventions that target primary photoreceptor loss, understanding the progression and time-course of retinal remodeling will be essential for the establishment of windows of therapeutic intervention and appropriate tuning and design of interventions. Finally, the development of protein aggregates and widespread neurodegeneration in numerous retinal degenerative diseases positions the retina as a ideal platform for the study of proteinopathies, and mechanisms of neurodegeneration that drive devastating CNS diseases.
Collapse
Affiliation(s)
- Rebecca L Pfeiffer
- Dept of Ophthalmology, Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, USA.
| | - Robert E Marc
- Dept of Ophthalmology, Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, USA
| | - Bryan William Jones
- Dept of Ophthalmology, Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
36
|
Liu Y, Wei W, Baazaoui N, Liu F, Iqbal K. Inhibition of AMD-Like Pathology With a Neurotrophic Compound in Aged Rats and 3xTg-AD Mice. Front Aging Neurosci 2019; 11:309. [PMID: 31803044 PMCID: PMC6877482 DOI: 10.3389/fnagi.2019.00309] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 10/28/2019] [Indexed: 11/17/2022] Open
Abstract
Age-associated macular degeneration (AMD), which leads to loss of vision at its end stage, is one of the most common neurodegenerative diseases among the elderly. However, to date, no effective drug therapy is available for the prevention of AMD. Here, we report the occurrence of AMD pathology and its prevention by chronic treatment with the neurotrophic peptidergic compound P021, in aged rats and 3xTg-AD mice. We found photoreceptor degeneration, lipofuscin granules, vacuoles, and atrophy in retinal pigment epithelium (RPE) as well as Bruch’s membrane (BM) thickening; in aged rats, we even found rosette-like structure formation. Microgliosis and astrogliosis were observed in different retinal layers. In addition, we also found that total tau, phosphorylated tau, Aβ/APP, and VEGF were widely distributed in the sub-retina of aged rats and 3xTg mice. Importantly, chronic treatment with P021 for 3 months in rats and for 18 months in 3xTg mice ameliorated the pathological changes above. These findings indicate the therapeutic potential of P021 for prevention and treatment of AMD and retinal changes associated with aging and Alzheimer’s disease.
Collapse
Affiliation(s)
- Yinghua Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States.,Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Molecular Clinical Pharmacology, Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Wei Wei
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States.,Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, China
| | - Narjes Baazaoui
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States
| |
Collapse
|
37
|
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
|
38
|
Chu L, Xiao L, Xu B, Xu J. Dissociation of HKII in retinal epithelial cells induces oxidative stress injury in the retina. Int J Mol Med 2019; 44:1377-1387. [PMID: 31432102 PMCID: PMC6713434 DOI: 10.3892/ijmm.2019.4304] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 06/27/2019] [Indexed: 12/20/2022] Open
Abstract
The retina is sensitive to injury resulting from oxidative stress (OS) due to its high oxygen consumption. Patients with retinitis pigmentosa suffer from excessive OS. N‑acetylcysteine (NAC) is used as a mucolytic agent for the clinical treatment of disorders, such as chronic bronchitis and other pulmonary diseases. The aim of the present study was to investigate the role of hexokinase 2 (HKII) in retinal OS injury. Amyloid β (Aβ)1‑40 was used to establish a cellular model of OS. Cell viability was measured with a Cell Counting Kit‑8 assay, and the apoptosis, reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) of cells were analyzed via flow cytometry with corresponding kits. The mRNA and protein levels were detected by reverse transcription‑quantitative PCR and western blot analyses, respectively. It was observed that Aβ1‑40 reduced the expression of HKII in the mitochondria of retinal pigment epithelial ARPE cells and impaired mitochondrial antioxidant functions. Additionally, knockdown of HKII promoted apoptosis, and increased ROS levels and the MMP. NAC attenuated the inhibition of mitochondrial functions induced by Aβ1‑40. The knockdown of HKII was revealed to decrease the levels of Bcl‑2, manganese superoxide dismutase (SOD) and copper‑zinc‑SOD, and increase the levels of cleaved caspase‑3, Bax and cytochrome c. The present findings suggested that the dissociation of HKII induced by OS induces apoptosis and mitochondrial damage. This study provided improved understanding of the mechanisms underlying the effects of OS on retinal epithelial cells.
