1
|
Jin Q, Wang S, Yao Y, Jiang Q, Li K. The gut-eye axis: from brain neurodegenerative diseases to age-related macular degeneration. Neural Regen Res 2025; 20:2741-2757. [PMID: 39435619 PMCID: PMC11826455 DOI: 10.4103/nrr.nrr-d-24-00531] [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: 05/09/2024] [Revised: 07/25/2024] [Accepted: 09/21/2024] [Indexed: 10/23/2024] Open
Abstract
Age-related macular degeneration is a serious neurodegenerative disease of the retina that significantly impacts vision. Unfortunately, the specific pathogenesis remains unclear, and effective early treatment options are consequently lacking. The microbiome is defined as a large ecosystem of microorganisms living within and coexisting with a host. The intestinal microbiome undergoes dynamic changes owing to age, diet, genetics, and other factors. Such dysregulation of the intestinal flora can disrupt the microecological balance, resulting in immunological and metabolic dysfunction in the host, and affecting the development of many diseases. In recent decades, significant evidence has indicated that the intestinal flora also influences systems outside of the digestive tract, including the brain. Indeed, several studies have demonstrated the critical role of the gut-brain axis in the development of brain neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. Similarly, the role of the "gut-eye axis" has been confirmed to play a role in the pathogenesis of many ocular disorders. Moreover, age-related macular degeneration and many brain neurodegenerative diseases have been shown to share several risk factors and to exhibit comparable etiologies. As such, the intestinal flora may play an important role in age-related macular degeneration. Given the above context, the present review aims to clarify the gut-brain and gut-eye connections, assess the effect of intestinal flora and metabolites on age-related macular degeneration, and identify potential diagnostic markers and therapeutic strategies. Currently, direct research on the role of intestinal flora in age-related macular degeneration is still relatively limited, while studies focusing solely on intestinal flora are insufficient to fully elucidate its functional role in age-related macular degeneration. Organ-on-a-chip technology has shown promise in clarifying the gut-eye interactions, while integrating analysis of the intestinal flora with research on metabolites through metabolomics and other techniques is crucial for understanding their potential mechanisms.
Collapse
Affiliation(s)
- Qianzi Jin
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Suyu Wang
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yujia Yao
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Qin Jiang
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Keran Li
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
2
|
Fortmann SD, Frey BF, Rosencrans RF, Adu-Rutledge Y, Ready V E, Kilchrist KV, Welner RS, Boulton ME, Saban DR, Grant MB. Prenatally derived macrophages support choroidal health and decline in age-related macular degeneration. J Exp Med 2025; 222:e20242007. [PMID: 40261298 PMCID: PMC12013653 DOI: 10.1084/jem.20242007] [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: 10/28/2024] [Revised: 02/02/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
Hallmark findings in age-related macular degeneration (AMD) include the accumulation of extracellular lipid and vasodegeneration of the choriocapillaris. Choroidal inflammation has long been associated with AMD, but little is known about the immune landscape of the human choroid. Using 3D multiplex immunofluorescence, single-cell RNA sequencing, and flow cytometry, we unravel the cellular composition and spatial organization of the human choroid and the immune cells within it. We identify two populations of choroidal macrophages with distinct FOLR2 expression that account for the majority of myeloid cells. FOLR2+ macrophages predominate in the nondiseased eye, express lipid-handling machinery, uptake lipoprotein particles, and contain high amounts of lipid. In AMD, FOLR2+ macrophages are decreased in number and exhibit dysfunctional lipoprotein metabolism. In mice, FOLR2+ macrophages are negative for the postnatal fate-reporter Ms4a3, and their depletion causes an accelerated AMD-like phenotype. Our results show that prenatally derived resident macrophages decline in AMD and are implicated in multiple hallmark functions known to be compromised in the disease.
Collapse
Affiliation(s)
- Seth D. Fortmann
- Medical Scientist Training Program (MSTP), University of Alabama at Birmingham (UAB), Birmingham, AL, USA
- Department of Ophthalmology, UAB, Birmingham, AL, USA
| | - Blake F. Frey
- Medical Scientist Training Program (MSTP), University of Alabama at Birmingham (UAB), Birmingham, AL, USA
- Department of Pathology, UAB, Birmingham, AL, USA
| | - Robert F. Rosencrans
- Medical Scientist Training Program (MSTP), University of Alabama at Birmingham (UAB), Birmingham, AL, USA
- Department of Ophthalmology, UAB, Birmingham, AL, USA
| | | | - Edgar Ready V
- Department of Ophthalmology, UAB, Birmingham, AL, USA
| | | | - Robert S. Welner
- Division of Hematology/Oncology, Department of Medicine, UAB, Birmingham, AL, USA
| | | | - Daniel R. Saban
- Department of Ophthalmology, Duke University, Durham, NC, USA
| | | |
Collapse
|
3
|
Biber J, Gandor C, Becirovic E, Michalakis S. Retina-directed gene therapy: Achievements and remaining challenges. Pharmacol Ther 2025; 271:108862. [PMID: 40268248 DOI: 10.1016/j.pharmthera.2025.108862] [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: 10/07/2024] [Revised: 02/07/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
Gene therapy is an innovative medical approach that offers new treatment options for congenital and acquired diseases by transferring, correcting, inactivating or regulating genes to supplement, replace or modify a gene function. The approval of voretigene neparvovec (Luxturna), a gene therapy for RPE65-associated retinopathy, has marked a milestone for the field of retinal gene therapy, but has also helped to accelerate the development of gene therapies for genetic diseases affecting other organs. Voretigene neparvovec is a vector based on adeno-associated virus (AAV) that delivers a functional copy of RPE65 to supplement the missing function of this gene. The AAV-based gene delivery has proven to be versatile and safe for the transfer of genetic material to retinal cells. However, challenges remain in treating additional inherited as well as acquired retinopathies with this technology. Despite the high level of activity in this field, no other AAV gene therapy for retinal diseases has been approved since voretigene neparvovec. Ongoing research focuses on overcoming the current restraints through innovative strategies like AAV capsid engineering, dual-AAV vector systems, or CRISPR/Cas-mediated genome editing. Additionally, AAV gene therapy is being explored for the treatment of complex acquired diseases like age-related macular degeneration (AMD) and diabetic retinopathy (DR) by targeting molecules involved in the pathobiology of the degenerative processes. This review outlines the current state of retinal gene therapy, highlighting ongoing challenges and future directions.
Collapse
Affiliation(s)
- Josef Biber
- Department of Ophthalmology, LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Catharina Gandor
- Laboratory for Retinal Gene Therapy, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Schlieren 8952, Switzerland
| | - Elvir Becirovic
- Laboratory for Retinal Gene Therapy, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Schlieren 8952, Switzerland
| | - Stylianos Michalakis
- Department of Ophthalmology, LMU University Hospital, LMU Munich, 80336 Munich, Germany.
| |
Collapse
|
4
|
Reddy A, Greene C, Hashimoto Y, Kiang AS, Hudson N, Adamson P, Santos-Ferreira T, Campbell M. Enhanced retinal pigment epithelial cells as a delivery vehicle for retinal disease. Mol Ther Methods Clin Dev 2025; 33:101450. [PMID: 40231247 PMCID: PMC11995081 DOI: 10.1016/j.omtm.2025.101450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 03/11/2025] [Indexed: 04/16/2025]
Abstract
Age-related macular degeneration (AMD) represents a major global health burden, with current estimates suggesting that up to 200 million people are affected globally. While effective treatments exist for the exudative form of the disease termed choroidal neovascular AMD, there remain challenges associated with long-term responses to treatment and the ongoing parallel development of the non-exudative form of AMD. Here, we sought to develop an approach for long-term delivery of both aflibercept, a decoy receptor that neutralises vascular endothelial growth factor and a concomitant treatment focused on treating the non-exudative form of AMD. To this end, we developed a series of induced pluripotent stem cell (iPS)-derived retinal pigment epithelial (RPE) cell lines that stably expressed aflibercept and/or sCD59. These cell lines were shown to produce high concentrations of both proteins. Sub-retinal injection of enhanced RPE cells potently prevented leakage of neovascular lesions in the JR5558 mouse model of retinal and choroidal neovascularization. Early results described here suggest that enhanced iPS-derived RPE cells could represent a novel approach to the long-term delivery of therapeutic agents to the eye.
Collapse
Affiliation(s)
- Avril Reddy
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Chris Greene
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Yosuke Hashimoto
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Anna-Sophia Kiang
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Natalie Hudson
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Peter Adamson
- UCL, Institute of Ophthalmology, University College London, London, UK
| | - Tiago Santos-Ferreira
- Tenpoint Therapeutics, Switzerland Innovation Park, Hegenheimermattweg 167A, 4123 Allschwil, Switzerland
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
5
|
Chen JS, Esko JD, Walker E, Gordts PLSM, Baxter SL, Toomey CB. High-Density Lipoproteins Associated with Age-Related Macular Degeneration in the All of Us Research Program. Ophthalmology 2025; 132:684-691. [PMID: 39756691 PMCID: PMC12097936 DOI: 10.1016/j.ophtha.2024.12.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 12/16/2024] [Accepted: 12/31/2024] [Indexed: 01/07/2025] Open
Abstract
PURPOSE Extracellular lipoprotein aggregation is a critical event in age-related macular degeneration (AMD) pathogenesis. In this study, we sought to analyze associations between clinical and genetic-based factors related to lipoprotein metabolism and risk for AMD in the All of Us research program. DESIGN Cross-sectional retrospective data analysis. PARTICIPANTS A total of 5028 healthy participants and 2328 patients with AMD from All of Us. METHODS Participants with and without AMD were age, race, and sex matched in a 1:2 ratio, respectively. Smoking status, history of hyperlipidemia, and statin use were extracted in a binary manner. Statin use was further subcategorized into hepatically versus nonhepatically metabolized statins. Laboratory values for low-density lipoprotein (LDL), high-density lipoprotein (HDL), and triglycerides (TGs) were also extracted, and outliers were excluded from analysis. The PLINK toolkit was used to extract single nucleotide polymorphisms (SNPs) associated with LDL and HDL dysregulation, as published in prior work. Odds ratio curves were computed to assess the risk between LDL, TG, and HDL versus AMD. All clinical and genetic variables were input into a multivariable logistic regression model, and odds ratios and P values were generated. MAIN OUTCOME MEASURES Statistical significance of risk factors for AMD, thresholded at P ≤ 0.05. RESULTS On multivariable regression analysis, statin use and low and high HDL were significantly associated with increased AMD risk (P < 0.001 for all variables). Additionally, the multivariable regression implicated HDL-associated SNP's increased risk for AMD. Last, LPA was identified (P = 0.007) as a novel SNP associated with increased AMD risk. CONCLUSIONS There exists a U-shaped relationship between HDL and AMD risk, such that high and low HDL are significantly associated with increased AMD risk. Additionally, SNPs associated with HDL metabolism are associated with AMD risk. This analysis further establishes the role of HDL in AMD pathogenesis. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosure may be found after the references.
Collapse
Affiliation(s)
- Jimmy S Chen
- Division of Ophthalmology Informatics and Data Science, Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, La Jolla, California
| | - Jeffrey D Esko
- Glycobiology Research and Training Center, University of California San Diego, La Jolla, California
| | - Evan Walker
- Division of Ophthalmology Informatics and Data Science, Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, La Jolla, California
| | - Philip L S M Gordts
- Glycobiology Research and Training Center, University of California San Diego, La Jolla, California
| | - Sally L Baxter
- Division of Ophthalmology Informatics and Data Science, Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, La Jolla, California; Division of Biomedical Informatics, Department of Medicine, University of California San Diego, La Jolla, California
| | - Christopher B Toomey
- Division of Ophthalmology Informatics and Data Science, Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, La Jolla, California; Glycobiology Research and Training Center, University of California San Diego, La Jolla, California.
| |
Collapse
|
6
|
Schwartz R, Warwick AN, Khawaja AP, Luben R, Khalid H, Phatak S, Jhingan M, de Vente C, Valmaggia P, Liakopoulos S, Olvera-Barrios A, Sánchez CI, Egan C, Bonelli R, Tufail A. Genetic Distinctions Between Reticular Pseudodrusen and Drusen: A Genome-Wide Association Study. Am J Ophthalmol 2025; 274:286-295. [PMID: 40064387 DOI: 10.1016/j.ajo.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 04/07/2025]
Abstract
OBJECTIVE To identify genetic determinants specific to reticular pseudodrusen (RPD) compared with drusen. DESIGN Genome-wide association study (GWAS) SUBJECTS: Participants with RPD, drusen, and controls from the UK Biobank (UKBB), a large, multisite, community-based cohort. METHODS Participants with RPD, drusen, and controls from the UK Biobank (UKBB), a large, multisite, community-based cohort, were included. A deep learning framework analyzed 169,370 optical coherence tomography (OCT) volumes to identify cases and controls within the UKBB. Five retina specialists validated the cohorts using OCT and color fundus photographs. Several GWAS were undertaken utilizing the quantity and presence of RPD and drusen. Genome-wide significance was defined as P < 5e-8. MAIN OUTCOMES MEASURES Genetic associations were examined with the number of RPD and drusen within 'pure' cases, where only RPD or drusen were present in either eye. A candidate approach assessed 46 previously known AMD loci. Secondary GWAS were conducted for number of RPD and drusen in mixed cases, and binary case-control analyses for pure RPD and pure drusen. RESULTS The study included 1787 participants: 1037 controls, 361 pure drusen, 66 pure RPD, and 323 mixed cases. The primary pure RPD GWAS identified four genome-wide significant loci: rs11200630 near ARMS2-HTRA1 (P = 1.9e-09), rs79641866 at PARD3B (P = 1.3e-08), rs143184903 near ITPR1 (P = 8.1e-09), and rs76377757 near SLN (P = 4.3e-08). The latter three are uncommon variants (minor allele frequency <5%). A significant association at the CFH locus was also observed using a candidate approach (P = 1.8e-04). For pure drusen, two loci reached genome-wide significance: rs10801555 at CFH (P = 6.0e-33) and rs61871744 at ARMS2-HTRA1 (P = 4.2e-20). CONCLUSIONS The study highlights a clear association between the ARMS2-HTRA1 locus and higher RPD load. Although the CFH locus association did not achieve genome-wide significance, a suggestive link was observed. Three novel associations unique to RPD were identified, albeit for uncommon genetic variants. Further studies with larger sample sizes are needed to explore these findings.
