1
|
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
|
2
|
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 DOI: 10.1167/tvst.14.5.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [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
|
3
|
Ong J, Selvam A, Driban M, Zarnegar A, Morgado Mendes Antunes Da Silva SI, Joy J, Rossi EA, Vande Geest JP, Sahel JA, Chhablani J. Characterizing Bruch's membrane: State-of-the-art imaging, computational segmentation, and biologic models in retinal disease and health. Prog Retin Eye Res 2025; 106:101358. [PMID: 40254245 DOI: 10.1016/j.preteyeres.2025.101358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
The Bruch's membrane (BM) is an acellular, extracellular matrix that lies between the choroid and retinal pigment epithelium (RPE). The BM plays a critical role in retinal health, performing various functions including biomolecule diffusion and RPE support. The BM is also involved in many retinal diseases, and insights into BM dysfunction allow for further understanding of the pathophysiology of various chorioretinal pathologies. Thus, characterization of the BM serves as an important area of research to further understand its involvement in retinal disease. In this article, we provide a review of various advancements in characterizing and visualizing the BM. We provide an overview of the BM in retinal health, as well as changes observed in aging and disease. We then describe current state-of-the-art imaging modalities and advances to further visualize the BM including various types of optical coherence tomography imaging, near-infrared reflectance (NIR), and autofluorescence imaging and tissue matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI-IMS). Following advances in imaging of the BM, we describe animal, cellular, and synthetic models that have been developed to further visualize the BM. Following this section, we provide an overview of deep learning in retinal imaging and describe advances in computational and artificial intelligence (AI) techniques to provide automated segmentation of the BM and BM opening. We conclude this section considering the clinical implications of these segmentation techniques. Ultimately, the diverse advances aimed to further characterize the BM may allow for deeper insights into the involvement of this critical structure in retinal health and disease.
Collapse
Affiliation(s)
- Joshua Ong
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, United States
| | - Amrish Selvam
- Illinois Eye and Ear Infirmary, University of Illinois College of Medicine, Chicago, IL, United States
| | - Matthew Driban
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, United States
| | - Arman Zarnegar
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | | | - Jincy Joy
- Karunya Eye Hospital, Kottarakara, Kerala, India
| | - Ethan A Rossi
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | | | - José-Alain Sahel
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jay Chhablani
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.
| |
Collapse
|
4
|
Álvarez-Barrios A, Álvarez L, Sáenz de Santa María P, García M, Álvarez-Buylla JR, Pereiro R, González-Iglesias H. Dysregulated lipid metabolism in a retinal pigment epithelial cell model and serum of patients with age-related macular degeneration. BMC Biol 2025; 23:96. [PMID: 40221802 PMCID: PMC11993946 DOI: 10.1186/s12915-025-02198-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 03/26/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Age-related macular degeneration (AMD) is a leading cause of blindness, characterized by retinal pigment epithelium (RPE) dysfunction, extracellular deposit formation, and disrupted lipid metabolism. Understanding the molecular changes underlying AMD is essential for identifying diagnostic markers and therapeutic targets. RESULTS This multiomic study employed a primary RPE culture model to investigate age-related changes associated with AMD. Over 25 weeks, RPE cells exhibited phenotypic deterioration, including depigmentation, cell shape deformation, and barrier integrity loss, accompanied by extracellular deposit formation. Transcriptomic analysis revealed dysregulation of genes involved in lipid metabolism, including pathways for cholesterol transport, glycerophospholipids, and ceramide biosynthesis. Metabolomic profiling further identified significant changes in glycerophospholipid and sphingolipid metabolism, highlighting a decline in phospholipid species and ceramide accumulation. Serum analysis of AMD patients revealed altered levels of 18 lipids identified in RPE cultures. Four lipids showed significant differences compared to controls: GlcCer(d16:1/18:0) (1.23-fold increase, adj. p value < 0.001), PE(19:1(9Z)/22:2(13Z,16Z)) (0.34-fold decrease, adj. p value < 0.001), PE(15:0/20:3(5Z,8Z,11Z)) (0.66-fold decrease, adj. p value < 0.05), and PC(22:2(13Z,16Z)/13:0) (0.71-fold decrease, adj. p value < 0.05). These findings underscore the systemic nature of lipid dysregulation in AMD and the translational relevance of the RPE model. CONCLUSIONS This study highlights the significant role of lipid metabolism dysregulation in AMD pathogenesis. The consistent lipidomic alterations observed in RPE cultures and AMD patient serum reinforce their potential as biomarkers for disease progression and therapeutic targets. These findings provide a robust framework for understanding AMD-associated lipid metabolism changes and their systemic impact.
Collapse
Affiliation(s)
- Ana Álvarez-Barrios
- Fundación de Investigación Oftalmológica, Oviedo, Spain
- Department of Physical and Analytical Chemistry, University of Oviedo, Oviedo, Spain
| | - Lydia Álvarez
- Fundación de Investigación Oftalmológica, Oviedo, Spain.
| | - Pilar Sáenz de Santa María
- Fundación de Investigación Oftalmológica, Oviedo, Spain
- Instituto Oftalmológico Fernández-Vega, Oviedo, Spain
| | | | - Jorge R Álvarez-Buylla
- Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Oviedo, Spain
| | - Rosario Pereiro
- Department of Physical and Analytical Chemistry, University of Oviedo, Oviedo, Spain
| | - Héctor González-Iglesias
- Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Oviedo, Spain.
| |
Collapse
|
5
|
Yuan Y, Biswas P, Zemke NR, Dang K, Wu Y, D’Antonio M, Xie Y, Yang Q, Dong K, Lau PK, Li D, Seng C, Bartosik W, Buchanan J, Lin L, Lancione R, Wang K, Lee S, Gibbs Z, Ecker J, Frazer K, Wang T, Preissl S, Wang A, Ayyagari R, Ren B. Single-cell analysis of the epigenome and 3D chromatin architecture in the human retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.28.630634. [PMID: 39764062 PMCID: PMC11703273 DOI: 10.1101/2024.12.28.630634] [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: 01/11/2025]
Abstract
Most genetic risk variants linked to ocular diseases are non-protein coding and presumably contribute to disease through dysregulation of gene expression, however, deeper understanding of their mechanisms of action has been impeded by an incomplete annotation of the transcriptional regulatory elements across different retinal cell types. To address this knowledge gap, we carried out single-cell multiomics assays to investigate gene expression, chromatin accessibility, DNA methylome and 3D chromatin architecture in human retina, macula, and retinal pigment epithelium (RPE)/choroid. We identified 420,824 unique candidate regulatory elements and characterized their chromatin states in 23 sub-classes of retinal cells. Comparative analysis of chromatin landscapes between human and mouse retina cells further revealed both evolutionarily conserved and divergent retinal gene-regulatory programs. Leveraging the rapid advancements in deep-learning techniques, we developed sequence-based predictors to interpret non-coding risk variants of retina diseases. Our study establishes retina-wide, single-cell transcriptome, epigenome, and 3D genome atlases, and provides a resource for studying the gene regulatory programs of the human retina and relevant diseases.