Collapse
Affiliation(s)
- Liqun Chu
- Department of Ophthalmology, Xiyuan Hospital, China Academy of Traditional Chinese Medicine, Beijing 100091, P.R. China
| | - Lin Xiao
- Department of Ophthalmology, Beijing Shijitan Hospital, CMU, Beijing 100038, P.R. China
| | - Bing Xu
- Department of Ophthalmology, Beijing Shijitan Hospital, CMU, Beijing 100038, P.R. China
| | - Jingmei Xu
- Department of Ophthalmology, Beijing Shijitan Hospital, CMU, Beijing 100038, P.R. China
| |
Collapse
|
39
|
Shoda C, Kitagawa Y, Shimada H, Yuzawa M, Tateno A, Okubo Y. Relationship of Area of Soft Drusen in Retina with Cerebral Amyloid-β Accumulation and Blood Amyloid-β Level in the Elderly. J Alzheimers Dis 2019; 62:239-245. [PMID: 29439351 DOI: 10.3233/jad-170956] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Histopathological studies have confirmed that soft drusen contains amyloid-β (Aβ). OBJECTIVE To examine the relationship between the area of soft drusen in the macular area and cerebral Aβ accumulation or plasma Aβ level in elderly persons without dementia. METHODS Fourteen consecutive patients (18 eyes) aged ≥50 years with macular soft drusen were studied prospectively. From color fundus photographs, the area of soft drusen (pixel) within a 6,000 μm diameter with the macula as center was measured. Standard uptake value ratio (SUVR) was obtained from positron emission tomography using florbetapir, which indicates the ratio of cerebral cortical-to-cerebellar Aβ accumulation. Ratio of plasma Aβ1-42 to Aβ1-40 level was calculated. RESULTS Mean age was 73.3±7.6 years. The soft drusen area was 4.32±2.42 mm2. The SUVR was 1.08±0.15. Plasma Aβ1-42/Aβ1-40 ratio was 0.17±0.08. When SUVR ≥1.10 was defined as positive and <1.10 as negative, the soft drusen area in SUVR-positive patients (6.19±1.14 mm2) was significantly (p = 0.0043) larger than that in SUVR-negative patients (3.13±2.27 mm2). Multivariate regression analysis showed that SUVR positivity correlated with soft drusen area (p = 0.0484) and with Voxel-based Specific Regional Analysis System for Alzheimer's Disease score (p = 0.0360). However, there was no correlation with gender (p = 0.1921), age (p = 0.2361), Alzheimer's Disease Assessment Scale score (p = 0.6310), Mini-Mental State Examination score (p = 0.4246), or plasma Aβ1-42/Aβ1-40 ratio (p = 0.8398). CONCLUSION Among elderly persons without dementia, the area of soft drusen was larger in those with more extensive cerebral Aβ accumulation. The area of soft drusen may be a biomarker of cerebral Aβ accumulation.
Collapse
Affiliation(s)
- Chiho Shoda
- Department of Ophthalmology, School of Medicine, Nihon University, Chiyoda-ku, Tokyo, Japan
| | - Yorihisa Kitagawa
- Department of Ophthalmology, School of Medicine, Nihon University, Chiyoda-ku, Tokyo, Japan
| | - Hiroyuki Shimada
- Department of Ophthalmology, School of Medicine, Nihon University, Chiyoda-ku, Tokyo, Japan
| | - Mitsuko Yuzawa
- Department of Ophthalmology, School of Medicine, Nihon University, Chiyoda-ku, Tokyo, Japan
| | - Amane Tateno
- Department of Neuropsychiatry, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Yoshiro Okubo
- Department of Neuropsychiatry, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
40
|
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
|
41
|
Pead E, Megaw R, Cameron J, Fleming A, Dhillon B, Trucco E, MacGillivray T. Automated detection of age-related macular degeneration in color fundus photography: a systematic review. Surv Ophthalmol 2019; 64:498-511. [PMID: 30772363 PMCID: PMC6598673 DOI: 10.1016/j.survophthal.2019.02.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 01/31/2019] [Accepted: 02/04/2019] [Indexed: 12/13/2022]
Abstract
The rising prevalence of age-related eye diseases, particularly age-related macular degeneration, places an ever-increasing burden on health care providers. As new treatments emerge, it is necessary to develop methods for reliably assessing patients' disease status and stratifying risk of progression. The presence of drusen in the retina represents a key early feature in which size, number, and morphology are thought to correlate significantly with the risk of progression to sight-threatening age-related macular degeneration. Manual labeling of drusen on color fundus photographs by a human is labor intensive and is where automatic computerized detection would appreciably aid patient care. We review and evaluate current artificial intelligence methods and developments for the automated detection of drusen in the context of age-related macular degeneration.
