1
|
Liu GM, Liu Y. Exosomes derived from human umbilical cord blood mesenchymal stem cells protect against blue light-induced damage to retinal pigment epithelial cells by inhibiting FGF2 expression. Cytotechnology 2025; 77:88. [PMID: 40225792 PMCID: PMC11982010 DOI: 10.1007/s10616-025-00752-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 07/17/2024] [Indexed: 04/15/2025] Open
Abstract
Age-related macular degeneration (AMD) is a debilitating retinal disorder that may lead to progressive vision loss. One contributing factor to AMD pathogenesis is excessive blue light (BL) exposure. In this study, we investigated the therapeutic potential of exosomes derived from human umbilical cord blood mesenchymal stem cells (hUCMSC-EXs) in addressing BL-induced damage to ARPE-19 human retinal pigment epithelial (RPE) cells and explored the underlying mechanisms. Our findings revealed that BL exposure induced morphological alterations in ARPE-19 cells, accompanied by a time-dependent decline in cell viability, increased apoptosis, heightened oxidative stress, and inflammatory responses; however, hUCMSC-EXs dose-dependently mitigated BL-induced ARPE-19 cell damage. Interestingly, hUCMSC-EXs were found to suppress the upregulation of fibroblast growth factor 2 (FGF2) in BL-exposed ARPE-19 cells. Furthermore, FGF2 overexpression partially counteracted the inhibitory effects of hUCMSC-EXs on FGF2 expression and compromised the protective benefits of hUCMSC-EXs against BL-induced ARPE-19 cell damage. In conclusion, our results suggest that hUCMSC-EXs shield ARPE-19 cells from BL-induced harm by inhibiting FGF2 expression.
Collapse
Affiliation(s)
- Guang-ming Liu
- Department of Ophthalmology, The First People’s Hospital of Changzhou, 185 Juqian Street, Tianning District, Changzhou, 213000 Jiangsu China
| | - Yan Liu
- Department of Ophthalmology, The First People’s Hospital of Changzhou, 185 Juqian Street, Tianning District, Changzhou, 213000 Jiangsu China
| |
Collapse
|
2
|
Josifova T, Konieczka K, Schötzau A, Flammer J. The effect of a specific vitamin supplement containing L-methylfolate (Ocufolin forte) in patients with neovascular age-related macular degeneration. ADVANCES IN OPHTHALMOLOGY PRACTICE AND RESEARCH 2025; 5:135-141. [PMID: 40276027 PMCID: PMC12020837 DOI: 10.1016/j.aopr.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 04/26/2025]
Abstract
Background Patients with nAMD often have pathologically elevated homocysteine (Hcy) and increased retinal venous pressure (RVP). We tested whether the administration of a specific vitamin preparation containing L-methylfolate (Ocufolin forte) as an addition to anti-VEGF therapy reduces these two risk factors and favorably influences the disease course. Methods A total of 27 eyes/27 patients with intra- and subretinal fluid, Hcy above 12 μmol/L, RVP of at least 8 mm above the IOP, and an IOP between 10 and 20 mmHg were included in this study. All eyes received three injections of 0.05 ml aflibercept at one-month intervals as clinically indicated. Fifteen patients additionally received one capsule of Ocufolin forte per day (Ocufolin group, OG), and the other twelve patients served as a control group (control group, CG). The following factors were measured before therapy and four months later: blood Hcy, best-corrected visual acuity (BCVA), intra-ocular pressure (IOP), RVP, optical coherence tomography (OCT), and optical coherence tomography - angiography (OCTA). Results Hcy decreased on average by 5.58 μmol/L in the OG and by 0.57 μmol/L in the CG. The RVP decreased on average by 4.60 mmHg in the OG and by 0.75 mmHg in the CG. The difference between the two groups was significant for both parameters (P <0.001); 66% of the OG and 41% of the CG had no retinal fluid at the end of the study. After the completion of the study, the injection intervals could be extended more often in the OG patients than in the CG patients. Conclusions When Ocufolin forte was added to the standard therapy, RVP and Hcy were reduced to a significantly greater extent than without Ocufolin forte. In addition, Ocufolin had a positive influence on morphology and future treatment intervals with anti-VEGF therapy.
Collapse
Affiliation(s)
- Tatjana Josifova
- Eye Clinic Orasis, Department for Medical and Surgical Retina, Reinach, Switzerland
| | - Katarzyna Konieczka
- Department of Ophthalmology, University of Basel, Switzerland
- Glaucoma Eye Practice Dr. K. Konieczka, Basel, Switzerland
| | | | - Josef Flammer
- Department of Ophthalmology, University of Basel, Switzerland
| |
Collapse
|
3
|
Deng L, Wang L, Meng Y, Zheng J, Dong X, Chen Y, Huang H. A Novel Bispecific Anti-IL17/VEGF Fusion Trap Exhibits Potent and Long-Lasting Inhibitory Effects on the Development of Age-Related Macular Degeneration. Biochem Res Int 2024; 2024:1405338. [PMID: 39735855 PMCID: PMC11681983 DOI: 10.1155/bri/1405338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 10/13/2024] [Accepted: 11/30/2024] [Indexed: 12/31/2024] Open
Abstract
Age-related macular degeneration (AMD) is a severe eye disease in people aged 60 years and older. Although anti-VEGF therapies are effective in treating neovascular AMD (NvAMD) in the clinic, up to 60% of patients do not completely respond to the therapies. Recent studies have shown that blood-derived macrophages and their associated proinflammatory cytokines may play important roles in the development of persistent disease and resistance to anti-VEGF therapy. To address this issue, we constructed an antibody-based bispecific fusion protein that can simultaneously inhibit IL-17-induced inflammation and VEGF-mediated neovascularization. As a result, the bispecific fusion protein 17V05 effectively inhibited multiple proinflammatory cytokines and chemokines, as well as laser-induced choroidal neovascularization (CNV). More importantly, 17V05 also exhibited stronger and longer inhibitory effects than conbercept in vivo. Thus, we provide a novel and promising strategy for treating AMD patients who are not sensitive to anti-VEGF therapies.
Collapse
Affiliation(s)
- Lan Deng
- Development of Research and Development, Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd., a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Lihua Wang
- Development of Research and Development, Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd., a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Yun Meng
- Development of Research and Development, Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd., a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Jidai Zheng
- Development of Research and Development, Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd., a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Xia Dong
- Development of Research and Development, Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd., a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Ying Chen
- Development of Research and Development, Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd., a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| | - Haomin Huang
- Development of Research and Development, Sunshine Guojian Pharmaceutical (Shanghai) Co. Ltd., a 3SBio Inc. Company, 399 Libing Road, Shanghai 201203, China
| |
Collapse
|
4
|
Zeng L, Yang Z, Pan W, Lin D, Tang Y, Chen B, Xu H, Li X. Higher Intraocular Levels of Inflammatory Factors are Related to Retinal Vascular and Neurodegeneration in Myopic Retinopathy. J Inflamm Res 2024; 17:10889-10900. [PMID: 39677296 PMCID: PMC11646394 DOI: 10.2147/jir.s484338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024] Open
Abstract
Purpose In this study, we aimed to investigate the relationship between the intraocular levels of inflammatory factors and myopia-related retinal vascular and neuronal degeneration. Patients and Methods One hundred and forty-seven patients with Implantable Collamer Lens (ICL) implantation were enrolled and all participants received comprehensive ophthalmic examination. About 100~150 ul of aqueous humor was collected immediately before ICL surgery. The levels of inflammatory factors including Aggrecan, April, BAFF, CCL5, CD163, Chi3l1, gp130, IL-6Rα, IL-8, IL-10, IL-11, IL-12, IL-19, IL-27, IL-28A, IL-34, IFN-β, IFN-γ, MMP-1, MMP-2, MMP-3 and PTX3 in the aqueous humor were measured using the Luminex Multiplexing system. Results Results showed that aqueous humor levels of pro-inflammatory factors Chi3l1, IL-6Rα, IL-8, IL-12, IL-27, inflammation-related cytokines April, BAFF and IL-34 progressively increased from the progression of myopic retinopathy. Conversely, the aqueous levels of IL-11 and Aggrecan gradually decreased from the progression of myopic retinopathy. Correlation analysis showed that the intraocular levels of Chi3l1, IL-6Rα, IL-8, IL-27 and BAFF were negatively correlated with retinal vascular density. The intraocular level of IL-6Rα was negatively correlated with retinal neuronal thickness. Protein-Protein Interaction (PPI) analysis revealed that Chi3l1 and Aggrecan were the upstream cytokines that affect IL-10 and IL-8 in the pathological myopic eyes. KEGG pathway analysis showed that cytokine-cytokine receptor interaction, JAK-STAT signaling pathway, rheumatoid arthritis, and chagas disease were influenced by these altered inflammatory factors (adjusted p-value<0.001). Conclusion The production of inflammatory factors in the eyes of individuals with high myopia and pathological myopia was altered, and the elevated levels of intraocular pro-inflammatory factors such as Chi3l1, IL-6Rα, and IL-8 were closely associated with myopia-related retinal microvascular and neurodegeneration.
Collapse
Affiliation(s)
- Ling Zeng
- Aier Academy of Ophthalmology, Central South University, Changsha, People’s Republic of China
- Changsha Aier Eye Hospital, Changsha, People’s Republic of China
- Aier Institute of Optometry and Vision Science, Aier Eye Hospital Group, Changsha, People’s Republic of China
- The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Zhikuan Yang
- Aier Academy of Ophthalmology, Central South University, Changsha, People’s Republic of China
- Changsha Aier Eye Hospital, Changsha, People’s Republic of China
- Aier Institute of Optometry and Vision Science, Aier Eye Hospital Group, Changsha, People’s Republic of China
| | - Wei Pan
- Aier Academy of Ophthalmology, Central South University, Changsha, People’s Republic of China
- Aier Institute of Optometry and Vision Science, Aier Eye Hospital Group, Changsha, People’s Republic of China
| | - Ding Lin
- Changsha Aier Eye Hospital, Changsha, People’s Republic of China
| | - Yao Tang
- Aier Academy of Ophthalmology, Central South University, Changsha, People’s Republic of China
- Changsha Aier Eye Hospital, Changsha, People’s Republic of China
- Aier Institute of Optometry and Vision Science, Aier Eye Hospital Group, Changsha, People’s Republic of China
| | - Baihua Chen
- The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Heping Xu
- Aier Academy of Ophthalmology, Central South University, Changsha, People’s Republic of China
- Aier Institute of Optometry and Vision Science, Aier Eye Hospital Group, Changsha, People’s Republic of China
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast, UK
| | - Xiaoning Li
- Aier Academy of Ophthalmology, Central South University, Changsha, People’s Republic of China
- Changsha Aier Eye Hospital, Changsha, People’s Republic of China
- Aier Institute of Optometry and Vision Science, Aier Eye Hospital Group, Changsha, People’s Republic of China
- Aier School of Optometry and Vision Science, Hubei University of Science and Technology, Xianning, People’s Republic of China
| |
Collapse
|
5
|
Shelton DA, Papania JT, Getz TE, Sellers JT, Giradot PE, Chrenek MA, Grossniklaus HE, Boatright JH, Nickerson JM. Loss of Pigment Epithelium Derived Factor Sensitizes C57BL/6J Mice to Light-Induced Retinal Damage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.04.626802. [PMID: 39679905 PMCID: PMC11643041 DOI: 10.1101/2024.12.04.626802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Purpose Pigment epithelium-derived factor (PEDF) is a neurotrophic glycoprotein secreted by the retinal pigment epithelium (RPE) that supports retinal photoreceptor health. Deficits in PEDF are associated with increased inflammation and retinal degeneration in aging and diabetic retinopathy. We hypothesized that light-induced stress in C57BL/6J mice deficient in PEDF would lead to increased retinal neuronal and RPE defects, impaired expression of neurotrophic factor Insulin-like growth factor 1 (IGF-1), and overactivation of Galectin-3-mediated inflammatory signaling. Methods C57BL/6J mice expressing the RPE65 M450/M450 allele were crossed to PEDF KO/KO and wildtype (PEDF +/+) littermates. Mice were exposed to 50,000 lux light for 5 hours to initiate acute damage. Changes in visual function outcomes were tracked via electroretinogram (ERG), confocal scanning laser ophthalmoscopy(cSLO), and spectral domain optical coherence tomography (SD-OCT) on days 3, 5, and 7 post-light exposure. Gene and protein expression of Galectin-3 were measured by digital drop PCR (ddPCR) and western blot. To further investigate the role of galectin-3 on visual outcomes and PEDF expression after damage, we also used a small-molecule inhibitor to reduce its activity. Results Following light damage, PEDF KO/KO mice showed more severe retinal thinning, impaired visual function (reduced a-, b-, and c-wave amplitudes), and increased Galectin-3 expressing immune cell infiltration compared to PEDF +/+. PEDF KO/KO mice had suppressed damage-associated increases in IGF-1 expression. Additionally, baseline Galectin-3 mRNA and protein expression were reduced in PEDF KO/KO mice compared to PEDF +/+. However, after light damage, Galectin-3 expression decreases in PEDF +/+ mice but increases in PEDF KO/KO mice without reaching PEDF +/+ levels. Galectin-3 inhibition worsens retinal degeneration, reduces PEDF expression in PEDF +/+ mice, and mimics the effects seen in PEDF knockouts. Conclusions Loss of PEDF alone does not elicit functional defects in C57BL/6J mice. However, under light stress, PEDF deficiency significantly increases severe retinal degeneration, visual deficits, Galectin-3 expression, and suppression of IGF-1 than PEDF +/+. PEDF deficiency reduced baseline expression of Galectin-3, and pharmacological inhibition of Galectin-3 worsens outcomes and suppresses PEDF expression in PEDF +/+, suggesting a novel co-regulatory relationship between the two proteins in mitigating light-induced retinal damage.
Collapse
Affiliation(s)
- Debresha A. Shelton
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Jack T. Papania
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Tatiana E. Getz
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Jana T. Sellers
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Preston E. Giradot
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Micah A. Chrenek
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | | | - Jeffrey H. Boatright
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Atlanta Veterans Administration Center for Visual and Neurocognitive Rehabilitation, Decatur, Georgia, United States
| | - John M. Nickerson
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| |
Collapse
|
6
|
Chen H, Lai H, Chi H, Fan W, Huang J, Zhang S, Jiang C, Jiang L, Hu Q, Yan X, Chen Y, Zhang J, Yang G, Liao B, Wan J. Multi-modal transcriptomics: integrating machine learning and convolutional neural networks to identify immune biomarkers in atherosclerosis. Front Cardiovasc Med 2024; 11:1397407. [PMID: 39660117 PMCID: PMC11628520 DOI: 10.3389/fcvm.2024.1397407] [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: 03/27/2024] [Accepted: 11/06/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Atherosclerosis, a complex chronic vascular disorder with multifactorial etiology, stands as the primary culprit behind consequential cardiovascular events, imposing a substantial societal and economic burden. Nevertheless, our current understanding of its pathogenesis remains imprecise. In this investigation, our objective is to establish computational models elucidating molecular-level markers associated with atherosclerosis. This endeavor involves the integration of advanced machine learning techniques and comprehensive bioinformatics analyses. MATERIALS AND METHODS Our analysis incorporated data from three publicly available the Gene Expression Omnibus (GEO) datasets: GSE100927 (104 samples, 30,558 genes), which includes atherosclerotic lesions and control arteries from carotid, femoral, and infra-popliteal arteries of deceased organ donors; GSE43292 (64 samples, 23,307 genes), consisting of paired carotid endarterectomy samples from 32 hypertensive patients, comparing atheroma plaques and intact tissues; and GSE159677 (30,498 single cells, 33,538 genes), examining single-cell transcriptomes of calcified atherosclerotic core plaques and adjacent carotid artery tissues from patients undergoing carotid endarterectomy. Utilizing single-cell sequencing, highly variable atherosclerotic monocyte subpopulations were systematically identified. We analyzed cellular communication patterns with temporal dynamics. The bioinformatics approach Weighted Gene Co-expression Network Analysis (WGCNA) identified key modules, constructing a Protein-Protein Interaction (PPI) network from module-associated genes. Three machine-learning models derived marker genes, formulated through logistic regression and validated via convolutional neural network(CNN) modeling. Subtypes were clustered based on Gene Set Variation Analysis (GSVA) scores, validated through immunoassays. RESULTS Three pivotal atherosclerosis-associated genes-CD36, S100A10, CSNK1A1-were unveiled, offering valuable clinical insights. Profiling based on these genes delineated two distinct isoforms: C2 demonstrated potent microbicidal activity, while C1 engaged in inflammation regulation, tissue repair, and immune homeostasis. Molecular docking analyses explored therapeutic potential for Estradiol, Zidovudine, Indinavir, and Dronabinol for clinical applications. CONCLUSION This study introduces three signature genes for atherosclerosis, shaping a novel paradigm for investigating clinical immunological medications. It distinguishes the high biocidal C2 subtype from the inflammation-modulating C1 subtype, utilizing identified signature gene as crucial targets.