Collapse
Affiliation(s)
- Ying Yuan
- Department of Material Science, UC San Diego, La Jolla, CA 92037, USA
| | - Pooja Biswas
- Ophthalmology, Shiley Eye Institute, UC San Diego, La Jolla, CA 92037, USA
| | - Nathan R. Zemke
- Center for Epigenomics, UC San Diego, La Jolla, CA 92037, USA
| | - Kelsey Dang
- Center for Epigenomics, UC San Diego, La Jolla, CA 92037, USA
| | - Yue Wu
- Department of Biological Science, UC San Diego, La Jolla, CA 92037, USA
| | - Matteo D’Antonio
- Department of Biomedical Informatics, UC San Diego, La Jolla, CA 92037, USA
| | - Yang Xie
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA 92037, USA
| | - Qian Yang
- Center for Epigenomics, UC San Diego, La Jolla, CA 92037, USA
| | - Keyi Dong
- Center for Epigenomics, UC San Diego, La Jolla, CA 92037, USA
| | - Pik Ki Lau
- Center for Epigenomics, UC San Diego, La Jolla, CA 92037, USA
| | - Daofeng Li
- Department of Genetics, Washington University School of Medicine in St.Louis, St. Louis, MO 63130, USA
| | - Chad Seng
- Department of Genetics, Washington University School of Medicine in St.Louis, St. Louis, MO 63130, USA
| | | | - Justin Buchanan
- Center for Epigenomics, UC San Diego, La Jolla, CA 92037, USA
| | - Lin Lin
- Center for Epigenomics, UC San Diego, La Jolla, CA 92037, USA
| | - Ryan Lancione
- Center for Epigenomics, UC San Diego, La Jolla, CA 92037, USA
| | - Kangli Wang
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA 92037, USA
| | - Seoyeon Lee
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA 92037, USA
| | - Zane Gibbs
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA 92037, USA
| | - Joseph Ecker
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA,USA
- Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Kelly Frazer
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
- Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Ting Wang
- Department of Genetics, Washington University School of Medicine in St.Louis, St. Louis, MO 63130, USA
| | | | - Allen Wang
- Center for Epigenomics, UC San Diego, La Jolla, CA 92037, USA
| | - Radha Ayyagari
- Ophthalmology, Shiley Eye Institute, UC San Diego, La Jolla, CA 92037, USA
| | - Bing Ren
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA 92037, USA
- Center for Epigenomics, UC San Diego, La Jolla, CA 92037, USA
| |
Collapse
|
6
|
Wei P, Gao S, Han G. Evidence for Genetic Causal Association Between the Gut Microbiome, Derived Metabolites, and Age-Related Macular Degeneration: A Mediation Mendelian Randomization Analysis. Biomedicines 2025; 13:639. [PMID: 40149615 PMCID: PMC11940807 DOI: 10.3390/biomedicines13030639] [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: 01/24/2025] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Despite substantial research, the causal relationships between gut microbiota (GM) and age-related macular degeneration (AMD) remain unclear. We aimed to explore these causal associations using Mendelian randomization (MR) and elucidate the potential mechanisms mediated by blood metabolites. Methods: We utilized the 211 GM dataset (n = 18,340) provided by the MiBioGen consortium. AMD outcome data were sourced from the MRC Integrated Epidemiology Unit (IEU) OpenGWAS Project. We performed bidirectional MR, two mediation analyses, and two-step MR to assess the causal links between GM and different stages of AMD (early, dry, and wet). Results: Our findings indicate that the Bacteroidales S24.7 group and genus Dorea are associated with an increased risk of early AMD, while Ruminococcaceae UCG011 and Parasutterella are linked to a higher risk of dry AMD. Conversely, Lachnospiraceae UCG004 and Anaerotruncus are protective against dry AMD. In the case of wet AMD, Intestinimonas and Sellimonas increase risk, whereas Anaerotruncus and Rikenellaceae RC9 reduce it. Additionally, various blood metabolites were implicated: valine, arabinose, creatine, lysine, alanine, and apolipoprotein A1 were associated with early AMD; glutamine and hyodeoxycholate-with a reduced risk of dry AMD; and androsterone sulfate, epiandrosterone sulfate, and lipopolysaccharide-with a reduced risk of wet AMD. Notably, the association between family Oxalobacteraceae and early AMD was mediated by valine, accounting for 19.1% of the association. Conclusions: This study establishes causal links between specific gut microbiota and AMD, mediated by blood metabolites, thereby enhancing our understanding of the gut-retina axis in AMD pathophysiology.
Collapse
Affiliation(s)
- Pinghui Wei
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin 300020, China; (P.W.); (S.G.)
- Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin 300071, China
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin 300020, China
| | - Shan Gao
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin 300020, China; (P.W.); (S.G.)
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Guoge Han
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin 300020, China; (P.W.); (S.G.)
- Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin 300071, China
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin 300020, China
| |
Collapse
|
7
|
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
|
8
|
Deng J, Qin Y. Investigating the Link between Psychological Well-Being and Early-Stage Age-Related Macular Degeneration: A Mendelian Randomization Analysis. Curr Eye Res 2025; 50:190-202. [PMID: 39329215 DOI: 10.1080/02713683.2024.2408757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/03/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024]
Abstract
PURPOSE While some studies have started to focus on the link between psychological well-being and age-related macular degeneration (AMD), the relationship remains uncertain. Our research aims to provide new insights into this association, laying a foundation for future interventions and addressing existing knowledge gaps. METHODS We utilized the "TwoSampleMR" package in R for a bidirectional Mendelian randomization analysis of psychological well-being (subjective well-being, depression, neuroticism, and Sensitivity to Environmental Stress and Adversity) and early-stage AMD. Causal effects were estimated using the inverse-variance weighted method, and additional methods included weighted median and MR-Egger regression. Sensitivity analyses included Cochran's Q test, MR-Egger intercept analysis, MR-PRESSO, and leave-one-out analysis. RESULTS The study found that the population with genetic predisposition to neuroticism had a 39.7% lower risk of early-stage AMD (OR = 0.603, 95% CI = 0.385-0.945, p = 0.027). Conversely, the population with genetic predisposition to subjective well-being had a 3.2% increased risk of early-stage AMD (OR = 1.032, 95% CI = 1.003-1.063, p = 0.029). No significant causal relationships were found from depression or Sensitivity to Environmental Stress and Adversity to early-stage AMD, nor from early-stage AMD to psychological well-being. CONCLUSION This study provides preliminary evidence that the relationship between psychological well-being and early-stage AMD may be complex and multifaceted. It suggests that moderate neuroticism levels might reduce early-stage AMD risk through health behaviors, pathophysiological mechanisms, and other factors, while high subjective well-being levels might increase this risk similarly. However, these findings are insufficient for preventive strategies due to a lack of substantial evidence and still require extensive experimental research for further validation.
Collapse
Affiliation(s)
- Jie Deng
- First Clinical College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - YuHui Qin
- First Clinical College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Graduate School, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
9
|
Garnsey MR, Wang Y, Edmonds DJ, Sammons MF, Reidich B, Ahn Y, Ashkenazi Y, Carlo A, Cerny MA, Coffman KJ, Culver JA, Dechert Schmitt AM, Eng H, Fisher EL, Gutierrez JA, James L, Jordan S, Kohrt JT, Kramer M, LaChapelle EA, Lee JC, Lee J, Li D, Li Z, Liu S, Liu J, Magee TV, Miller MR, Moran M, Nason DM, Nedoma NL, O'Neil SV, Piotrowski MA, Racich J, Sommese RF, Stevens LM, Wright AS, Xiao J, Zhang L, Zhou D, Barrandon O, Clasquin MF. Design and application of synthetic 17B-HSD13 substrates reveals preserved catalytic activity of protective human variants. Nat Commun 2025; 16:297. [PMID: 39746932 PMCID: PMC11697577 DOI: 10.1038/s41467-024-54487-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 11/13/2024] [Indexed: 01/04/2025] Open
Abstract
Several hydroxysteroid dehydrogenase 17-beta 13 variants have previously been identified as protective against metabolic dysfunction-associated steatohepatitis (MASH) fibrosis, ballooning and inflammation, and as such this target holds significant therapeutic potential. However, over 5 years later, the function of 17B-HSD13 remains unknown. Structure-aided design enables the development of potent and selective sulfonamide-based 17B-HSD13 inhibitors. In order to probe their inhibitory potency in endogenous expression systems like primary human hepatocytes, inhibitors are transformed into synthetic surrogate substrates with distinct selectivity advantages over substrates previously published. Their application to cells endogenously expressing 17B-HSD13 enables quantitative measures of enzymatic inhibition in primary human hepatocytes which has never been reported to date. Application to multiple cellular systems expressing the protective human variants reveals that the most prevalent IsoD variant maintains NAD-dependent catalytic activity towards some but not all substrates, contradicting reports that the truncation results in loss-of-function.
Collapse
Affiliation(s)
| | - Yang Wang
- Pfizer, Inc., Cambridge, MA, 02139, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Jisun Lee
- Pfizer, Inc., Groton, CT, 06340, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Jun Xiao
- Pfizer, Inc., Groton, CT, 06340, USA
| | | | | | | | | |
Collapse
|
10
|
Garzone D, Imtiaz MA, Mauschitz MM, Aziz NA, Holz FG, Breteler MMB, Finger RP. Age-Related Macular Degeneration and Its Genetic Risk: A Population-based Study. Curr Eye Res 2025; 50:82-86. [PMID: 39155542 DOI: 10.1080/02713683.2024.2388692] [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: 04/08/2024] [Revised: 07/11/2024] [Accepted: 07/31/2024] [Indexed: 08/20/2024]
Abstract
PURPOSE Specific genetic factors might serve as markers for risk stratification of AMD progression, but their association with key features of AMD has not been fully elucidated. Thus, we investigated the association between overall and pathway-specific genetic risk scores (GRS) and lead loci (ARMS2, CFH) with AMD stages and features of high-risk nonlate AMD, including reticular pseudodrusen (RPD) and large drusen area (LDA). METHODS We performed a cross-sectional analysis of data from the Rhineland Study, a population-based study in Bonn, Germany. We included 4016 individuals aged 50 years and older of European descent. GRS and pathway-specific subscores were constructed based on a large genome-wide association study of AMD. Subscores were generated based on gene-pathways associations (complement, extracellular matrix remodeling (ECM) and lipid metabolism). Associations were assessed using logistic and multinomial regression. RESULTS The mean age of participants was 63.36 years and 1813 (45.1%) were men. The GRS was positive in 48.1% of individuals and increased, but did not fully overlap, across AMD stages. Pathway-specific subscores increased across AMD stages except for the ECM subscore, which only showed a trend for increasing in late AMD. Increasing overall GRS was associated with RPD and LDA (OR [95%CI] for RPD: 1.70 [1.33-2.15], for LDA: 1.64 [1.29-2.07]) among individuals with AMD. Similarly, higher complement and ECM subscores was associated with RPD, while for LDA, only an association with complement subscore was observed. CONCLUSIONS In a population-based setting, we confirmed higher genetic risk to be associated with more severe AMD and identified associations with high-risk features of intermediate AMD. Conjoint analyses suggested that high-risk features and late AMD might be differentially associated with genetic architecture in AMD, such as ECM remodeling. Incorporation of genetic information such as GRSs might improve AMD risk prediction strategies.