Collapse
Affiliation(s)
- Emma Pead
- VAMPIRE Project, Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, Scotland.
| | - Roly Megaw
- Princess Alexandra Eye Pavilion, Edinburgh, Scotland
| | - James Cameron
- MRC Human Genetics Unit, The University of Edinburgh, Edinburgh, Scotland
| | - Alan Fleming
- Optos plc, Queensferry House, Carnegie Campus, Dunfermline
| | | | - Emanuele Trucco
- VAMPIRE Project, Computing (School of Science and Engineering), University of Dundee, UK
| | - Thomas MacGillivray
- VAMPIRE Project, Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, Scotland
| |
Collapse
|
42
|
Huang P, Sun J, Wang F, Luo X, Zhu H, Gu Q, Sun X, Liu T, Sun X. DNMT1 and Sp1 competitively regulate the expression of BACE1 in A2E-mediated photo-oxidative damage in RPE cells. Neurochem Int 2018; 121:59-68. [PMID: 30273642 DOI: 10.1016/j.neuint.2018.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 09/02/2018] [Accepted: 09/03/2018] [Indexed: 02/03/2023]
Abstract
Numerous studies have focused on the deteriorate role of amyloid-β (Aβ) on retina, implying the potential pathogenic mechanism underlying age-related macular degeneration (AMD). However, the mechanism underlying the Aβ deposition in AMD patients remains unknown. Beta-site amyloid precursor protein-cleaving enzyme 1 (BACE1), rate-limiting enzyme for Aβ production, plays an important role in Aβ deposition in the brain. In the current study, we aimed to clarify the regulation mechanism of BACE1 and explore potential drug targets using a lipofuscinfluorophore A2E-mediated photo-oxidation model. In this model, Aβ1-40 and Aβ1-42 levels increased simultaneously with the enhanced BACE1 expression. These changes were associated with the hypomethylation of specific loci within the BACE1 gene promoter and the decreased levels of DNA methyltransferase 1 (DNMT1). Furthermore, we noticed overlapping regions of differentially methylated CpG islands and specificity protein (Sp1) binding sites within the BACE1 promoter. We employed chromatin immunoprecipitation (ChIP) assay to verify that the decreased BACE1 promoter methylation by DNMT1 enabled increased binding between Sp1 and the BACE1 promoter, which further enhanced BACE1 transcription. The inhibition of Sp1 with mithramycin A (MTM) could down-regulate the expression of BACE1 as well as alleviate the RPE barrier morphology and function impairment. Our results for the first time show the competitive regulation of BACE1 by transcription factor Sp1 and DNMT1 after photo-oxidation and confirm the potential novel protective role of MTM on RPE cells.