Collapse
Affiliation(s)
- Haiqing Chen
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Haotian Lai
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Hao Chi
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Wei Fan
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of Cardiovascular Remodeling and Dysfunction, Department of Cardiovascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Jinbang Huang
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Shengke Zhang
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Chenglu Jiang
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Lai Jiang
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Qingwen Hu
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Xiuben Yan
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Yemeng Chen
- New York College of Traditional Chinese Medicine, Mineola, NY, United States
| | - Jieying Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH, United States
| | - Bin Liao
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of Cardiovascular Remodeling and Dysfunction, Department of Cardiovascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Juyi Wan
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of Cardiovascular Remodeling and Dysfunction, Department of Cardiovascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
7
|
Almalki WH, Almujri SS. The impact of NF-κB on inflammatory and angiogenic processes in age-related macular degeneration. Exp Eye Res 2024; 248:110111. [PMID: 39326776 DOI: 10.1016/j.exer.2024.110111] [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: 06/16/2024] [Revised: 09/09/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
Age-related macular degeneration (AMD) is a prominent cause of vision loss, characterized by two different types, dry (atrophic) and wet (neovascular). Dry AMD is distinguished by the progressive deterioration of retinal cells, which ultimately causes a decline in vision. In contrast, wet AMD is defined by the abnormal development of blood vessels underneath the retina, leading to a sudden and severe vision impairment. The course of AMD is primarily driven by chronic inflammation and pathological angiogenesis, in which the NF-κB signaling pathway plays a crucial role. The activation of NF-κB results in the generation of pro-inflammatory cytokines, chemokines, and angiogenic factors like VEGF, which contribute to inflammation and the formation of new blood vessels in AMD. This review analyzes the intricate relationship between NF-κB signaling, inflammation, and angiogenesis in AMD and assesses the possibility of using NF-κB as a target for therapy. The evaluation involves a comprehensive examination of preclinical and clinical evidence that substantiates the effectiveness of NF-κB inhibitors in treating AMD by diminishing inflammation and pathological angiogenesis.
Collapse
Affiliation(s)
- Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 61421, Aseer, Saudi Arabia
| |
Collapse
|
8
|
Adegboro CO, Luo W, Kabra M, McAdams RM, York NW, Wijenayake RI, Suchla KM, Pillers DAM, Pattnaik BR. Transplacental Transfer of Oxytocin and Its Impact on Neonatal Cord Blood and In Vitro Retinal Cell Activity. Cells 2024; 13:1735. [PMID: 39451253 PMCID: PMC11506339 DOI: 10.3390/cells13201735] [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: 09/06/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
The development of fetal organs can be impacted by systemic changes in maternal circulation, with the placenta playing a pivotal role in maintaining pregnancy homeostasis and nutrient exchange. In clinical obstetrics, oxytocin (OXT) is commonly used to induce labor. To explore the potential role of OXT in the placental homeostasis of OXT, we compared OXT levels in neonatal cord blood among neonates (23-42 weeks gestation) whose mothers either received prenatal OXT or experienced spontaneous labor. Our previous research revealed that the oxytocin receptor (OXTR), essential in forming the blood-retina barrier, is expressed in the retinal pigment epithelium (RPE). We hypothesized that perinatal OXT administration might influence the development of the neural retina and its vasculature, offering therapeutic potential for retinal diseases such as retinopathy of prematurity (ROP). Plasma OXT levels were measured using a commercial OXT ELISA kit. Human fetal RPE (hfRPE) cells treated with OXT (10 µM) were assessed for gene expression via RNA sequencing, revealing 14 downregulated and 32 upregulated genes. To validate these differentially expressed genes (DEGs), hfRPE cells were exposed to OXT (0.01, 0.1, 1, or 10 µM) for 12 h, followed by RNA analysis via real-time PCR. Functional, enrichment, and network analyses (Gene Ontology term, FunRich, Cytoscape) were performed to predict the affected pathways. This translational study suggests that OXT likely crosses the placenta, altering fetal OXT concentrations. RNA sequencing identified 46 DEGs involved in vital metabolic and signaling pathways and critical cellular components. Our results indicate that the perinatal administration of OXT may affect neural retina and retinal vessel development, making OXT a potential therapeutic option for developmental eye diseases, including ROP.
Collapse
Affiliation(s)
- Claudette O. Adegboro
- Department of Pediatrics, Division of Neonatology and Newborn Nursery, University of Wisconsin–Madison, 1300 University Avenue, SMI 112, Madison, WI 53706, USA; (C.O.A.); (M.K.); (R.M.M.); (N.W.Y.); (R.I.W.); (K.M.S.)
| | - Wenxiang Luo
- Department of Pediatrics, Division of Neonatology, University of Illinois at Chicago, Chicago, IL 60612, USA; (W.L.); (D.-A.M.P.)
| | - Meha Kabra
- Department of Pediatrics, Division of Neonatology and Newborn Nursery, University of Wisconsin–Madison, 1300 University Avenue, SMI 112, Madison, WI 53706, USA; (C.O.A.); (M.K.); (R.M.M.); (N.W.Y.); (R.I.W.); (K.M.S.)
- McPherson Eye Research Institute, University of Wisconsin–Madison, 1300 University Avenue, SMI 112, Madison, WI 53706, USA
| | - Ryan M. McAdams
- Department of Pediatrics, Division of Neonatology and Newborn Nursery, University of Wisconsin–Madison, 1300 University Avenue, SMI 112, Madison, WI 53706, USA; (C.O.A.); (M.K.); (R.M.M.); (N.W.Y.); (R.I.W.); (K.M.S.)
| | - Nathaniel W. York
- Department of Pediatrics, Division of Neonatology and Newborn Nursery, University of Wisconsin–Madison, 1300 University Avenue, SMI 112, Madison, WI 53706, USA; (C.O.A.); (M.K.); (R.M.M.); (N.W.Y.); (R.I.W.); (K.M.S.)
- Endocrine and Reproductive Physiology Program, University of Wisconsin–Madison, 1300 University Avenue, SMI 112, Madison, WI 53706, USA
| | - Ruwandi I. Wijenayake
- Department of Pediatrics, Division of Neonatology and Newborn Nursery, University of Wisconsin–Madison, 1300 University Avenue, SMI 112, Madison, WI 53706, USA; (C.O.A.); (M.K.); (R.M.M.); (N.W.Y.); (R.I.W.); (K.M.S.)
| | - Kiana M. Suchla
- Department of Pediatrics, Division of Neonatology and Newborn Nursery, University of Wisconsin–Madison, 1300 University Avenue, SMI 112, Madison, WI 53706, USA; (C.O.A.); (M.K.); (R.M.M.); (N.W.Y.); (R.I.W.); (K.M.S.)
| | - De-Ann M. Pillers
- Department of Pediatrics, Division of Neonatology, University of Illinois at Chicago, Chicago, IL 60612, USA; (W.L.); (D.-A.M.P.)
- Children’s Hospital University of Illinois, University of Illinois Hospital & Health Sciences System, Chicago, IL 60612, USA
| | - Bikash R. Pattnaik
- Department of Pediatrics, Division of Neonatology and Newborn Nursery, University of Wisconsin–Madison, 1300 University Avenue, SMI 112, Madison, WI 53706, USA; (C.O.A.); (M.K.); (R.M.M.); (N.W.Y.); (R.I.W.); (K.M.S.)
- McPherson Eye Research Institute, University of Wisconsin–Madison, 1300 University Avenue, SMI 112, Madison, WI 53706, USA
- Endocrine and Reproductive Physiology Program, University of Wisconsin–Madison, 1300 University Avenue, SMI 112, Madison, WI 53706, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, 1300 University Avenue, SMI 112, Madison, WI 53706, USA
| |
Collapse
|
9
|
Peterson KM, Mishra S, Asaki E, Powell JI, He Y, Berger AE, Rajapakse D, Wistow G. Serum-deprivation response of ARPE-19 cells; expression patterns relevant to age-related macular degeneration. PLoS One 2024; 19:e0293383. [PMID: 39325754 PMCID: PMC11426544 DOI: 10.1371/journal.pone.0293383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/26/2024] [Indexed: 09/28/2024] Open
Abstract
ARPE-19 cells are derived from adult human retinal pigment epithelium (RPE). The response of these cells to the stress of serum deprivation mimics some important processes relevant to age-related macular degeneration (AMD). Here we extend the characterization of this response using RNASeq and EGSEA gene set analysis of ARPE-19 cells over nine days of serum deprivation. This experiment confirmed the up-regulation of cholesterol and lipid-associated pathways that increase cholesterol levels in these cells. The gene expression analysis also identified other pathways relevant to AMD progression. There were significant changes in extracellular matrix gene expression, notably a switch from expression of collagen IV, a key component of Bruch's membrane (part of the blood-retina barrier), to expression of a fibrosis-like collagen type I matrix. Changes in the expression profile of the extracellular matrix led to the discovery that amelotin is induced in AMD and is associated with the development of the calcium deposits seen in late-stage geographic atrophy. The transcriptional profiles of other pathways, including inflammation, complement, and coagulation, were also modified, consistent with immune response patterns seen in AMD. As previously noted, the cells resist apoptosis and autophagy but instead initiate a gene expression pattern characteristic of senescence, consistent with the maintenance of barrier function even as other aspects of RPE function are compromised. Other differentially regulated genes were identified that open new avenues for investigation. Our results suggest that ARPE-19 cells maintain significant stress responses characteristic of native RPE that are informative for AMD. As such, they provide a convenient system for discovery and for testing potential therapeutic interventions.
Collapse
Affiliation(s)
- Katherine M. Peterson
- Molecular Structure and Functional Genomics Section, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Sanghamitra Mishra
- Molecular Structure and Functional Genomics Section, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Esther Asaki
- Office of Intramural Research, Center for Information Technology, National Institutes of Health, Bethesda, Maryland, United States of America
| | - John I. Powell
- Office of Intramural Research, Center for Information Technology, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yiwen He
- Office of Intramural Research, Center for Information Technology, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alan E. Berger
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Dinusha Rajapakse
- Molecular Structure and Functional Genomics Section, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Graeme Wistow
- Molecular Structure and Functional Genomics Section, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
10
|
Wu Y, Qin X, Lu X, Ji C, Ling Y, Zhang J, Shi H, Chu B, Song B, Wang H, He Y. Enzyme-Responsive DNA Origami-Antibody Conjugates for Targeted and Combined Therapy of Choroidal Neovascularization. ACS NANO 2024; 18:22194-22207. [PMID: 39116033 DOI: 10.1021/acsnano.4c05635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Monotherapy, especially the use of antibodies targeting vascular endothelial growth factor (VEGF), has shown limitations in treating choroidal neovascularization (CNV) since reactive oxygen species (ROS) also exacerbate CNV formation. Herein, we developed a combination therapy based on a DNA origami platform targeting multiple components of ocular neovascularization. Our study demonstrated that ocular neovascularization was markedly suppressed by intravitreal injection of a rectangular DNA origami sheet modified with VEGF aptamers (Ap) conjugated to an anti-VEGF antibody (aV) via matrix metalloproteinase (MMP)-cleavable peptide linkers in a mouse model of CNV. Typically, the DNA origami-based therapeutic platform selectively accumulates in neovascularization lesions owing to the dual-targeting ability of the aV and Ap, followed by the cleavage of the peptide linker by MMPs to release the antibody. Together, the released antibody and Ap inhibited VEGF activity. Moreover, the residual bare DNA origami could effectively scavenge ROS, reducing oxidative stress at CNV sites and thus maximizing the synergistic effects of inhibiting neovascularization.
Collapse
Affiliation(s)
- Yuqi Wu
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123, China
| | - Xuan Qin
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123, China
| | - Xing Lu
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123, China
| | - Chen Ji
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123, China
| | - Yufan Ling
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Jiawei Zhang
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123, China
| | - Haoliang Shi
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123, China
| | - Binbin Chu
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123, China
| | - Bin Song
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123, China
| | - Houyu Wang
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123, China
| | - Yao He
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123, China
- Macao Translational Medicine Center, Macau University of Science and Technology, Taipa, 999078 Macau SAR, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, 999078 Macau SAR, China
| |
Collapse
|
11
|
Qi S, Zhang Y, Kong L, Bi D, Kong H, Zhang S, Zhao C. SPI1-mediated macrophage polarization aggravates age-related macular degeneration. Front Immunol 2024; 15:1421012. [PMID: 38979414 PMCID: PMC11228255 DOI: 10.3389/fimmu.2024.1421012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 05/28/2024] [Indexed: 07/10/2024] Open
Abstract
Objective This study revealed a core regulator and common upstream mechanisms for the multifaceted pathological processes of age-related macular degeneration (AMD) and provided proof-of-concept for this new therapeutic target. Methods Comprehensive gene expression analysis was performed using RNA sequencing of eye cup from old mice as well as laser-induced choroidal neovascularization (CNV) mouse model. Through integrative analysis and protein-protein interaction (PPI) analysis, common pathways and key transcription factor was identified simultaneously engaged in age-related retinal degeneration and CNV, the two typical pathological process of AMD. Subsequently, the expression changes of Spi1, the key regulator, as well as the alternation of the downstream mechanisms were validated in both models through qRT-PCR, Elisa, flow cytometry and immunofluorescence. Further, we assessed the impact of Spi1 knockdown in vitro and in vivo using gene intervention vectors carried by adeno-associated virus or lentivirus to test its potential as a therapeutic target. Results Compared to corresponding controls, we found 1,939 and 1,319 genes differentially expressed in eye cups of old and CNV mice respectively. The integrative analysis identified a total of 275 overlapping DEGs, of which 150 genes were co-upregulated. PPI analysis verified a central transcription factor, SPI1. The significant upregulation of Spi1 expression was then validated in both models, accompanied by macrophage polarization towards the M1 phenotype. Finally, SPI1 suppression significantly inhibited M1 polarization of BMDMs and attenuated neovascularization in CNV mice. Conclusion This study demonstrates that SPI1 exerts a pivotal role in AMD by regulation of macrophage polarization and innate immune response, offering promise as an innovative target for treating AMD.
Collapse
Affiliation(s)
- Siyi Qi
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, NHC, Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Yihan Zhang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, NHC, Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Lingjie Kong
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, NHC, Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Daode Bi
- Department of Neurology and Geriatrics, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Hongyu Kong
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, NHC, Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Shujie Zhang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, NHC, Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Chen Zhao
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, NHC, Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| |
Collapse
|
12
|
Cui K, Tang X, Yang B, Fan M, Hu A, Wu P, Yang F, Lin J, Kong H, Lu X, Yu S, Xu Y, Liang X. Sema4D Knockout Attenuates Choroidal Neovascularization by Inhibiting M2 Macrophage Polarization Via Regulation of the RhoA/ROCK Pathway. Invest Ophthalmol Vis Sci 2024; 65:34. [PMID: 38913005 PMCID: PMC11204059 DOI: 10.1167/iovs.65.6.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/01/2024] [Indexed: 06/25/2024] Open
Abstract
Purpose The aim of this study was to elucidate the role of Sema4D in the pathogenesis of senescence-associated choroidal neovascularization (CNV) and to explore its underlying mechanisms. Methods In this study, we utilized a model of laser-induced CNV in both young (3 months old) and old (18 months old) mice, including those with or without Sema4D knockout. The expression and localization of Sema4D in CNV were assessed using PCR, Western blot, and immunostaining. Subsequently, the morphological and imaging examinations were used to evaluate the size of CNV and vascular leakage. Finally, the expression of M2 markers, senescence-related markers, and molecules involved in the RhoA/ROCK pathway was detected. Results We found that Sema4D was predominantly expressed in macrophages within CNV lesions, and both the mRNA and protein levels of Sema4D progressively increased following laser photocoagulation, a trend more pronounced in old mice. Moreover, Sema4D knockout markedly inhibited M2 polarization in senescent macrophages and reduced the size and leakage of CNV, particularly in aged mice. Mechanistically, aging was found to upregulate RhoA/ROCK signaling, and knockout of Sema4D effectively suppressed the activation of this pathway, with more significant effects observed in aged mice. Conclusions Our findings revealed that the deletion of Sema4D markedly inhibited M2 macrophage polarization through the suppression of the RhoA/ROCK pathway, ultimately leading to the attenuation of senescence-associated CNV. These data indicate that targeting Sema4D could offer a promising approach for gene editing therapy in patients with neovascular age-related macular degeneration.