Collapse
Affiliation(s)
- Davide Garzone
- Department of Ophthalmology, University Hospital Bonn, Bonn, Germany
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Mohammed Aslam Imtiaz
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Matthias M Mauschitz
- Department of Ophthalmology, University Hospital Bonn, Bonn, Germany
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - N Ahmad Aziz
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Frank G Holz
- Department of Ophthalmology, University Hospital Bonn, Bonn, Germany
| | - Monique M B Breteler
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Institute for Medical Biometry, Informatics and Epidemiology (IMBIE), Faculty of Medicine, University of Bonn, Germany
| | - Robert P Finger
- Department of Ophthalmology, University Hospital Bonn, Bonn, Germany
- Department of Ophthalmology, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
11
|
Ellervik C, Boulakh L, Teumer A, Marouli E, Kuś A, Buch Hesgaard H, Heegaard S, Blankers L, Sterenborg R, Åsvold BO, Winkler TW, Medici M, Kjaergaard AD. Thyroid Function, Diabetes, and Common Age-Related Eye Diseases: A Mendelian Randomization Study. Thyroid 2024; 34:1414-1423. [PMID: 39283829 PMCID: PMC11958925 DOI: 10.1089/thy.2024.0257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Background: Previous Mendelian randomization (MR) studies showed an association between hypothyroidism and cataract and between high-normal free thyroxine (FT4) and late age-related macular degeneration (AMD), but not between FT4, thyroid stimulating hormone (TSH), or hyperthyroidism and diabetic retinopathy or cataract. These studies included a limited number of genetic variants for thyroid function and did not investigate autoimmune thyroid disease (AITD) or glaucoma, include bidirectional and multivariable MR (MVMR), and examine sex differences or potential mediation effects of diabetes. We aimed to address this knowledge gap. Methods: We examined the causality and directionality of the associations of AITD, and FT4 and TSH within the reference range with common age-related eye diseases (diabetic retinopathy, cataract, early and late AMD, and primary open-angle glaucoma). We conducted a bidirectional two-sample MR study utilizing publicly available genome-wide association study (GWAS) summary statistics from international consortia (ThyroidOmics, International AMD Genetics Consortium, deCODE, UK Biobank, FinnGen, and DIAGRAM). Bidirectional MR tested directionality, whereas MVMR estimated independent causal effects. Furthermore, we investigated type 1 diabetes (T1D) and type 2 diabetes (T2D) as potential mediators. Results: Genetic predisposition to AITD was associated with increased risk of diabetic retinopathy (p = 3 × 10-4), cataract (p = 3 × 10-3), and T1D (p = 1 × 10-3), but less likely T2D (p = 0.01). MVMR showed attenuated estimates for diabetic retinopathy and cataract when adjusting for T1D, but not T2D. We found pairwise bidirectional associations between AITD, T1D, and diabetic retinopathy. Genetic predisposition to both T1D and T2D increased the risk of diabetic retinopathy and cataract (p < 4 × 10-4). Moreover, genetically predicted higher FT4 within the reference range was associated with an increased risk of late AMD (p = 0.01), particularly in women (p = 7 × 10-3). However, we neither found any association between FT4 and early AMD nor between TSH and early and late AMD. No other associations were observed. Conclusions: Genetic predisposition to AITD is associated with risk of diabetic retinopathy and cataract, mostly mediated through increased T1D risk. Reciprocal associations between AITD, diabetic retinopathy, and T1D imply a shared autoimmune origin. The role of FT4 in AMD and potential sex discrepancies needs further investigation.
Collapse
Affiliation(s)
- Christina Ellervik
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Pathology, Harvard Medical School, Boston, MA, USA
- Department of Laboratory Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Clinical Biochemistry, Zealand University Hospital, Køge, Denmark
| | - Lena Boulakh
- Department of Ophthalmology, Rigshospitalet-Glostrup, Glostrup, Denmark
| | - Alexander Teumer
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Eirini Marouli
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Aleksander Kuś
- Department of Internal Medicine and Endocrinology, Medical University of Warsaw, Warsaw, Poland
| | - Helena Buch Hesgaard
- Institute of Neuroscience and Physiology, Gothenburg University, Sweden
- Department of Ophthalmology, Sahlgrenska University Hospital, Sweden
| | - Steffen Heegaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Ophthalmology, Rigshospitalet-Glostrup, Glostrup, Denmark
- Department of Pathology, Rigshospitalet-Glostrup, Glostrup, Denmark
| | - Lizette Blankers
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Rosalie Sterenborg
- Department of Internal Medicine, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Bjørn Olav Åsvold
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | | | - Marco Medici
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - The International AMD Genomics Consortium, and The Mendelian Randomization Study Group for the ThyroidOmics Consortium
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Pathology, Harvard Medical School, Boston, MA, USA
- Department of Laboratory Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Clinical Biochemistry, Zealand University Hospital, Køge, Denmark
- Department of Ophthalmology, Rigshospitalet-Glostrup, Glostrup, Denmark
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
- Department of Internal Medicine and Endocrinology, Medical University of Warsaw, Warsaw, Poland
- Institute of Neuroscience and Physiology, Gothenburg University, Sweden
- Department of Ophthalmology, Sahlgrenska University Hospital, Sweden
- Department of Pathology, Rigshospitalet-Glostrup, Glostrup, Denmark
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
12
|
Spaide RF. PATHWAYS TO GEOGRAPHIC ATROPHY IN NONNEOVASCULAR AGE-RELATED MACULAR DEGENERATION. Retina 2024; 44:1655-1665. [PMID: 39121492 DOI: 10.1097/iae.0000000000004242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2024]
Abstract
PURPOSE To characterize and quantify the precursor lesions of geographic atrophy in eyes with age-related macular degeneration. METHODS A retrospective study of eyes with a minimum of 6-month follow-up before developing geographic atrophy. Evaluations included color and autofluorescence imaging, along with spectral-domain optical coherence tomography, employing definitions from the Consensus of Atrophy Meeting Group and Consensus on Neovascular Age-Related Macular Degeneration Nomenclature Study Group. RESULTS There were 55 eyes of 44 patients, who had a mean age of 81.3 years at onset of atrophy; 35 (63.6%) were female. The mean duration of follow-up before and after the advent of geographic atrophy was 4.9 years and 1.2 years, respectively. Geographic atrophy was preceded by collapse of a druse in 41 eyes (74.5%). Of these, 29 (70.7%) were drusenoid pigment epithelial detachments. Among the eyes with regressing drusen, there were 9 with overlying vitelliform deposit, and all had concurrent subretinal drusenoid deposit; however, 19 of 30 eyes with no vitelliform deposit overlying the druse had concurrent subretinal drusenoid deposit, a difference that was significant ( P < 0.001). Regression of subretinal drusenoid deposit was found in 4 eyes (7.3%), regression of vitelliform deposit associated with subretinal drusenoid deposit in 5 (9.1%), and regression of vitelliform deposit in eyes concurrently harboring drusen was found in 3 (5.4%) and regression of vitelliform deposit alone in 2 (3.6%) at the site of eventual development of geographic atrophy. CONCLUSION Geographic atrophy appears to develop from multiple pathways as manifested by the many precursor lesions, all various forms of extracellular deposit, that upon regression, result in a common end-stage appearance.