Collapse
Affiliation(s)
- Peirong Huang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China; Shanghai Key Laboratory of Fundus Disease, Shanghai, People's Republic of China
| | - Junran Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China; Shanghai Key Laboratory of Fundus Disease, Shanghai, People's Republic of China
| | - Fenghua Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China; Shanghai Key Laboratory of Fundus Disease, Shanghai, People's Republic of China
| | - Xueting Luo
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China; Shanghai Key Laboratory of Fundus Disease, Shanghai, People's Republic of China
| | - Hong Zhu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China; Shanghai Key Laboratory of Fundus Disease, Shanghai, People's Republic of China
| | - Qing Gu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China; Shanghai Key Laboratory of Fundus Disease, Shanghai, People's Republic of China
| | - Xiangjun Sun
- School of Biology and Agriculture, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Te Liu
- Department of Pathology, Yale University School of Medicine, New Haven, USA; Shanghai Geriatric Institute of Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China.
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China; Shanghai Key Laboratory of Fundus Disease, Shanghai, People's Republic of China.
| |
Collapse
|
43
|
Farjood F, Vargis E. Novel devices for studying acute and chronic mechanical stress in retinal pigment epithelial cells. LAB ON A CHIP 2018; 18:3413-3424. [PMID: 30328441 DOI: 10.1039/c8lc00659h] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Choroidal neovascularization (CNV) is a major cause of blindness in patients with age-related macular degeneration (AMD). Overexpression of vascular endothelial growth factor (VEGF), a potent angiogenic protein, by retinal pigment epithelial (RPE) cells is a key stimulator of CNV. Mechanical stress occurs during different stages of AMD and is a possible inducer of VEGF expression in RPE cells. However, robust and realistic approaches to studying acute and chronic mechanical stress under various AMD stages do not exist. The majority of previous work has studied cyclic stretching of RPE cells grown on flexible substrates, but an ideal model must be able to mimic localized and continuous stretching of the RPE as would occur in AMD in vivo. To bridge this gap, we developed two in vitro devices to model chronic and acute mechanical stress on RPE cells during different stages of AMD. In one device, high levels of continuous mechanical stress were applied to focal regions of the RPE monolayer by stretching the underlying silicon substrate to study the role of chronic mechanical stimulation. In the second device, RPE cells were grown on porous plastic substrates and acute stress was studied by stretching small areas. Using these devices, we studied the effect of mechanical stress on VEGF expression in RPE cells. Our results suggest that mechanical stress in RPE cells induces VEGF expression and promotes in vitro angiogenesis. These results confirm the hypothesis that mechanical stress is involved in the initiation and progression of CNV.
Collapse
Affiliation(s)
- Farhad Farjood
- Department of Biological Engineering, Utah State University, 4105 Old Main Hill, Logan, UT 84322, USA.
| | | |
Collapse
|
44
|
Cerquera-Jaramillo MA, Nava-Mesa MO, González-Reyes RE, Tellez-Conti C, de-la-Torre A. Visual Features in Alzheimer's Disease: From Basic Mechanisms to Clinical Overview. Neural Plast 2018; 2018:2941783. [PMID: 30405709 PMCID: PMC6204169 DOI: 10.1155/2018/2941783] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 08/07/2018] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia worldwide. It compromises patients' daily activities owing to progressive cognitive deterioration, which has elevated direct and indirect costs. Although AD has several risk factors, aging is considered the most important. Unfortunately, clinical diagnosis is usually performed at an advanced disease stage when dementia is established, making implementation of successful therapeutic interventions difficult. Current biomarkers tend to be expensive, insufficient, or invasive, raising the need for novel, improved tools aimed at early disease detection. AD is characterized by brain atrophy due to neuronal and synaptic loss, extracellular amyloid plaques composed of amyloid-beta peptide (Aβ), and neurofibrillary tangles of hyperphosphorylated tau protein. The visual system and central nervous system share many functional components. Thus, it is plausible that damage induced by Aβ, tau, and neuroinflammation may be observed in visual components such as the retina, even at an early disease stage. This underscores the importance of implementing ophthalmological examinations, less invasive and expensive than other biomarkers, as useful measures to assess disease progression and severity in individuals with or at risk of AD. Here, we review functional and morphological changes of the retina and visual pathway in AD from pathophysiological and clinical perspectives.