Collapse
Affiliation(s)
- Kaixuan Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xiaoyu Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Boyu Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Matthew Fan
- Yale College, Yale University, New Haven, Connecticut, United States
| | - Andina Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Peiqi Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Fengmei Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jicheng Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Haolin Kong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xi Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Shanshan Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yue Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xiaoling Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
13
|
Li C, Zhou L, Sun H, Yang MM. Age-Related Macular Degeneration: A Disease of Cellular Senescence and Dysregulated Immune Homeostasis. Clin Interv Aging 2024; 19:939-951. [PMID: 38807637 PMCID: PMC11130992 DOI: 10.2147/cia.s463297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/11/2024] [Indexed: 05/30/2024] Open
Abstract
Age-related macular degeneration (AMD) is a degenerative ocular disease primarily affecting central vision in the elderly. Its pathogenesis is complex, involving cellular senescence and immune homeostasis dysregulation. This review investigates the interaction between these two critical biological processes in AMD pathogenesis and their impact on disease progression. Initially, cellular senescence is analyzed, with particular emphasis on retinal damage induced by senescent retinal pigment epithelial cells. Subsequently, the occurrence of immune homeostasis dysregulation within the retina and its mechanistic role in AMD areis explored. Furthermore, the paper also discusses in detail the interplay between cellular senescence and immune responses, forming a vicious cycle that exacerbates retinal damage and may influence treatment outcomes. In summary, a deeper understanding of the interrelation between cellular senescence and immune dysregulation is vital for the developing innovative therapeutic strategies for AMD.
Collapse
Affiliation(s)
- Cunzi Li
- The Second Clinical Medical College of Jinan University (Shenzhen People’s Hospital), Shenzhen, 518020, People’s Republic of China
| | - Lan Zhou
- Department of Ophthalmology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, People’s Republic of China
- Post-Doctoral Scientific Research Station of Basic Medicine, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Hongyan Sun
- Department of Ophthalmology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, People’s Republic of China
| | - Ming Ming Yang
- Department of Ophthalmology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, People’s Republic of China
| |
Collapse
|
14
|
Brito M, Sorbier C, Mignet N, Boudy V, Borchard G, Vacher G. Understanding the Impact of Polyunsaturated Fatty Acids on Age-Related Macular Degeneration: A Review. Int J Mol Sci 2024; 25:4099. [PMID: 38612907 PMCID: PMC11012607 DOI: 10.3390/ijms25074099] [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: 03/01/2024] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Age-related Macular Degeneration (AMD) is a multifactorial ocular pathology that destroys the photoreceptors of the macula. Two forms are distinguished, dry and wet AMD, with different pathophysiological mechanisms. Although treatments were shown to be effective in wet AMD, they remain a heavy burden for patients and caregivers, resulting in a lack of patient compliance. For dry AMD, no real effective treatment is available in Europe. It is, therefore, essential to look for new approaches. Recently, the use of long-chain and very long-chain polyunsaturated fatty acids was identified as an interesting new therapeutic alternative. Indeed, the levels of these fatty acids, core components of photoreceptors, are significantly decreased in AMD patients. To better understand this pathology and to evaluate the efficacy of various molecules, in vitro and in vivo models reproducing the mechanisms of both types of AMD were developed. This article reviews the anatomy and the physiological aging of the retina and summarizes the clinical aspects, pathophysiological mechanisms of AMD and potential treatment strategies. In vitro and in vivo models of AMD are also presented. Finally, this manuscript focuses on the application of omega-3 fatty acids for the prevention and treatment of both types of AMD.
Collapse
Affiliation(s)
- Maëlis Brito
- Unither Développement Bordeaux, Avenue Toussaint Catros, 33185 Le Haillan, France
- Université Paris Cité, CNRS, INSERM, UTCBS, Unité de Technologies Chimiques et Biologiques pour la Santé, F-75006 Paris, France
- Département de Recherche et Développement (DRDP), Agence Générale des Equipements et Produits de Santé (AGEPS), Assistance Publique Hôpitaux de Paris (AP-HP), 7 Rue du Fer-à-Moulin, 75005 Paris, France
- Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1206 Geneva, Switzerland
| | - Capucine Sorbier
- Unither Développement Bordeaux, Avenue Toussaint Catros, 33185 Le Haillan, France
| | - Nathalie Mignet
- Université Paris Cité, CNRS, INSERM, UTCBS, Unité de Technologies Chimiques et Biologiques pour la Santé, F-75006 Paris, France
| | - Vincent Boudy
- Université Paris Cité, CNRS, INSERM, UTCBS, Unité de Technologies Chimiques et Biologiques pour la Santé, F-75006 Paris, France
- Département de Recherche et Développement (DRDP), Agence Générale des Equipements et Produits de Santé (AGEPS), Assistance Publique Hôpitaux de Paris (AP-HP), 7 Rue du Fer-à-Moulin, 75005 Paris, France
| | - Gerrit Borchard
- Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1206 Geneva, Switzerland
| | - Gaëlle Vacher
- Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1206 Geneva, Switzerland
| |
Collapse
|
15
|
Dervenis N, Dervenis P, Agorogiannis E. Neovascular age-related macular degeneration: disease pathogenesis and current state of molecular biomarkers predicting treatment response-a scoping review. BMJ Open Ophthalmol 2024; 9:e001516. [PMID: 38341189 PMCID: PMC10862285 DOI: 10.1136/bmjophth-2023-001516] [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: 09/27/2023] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Age-related macular degeneration is a major cause of blindness, and the development of anti-vascular endothelial growth factor (VEGF) intravitreal treatments has revolutionised the management of the disease. At the same time, new challenges and unmet needs arose due to the limitations of the current therapeutic options. Neovascularisation development during the course of the disease has a complex pathogenetic mechanism, and several biomarkers and their association with treatment outcomes have been investigated. We reviewed the relevant literature about neovascularisation development and biomarkers related to response to treatment. Improving our knowledge on the field can improve patient outcomes and offer personalised care.
Collapse
Affiliation(s)
- Nikolaos Dervenis
- Aristotle University of Thessaloniki, Thessaloniki, Greece
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | | | | |
Collapse
|
16
|
Orihara K, Kobayashi-Otsugu M, Nakajima E, Walkup RD, Wilson DJ, Shearer TR, Azuma M. Calpain-specific breakdown fragment in human drusen. Histol Histopathol 2024; 39:165-175. [PMID: 37314158 DOI: 10.14670/hh-18-635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
PURPOSE With aging and age-related macular dystrophy (AMD), proteolytic fragments are deposited in extracellular drusen located between the RPE and Bruch's membrane. Localized hypoxia may be a risk factor for AMD. Our hypothesis is that following hypoxia, activation of proteolytic enzymes called calpains may cause proteolysis/degeneration of retinal cells and RPE. No direct evidence has yet demonstrated activation of calpains in AMD. The purpose of the present study was to identify calpain-cleaved proteins in drusen. METHODS Seventy-six (76) drusen were analyzed in human eye sections from six normal and twelve AMD human donor eyes. The sections were subjected to immunofluorescence for the calpain-specific 150 kDa breakdown product from α-spectrin, SBDP150 - a marker for calpain activation, and for recoverin - a marker for photoreceptor cells. RESULTS Among 29 nodular drusen, 80% from normal eyes and 90% from AMD eyes stained positive for SBDP150. Among 47 soft drusen, mostly from AMD eyes, 72% stained positive for SBDP150. Thus, the majority of both soft and nodular drusen from AMD donors contained SBDP150. CONCLUSIONS SBDP150 was detected for the first time in soft and nodular drusen from human donors. Our results suggest that calpain-induced proteolysis participates in the degeneration of photoreceptors and/or RPE cells during aging and AMD. Calpain inhibitors may ameliorate AMD progression.
Collapse
Affiliation(s)
- Kana Orihara
- Senju Laboratory of Ocular Sciences, Senju Pharmaceutical Corporation Ltd., Portland, OR, USA
| | - Momoko Kobayashi-Otsugu
- Senju Laboratory of Ocular Sciences, Senju Pharmaceutical Corporation Ltd., Portland, OR, USA
| | - Emi Nakajima
- Senju Laboratory of Ocular Sciences, Senju Pharmaceutical Corporation Ltd., Portland, OR, USA
- Department of Oral Rehabilitation and Biosciences, Oregon Health and Science University, Portland, OR, USA
| | - Ryan D Walkup
- Senju Laboratory of Ocular Sciences, Senju Pharmaceutical Corporation Ltd., Portland, OR, USA
- Department of Oral Rehabilitation and Biosciences, Oregon Health and Science University, Portland, OR, USA
| | - David J Wilson
- Department of Ophthalmology, School of Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Thomas R Shearer
- Department of Oral Rehabilitation and Biosciences, Oregon Health and Science University, Portland, OR, USA
| | - Mitsuyoshi Azuma
- Senju Laboratory of Ocular Sciences, Senju Pharmaceutical Corporation Ltd., Portland, OR, USA
- Department of Oral Rehabilitation and Biosciences, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
17
|
Shelton DA, Gefke I, Summers V, Kim YK, Yu H, Getz Y, Ferdous S, Donaldson K, Liao K, Papania JT, Chrenek MA, Boatright JH, Nickerson JM. Age-Related RPE changes in Wildtype C57BL/6J Mice between 2 and 32 Months. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.30.574142. [PMID: 38352604 PMCID: PMC10862734 DOI: 10.1101/2024.01.30.574142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Purpose This study provides a systematic evaluation of age-related changes in RPE cell structure and function using a morphometric approach. We aim to better capture nuanced predictive changes in cell heterogeneity that reflect loss of RPE integrity during normal aging. Using C57BL6/J mice ranging from P60-P730, we sought to evaluate how regional changes in RPE shape reflect incremental losses in RPE cell function with advancing age. We hypothesize that tracking global morphological changes in RPE is predictive of functional defects over time. Methods We tested three groups of C57BL/6J mice (young: P60-180; Middle-aged: P365-729; aged: 730+) for function and structural defects using electroretinograms, immunofluorescence, and phagocytosis assays. Results The largest changes in RPE morphology were evident between the young and aged groups, while the middle-aged group exhibited smaller but notable region-specific differences. We observed a 1.9-fold increase in cytoplasmic alpha-catenin expression specifically in the central-medial region of the eye between the young and aged group. There was an 8-fold increase in subretinal, IBA-1-positive immune cell recruitment and a significant decrease in visual function in aged mice compared to young mice. Functional defects in the RPE corroborated by changes in RPE phagocytotic capacity. Conclusions The marked increase of cytoplasmic alpha-catenin expression and subretinal immune cell deposition, and decreased visual output coincide with regional changes in RPE cell morphometrics when stratified by age. These cumulative changes in the RPE morphology showed predictive regional patterns of stress associated with loss of RPE integrity.
Collapse
Affiliation(s)
- Debresha A. Shelton
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Isabelle Gefke
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Vivian Summers
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Yong-Kyu Kim
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Department of Ophthalmology, Hallym University College of Medicine, Kangdong Sacred Heart Hospital, Seoul, South Korea
| | - Hanyi Yu
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Department of Computer Science, Emory University, Atlanta, Georgia, United States
| | - Yana Getz
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Salma Ferdous
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States
| | - Kevin Donaldson
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Kristie Liao
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Jack T. Papania
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Micah A. Chrenek
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Jeffrey H. Boatright
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Decatur, Georgia, United States
| | - John M. Nickerson
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| |
Collapse
|
18
|
Li B, Fan K, Zhang T, Wu Z, Zeng S, Zhao M, Ren Q, Zheng D, Wang L, Liu X, Han M, Song Y, Ye J, Pei C, Yi J, Wang X, Peng H, Zhang H, Zhou Z, Liang X, Yu F, Wu M, Li C, Lei C, Hao J, Tang L, Yuan H, Cai S, Li Q, Zhong J, Li S, Liu L, Ke M, Wang J, Wang H, Zhu M, Wang Z, Yan Y, Wang F, Chen Y. Efficacy and Safety of Biosimilar QL1207 vs. the Reference Aflibercept for Patients with Neovascular Age-Related Macular Degeneration: A Randomized Phase 3 Trial. Ophthalmol Ther 2024; 13:353-366. [PMID: 37987893 PMCID: PMC10776547 DOI: 10.1007/s40123-023-00836-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 10/11/2023] [Indexed: 11/22/2023] Open
Abstract
INTRODUCTION This trial aimed to compare the efficacy and safety between biosimilar QL1207 and the reference aflibercept for the treatment of neovascular age-related macular degeneration (nAMD). METHODS This randomized, double-blind, phase 3 trial was conducted at 35 centers in China. Patients aged ≥ 50 years old with untreated subfoveal choroidal neovascularization secondary to nAMD and best-corrected visual acuity (BCVA) letter score of 73-34 were eligible. Patients were randomly assigned to receive intravitreous injections of QL1207 or aflibercept 2 mg (0.05 ml) in the study eye every 4 weeks for the first 3 months, followed by 2 mg every 8 weeks until week 48, stratified by baseline BCVA ≥ or < 45 letters. The primary endpoint was BCVA change from baseline at week 12. The equivalence margin was ± 5 letters. The safety, immunogenicity, pharmacokinetics (PK), and plasma vascular endothelial growth factor (VEGF) concentration were also evaluated. RESULTS A total of 366 patients were enrolled (QL1207 group, n = 185; aflibercept group, n = 181) from Aug 2019 to Jan 2022 with comparable baseline characteristics. The least-squares mean difference in BCVA changes was - 1.1 letters (95% confidence interval - 3.0 to 0.7; P = 0.2275) between the two groups, within the equivalence margin. The incidences of treatment-emergent adverse events (TEAE; QL1207: 71.4% [132/185] vs. aflibercept: 71.8% [130/181]) and serious TEAE (QL1207: 14.1% [26] vs. aflibercept: 12.7% [23]) appeared comparable between treatment groups, and no new safety signal was found. Anti-drug antibody, PK profiles, and VEGF concentration were similar between the two groups. CONCLUSIONS QL1207 has equivalent efficacy to aflibercept for nAMD with similar safety profiles. It could be used as an alternative anti-VEGF agent for clinical practice. TRIAL REGISTRATION ClinicalTrials.gov: NCT05345236 (retrospectively registered on April 25, 2022); National Medical Products Administration of China: CTR20190937 (May 20, 2019).