Collapse
Affiliation(s)
- Roy Schwartz
- From the Moorfields Eye Hospital NHS Foundation Trust (R.S., A.N.W., H.K., S.P., M.J., C.E., A.T.), London, UK; Institute of Health Informatics (R.S.), University College London, London, UK.
| | - Alasdair N Warwick
- From the Moorfields Eye Hospital NHS Foundation Trust (R.S., A.N.W., H.K., S.P., M.J., C.E., A.T.), London, UK; University College London Institute of Cardiovascular Science (A.N.W.), London, UK
| | - Anthony P Khawaja
- NIHR Biomedical Research Centre (A.P.K., R.L., P.V., A.O.B., C.E., A.T.), Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology, London, UK
| | - Robert Luben
- NIHR Biomedical Research Centre (A.P.K., R.L., P.V., A.O.B., C.E., A.T.), Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology, London, UK; MRC Epidemiology Unit (R.L.), University of Cambridge, Cambridge, UK
| | - Hagar Khalid
- From the Moorfields Eye Hospital NHS Foundation Trust (R.S., A.N.W., H.K., S.P., M.J., C.E., A.T.), London, UK
| | - Sumita Phatak
- From the Moorfields Eye Hospital NHS Foundation Trust (R.S., A.N.W., H.K., S.P., M.J., C.E., A.T.), London, UK
| | - Mahima Jhingan
- From the Moorfields Eye Hospital NHS Foundation Trust (R.S., A.N.W., H.K., S.P., M.J., C.E., A.T.), London, UK
| | - Coen de Vente
- Quantitative Healthcare Analysis (qurAI) Group (C.D.V., C.I.S.), Informatics Institute, University of Amsterdam, Amsterdam, Netherlands; Biomedical Engineering and Physics (C.D.V., C.I.S.), Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
| | - Philippe Valmaggia
- NIHR Biomedical Research Centre (A.P.K., R.L., P.V., A.O.B., C.E., A.T.), Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology, London, UK; Department of Biomedical Engineering (P.V.), University of Basel, Basel, Switzerland; Department of Ophthalmology (P.V.), University Hospital Basel, Basel, Switzerland
| | - Sandra Liakopoulos
- Cologne Image Reading Center (S.L.), Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Department of Ophthalmology (S.L.), Goethe University, Frankfurt, Germany
| | - Abraham Olvera-Barrios
- NIHR Biomedical Research Centre (A.P.K., R.L., P.V., A.O.B., C.E., A.T.), Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology, London, UK; Institute of Ophthalmology (A.O.B), University College London, London, UK
| | - Clara I Sánchez
- Quantitative Healthcare Analysis (qurAI) Group (C.D.V., C.I.S.), Informatics Institute, University of Amsterdam, Amsterdam, Netherlands; Biomedical Engineering and Physics (C.D.V., C.I.S.), Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
| | - Catherine Egan
- From the Moorfields Eye Hospital NHS Foundation Trust (R.S., A.N.W., H.K., S.P., M.J., C.E., A.T.), London, UK; NIHR Biomedical Research Centre (A.P.K., R.L., P.V., A.O.B., C.E., A.T.), Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology, London, UK
| | - Roberto Bonelli
- Lowy Medical Research Institute (R.B.), La Jolla, California, USA
| | - Adnan Tufail
- From the Moorfields Eye Hospital NHS Foundation Trust (R.S., A.N.W., H.K., S.P., M.J., C.E., A.T.), London, UK; NIHR Biomedical Research Centre (A.P.K., R.L., P.V., A.O.B., C.E., A.T.), Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology, London, UK
| |
Collapse
|
7
|
Wang Z, Zhang Y, Xu C, Peng A, Qin H, Yao K. Advancements in age-related macular degeneration treatment: From traditional anti-VEGF to emerging therapies in gene, stem cell, and nanotechnology. Biochem Pharmacol 2025; 236:116902. [PMID: 40158818 DOI: 10.1016/j.bcp.2025.116902] [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: 12/30/2024] [Revised: 02/18/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Age-related macular degeneration (AMD) is the leading cause of central vision loss in older adults and is projected to affect approximately 400 million individuals worldwide by 2040. Its pathological characteristics include retinal extracellular deposits, such as drusen, which trigger photoreceptor degeneration and damage to the retinal pigment epithelium (RPE), resulting in irreversible vision loss. The pathogenesis of AMD involves genetic, environmental, and aging-related factors. Anti-vascular endothelial growth factor (anti-VEGF) therapy for wet AMD significantly inhibits choroidal neovascularization and delays visual deterioration. However, its high cost, frequent injections, and poor patient compliance limit application, and there remains no effective intervention for dry AMD. In recent years, emerging strategies, such as gene therapy, stem cell therapy, and nanotechnology-based drug delivery systems, offer hope for slowing disease progression by improving targeting, drug stability, and reducing treatment frequency. Nanoparticles, including polymeric and lipid systems, have shown promise for enhancing drug delivery and bioavailability, particularly for dry AMD, where existing therapies are inadequate. These strategies also have the potential to improve patient compliance. This review summarizes AMD epidemiology and examines the limitations of current therapies. It emphasizes the mechanisms and clinical advancements of gene therapy, stem cell therapy, and nanotechnology in AMD treatment. These emerging technologies offer promising opportunities for precision medicine and lay a solid foundation for the future development of multifaceted therapeutic strategies.
Collapse
Affiliation(s)
- Zhanfei Wang
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yaqin Zhang
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Chunxiu Xu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Anna Peng
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Huan Qin
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Kai Yao
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China.
| |
Collapse
|
8
|
Larsen PP, Delyfer MN, Schweitzer C, Korobelnik JF, Delcourt C. Neuroretinal and Retinal Pigment Epithelium Changes and Susceptibility to Age-Related Macular Degeneration: Insights from the Longitudinal ALIENOR Study. Ophthalmology 2025; 132:671-683. [PMID: 39793657 DOI: 10.1016/j.ophtha.2025.01.002] [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: 09/27/2024] [Revised: 12/23/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025] Open
Abstract
PURPOSE We assessed the associations of macular layer thicknesses, measured using spectral-domain (SD) OCT, with incident age-related macular degeneration (AMD) and AMD polygenic risk scores (PRSs). DESIGN Population-based cohort study. PARTICIPANTS A total of 653 participants from the ALIENOR study, with biennial eye imaging from 2009 through 2024. METHODS Macular layer thicknesses of 8 distinct layers were automatically segmented based on SD-OCT imaging. Total and pathway-specific PRSs were calculated from previous AMD genome-wide association studies summary statistics. Associations of macular layer thicknesses with incident intermediate and advanced AMD were analyzed using time-dependent Cox proportional hazards models. Associations of macular layer thicknesses with PRS were assessed using linear mixed models. MAIN OUTCOME MEASURES Incident intermediate and advanced AMD based on fundus color photographs and SD-OCT. RESULTS Mean age at first OCT examination of the 653 participants was 82.2 ± 4.2 years, 61.3% of which were women. In multivariable adjusted models, incident intermediate AMD was associated with thicker retinal pigment epithelium (RPE)-Bruch membrane (BM) complex in the 1-mm central circle (hazard ratio [HR], 1.13 for 1-μm increase; P = 8.08 × 10-4 with false discovery rate [FDR] correction). Incident advanced AMD was associated with thicker RPE-BM complex in both the central (HR, 1.09; PFDR = 0.005) and inner circle (1- to 3 mm; HR, 1.28; PFDR = 1.61 × 10-5). Over the study period, RPE-BM complex thickening in the inner circle was more pronounced in individuals with high total PRS (β = 0.06 μm/year for 1-standard deviation increase; PFDR = 1.61 × 10-10), high complement pathway PRS (β = 0.04 μm/year; PFDR = 3.23 × 10-5), high lipid pathway PRS (β = 0.03 μm/year; PFDR = 3.74 × 10-4), and ARMS2 (β = 0.03 μm/year, PFDR = 0.002). High total PRS and high complement-specific PRS were associated with thinner photoreceptor segment layer (PSL) at baseline and longitudinal thinning of the outer nuclear layer. CONCLUSIONS These findings highlight RPE-BM complex thickening in the pathophysiologic sequence of AMD. Further longitudinal studies are needed, in particular to determine the value of RPE-BM thickening and PSL thinning measured using SD-OCT for the clinical follow-up of patients with AMD. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Petra P Larsen
- INSERM, BPH, U1219, University of Bordeaux, Bordeaux, France.
| | - Marie-Noëlle Delyfer
- INSERM, BPH, U1219, University of Bordeaux, Bordeaux, France; Service d'Ophtalmologie, CHU de Bordeaux, Bordeaux, France; FRCRnet, F-CRIN Network, Bordeaux, France
| | - Cédric Schweitzer
- INSERM, BPH, U1219, University of Bordeaux, Bordeaux, France; Service d'Ophtalmologie, CHU de Bordeaux, Bordeaux, France
| | - Jean-François Korobelnik
- INSERM, BPH, U1219, University of Bordeaux, Bordeaux, France; Service d'Ophtalmologie, CHU de Bordeaux, Bordeaux, France
| | - Cécile Delcourt
- INSERM, BPH, U1219, University of Bordeaux, Bordeaux, France; FRCRnet, F-CRIN Network, Bordeaux, France
| |
Collapse
|
9
|
Tang F, Hogg RE, Higgins BE, Wright DM, Smyth L, Sivaprasad S. Polygenic Risk Score Impact on Visual Function in Older Individuals with Healthy Macula: The Northern Ireland Sensory Ageing Study. Eye (Lond) 2025; 39:1508-1516. [PMID: 39962206 DOI: 10.1038/s41433-025-03642-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/13/2025] [Accepted: 01/23/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND/OBJECTIVES Although polygenic risk scores (PRSs) have been developed for age-related macular degeneration (AMD), it is not known whether these scores are associated with impairment of visual functions in older individuals with healthy macula. We evaluated age-related changes in visual function in people aged 55 years or above with healthy macula and determined the associations of age-related visual function changes with AMD PRS in people with healthy macula. SUBJECTS/METHODS Participants aged 55 years or above with healthy macula and a comparative group of people with early or intermediate AMD from the Northern Ireland Sensory Ageing study were included. 45 SNPs were included for PRS calculation. RESULTS A total of 470 participants with healthy macula were included (Beckman grade 0 or 1). The comparator group consisted of participants with early AMD (n = 87) or intermediate AMD (n = 48). All visual functions except metrics of central visual field assessment showed a significant decline with age in adjusted linear regression models. Rod intercept time (RIT) was the only visual function significantly associated with PRS with Beta = 0.12 (95% confidence interval: 0.01-0.23), P = 0.03. A PRS integrated model achieved the highest area under the receiver operating characteristic curve (AUC) of 0.803 (0.732 to 0.874) to distinguish between normal or increased RIT. CONCLUSIONS AND RELEVANCE We observed a significant decline in multiple visual functions with increasing age. However, PRS was significantly associated with RIT only, highlighting the genetic association of age-related decline in rod function.
Collapse
Affiliation(s)
- Fangyao Tang
- Institute of Ophthalmology, University College London, London, UK
| | - Ruth E Hogg
- Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Bethany E Higgins
- Optometry and Vision Sciences, City, University of London, London, UK
| | - David M Wright
- Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Laura Smyth
- Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Sobha Sivaprasad
- Institute of Ophthalmology, University College London, London, UK.
- National Institute for Health Research Biomedical Research Centre, Moorfields Eye Hospital National Health Service Foundation Trust, London, UK.
| |
Collapse
|
10
|
Koçyiğit E, Gövez NE, Arslan S, Ağagündüz D. A narrative review on dietary components and patterns and age-related macular degeneration. Nutr Res Rev 2025; 38:143-170. [PMID: 38221852 DOI: 10.1017/s0954422424000015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Age-related macular degeneration (AMD) is one of the most prevalent eye diseases among the ageing population worldwide. It is a leading cause of blindness in individuals over 55, particularly in industrialised Western countries. The prevalence of AMD increases with age, and genetic factors and environmental influences are believed to contribute to its development. Among the environmental factors, diet plays a significant role in AMD. This review explores the association between dietary components, dietary patterns and AMD. Various nutrients, non-nutrient substances and dietary models that have the potential to counteract oxidative stress and inflammation, which are underlying mechanisms of AMD, are discussed. Consuming fruits, vegetables, fish and seafood, whole grains, olive oil, nuts and low-glycaemic-index foods has been highlighted as beneficial for reducing the risk of AMD. Adhering to the Mediterranean diet, which encompasses these elements, can be recommended as a dietary pattern for AMD. Furthermore, the modulation of the gut microbiota through dietary interventions and probiotics has shown promise in managing AMD.
Collapse
Affiliation(s)
- Emine Koçyiğit
- Department of Nutrition and Dietetics, Ordu University, Ordu, Türkiye
| | - Nazlıcan Erdoğan Gövez
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara, Türkiye
| | - Sabriye Arslan
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara, Türkiye
| | - Duygu Ağagündüz
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara, Türkiye
| |
Collapse
|
11
|
Peng M, Zeng Q, Zheng W, Xia X. Peripheral Choroid/RPE/Sclera as a Shared Pathogenic Hub: Multi-Tissue Transcriptomic Profiling Identifies Common Differentially Expressed Genes in Age-Related Macular Degeneration and Alzheimer's Disease. Mol Neurobiol 2025:10.1007/s12035-025-05078-y. [PMID: 40411685 DOI: 10.1007/s12035-025-05078-y] [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: 12/17/2023] [Accepted: 05/15/2025] [Indexed: 05/26/2025]
Abstract
BACKGROUND Age-related macular degeneration (AMD) and Alzheimer's disease (AD), two prevalent neurodegenerative disorders, share overlapping pathophysiological features yet lack cross-disease therapeutic strategies. This study systematically investigates their parallel genes and shared molecular mechanisms to identify potential therapeutic targets for dry AMD, a condition with limited treatment options. METHODS Transcriptomic datasets for AMD (GSE155154) and AD (GSE95587) were retrieved from the GEO database. AMD tissues were stratified into four subgroups: macular retina (MR), macular choroid/RPE/sclera (MCRS), peripheral retina (PR), and peripheral choroid/RPE/sclera (PCRS). Common differentially expressed genes (DEGs) were identified and analyzed via functional enrichment (GO, KEGG), gene set enrichment analysis (GSEA), and protein-protein interaction (PPI) networks. Drug-gene interactions and competing endogenous RNA (ceRNA) networks were constructed to prioritize therapeutic targets. Key hub genes were experimentally validated in a sodium iodate-induced AMD murine model using RT-qPCR. RESULTS Comparative analysis revealed 89, 56, 4, and 130 common DEGs between AD and MR, MCRS, PR, and PCRS subgroups, respectively. Neuroactive ligand-receptor interactions were prioritized in MR/MCRS-AD analyses, while extracellular matrix organization emerged as the dominant pathway in PCRS-AD comparisons. GSEA identified conserved the TNFα signaling pathway via NF-κB across both diseases. PCRS exhibited consistent expression trends for shared genes and pathways with AD. Computational screening prioritized seven druggable targets (COL1A1, COL1A2, COL3A1, MMP2, MMP9, VCAN, ITGA5) with dual therapeutic potential, along with a reconstructed circRNA (circRNA_002179)-miRNA (miR-124)-mRNA (VCAN) regulatory axis. Experimental validation in a sodium iodate-induced AMD murine model confirmed region-specific dysregulation: hub genes were significantly downregulated in MCRS but upregulated in PCRS. CONCLUSIONS Our study delineates both convergent and divergent molecular landscapes of AMD and AD, with PCRS emerging as a critical locus for shared pathophysiology. These findings bridge a critical gap in understanding AMD-AD comorbidity, offering actionable strategies for targeted drug development.
Collapse
Affiliation(s)
- Manjuan Peng
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Ophthalmology, Hunan Provincial People's Hospital and The First-Affiliated Hospital of Hunan Normal University, Changsha, China
- Department of Ophthalmology, the 921, Hospital of PLA (Second Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Qi Zeng
- Department of Ophthalmology, Hunan Provincial People's Hospital and The First-Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Wei Zheng
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaobo Xia
- Eye Center of Xiangya Hospital, Central South University, Changsha, China.
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
12
|
Chronopoulos A, Vidal-Oliver L, Sas LK, Holzwarth JW, Schutz J, Hattenbach LO. The effect of brolucizumab on diabetic macular edema and ischemia; a real-world analysis. Eur J Ophthalmol 2025:11206721251340436. [PMID: 40398452 DOI: 10.1177/11206721251340436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
ObjectiveThe response to intravitreal brolucizumab on diabetic macular edema (DME) and macular ischemia was studied in treatment-refractory and treatment-naïve patients.MethodsPatients with either recalcitrant (group 1) or treatment-naïve (group 2) DME were studied before and after intravitreal brolucizumab. All patients underwent a comprehensive ophthalmological examination including optical coherence tomography (OCT) and OCT-angiography (OCT-A). We compared the DME and macular/choriocapillaris ischemia before and after the loading dose.ResultsA total of 33 eyes of 26 patients were examined. 18 eyes were switched to brolucizumab, 15 eyes were treatment naive. All eyes responded with anatomical and functional improvement: group 1: 391 vs 298 μm (p = 0.005), 0.6 ± 0.4 logMAR vs. 0.4 ± 0.4 logMAR) (p = 0.01), group 2: 430 vs 281 μm (p = 0.0007), 0.5 ± 0.2 logMAR vs. 0.2 ± 0.2 logMAR (p = 0.004). In group 2 there was also significant improvement in retinal vascular parameters, especially in the central and inner circles of the ETDRS grid: central capillary density 6.55 vs 9.03 mm/mm2 (p = 0.008), inner 11.2 vs 13, 9 mm/mm2 (p = 0.02), central perfusion density 15.5% vs 20.7% (p = 0.02). Choriocapillaris flow deficit (CCFD%) also improved after the loading dose (40.3 vs. 35.3, p = 0.03). Although not statistically significant, there was also an improved foveal circularity.ConclusionIntravitreal brolucizumab leads to significant functional and anatomical improvement in diabetic macular edema and has also a beneficial effect on macular ischemia as shown by improvement in both retinal and choroidal perfusion.