Collapse
Affiliation(s)
- Richard F Spaide
- Vitreous, Retina, Macula Consultants of New York, New York, New York
| |
Collapse
|
13
|
Tang S, Yang J, Xiao B, Wang Y, Lei Y, Lai D, Qiu Q. Aberrant Lipid Metabolism and Complement Activation in Age-Related Macular Degeneration. Invest Ophthalmol Vis Sci 2024; 65:20. [PMID: 39405051 PMCID: PMC11482642 DOI: 10.1167/iovs.65.12.20] [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: 06/25/2024] [Accepted: 09/10/2024] [Indexed: 10/19/2024] Open
Abstract
Age-related macular degeneration (AMD) stands as a leading cause of severe visual impairment and blindness among the elderly globally. As a multifactorial disease, AMD's pathogenesis is influenced by genetic, environmental, and age-related factors, with lipid metabolism abnormalities and complement system dysregulation playing critical roles. This review delves into recent advancements in understanding the intricate interaction between these two crucial pathways, highlighting their contribution to the disease's progression through chronic inflammation, drusen formation, and retinal pigment epithelium dysfunction. Importantly, emerging evidence points to dysregulated lipid profiles, particularly alterations in high-density lipoprotein levels, oxidized lipid deposits, and intracellular lipofuscin accumulation, as exacerbating factors that enhance complement activation and subsequently amplify tissue damage in AMD. Furthermore, genetic studies have revealed significant associations between AMD and specific genes involved in lipid transport and complement regulation, shedding light on disease susceptibility and underlying mechanisms. The review further explores the clinical implications of these findings, advocating for a novel therapeutic approach that integrates lipid metabolism modulators with complement inhibitors. By concurrently targeting these pathways, the dual-targeted approach holds promise in significantly improving outcomes for AMD patients, heralding a new horizon in AMD management and treatment.
Collapse
Affiliation(s)
- Siao Tang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, PR China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Jiaqi Yang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, PR China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Bingqing Xiao
- Xiangya School of Medicine, Central South University, Changsha, Hunan, PR China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Yani Wang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, PR China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Yiou Lei
- Xiangya School of Medicine, Central South University, Changsha, Hunan, PR China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Dongwei Lai
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Qinghua Qiu
- Department of Ophthalmology, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| |
Collapse
|
14
|
Lin J, Lin L. Genetic liability to higher frailty index may increase the risk of ophthalmic disease. Int Ophthalmol 2024; 44:397. [PMID: 39347840 DOI: 10.1007/s10792-024-03319-y] [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: 04/19/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
PURPOSE Frailty and age-related eye diseases are common in older people; however, whether there is a causal link remains unknown. We aimed to explore the causal associations between the frailty index (FI) and ophthalmic traits and identify modifiable mediators. METHODS Linkage disequilibrium score regression and two-sample Mendelian randomization were applied to identify genetic correlations and causal associations between FI and ophthalmic traits. Summary data for FI was obtained from a genome-wide association study that included 175,226 individuals of European ancestry. Summary-level statistics for ophthalmic traits were obtained from relative GWASs. Summary-level data for cardiovascular risk factors, inflammatory biomarkers, and the central nervous system were used to identify the possible mediators. RESULTS FI had a significant genetic correlation with 10 ophthalmic traits. Per SD increment of FI, the odds ratio was 1.329 (95% CI, 1.123, 1.573; P = 9.5 × 10-4) for cataracts, 1.825 (95% CI, 1.115, 2.986; P = 0.016) for keratitis, 1.798 (95% CI, 1.039, 3.11; P = 0.036) for disorders of vitreous body and 1.478 (95% CI, 1.005, 2.173; P = 0.046) for disorders of sclera, cornea, iris and ciliary body. The MR effect estimates of FI on ophthalmic traits were attenuated after adjusting for mental disorders, type 2 diabetes, triglyceride, and interleukin-8 (IL-8) levels. CONCLUSION This study reports a genetic correlation and causal association between FI and ophthalmic traits, in which mental disorders, type 2 diabetes, triglycerides, and IL-8 may play a mediating role. These findings highlight a possible method to reduce the risk of FI-related ophthalmic diseases.
Collapse
Affiliation(s)
- Jianwei Lin
- Big Data Laboratory, Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Liling Lin
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
15
|
Lin J, Lin L. Genetic liability to higher frailty index may increase the risk of ophthalmic disease. Int Ophthalmol 2024; 44:397. [PMID: 39347840 DOI: 10.1007/s10792-024-03319-y.pmid:] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
PURPOSE Frailty and age-related eye diseases are common in older people; however, whether there is a causal link remains unknown. We aimed to explore the causal associations between the frailty index (FI) and ophthalmic traits and identify modifiable mediators. METHODS Linkage disequilibrium score regression and two-sample Mendelian randomization were applied to identify genetic correlations and causal associations between FI and ophthalmic traits. Summary data for FI was obtained from a genome-wide association study that included 175,226 individuals of European ancestry. Summary-level statistics for ophthalmic traits were obtained from relative GWASs. Summary-level data for cardiovascular risk factors, inflammatory biomarkers, and the central nervous system were used to identify the possible mediators. RESULTS FI had a significant genetic correlation with 10 ophthalmic traits. Per SD increment of FI, the odds ratio was 1.329 (95% CI, 1.123, 1.573; P = 9.5 × 10-4) for cataracts, 1.825 (95% CI, 1.115, 2.986; P = 0.016) for keratitis, 1.798 (95% CI, 1.039, 3.11; P = 0.036) for disorders of vitreous body and 1.478 (95% CI, 1.005, 2.173; P = 0.046) for disorders of sclera, cornea, iris and ciliary body. The MR effect estimates of FI on ophthalmic traits were attenuated after adjusting for mental disorders, type 2 diabetes, triglyceride, and interleukin-8 (IL-8) levels. CONCLUSION This study reports a genetic correlation and causal association between FI and ophthalmic traits, in which mental disorders, type 2 diabetes, triglycerides, and IL-8 may play a mediating role. These findings highlight a possible method to reduce the risk of FI-related ophthalmic diseases.
Collapse
Affiliation(s)
- Jianwei Lin
- Big Data Laboratory, Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Liling Lin
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
16
|
Dascalu AM, Grigorescu CC, Serban D, Tudor C, Alexandrescu C, Stana D, Jurja S, Costea AC, Alius C, Tribus LC, Dumitrescu D, Bratu D, Cristea BM. Complement Inhibitors for Geographic Atrophy in Age-Related Macular Degeneration-A Systematic Review. J Pers Med 2024; 14:990. [PMID: 39338244 PMCID: PMC11432754 DOI: 10.3390/jpm14090990] [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: 07/25/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND/OBJECTIVES Age-related macular degeneration (AMD) is one of the main causes of blindness and visual impairment worldwide. Intravitreal complement inhibitors are an emergent approach in the treatment of AMD, which have had encouraging results. This systematic review analyzes the outcomes and safety of complement inhibitor therapies for GA in AMD cases. METHODS A comprehensive search on the PubMed and Web of Science databases returned 18 studies involving various complement inhibitor agents, with a total of 4272 patients and a mean follow-up of 68.2 ± 20.4 weeks. RESULTS Most treated patients were white (96.8%) and female (55.8%), with a mean age of 78.3 ± 7.8 years and a mean GA area of 8.0 ± 3.9 mm2. There were no differences in visual function change between treated and control participants. The mean GA area change was 2.4 ± 0.7 mm2 in treated participants vs. 2.7 ± 0.8 mm2 in control groups (p < 0.001). The ocular and systemic side effects were similar to those of intravitreal anti-VEGF. A less-understood effect was that of the onset of choroidal neovascularization (CNV) in 1.1-13% of patients; this effect was found to be more frequent in patients with neovascular AMD in the fellow eye or nonexudative CNV in the study eye at baseline. CONCLUSIONS Complement inhibitors may represent a useful therapy for GA in AMD, but a personalized approach to patient selection is necessary to optimize the outcomes.