Collapse
Affiliation(s)
- Bing Li
- Ophthalmology Department, Peking Union Medical College Hospital, No. 1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Ke Fan
- Ophthalmology Department, Henan Provincial Eye Hospital, Zhengzhou, China
| | - Tonghe Zhang
- Ophthalmology Department, Jinan Second People's Hospital, Jinan, China
| | - Zhifeng Wu
- Ophthalmology Department, Wuxi Second People's Hospital, Wuxi, China
| | - Siming Zeng
- Ophthalmology Department, Guangxi Zhuang Autonomous Region People's Hospital, Nanning, China
| | - Mingwei Zhao
- Ophthalmology Department, Peking University People's Hospital, Beijing, China
| | - Qian Ren
- Ophthalmology Department, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Dongping Zheng
- Ophthalmology Department, Shanxi Ophthalmic Hospital, Taiyuan, China
| | - Lifei Wang
- Ophthalmology Department, Hebei Eye Hospital, Xingtai, China
| | - Xiaoling Liu
- Ophthalmology Department, Eye Hospital, WMU Zhejiang Eye Hospital, Wenzhou, China
| | - Mei Han
- Ophthalmology Department, Tianjin Eye Hospital, Tianjin, China
| | - Yanping Song
- Ophthalmology Department, General Hospital of Central Theater Command, Wuhan, China
| | - Jian Ye
- Ophthalmology Department, Army Medical Center of PLA, Chongqing, China
| | - Cheng Pei
- Ophthalmology Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jinglin Yi
- Ophthalmology Department, Affiliated Eye Hospital of Nanchang University, Nanchang, China
| | - Xian Wang
- Ophthalmology Department, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Hui Peng
- Ophthalmology Department, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Zhang
- Ophthalmology Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhanyu Zhou
- Ophthalmology Department, Qingdao Municipal Hospital, Qingdao, China
| | - Xiaoling Liang
- Ophthalmology Department, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Fangliang Yu
- Ophthalmology Department, The First Hospital of Nanchang, Nanchang, China
| | - Miaoqin Wu
- Ophthalmology Department, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Chaopeng Li
- Ophthalmology Department, Huai'an First People's Hospital, Huai'an, China
| | - Chunling Lei
- Ophthalmology Department, Xi'an People's Hospital, Xi'an, China
| | - Jilong Hao
- Ophthalmology Department, First Hospital of Jilin University, Changchun, China
| | - Luosheng Tang
- Ophthalmology Department, The Second Xiangya Hospital of Center South University, Changsha, China
| | - Huiping Yuan
- Ophthalmology Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shanjun Cai
- Ophthalmology Department, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Qiuming Li
- Ophthalmology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingxiang Zhong
- Ophthalmology Department, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Suyan Li
- Ophthalmology Department, Xuzhou First People's Hospital, Xuzhou, China
| | - Lin Liu
- Ophthalmology Department, Renji Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Min Ke
- Ophthalmology Department, Wuhan University Zhongnan Hospital, Wuhan, China
| | - Jing Wang
- Clinical Research Center, Qilu Pharmaceutical Co., Ltd., Jinan, China
| | - Hui Wang
- Clinical Research Center, Qilu Pharmaceutical Co., Ltd., Jinan, China
| | - Mengli Zhu
- Clinical Research Center, Qilu Pharmaceutical Co., Ltd., Jinan, China
| | - Zenghua Wang
- Clinical Research Center, Qilu Pharmaceutical Co., Ltd., Jinan, China
| | - Yang Yan
- Clinical Research Center, Qilu Pharmaceutical Co., Ltd., Jinan, China
| | - Feng Wang
- Ophthalmology Department, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi'an, 710004, China.
| | - Youxin Chen
- Ophthalmology Department, Peking Union Medical College Hospital, No. 1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
19
|
Tolentino MJ, Tolentino AJ, Tolentino EM, Krishnan A, Genead MA. Sialic Acid Mimetic Microglial Sialic Acid-Binding Immunoglobulin-like Lectin Agonism: Potential to Restore Retinal Homeostasis and Regain Visual Function in Age-Related Macular Degeneration. Pharmaceuticals (Basel) 2023; 16:1735. [PMID: 38139861 PMCID: PMC10747662 DOI: 10.3390/ph16121735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/29/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Age-related macular degeneration (AMD), a leading cause of visual loss and dysfunction worldwide, is a disease initiated by genetic polymorphisms that impair the negative regulation of complement. Proteomic investigation points to altered glycosylation and loss of Siglec-mediated glyco-immune checkpoint parainflammatory and inflammatory homeostasis as the main determinant for the vision impairing complications of macular degeneration. The effect of altered glycosylation on microglial maintained retinal para-inflammatory homeostasis and eventual recruitment and polarization of peripheral blood monocyte-derived macrophages (PBMDMs) into the retina can explain the phenotypic variability seen in this clinically heterogenous disease. Restoring glyco-immune checkpoint control with a sialic acid mimetic agonist targeting microglial/macrophage Siglecs to regain retinal para-inflammatory and inflammatory homeostasis is a promising therapeutic that could halt the progression of and improve visual function in all stages of macular degeneration.
Collapse
Affiliation(s)
- Michael J. Tolentino
- Department of Ophthalmology, University of Central Florida College of Medicine, Orlando, FL 32827, USA
- Department of Ophthalmology, Orlando College of Osteopathic Medicine, Orlando, FL 34787, USA
- Aviceda Therapeutics, Cambridge, MA 02142, USA; (A.K.); (M.A.G.)
| | - Andrew J. Tolentino
- Department of Biology, University of California Berkeley, Berkeley, CA 94720, USA;
| | | | - Anitha Krishnan
- Aviceda Therapeutics, Cambridge, MA 02142, USA; (A.K.); (M.A.G.)
| | | |
Collapse
|
20
|
Nadeem A, Malik IA, Shariq F, Afridi EK, Taha M, Raufi N, Naveed AK, Iqbal J, Habte A. Advancements in the treatment of geographic atrophy: focus on pegcetacoplan in age-related macular degeneration. Ann Med Surg (Lond) 2023; 85:6067-6077. [PMID: 38098608 PMCID: PMC10718344 DOI: 10.1097/ms9.0000000000001466] [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: 07/12/2023] [Accepted: 10/23/2023] [Indexed: 12/17/2023] Open
Abstract
Geographic atrophy (GA) is a progressive form of age-related macular degeneration characterized by the degeneration of retinal pigment epithelial cells and photoreceptor death. The dysregulation of the complement cascade has been implicated in GA progression. This review provides a comprehensive overview of the pathophysiology of age-related macular degeneration and GA, discusses current therapeutic options, and focuses on the recent breakthrough drug, pegcetacoplan (SYFOVRE). Pegcetacoplan is a complement inhibitor that selectively targets the C3 complement protein, effectively modulating complement activation. Clinical trials, including the OAKS and DERBY studies, have demonstrated the efficacy of SYFOVRE in reducing the growth of GA lesions compared to placebo. The FDA approval of SYFOVRE as the first and only definitive therapy for GA marks a significant milestone in the management of this debilitating condition. The review also explores potential future treatment strategies, including immune-modulating agents and ocular gene therapy. While SYFOVRE offers new hope for GA patients, further research is needed to evaluate its long-term benefits, safety profile, and optimal treatment regimens.
Collapse
Affiliation(s)
| | | | | | | | | | - Nahid Raufi
- Department of Medicine, Kabul Medical University, Afghanistan
| | - Ahmed K. Naveed
- Department of Medicine, Dow University of Health Sciences, Karachi
| | - Javed Iqbal
- King Edward Medical University Lahore, Pakistan
| | - Alexander Habte
- Department of Surgery, Assab Military Hospital, Assab, Eritrea
| |
Collapse
|
21
|
Cozzi M, Monteduro D, Airaldi M, Parrulli S, Cinus F, Trinco A, Staurenghi G, Invernizzi A. Retromode Imaging Technology for Detecting Drusen-Like Deposits in Healthy Adults. Ophthalmol Retina 2023; 7:1051-1058. [PMID: 37479086 DOI: 10.1016/j.oret.2023.07.012] [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: 03/08/2023] [Revised: 06/17/2023] [Accepted: 07/13/2023] [Indexed: 07/23/2023]
Abstract
PURPOSE To investigate the ability of retromode imaging technology to visualize drusen-like deposits (DLDs) in the macular region of healthy individuals without retinal diseases. Additionally, the correlation between subject age and the density of DLDs was assessed and their topographic distribution was evaluated. DESIGN Prospective, observational, cross-sectional study SUBJECTS: Healthy volunteers (aged ≥ 35 years) without macular diseases. METHODS This study evaluated macular images in healthy adults using color fundus photography (FP) and retromode imaging. Two masked graders counted the number of DLDs identifiable with each modality. The standardized ETDRS concentric rings were adopted to divide DLDs based on their topographic distribution. MAIN OUTCOME MEASURES Comparison of the number of DLDs detected with each imaging modality. The association between DLDs and age. The topographic distribution of macular DLDs with retromode imaging. RESULTS The study included 91 eyes of 52 healthy volunteers (mean ± standard deviation age, 57.9 ± 10.9 years; range, 36-82 years). Overall, at least 1 DLD was present in 63.74% of eyes on color FP and 96.71% on retromode. Retromode imaging allowed detection of significantly more DLDs compared with color FP within the ETDRS grid (median [interquartile range], 4 [1-14] vs. 0 [0-0] respectively; P < 0.001). The density of DLDs was higher in the outer and inner rings compared with the central subfield (relative risk [RR], 16.70; 95% confidence interval [CI], 10.3-27.3 vs. RR 17.1; 95% CI, 10.5-27.6, respectively). Age was significantly correlated with DLDs density in all 3 sectors (all P < 0.05). CONCLUSIONS Retromode technology allowed the detection of a significantly higher number of DLDs compared with FP in the macula of healthy individuals. This noninvasive imaging modality could be used to investigate the effect of the aging process on the macula, fostering a better understanding of the pathophysiology of age-related macular diseases. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Mariano Cozzi
- Eye Clinic, Department of Biomedical and Clinical Science, University of Milan, Milan, Italy
| | - Davide Monteduro
- Eye Clinic, Department of Biomedical and Clinical Science, University of Milan, Milan, Italy
| | - Matteo Airaldi
- Eye Clinic, Department of Biomedical and Clinical Science, University of Milan, Milan, Italy
| | - Salvatore Parrulli
- Eye Clinic, Department of Biomedical and Clinical Science, University of Milan, Milan, Italy
| | - Federico Cinus
- Eye Clinic, Department of Biomedical and Clinical Science, University of Milan, Milan, Italy
| | - Andrea Trinco
- Eye Clinic, Department of Biomedical and Clinical Science, University of Milan, Milan, Italy
| | - Giovanni Staurenghi
- Eye Clinic, Department of Biomedical and Clinical Science, University of Milan, Milan, Italy
| | - Alessandro Invernizzi
- Eye Clinic, Department of Biomedical and Clinical Science, University of Milan, Milan, Italy; The University of Sydney, Save Sight Institute, Discipline of Ophthalmology, Sydney Medical School, Sydney, NSW, Australia.
| |
Collapse
|
22
|
Wu J, Lin C, Du Y, Fan SJ, Pan L, Pan Q, Cao K, Wang N. Macular thickness and its associated factors in a Chinese rural adult population: the Handan Eye Study. Br J Ophthalmol 2023; 107:1864-1872. [PMID: 36162970 DOI: 10.1136/bjo-2022-321766] [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: 05/06/2022] [Accepted: 09/04/2022] [Indexed: 11/04/2022]
Abstract
PURPOSE To describe the normal macular thickness and assess its associations. METHODS The Handan Eye Follow-up Study was conducted between 2012 and 2013. Macular thickness was scanned by spectral-domain optical coherence tomography (OCT). The built-in software generated a retinal thickness (RT) map, which was divided into three regions (central, internal and external regions) and nine quadrants (one in central and four in internal and external regions each). RESULTS For 5394 subjects in the Handan Eye Follow-up Study, 4793 received OCT examination, 2946 of whom (accounting for 61.46% of the total subjects, mean age 58.91±10.95, 55.6% were women) were included for analysis. The mean RT in central macula, inner and outer rings were (237.38 µm±23.05 µm), (309.77 µm±18.36 µm) and (278.29 µm±14.38 µm), respectively (overall difference, p<0.001). In inner ring, the RT in temporal was thinnest, followed by nasal, superior and inferior. In outer ring, the RT in superior was thinnest, with the next subfields being temporal, inferior and nasal, respectively. The RT in central macula, inner and outer rings were significantly thicker in men than in women. Multivariate linear regression analysis showed that in central macula, RT increased in subjects younger than 60 years and thinned above the age of 60. In inner and outer rings, RT thinned along with age (p<0.001). CONCLUSIONS This study finds that RT in central macula is the thinnest, followed by the outer ring, the RT in the inner ring is the thickest. Age and gender are related to RT. These associated factors need to be considered when explaining RT.
Collapse
Affiliation(s)
- Jian Wu
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Science Key Lab, Beijing, Beijing, China
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, Stanford, California, USA
| | - Caixia Lin
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Science Key Lab, Beijing, Beijing, China
| | - Yifan Du
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Science Key Lab, Beijing, Beijing, China
| | - Su Jie Fan
- Department of Ophthalmology, Handan City Eye Hospital, Handan, Hebei, China
| | - Lijie Pan
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Science Key Lab, Beijing, Beijing, China
| | - Qing Pan
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kai Cao
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Science Key Lab, Beijing, Beijing, China
| | - Ningli Wang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Science Key Lab, Beijing, Beijing, China
| |
Collapse
|
23
|
Krishnan A, Sendra VG, Patel D, Lad A, Greene MK, Smyth P, Gallaher SA, Herron ÚM, Scott CJ, Genead M, Tolentino M. PolySialic acid-nanoparticles inhibit macrophage mediated inflammation through Siglec agonism: a potential treatment for age related macular degeneration. Front Immunol 2023; 14:1237016. [PMID: 38045700 PMCID: PMC10690618 DOI: 10.3389/fimmu.2023.1237016] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 10/23/2023] [Indexed: 12/05/2023] Open
Abstract
Age-related macular degeneration (AMD) is a chronic, progressive retinal disease characterized by an inflammatory response mediated by activated macrophages and microglia infiltrating the inner layer of the retina. In this study, we demonstrate that inhibition of macrophages through Siglec binding in the AMD eye can generate therapeutically useful effects. We show that Siglecs-7, -9 and -11 are upregulated in AMD associated M0 and M1 macrophages, and that these can be selectively targeted using polysialic acid (PolySia)-nanoparticles (NPs) to control dampen AMD-associated inflammation. In vitro studies showed that PolySia-NPs bind to macrophages through human Siglecs-7, -9, -11 as well as murine ortholog Siglec-E. Following treatment with PolySia-NPs, we observed that the PolySia-NPs bound and agonized the macrophage Siglecs resulting in a significant decrease in the secretion of IL-6, IL-1β, TNF-α and VEGF, and an increased secretion of IL-10. In vivo intravitreal (IVT) injection of PolySia-NPs was found to be well-tolerated and safe making it effective in preventing thinning of the retinal outer nuclear layer (ONL), inhibiting macrophage infiltration, and restoring electrophysiological retinal function in a model of bright light-induced retinal degeneration. In a clinically validated, laser-induced choroidal neovascularization (CNV) model of exudative AMD, PolySia-NPs reduced the size of neovascular lesions with associated reduction in macrophages. The PolySia-NPs described herein are therefore a promising therapeutic strategy for repolarizing pro-inflammatory macrophages to a more anti-inflammatory, non-angiogenic phenotype, which play a key role in the pathophysiology of non-exudative AMD.