Collapse
Affiliation(s)
| | - Mauricio O. Nava-Mesa
- Grupo de Investigación en Neurociencias (NeURos), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Rodrigo E. González-Reyes
- Grupo de Investigación en Neurociencias (NeURos), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Carlos Tellez-Conti
- Escuela Superior de Oftalmología-Instituto Barraquer de América, Bogotá, Colombia
| | - Alejandra de-la-Torre
- Grupo de Investigación en Neurociencias (NeURos), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
45
|
Jun S, Datta S, Wang L, Pegany R, Cano M, Handa JT. The impact of lipids, lipid oxidation, and inflammation on AMD, and the potential role of miRNAs on lipid metabolism in the RPE. Exp Eye Res 2018; 181:346-355. [PMID: 30292489 DOI: 10.1016/j.exer.2018.09.023] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 08/31/2018] [Accepted: 09/30/2018] [Indexed: 12/17/2022]
Abstract
The accumulation of lipids within drusen, the epidemiologic link of a high fat diet, and the identification of polymorphisms in genes involved in lipid metabolism that are associated with disease risk, have prompted interest in the role of lipid abnormalities in AMD. Despite intensive investigation, our understanding of how lipid abnormalities contribute to AMD development remains unclear. Lipid metabolism is tightly regulated, and its dysregulation can trigger excess lipid accumulation within the RPE and Bruch's membrane. The high oxidative stress environment of the macula can promote lipid oxidation, impairing their original function as well as producing oxidation-specific epitopes (OSE), which unless neutralized, can induce unwanted inflammation that additionally contributes to AMD progression. Considering the multiple layers of lipid metabolism and inflammation, and the ability to simultaneously target multiple pathways, microRNA (miRNAs) have emerged as important regulators of many age-related diseases including atherosclerosis and Alzheimer's disease. These diseases have similar etiologic characteristics such as lipid-rich deposits, oxidative stress, and inflammation with AMD, which suggests that miRNAs might influence lipid metabolism in AMD. In this review, we discuss the contribution of lipids to AMD pathobiology and introduce how miRNAs might affect lipid metabolism during lesion development. Establishing how miRNAs contribute to lipid accumulation in AMD will help to define the role of lipids in AMD, and open new treatment avenues for this enigmatic disease.
Collapse
Affiliation(s)
- Sujung Jun
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, 21287, United States
| | - Sayantan Datta
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, 21287, United States
| | - Lei Wang
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, 21287, United States
| | - Roma Pegany
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, 21287, United States
| | - Marisol Cano
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, 21287, United States
| | - James T Handa
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, 21287, United States.
| |
Collapse
|
46
|
Ocular amyloid imaging at the crossroad of Alzheimer's disease and age-related macular degeneration: implications for diagnosis and therapy. J Neurol 2018; 266:1566-1577. [PMID: 30155741 DOI: 10.1007/s00415-018-9028-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/18/2018] [Accepted: 08/20/2018] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) and age-related macular degeneration (AMD) are important disorders of aging, but significant challenges remain in diagnosis and therapy. Amyloid-beta (Aβ), found in the brain and a defining feature of AD, has also been observed in the retina in both AD and AMD. While current diagnostic modalities for detecting Aβ in the brain are costly or invasive, Aβ in the retina can be noninvasively and conveniently imaged using modern photonic imaging systems such as optical coherence tomography (OCT). Moreover, since many of these retinal changes occur before degenerative changes can be detected in the brain, ocular amyloid biomarkers could be utilized to detect AD as well as AMD in their earliest stages when therapy may be most effective in halting disease progression. Novel technologies to quantify retinal biomarkers have the potential to facilitate early diagnosis and noninvasive monitoring of disease progression with important therapeutic implications.