Collapse
Affiliation(s)
- Ana Maria Dascalu
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Ophthalmology Department, Emergency University Hospital Bucharest, 050098 Bucharest, Romania
| | | | - Dragos Serban
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Fourth Surgery Department, Emergency University Hospital Bucharest, 050098 Bucharest, Romania
| | - Corneliu Tudor
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Fourth Surgery Department, Emergency University Hospital Bucharest, 050098 Bucharest, Romania
| | - Cristina Alexandrescu
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Ophthalmology Department, Emergency University Hospital Bucharest, 050098 Bucharest, Romania
| | - Daniela Stana
- Ophthalmology Department, Emergency University Hospital Bucharest, 050098 Bucharest, Romania
| | - Sanda Jurja
- Faculty of Medicine, Ovidius University, 900470 Constanta, Romania
- Ophthalmology Department, Emergency County Hospital Constanta, 900591 Constanta, Romania
| | | | - Catalin Alius
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Fourth Surgery Department, Emergency University Hospital Bucharest, 050098 Bucharest, Romania
| | - Laura Carina Tribus
- Faculty of Dental Medicine, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Internal Medicine, Ilfov Emergency Clinic Hospital, 022113 Bucharest, Romania
| | - Dan Dumitrescu
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Fourth Surgery Department, Emergency University Hospital Bucharest, 050098 Bucharest, Romania
| | - Dan Bratu
- Faculty of Medicine, University "Lucian Blaga", 550169 Sibiu, Romania
- Department of Surgery, Emergency County Hospital Sibiu, 550245 Sibiu, Romania
| | - Bogdan Mihai Cristea
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
17
|
Cebatoriene D, Vilkeviciute A, Gedvilaite-Vaicechauskiene G, Duseikaite M, Bruzaite A, Kriauciuniene L, Zaliuniene D, Liutkeviciene R. The Impact of ARMS2 (rs10490924), VEGFA (rs3024997), TNFRSF1B (rs1061622), TNFRSF1A (rs4149576), and IL1B1 (rs1143623) Polymorphisms and Serum Levels on Age-Related Macular Degeneration Development and Therapeutic Responses. Int J Mol Sci 2024; 25:9750. [PMID: 39273697 PMCID: PMC11396313 DOI: 10.3390/ijms25179750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
Age-related macular degeneration (AMD) is a major global health problem as it is the leading cause of irreversible loss of central vision in the aging population. Anti-vascular endothelial growth factor (anti-VEGF) therapies are effective but do not respond optimally in all patients. This study investigates the genetic factors associated with susceptibility to AMD and response to treatment, focusing on key polymorphisms in the ARMS2 (rs10490924), IL1B1 (rs1143623), TNFRSF1B (rs1061622), TNFRSF1A (rs4149576), VEGFA (rs3024997), ARMS2, IL1B1, TNFRSF1B, TNFRSF1A, and VEGFA serum levels in AMD development and treatment efficacy. This study examined the associations of specific genetic polymorphisms and serum protein levels with exudative and early AMD and the response to anti-VEGF treatment. The AA genotype of VEGFA (rs3024997) was significantly associated with a 20-fold reduction in the odds of exudative AMD compared to the GG + GA genotypes. Conversely, the TT genotype of ARMS2 (rs10490924) was linked to a 4.2-fold increase in the odds of exudative AMD compared to GG + GT genotypes. In females, each T allele of ARMS2 increased the odds by 2.3-fold, while in males, the TT genotype was associated with a 5-fold increase. Lower serum IL1B levels were observed in the exudative AMD group compared to the controls. Early AMD patients had higher serum TNFRSF1B levels than controls, particularly those with the GG genotype of TNFRSF1B rs1061622. Exudative AMD patients with the CC genotype of TNFRSF1A rs4149576 had lower serum TNFRSF1A levels compared to the controls. Visual acuity (VA) analysis showed that non-responders had better baseline VA than responders but experienced decreased VA after treatment, whereas responders showed improvement. Central retinal thickness (CRT) reduced significantly in responders after treatment and was lower in responders compared to non-responders after treatment. The T allele of TNFRSF1B rs1061622 was associated with a better response to anti-VEGF treatment under both dominant and additive genetic models. These findings highlight significant genetic and biochemical markers associated with AMD and treatment response. This study found that the VEGFA rs3024997 AA genotype reduces the odds of exudative AMD, while the ARMS2 rs10490924 TT genotype increases it. Lower serum IL1B levels and variations in TNFRSF1B and TNFRSF1A levels were linked to AMD. The TNFRSF1B rs1061622 T allele was associated with better anti-VEGF treatment response. These markers could potentially guide risk assessment and personalized treatment for AMD.
Collapse
Affiliation(s)
- Dzastina Cebatoriene
- Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus St. 9, LT-44307 Kaunas, Lithuania
| | - Alvita Vilkeviciute
- Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu St. 2, LT-50161 Kaunas, Lithuania
| | | | - Monika Duseikaite
- Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu St. 2, LT-50161 Kaunas, Lithuania
| | - Akvile Bruzaite
- Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu St. 2, LT-50161 Kaunas, Lithuania
| | - Loresa Kriauciuniene
- Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu St. 2, LT-50161 Kaunas, Lithuania
- Department of Ophthalmology, Medical Academy, Lithuanian University of Health Sciences, Eiveniu St. 2, LT-50161 Kaunas, Lithuania
| | - Dalia Zaliuniene
- Department of Ophthalmology, Medical Academy, Lithuanian University of Health Sciences, Eiveniu St. 2, LT-50161 Kaunas, Lithuania
| | - Rasa Liutkeviciene
- Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu St. 2, LT-50161 Kaunas, Lithuania
- Department of Ophthalmology, Medical Academy, Lithuanian University of Health Sciences, Eiveniu St. 2, LT-50161 Kaunas, Lithuania
| |
Collapse
|
18
|
Kang H, Park SK, Kim DH, Choi YH. Exposure to per- and polyfluoroalkyl substances and age-related macular degeneration in U.S. middle-aged and older adults. CHEMOSPHERE 2024; 364:143167. [PMID: 39181460 DOI: 10.1016/j.chemosphere.2024.143167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 07/24/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Despite various health effects of per- and polyfluoroalkyl substances (PFAS) exposure, the association between PFAS exposure and age-related macular degeneration (AMD) has not been investigated. We aimed to assess associations of PFAS exposure with AMD, using data from 1722 U.S. adults aged 40 years or more participating in the National Health and Nutrition Examination Survey 2005-2008 with complete data on PFAS measurement, AMD diagnosis, and covariates. Serum concentrations of PFAS, including perfluorooctanoic acid (PFOA), perfluorononanoic acid (PFNA), perfluorohexane sulfonic acid (PFHxS), and perfluorooctane sulfonic acid (PFOS), were measured. An overall PFAS burden score was calculated using item response theory scoring. Individual PFAS concentration and overall PFAS burden score were categorized into low (reference), medium, and high groups. Diagnosis of AMD was based on retinal image examination. Any AMD was defined as the presence of early or late AMD. Survey-weighted logistic regression adjusted for potential confounders was used to calculate odds ratios (ORs) and 95% confidence intervals (CIs) for presence of AMD according to PFAS exposure. Overall, 132 (6.5%) individuals were diagnosed as any AMD, including 115 (5.7%) individuals with early AMD. A significant dose-response association was observed between serum PFOS concentration and any AMD (p-trend = 0.03), with a significant OR of 1.99 (95% CI: 1.05, 3.79) for the high group compared to the reference. Overall PFAS burden showed a non-monotonic association with any AMD, with a significant OR of 2.18 (95% CI: 1.18, 4.04) for the medium. Inverted U-shaped associations were observed by restricted cubic spline analyses. Also, early AMD showed similar patterns in PFOS and overall PFAS burden and additionally an inverted U-shape association in PFNA. Our findings suggest that exposure to PFAS estimated by serum PFOS and PFNA as well as overall PFAS burden might be a risk factor for AMD in middle-aged and older population.
Collapse
Affiliation(s)
- Habyeong Kang
- Institute of Health Sciences, Korea University, Seoul, South Korea; School of Health and Environmental Science, Korea University, Seoul, South Korea; Department of Preventive Medicine, College of Medicine, Hanyang University, Seoul, South Korea
| | - Sung Kyun Park
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Dong Hyun Kim
- Department of Ophthalmology, Korea University College of Medicine, Seoul, South Korea.
| | - Yoon-Hyeong Choi
- Institute of Health Sciences, Korea University, Seoul, South Korea; School of Health and Environmental Science, Korea University, Seoul, South Korea.
| |
Collapse
|
19
|
Honda S, Misawa N, Sato Y, Oikawa D, Tokunaga F. The hypothetical molecular mechanism of the ethnic variations in the manifestation of age-related macular degeneration; focuses on the functions of the most significant susceptibility genes. Graefes Arch Clin Exp Ophthalmol 2024; 262:2799-2811. [PMID: 38507046 DOI: 10.1007/s00417-024-06442-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 02/27/2024] [Accepted: 03/08/2024] [Indexed: 03/22/2024] Open
Abstract
Age-related macular degeneration (AMD) is the leading sight-threatening disease in developed countries. On the other hand, recent studies indicated an ethnic variation in the phenotype of AMD. For example, several reports demonstrated that the incidence of drusen in AMD patients is less in Asians compared to Caucasians though the reason has not been clarified yet. In the last decades, several genome association studies have disclosed many susceptible genes of AMD and revealed that the association strength of some genes was different among races and AMD phenotypes. In this review article, the essential findings of the clinical studies and genome association studies for the most significant genes CFH and ARMS2/HTRA1 in AMD of different races are summarized, and theoretical hypotheses about the molecular mechanisms underlying the ethnic variation in the AMD manifestation mainly focused on those genes between Caucasians and Asians are discussed.