Collapse
Affiliation(s)
| | | | - Diyan Patel
- Aviceda Therapeutics Inc., Cambridge, MA, United States
| | - Amit Lad
- Aviceda Therapeutics Inc., Cambridge, MA, United States
| | - Michelle K. Greene
- Aviceda Therapeutics Inc., Cambridge, MA, United States
- The Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry & Biomedical Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Peter Smyth
- Aviceda Therapeutics Inc., Cambridge, MA, United States
- The Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry & Biomedical Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Samantha A. Gallaher
- The Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry & Biomedical Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Úna M. Herron
- Aviceda Therapeutics Inc., Cambridge, MA, United States
- The Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry & Biomedical Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Christopher J. Scott
- Aviceda Therapeutics Inc., Cambridge, MA, United States
- The Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry & Biomedical Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | | | - Michael Tolentino
- Aviceda Therapeutics Inc., Cambridge, MA, United States
- Department of Ophthalmology, University of Central Florida School of Medicine, Orlando, FL, United States
| |
Collapse
|
24
|
Korb CA, Lackner KJ, Wolters D, Schuster AK, Nickels S, Beutgen VM, Münzel T, Wild PS, Beutel ME, Schmidtmann I, Pfeiffer N, Grus FH. Association of autoantibody levels with different stages of age-related macular degeneration (AMD): Results from the population-based Gutenberg Health Study (GHS). Graefes Arch Clin Exp Ophthalmol 2023; 261:2763-2773. [PMID: 37160502 PMCID: PMC10543519 DOI: 10.1007/s00417-023-06085-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 04/17/2023] [Accepted: 04/21/2023] [Indexed: 05/11/2023] Open
Abstract
PURPOSE Anti-retinal autoantibodies are assumed to be associated with age-related macular degeneration (AMD). To our knowledge, this is the first evaluation of autoantibodies in human sera of participants with different stages of AMD in a large population-based, observational cohort study in Germany. METHODS The Gutenberg Health Study (GHS) is a population-based, observational cohort study in Germany, including 15,010 participants aged between 35 and 74. Amongst others, non-mydriatic fundus photography (Visucam PRO NM™, Carl Zeiss Meditec AG, Jena, Germany) was performed. Fundus images of the first 5000 participants were graded based on the Rotterdam Eye Study classification. Sera of participants with AMD (n=541) and sera of age-matched participants without AMD (n=490) were analyzed by antigen-microarrays. Besides descriptive statistics, autoantibody-levels were compared by Mann-Whitney-U test and the associations of level of autoantibodies with AMD were calculated by logistic regression analysis. Likewise, possible associations of the autoantibodies and both clinical and laboratory parameters on AMD subjects were analyzed. RESULTS Autoantibodies against transferrin (p<0.001) were significantly downregulated in participants with early AMD and soft, distinct drusen (≥63 μm) or pigmentary abnormalities only compared to Controls. Mitogen-activated protein kinase 3 (p=0.041), glutathione peroxidase 4 (p=0.048), clusterin (p=0.045), lysozyme (p=0.19), protein kinase C substrate 80K-H (p=0.02), heat shock 70 kDa protein 1A (p=0.04) and insulin (p=0.018) show a trend between Control and participants with early AMD and soft, distinct drusen (≥63 μm) or pigmentary abnormalities only. CONCLUSIONS This study contributes to a growing knowledge of autoantibodies in association with different AMD stages compared to controls in the context of a large population-based study in Germany. Especially autoantibodies against inflammatory proteins were downregulated in participants with early AMD and soft, distinct drusen (≥63 μm) or pigmentary abnormalities only.
Collapse
Affiliation(s)
- Christina A Korb
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Karl J Lackner
- Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Dominik Wolters
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Alexander K Schuster
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Stefan Nickels
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Vanessa M Beutgen
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Thomas Münzel
- Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Philipp S Wild
- Preventive Cardiology and Preventive Medicine, Department of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), partner site Rhine Main, Mainz, Germany
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Manfred E Beutel
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Irene Schmidtmann
- Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Franz H Grus
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| |
Collapse
|
25
|
Keenan TD. Geographic Atrophy in Age-Related Macular Degeneration: A Tale of Two Stages. OPHTHALMOLOGY SCIENCE 2023; 3:100306. [PMID: 37197703 PMCID: PMC10183660 DOI: 10.1016/j.xops.2023.100306] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 05/19/2023]
Abstract
Purpose To examine disease progression in age-related macular degeneration (AMD) at 2 distinct stages, progression to geographic atrophy (GA) versus GA expansion, by comparison of the risk and protective factors at each stage. Design Perspective. Subjects Individuals at risk of GA or with GA. Main Outcome Measures Progression to GA and GA expansion rate. Methods Critical synthesis of the literature on risk and protective factors, both environmental and genetic, for progression to GA versus GA expansion in AMD. Results Comparison of the risk and protective factors demonstrates partially overlapping but partially distinct risk and protective factors for progression to GA versus GA expansion. Some factors are shared (i.e., operating in the same direction at both stages), others are not shared, and others seem to operate in different directions at each stage. Risk variants at ARMS2/HTRA1 increase both risk of progression to GA and GA expansion rate, presumably through the same mechanism. By contrast, risk and protective variants at CFH/CFHR alter risk of GA but not GA expansion rate. A risk variant at C3 increases risk of GA but is associated with slower GA expansion. In environmental factors, cigarette smoking is associated with increased risk of GA and faster GA expansion, whereas increased age is associated with the former but not the latter. The Mediterranean diet is associated with decreased progression at both stages, although the food components with the largest contributions seem to differ between the 2 stages. Some phenotypic features, such as reticular pseudodrusen and hyperreflective foci, are associated with increased progression at both stages. Conclusions Analysis of the risk and protective factors for progression to GA and GA expansion demonstrates partially overlapping but partially distinct elements at each stage: some are shared, some are relevant to 1 stage only, and some even seem active in opposite directions at each stage. Aside from ARMS2/HTRA1, the overlap between the genetic risk factors for the 2 stages is minimal. This suggests that the biologic mechanisms differ at least partially between the 2 disease stages. This has implications for therapeutic approaches and suggests that treatment aimed at the underlying disease processes may need to be tailored by stage. Financial Disclosures Proprietary or commercial disclosure may be found after the references.
Collapse
Affiliation(s)
- Tiarnan D.L. Keenan
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland
- Correspondence: Tiarnan D. L. Keenan, BM BCh, PhD, NIH, Building 10, CRC, Room 10D45, 10 Center Dr, MSC 1204, Bethesda, MD 20892-1204.
| |
Collapse
|
26
|
Sharma SN, Marsh JW, Tsipursky MS, Boppart SA. Ratiometric Analysis of In Vivo Optical Coherence Tomography Retinal Layer Thicknesses for Detection of Changes in Alzheimer's Disease. TRANSLATIONAL BIOPHOTONICS 2023; 5:e202300003. [PMID: 38617043 PMCID: PMC11013958 DOI: 10.1002/tbio.202300003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/22/2023] [Indexed: 04/16/2024] Open
Abstract
We analyzed ophthalmic retinal optical coherence tomography (OCT) images from patients with Alzheimer's disease (AD) to identify retinal layer thickness and ratio changes that may serve as image-based biomarkers for the disease. One three-dimensional volume before and one after diagnosis for each of 48 patients were segmented to identify retinal layer and total retinal thicknesses. Between before- and after-diagnosis retinal OCT images, there were significant thickness changes in six of ten (60%) retinal layers across all 48 patients. Through a comparison with age-matched healthy subjects, the significant changes were attributed to AD only (NFL and PR2 layers), age only (GCL, IPL, and RPE layers), or both AD and age (OPL layer). Analyzing ratios of retinal layer thicknesses, 53 of 90 (58.89%) ratios had significant changes. The four independently non-significant layers were assessed to be affected by neither AD nor age (INL layer) or both AD and age (ELM, PR1, and BM layers). The demonstrated image segmentation, measurement, and ratiometric analysis of retinal layers in AD patients may yield a noninvasive OCT image-based retinal biomarker that can be used to detect retinal changes associated with this disease.
Collapse
Affiliation(s)
- Shonit N Sharma
- Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Jordan W Marsh
- Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Michael S Tsipursky
- Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Stephen A Boppart
- Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA
- NIH Center for Label-free Imaging and Multiscale Biophotonics (CLIMB), University of Illinois Urbana-Champaign, Urbana, IL, USA
- Interdisciplinary Health Sciences Institute, University of Illinois Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
27
|
Cui K, Tang X, Hu A, Fan M, Wu P, Lu X, Lin J, Yang F, Zhao X, Huang J, Yu S, Xu Y, Liang X. Therapeutic Benefit of Melatonin in Choroidal Neovascularization During Aging Through the Regulation of Senescent Macrophage/Microglia Polarization. Invest Ophthalmol Vis Sci 2023; 64:19. [PMID: 37578424 PMCID: PMC10431207 DOI: 10.1167/iovs.64.11.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 07/27/2023] [Indexed: 08/15/2023] Open
Abstract
Purpose This study aimed to investigate the age-dependent anti-angiogenic capability of melatonin in choroidal neovascularization (CNV) and to explore the underlying molecular mechanisms. Methods In the present study, a laser-induced CNV model was established in both young (three months of age) and old (18 months of age) mice, and the size of CNV lesions and vascular leakage was detected by morphological and imaging examination. Next, Western blot and immunostaining were used to observe the levels of M2 markers, senescence-related markers, and molecules involved in IL-10/STAT3 pathway. Additionally, colivelin was used to study the effect of IL-10/STAT3 pathway activation on the expression of M2 markers and senescence-related markers by Western blot and immunostaining. Finally, the effects of colivelin on melatonin-induced reduction of CNV size and vascular leakage in mice at different ages were assessed using morphological and imaging examination. Results Our results revealed that aging promoted M2 macrophage/microglia polarization, and aggravated CNV and vascular leakage. Melatonin significantly inhibited the M2 polarization of senescent macrophage/microglia and reduced the CNV area and vascular leakage. Moreover, melatonin markedly suppressed IL-10/STAT3 pathway activation in the macrophage/microglia of old mice, and STAT3 activator colivelin reversed the suppressive effect of melatonin on M2 polarization of senescent macrophage/microglia and laser-induced CNV in old mice. Conclusions Our data demonstrated that melatonin significantly prevented the M2 polarization of senescent macrophage/microglia by inhibiting the IL-10/STAT3 pathway, and eventually attenuated senescence-associated CNV. These findings suggested that melatonin could serve as a promising therapeutic agent to treat CNV and other age-related ocular diseases.
Collapse
Affiliation(s)
- Kaixuan Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xiaoyu Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Andina Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Matthew Fan
- Yale College, Yale University, New Haven, Connecticut, United States
| | - Peiqi Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xi Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jicheng Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Fengmei Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xinyu Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jingjing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Shanshan Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yue Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xiaoling Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
28
|
Abokyi S, Ghartey-Kwansah G, Tse DYY. TFEB is a central regulator of the aging process and age-related diseases. Ageing Res Rev 2023; 89:101985. [PMID: 37321382 DOI: 10.1016/j.arr.2023.101985] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/25/2023] [Accepted: 06/12/2023] [Indexed: 06/17/2023]
Abstract
Old age is associated with a greater burden of disease, including neurodegenerative disorders such as Alzheimer's disease and Parkinson's disease, as well as other chronic diseases. Coincidentally, popular lifestyle interventions, such as caloric restriction, intermittent fasting, and regular exercise, in addition to pharmacological interventions intended to protect against age-related diseases, induce transcription factor EB (TFEB) and autophagy. In this review, we summarize emerging discoveries that point to TFEB activity affecting the hallmarks of aging, including inhibiting DNA damage and epigenetic modifications, inducing autophagy and cell clearance to promote proteostasis, regulating mitochondrial quality control, linking nutrient-sensing to energy metabolism, regulating pro- and anti-inflammatory pathways, inhibiting senescence and promoting cell regenerative capacity. Furthermore, the therapeutic impact of TFEB activation on normal aging and tissue-specific disease development is assessed in the contexts of neurodegeneration and neuroplasticity, stem cell differentiation, immune responses, muscle energy adaptation, adipose tissue browning, hepatic functions, bone remodeling, and cancer. Safe and effective strategies of activating TFEB hold promise as a therapeutic strategy for multiple age-associated diseases and for extending lifespan.
Collapse
Affiliation(s)
- Samuel Abokyi
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR of China; Research Centre for SHARP Vision, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR of China.
| | - George Ghartey-Kwansah
- Department of Biomedical Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Dennis Yan-Yin Tse
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR of China; Research Centre for SHARP Vision, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR of China; Centre for Eye and Vision Research, 17W Hong Kong Science Park, Hong Kong SAR of China.
| |
Collapse
|
29
|
Goodman D, Ness S. The Role of Oxidative Stress in the Aging Eye. Life (Basel) 2023; 13:life13030837. [PMID: 36983992 PMCID: PMC10052045 DOI: 10.3390/life13030837] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Given the expanding elderly population in the United States and the world, it is important to understand the processes underlying both natural and pathological age-related changes in the eye. Both the anterior and posterior segment of the eye undergo changes in biological, chemical, and physical properties driven by oxidative stress. With advancing age, changes in the anterior segment include dermatochalasis, blepharoptosis, thickening of the sclera, loss of corneal endothelial cells, and stiffening of the lens. Changes in the posterior segment include lowered viscoelasticity of the vitreous body, photoreceptor cell loss, and drusen deposition at the macula and fovea. Age-related ocular pathologies including glaucoma, cataracts, and age-related macular degeneration are largely mediated by oxidative stress. The prevalence of these diseases is expected to increase in the coming years, highlighting the need to develop new therapies that address oxidative stress and slow the progression of age-related pathologies.
Collapse
Affiliation(s)
- Deniz Goodman
- Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Steven Ness
- Department of Ophthalmology, Boston Medical Center, Boston, MA 02118, USA
| |
Collapse
|
30
|
Zhao B, Zhu L, Ye M, Lou X, Mou Q, Hu Y, Zhang H, Zhao Y. Oxidative stress and epigenetics in ocular vascular aging: an updated review. Mol Med 2023; 29:28. [PMID: 36849907 PMCID: PMC9972630 DOI: 10.1186/s10020-023-00624-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 02/16/2023] [Indexed: 03/01/2023] Open
Abstract
Vascular aging is an inevitable process with advancing age, which plays a crucial role in the pathogenesis of cardiovascular and microvascular diseases. Diabetic retinopathy (DR) and age-related macular degeneration (AMD), characterized by microvascular dysfunction, are the common causes of irreversible blindness worldwide, however there is still a lack of effective therapeutic strategies for rescuing the visual function. In order to develop novel treatments, it is essential to illuminate the pathological mechanisms underlying the vascular aging during DR and AMD progression. In this review, we have summarized the recent discoveries of the effects of oxidative stress and epigenetics on microvascular degeneration, which could provide potential therapeutic targets for DR and AMD.
Collapse
Affiliation(s)
- Bowen Zhao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lijia Zhu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Meng Ye
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaotong Lou
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qianxue Mou
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuanyuan Hu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hong Zhang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Yin Zhao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
31
|
Santos FM, Ciordia S, Mesquita J, Cruz C, Sousa JPCE, Passarinha LA, Tomaz CT, Paradela A. Proteomics profiling of vitreous humor reveals complement and coagulation components, adhesion factors, and neurodegeneration markers as discriminatory biomarkers of vitreoretinal eye diseases. Front Immunol 2023; 14:1107295. [PMID: 36875133 PMCID: PMC9978817 DOI: 10.3389/fimmu.2023.1107295] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 02/01/2023] [Indexed: 02/18/2023] Open
Abstract
Introduction Diabetic retinopathy (DR) and age-related macular degeneration (AMD) are leading causes of visual impairment and blindness in people aged 50 years or older in middle-income and industrialized countries. Anti-VEGF therapies have improved the management of neovascular AMD (nAMD) and proliferative DR (PDR), no treatment options exist for the highly prevalent dry form of AMD. Methods To unravel the biological processes underlying these pathologies and to find new potential biomarkers, a label-free quantitative (LFQ) method was applied to analyze the vitreous proteome in PDR (n=4), AMD (n=4) compared to idiopathic epiretinal membranes (ERM) (n=4). Results and discussion Post-hoc tests revealed 96 proteins capable of differentiating among the different groups, whereas 118 proteins were found differentially regulated in PDR compared to ERM and 95 proteins in PDR compared to dry AMD. Pathway analysis indicates that mediators of complement, coagulation cascades and acute phase responses are enriched in PDR vitreous, whilst proteins highly correlated to the extracellular matrix (ECM) organization, platelet degranulation, lysosomal degradation, cell adhesion, and central nervous system development were found underexpressed. According to these results, 35 proteins were selected and monitored by MRM (multiple reaction monitoring) in a larger cohort of patients with ERM (n=21), DR/PDR (n=20), AMD (n=11), and retinal detachment (n=13). Of these, 26 proteins could differentiate between these vitreoretinal diseases. Based on Partial least squares discriminant and multivariate exploratory receiver operating characteristic (ROC) analyses, a panel of 15 discriminatory biomarkers was defined, which includes complement and coagulation components (complement C2 and prothrombin), acute-phase mediators (alpha-1-antichymotrypsin), adhesion molecules (e.g., myocilin, galectin-3-binding protein), ECM components (opticin), and neurodegeneration biomarkers (beta-amyloid, amyloid-like protein 2).