Collapse
|
47
|
Schottler J, Randoll N, Lucius R, Caliebe A, Roider J, Klettner A. Long-term treatment with anti-VEGF does not induce cell aging in primary retinal pigment epithelium. Exp Eye Res 2018. [DOI: 10.1016/j.exer.2018.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
48
|
Inanc Tekin M, Sekeroglu MA, Demirtas C, Tekin K, Doguizi S, Bayraktar S, Yilmazbas P. Brain-Derived Neurotrophic Factor in Patients With Age-Related Macular Degeneration and Its Correlation With Retinal Layer Thicknesses. Invest Ophthalmol Vis Sci 2018; 59:2833-2840. [PMID: 30025135 DOI: 10.1167/iovs.18-24030] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To determine brain-derived neurotrophic factor (BDNF) levels in serum and aqueous humor (AH) and to assess the relationship between BDNF levels and retinal layer thicknesses in age-related macular degeneration (AMD). METHODS A total of 48 AMD patients (AMD group) that was composed of twenty-three nonexudative and 25 exudative patients and 26 control subjects (control group) were included in the study. Serum and AH BDNF levels were assessed by ELISA method. Retinal layer thicknesses were calculated by segmentation analysis of optical coherence tomography. RESULTS The mean BDNF levels in AH were found to be significantly lower in both the nonexudative and exudative AMD groups than in the control group (P = 0.003 and P < 0.001, respectively). Optical coherence tomography segmentation analysis revealed that the total average retina pigment epithelium thickness was statistically significantly thinner in the nonexudative AMD group compared with the exudative AMD and control groups (P = 0.001 and P = 0.040, respectively). The total average outer nuclear layer (ONL) thicknesses of nonexudative and exudative AMD cases were reduced compared to control group; however, the decrement was statistically significant only in the nonexudative AMD group (P = 0.009). In the correlation analysis of BDNF levels with retinal layer thicknesses, statistically significant correlations exist between BDNF levels of AH with ONL thicknesses in cases of AMD and with retina pigment epithelium thicknesses in the nonexudative AMD group. CONCLUSIONS BDNF concentrations in AH decreased in the AMD group and this decrease correlates with outer retinal layer thicknesses. Low BDNF levels detected in the AMD group may be insufficient to protect the photoreceptors, resulting in thinning of ONL.
Collapse
Affiliation(s)
| | | | - Canan Demirtas
- Department of Medical Biochemistry, Gazi University, Ankara, Turkey
| | - Kemal Tekin
- Department of Ophthalmology, Kars State Hospital, Kars, Turkey
| | - Sibel Doguizi
- Ankara Ulucanlar Eye Training and Research Hospital, Ankara, Turkey
| | - Serdar Bayraktar
- Ankara Ulucanlar Eye Training and Research Hospital, Ankara, Turkey
| | - Pelin Yilmazbas
- Ankara Ulucanlar Eye Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
49
|
Lashkari K, Teague G, Chen H, Lin YQ, Kumar S, McLaughlin MM, López FJ. A monoclonal antibody targeting amyloid β (Aβ) restores complement factor I bioactivity: Potential implications in age-related macular degeneration and Alzheimer's disease. PLoS One 2018; 13:e0195751. [PMID: 29782502 PMCID: PMC5962057 DOI: 10.1371/journal.pone.0195751] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/28/2018] [Indexed: 11/18/2022] Open
Abstract
Activation of the alternative complement cascade has been implicated in the pathogenesis of age related macular degeneration (AMD) and Alzheimer’s disease (AD). Amyloid β (Aβ), a component of drusen, may promote complement activation by inhibiting CFI bioactivity. We determined whether Aβ reduced CFI bioactivity and whether antibodies against Aβ including a monoclonal antibody, GSK933776 could restore CFI bioactivity. We also measured CFI bioactivity in plasma of subjects with AMD and AD. In support of the GSK933776 development program in AMD (geographic atrophy), we developed a quantitative assay to measure CFI bioactivity based on its ability to cleave C3b to iC3b, and repeated it in presence or absence of Aβ and anti-Aβ antibodies. Using this assay, we measured CFI bioactivity in plasma of 194 subjects with AMD, and in samples from subjects with AD that had been treated with GSK933776 as part of the GSK933776 development program in AD. Aβ reduced the CFI bioactivity by 5-fold and pre-incubation with GSK933776 restored CFI bioactivity. In subjects with AMD, plasma CFI levels and bioactivity were not significantly different from non-AMD controls. However, we detected a positive linear trend, suggesting increasing activity with disease severity. In subjects with AD, we observed a 10% and 27% increase in overall CFI bioactivity after treatment with GSK933776 during the second and third dose. Our studies indicate that CFI enzymatic activity can be inhibited by Aβ and be altered in proinflammatory diseases such as AMD and AD, in which deposition of Aβ and activation of the alternative complement cascade are believed to play a key role in the disease process.