Collapse
Affiliation(s)
- Argyrios Chronopoulos
- Department of Ophthalmology, Ludwigshafen City Hospital, Ludwigshafen am Rhein, Germany
- Department of Ophthalmology, Medical Faculty Mannheim, Ruprecht-Karls-University Heidelberg, Mannheim, Germany
| | - Lourdes Vidal-Oliver
- Department of Ophthalmology, Medical Faculty Mannheim, Ruprecht-Karls-University Heidelberg, Mannheim, Germany
| | - Lea K Sas
- Department of Ophthalmology, Ludwigshafen City Hospital, Ludwigshafen am Rhein, Germany
| | - Jakob W Holzwarth
- Department of Ophthalmology, Ludwigshafen City Hospital, Ludwigshafen am Rhein, Germany
| | - James Schutz
- Department of Ophthalmology, Ludwigshafen City Hospital, Ludwigshafen am Rhein, Germany
| | - Lars-Olof Hattenbach
- Department of Ophthalmology, Ludwigshafen City Hospital, Ludwigshafen am Rhein, Germany
| |
Collapse
|
13
|
Lerga-Jaso J, Terpolovsky A, Novković B, Osama A, Manson C, Bohn S, De Marino A, Kunitomi M, Yazdi PG. Optimization of multi-ancestry polygenic risk score disease prediction models. Sci Rep 2025; 15:17495. [PMID: 40394127 PMCID: PMC12092622 DOI: 10.1038/s41598-025-02903-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 05/16/2025] [Indexed: 05/22/2025] Open
Abstract
Polygenic risk scores (PRS) have ushered in a new era in genetic epidemiology, offering insights into individual predispositions to a wide range of diseases. However, despite recent marked enhancements in predictive power, PRS-based models still need to overcome several hurdles before they can be broadly applied in the clinic. Chiefly, they need to achieve sufficient accuracy, easy interpretability and portability across diverse populations. Leveraging trans-ancestry genome-wide association study (GWAS) meta-analysis, we generated novel, diverse summary statistics for 30 medically-related traits and benchmarked the performance of six existing PRS algorithms using UK Biobank. We built an ensemble model using logistic regression to combine outputs of top-performing algorithms and validated it on the diverse eMERGE and PAGE MEC cohorts. It surpassed current state-of-the-art PRS models, with minimal performance drops in external cohorts, indicating good calibration. To enhance predictive accuracy for clinical application, we incorporated easily-accessible clinical characteristics such as age, gender, ancestry and risk factors, creating disease prediction models intended as prospective diagnostic tests, with easily interpretable positive or negative outcomes. After adding clinical characteristics, 12 out of 30 models surpassed 80% AUC. Further, 25 traits exceeded the diagnostic odds ratio (DOR) of five, and 19 traits exceeded DOR of 10 for all ancestry groups, indicating high predictive value. Our PRS model for coronary artery disease identified 55-80 times more true coronary events than rare pathogenic variant models, reinforcing its clinical potential. The polygenic component modulated the effect of high-risk rare variants, stressing the need to consider all genetic components in clinical settings. These findings show that newly developed PRS-based disease prediction models have sufficient accuracy and portability to warrant consideration of being used in the clinic.
Collapse
Affiliation(s)
| | | | | | - Alex Osama
- Research & Development, Omics Edge, Miami, FL, USA
| | | | - Sandra Bohn
- Research & Development, Omics Edge, Miami, FL, USA
| | | | | | - Puya G Yazdi
- Research & Development, Omics Edge, Miami, FL, USA.
| |
Collapse
|
14
|
Hushmandi K, Lam HY, Wong WM, Tan W, Daryabari SH, Reiter RJ, Farahani N, Kumar AP. Gene therapy for age-related macular degeneration: a promising frontier in vision preservation. Cell Commun Signal 2025; 23:233. [PMID: 40394614 PMCID: PMC12090701 DOI: 10.1186/s12964-025-02246-4] [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: 01/29/2025] [Accepted: 05/11/2025] [Indexed: 05/22/2025] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of central vision loss, progressively impairing the retina and affecting millions worldwide. By 2040, global cases of AMD are projected to reach 300 million, posing a significant public health challenge. While early AMD may only cause mild visual impairment, advanced stages, particularly neovascular (wet) and non-neovascular (dry) AMD, can lead to severe vision loss or legal blindness, substantially affecting daily life. The introduction of anti-angiogenic therapies has revolutionized wet AMD treatment, offering a high probability of preserving or improving vision. However, these therapies do not halt AMD progression, and no definitive treatments exist for dry AMD. The limitations of current therapies, such as frequent injections and treatment resistance, underscore the urgent need for novel strategies. Gene therapy, which has shown success in treating other hereditary retinal diseases, offers a promising long-term solution for AMD by targeting retinal cells to produce therapeutic proteins. This review explores the potential of gene therapy for AMD, examining recent clinical trials and future treatment directions.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- Yong Loo Lin School of Medicine, NUS Centre for Cancer Research (N2CR), National University of Singapore, Singapore, 117599, Singapore
| | - Wendy Meihua Wong
- Centre for Innovation & Precision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Ophthalmology, National University Hospital, National University Health System, Singapore, Singapore
| | - Wency Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- Yong Loo Lin School of Medicine, NUS Centre for Cancer Research (N2CR), National University of Singapore, Singapore, 117599, Singapore
- School of Chemical & Life Sciences, Singapore Polytechnic, 500 Dover Road, Singapore, 139651, Singapore
| | - Seyed-Hashem Daryabari
- Basir Eye Health Research Center, Tehran, Iran
- Trauma Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, Long School of Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- Yong Loo Lin School of Medicine, NUS Centre for Cancer Research (N2CR), National University of Singapore, Singapore, 117599, Singapore.
| |
Collapse
|
15
|
Hulleman JD, Jeon S, Bali S, DiCesare SM, Abbas A, Daniel S, Ortega AJ, Collier GE, Yang J, Bhattacharyaa A, McCoy MK, Joachimiak LA, Posner BA. Select azo compounds post-translationally modulate HTRA1 abundance and activity potentially through interactions at the trimer interface. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.05.13.651909. [PMID: 40463243 PMCID: PMC12132422 DOI: 10.1101/2025.05.13.651909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
High-temperature requirement protein A1 (HTRA1) is a secreted serine protease with diverse substrates, including extracellular matrix proteins, proteins involved in amyloid deposition, and growth factors. Accordingly, HTRA1 has been implicated in a variety of neurodegenerative diseases including a leading cause of blindness in the elderly, age-related macular degeneration (AMD). In fact, genome wide association studies have identified that the 10q26 locus which contains HTRA1 confers the strongest genetic risk factor for AMD. A recent study has suggested that AMD-associated risk alleles in HTRA1 correlate with a significant age-related defect in HTRA1 synthesis in the retinal pigmented epithelium (RPE) within the eye, possibly accounting for AMD susceptibility. Thus, we sought to identify small molecule enhancers of HTRA1 transcription and/or protein abundance using an unbiased high-throughput screening approach. To accomplish this goal, we used CRISPR/Sp.Cas9 engineering to introduce an 11 amino acid luminescent peptide tag (HiBiT) onto the C-terminus of HTRA1 in immortalized ARPE-19 cells. Editing was very efficient (~88%), verified by genomic DNA analysis, short interfering RNA (siRNA), and HiBiT blotting. Nineteen-hundred and twenty compounds from two libraries were screened. An azo compound with reported anti-amyloidogenic and cardioprotective activity, Chicago Sky Blue 6B (CSB), was identified as an enhancer of endogenous HTRA1 secretion (2.0 ± 0.3 fold) and intracellular levels (1.7 ± 0.2 fold). These results were counter-screened using HiBiT complement factor H (CFH) edited ARPE-19 cells, verified using HiBiT blotting, and were not due to HTRA1 transcriptional changes. Importantly, serine hydrolase activity-based protein profiling (SH-ABPP) demonstrated that CSB does not affect HTRA1's specific activity. However, interestingly, in follow-up studies, Congo Red, another azo compound structurally similar to CSB, also substantially increased intracellular HTRA1 levels (up to 3.6 ± 0.3 fold) but was found to significantly impair HTRA1 enzymatic reactivity (0.45 ± 0.07 fold). Computational modeling of potential azo dye interaction with HTRA1 suggests that CSB and Congo Red can bind to the non-catalytic face of the trimer interface but with different orientation tolerances and interaction energies. These studies identify select azo dyes as HTRA1 chemical probes which may serve as starting points for future HTRA1-centered small molecule therapeutics.
Collapse
Affiliation(s)
- John D. Hulleman
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6 St. SE, Minneapolis, Minnesota, 55455, United States
| | - Seungje Jeon
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
| | - Sofia Bali
- Center for Alzheimer’s and Neurodegenerative Diseases, O’Donnell Brain Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
| | - Sophia M. DiCesare
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
| | - Ali Abbas
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
| | - Steffi Daniel
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6 St. SE, Minneapolis, Minnesota, 55455, United States
| | - Antonio J. Ortega
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6 St. SE, Minneapolis, Minnesota, 55455, United States
| | - Gracen E. Collier
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
| | - Julian Yang
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6 St. SE, Minneapolis, Minnesota, 55455, United States
| | - Archishman Bhattacharyaa
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
| | - Melissa K. McCoy
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
| | - Lukasz A. Joachimiak
- Center for Alzheimer’s and Neurodegenerative Diseases, O’Donnell Brain Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
| | - Bruce A. Posner
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
| |
Collapse
|
16
|
Zeng B, Zhang C, Liang Y, Huang J, Li D, Liu Z, Liao H, Yang T, Liu M, Zou C, Liu D, Qin B. Single-cell RNA sequencing highlights a significant retinal Müller glial population in dry age-related macular degeneration. iScience 2025; 28:112464. [PMID: 40343286 PMCID: PMC12059717 DOI: 10.1016/j.isci.2025.112464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 12/21/2024] [Accepted: 04/14/2025] [Indexed: 05/11/2025] Open
Abstract
The main challenge in dissecting the cells and pathways involved in the pathogenesis of age-related macular degeneration (AMD) is the highly heterogeneous and dynamic nature of the retinal microenvironment. This study aimed to describe the comprehensive landscape of the dry AMD (dAMD) model and identify the key cell cluster contributing to dAMD. We identified a subset of Müller cells that express high levels of Sox2, which play crucial roles in homeostasis and neuroprotection in both mouse models of AMD and patients with dAMD. Additionally, the number of Sox2+ Müller cells decreased significantly during the progression of AMD, indicating these cells were damaged and underwent cell death. Interestingly, ferroptosis and apoptosis were identified as contributors to the damage of Sox2+ Müller cells. Our findings are potentially valuable not only for advancing the current understanding of dAMD progression but also for the development of treatment strategies through the protection of Müller cells.
Collapse
Affiliation(s)
- Bing Zeng
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Chuanhe Zhang
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Yifan Liang
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Jianguo Huang
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Deshuang Li
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Ziling Liu
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Hongxia Liao
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
| | - Tedu Yang
- Shenzhen Shendong Aier Eye Hospital, Shenzhen, China
| | - Muyun Liu
- National Engineering Research Center of Foundational Technologies for CGT Industry, Shenzhen Kenuo Medical Laboratory, Shenzhen, Guangdong, China
| | - Chang Zou
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, P.R. China
| | - Dongcheng Liu
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
- Aier School of Ophthalmology, Central South University, Changsha, China
| | - Bo Qin
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China
- Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, China
- Aier School of Ophthalmology, Central South University, Changsha, China
| |
Collapse
|
17
|
Butovsky O, Rosenzweig N. Alzheimer's disease and age-related macular degeneration: Shared and distinct immune mechanisms. Immunity 2025; 58:1120-1139. [PMID: 40324382 DOI: 10.1016/j.immuni.2025.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 05/07/2025]
Abstract
Alzheimer's disease (AD) and age-related macular degeneration (AMD) represent the leading causes of dementia and vision impairment in the elderly, respectively. The retina is an extension of the brain, yet these two central nervous system (CNS) compartments are often studied separately. Despite affecting cognition vs. vision, AD and AMD share neuroinflammatory pathways. By comparing these diseases, we can identify converging immune mechanisms and potential cross-applicable therapies. Here, we review immune mechanisms highlighting the shared and distinct aspects of these two age-related neurodegenerative conditions, focusing on responses to hallmark disease manifestations, the opposite role of overlapping immune risk loci, and potential unified therapeutic approaches. We also discuss unique tissue requirements that may dictate different outcomes of conserved immune mechanisms and how we can reciprocally utilize lessons from AD therapeutics to AMD. Looking forward, we suggest promising directions for research, including the exploration of regenerative medicine, gene therapies, and innovative diagnostics.
Collapse
Affiliation(s)
- Oleg Butovsky
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Neta Rosenzweig
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
18
|
Bairagi RD, Reon RR, Hasan MM, Sarker S, Debnath D, Rahman MT, Rahman S, Islam MA, Siddique MAT, Bokshi B, Rahman MM, Acharzo AK. Ocular drug delivery systems based on nanotechnology: a comprehensive review for the treatment of eye diseases. DISCOVER NANO 2025; 20:75. [PMID: 40317427 PMCID: PMC12049359 DOI: 10.1186/s11671-025-04234-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/07/2025] [Indexed: 05/07/2025]
Abstract
Ocular drug delivery is a significant challenge due to the intricate anatomy of the eye and the various physiological barriers. Conventional therapeutic approaches, while effective to some extent, often fall short in effectively targeting ocular diseases, resulting in suboptimal therapeutic outcomes due to factors such as poor ocular bioavailability, frequent dosing requirements, systemic side effects, and limited penetration through ocular barriers. This review elucidates the eye's intricate anatomy and physiology, prevalent ocular diseases, traditional therapeutic modalities, and the inherent pharmacokinetic and pharmacodynamic limitations associated with these modalities. Subsequently, it delves into nanotechnology-based solutions, presenting breakthroughs in nanoformulations such as nanocrystals, liposomes, dendrimers, and nanoemulsions that have demonstrated enhanced drug stability, controlled release, and deeper ocular penetration. Additionally, it explores a range of nanosized carriers, including nano-structured lipid carriers, hydrogels, nanogels, nanoenzymes, microparticles, conjugates, exosomes, nanosuspensions, viral vectors, and polymeric nanoparticles, and their applications. Unique insights include emerging innovations such as nanowafers and transcorneal iontophoresis, which indicate paradigm shifts in non-invasive ocular drug delivery. Furthermore, it sheds light on the advantages and limitations of these nanotechnology-based platforms in addressing the challenges of ocular drug delivery. Though nano-based drug delivery systems are drawing increasing attention due to their potential to enhance bioavailability and therapeutic efficacy, the review ends up emphasizing the imperative need for further research to drive innovation and improve patient outcomes in ophthalmology.
Collapse
Affiliation(s)
- Rahul Dev Bairagi
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, 9208, Bangladesh
| | - Raiyan Rahman Reon
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, 9208, Bangladesh
| | - Md Mahbub Hasan
- Department of Biomedical Engineering, Khulna University of Engineering and Technology (KUET), Khulna, 9203, Bangladesh
| | - Sumit Sarker
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Bara Phool, Punjab, 140001, India
| | - Dipa Debnath
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology BHU, Varanasi, Uttar Pradesh, 221005, India
| | - Md Tawhidur Rahman
- Department of Pharmacy, Northern University of Bangladesh, Dhaka, 1230, Bangladesh
| | - Sinthia Rahman
- Department of Chemistry, University of Wyoming, Laramie, WY, USA
| | - Md Amirul Islam
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, 9208, Bangladesh
- Department of Pharmacy, East West University, Dhaka, 1212, Bangladesh
| | - Md Abu Talha Siddique
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Bishwajit Bokshi
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, 9208, Bangladesh
| | - Md Mustafizur Rahman
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, 9208, Bangladesh
| | | |
Collapse
|
19
|
Skowronska-Krawczyk D, Finnemann SC, Grant MB, Held K, Hu Z, Lu YR, Malek G, Sennlaub F, Sparrow J, D'Amore PA. Features that distinguish age-related macular degeneration from aging. Exp Eye Res 2025; 254:110303. [PMID: 39986366 PMCID: PMC11975485 DOI: 10.1016/j.exer.2025.110303] [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: 02/11/2025] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Age-related macular degeneration (AMD) is a complex, multifactorial retinal degenerative disease that is influenced by both genetic and environmental factors. However, the strongest risk factor for AMD is advanced age. Several physiological processes are observed in aging tissues including a low level of chronic inflammation (inflammaging), changed lipid and energy metabolism, and senescence. Nevertheless, whereas everyone ages, only a subset of the population develops AMD. The purpose of this review is to delineate the differences on a cellular and molecular level between natural aging changes and those observed in AMD. We provide a unique perspective on how genetic and environmental components modulate aging in the eye, as well as the specific role of the aging RPE and retina in the pathogenesis of AMD. Topics discussed include the mechanism of aging and its relation to the mechanism of AMD, current animal models that can be used to recapitulate some aspects of the pathology, and potential interventions that shift the balance towards healthy aging and therefore attenuate, prevent or delay the initiation of the disease.