Collapse
Affiliation(s)
- Shigeru Honda
- Department of Ophthalmology and Visual Sciences, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-Ku, Osaka, Japan.
| | - Norihiko Misawa
- Department of Ophthalmology and Visual Sciences, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-Ku, Osaka, Japan
| | - Yusuke Sato
- Center for Research On Green Sustainable Chemistry, Graduate School of Engineering, Tottori University, Tottori, Japan
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori, Japan
| | - Daisuke Oikawa
- Department of Medical Biochemistry, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Fuminori Tokunaga
- Department of Medical Biochemistry, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
20
|
DiCesare SM, Ortega AJ, Collier GE, Daniel S, Thompson KN, McCoy MK, Posner BA, Hulleman JD. GSK3 inhibition reduces ECM production and prevents age-related macular degeneration-like pathology. JCI Insight 2024; 9:e178050. [PMID: 39114980 PMCID: PMC11383595 DOI: 10.1172/jci.insight.178050] [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: 11/30/2023] [Accepted: 06/20/2024] [Indexed: 08/22/2024] Open
Abstract
Malattia Leventinese/Doyne honeycomb retinal dystrophy (ML/DHRD) is an age-related macular degeneration-like (AMD-like) retinal dystrophy caused by an autosomal dominant R345W mutation in the secreted glycoprotein, fibulin-3 (F3). To identify new small molecules that reduce F3 production in retinal pigmented epithelium (RPE) cells, we knocked-in a luminescent peptide tag (HiBiT) into the endogenous F3 locus that enabled simple, sensitive, and high-throughput detection of the protein. The GSK3 inhibitor, CHIR99021 (CHIR), significantly reduced F3 burden (expression, secretion, and intracellular levels) in immortalized RPE and non-RPE cells. Low-level, long-term CHIR treatment promoted remodeling of the RPE extracellular matrix, reducing sub-RPE deposit-associated proteins (e.g., amelotin, complement component 3, collagen IV, and fibronectin), while increasing RPE differentiation factors (e.g., tyrosinase, and pigment epithelium-derived factor). In vivo, treatment of 8-month-old R345W+/+ knockin mice with CHIR (25 mg/kg i.p., 1 mo) was well tolerated and significantly reduced R345W F3-associated AMD-like basal laminar deposit number and size, thereby preventing the main pathological feature in these mice. This is an important demonstration of small molecule-based prevention of AMD-like pathology in ML/DHRD mice and may herald a rejuvenation of interest in GSK3 inhibition for the treatment of retinal degenerative diseases, including potentially AMD itself.
Collapse
Affiliation(s)
- Sophia M. DiCesare
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Antonio J. Ortega
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Gracen E. Collier
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Steffi Daniel
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Krista N. Thompson
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Melissa K. McCoy
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Bruce A. Posner
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - John D. Hulleman
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
21
|
Ye S, Ma S, Liu S, Huang Y, Li D, Li M, Su T, Luo J, Zhang C, Shi D, Hu L, Zhang L, Yu H, He M, Shang X, Zhang X. Shared whole environmental etiology between Alzheimer's disease and age-related macular degeneration. NPJ AGING 2024; 10:36. [PMID: 39103390 DOI: 10.1038/s41514-024-00162-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 07/22/2024] [Indexed: 08/07/2024]
Abstract
The comorbidity of Alzheimer's disease (AD) and age-related macular degeneration (AMD) has been established in clinical and genetic studies. There is growing interest in determining the shared environmental factors associated with both conditions. Recent advancements in record linkage techniques enable us to identify the contributing factors to AD and AMD from a wide range of variables. As such, we first constructed a knowledge graph based on the literature, which included all statistically significant risk factors for AD and AMD. An environment-wide association study (EWAS) was conducted to assess the contribution of various environmental factors to the comorbidity of AD and AMD based on the UK biobank. Based on the conditional Q-Q plots and Bayesian algorithm, several shared environmental factors were identified, which could be categorized into the domains of health condition, biological sample parameters, body index, and attendance availability. Finally, we generated a shared etiology landscape for AD and AMD by combining existing knowledge with our novel findings.
Collapse
Affiliation(s)
- Siting Ye
- Department of Ultrasound, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Orthopaedics, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuo Ma
- Clinical Data Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Ethicon Minimally Invasive Procedures and Advanced Energy, Johnson & Johnson Medical (Shanghai) Device Company, Shanghai, China
| | - Shunming Liu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
| | - Yu Huang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
| | - Dantong Li
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
| | - Min Li
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
| | - Ting Su
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
| | - Jing Luo
- Medical Big Data Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Chi Zhang
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Danli Shi
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China
| | - Lianting Hu
- Medical Big Data Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Lei Zhang
- China-Australia Joint Research Center for Infectious Diseases, School of Public Health, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China
- Central Clinical School, Faculty of Medicine, Monash University, 3800, Melbourne, Australia
| | - Honghua Yu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
| | - Mingguang He
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China.
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Xianwen Shang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China.
| | - Xueli Zhang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China.
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, 510080, Guangzhou, China.
| |
Collapse
|
22
|
Terao R, Sohn BS, Yamamoto T, Lee TJ, Colasanti J, Pfeifer CW, Lin JB, Santeford A, Yamaguchi S, Yoshida M, Apte RS. Cholesterol Accumulation Promotes Photoreceptor Senescence and Retinal Degeneration. Invest Ophthalmol Vis Sci 2024; 65:29. [PMID: 39167399 PMCID: PMC11343002 DOI: 10.1167/iovs.65.10.29] [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: 04/28/2024] [Accepted: 08/01/2024] [Indexed: 08/23/2024] Open
Abstract
Purpose Dysregulated cholesterol metabolism is critical in the pathogenesis of AMD. Cellular senescence contributes to the development of numerous age-associated diseases. In this study, we investigated the link between cholesterol burden and the cellular senescence of photoreceptors. Methods Retinas from rod-specific ATP binding cassette subfamily A member 1 (Abca1) and G member 1 (Abcg1) (Abca1/g1-rod/-rod) knockout mice fed with a high-fat diet were analyzed for the signs of cellular senescence. Real-time quantitative PCR and immunofluorescence were used to characterize the senescence profile of the retina and cholesterol-treated photoreceptor cell line (661W). Inducible elimination of p16(Ink4a)-positive senescent cells (INK-ATTAC) mice or the administration of senolytic drugs (dasatinib and quercetin: D&Q) were used to examine the impact of senolytics on AMD-like phenotypes in Abca1/g1-rod/-rod retina. Results Increased accumulation of senescent cells as measured by markers of cellular senescence was found in Abca1/g1-rod/-rod retina. Exogenous cholesterol also induced cellular senescence in 661W cells. Selective elimination of senescent cells in Abca1/g1-rod/-rod;INK-ATTAC mice or by administration of D&Q improved visual function, lipid accumulation in retinal pigment epithelium, and Bruch's membrane thickening. Conclusions Cholesterol accumulation promotes cellular senescence in photoreceptors. Eliminating senescent photoreceptors improves visual function in a model of retinal neurodegeneration, and senotherapy offers a novel therapeutic avenue for further investigation.
Collapse
Affiliation(s)
- Ryo Terao
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Ophthalmology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Brian S. Sohn
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Taku Yamamoto
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Tae Jun Lee
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Jason Colasanti
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Charles W. Pfeifer
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Joseph B. Lin
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Andrea Santeford
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Shinobu Yamaguchi
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Mitsukuni Yoshida
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Rajendra S. Apte
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States
| |
Collapse
|
23
|
Lee PC, Jung IH, Thussu S, Patel V, Wagoner R, Burks KH, Amrute J, Elenbaas JS, Kang CJ, Young EP, Scherer PE, Stitziel NO. Instrumental variable and colocalization analyses identify endotrophin and HTRA1 as potential therapeutic targets for coronary artery disease. iScience 2024; 27:110104. [PMID: 38989470 PMCID: PMC11233907 DOI: 10.1016/j.isci.2024.110104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/26/2024] [Accepted: 05/22/2024] [Indexed: 07/12/2024] Open
Abstract
Coronary artery disease (CAD) remains a leading cause of disease burden globally, and there is a persistent need for new therapeutic targets. Instrumental variable (IV) and genetic colocalization analyses can help identify novel therapeutic targets for human disease by nominating causal genes in genome-wide association study (GWAS) loci. We conducted cis-IV analyses for 20,125 genes and 1,746 plasma proteins with CAD using molecular trait quantitative trait loci variant (QTLs) data from three different studies. 19 proteins and 119 genes were significantly associated with CAD risk by IV analyses and demonstrated evidence of genetic colocalization. Notably, our analyses validated well-established targets such as PCSK9 and ANGPTL4 while also identifying HTRA1 and endotrophin (a cleavage product of COL6A3) as proteins whose levels are causally associated with CAD risk. Further experimental studies are needed to confirm the causal role of the genes and proteins identified through our multiomic cis-IV analyses on human disease.