Collapse
Affiliation(s)
- Fátima M. Santos
- CICS-UBI – Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
- Functional Proteomics Laboratory, Centro Nacional de Biotecnología, CSIC, Madrid, Spain
| | - Sergio Ciordia
- Functional Proteomics Laboratory, Centro Nacional de Biotecnología, CSIC, Madrid, Spain
| | - Joana Mesquita
- CICS-UBI – Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Carla Cruz
- CICS-UBI – Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
- Chemistry Department, Faculty of Sciences, University of Beira Interior, Covilhã, Portugal
| | - João Paulo Castro e Sousa
- CICS-UBI – Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
- Department of Ophthalmology, Centro Hospitalar de Leiria, Leiria, Portugal
| | - Luís A. Passarinha
- CICS-UBI – Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA, Caparica, Portugal
- UCIBIO–Applied Molecular Biosciences Unit, Departamento de Química/Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- Laboratório de Fármaco-Toxicologia, UBIMedical, Universidade da Beira Interior, Covilhã, Portugal
| | - Cândida T. Tomaz
- CICS-UBI – Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
- Chemistry Department, Faculty of Sciences, University of Beira Interior, Covilhã, Portugal
| | - Alberto Paradela
- Functional Proteomics Laboratory, Centro Nacional de Biotecnología, CSIC, Madrid, Spain
| |
Collapse
|
32
|
Kakihara S, Matsuda Y, Hirabayashi K, Imai A, Iesato Y, Sakurai T, Kamiyoshi A, Tanaka M, Ichikawa-Shindo Y, Kawate H, Zhao Y, Zhang Y, Guo Q, Li P, Onishi N, Murata T, Shindo T. Role of Adrenomedullin 2/Intermedin in the Pathogenesis of Neovascular Age-Related Macular Degeneration. J Transl Med 2023; 103:100038. [PMID: 36870288 DOI: 10.1016/j.labinv.2022.100038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/11/2022] [Accepted: 11/22/2022] [Indexed: 01/11/2023] Open
Abstract
Adrenomedullin 2 (AM2; also known as intermedin) is a member of the adrenomedullin (AM) peptide family. Similarly to AM, AM2 partakes in a variety of physiological activities. AM2 has been reported to exert protective effects on various organ disorders; however, its significance in the eye is unknown. We investigated the role of AM2 in ocular diseases. The receptor system of AM2 was expressed more abundantly in the choroid than in the retina. In an oxygen-induced retinopathy model, physiological and pathologic retinal angiogenesis did not differ between AM2-knockout (AM2-/-) and wild-type mice. In contrast, in laser-induced choroidal neovascularization, a model of neovascular age-related macular degeneration, AM2-/- mice had enlarged and leakier choroidal neovascularization lesions, with exacerbated subretinal fibrosis and macrophage infiltration. Contrary to this, exogenous administration of AM2 ameliorated the laser-induced choroidal neovascularization-associated pathology and suppressed gene expression associated with inflammation, fibrosis, and oxidative stress, including that of VEGF-A, VEGFR-2, CD68, CTGF, and p22-phox. The stimulation of human adult retinal pigment epithelial (ARPE) cell line 19 cells with TGF-β2 and TNF-α induced epithelial-to-mesenchymal transition (EMT), whereas AM2 expression was also elevated. The induction of EMT was suppressed when the ARPE-19 cells were pretreated with AM2. A transcriptome analysis identified 15 genes, including mesenchyme homeobox 2 (Meox2), whose expression was significantly altered in the AM2-treated group compared with that in the control group. The expression of Meox2, a transcription factor that inhibits inflammation and fibrosis, was enhanced by AM2 treatment and attenuated by endogenous AM2 knockout in the early phase after laser irradiation. The AM2 treatment of endothelial cells inhibited endothelial to mesenchymal transition and NF-κB activation; however, this effect tended to be canceled following Meox2 gene knockdown. These results indicate that AM2 suppresses the neovascular age-related macular degeneration-related pathologies partially via the upregulation of Meox2. Thus, AM2 may be a promising therapeutic target for ocular vascular diseases.
Collapse
Affiliation(s)
- Shinji Kakihara
- Department of Cardiovascular Research, Shinshu University School of Medicine, Nagano, Japan; Department of Ophthalmology, Shinshu University School of Medicine, Nagano, Japan
| | - Yorishige Matsuda
- Department of Cardiovascular Research, Shinshu University School of Medicine, Nagano, Japan; Department of Ophthalmology, Shinshu University School of Medicine, Nagano, Japan
| | - Kazutaka Hirabayashi
- Department of Cardiovascular Research, Shinshu University School of Medicine, Nagano, Japan; Department of Ophthalmology, Shinshu University School of Medicine, Nagano, Japan
| | - Akira Imai
- Department of Cardiovascular Research, Shinshu University School of Medicine, Nagano, Japan; Department of Ophthalmology, Shinshu University School of Medicine, Nagano, Japan
| | - Yasuhiro Iesato
- Department of Cardiovascular Research, Shinshu University School of Medicine, Nagano, Japan; Department of Ophthalmology, Shinshu University School of Medicine, Nagano, Japan
| | - Takayuki Sakurai
- Department of Cardiovascular Research, Shinshu University School of Medicine, Nagano, Japan; Department of Life Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, Japan
| | - Akiko Kamiyoshi
- Department of Cardiovascular Research, Shinshu University School of Medicine, Nagano, Japan; Department of Life Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, Japan
| | - Megumu Tanaka
- Department of Cardiovascular Research, Shinshu University School of Medicine, Nagano, Japan
| | - Yuka Ichikawa-Shindo
- Department of Cardiovascular Research, Shinshu University School of Medicine, Nagano, Japan
| | - Hisaka Kawate
- Department of Cardiovascular Research, Shinshu University School of Medicine, Nagano, Japan
| | - Yunlu Zhao
- Department of Cardiovascular Research, Shinshu University School of Medicine, Nagano, Japan
| | - Yan Zhang
- Department of Cardiovascular Research, Shinshu University School of Medicine, Nagano, Japan
| | - QianQian Guo
- Department of Cardiovascular Research, Shinshu University School of Medicine, Nagano, Japan
| | - Peixuan Li
- Department of Cardiovascular Research, Shinshu University School of Medicine, Nagano, Japan
| | - Naho Onishi
- Department of Cardiovascular Research, Shinshu University School of Medicine, Nagano, Japan
| | - Toshinori Murata
- Department of Ophthalmology, Shinshu University School of Medicine, Nagano, Japan
| | - Takayuki Shindo
- Department of Cardiovascular Research, Shinshu University School of Medicine, Nagano, Japan; Department of Life Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, Japan.
| |
Collapse
|
33
|
García-Montalvo IA, Matías-Pérez D, Hernández-Bautista E, Pérez-Campos E. Inclusion of carotenoids in dietary habits as an alternative to prevent age-related macular degeneration. Front Nutr 2023; 9:1063517. [PMID: 36698471 PMCID: PMC9868752 DOI: 10.3389/fnut.2022.1063517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/14/2022] [Indexed: 01/11/2023] Open
Affiliation(s)
- Iván Antonio García-Montalvo
- Division of Graduate Studies and Research, National Institute of Technology of Mexico/Technological Institute of Oaxaca, Oaxaca, Mexico
| | | | | | | |
Collapse
|
34
|
Saddala MS, Mundla S, Patyal N, Dash S. Single-Cell RNA Sequencing (scRNA-Seq) Data Analysis of Retinal Homeostasis and Degeneration of Microglia. Methods Mol Biol 2023; 2678:91-106. [PMID: 37326706 DOI: 10.1007/978-1-0716-3255-0_6] [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: 06/17/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) experiment reveals previously unseen molecular features. The number of sequencing procedures and computational data analysis approaches has been increasing rapidly in recent years. This chapter provides a general idea of the single-cell data analysis and visualization. An introduction and practical guidance for the 10× sequencing data analysis and visualization are presented. Basic data analysis approaches are highlighted, followed by quality control of data, filtering in cell level and gene level, normalization, dimensional reduction, clustering analysis, and marker identification.
Collapse
Affiliation(s)
- Madhu Sudhana Saddala
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Srilatha Mundla
- Department of Biotechnology, Sri Venkateswara University, Tirupati, AP, India
| | - Naina Patyal
- Department of Bioinformatics, Central University of Himachal Pradesh, Kangra, HP, India
| | - Srujanika Dash
- Department of Bioinformatics, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha, India
| |
Collapse
|
35
|
Lin R, Yu J. The role of NAD + metabolism in macrophages in age-related macular degeneration. Mech Ageing Dev 2023; 209:111755. [PMID: 36435209 DOI: 10.1016/j.mad.2022.111755] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/05/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022]
Abstract
Age-related macular degeneration (AMD) is a leading cause of legal blindness and moderate and severe vision impairment (MSVI) in people older than 50 years. It is classified in various stages including early, intermediate, and late stage. In the early stages, innate immune system, especially macrophages, play an essential part in disease onset and progression. NAD+ is an essential coenzyme involved in cellular senescence and immune cell function, and its role in age-related diseases is gaining increasing attention. The imbalance between the NAD+ synthesis and consumption causes the fluctuation of intracellular NAD+ level which determines the polarization fate of macrophages. In AMD, the over-expression of NAD+-consuming enzymes in macrophages leads to declining of NAD+ concentrations in the microenvironment. This phenomenon triggers the activation of inflammatory pathways in macrophages, positive feedback aggregation of inflammatory cells and accumulation of reactive oxygen species (ROS). This review details the role of NAD+ metabolism in macrophages and molecular mechanisms during AMD. The selected pathways were identified as potential targets for intervention in AMD, pending further investigation.
Collapse
Affiliation(s)
- Ruoyi Lin
- Department of Ophthalmology, the Tenth People's Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai 200072, China
| | - Jing Yu
- Department of Ophthalmology, the Tenth People's Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai 200072, China; Department of Ophthalmology, Bengbu Third People's Hospital, Bengbu, Anhui 233099, China.
| |
Collapse
|
36
|
Aggio-Bruce R, Schumann U, Cioanca AV, Chen FK, McLenachan S, Heath Jeffery RC, Das S, Natoli R. Serum miRNA modulations indicate changes in retinal morphology. Front Mol Neurosci 2023; 16:1130249. [PMID: 36937046 PMCID: PMC10020626 DOI: 10.3389/fnmol.2023.1130249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
Background Age-related macular degeneration (AMD) is the leading cause of vision loss in the developed world and the detection of its onset and progression are based on retinal morphological assessments. MicroRNA (miRNA) have been explored extensively as biomarkers for a range of neurological diseases including AMD, however differences in experimental design and the complexity of human biology have resulted in little overlap between studies. Using preclinical animal models and clinical samples, this study employs a novel approach to determine a serum signature of AMD progression. Methods Serum miRNAs were extracted from mice exposed to photo-oxidative damage (PD; 0, 1, 3 and 5 days), and clinical samples from patients diagnosed with reticular pseudodrusen or atrophic AMD. The expression of ~800 miRNAs was measured using OpenArray™, and differential abundance from controls was determined using the HTqPCR R package followed by pathway analysis with DAVID. MiRNA expression changes were compared against quantifiable retinal histological indicators. Finally, the overlap of miRNA changes observed in the mouse model and human patient samples was investigated. Results Differential miRNA abundance was identified at all PD time-points and in clinical samples. Importantly, these were associated with inflammatory pathways and histological changes in the retina. Further, we were able to align findings in the mouse serum to those of clinical patients. Conclusion In conclusion, serum miRNAs are a valid tool as diagnostics for the early detection of retinal degeneration, as they reflect key changes in retinal health. The combination of pre-clinical animal models and human patient samples led to the identification of a preliminary serum miRNA signature for AMD. This study is an important platform for the future development of a diagnostic serum miRNA panel for the early detection of retinal degeneration.
Collapse
Affiliation(s)
- Riemke Aggio-Bruce
- The John Curtin School of Medical Research, The Australian National University, Acton, ACT, Australia
- The School of Medicine and Psychology, Acton, ACT, Australia
| | - Ulrike Schumann
- The John Curtin School of Medical Research, The Australian National University, Acton, ACT, Australia
- The Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Adrian V. Cioanca
- The John Curtin School of Medical Research, The Australian National University, Acton, ACT, Australia
| | - Fred K. Chen
- Centre of Ophthalmology and Visual Science, The University of Western Australia, Perth, WA, Australia
- Lions Eye Institute, Perth, WA, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
| | - Samuel McLenachan
- Centre of Ophthalmology and Visual Science, The University of Western Australia, Perth, WA, Australia
- Lions Eye Institute, Perth, WA, Australia
| | - Rachael C. Heath Jeffery
- Centre of Ophthalmology and Visual Science, The University of Western Australia, Perth, WA, Australia
- Lions Eye Institute, Perth, WA, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
| | - Shannon Das
- The John Curtin School of Medical Research, The Australian National University, Acton, ACT, Australia
| | - Riccardo Natoli
- The John Curtin School of Medical Research, The Australian National University, Acton, ACT, Australia
- The School of Medicine and Psychology, Acton, ACT, Australia
- *Correspondence: Riccardo Natoli,
| |
Collapse
|
37
|
Wilson GA, Cheyne K, Ramrakha S, Ambler A, Tan GS, Caspi A, Williams B, Sugden K, Houts R, Niederer RL, Wong TY, Moffitt TE, Poulton R. Are macular drusen in midlife a marker of accelerated biological ageing? Clin Exp Optom 2023; 106:41-46. [PMID: 34902293 DOI: 10.1080/08164622.2021.2012428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/04/2021] [Accepted: 11/15/2021] [Indexed: 01/13/2023] Open
Abstract
CLINICAL RELEVANCE Macular drusen are associated with age-related maculopathy but are not an ocular manifestation or biomarker of systemic ageing. BACKGROUND Macular drusen are the first sign of age-related maculopathy, an eye disease for which age is the strongest risk factor. The aim of this cohort study was to investigate whether macular drusen in midlife - a sign of the earliest stages of age-related macular degeneration (AMD) - are associated with accelerated biological ageing more generally. METHODS Members of the long-running Dunedin Multidisciplinary Health and Development Study (hereafter the Dunedin Study, n = 1037) underwent retinal photography at their most recent assessment at the age of 45 years. Images were graded for the presence of AMD using a simplified scale from the Age-Related Eye Disease Study (AREDS). Accelerated ageing was assessed by (i) a measure of Pace of Ageing defined from a combination of clinical and serum biomarkers obtained at ages 26, 32, 38, and 45 years and (ii) Facial Ageing, defined from photographs obtained at age 38 and 45 years. RESULTS Of the 938 participants who participated at the age 45 assessments, 834 had gradable retinal photographs, and of these 165 (19.8%) had macular drusen. There was no significant difference in Pace of Ageing (p = .743) or Facial Ageing (p = .945) among participants with and without macular drusen. CONCLUSIONS In this representative general population sample, macular drusen in midlife were not associated with accelerated ageing. Future studies tracking longitudinal changes in drusen number and severity at older ages may reveal whether drusen are a biomarker of accelerated ageing.
Collapse
Affiliation(s)
- Graham A Wilson
- Dunedin Multidisciplinary Health and Development Research Unit, Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Kirsten Cheyne
- Dunedin Multidisciplinary Health and Development Research Unit, Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Sandhya Ramrakha
- Dunedin Multidisciplinary Health and Development Research Unit, Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Antony Ambler
- Dunedin Multidisciplinary Health and Development Research Unit, Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Gavin Sw Tan
- Singapore Eye Research Institute, Singapore National Eye Centre, Duke-NUS Medical School, National University of Singapore, Singapore
| | - Avshalom Caspi
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina, USA
| | - Ben Williams
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina, USA
| | - Karen Sugden
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina, USA
| | - Renate Houts
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina, USA
| | - Rachael L Niederer
- Department of Ophthalmology, University of Auckland, Auckland, New Zealand
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Duke-NUS Medical School, National University of Singapore, Singapore
| | - Terrie E Moffitt
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina, USA
| | - Richie Poulton
- Dunedin Multidisciplinary Health and Development Research Unit, Department of Psychology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
38
|
Dos Santos FM, Ciordia S, Mesquita J, de Sousa JPC, Paradela A, Tomaz CT, Passarinha LAP. Vitreous humor proteome: unraveling the molecular mechanisms underlying proliferative and neovascular vitreoretinal diseases. Cell Mol Life Sci 2022; 80:22. [PMID: 36585968 PMCID: PMC11072707 DOI: 10.1007/s00018-022-04670-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 11/09/2022] [Accepted: 12/12/2022] [Indexed: 01/01/2023]
Abstract
Proliferative diabetic retinopathy (PDR), proliferative vitreoretinopathy (PVR), and neovascular age-related macular degeneration (nAMD) are among the leading causes of blindness. Due to the multifactorial nature of these vitreoretinal diseases, omics approaches are essential for a deeper understanding of the pathophysiologic processes underlying the evolution to a proliferative or neovascular etiology, in which patients suffer from an abrupt loss of vision. For many years, it was thought that the function of the vitreous was merely structural, supporting and protecting the surrounding ocular tissues. Proteomics studies proved that vitreous is more complex and biologically active than initially thought, and its changes reflect the physiological and pathological state of the eye. The vitreous is the scenario of a complex interplay between inflammation, fibrosis, oxidative stress, neurodegeneration, and extracellular matrix remodeling. Vitreous proteome not only reflects the pathological events that occur in the retina, but the changes in the vitreous itself play a central role in the onset and progression of vitreoretinal diseases. Therefore, this review offers an overview of the studies on the vitreous proteome that could help to elucidate some of the pathological mechanisms underlying proliferative and/or neovascular vitreoretinal diseases and to find new potential pharmaceutical targets.