Collapse
Affiliation(s)
- Kameran Lashkari
- Schepens Eye Research Institute, Mass Eye & Ear, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| | - Gianna Teague
- Schepens Eye Research Institute, Mass Eye & Ear, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Hong Chen
- Alliance Pharma, Malvern, Pennsylvania, United States of America
| | - Yong-Qing Lin
- Alliance Pharma, Malvern, Pennsylvania, United States of America
| | - Sanjay Kumar
- Alternative Discovery & Development, GlaxoSmithKline, King of Prussia, Pennsylvania, United States of America
| | - Megan M. McLaughlin
- Alternative Discovery & Development, GlaxoSmithKline, King of Prussia, Pennsylvania, United States of America
| | - Francisco J. López
- Alternative Discovery & Development, GlaxoSmithKline, King of Prussia, Pennsylvania, United States of America
| |
Collapse
|
50
|
Dong ZZ, Li J, Gan YF, Sun XR, Leng YX, Ge J. Amyloid beta deposition related retinal pigment epithelium cell impairment and subretinal microglia activation in aged APPswePS1 transgenic mice. Int J Ophthalmol 2018; 11:747-755. [PMID: 29862171 DOI: 10.18240/ijo.2018.05.06] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 03/21/2018] [Indexed: 11/23/2022] Open
Abstract
AIM To identify the pathological role of amyloid beta (Aβ) deposition in retinal degeneration, and explore Aβ deposition on the retinal pigment epithelium cells (RPE) layer and the associated structural and functional changes in Alzheimer's disease transgenic mice. METHODS RPE changes in the eyes of APPswe/PS1 transgenic and none transgenic (NTG) mice over 20 months old were examined. Histological changes were investigated via hematoxylin and eosin (H&E) staining and transmission electron microscopy (TEM) examination, whereas the expression of amyloid precursor protein (APP), Aβ, Zonula occludens-1 (ZO-1) and Ionized calcium binding adaptor molecule-1 (IBA-1) were investigated using immunohistochemistry and immunofluorescence techniques. All of the obtained results were quantitatively and statistically analyzed. RESULTS In aged transgenic mice, an APP-positive immunoreaction and Aβ deposition were detected on the RPE layer but were undetectable in NTG mice. The RPE demonstrated some vacuole changes, shortened basal infoldings and basal deposition in histopathological examination and TEM tests, wherein irregular shapes were indicated by ZO-1 disorganization through fluorescence. Furthermore, IBA-1 positive cells were observed to have accumulated and infiltrated into the RPE layer and localized beneath the RPE/Bruch's membrane (BrM) complex, which was accompanied by an increase in BrM thickness in aged transgenic mice in comparison to NTG mice. The IBA-1 positive cells were found to be co-stained with Aβ deposition on the RPE flat mounts. CONCLUSION The observed Aβ deposition in the RPE layer may cause RPE dysfunction, which is associated with microglia cells infiltration into the retina of aged transgenic mice, suggesting that Aβ deposition probably plays a significant role in RPE-related degenerative disease.
Collapse
Affiliation(s)
- Zhi-Zhang Dong
- The 2nd Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325200, Zhejiang Province, China
| | - Juan Li
- Xi'an No.4 Hospital, Xi'an 710000, Shaanxi Province, China
| | - Yi-Feng Gan
- The 2nd Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325200, Zhejiang Province, China
| | - Xue-Rong Sun
- Institute of Aging Research, Guangdong Medical College, Dongguan 523000, Guangdong Province, China
| | - Yun-Xia Leng
- Guangzhou First Municipal People's Hospital, Guangzhou 510060, Guangdong Province, China
| | - Jian Ge
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| |
Collapse
|