Collapse
Affiliation(s)
- Paul C. Lee
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - In-Hyuk Jung
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Shreeya Thussu
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Ved Patel
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Ryan Wagoner
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Kendall H. Burks
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Junedh Amrute
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Jared S. Elenbaas
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Chul Joo Kang
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO 63108, USA
| | - Erica P. Young
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO 63108, USA
| | - Philipp E. Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nathan O. Stitziel
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO 63108, USA
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO 63110, USA
| |
Collapse
|
24
|
Guymer RH, Silva R, Ghadessi M, Leal S, Gashaw I, Damask A, Paulding C, Rittenhouse KD. ANO2 Genetic Variants and Anti-VEGF Treatment Response in Neovascular AMD: A Pharmacogenetic Substudy of VIEW 1 and VIEW 2. Invest Ophthalmol Vis Sci 2024; 65:17. [PMID: 38980270 PMCID: PMC11244643 DOI: 10.1167/iovs.65.8.17] [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: 09/01/2023] [Accepted: 06/11/2024] [Indexed: 07/10/2024] Open
Abstract
Purpose This analysis investigated potential associations between gene variants and clinical end points in the VIEW 1 and 2 randomized clinical trials of intravitreal aflibercept and ranibizumab in neovascular age-related macular degeneration (AMD). Methods A genome-wide association analysis was conducted in a subgroup of patients from VIEW 1 and 2 consenting to the optional pharmacogenetic analysis. Results Data were pooled from 780 samples from patients representative of the overall VIEW 1 and 2 populations. After Bonferroni correction for multiplicity and statistical adjustment for baseline risk factors, no significant associations were found between previously identified prognostic AMD gene variants and treatment response according to key prespecified VIEW 1 and 2 end points. Genome-wide, there were no significant genetic associations in patients experiencing gains of ≥15 Early Treatment of Diabetic Retinopathy Study letters after 1 or 2 years of treatment. A cluster of variants in ANO2 (encoding anoctamin 2, a calcium-activated chloride channel expressed on photoreceptor cells) on chromosome 12 reached the level of significance for loss of ≥5 letters after 1 year of treatment (P < 5 × 10-8), with the ANO2 rs2110166 SNP demonstrating highly significant association (P = 1.99 × 10-8). Carriers of the ANO2 rs2110166 TT genotype showed a robust increase in visual acuity versus baseline compared with a small decrease in those with the TC genotype. Conclusions None of the potential prognostic candidate genes were associated with the clinical end points for treated patients. Preliminary analyses suggest an association of ANO2 with retinal function, with a potential impact on vision of approximately one line over at least the first year. Further investigation of the function of ANO2 in retinal pathophysiology is merited.
Collapse
Affiliation(s)
- Robyn H. Guymer
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
- The University of Melbourne, Melbourne, Australia
| | - Rufino Silva
- Faculty of Medicine, University of Coimbra (FMUC-UC), Coimbra, Portugal
- Unidade Local de Saude de Coimbra (ULS-Coimbra), Coimbra, Portugal
- Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra (CACC), Coimbra, Portugal
| | | | | | | | - Amy Damask
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, United States
| | | | | |
Collapse
|
25
|
Han G, Wei P, He M, Jia L, Su Q, Yang X, Hao R. Role of plasma fatty acid in age-related macular degeneration: insights from a mendelian randomization analysis. Lipids Health Dis 2024; 23:206. [PMID: 38951820 PMCID: PMC11218068 DOI: 10.1186/s12944-024-02197-8] [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: 04/04/2024] [Accepted: 06/20/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND An imbalance in lipid metabolism has been linked to the development of AMD, but the causal relationship between AMD and plasma fatty acids (FAs) remains controversial. Using a two-sample Mendelian randomization (MR) approach, we sought to evaluate the impact of specific FA plasma levels on the risk of different AMD subtypes. METHODS We analysed genome-wide association data of circulating FAs from 115,006 European-descended individuals in the UK Biobank. These data were used in a two-sample MR framework to assess the potential role of circulating FAs in developing wet and dry AMD. Sensitivity analyses were conducted to ensure the robustness of our findings. Additional multivariable and locus-specific MR analyses were conducted to evaluate direct effects of FA on AMD subtypes, minimizing biases from lipoprotein-related traits and triglycerides. RESULTS Mendelian randomization revealed associations of omega-3 was associated with decreased wet (OR 0.78, 95%CI 0.66-0.92) and dry AMD (0.85, 0.74-0.97) risk, showed a protective effect on AMD. Notably, the omega-6 to omega-3 ratio showed potential causal effects on both wet (1.27, 1.03-1.56) and dry AMD (1.18, 1.02-1.37). Multivariable MR suggested that the causal relationship of omega-3, omega-6 to omega-3 ratio on wet AMD persists after conditioning on HDL, LDL and triglycerides, albeit with slightly diminished evidence strength. Locus-specific MR linked to omega-3(FADS1, 0.89, 0.82-0.98; FADS2, 0.88, 0.81-0.96) and omega-6 to omega-3 ratio (FADS1, 1.10, 1.02-1.20; FADS2, 1.11, 1.03-1.20) suggests causal effects of these factors on wet AMD. CONCLUSIONS The associations between plasma FA concentrations and AMD, suggest potential causal role of omega-3, and the omega-6 to omega-3 ratio in wet AMD. These results underscore the impact of an imbalanced circulating omega-3 and omega-6 FA ratio on AMD pathophysiology from MR perspective.
Collapse
Affiliation(s)
- Guoge Han
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, PR China.
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Nankai University, Tianjin, PR China.
- Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin, China.
| | - Pinghui Wei
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, PR China
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Nankai University, Tianjin, PR China
- Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin, China
| | - Meiqin He
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, PR China
| | - Lanbo Jia
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Nankai University, Tianjin, PR China
- Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin, China
| | - Qi Su
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Nankai University, Tianjin, PR China
- Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin, China
| | - Xiru Yang
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Nankai University, Tianjin, PR China
- Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin, China
| | - Rui Hao
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, PR China
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Nankai University, Tianjin, PR China
- Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin, China
| |
Collapse
|
26
|
Liang H, Sedillo JC, Schrodi SJ, Ikeda A. Structural variants in linkage disequilibrium with GWAS-significant SNPs. Heliyon 2024; 10:e32053. [PMID: 38882374 PMCID: PMC11177133 DOI: 10.1016/j.heliyon.2024.e32053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/17/2024] [Accepted: 05/28/2024] [Indexed: 06/18/2024] Open
Abstract
With the recent expansion of structural variant identification in the human genome, understanding the role of these impactful variants in disease architecture is critically important. Currently, a large proportion of genome-wide-significant genome-wide association study (GWAS) single nucleotide polymorphisms (SNPs) are functionally unresolved, raising the possibility that some of these SNPs are associated with disease through linkage disequilibrium with causal structural variants. Hence, understanding the linkage disequilibrium between newly discovered structural variants and statistically significant SNPs may provide a resource for further investigation into disease-associated regions in the genome. Here we present a resource cataloging structural variant-significant SNP pairs in high linkage disequilibrium. The database is composed of (i) SNPs that have exhibited genome-wide significant association with traits, primarily disease phenotypes, (ii) newly released structural variants (SVs), and (iii) linkage disequilibrium values calculated from unphased data. All data files including those detailing SV and GWAS SNP associations and results of GWAS-SNP-SV pairs are available at the SV-SNP LD Database and can be accessed at 'https://github.com/hliang-SchrodiLab/SV_SNPs. Our analysis results represent a useful fine mapping tool for interrogating SVs in linkage disequilibrium with disease-associated SNPs. We anticipate that this resource may play an important role in subsequent studies which investigate incorporating disease causing SVs into disease risk prediction models.
Collapse
Affiliation(s)
- Hao Liang
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, USA
| | - Joni C Sedillo
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, USA
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Steven J Schrodi
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, USA
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Akihiro Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
27
|
Liu J, Copland DA, Clare AJ, Gorski M, Richards BT, Scott L, Theodoropoulou S, Greferath U, Cox K, Shi G, Bell OH, Ou K, Powell JLB, Wu J, Robles LM, Li Y, Nicholson LB, Coffey PJ, Fletcher EL, Guymer R, Radeke MJ, Heid IM, Hageman GS, Chan YK, Dick AD. Replenishing IRAK-M expression in retinal pigment epithelium attenuates outer retinal degeneration. Sci Transl Med 2024; 16:eadi4125. [PMID: 38838135 DOI: 10.1126/scitranslmed.adi4125] [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: 04/25/2023] [Accepted: 05/14/2024] [Indexed: 06/07/2024]
Abstract
Chronic inflammation is a constitutive component of many age-related diseases, including age-related macular degeneration (AMD). Here, we identified interleukin-1 receptor-associated kinase M (IRAK-M) as a key immunoregulator in retinal pigment epithelium (RPE) that declines during the aging process. Rare genetic variants of IRAK3, which encodes IRAK-M, were associated with an increased likelihood of developing AMD. In human samples and mouse models, IRAK-M abundance in the RPE declined with advancing age or exposure to oxidative stress and was further reduced in AMD. Irak3-knockout mice exhibited an increased incidence of outer retinal degeneration at earlier ages, which was further exacerbated by oxidative stressors. The absence of IRAK-M led to a disruption in RPE cell homeostasis, characterized by compromised mitochondrial function, cellular senescence, and aberrant cytokine production. IRAK-M overexpression protected RPE cells against oxidative or immune stressors. Subretinal delivery of adeno-associated virus (AAV)-expressing human IRAK3 rescued light-induced outer retinal degeneration in wild-type mice and attenuated age-related spontaneous retinal degeneration in Irak3-knockout mice. Our data show that replenishment of IRAK-M in the RPE may redress dysregulated pro-inflammatory processes in AMD, suggesting a potential treatment for retinal degeneration.