Collapse
Affiliation(s)
- Fátima Milhano Dos Santos
- Health Sciences Research Centre (CICS-UBI), Universidade da Beira Interior, 6201-001, Covilhã, Portugal.
- Functional Proteomics Laboratory, Centro Nacional de Biotecnología (CNB-CSIC), Unidad de Proteomica, Calle Darwin 3, Campus de Cantoblanco, 28049, Madrid, Spain.
| | - Sergio Ciordia
- Functional Proteomics Laboratory, Centro Nacional de Biotecnología (CNB-CSIC), Unidad de Proteomica, Calle Darwin 3, Campus de Cantoblanco, 28049, Madrid, Spain
| | - Joana Mesquita
- Health Sciences Research Centre (CICS-UBI), Universidade da Beira Interior, 6201-001, Covilhã, Portugal
| | - João Paulo Castro de Sousa
- Health Sciences Research Centre (CICS-UBI), Universidade da Beira Interior, 6201-001, Covilhã, Portugal
- Department of Ophthalmology, Centro Hospitalar de Leiria, 2410-197, Leiria, Portugal
| | - Alberto Paradela
- Functional Proteomics Laboratory, Centro Nacional de Biotecnología (CNB-CSIC), Unidad de Proteomica, Calle Darwin 3, Campus de Cantoblanco, 28049, Madrid, Spain
| | - Cândida Teixeira Tomaz
- Health Sciences Research Centre (CICS-UBI), Universidade da Beira Interior, 6201-001, Covilhã, Portugal
- C4-UBI, Cloud Computing Competence Centre, University of Beira Interior, 6200-501, Covilhã, Portugal
- Chemistry Department, Faculty of Sciences, Universidade da Beira Interior, 6201-001, Covilhã, Portugal
| | - Luís António Paulino Passarinha
- Health Sciences Research Centre (CICS-UBI), Universidade da Beira Interior, 6201-001, Covilhã, Portugal.
- Associate Laboratory i4HB, Faculdade de Ciências e Tecnologia, Institute for Health and Bioeconomy, Universidade NOVA, 2819-516, Caparica, Portugal.
- UCIBIO-Applied Molecular Biosciences Unit, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal.
- Pharmaco-Toxicology Laboratory, UBIMedical, Universidade da Beira Interior, 6200-000, Covilhã, Portugal.
| |
Collapse
|
39
|
Hess K, de Silva T, Grisso P, Wiley H, Thavikulwat AT, Keenan TDL, Chew EY, Cukras CA. Evaluation of Cone- and Rod-Mediated Parameters in Dark Adaptation Testing as Outcome Measures in Age-Related Macular Degeneration. Ophthalmol Retina 2022; 6:1173-1184. [PMID: 35643387 DOI: 10.1016/j.oret.2022.05.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 06/15/2023]
Abstract
PURPOSE To investigate the suitability of 6 rod- or cone-mediated dark adaptation (DA) parameters as outcome measures for clinical trials in age-related macular degeneration (AMD), including their retest reliability, association with age and disease severity, and measurable longitudinal change over time. DESIGN Prospective, longitudinal study (Clinicaltrials.gov: NCT01352975). PARTICIPANTS A total of 191 patients with AMD and older participants followed longitudinally over 5 years. METHODS Dark adaptation testing was performed using the AdaptDx dark adaptometer with a maximum test time of 40 minutes. A 2-part exponential-linear curve was fitted to obtain values for cone decay, cone plateau, time to rod-cone break, rod intercept time (RIT), rod adaptation rate (S2), and area under the curve. Intersession retest reliability was assessed in tests performed within 2 weeks using the Bland-Altman analysis. The relationship of DA parameters with age, AMD severity, and reticular pseudodrusen (RPD) presence was evaluated using linear mixed models. MAIN OUTCOME MEASURES Retest reliability, association with disease severity, and longitudinal change of 6 DA parameters. RESULTS A total of 1329 DA curves were analyzed. Rod intercept time was the parameter that showed the greatest reliability (intraclass correlation coefficient of 0.88) and greatest association with age, AMD severity, and RPD (marginal R2 of 0.38), followed by the rod-mediated parameters area under the curve and rod-cone break. Cone plateau appeared constant at lower RIT values but increased with progressive rod dysfunction (RIT > 22.8 minutes) with a slope of 0.07 log units per 10 minutes RIT prolongation. Therefore, it might provide additional information in the advanced stages of AMD. CONCLUSIONS Age-related macular degeneration severity and RPD presence are each associated with large differences in multiple DA curve parameters. In addition, substantial differences in some parameters occur with age, even accounting for AMD severity and RPD status. This supports the 2-hit hypothesis of age and disease status on DA (and perhaps AMD pathophysiology itself). Of the DA parameters, RIT has the highest retest reliability, closest correlation with AMD severity and RPD, and largest longitudinal changes. This underscores the suitability of RIT as an outcome measure in clinical trials. The cone plateau increases only in advanced stages of kinetic rod dysfunction, indicating rod dysfunction preceding cone dysfunction and degeneration in the temporal sequence of pathology in AMD.
Collapse
Affiliation(s)
- Kristina Hess
- Unit on Clinical Investigation of Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland; Department of Ophthalmology, University Hospital Bonn, Bonn, Germany
| | - Tharindu de Silva
- Unit on Clinical Investigation of Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Peyton Grisso
- Unit on Clinical Investigation of Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Henry Wiley
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Alisa T Thavikulwat
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Tiarnan D L Keenan
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Emily Y Chew
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Catherine A Cukras
- Unit on Clinical Investigation of Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
40
|
Fietz A, Hurst J, Schnichels S. Out of the Shadow: Blue Light Exposure Induces Apoptosis in Müller Cells. Int J Mol Sci 2022; 23:ijms232314540. [PMID: 36498867 PMCID: PMC9739907 DOI: 10.3390/ijms232314540] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
Awareness toward the risks of blue light (BL) exposure is rising due to increased use of BL-enriched LEDs in displays. Short-wave BL (400-500 nm) has a high photochemical energy, leading to the enhanced production of reactive oxygen species (ROS). BL potentially plays a role in causing dry eye, cataracts, and age-related macular degeneration (AMD). The effect of BL on retinal pigment epithelium cells (RPEs) or photoreceptors has been extensively investigated. In contrast, only a few studies have investigated the effects of BL exposure on Müller cells (MCs). This is mainly due to their lack of photosensitive elements and the common assumption that their reaction to stress is only secondary in disease development. However, MCs perform important supportive, secretory, and immune functions in the retina, making them essential for retinal survival. Increased oxidative stress is a key player in many retinal diseases such as AMD or glaucoma. We hypothesize that increased oxidative stress can also affect MCs. Thus, we simulated oxidative stress levels by exposing primary porcine MCs and human MIO-M1 cells to BL. To confirm the wavelength-specificity, the cells were further exposed to red (RL), purple (PL), and white light (WL). BL and WL exposure increased ROS levels, but only BL exposure led to apoptosis in primary MCs. Thus, BL accounted for the harmful part of WL exposure. When cells were simultaneously exposed to BL and RL (i.e., PL), cell damage due to BL could be partly prevented, as could the inhibition of p53, demonstrating the protective effect of RL and p53 dependency. In contrast, BL hardly induced apoptosis in MIO-M1 cells, which is likely due to the immortalization of the cells. Therefore, enhanced oxidative stress levels can significantly harm MC function, probably leading to decreased retinal survival and, thus, further enhancing the progression of retinal diseases. Preventing the cell death of these essential retinal cells represents a promising therapy option to enhance retinal survival.
Collapse
|
41
|
Palevski D, Ben-David G, Weinberger Y, Haj Daood R, Fernández JA, Budnik I, Levy-Mendelovich S, Kenet G, Nisgav Y, Weinberger D, Griffin JH, Livnat T. 3K3A-Activated Protein C Prevents Microglia Activation, Inhibits NLRP3 Inflammasome and Limits Ocular Inflammation. Int J Mol Sci 2022; 23:ijms232214196. [PMID: 36430674 PMCID: PMC9694680 DOI: 10.3390/ijms232214196] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/07/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
3K3A-Activated Protein C (APC) is a recombinant variant of the physiological anticoagulant APC with pleiotropic cytoprotective properties albeit without the bleeding risks. The anti-inflammatory activities of 3K3A-APC were demonstrated in multiple preclinical injury models, including various neurological disorders. We determined the ability of 3K3A-APC to inhibit ocular inflammation in a murine model of lipopolysaccharide (LPS)-induced uveitis. Leukocyte recruitment, microglia activation, NLRP3 inflammasome and IL-1β levels were assessed using flow cytometry, retinal cryosection histology, retinal flatmount immunohistochemistry and vascular imaging, with and without 3K3A-APC treatment. LPS triggered robust inflammatory cell recruitment in the posterior chamber. The 3K3A-APC treatment significantly decreased leukocyte numbers and inhibited leukocyte extravasation from blood vessels into the retinal parenchyma to a level similar to controls. Resident microglia, which underwent an inflammatory transition following LPS injection, remained quiescent in eyes treated with 3K3A-APC. An inflammation-associated increase in retinal thickness, observed in LPS-injected eyes, was diminished by 3K3A-APC treatment, suggesting its clinical relevancy. Finally, 3K3A-APC treatment inhibited inflammasome activation, determined by lower levels of NLRP3 and its downstream effector IL-1β. Our results highlight the anti-inflammatory properties of 3K3A-APC in ocular inflammation and suggest its potential use as a novel treatment for retinal diseases associated with inflammation.
Collapse
Affiliation(s)
- Dahlia Palevski
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Petah-Tikva 49100, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Gil Ben-David
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Petah-Tikva 49100, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Yehonatan Weinberger
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Petah-Tikva 49100, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Rabeei Haj Daood
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Petah-Tikva 49100, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - José A. Fernández
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ivan Budnik
- Sheba Medical Center, The Amalia Biron Thrombosis Research Institute, Tel-Hashomer 52621, Israel
| | - Sarina Levy-Mendelovich
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
- Sheba Medical Center, The Amalia Biron Thrombosis Research Institute, Tel-Hashomer 52621, Israel
| | - Gili Kenet
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
- Sheba Medical Center, The Amalia Biron Thrombosis Research Institute, Tel-Hashomer 52621, Israel
| | - Yael Nisgav
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Petah-Tikva 49100, Israel
| | - Dov Weinberger
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Petah-Tikva 49100, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - John H. Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Tami Livnat
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Petah-Tikva 49100, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
- Sheba Medical Center, The Amalia Biron Thrombosis Research Institute, Tel-Hashomer 52621, Israel
- Correspondence:
| |
Collapse
|
42
|
Karunadharma PP, Kapphahn RJ, Stahl MR, Olsen TW, Ferrington DA. Dissecting Regulators of Aging and Age-Related Macular Degeneration in the Retinal Pigment Epithelium. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6009787. [PMID: 36439688 PMCID: PMC9683958 DOI: 10.1155/2022/6009787] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022]
Abstract
Age-related macular degeneration (AMD), the leading cause of blindness in elderly populations, involves the loss of central vision due to progressive dysfunction of the retinal pigment epithelium (RPE) and subsequent loss of light-sensing photoreceptors. While age is a key risk factor, not every aged individual develops AMD. Thus, the critical question is what specific cellular changes tip the balance from healthy aging to disease. To distinguish between changes associated with aging and AMD, we compared the RPE proteome in human eye bank tissue from nondiseased donors during aging (n = 50, 29-91 years) and in donors with AMD (n = 36) compared to age-matched donors without disease (n = 28). Proteins from RPE cells were separated on two-dimensional gels, analyzed for content, and identified using mass spectrometry. A total of 58 proteins displayed significantly altered content with either aging or AMD. Proteins involved in metabolism, protein turnover, stress response, and cell death were altered with both aging and AMD. However, the direction of change was predominantly opposite. With aging, we detected an overall decrease in metabolism and reductions in stress-associated proteins, proteases, and chaperones. With AMD, we observed upregulation of metabolic proteins involved in glycolysis, TCA, and fatty acid metabolism, with a concurrent decline in oxidative phosphorylation, suggesting a reprogramming of energy utilization. Additionally, we detected upregulation of proteins involved in the stress response and protein turnover. Predicted upstream regulators also showed divergent results, with inhibition of inflammation and immune response with aging and activation of these processes with AMD. Our results support the idea that AMD is not simply advanced aging but rather the culmination of perturbed protein homeostasis, defective bioenergetics, and increased oxidative stress within the aging RPE, exacerbated by environmental factors and the genetic background of an individual.
Collapse
Affiliation(s)
- Pabalu P. Karunadharma
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota Twin Cities, MN 55455, USA
- Graduate Program in Biochemistry, Molecular Biology, And Biophysics, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Rebecca J. Kapphahn
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota Twin Cities, MN 55455, USA
| | - Madilyn R. Stahl
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota Twin Cities, MN 55455, USA
| | - Timothy W. Olsen
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota Twin Cities, MN 55455, USA
| | - Deborah A. Ferrington
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota Twin Cities, MN 55455, USA
- Graduate Program in Biochemistry, Molecular Biology, And Biophysics, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| |
Collapse
|
43
|
Dhirachaikulpanich D, Lagger C, Chatsirisupachai K, de Magalhães JP, Paraoan L. Intercellular communication analysis of the human retinal pigment epithelial and choroidal cells predicts pathways associated with aging, cellular senescence and age-related macular degeneration. Front Aging Neurosci 2022; 14:1016293. [PMID: 36408112 PMCID: PMC9669800 DOI: 10.3389/fnagi.2022.1016293] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
The retinal pigment epithelium (RPE) and the choroid are ocular tissues with fundamental roles in supporting neuroretinal function. The pathogenesis of age-related macular degeneration (AMD), a leading cause of irreversible blindness for which aging is the highest risk factor is closely linked with progressive impairment of various functions of these tissues. Cellular senescence, marked by cell cycle arrest and secretion of proinflammatory factors, is known to be associated with aging and has been proposed as a potential driver of AMD. Here, we investigated the role played by intercellular communication in the RPE/choroid within the context of aging, senescence and AMD. We inferred cell–cell interactions in the RPE/choroid by applying CellChat and scDiffCom on a publicly available scRNA-seq dataset from three human donors with and without AMD. We identified age-regulated ligand and receptor genes by using limma on a separate publicly available bulk microarray dataset providing RPE/choroid samples at multiple time points. Cellular senescence was investigated by assigning a score to each cell and each sample of these scRNA-seq and microarray datasets, respectively, based on the expression of key signature genes determined by a previous senescence meta-analysis. We identified VEGF-, BMP-and tenascin-mediated pathways supporting some of the strongest cell–cell interactions between RPE cells, fibroblasts and choroidal endothelial cells and as strong intercellular communication pathways related to both aging and senescence. Their signaling strength was enhanced between subpopulations of cells having high senescence scores. Predominant ligands of these pathways were upregulated with age whereas predominant receptors were downregulated. Globally, we also observed that cells from AMD samples presented slightly bigger senescence scores than normal cells and that the senescence score positively correlated with age in bulk samples (R = 0.26, value of p < 0.01). Hence, our analysis provides novel information on RPE/choroid intercellular communication that gives insights into the connection between aging, senescence and AMD.