Collapse
Affiliation(s)
| | | | - Maria B Grant
- Department of Ophthalmology and Visual Sciences, Marnix E. Heersink School of Medicine University of Alabama at Birmingham, Alabama, USA
| | - Katherine Held
- Ophthalmology Discovery Research, AbbVie Inc., Irvine, CA, USA
| | - Zhengping Hu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA; Departments of Ophthalmology and Pathology, Harvard Medical School, Boston, MA, USA
| | | | - Goldis Malek
- Duke University, Departments of Ophthalmology, Pathology, and Cell Biology, Albert Eye Research Institute, Durham, NC, USA
| | - Florian Sennlaub
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Janet Sparrow
- Departments of Ophthalmology and Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Patricia A D'Amore
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA; Departments of Ophthalmology and Pathology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
20
|
Ratnapriya R, Grassman F, Chen R, Hewitt A, Du J, Saban DR, Klaver CCW, Ash J, Stambolian D, Tumminia SJ, Qian J, Husain D, Iyengar SK, den Hollander AI. Functional genomics in age-related macular degeneration: From genetic associations to understanding disease mechanisms. Exp Eye Res 2025; 254:110344. [PMID: 40089136 PMCID: PMC12048874 DOI: 10.1016/j.exer.2025.110344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 03/17/2025]
Abstract
Genome-wide association studies have been remarkably successful in identifying genetic variants associated with age-related macular degeneration (AMD), demonstrating a strong genetic component largely driven by common variants. However, progress in translating these genetic findings into a deeper understanding of disease mechanisms and new therapies has been slow. Slow progress in this area can be attributed to limited knowledge of the functional impact of AMD-associated non-coding variants on gene function, the molecular mechanisms and cell types underlying disease. This review offers a comprehensive overview of functional genomics approaches to uncover the genetic, epigenetic, cellular and molecular mechanisms underlying AMD and outlines future directions for research.
Collapse
Affiliation(s)
- Rinki Ratnapriya
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - Felix Grassman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Institute for Clinical Research and Systems Medicine, Health and Medical University, Potsdam, Germany
| | - Rui Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Alex Hewitt
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, USA
| | - Daniel R Saban
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA; Department of Integrative Immunobiology, Duke University, Durham, NC, USA
| | - Caroline C W Klaver
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Ophthalmology, Radboud University Medical Center, Nijmegen, the Netherlands; Institute of Molecular and Clinical Ophthalmology, University of Basel, Basel, Switzerland
| | - John Ash
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Jiang Qian
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Deeba Husain
- Retina Service, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Sudha K Iyengar
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Anneke I den Hollander
- Genomics Research Center, AbbVie, North Chicago, IL, USA; Genomics Research Center, AbbVie, Cambridge, MA, USA.
| |
Collapse
|
21
|
Chen C, Lan Y, Yan W, Zhang X, Li T, Han J. Exploring Therapeutic Targets for Age-Related Macular Degeneration From Circulating Proteins to Plasma Metabolites in the European Population. Transl Vis Sci Technol 2025; 14:8. [PMID: 40327005 PMCID: PMC12063708 DOI: 10.1167/tvst.14.5.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 04/02/2025] [Indexed: 05/07/2025] Open
Abstract
Purpose To explore the causal associations among circulating proteins, plasma metabolites, and age-related macular degeneration (AMD). Methods We employed Mendelian randomization (MR) analysis and colocalization analysis to discern the causal relationship between proteomes and AMD. This investigation utilized data from protein quantitative trait loci (pQTL) studies in deCODE and the UK Biobank. Additionally, plasma metabolite-related genome-wide association studies (GWAS) data and AMD-related GWAS data were incorporated. Results Our findings confirmed a potential causal relationship between cytoplasmic tryptophanyl-tRNA synthetase 1 (WARS1) and a higher risk of AMD. The observed causal impact of WARS1 on the two subtypes of AMD (dry and wet) align consistently with the aforementioned outcomes. Three plasma metabolites-N-acetyl-kynurenine, N-acetyltyrosine, and caproate (6:0)-were identified as mediators of the causal effect of WARS1 on AMD, and subgroup analysis revealed that N-acetyltyrosine is a specific negative metabolite associated with WARS1 and dry AMD, whereas X-16580 is a specific positive metabolite linked to WARS1 and wet AMD. Conclusions The outcomes of this study suggest a potential causal role of specific circulating proteins in AMD and identified the mediating role of plasma metabolites between WARS1 and AMD by integrating multiple genetic analyses. Nevertheless, further research is essential to validate and strengthen these conclusions. Translational Relevance This study establishes the causal role of specific circulating proteins in AMD and identified the mediating role of plasma metabolites between WARS1 and AMD.
Collapse
Affiliation(s)
- Chengming Chen
- Department of Ophthalmology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
- Department of Ophthalmology, The 900th Hospital of Joint Logistic Support Force, PLA (Clinical Medical College of Fujian Medical University, Dongfang Hospital Affiliated to Xiamen University), Fuzhou, China
| | - Yanyan Lan
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Weiming Yan
- Department of Ophthalmology, The 900th Hospital of Joint Logistic Support Force, PLA (Clinical Medical College of Fujian Medical University, Dongfang Hospital Affiliated to Xiamen University), Fuzhou, China
| | - Xiaohong Zhang
- Department of Ophthalmology, The 900th Hospital of Joint Logistic Support Force, PLA (Clinical Medical College of Fujian Medical University, Dongfang Hospital Affiliated to Xiamen University), Fuzhou, China
| | - Tian Li
- Tianjin Medical University, Tianjin, China
| | - Jing Han
- Department of Ophthalmology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
22
|
Holleman AM, Deaton AM, Hoffing RA, Krohn L, LoGerfo P, Nioi P, Plekan ME, Akle Serrano S, Ticau S, Walshe TE, Borodovsky A, Ward LD. Rare predicted loss-of-function and damaging missense variants in CFHR5 associate with protection from age-related macular degeneration. Am J Hum Genet 2025; 112:1062-1080. [PMID: 40250423 PMCID: PMC12120177 DOI: 10.1016/j.ajhg.2025.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 04/20/2025] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of blindness among older adults worldwide, but treatment options are limited. Genetics studies have implicated the CFH locus, containing CFH and five CFHR genes, CFHR1-5, in AMD. While CFH has been robustly linked with AMD risk, potential additional roles for the CFHR genes remain unclear, obscured by strong linkage disequilibrium across the locus. Investigating rare coding variants can help to identify causal genes in such regions. We used whole-exome sequencing data from 406,952 UK Biobank participants to examine AMD associations with genes at the CFH locus. For each gene, we used burden testing to examine associations of rare (minor-allele frequency [MAF] < 1%) predicted loss-of-function (pLoF) and predicted damaging missense variants with AMD. We considered "broadly defined AMD" (ICD-10 35.3; ncases = 10,700) and "strictly defined AMD" (dry or wet AMD; ncases = 346). Adjusting for CFH-region variants known to independently associate with AMD, we find that CFHR5 rare variant burden significantly associates with a decreased risk of broadly defined AMD (odds ratio [OR] = 0.75, p = 7 × 10-4), with this association primarily driven by pLoF variants. Furthermore, the association of CFHR5 rare variants with AMD protection is estimated to be stronger for individuals with the CFH rs1061170 AMD risk allele (p.Tyr402His [p.Y402H]; interaction p = 0.04). Corresponding analyses of strict AMD were underpowered. However, we observe that thinning of the photoreceptor layer outer segment strongly predicts strict AMD and find that CFHR5 rare variant burden is significantly associated with increased thickness of this retinal layer (+0.34 SD, p = 4 × 10-4, n = 45,365). These findings suggest CFHR5 inhibition as a potential therapeutic approach for AMD.
Collapse
Affiliation(s)
| | | | | | - Lynne Krohn
- Alnylam Pharmaceuticals, Cambridge, MA 02142, USA
| | | | - Paul Nioi
- Alnylam Pharmaceuticals, Cambridge, MA 02142, USA
| | | | | | - Simina Ticau
- Alnylam Pharmaceuticals, Cambridge, MA 02142, USA
| | | | | | - Lucas D Ward
- Alnylam Pharmaceuticals, Cambridge, MA 02142, USA
| |
Collapse
|
23
|
Papadam A, Lionikas A, Grassmann F. Differential Organ Ageing Is Associated With Age-Related Macular Degeneration. Aging Cell 2025; 24:e14473. [PMID: 39757747 PMCID: PMC12073918 DOI: 10.1111/acel.14473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 12/15/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025] Open
Abstract
Age-related macular degeneration (AMD) is a progressive disorder and the leading cause of central vision loss. Age is the most important risk factor, followed by genetics and smoking. However, ageing is a complex process, and biological age can deviate from chronological age between individuals and within different organ systems. Initially, we used machine learning to predict the biological age of the immune, cardiovascular, pulmonary, renal, musculoskeletal, metabolic and hepatic systems by analysing various physiological and physical markers in the UK Biobank cohort. Then, we investigated the association of each organ's biological age with incident AMD derived from electronic health record data as well as with different AMD genetic risk scores. We observed that most organ systems in participants who developed AMD after recruitment showed accelerated ageing compared with controls, with the immune system being the most affected, especially in younger males. Surprisingly, we found that AMD patients showed slower ageing of their hepatic system compared to controls, particularly in female patients. The overall AMD genetic risk score was associated with faster organ ageing across all tissues except cardiovascular and pulmonary, while genetic risk scores stratified by pathways differently influenced each organ system. In conclusion, we found differential organ ageing associated with AMD. Significantly, genetic risk variants of AMD are associated with differential ageing of various organ systems.
Collapse
Affiliation(s)
| | | | - Felix Grassmann
- Institute of Medical Sciences, University of AberdeenAberdeenUK
- Institute for Clinical Research and Systems MedicineHealth and Medical UniversityPotsdamGermany
| |
Collapse
|
24
|
Miller JML, Thompson BR, Handa JT, Luthert P, Chakravarthy U, Csaky KG, Bird A, Young BK, Iyengar SK, Baek J, Zouache MA, Richards BT, Hageman GS, Rodrigues G, Bharti K, Flannery JG, Gorin MB, Bowes Rickman C. Dissecting the biological complexity of age-related macular degeneration: Is it one disease, multiple separate diseases, or a spectrum? Exp Eye Res 2025; 254:110304. [PMID: 39983974 PMCID: PMC12066171 DOI: 10.1016/j.exer.2025.110304] [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: 10/23/2024] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Clinicians recognize the heterogeneity of age-related macular degeneration (AMD) in presentation, progression, and treatment response, as well as the challenges in distinguishing it from other macular degenerations. As part of the 2024 Ryan Initiative for Macular Research meeting, a group of clinician-scientists and basic scientists were convened to consider the question of whether AMD should be classified as a single disorder or a spectrum of conditions. To answer this question, we reviewed research on several "dimensions" that constitute AMD risk or pathogenesis: genetics, ancestry, retinal imaging findings, diet and environment, aging, and outer retinal molecular and cellular pathways. The group reached a consensus that AMD represents a heterogeneous collection of disease states arising from the interplay of these dimensions. This heterogeneity can be conceived of as a "cloud" of AMD phenotypes. Defining subtypes within this "cloud" requires longitudinal cohorts of well-genotyped and phenotyped patients who progress from no AMD through late AMD, analyzed by unsupervised learning. Comparing the AMD subtypes that emerge from this analysis, especially -omics data from each subtype, will illuminate biology that is applicable to certain subtypes of AMD patients and molecular pathogenic mechanisms that universally apply to all AMD. This knowledge will, in turn, drive improved drug development.
Collapse
Affiliation(s)
- Jason M L Miller
- University of Michigan, Kellogg Eye Center and Cellular and Molecular Biology Program, Ann Arbor, MI, USA.
| | - Benjamin R Thompson
- Northwestern University Feinberg School of Medicine, Department of Ophthalmology and Feinberg Cardiovascular and Renal Research Inst., Chicago, IL, USA
| | - James T Handa
- Johns Hopkins Medical School, Wilmer Eye Institute, Baltimore, MD, USA
| | | | - Usha Chakravarthy
- Queens University of Belfast, Center for Public Health, Belfast, Ireland
| | - Karl G Csaky
- Retina Foundation of the Southwest, Dallas, TX, USA
| | - Alan Bird
- UCL Institute of Ophthalmology, London, United Kingdom
| | - Benjamin K Young
- Oregon Health Sciences University, Department of Ophthalmology, Portland, OR, USA
| | - Sudha K Iyengar
- Case Western Reserve University, Department of Population and Quantitative Health Sciences, Cleveland, OH, USA
| | - Jiwon Baek
- Department of Ophthalmology, College of Medicine, The Catholic University of Korea, South Korea
| | - Moussa A Zouache
- University of Utah, Department of Ophthalmology and Visual Sciences, Salt Lake City, UT, USA
| | - Burt T Richards
- University of Utah, Department of Ophthalmology and Visual Sciences, Salt Lake City, UT, USA
| | - Gregory S Hageman
- University of Utah, Department of Ophthalmology and Visual Sciences, Salt Lake City, UT, USA
| | - Gerry Rodrigues
- Allergan/Abbvie, Ophthalmology Discovery Research, Laguna Niguel, CA, USA
| | - Kapil Bharti
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - John G Flannery
- University of California, Berkeley, Department of Neuroscience, Berkeley, CA, USA
| | - Michael B Gorin
- David Geffen School of Medicine, Stein Eye Institute, Los Angeles, CA, USA.
| | - Catherine Bowes Rickman
- Duke University Medical Center, Departments of Ophthalmology and of Cell Biology, Durham, NC, USA.
| |
Collapse
|
25
|
Sirks MJ, van Dijk EHC, Ghalayini H, Bazdar S, Yu W, Yzer S, Martinez J, Schlingemann RO, Diederen RMH, Boon CJF. The clinical spectrum of polypoidal choroidal vasculopathy in Caucasian patients: a retrospective multicenter cohort study. Ophthalmol Retina 2025:S2468-6530(25)00205-2. [PMID: 40316047 DOI: 10.1016/j.oret.2025.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/16/2025] [Accepted: 04/22/2025] [Indexed: 05/04/2025]
Abstract
PURPOSE To describe clinical characteristics of polypoidal choroidal vasculopathy (PCV) in a large Caucasian cohort. DESIGN Multicenter retrospective cohort study in 3 tertiary referral centers in the Netherlands. SUBJECTS Caucasian patients with an indocyanine green angiography-confirmed diagnosis of PCV in one or both eyes. METHODS The medical charts and multimodal imaging (MMI) of the included patients were assessed retrospectively by 2 independent assessors. Any discrepancies between graders were resolved by a senior retinal specialist. A predefined set of phenotypic characteristics were graded on MMI, including optical coherence tomography, color fundus photography, fundus fluorescein angiography, and indocyanine green angiography. MAIN OUTCOME MEASURES PCV patients were distributed among 4 phenotypically different types, based on a previously published description: PCV-AMD: PCV with drusenoid age-related macular degeneration (AMD; type A); PCV-BNN: PCV without drusen but with a branching neovascular network (BNN; type B); PCV-i: isolated PCV without drusen or a BNN (type C); PCV-CSC: PCV with a background of central serous chorioretinopathy (CSC; type D). RESULTS We included 332 eyes of 305 PCV patients, with 179 out of 305 patients being female (58.7%). The average age at diagnosis was 73 years. The included eyes had the following types: PCV-AMD in 188 eyes (58.4%); PCV-BNN in 61 eyes (18.9%); PCV-i in 15 eyes (4.7%); PCV-CSC in 58 eyes (18.0%). Patients with PCV-AMD were older and more often female than patients with PCV-CSC. The median best-corrected visual acuity of affected eyes was 0.30 logMAR (interquartile range: 0.10 - 0.52), with a large range in each type. A median of 2 polypoidal lesions per eye was found (range: 1 - 12), with no significant differences between types. The choroidal thickness beneath the fovea and beneath polypoidal lesions was significantly higher in PCV-CSC than in PCV-AMD (both p<0.001). CONCLUSIONS PCV in Caucasian patients comprises a spectrum of different phenotypes: it may present with signs of drusenoid AMD, with a background of CSC, or without signs of either diseases. We found a different phenotype distribution when compared to published findings in Asian patients with PCV.
Collapse
Affiliation(s)
- Marc J Sirks
- Department of Ophthalmology, Amsterdam University Medical Center, Amsterdam, the Netherlands; Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands; Rotterdam Eye Hospital, Rotterdam, the Netherlands
| | - Elon H C van Dijk
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands; Rotterdam Eye Hospital, Rotterdam, the Netherlands
| | - Husein Ghalayini
- Department of Ophthalmology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Somayeh Bazdar
- Department of Ophthalmology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - WeiFeng Yu
- Department of Ophthalmology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Suzanne Yzer
- Rotterdam Eye Hospital, Rotterdam, the Netherlands; Department of Ophthalmology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Reinier O Schlingemann
- Department of Ophthalmology, Amsterdam University Medical Center, Amsterdam, the Netherlands; Department of Ophthalmology, Ocular Angiogenesis Group, Amsterdam University Medical Center, Amsterdam, the Netherlands; Department of Ophthalmology, Jules-Gonin Eye Hospital, Fondation Asile Des Aveugles, Lausanne, Switzerland
| | - Roselie M H Diederen
- Department of Ophthalmology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Camiel J F Boon
- Department of Ophthalmology, Amsterdam University Medical Center, Amsterdam, the Netherlands; Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
26
|
Sampson J, Segrè AV, Bujakowska KM, Haynes S, Baralle D, Banka S, Black GC, Sergouniotis PI, Ellingford JM. Paired DNA and RNA sequencing uncovers common and rare genetic variants regulating gene expression in the human retina. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.25.25326445. [PMID: 40313258 PMCID: PMC12045431 DOI: 10.1101/2025.04.25.25326445] [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: 05/03/2025]
Abstract
Genetic disorders impacting vision affect millions of individuals worldwide, including age-related macular degeneration (common) and inherited retinal disorders (rare). There is incomplete understanding of the impact of genetic variation on gene expression in the human retina, and its role in genetic disorders. Through the generation of whole genome sequencing and bulk RNA-sequencing of neurosensory retina (NSR) and retinal pigment epithelium (RPE) from 201 post-mortem eyes, we uncovered common and rare genetic variants shaping retinal expression profiles. This includes 1,483,595 significant cis-expression quantitative trait loci (eQTLs) impacting 9,959 and 3,699 genes in NSR and RPE, respectively, with associated genetic variants enriched to cis-candidate regulatory elements and notable shared eGenes between NSR and RPE. We also detected 1051 expression outliers and prioritised 299 rare non-coding single-nucleotide, structural variants or copy number variants as plausible drivers for 28% of outlier events. This study increases understanding of gene expression regulation in the human retina.