Collapse
Affiliation(s)
- Dhanach Dhirachaikulpanich
- Ocular Molecular Biology and Mechanisms of Disease Group, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Cyril Lagger
- Integrative Genomics of Ageing Group, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Kasit Chatsirisupachai
- Integrative Genomics of Ageing Group, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- *Correspondence: João Pedro de Magalhães,
| | - Luminita Paraoan
- Ocular Molecular Biology and Mechanisms of Disease Group, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- Luminita Paraoan,
| |
Collapse
|
44
|
Geng Z, Ling L, Li B, Yuan L, Zhang B. Electroacupuncture for blindness in age-related macular degeneration: a case report. Acupunct Med 2022; 40:496-497. [PMID: 35765832 DOI: 10.1177/09645284221105528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Zixiang Geng
- Department of Acupuncture and Moxibustion, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lele Ling
- Department of Acupuncture and Moxibustion, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bingrong Li
- Department of Acupuncture and Moxibustion, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Long Yuan
- Department of Acupuncture and Moxibustion, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bimeng Zhang
- Department of Acupuncture and Moxibustion, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
45
|
Martínez-Gil N, Maneu V, Kutsyr O, Fernández-Sánchez L, Sánchez-Sáez X, Sánchez-Castillo C, Campello L, Lax P, Pinilla I, Cuenca N. Cellular and molecular alterations in neurons and glial cells in inherited retinal degeneration. Front Neuroanat 2022; 16:984052. [PMID: 36225228 PMCID: PMC9548552 DOI: 10.3389/fnana.2022.984052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/29/2022] [Indexed: 11/19/2022] Open
Abstract
Multiple gene mutations have been associated with inherited retinal dystrophies (IRDs). Despite the spectrum of phenotypes caused by the distinct mutations, IRDs display common physiopathology features. Cell death is accompanied by inflammation and oxidative stress. The vertebrate retina has several attributes that make this tissue vulnerable to oxidative and nitrosative imbalance. The high energy demands and active metabolism in retinal cells, as well as their continuous exposure to high oxygen levels and light-induced stress, reveal the importance of tightly regulated homeostatic processes to maintain retinal function, which are compromised in pathological conditions. In addition, the subsequent microglial activation and gliosis, which triggers the secretion of pro-inflammatory cytokines, chemokines, trophic factors, and other molecules, further worsen the degenerative process. As the disease evolves, retinal cells change their morphology and function. In disease stages where photoreceptors are lost, the remaining neurons of the retina to preserve their function seek out for new synaptic partners, which leads to a cascade of morphological alterations in retinal cells that results in a complete remodeling of the tissue. In this review, we describe important molecular and morphological changes in retinal cells that occur in response to oxidative stress and the inflammatory processes underlying IRDs.
Collapse
Affiliation(s)
- Natalia Martínez-Gil
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Victoria Maneu
- Department of Optics, Pharmacology and Anatomy, University of Alicante, Alicante, Spain
| | - Oksana Kutsyr
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | | | - Xavier Sánchez-Sáez
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Carla Sánchez-Castillo
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Laura Campello
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Pedro Lax
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - Isabel Pinilla
- Aragón Institute for Health Research (IIS Aragón), Zaragoza, Spain
- Department of Ophthalmology, Lozano Blesa University Hospital, Zaragoza, Spain
- Department of Surgery, University of Zaragoza, Zaragoza, Spain
- Isabel Pinilla,
| | - Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Institute Ramón Margalef, University of Alicante, Alicante, Spain
- *Correspondence: Nicolás Cuenca,
| |
Collapse
|
46
|
Shughoury A, Sevgi DD, Ciulla TA. Molecular Genetic Mechanisms in Age-Related Macular Degeneration. Genes (Basel) 2022; 13:1233. [PMID: 35886016 PMCID: PMC9316037 DOI: 10.3390/genes13071233] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/30/2022] [Accepted: 07/05/2022] [Indexed: 11/29/2022] Open
Abstract
Age-related macular degeneration (AMD) is among the leading causes of irreversible blindness worldwide. In addition to environmental risk factors, such as tobacco use and diet, genetic background has long been established as a major risk factor for the development of AMD. However, our ability to predict disease risk and personalize treatment remains limited by our nascent understanding of the molecular mechanisms underlying AMD pathogenesis. Research into the molecular genetics of AMD over the past two decades has uncovered 52 independent gene variants and 34 independent loci that are implicated in the development of AMD, accounting for over half of the genetic risk. This research has helped delineate at least five major pathways that may be disrupted in the pathogenesis of AMD: the complement system, extracellular matrix remodeling, lipid metabolism, angiogenesis, and oxidative stress response. This review surveys our current understanding of each of these disease mechanisms, in turn, along with their associated pathogenic gene variants. Continued research into the molecular genetics of AMD holds great promise for the development of precision-targeted, personalized therapies that bring us closer to a cure for this debilitating disease.
Collapse
Affiliation(s)
- Aumer Shughoury
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.S.); (D.D.S.)
| | - Duriye Damla Sevgi
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.S.); (D.D.S.)
| | - Thomas A. Ciulla
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.S.); (D.D.S.)
- Clearside Biomedical, Inc., Alpharetta, GA 30005, USA
- Midwest Eye Institute, Indianapolis, IN 46290, USA
| |
Collapse
|
47
|
Yang S, Li T, Jia H, Gao M, Li Y, Wan X, Huang Z, Li M, Zhai Y, Li X, Yang X, Wang T, Liang J, Gu Q, Luo X, Qian L, Lu S, Liu J, Song Y, Wang F, Sun X, Yu D. Targeting C3b/C4b and VEGF with a bispecific fusion protein optimized for neovascular age-related macular degeneration therapy. Sci Transl Med 2022; 14:eabj2177. [PMID: 35648811 DOI: 10.1126/scitranslmed.abj2177] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Antiangiogenesis therapies targeting vascular endothelial growth factor (VEGF) have revolutionized the treatment of neovascular ocular diseases, including neovascular age-related macular degeneration (nAMD). Compelling evidence has implicated the vital role of complement system dysregulation in AMD pathogenesis, implying it as a potential therapeutic strategy for geographic atrophy in dry AMD and to enhance the efficacy of anti-VEGF monotherapies in nAMD. This study reports the preclinical assessment and phase 1 clinical outcomes of a bispecific fusion protein, efdamrofusp alfa (code: IBI302), which is capable of neutralizing both VEGF isoforms and C3b/C4b. Efdamrofusp alfa showed superior efficacy over anti-VEGF monotherapy in a mouse laser-induced choroidal neovascularization (CNV) model after intravitreal delivery. Dual inhibition of VEGF and the complement activation was found to further inhibit macrophage infiltration and M2 macrophage polarization. Intravitreal efdamrofusp alfa demonstrated favorable safety profiles and exhibited antiangiogenetic efficacy in a nonhuman primate laser-induced CNV model. A phase 1 dose-escalating clinical trial (NCT03814291) was thus conducted on the basis of the preclinical data. Preliminary results showed that efdamrofusp alfa was well tolerated in patients with nAMD. These data suggest that efdamrofusp alfa might be effective for treating nAMD and possibly other complement-related ocular conditions.
Collapse
Affiliation(s)
- Shiqi Yang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Ophthalmic Diseases, Shanghai 200080, China
| | - Tong Li
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Ophthalmic Diseases, Shanghai 200080, China
| | - Huixun Jia
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Ophthalmic Diseases, Shanghai 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
| | - Min Gao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yiming Li
- Innovent Biologics Inc., Suzhou 215000, China
| | - Xiaoling Wan
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China
| | - Zhen Huang
- Department of Ophthalmology, Wuhan General Hospital of Guangzhou Military Region, Wuhan 430070, China
| | - Min Li
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China
| | - Yuanqi Zhai
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China
| | - Xiaomeng Li
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Ophthalmic Diseases, Shanghai 200080, China
| | - Xiaotong Yang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Ophthalmic Diseases, Shanghai 200080, China
| | - Tao Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China
| | - Jian Liang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China
| | - Qing Gu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China
| | - Xueting Luo
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China
| | - Lei Qian
- Innovent Biologics Inc., Suzhou 215000, China
| | - Shujie Lu
- Innovent Biologics Inc., Suzhou 215000, China
| | - Junjian Liu
- Innovent Biologics Inc., Suzhou 215000, China
| | - Yanping Song
- Department of Ophthalmology, Wuhan General Hospital of Guangzhou Military Region, Wuhan 430070, China
| | - Fenghua Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Ophthalmic Diseases, Shanghai 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Ophthalmic Diseases, Shanghai 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China
| | - Dechao Yu
- Innovent Biologics Inc., Suzhou 215000, China
| |
Collapse
|
48
|
Garcia-Garcia J, Usategui-Martin R, Sanabria MR, Fernandez-Perez E, Telleria JJ, Coco-Martin RM. Pathophysiology of Age-Related Macular Degeneration: Implications for Treatment. Ophthalmic Res 2022; 65:615-636. [PMID: 35613547 DOI: 10.1159/000524942] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/29/2022] [Indexed: 12/16/2022]
Abstract
Age-related macular degeneration (AMD) is a complex, multifactorial, progressive retinal disease that affects millions of people worldwide and has become the leading cause of visual impairment in developed countries. The disease etiopathogenesis is not understood fully, although many triggers and processes that lead to dysfunction and degeneration of the retinal pigment epithelium (RPE) have already been identified. Thus, the lack of cellular control of oxidative stress, altered proteostasis, dysfunction of lipid homeostasis, and mitochondrial dysfunction form an internal feedback loop that causes the RPE to fail and allows accumulation of abnormal misfolded proteins and abnormal lipids that will form drusen. An inadequate antioxidant response, deficits in autophagy mechanisms, and dysregulation of the extracellular matrix (ECM) help to increase the deposition of abnormal drusen material over time. The drusen then act as inflammatory centers that trigger chronic inflammation of the subretinal space in which microglia and recruited macrophages are also involved, and where the complement system is a key component. Choriocapillaris degeneration and nutritional influences are also classic elements recognized in the AMD pathophysiology. The genetic component of the disease is embodied in the recognition of the described risk or protective polymorphisms of some complement and ECM related genes (mainly CFH and ARMS2/HTRA1). Thus, carriers of the risk haplotype at ARMS2/HTRA1 have a higher risk of developing late AMD at a younger age. Finally, gut microbiota and epigenetics may play a role in modulating the progression to advanced AMD with the presence of local inflammatory conditions. Because of multiple implicated processes, different complex combinations of treatments will probably be the best option to obtain the best visual results; they in turn will differ depending on the type and spectrum of disease affecting individual patients or the disease stage in each patient at a specific moment. This will undoubtedly lead to personalized medicine for control and hopefully find a future cure. This necessitates the continued unraveling of all the processes involved in the pathogenesis of AMD that must be understood to devise the combinations of treatments for different concurrent or subsequent problems.
Collapse
Affiliation(s)
- Julián Garcia-Garcia
- Instituto de Oftalmobiologia Aplicada (IOBA), University of Valladolid, Valladolid, Spain
| | - Ricardo Usategui-Martin
- Instituto de Oftalmobiologia Aplicada (IOBA), University of Valladolid, Valladolid, Spain
- RICORS of Inflammation and Immunopathology of Organs and Systems Network, ISCIII, Madrid, Spain
- Dpto. de Biología Celular, Histología y Farmacología, University of Valladolid, Valladolid, Spain
| | - Maria Rosa Sanabria
- Instituto de Oftalmobiologia Aplicada (IOBA), University of Valladolid, Valladolid, Spain
- RICORS of Inflammation and Immunopathology of Organs and Systems Network, ISCIII, Madrid, Spain
- Ophthalmology Department, Palencia University Hospital Complex, Palencia, Spain
| | - Esther Fernandez-Perez
- Instituto de Oftalmobiologia Aplicada (IOBA), University of Valladolid, Valladolid, Spain
| | - Juan Jose Telleria
- Institute of Biology and Molecular Genetics (IBGM) University of Valladolid, Valladolid, Spain
- Dpto. de Biología Celular, Histología y Farmacología, University of Valladolid, Valladolid, Spain
| | - Rosa M Coco-Martin
- Instituto de Oftalmobiologia Aplicada (IOBA), University of Valladolid, Valladolid, Spain
- RICORS of Inflammation and Immunopathology of Organs and Systems Network, ISCIII, Madrid, Spain
| |
Collapse
|
49
|
Zhong H, Sun X. Contribution of Interleukin-17A to Retinal Degenerative Diseases. Front Immunol 2022; 13:847937. [PMID: 35392087 PMCID: PMC8980477 DOI: 10.3389/fimmu.2022.847937] [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: 01/03/2022] [Accepted: 02/25/2022] [Indexed: 12/26/2022] Open
Abstract
Retinal degenerative diseases are a leading cause of vision loss and blindness throughout the world, characterized by chronic and progressive loss of neurons and/or myelin. One of the common features of retinal degenerative diseases and central neurodegenerative diseases is chronic neuroinflammation. Interleukin-17A (IL-17A) is the cytokine most closely related to disease in its family. Accumulating evidence suggests that IL-17A plays a key role in human retinal degenerative diseases, including age-related macular degeneration, diabetic retinopathy and glaucoma. This review aims to provide an overview of the role of IL-17A participating in the pathogenesis of retinal degenerative diseases, which may open new avenues for potential therapeutic interventions.
Collapse
Affiliation(s)
- Huimin Zhong
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Xiaodong Sun
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| |
Collapse
|
50
|
Tzoumas N, Kavanagh D, Cordell HJ, Lotery AJ, Patel PJ, Steel DH. Rare complement factor I variants associated with reduced macular thickness and age-related macular degeneration in the UK biobank. Hum Mol Genet 2022; 31:2678-2692. [PMID: 35285476 PMCID: PMC9402241 DOI: 10.1093/hmg/ddac060] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 11/24/2022] Open
Abstract
To evaluate potential diagnostic and therapeutic biomarkers for age-related macular degeneration (AMD), we identified 8433 UK Biobank participants with rare complement Factor I gene (CFI) variants, 579 with optical coherence tomography-derived macular thickness data. We stratified these variants by predicted gene expression and measured their association with retinal pigment epithelium-Bruch’s membrane (RPE-BM) complex and retinal thicknesses at nine macular subfields, as well as AMD risk, using multivariable regression models adjusted for the common complement Factor H gene (CFH) p.Y402H and age-related maculopathy susceptibility protein 2 gene (ARMS2) p.A69S risk genotypes. CFI variants associated with low Factor I levels predicted a thinner mean RPE-BM (95% confidence interval [CI] −1.66 to −0.37 μm, P = 0.002) and retina (95% CI −5.88 to −0.13 μm, P = 0.04) and a higher AMD risk (odds ratio [OR] = 2.26, 95% CI 1.56 to 3.27, P < 0.001). CFI variants associated with normal Factor I levels did not impact mean RPE-BM/retinal thickness (P = 0.28; P = 0.99) or AMD risk (P = 0.97). CFH p.Y402H was associated with a thinner RPE-BM (95% CI −0.31 to −0.18 μm, P < 0.001 heterozygous; 95% CI −0.62 to −0.42 μm, P < 0.001 homozygous) and retina (95% CI −0.73 to −0.12 μm, P = 0.007 heterozygous; 95% CI −1.08 to −0.21 μm, P = 0.004 homozygous). ARMS2 p.A69S did not influence RPE-BM (P = 0.80 heterozygous; P = 0.12 homozygous) or retinal thickness (P = 0.75 heterozygous; P = 0.07 homozygous). p.Y402H and p.A69S exhibited a significant allele–dose response with AMD risk. Thus, CFI rare variants associated with low Factor I levels are robust predictors of reduced macular thickness and AMD. The observed association between macular thickness and CFH p.Y402H, but not ARMS2 p.A69S, highlights the importance of complement dysregulation in early pathogenesis.
Collapse
Affiliation(s)
- Nikolaos Tzoumas
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Complement Therapeutics Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - David Kavanagh
- Complement Therapeutics Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- National Renal Complement Therapeutics Centre, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | - Heather J Cordell
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Andrew J Lotery
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Praveen J Patel
- NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, United Kingdom
| | - David H Steel
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Sunderland Eye Infirmary, Sunderland, United Kingdom
| |
Collapse
|