Collapse
Affiliation(s)
- Jacob Sampson
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Ayellet V Segrè
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Kinga M Bujakowska
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Steve Haynes
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Diana Baralle
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Siddharth Banka
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Health Innovation Manchester, Manchester, UK
| | - Graeme C Black
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Health Innovation Manchester, Manchester, UK
| | - Panagiotis I Sergouniotis
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Health Innovation Manchester, Manchester, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL- EBI), Wellcome Genome Campus, Cambridge, UK
- Manchester Royal Eye Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Jamie M Ellingford
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
27
|
Hector M, Behnke V, Dabrowska-Schlepp P, Busch A, Schaaf A, Langmann T, Wolf A. Moss-derived human complement factor H modulates retinal immune response and attenuates retinal degeneration. J Neuroinflammation 2025; 22:104. [PMID: 40217267 PMCID: PMC11992837 DOI: 10.1186/s12974-025-03418-2] [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: 04/22/2024] [Accepted: 03/12/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND AMD is a multifactorial progressive disease of the central retina that leads to severe vision loss among the elderly. Genome-wide association studies in AMD patients and preclinical data have identified a dysregulated complement system and aberrant microglia responses in the pathogenesis of AMD. Specifically, a genetic variant in the complement factor H (CFH) gene, an important inhibitor of the alternative complement pathway, confers the strongest risk for AMD. Here, we investigated whether moss-derived recombinant human CFH proteins, termed CPV-101 and CPV-104, can modulate microglia reactivity and limit retinal degeneration in a murine light damage paradigm mimicking important features of AMD. METHODS Two glycosylated human recombinant CFH proteins CPV101, and CPV-104 were produced in moss suspension cultures. In addition, glycans of the CPV-104 variant are sialylated, an optimization that makes CPV-104 an analog of human CFH. BALB/cJ mice received intravitreal injections of 5 µg CPV-101 and CPV-104 or vehicle, starting 1 day prior to exposure to 10,000 lx white light for 30 min. The effects of CPV-101 and CPV-104 treatment on mononuclear phagocyte and Müller cell reactivity were analyzed by immunostainings of retinal sections and flat mounts. Gene expression of microglia markers was analyzed using quantitative real-time PCR (qRT-PCR). Optical coherence tomography (OCT); Blue Peak Autofluorescence (BAF); terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining, and morphometric analyses were used to quantify the extent of retinal degeneration and photoreceptor apoptosis. RESULTS Light-exposed mice treated with moss-derived recombinant human full-length CFH showed reduced complement activation and MAC deposition in the retina. Concomitantly, mononuclear phagocyte and Müller cell reactivity in light-exposed retinas were also ameliorated upon CFH substitution. Moreover, attenuated light-induced retinal degeneration was detected in mice that received moss-derived CFH. CONCLUSION Modulating the alternative complement pathway using moss-derived recombinant human full-length CFH variant CPV-101 and CPV-104 counter-regulate gliosis and attenuates light-induced retinal degeneration, highlighting a promising concept for the treatment of AMD patients.
Collapse
Affiliation(s)
- Mandy Hector
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50931, Cologne, Germany
| | - Verena Behnke
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50931, Cologne, Germany
| | | | - Andreas Busch
- Eleva GmbH, Hans-Bunte-Straße 19, 79108, Freiburg, Germany
| | - Andreas Schaaf
- Eleva GmbH, Hans-Bunte-Straße 19, 79108, Freiburg, Germany
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50931, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Anne Wolf
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50931, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany.
| |
Collapse
|
28
|
Cheng SY, Giguere D, Silverstein I, Conza A, Seddon JM, Kim S, Iwata T, Mueller C, Punzo C. Role of alpha-1 antitrypsin in Bruch's membrane integrity. Sci Rep 2025; 15:12223. [PMID: 40210893 PMCID: PMC11985914 DOI: 10.1038/s41598-025-96570-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 03/31/2025] [Indexed: 04/12/2025] Open
Abstract
Alpha-1 antitrypsin (AAT) is a serine protease inhibitor that plays a crucial role in maintaining extracellular matrix integrity. Studies suggest that AAT augmentation therapy may benefit multiple eye diseases, including age-related macular degeneration (AMD). However, the function of endogenous AAT in the eye remains unclear. Here we used genetic knockout mice to study the role of AAT in eye health. We show that loss of AAT results in Bruch's membrane (BrM) thickening driven in part by increased laminin deposition with a concomitant decrease in collagen and elastin, which are two other critical BrM components. Interestingly, BrM remodeling due to excess extracellular protease activity reduced the age-related deposition at the BrM of apolipoprotein E, while increasing complement factor H and lowering secretion of the proangiogenic vascular endothelial growth factor. Despite these changes, the phagocytic function of the retinal pigment epithelium was not affected nor was the expression of genes that partake in photoreceptor cell metabolism. Consistent with loss of AAT resulting in changes that should alleviate AMD pathologies, human AMD donor eyes exhibited lower AAT expression levels in the BrM/choroid layer when compared to healthy donor eyes. Together, the study provides insight into AAT's function and its potential involvement in AMD.
Collapse
Affiliation(s)
- Shun-Yun Cheng
- Department of Ophthalmology and Visual Sciences, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Delaney Giguere
- Department of Ophthalmology and Visual Sciences, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Ilana Silverstein
- Department of Ophthalmology and Visual Sciences, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Adrienne Conza
- Department of Ophthalmology and Visual Sciences, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Johanna M Seddon
- Department of Ophthalmology and Visual Sciences, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - San Kim
- Department of Ophthalmology and Visual Sciences, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Takeshi Iwata
- Divivion of Molecular and Cellular Biology, National Institute of Sensory Organ, NHO Tokyo Medical Center, 2-5-1, Higashigaoka, Meguro-ku, Tokyo, 152-8902, Japan
| | | | - Claudio Punzo
- Department of Ophthalmology and Visual Sciences, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
- Department of Genetics and Cellular Medicine and Horae Gene Therapy Center, Worcester, MA, 01605, USA.
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
29
|
Chandler LC, Gardner A, Cepko CL. RPE-specific MCT2 expression promotes cone survival in models of retinitis pigmentosa. Proc Natl Acad Sci U S A 2025; 122:e2421978122. [PMID: 40178895 PMCID: PMC12002273 DOI: 10.1073/pnas.2421978122] [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: 10/24/2024] [Accepted: 02/18/2025] [Indexed: 04/05/2025] Open
Abstract
Retinitis pigmentosa (RP) is the most common cause of inherited retinal degeneration worldwide. It is characterized by the sequential death of rod and cone photoreceptors, the cells responsible for night and daylight vision, respectively. Although the expression of most RP genes occurs only in rods, there is a secondary degeneration of cones. One possible mechanism of cone death is metabolic dysregulation. Photoreceptors are highly metabolically active, consuming large quantities of glucose and producing substantial amounts of lactate. The retinal pigment epithelium (RPE) mediates the transport of glucose from the blood to photoreceptors and, in turn, removes lactate, which can influence the rate of consumption of glucose by the RPE. One model for metabolic dysregulation in RP suggests that following the death of rods, lactate levels are substantially diminished causing the RPE to withhold glucose, resulting in nutrient deprivation for cones. Here, we present adeno-associated viral vector-mediated delivery of monocarboxylate transporter 2 (MCT2, Slc16a7) into the eye, with expression limited to RPE cells, with the aim of promoting lactate uptake from the blood and encouraging the passage of glucose to cones. We demonstrate prolonged survival and function of cones in rat and mouse RP models, revealing a possible gene-agnostic therapy for preserving vision in RP. We also present the use of fluorescence lifetime imaging-based biosensors for lactate and glucose within the eye. Using this technology, we show changes to lactate and glucose levels within MCT2-expressing RPE, suggesting that cone survival is impacted by changes in RPE metabolism.
Collapse
Affiliation(s)
- Laurel C. Chandler
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA02115
- Department of Ophthalmology, Harvard Medical School, Boston, MA02115
- HHMI, Chevy Chase, MD20815
| | - Apolonia Gardner
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA02115
- Department of Ophthalmology, Harvard Medical School, Boston, MA02115
- HHMI, Chevy Chase, MD20815
- Virology Program, Harvard Medical School, Boston, MA02115
| | - Constance L. Cepko
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA02115
- Department of Ophthalmology, Harvard Medical School, Boston, MA02115
- HHMI, Chevy Chase, MD20815
| |
Collapse
|
30
|
Ochoa Hernández ME, Lewis-Luján LM, Burboa Zazueta MG, Del Castillo Castro T, De La Re Vega E, Gálvez-Ruiz JC, Trujillo-López S, López Torres MA, Iloki-Assanga SB. Role of Oxidative Stress and Inflammation in Age Related Macular Degeneration: Insights into the Retinal Pigment Epithelium (RPE). Int J Mol Sci 2025; 26:3463. [PMID: 40331961 PMCID: PMC12026614 DOI: 10.3390/ijms26083463] [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: 03/15/2025] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 05/08/2025] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of visual impairment worldwide, characterized by the accumulation of extracellular drusen deposits within the macula. The pathogenesis of AMD is multifactorial, involving oxidative stress, chronic inflammation, immune system dysregulation, and genetic predisposition. A key contributor to disease progression is the excessive accumulation of reactive oxygen species (ROS), which damage retinal pigment epithelium (RPE) cells and disrupt cellular homeostasis. Additionally, immunosenescence and chronic low-grade inflammation exacerbate AMD pathology, further impairing retinal integrity. Despite ongoing research, effective therapeutic options remain limited, and there is no definitive cure for AMD. This review explores the intricate molecular mechanisms underlying AMD, including the role of oxidative stress, chronic inflammation, and genetic factors in RPE dysfunction. Furthermore, we highlight potential therapeutic strategies targeting these pathways, as well as the emerging role of bioinformatics and artificial intelligence in AMD diagnosis and treatment development. By improving our understanding of AMD pathophysiology, we can advance the search for novel therapeutic interventions and preventative strategies.
Collapse
Affiliation(s)
- María Elena Ochoa Hernández
- Department of Scientific and Technological Research, University of Sonora, Luis Encinas y Rosales, Centro, Hermosillo 83000, Sonora, Mexico; (M.E.O.H.); (M.G.B.Z.); (E.D.L.R.V.); (M.A.L.T.)
| | - Lidianys María Lewis-Luján
- Department of Biological Chemical Sciences, University of Sonora, Luis Encinas y Rosales, Centro, Hermosillo 83000, Sonora, Mexico; (L.M.L.-L.); (J.C.G.-R.)
| | - María Guadalupe Burboa Zazueta
- Department of Scientific and Technological Research, University of Sonora, Luis Encinas y Rosales, Centro, Hermosillo 83000, Sonora, Mexico; (M.E.O.H.); (M.G.B.Z.); (E.D.L.R.V.); (M.A.L.T.)
| | - Teresa Del Castillo Castro
- Department of Research in Polymers and Materials, University of Sonora, Calle de la Sabiduría, Centro, Hermosillo 83000, Sonora, Mexico;
| | - Enrique De La Re Vega
- Department of Scientific and Technological Research, University of Sonora, Luis Encinas y Rosales, Centro, Hermosillo 83000, Sonora, Mexico; (M.E.O.H.); (M.G.B.Z.); (E.D.L.R.V.); (M.A.L.T.)
| | - Juan Carlos Gálvez-Ruiz
- Department of Biological Chemical Sciences, University of Sonora, Luis Encinas y Rosales, Centro, Hermosillo 83000, Sonora, Mexico; (L.M.L.-L.); (J.C.G.-R.)
| | - Sergio Trujillo-López
- Department of Medicine and Health Sciences, University of Sonora, Luis Encinas y Rosales, Centro, Hermosillo 83000, Sonora, Mexico;
| | - Marco Antonio López Torres
- Department of Scientific and Technological Research, University of Sonora, Luis Encinas y Rosales, Centro, Hermosillo 83000, Sonora, Mexico; (M.E.O.H.); (M.G.B.Z.); (E.D.L.R.V.); (M.A.L.T.)
| | - Simon Bernard Iloki-Assanga
- Department of Biological Chemical Sciences, University of Sonora, Luis Encinas y Rosales, Centro, Hermosillo 83000, Sonora, Mexico; (L.M.L.-L.); (J.C.G.-R.)
- Department of Medicine and Health Sciences, University of Sonora, Luis Encinas y Rosales, Centro, Hermosillo 83000, Sonora, Mexico;
| |
Collapse
|
31
|
Allaire P, Fox J, Kitchner T, Gabor R, Folz C, Bettadahalli S, Hebbring S. Familial Renal Glucosuria and Potential Pharmacogenetic Impact on Sodium-Glucose Cotransporter-2 Inhibitors. KIDNEY360 2025; 6:521-530. [PMID: 39412882 PMCID: PMC12045503 DOI: 10.34067/kid.0000000621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 10/09/2024] [Indexed: 10/18/2024]
Abstract
Key Points A significant knowledge gap exists in SLC5A2 's role in familial renal glycosuria and sodium-glucose cotransporter-2 inhibitors' efficacy. Two percent of individuals in the All-of-Us cohort harbor rare genetic variants in SLC5A2 , potentially increasing the risk of familial renal glycosuria. Our trial suggests differential responses to sodium-glucose cotransporter-2 inhibitors in individuals with rare SLC5A2 alleles compared with wild types. Background Renal glucosuria is a rare inheritable trait caused by loss-of-function variants in the gene that encodes sodium-glucose cotransporter-2 (SGLT2) (i.e ., SLC5A2 ). The genetics of renal glucosuria is poorly understood, and even less is known on how loss-of-function variants in SLC5A2 may affect response to SGLT2 inhibitors, a new class of medication gaining popularity to treat diabetes by artificially inducing glucosuria. Methods We used two biobanks that link genomic with electronic health record data to study the genetics of renal glucosuria. This included 245,394 participants enrolled in the All of Us Research Program and 11,011 enrolled in Marshfield Clinic's Personalized Research Project (PMRP). Association studies in All of Us and PMRP identified ten variants that reached an experiment-wise Bonferroni threshold in either cohort, of which nine were novel. PMRP was further used as a recruitment source for a prospective SGLT2 pharmacogenetic trial. During a glucose tolerance test, the trial measured urine glucose concentrations in 15 SLC5A2 variant–positive individuals and 15 matched wild types with and without an SGLT2 inhibitor. Results This trial demonstrated that carriers of SLC5A2 risk variants may be more sensitive to SGLT2 inhibitors compared with wild types (P = 0.075). On the basis of population data, 2% of an ethnically diverse population carried rare variants in SLC5A2 and are at risk of renal glucosuria. Conclusions As a result, 2% of individuals being treated with SGLT2 inhibitors may respond differently to this new class of medication compared with the general population, suggesting that a larger investigation into SLC5A2 variants and SGLT2 inhibitors is needed.
Collapse
Affiliation(s)
- Patrick Allaire
- Center for Precision Medicine Research, Marshfield Clinic Health System , Marshfield, Wisconsin
| | | | | | | | | | | | | |
Collapse
|
32
|
Furundaoturan O, Degirmenci C, Afrashi F, Atik T, Akkin C, Mentes J, Nalcaci S. Association between the ARMS2 rs10490924 risk genotype and dry-age related macular degeneration patients with and without reticular pseudodrusen in a Turkish population: findings from a study conducted at a tertiary clinic. Graefes Arch Clin Exp Ophthalmol 2025; 263:1167-1173. [PMID: 39601844 DOI: 10.1007/s00417-024-06699-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/14/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024] Open
Abstract
PURPOSE To evaluate the relationship between the presence of reticular pseudodrusen (RPD) and the risk allele of ARMS2 rs10490924 variation in dry-AMD patients by using multimodal imaging. Also, to compare patients with and without RPD and healthy volunteers according to the distribution of the risk allele. METHODS In this cross-sectional study, dry-AMD patients with (Group A, n = 50) and without (Group B, n = 50) RPD and healthy volunteers (Group C, n = 50) were enrolled. After detailed ophthalmologic examination, confocal scanning laser ophthalmoscope (Heidelberg, Germany) was used to acquire near infra-red (NIR) imaging for RPD and the diagnosis was confirmed by Spectral Domain-Optical coherence tomography (Heidelberg, Germany). In silent choroidal neovascularization suspicion, optical coherence tomography angiography (Optovue, Fremont, CA) was performed and those were excluded. For genetic assessment, peripheric blood sampling was performed. Using next-generation sequencing technique (NGS), ARMS2 rs10490924 single nucleotide polymorphism was investigated. Groups were compared according to the distribution of the risky allele. RESULTS 150 eyes of 150 participants were included. In Group A, 42% (21) of patients were heterozygous for the T risk allele, 30% (15) were homozygous, and the risk allele was not detected in 28% (14). In Group B, 44% (22) of patients were heterozygous, 17% (8) were homozygous, and the risk allele was not detected in 39% (20). In Group C, 30% (15) of participants were heterozygous, 4% (2) were homozygous, and variation was not observed in 64% (32). Homozygous participants in Group A were significantly higher than other two groups (Group A-B: OR = 2.67, 95% CI: 0.895, 8.020; Group A-C: OR = 17.14, 95% CI: 3.449, 85.208) while in Group B, homozygous individuals were higher than Group C (respectively, p values 0.0039, 0.0002, 0.013). T risky allele frequencies were 51%, 38%, and 20% in Groups A, B, and C, respectively, which was significantly higher in Group A (p = 0.02). CONCLUSION Genetic influence in AMD is inevitable while certain differences according to different ethnicities may apply. Association of genetic variations and imaging findings like RPD is lacking among literature for different populations. By the aspect of this study, the relationship between RPD and ARMS2 rs10490924 polymorphism in dry-AMD patients were highlighted among Turkish population by using multimodal imaging for the first time. KEY MESSAGES What is Known? Pathophysiology of age-related macular degeneration is influenced from multiple factors including single nucleotide polymorphisms. The variations of ARMS2 are suspected well in the current literature. Reticular pseudodrusen is related to advanced stages of age related macular degeneration disease. What is New? The ARMS2 rs10490924 risk genotype is associated with the presence of reticular pseudodrusen in dry age related macular degeneration patients. Homozygous genotype of T risk allele is evaluated significantly higher in dry age related macular degeneration patients with reticular pseudodrusen.
Collapse
Affiliation(s)
| | | | - Filiz Afrashi
- Department of Ophthalmology, Ege University, Izmir, Turkey
| | - Tahir Atik
- Department of Medical Genetics, Ege University, Izmir, Turkey
| | - Cezmi Akkin
- Department of Ophthalmology, Ege University, Izmir, Turkey
| | - Jale Mentes
- Department of Ophthalmology, Ege University, Izmir, Turkey
| | - Serhad Nalcaci
- Department of Ophthalmology, Ege University, Izmir, Turkey
| |
Collapse
|
33
|
Kiel C, Weber BHF. Diagnostic testing in the genetically complex age-related macular degeneration. MED GENET-BERLIN 2025; 37:27-35. [PMID: 39943980 PMCID: PMC11812471 DOI: 10.1515/medgen-2024-2064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Age-related macular degeneration (AMD) is a leading cause of visual impairment with the risk of developing the disease influenced by a combination of genetic and environmental factors. With the recent expansion of treatment options, enhancing diagnostic accuracy and improving access to treatment are increasingly becoming the focus of interest. By using data from genome-wide association studies (GWAS) to generate polygenic risk scores (PRS), an assessment of an individual's genetic risk for AMD is feasible. While the predictive accuracy of the AMD-PRS is most robust for individuals at very high genetic risk, genetic diagnostic testing is warranted due to the large number of affected individuals resulting from the high prevalence of AMD. Early genetic confirmation of AMD-related pathology can facilitate timely treatment initiation, potentially improving patient outcomes.
Collapse
Affiliation(s)
- Christina Kiel
- University of RegensburgInstitute of Human GeneticsFranz-Josef-Strauss-Allee 1193053RegensburgGermany
| | - Bernhard H. F. Weber
- University of RegensburgInstitute of Human GeneticsFranz-Josef-Strauß-Allee 1193053RegensburgGermany
| |
Collapse
|
34
|
Vujosevic S, Limoli C, Kozak I. Hallmarks of aging in age-related macular degeneration and age-related neurological disorders: novel insights into common mechanisms and clinical relevance. Eye (Lond) 2025; 39:845-859. [PMID: 39289517 PMCID: PMC11933422 DOI: 10.1038/s41433-024-03341-5] [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: 04/14/2024] [Revised: 08/13/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024] Open
Abstract
Age-related macular degeneration (AMD) and age-related neurological diseases (ANDs), such as Alzheimer's and Parkinson's Diseases, are increasingly prevalent conditions that significantly contribute to global morbidity, disability, and mortality. The retina, as an accessible part of the central nervous system (CNS), provides a unique window to study brain aging and neurodegeneration. By examining the associations between AMD and ANDs, this review aims to highlight novel insights into fundamental mechanisms of aging and their role in neurodegenerative disease progression. This review integrates knowledge from the emerging field of aging research, which identifies common denominators of biological aging, specifically loss of proteostasis, impaired macroautophagy, mitochondrial dysfunction, and inflammation. Finally, we emphasize the clinical relevance of these pathways and the potential for cross-disease therapies that target common aging hallmarks. Identifying these shared pathways could open avenues to develop therapeutic strategies targeting mechanisms common to multiple degenerative diseases, potentially attenuating disease progression and promoting the healthspan.
Collapse
Affiliation(s)
- Stela Vujosevic
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy.
- Eye Clinic, IRCCS MultiMedica, Milan, Italy.
| | - Celeste Limoli
- Eye Clinic, IRCCS MultiMedica, Milan, Italy
- University of Milan, Milan, Italy
| | - Igor Kozak
- Moorfields Eye Hospital Centre, Abu Dhabi, UAE
- Ophthalmology and Vision Science, University of Arizona, Tucson, USA
| |
Collapse
|
35
|
Morino K, Miyake M, Nagasaki M, Kawaguchi T, Numa S, Mori Y, Yasukura S, Akada M, Nakao SY, Nakata A, Hashimoto H, Otokozawa R, Kamoi K, Takahashi H, Tabara Y, Matsuda F, Ohno-Matsui K, Tsujikawa A, Nagahama Study Group. Genome-wide Meta-analysis for Myopic Macular Neovascularization Identified a Novel Susceptibility Locus and Revealed a Shared Genetic Susceptibility with Age-Related Macular Degeneration. Ophthalmol Retina 2025; 9:367-377. [PMID: 39489378 DOI: 10.1016/j.oret.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 11/05/2024]
Abstract
PURPOSE To identify the susceptibility loci for myopic macular neovascularization (mMNV) in patients with high myopia. DESIGN A genome-wide association study (GWAS) meta-analysis (meta-GWAS). PARTICIPANTS We included 2783 highly myopic individuals, including 608 patients with mMNV and 2175 control participants without mMNV. METHODS We performed a meta-analysis of 3 independent GWASs conducted according to the genotyping platform (Illumina Asian Screening Array [ASA] data set, Illumina Human610 BeadChip [610K] data set, and whole genome sequencing [WGS] data set), adjusted for age, sex, axial length, and the first to third principal components. We used DeltaSVM to evaluate the binding affinity of transcription factors (TFs) to DNA sequences around the susceptibility of single nucleotide polymorphisms (SNPs). In addition, we evaluated the contribution of previously reported age-related macular degeneration (AMD) susceptibility loci. MAIN OUTCOME MEASURES The association between SNPs and mMNV in patients with high myopia. RESULTS The meta-GWAS identified rs56257842 at TEX29- LINC02337 as a novel susceptibility SNP for mMNV (odds ratio [OR]meta = 0.62, Pmeta = 4.63 × 10-8, I2 = 0.00), which was consistently associated with mMNV in all data sets (ORASA = 0.59, PASA = 1.71 × 10-4; OR610K = 0.63, P610K = 5.53 × 10-4; ORWGS = 0.66, PWGS = 4.38 × 10-2). Transcription factor-wide analysis showed that the TFs ZNF740 and EGR1 lost their binding affinity to this locus when rs56257842 had the C allele (alternative allele), and the WNT signaling-related TF ZBTB33 gained binding affinity when rs56257842 had the C allele. When we examined the associations of AMD susceptibility loci, rs12720922 at CETP showed a statistically significant association with mMNV (ORmeta = 0.52, Pmeta = 1.55 × 10-5), whereas rs61871745 near ARMS2 showed a marginal association (ORmeta = 1.25, Pmeta = 7.79 × 10-3). CONCLUSIONS Our study identified a novel locus associated with mMNV in high myopia. Subsequent analyses offered important insights into the molecular biology of mMNV, providing the potential therapeutic targets for mMNV. Furthermore, our findings imply shared genetic susceptibility between mMNV and AMD. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Kazuya Morino
- Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masahiro Miyake
- Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Masao Nagasaki
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Division of Biomedical Information Analysis, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Takahisa Kawaguchi
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shogo Numa
- Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuki Mori
- Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shota Yasukura
- Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masahiro Akada
- Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shin-Ya Nakao
- Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ai Nakata
- Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroki Hashimoto
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Division of Biomedical Information Analysis, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Ryoko Otokozawa
- Division of Biomedical Information Analysis, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Koju Kamoi
- Department of Ophthalmology and Visual Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Takahashi
- Department of Ophthalmology and Visual Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yasuharu Tabara
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, Shizuoka, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kyoko Ohno-Matsui
- Department of Ophthalmology and Visual Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akitaka Tsujikawa
- Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | |
Collapse
Collaborators
Takeo Nakayama, Akihiro Sekine, Shinji Kosugi, Yasuharu Tabara, Fumihiko Matsuda,
Collapse
|
36
|
Ottensmann L, Tabassum R, Ruotsalainen SE, Gerl MJ, Klose C, McCartney DL, Widén E, Simons K, Ripatti S, Vitart V, Hayward C, Pirinen M. Examining the link between 179 lipid species and 7 diseases using genetic predictors. EBioMedicine 2025; 114:105671. [PMID: 40157129 PMCID: PMC11995710 DOI: 10.1016/j.ebiom.2025.105671] [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: 12/05/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Genome-wide association studies of lipid species have identified several loci shared with various diseases, however, the relationship between lipid species and disease risk remains poorly understood. Here we investigated whether the plasma levels of lipid species are causally linked to disease risk. METHODS We built genetic predictors of 179 lipid species, measured in 7174 Finnish individuals, by utilising either 11 high-impact genomic loci or genome-wide polygenic scores (PGS). We assessed the impact of the lipid species on seven diseases by performing disease association across FinnGen (n = 500,348), UK Biobank (n = 420,531), and Generation Scotland (n = 20,032). We performed univariable Mendelian randomisation (MR) and multivariable MR (MVMR) analyses to examine whether lipid species impact disease risk independently of standard lipids. FINDINGS PGS explained >4% of the variance for 34 lipid species but variants outside the high-impact loci had only a marginal contribution. Variants within the high-impact loci showed association with all seven diseases. MVMR supported a causal role of ApoB in ischaemic heart disease after accounting for lipid species. Phosphatidylethanolamine-increasing LIPC variants seemed to lower age-related macular degeneration risk independently of HDL-cholesterol. MVMR suggested a protective effect of four lipid species containing arachidonic acid on cholelithiasis risk independently of Total Cholesterol. INTERPRETATION Our study demonstrates how genetic predictors of lipid species can be utilised to gain insights into disease risk. We report potential links between lipid species and age-related macular degeneration and cholelithiasis risk, which can be explored for their utility in disease risk prediction and therapy. FUNDING The funders had no role in the study design, data analyses, interpretation, or writing of this article.
Collapse
Affiliation(s)
- Linda Ottensmann
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland; Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom.
| | - Rubina Tabassum
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Sanni E Ruotsalainen
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | | | | | - Daniel L McCartney
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | - Elisabeth Widén
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | | | - Samuli Ripatti
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland; Department of Public Health, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Veronique Vitart
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | - Matti Pirinen
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland; Department of Public Health, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Mathematics and Statistics, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
37
|
Wang SK, Li J, Nair S, Korasaju R, Chen Y, Zhang Y, Kundaje A, Liu Y, Wang N, Chang HY. Single-cell multiome and enhancer connectome of human retinal pigment epithelium and choroid nominate pathogenic variants in age-related macular degeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.21.644670. [PMID: 40196652 PMCID: PMC11974679 DOI: 10.1101/2025.03.21.644670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Age-related macular degeneration (AMD) is a leading cause of vision loss worldwide. Genome-wide association studies (GWAS) of AMD have identified dozens of risk loci that may house disease targets. However, variants at these loci are largely noncoding, making it difficult to assess their function and whether they are causal. Here, we present a single-cell gene expression and chromatin accessibility atlas of human retinal pigment epithelium (RPE) and choroid to systematically analyze both coding and noncoding variants implicated in AMD. We employ HiChIP and Activity-by-Contact modeling to map enhancers in these tissues and predict cell and gene targets of risk variants. We further perform allele-specific self-transcribing active regulatory region sequencing (STARR-seq) to functionally test variant activity in RPE cells, including in the context of complement activation. Our work nominates new pathogenic variants and mechanisms in AMD and offers a rich and accessible resource for studying diseases of the RPE and choroid.
Collapse
Affiliation(s)
- Sean K Wang
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA
- Department of Ophthalmology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jiaying Li
- Department of Ophthalmology, Stanford University School of Medicine, Stanford, CA, USA
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Surag Nair
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Reshma Korasaju
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA
| | - Yun Chen
- Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yuanyuan Zhang
- Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Anshul Kundaje
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuwen Liu
- Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Kunpeng Institute of Modern Agriculture at Foshan, Chinese Academy of Agricultural Sciences, Foshan, China
| | - Ningli Wang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Henan Academy of Innovations in Medical Science, Henan, China
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Current address: Amgen Research, South San Francisco, CA, USA
- Lead contact
| |
Collapse
|
38
|
Chen SY, Xu YM, Tam POS, Pang CP, Tham CC, Yam JC, Chen LJ. Association of polymorphisms in the HTRA1 gene with myopia. Br J Ophthalmol 2025; 109:456-462. [PMID: 39406463 DOI: 10.1136/bjo-2024-325935] [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: 06/08/2024] [Accepted: 09/29/2024] [Indexed: 03/22/2025]
Abstract
PURPOSE To evaluate the associations of single-nucleotide polymorphisms (SNPs) in the high-temperature requirement protease A 1 (HTRA1) gene with myopia. METHODS 25 SNPs in HTRA1 were selected, including 23 haplotype-tagging SNPs, SNP rs2142308 from a previous genome-wide association study (GWAS) of myopia and rs11200638, a SNP strongly associated with age-related macular degeneration (AMD). All SNPs were genotyped in a Hong Kong Chinese cohort of 533 myopia subjects (including 175 high myopia, 189 moderate myopia and 189 mild myopia) and 280 non-myopic controls. The association of individual SNPs were evaluated in overall myopia and different subgroups of myopia using logistic regression. RESULTS A tagging SNP, rs11200647, was significantly associated with myopia (p=2.17×10-4, OR=0.67). Nominal associations were detected for the AMD-associated SNP rs11200638 (p=0.0042, OR=1.37) and tagging SNPs rs12266322 (p=0.0048, OR=0.59) and rs17103569 (p=0.047, OR=1.34). The association of rs11200647 with myopia remained significant after adjusting for rs11200638, rs12266322 and rs17103569. In sub-group analysis, two tagging SNPs, rs11200647 (p=2.24×10-4, OR=0.58) and rs12266322 (p=8.31×10-4, OR=0.39), showed significant association with moderate myopia. In haplotype association analysis, haplotypes AT (p=1.00×10-4, OR=1.77) and haplotype GT (p=0.0019, OR=0.64), defined by rs11200647 and rs66884382, were significantly associated with myopia. CONCLUSIONS This study provided new evidence to support HTRA1 as an associated gene for myopia, especially moderate myopia. The findings suggested that myopia and AMD may have shared genetic components.
Collapse
Affiliation(s)
- Shu Ying Chen
- Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - You Mei Xu
- Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Pancy O S Tam
- Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Chi Pui Pang
- Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Clement C Tham
- Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Jason C Yam
- Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Li Jia Chen
- Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, Hong Kong, Hong Kong
| |
Collapse
|
39
|
Lee SSY, Stapleton F, MacGregor S, Mackey DA. Genome-wide association studies, Polygenic Risk Scores and Mendelian randomisation: an overview of common genetic epidemiology methods for ophthalmic clinicians. Br J Ophthalmol 2025; 109:433-441. [PMID: 39622623 PMCID: PMC12013552 DOI: 10.1136/bjo-2024-326554] [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: 09/23/2024] [Accepted: 11/17/2024] [Indexed: 01/12/2025]
Abstract
Genetic information will be increasingly integrated into clinical eye care within the current generation of ophthalmologists. For monogenic diseases such as retinoblastoma, genetic studies have been relatively straightforward as these conditions result from pathogenic variants in a single gene resulting in large physiological effects. However, most eye diseases result from the cumulative effects of multiple genetic variants and environmental factors. In such diseases, because each variant usually has an individually small effect, genetic studies for complex diseases are comparatively more challenging. This article aims to provide an overview of three genetic epidemiology methods for polygenic (or complex) diseases: genome-wide association studies (GWAS), Polygenic Risk Scores (PRS) and Mendelian randomisation (MR). A GWAS systematically conducts association analyses of a trait of interest against millions of genetic variants, usually in the form of single nucleotide polymorphisms, across the genome. GWAS findings can then be used for PRS construction and MR analyses. To construct a PRS, the cumulative effect of many genetic variants associated with a trait from a prior GWAS is calculated and taken as a quantitative representation of an individual's genetic risk of a complex disease. MR studies analyse an outcome measure against the genetic variants of an exposure, and are particularly useful in investigating causal relations between two traits where randomised controlled trials are not possible or ethical. In addition to explaining the principles of these three genetic epidemiology concepts, this article provides a minimally technical description of their basic methodology that is accessible to the non-expert reader.
Collapse
Affiliation(s)
- Samantha Sze-Yee Lee
- Genetics and Epidemiology, Lions Eye Institute, Nedlands, Western Australia, Australia
- Centre for Ophthalmology and Visual Sciences, University of Western Australia, Nedlands, Western Australia, Australia
- School of Optometry and Vision Science, UNSW, Sydney, New South Wales, Australia
| | - Fiona Stapleton
- School of Optometry and Vision Science, UNSW, Sydney, New South Wales, Australia
| | - Stuart MacGregor
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - David A Mackey
- Genetics and Epidemiology, Lions Eye Institute, Nedlands, Western Australia, Australia
- Centre for Ophthalmology and Visual Sciences, University of Western Australia, Nedlands, Western Australia, Australia
- Centre for Ophthalmology and Visual Science, Lions Eye Institute, Nedlands, Western Australia, Australia
- Centre for Eye Research Australia, Department of Ophthalmology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
40
|
Li H, Sharma R, Bharti K. iPSC-derived retinal pigment epithelium: an in vitro platform to reproduce key cellular phenotypes and pathophysiology of retinal degenerative diseases. Stem Cells Transl Med 2025; 14:szae097. [PMID: 39729520 PMCID: PMC11954503 DOI: 10.1093/stcltm/szae097] [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: 09/18/2024] [Accepted: 11/30/2024] [Indexed: 12/29/2024] Open
Abstract
Retinal pigment epithelium (RPE) atrophy is a significant cause of human blindness worldwide, occurring in polygenic diseases such as age-related macular degeneration (AMD) and monogenic diseases such as Stargardt diseases (STGD1) and late-onset retinal degeneration (L-ORD). The patient-induced pluripotent stem cells (iPSCs)-derived RPE (iRPE) model exhibits many advantages in understanding the cellular basis of pathological mechanisms of RPE atrophy. The iRPE model is based on iPSC-derived functionally mature and polarized RPE cells that reproduce several features of native RPE cells, such as phagocytosis of photoreceptor outer segments (POS) and replenishment of visual pigment. When derived from patients, iRPE are able to recapitulate critical cellular phenotypes of retinal degenerative diseases, such as the drusen-like sub-RPE deposits in the L-ORD and AMD models; lipid droplets and cholesterol accumulation in the STGD1 and AMD models. The iRPE model has helped discover the unexpected role of RPE in understanding retinal degenerative diseases, such as a cell-autonomous function of ABCA4 in STGD1. The iRPE model has helped uncover the pathological mechanism of retinal degenerative diseases, including the roles of alternate complement cascades and oxidative stress in AMD pathophysiology, abnormal POS processing in STGD1 and L-ORD, and its association with lipid accumulation. These studies have helped better understand-the role of RPE in retinal degenerative diseases, and molecular mechanisms underlying RPE atrophy, and have provided a basis to discover therapeutics to target RPE-associated diseases.
Collapse
Affiliation(s)
- Huirong Li
- NEI/OSCTRS/OGVFB, Bethesda, MD, United States
| | | | | |
Collapse
|
41
|
Eichenbaum DA, Holekamp N, Khanani AM, Pieramici D, Hershberger V, Sheth V, Brunstein F, Ma L, Zou Y, Indjeian VB, Dere R, Maia M, Hsu JC, Gao SS, Yaspan B, Willis JR, Wiley H, Lai P, Chen H. Phase 2 Study of the Anti-High Temperature Requirement A1 (HtrA1) Fab Galegenimab (FHTR2163) in Geographic Atrophy Secondary to Age-Related Macular Degeneration. Am J Ophthalmol 2025; 275:14-26. [PMID: 40089174 DOI: 10.1016/j.ajo.2025.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/10/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
PURPOSE To investigate the safety, tolerability, and efficacy of intravitreal injection of galegenimab, an anti-HtrA1 FAb, in patients with geographic atrophy (GA) secondary to age-related macular degeneration (AMD). DESIGN Phase 2, single-masked, randomized clinical trial. METHODS Eligible GA patients with BCVA letter scores of ≥ 24 letters and baseline GA lesion size 2.54∼25.4 mm2 in the study eye were enrolled. Patients were randomized 2:1:2 to receive 20 mg galegenimab every 4 (Q4W) or 8 weeks (Q8W), or sham Q4/8 W. The primary endpoint was mean change in GA area from baseline to Week 72 measured by fundus autofluorescence. A data monitoring committee (DMC) conducted periodic unmasked review of cumulative safety/limited efficacy data of the ongoing study. RESULTS Among 337 patients who received ≥ 1 dose and have at least one postbaseline GA area measurement, the adjusted mean change in GA area from baseline to Week 72 was 2.67, 2.50, and 2.38 mm2 for the galegenimab Q4W, galegenimab Q8W, and pooled sham arms, respectively. Differences between the treated and sham groups were not statistically significant. However, the rate of intraocular inflammation was high (7.1%, 16/224 patients) among treated patients. The DMC recommended early termination of the study based on an early benefit/risk analysis. CONCLUSION Galegenimab administration did not show a difference in mean change in GA area from baseline to Week 72 compared with sham. Inhibition of HtrA1 with a Fab did not slow down GA progression.
Collapse
Affiliation(s)
- David A Eichenbaum
- From the Retina Vitreous Associates of Florida (D.A.E.), St. Petersburg, Florida, USA.
| | | | - Arshad M Khanani
- Sierra Eye Associates (A.M.K.), Reno, Nevada, USA; Reno School of Medicine (A.M.K.), University of Nevada, Reno, Nevada, USA
| | - Dante Pieramici
- California Retina Consultants (D.P.), Santa Barbara, California, USA
| | | | - Veeral Sheth
- University Retina and Macula Associates (V.S.), Oak Forest, Illinois, USA
| | - Flavia Brunstein
- Genentech, Inc. (F.B., L.M., Y.Z., Y.B.I., R.D., M.M., J.C.H., S.S.G., B.Y., J.R.W., H.W., P.L., H.C.), South San Francisco, California, USA
| | - Ling Ma
- Genentech, Inc. (F.B., L.M., Y.Z., Y.B.I., R.D., M.M., J.C.H., S.S.G., B.Y., J.R.W., H.W., P.L., H.C.), South San Francisco, California, USA
| | - Yixuan Zou
- Genentech, Inc. (F.B., L.M., Y.Z., Y.B.I., R.D., M.M., J.C.H., S.S.G., B.Y., J.R.W., H.W., P.L., H.C.), South San Francisco, California, USA
| | - Vahan B Indjeian
- Genentech, Inc. (F.B., L.M., Y.Z., Y.B.I., R.D., M.M., J.C.H., S.S.G., B.Y., J.R.W., H.W., P.L., H.C.), South San Francisco, California, USA
| | - Randall Dere
- Genentech, Inc. (F.B., L.M., Y.Z., Y.B.I., R.D., M.M., J.C.H., S.S.G., B.Y., J.R.W., H.W., P.L., H.C.), South San Francisco, California, USA
| | - Mauricio Maia
- Genentech, Inc. (F.B., L.M., Y.Z., Y.B.I., R.D., M.M., J.C.H., S.S.G., B.Y., J.R.W., H.W., P.L., H.C.), South San Francisco, California, USA
| | - Joy C Hsu
- Genentech, Inc. (F.B., L.M., Y.Z., Y.B.I., R.D., M.M., J.C.H., S.S.G., B.Y., J.R.W., H.W., P.L., H.C.), South San Francisco, California, USA
| | - Simon S Gao
- Genentech, Inc. (F.B., L.M., Y.Z., Y.B.I., R.D., M.M., J.C.H., S.S.G., B.Y., J.R.W., H.W., P.L., H.C.), South San Francisco, California, USA
| | - Brian Yaspan
- Genentech, Inc. (F.B., L.M., Y.Z., Y.B.I., R.D., M.M., J.C.H., S.S.G., B.Y., J.R.W., H.W., P.L., H.C.), South San Francisco, California, USA
| | - Jeffrey R Willis
- Genentech, Inc. (F.B., L.M., Y.Z., Y.B.I., R.D., M.M., J.C.H., S.S.G., B.Y., J.R.W., H.W., P.L., H.C.), South San Francisco, California, USA
| | - Henry Wiley
- Genentech, Inc. (F.B., L.M., Y.Z., Y.B.I., R.D., M.M., J.C.H., S.S.G., B.Y., J.R.W., H.W., P.L., H.C.), South San Francisco, California, USA
| | - Phillip Lai
- Genentech, Inc. (F.B., L.M., Y.Z., Y.B.I., R.D., M.M., J.C.H., S.S.G., B.Y., J.R.W., H.W., P.L., H.C.), South San Francisco, California, USA
| | - Hao Chen
- Genentech, Inc. (F.B., L.M., Y.Z., Y.B.I., R.D., M.M., J.C.H., S.S.G., B.Y., J.R.W., H.W., P.L., H.C.), South San Francisco, California, USA
| |
Collapse
|
42
|
Schikora J, Dort A, Wolf HN, Józsi M, Pouw RB, Bertelmann T, Bahlmann D, van Oterendorp C, Feltgen N, Hoerauf H, Pauly D, Klemming J. Decreased complement 4 and interleukin-10 as biomarkers in aqueous humour for non-exudative age-related macular degeneration: a case control study. J Transl Med 2025; 23:317. [PMID: 40075380 PMCID: PMC11905602 DOI: 10.1186/s12967-024-05909-x] [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: 07/09/2024] [Accepted: 11/21/2024] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND The development of age-related macular degeneration (AMD) is influenced by risk factors that contribute to inflammatory processes, cellular stress responses, and a dysregulation of the complement system. Given the incomplete understanding of the pathogenesis of AMD and the necessity for novel therapeutics, biomarker studies investigating aqueous humour from the anterior chamber of the eye serve as a valuable tool. This pilot study aimed to assess inflammatory mediators and complement components in aqueous humour of non-exudative AMD patients in comparison with a control group. METHODS The aqueous humour of 12 non-exudative AMD patients and 21 control subjects was collected during cataract surgery. Levels of 78 inflammatory proteins and complement components were measured using multiplex immunoassays. The influence of sex or smoking on the AMD status was assessed using Pearson's chi-square test. Biomarker levels between AMD patients vs. controls, smokers vs. non-smokers, and females vs. males were compared. Parametric datasets were analysed using independent-means t-test, while non-parametric data analysis was conducted utilising Wilcoxon's rank-sum test. Spearman's correlation investigated associations between drusen volume and biomarker levels, as well as biomarker levels and subject age. RESULTS All examined 78 immunological factors were detectable in aqueous humour. The proteins were categorised into high, medium, and low level groups. Aqueous humour contained high levels of complement proteins, including iC3b, FH/FHL-1, C4B, and FI. Non-exudative AMD patients exhibited decreased levels of C4 (P = 0.020), IL-10 (P = 0.033), and FI (P = 0.082). A positive correlation was observed between drusen volume and CCL4 levels (rS = 0.78, P = 0.013). Furthermore, smokers demonstrated significantly increased levels of pro-inflammatory proteins (CCL7, IL-7; P = 0.027, P = 0.030). MMP-1 was positively correlated with age (rS = 0.44, P = 0.010), while sex differences were observed in FB (P = 0.027) and C4B (P = 0.036) levels. CONCLUSIONS This pilot study presents an initial overview of inflammation-associated biomarkers in the aqueous humour, highlighting potential roles for C4 and IL-10 in the development of non-exudative AMD. A larger, more-focused follow-up study is in progress to further investigate biomarkers localised to the eye and refine our understanding of AMD.
Collapse
Affiliation(s)
- Juliane Schikora
- Experimental Ophthalmology, University Marburg, Marburg, Germany
| | - Aaron Dort
- Experimental Ophthalmology, University Marburg, Marburg, Germany
| | - Hannah N Wolf
- Experimental Ophthalmology, University Marburg, Marburg, Germany
| | - Mihály Józsi
- Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Richard B Pouw
- Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Thomas Bertelmann
- Department of Ophthalmology, University Medical Center Göttingen, Göttingen, Germany
| | - Dirk Bahlmann
- Department of Ophthalmology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Nicolas Feltgen
- Department of Ophthalmology, University Basel, Basel, Switzerland
| | - Hans Hoerauf
- Department of Ophthalmology, University Medical Center Göttingen, Göttingen, Germany
| | - Diana Pauly
- Experimental Ophthalmology, University Marburg, Marburg, Germany.
| | - Jannis Klemming
- Department of Ophthalmology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
43
|
Markakis D, Britten-Jones AC, Guymer RH, Edwards TL, Hall AJ, Kerr NM, Ng W, Skalicky S, Ayton LN, Mack HG. Retrospective audit reviewing accuracy of clinical diagnosis of geographic atrophy in a single centre private tertiary retinal practice in Australia. Sci Rep 2025; 15:8528. [PMID: 40074767 PMCID: PMC11903886 DOI: 10.1038/s41598-025-90516-z] [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: 12/15/2024] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
Accurate diagnosis of both age-related macular degeneration (AMD) and inherited retinal diseases (IRD) with macular atrophy is important because treatments for both conditions are emerging. Phenotypical similarities between macular atrophy associated with AMD (geographic atrophy, GA) and IRD-associated atrophy exist, which can make accurate diagnosis challenging in clinical practice. Misdiagnosis may lead to inappropriate treatment strategies and missed opportunities for disease-specific interventions. A retrospective clinical review of medical records of people diagnosed with AMD between 1995 and 2023 from a large multidisciplinary private ophthalmic practice in Australia was undertaken to identify cases of patients diagnosed with geographic atrophy without drusen, which was then further assessed for potentially missed IRD with macular atrophy. Flagged cases were presented to experts in AMD and IRD to establish a most-likely diagnosis. Cases without consensus between graders were grouped into most-likely diagnosis by a third senior retinal clinician. Of the 1136 cases reviewed, the possible rate of misdiagnosis observed was 1.9%, with 1.0% representing potentially missed IRDs, most commonly pattern dystrophy (0.5%). A multi-modal approach, including clinical features and patient history, is important to limit the possibility of misdiagnosis of GA, and identify a subset of patients who might benefit from genetic testing prior to considering possible treatments.
Collapse
Affiliation(s)
- Demi Markakis
- Cabrini Hospital, Malvern, Australia
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Australia
- The Alfred Hospital, Prahran, Australia
| | - Alexis Ceecee Britten-Jones
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
- Department of Optometry and Vision Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Robyn H Guymer
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Thomas L Edwards
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Anthony J Hall
- Cabrini Hospital, Malvern, Australia
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Australia
| | - Nathan M Kerr
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
- Eye Surgery Associates, 2/232 Victoria Pde, East Melbourne, VIC, 3002, Australia
| | - Weng Ng
- Eye Surgery Associates, 2/232 Victoria Pde, East Melbourne, VIC, 3002, Australia
| | - Simon Skalicky
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
- Eye Surgery Associates, 2/232 Victoria Pde, East Melbourne, VIC, 3002, Australia
| | - Lauren N Ayton
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
- Department of Optometry and Vision Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Heather G Mack
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Australia.
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia.
- Eye Surgery Associates, 2/232 Victoria Pde, East Melbourne, VIC, 3002, Australia.
| |
Collapse
|
44
|
Li J, Nagasaka Y, Shen H, Zhou X, Ma J, Trevisan-Silva D, Sherman NE, Ambati J, Gelfand BD, Guo LW. TMEM97 governs partial epithelial-mesenchymal transition of retinal pigment epithelial cells via the CTNND2-ADAM10 axis. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102460. [PMID: 39995975 PMCID: PMC11848774 DOI: 10.1016/j.omtn.2025.102460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/17/2025] [Indexed: 02/26/2025]
Abstract
Epithelial-mesenchymal transition (EMT) is associated with retinal pigment epithelium (RPE) dysfunction in degenerative retinal diseases. However, the role of partial EMT (pEMT), a hybrid state exhibiting both epithelial and mesenchymal markers, remains poorly understood in this context. Our previous research demonstrated that TMEM97 ablation in mice worsens photoreceptor loss in an oxidant-induced RPE damage model. Here, we link TMEM97 to pEMT in RPE cells and explore the underlying molecular mechanisms. We found that re-expressing TMEM97 in the RPE of TMEM97-knockout mice, via subretinal lentiviral delivery, mitigated oxidant (NaIO3)-induced photoreceptor loss. Interestingly, TMEM97 knockout in ARPE19 cells in vitro led to upregulation of cadherin/adhesion-binding pathways, even without oxidant exposure. Integrated proteomic, transcriptomic, segmentation, and immunoblot analyses revealed that TMEM97 ablation induces pEMT, marked by the concurrent expression of epithelial E-cadherin and mesenchymal N-cadherin, a process reversed upon TMEM97 re-expression. Furthermore, TMEM97 negatively regulated CTNND2 protein (catenin δ-2), but not the known EMT driver β-catenin, and CTNND2 was found to promote ADAM10, which sustains both E- and N-cadherin protein levels. These findings identify TMEM97 as a novel regulator of RPE-cell pEMT through the CTNND2-ADAM10 axis, highlighting potential new targets for therapeutic intervention in RPE-related pathophysiology.
Collapse
Affiliation(s)
- Jing Li
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Yosuke Nagasaka
- Department of Ophthalmology, University of Virginia, Charlottesville, VA 22903, USA
| | - Hongtao Shen
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Xinyu Zhou
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Jianjie Ma
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Dilza Trevisan-Silva
- School of Medicine Core Facilities, University of Virginia, Charlottesville, VA 22903, USA
| | - Nicholas E. Sherman
- School of Medicine Core Facilities, University of Virginia, Charlottesville, VA 22903, USA
| | - Jayakrishna Ambati
- Department of Ophthalmology, University of Virginia, Charlottesville, VA 22903, USA
| | - Bradley D. Gelfand
- Department of Ophthalmology, University of Virginia, Charlottesville, VA 22903, USA
| | - Lian-Wang Guo
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
- Department of Ophthalmology, University of Virginia, Charlottesville, VA 22903, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA
| |
Collapse
|
45
|
Jia R, Yin Y, Shan H. Systemic inflammatory response index as a novel biomarker for age-related macular degeneration: a cross-sectional study from NHANES (2005-2008). Front Nutr 2025; 12:1540933. [PMID: 40115389 PMCID: PMC11922706 DOI: 10.3389/fnut.2025.1540933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/24/2025] [Indexed: 03/23/2025] Open
Abstract
Background Chronic low-grade systemic inflammation plays a significant role in age-related macular degeneration (AMD) pathogenesis. The systemic inflammatory response index (SIRI), a novel inflammatory marker, may predict various diseases. However, data on the relationship between SIRI and AMD are limited. This study examines the relationship between SIRI and AMD and assesses its potential as a predictive biomarker. Methods A cross-sectional analysis of the National Health and Nutrition Examination Survey (NHANES) data from 2005 to 2008 was conducted on participants aged ≥40 years with SIRI and AMD status data. Multivariable logistic regression models adjusted for confounders were used to assess the association. Sensitivity and subgroup analyses, along with restricted cubic spline (RCS) curve analysis, were performed. Results Among 5,365 participants, 425 (7.9%) had AMD. The median SIRI was higher in AMD patients (1.23 vs. 1.04, p < 0.001). Higher SIRI was independently associated with increased odds (adjusted OR: 1.18, 95% CI:1.07-1.29, p = 0.001). RCS analyses revealed a dose-response relationship (p = 0.002). Subgroup analyses showed a positive association in male participants, individuals with hypertension, individuals with obesity, and non-smokers. Higher SIRI levels were independently associated with increased AMD risk (adjusted OR: 1.27, 95% CI: 1.03-1.56, p = 0.023). Conclusion Elevated SIRI is independently associated with increased AMD risk in the U.S. population. SIRI may serve as a biomarker for identifying high-risk individuals, enabling early intervention. The cross-sectional design limits causal inference, and unmeasured confounders may affect the results. SIRI could potentially serve as a non-invasive biomarker for AMD risk, pending further validation through longitudinal studies.
Collapse
Affiliation(s)
- Ruoshuang Jia
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yiqing Yin
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Huimin Shan
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
46
|
Shaw EM, Anderson DM, Periasamy R, Schey KL, Curcio CA, Lipinski DM. Porcine Sub-Retinal Pigment Epithelium Deposits: A Model for Dry Age-Related Macular Degeneration With Comparison to Human Drusen. Invest Ophthalmol Vis Sci 2025; 66:18. [PMID: 40048184 PMCID: PMC11895847 DOI: 10.1167/iovs.66.3.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/07/2025] [Indexed: 03/14/2025] Open
Abstract
Purpose Due to the slowly progressing nature of age-related macular degeneration (AMD) and critical differences in ocular anatomy between humans and animals, it has been difficult to model disease progression, hampering the development of novel therapeutics aimed at impacting drusen biogenesis. To determine whether "drusen-in-a-dish" model systems are of utility in screening potential therapeutics aimed at early-intermediate dry AMD, we developed a detailed characterization of the protein, glycoprotein, and lipid composition of sub-retinal pigment epithelium (RPE) deposits grown by monolayers of ex vivo porcine RPE with human drusen in AMD globes. Methods Immunohistochemistry and imaging mass spectrometry (IMS) were performed on 20-week aged monolayers of porcine RPE and human donor globes recovered from an 81-year-old non-transplant donor with confirmed diagnosis of bilateral dry AMD. The presence of major protein, glycoprotein, and lipid species was compared between porcine sub-RPE deposits and human drusen with reference to macular/peripheral eccentricity. Results The protein and glycoprotein composition of porcine sub-RPE deposits closely mimics human drusen identified in donor globes with dry AMD, including the presence of major complement components (C9, CFH, CHI), apolipoproteins (ApoE, ApoJ), extracellular matrix proteins (vitronectin, collagen VI), and calcification (hydroxyapatite). Sub-RPE deposits were additionally rich in long-chain ceramide species (Cer, CerPE, PI), which have only recently been described in human drusen. Conclusions Due to their compositional similarity to human drusen, ex vivo "drusen-in-a-dish" systems represent a potentially robust and cost-effective model for both studying the pathobiology of drusen biogenesis and screening novel therapeutics aimed at limiting drusen formation.
Collapse
Affiliation(s)
- Erika M. Shaw
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - David M. Anderson
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, United States
| | - Ramesh Periasamy
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Kevin L. Schey
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, United States
| | - Christine A. Curcio
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, United States
| | - Daniel M. Lipinski
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| |
Collapse
|
47
|
Larsen PP, Dinet V, Delcourt C, Helmer C, Linard M. Could Infectious Agents Play a Role in the Onset of Age-related Macular Degeneration? A Scoping Review. OPHTHALMOLOGY SCIENCE 2025; 5:100668. [PMID: 39906411 PMCID: PMC11791433 DOI: 10.1016/j.xops.2024.100668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/19/2024] [Accepted: 11/25/2024] [Indexed: 02/06/2025]
Abstract
Topic This scoping review aims to summarize the current state of knowledge on the potential involvement of infections in age-related macular degeneration (AMD). Clinical relevance Age-related macular degeneration is a multifactorial disease and the leading cause of vision loss among older adults in developed countries. Clarifying whether certain infections participate in its onset or progression seems essential, given the potential implications for treatment and prevention. Methods Using the PubMed database, we searched for articles in English, published until June 1, 2023, whose title and/or abstract contained terms related to AMD and infections. All types of study design, infectious agents, AMD diagnostic methods, and AMD stages were considered. Articles dealing with the oral and gut microbiota were not included but we provide a brief summary of high-quality literature reviews recently published on the subject. Results Two investigators independently screened the 868 articles obtained by our algorithm and the reference lists of selected studies. In total, 40 articles were included, among which 30 on human data, 9 animal studies, 6 in vitro experiments, and 1 hypothesis paper (sometimes with several data types in the same article). Of these, 27 studies were published after 2010, highlighting a growing interest in recent years. A wide range of infectious agents has been investigated, including various microbiota (nasal, pharyngeal), 8 bacteria, 6 viral species, and 1 yeast. Among them, most have been investigated anecdotally. Only Chlamydia pneumoniae, Cytomegalovirus, and hepatitis B virus received more attention with 17, 6, and 4 studies, respectively. Numerous potential pathophysiological mechanisms have been discussed, including (1) an indirect role of infectious agents (i.e. a role of infections located distant from the eye, mainly through their interactions with the immune system) and (2) a direct role of some infectious agents implying potential infection of various cells types within AMD-related tissues. Conclusions Overall, this review highlights the diversity of possible interactions between infectious agents and AMD and suggests avenues of research to enrich the data currently available, which provide an insufficient level of evidence to conclude whether or not infectious agents are involved in this pathology. Financial Disclosures Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Petra P. Larsen
- University of Bordeaux, INSERM, BPH, U1219, Bordeaux, France
| | - Virginie Dinet
- INSERM, Biologie des Maladies Cardiovasculaires, U1034, University of Bordeaux, Pessac, France
| | - Cécile Delcourt
- University of Bordeaux, INSERM, BPH, U1219, Bordeaux, France
| | | | - Morgane Linard
- University of Bordeaux, INSERM, BPH, U1219, Bordeaux, France
| |
Collapse
|
48
|
Salman A, Song WK, Storm T, McClements ME, MacLaren RE. CRISPR targeting of SNPs associated with age-related macular degeneration in ARPE-19 cells: a potential model for manipulating the complement system. Gene Ther 2025; 32:132-141. [PMID: 40102632 PMCID: PMC11946884 DOI: 10.1038/s41434-025-00522-z] [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: 07/17/2024] [Revised: 01/10/2025] [Accepted: 02/14/2025] [Indexed: 03/20/2025]
Abstract
Age-related Macular degeneration (AMD) is a major cause of vision loss and is linked to several predisposing single nucleotide polymorphisms (SNPs). CRISPR-mediated genome editing offers the potential to target negatively associated SNPs in an allele-specific manner, necessitating the need for a relevant cell model. The ARPE-19 cell line, with its stable monolayer growth and retinal pigment epithelium (RPE) characteristics, serves as an ideal model for AMD studies. Chronic inflammation and complement system dysregulation are implicated in AMD pathogenesis. Most genetic variations associated with AMD are in complement genes, suggesting their regulatory role. In this study, we conducted targeted PCRs to identify AMD-related SNPs in ARPE-19 cells and used CRISPR constructs to assess allele-specific activity. Guide RNA sequences were cloned into an EF-1-driven SpCas9 vector and packaged into lentivirus. Targeting efficiencies were evaluated with TIDE analysis, and allele-specificity was measured with NGS analysis 30 days post-transduction. Our results showed varying targeting efficiencies depending on guide RNA efficacy. For example, TIDE analysis of CFH SNPs rs1061170 and rs1410996 revealed efficiencies of 35.5% and 33.8%, respectively. CFB SNP rs4541862 showed efficiencies from 3% to 36.7%, and rs641153 ranged from 3.4% to 23.8%. Additionally, allele-specific targeting of AMD-related SNPs rs1061170, rs1410996, rs4541862, and rs641153 ranged from 48% to 52% in heterozygous differentiated ARPE-19 cells. These findings demonstrate the potential to manipulate the complement system in an AMD model by targeting disease-associated SNPs in an allele-specific manner, offering a promising therapeutic approach.
Collapse
Affiliation(s)
- Ahmed Salman
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.
| | - Won Kyung Song
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Gangnum Yonsei Eye Clinic, Seoul, Republic of South Korea
| | - Tina Storm
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | - Robert E MacLaren
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
49
|
Tang C, Zhou QQ, Huang XF, Ju YY, Rao BL, Liu ZC, Jia YA, Bai ZP, Lin QY, Liu L, Qu J, Zhang J, Gao ML. Integration and functionality of human iPSC-derived microglia in a chimeric mouse retinal model. J Neuroinflammation 2025; 22:53. [PMID: 40016767 PMCID: PMC11869422 DOI: 10.1186/s12974-025-03393-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/20/2025] [Indexed: 03/01/2025] Open
Abstract
INTRODUCTION Microglia, the resident immune cells of the central nervous system, play a pivotal role in maintaining homeostasis, responding to injury, and modulating neuroinflammation. However, the limitations of rodent models in accurately representing human microglia have posed significant challenges in the study of retinal diseases. METHODS PLX5622 was used to eliminate endogenous microglia in mice through oral and intraperitoneal administration, followed by transplantation of human induced pluripotent stem cell-derived microglia (hiPSC-microglia, iMG) into retinal explants to create a novel ex vivo chimeric model containing xenotransplanted microglia (xMG). The number and proportion of xMG in the retina were quantified using retinal flat-mounting and immunostaining. To evaluate the proliferative capacity and synaptic pruning ability of xMG, the expression of Ki-67 and the phagocytosis of synaptic proteins SV2 and PSD95 was assessed. The chimeric model was stimulated with LPS, and single-cell RNA sequencing (scRNA-seq) was used to analyze transcriptomic changes in iMG and xMG. Mouse IL-34 antibody neutralization experiments were performed, and the behavior of xMG in retinal degenerative Pde6b-/- mice was examined. RESULTS We demonstrated that xenotransplanted microglia (xMG) successfully migrated to and localized within the mouse retina, adopting homeostatic morphologies. Our approach achieved over 86% integration of human microglia, which maintained key functions including proliferation, immune responsiveness, and synaptic pruning over a 14-day culture period. scRNA-seq of xMG revealed a shift in microglial signatures compared to monoculture iMG, indicating a transition to a more in vivo-like phenotype. In retinal degenerative Pde6b-/- mice, xMG exhibited activation and migrated toward degenerated photoreceptors. CONCLUSION This model provides a powerful platform for studying human microglia in the retinal context, offering significant insights for advancing research into retinal degenerative diseases and developing potential therapeutic strategies. Future applications of this model include using patient-derived iPSCs to investigate disease-specific microglial behaviors, thereby enhancing our understanding of microglia-related pathogenesis.
Collapse
Affiliation(s)
- Chun Tang
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Qi-Qi Zhou
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiu-Feng Huang
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Ya-Yi Ju
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Bi-Lin Rao
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhi-Cong Liu
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yi-An Jia
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhan-Pei Bai
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Qing-Yang Lin
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Lin Liu
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jia Qu
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China.
- The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, 325027, China.
| | - Jun Zhang
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China.
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China.
- Lead Contact, Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| | - Mei-Ling Gao
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China.
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China.
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| |
Collapse
|
50
|
VandenBosch LS, Cherry TJ. Machine Learning Prediction of Non-Coding Variant Impact in Cell-Class-Specific Human Retinal Cis-Regulatory Elements. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.22.638679. [PMID: 40060626 PMCID: PMC11888276 DOI: 10.1101/2025.02.22.638679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Non-coding variants in cis-regulatory elements such as promoters and enhancers contribute to inherited retinal diseases (IRDs), however, characterizing the functional impact of most regulatory variants remains challenging. To improve identification of variants of interest, we implemented machine learning using a gapped k-mer support vector machine approach trained on single nucleus ATAC-seq data from specific cell classes of the adult and developing human retina. We developed 18 distinct ML models to predict the impact of non-coding variants on 39,437 cell-class-specific regulatory elements. These models demonstrate accuracy over 90% and a high degree of cell class specificity. Variant Impact Prediction (VIP) scores highlight specific sequences within candidate CREs, including putative transcription factor (TF) binding motifs, that are predicted to alter CRE function if mutated. Correlations to massively parallel reporter assays support the predictive value of VIP scores to model single nucleotide variants and indels in a cell-class-specific manner. These analyses demonstrate the capacity for single nucleus epigenomic data to predict the impact of non-coding sequence variants and allow for rapid prioritization of patient variants for further functional analysis.
Collapse
Affiliation(s)
- Leah S VandenBosch
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA
| | - Timothy J Cherry
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA
- University of Washington Department of Pediatrics, Seattle, Washington, USA
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
| |
Collapse
|