Collapse
Affiliation(s)
- Jian Liu
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol BS8 1TD, UK
| | - David A Copland
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol BS8 1TD, UK
| | - Alison J Clare
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol BS8 1TD, UK
| | - Mathias Gorski
- Department of Genetic Epidemiology, University of Regensburg, Regensburg 93053, Germany
| | - Burt T Richards
- Sharon Eccles Steele Center for Translational Medicine, John A. Moran Eye Center, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Louis Scott
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol BS8 1TD, UK
| | - Sofia Theodoropoulou
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol BS8 1TD, UK
| | - Ursula Greferath
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Katherine Cox
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol BS8 1TD, UK
| | - Gongyu Shi
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol BS8 1TD, UK
| | - Oliver H Bell
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol BS8 1TD, UK
| | - Kepeng Ou
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol BS8 1TD, UK
| | - Jenna Le Brun Powell
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 1TD, UK
| | - Jiahui Wu
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol BS8 1TD, UK
| | - Luis Martinez Robles
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Yingxin Li
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 1TD, UK
| | - Lindsay B Nicholson
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol BS8 1TD, UK
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Peter J Coffey
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Erica L Fletcher
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Robyn Guymer
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Monte J Radeke
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA
| | - Iris M Heid
- Department of Genetic Epidemiology, University of Regensburg, Regensburg 93053, Germany
| | - Gregory S Hageman
- Sharon Eccles Steele Center for Translational Medicine, John A. Moran Eye Center, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Ying Kai Chan
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol BS8 1TD, UK
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Andrew D Dick
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol BS8 1TD, UK
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
- National Institute for Health Research Biomedical Research Centre, Moorfields Eye Hospital, London EC1V 2PD, UK
| |
Collapse
|
28
|
Hou Y, Liu Q, Xiao Z, Li Y, Tian X, Wang Z. Association between chronic kidney disease and age-related macular degeneration: a Mendelian randomization study. Front Aging Neurosci 2024; 16:1399666. [PMID: 38872627 PMCID: PMC11169943 DOI: 10.3389/fnagi.2024.1399666] [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: 03/12/2024] [Accepted: 05/20/2024] [Indexed: 06/15/2024] Open
Abstract
Purpose Observational studies have reported inconsistent results on the relationship between chronic kidney disease (CKD) and age-related macular degeneration (AMD). The primary objective of this study was to investigate the causal relationships between estimated glomerular filtration rate (eGFR), CKD, its common causes, and AMD among participants of European descent. Methods Genetic variants associated with eGFR, CKD and its common causes, encompassing diabetic nephropathy (DN), immunoglobulin A nephropathy (IgAN), and membranous nephropathy (MN) were obtained from previously published genome-wide association studies (GWAS) and FinnGen database. Summary statistics for early AMD, AMD, dry AMD, and wet AMD were acquired from the GWAS and FinnGen database. Inverse-variance-weighted (IVW) method was the main MR analysis. Sensitivity analyses were performed with Cochran's Q, MR-Egger intercept, and leave-one-out analysis. In addition, RadialMR was utilized to identify and remove outliers. Results IVW results showed that CKD, eGFR were not associated with any type of AMD (p > 0.05). DN (OR: 1.042, 95% CI: 1.002-1.083, p = 0.037) and MN (OR: 1.023, 95% CI: 1.007-1.040, p = 0.005) were associated with an increased risk of earl AMD. DN (OR: 1.111, 95% CI: 1.07-1.154, p = 4.87 × 10-8), IgAN (OR: 1.373, 95% CI: 1.097-1.719, p = 0.006), and MN (OR: 1.036, 95% CI: 1.008-1.064, p = 0.012) were associated with an increased risk of AMD. DN (OR: 1.090, 95% CI: 1.042-1.140, p = 1.57 × 10-4) and IgAN (OR: 1.480, 95% CI: 1.178-1.858, p = 7.55 × 10-4) were associated with an increased risk of dry AMD. The risk of wet AMD was associated with DN (OR: 1.107, 95% CI: 1.043-1.174, p = 7.56 × 10-4) and MN (OR: 1.071, 95% CI: 1.040-1.103, p = 5.48 × 10-6). Conclusion This MR study found no evidence of causal relationship between CKD and AMD. DN, IgAN, and MN may increase risk of AMD. This findings underscore the importance of ocular examinations in patients with DN, MN, and IgAN. More studies are needed to support the findings of our current study.
Collapse
Affiliation(s)
- Yawei Hou
- Institute of Chinese Medical Literature and Culture, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qinglin Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhenwei Xiao
- Department of Nephrology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yameng Li
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinyang Tian
- Institute of Chinese Medical Literature and Culture, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhenguo Wang
- Institute of Chinese Medical Literature and Culture, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
29
|
Terao R, Lee TJ, Colasanti J, Pfeifer CW, Lin JB, Santeford A, Hase K, Yamaguchi S, Du D, Sohn BS, Sasaki Y, Yoshida M, Apte RS. LXR/CD38 activation drives cholesterol-induced macrophage senescence and neurodegeneration via NAD + depletion. Cell Rep 2024; 43:114102. [PMID: 38636518 PMCID: PMC11223747 DOI: 10.1016/j.celrep.2024.114102] [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: 09/15/2023] [Revised: 02/23/2024] [Accepted: 03/27/2024] [Indexed: 04/20/2024] Open
Abstract
Although dysregulated cholesterol metabolism predisposes aging tissues to inflammation and a plethora of diseases, the underlying molecular mechanism remains poorly defined. Here, we show that metabolic and genotoxic stresses, convergently acting through liver X nuclear receptor, upregulate CD38 to promote lysosomal cholesterol efflux, leading to nicotinamide adenine dinucleotide (NAD+) depletion in macrophages. Cholesterol-mediated NAD+ depletion induces macrophage senescence, promoting key features of age-related macular degeneration (AMD), including subretinal lipid deposition and neurodegeneration. NAD+ augmentation reverses cellular senescence and macrophage dysfunction, preventing the development of AMD phenotype. Genetic and pharmacological senolysis protect against the development of AMD and neurodegeneration. Subretinal administration of healthy macrophages promotes the clearance of senescent macrophages, reversing the AMD disease burden. Thus, NAD+ deficit induced by excess intracellular cholesterol is the converging mechanism of macrophage senescence and a causal process underlying age-related neurodegeneration.
Collapse
Affiliation(s)
- Ryo Terao
- John F. Hardesty, MD Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA; Department of Ophthalmology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Tae Jun Lee
- John F. Hardesty, MD Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Jason Colasanti
- John F. Hardesty, MD Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Charles W Pfeifer
- John F. Hardesty, MD Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Joseph B Lin
- John F. Hardesty, MD Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrea Santeford
- John F. Hardesty, MD Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Keitaro Hase
- John F. Hardesty, MD Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA; Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Shinobu Yamaguchi
- John F. Hardesty, MD Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel Du
- John F. Hardesty, MD Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian S Sohn
- John F. Hardesty, MD Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Yo Sasaki
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Mitsukuni Yoshida
- John F. Hardesty, MD Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA.
| | - Rajendra S Apte
- John F. Hardesty, MD Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
30
|
Farinha C, Barreto P, Coimbra R, Machado MB, Figueiredo I, Cachulo ML, Cunha-Vaz J, Silva R. Age-Related Macular Degeneration and Extramacular Drusen: Genetic Associations in the Coimbra Eye Study. Invest Ophthalmol Vis Sci 2024; 65:35. [PMID: 38776116 PMCID: PMC11127495 DOI: 10.1167/iovs.65.5.35] [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: 01/22/2024] [Accepted: 05/04/2024] [Indexed: 05/27/2024] Open
Abstract
Purpose To explore the association between the genetics of age-related macular degeneration (AMD) and extramacular drusen (EMD) in patients with and without AMD. Methods We included 1753 eyes (912 subjects) with phenotypic characterization regarding AMD and EMD. Genetic sequencing and the genetic risk score (GRS) for AMD were performed according to the EYE-RISK consortium methodology. To test for differences in the GRS from EMD cases, AMD cases, and controls, a clustered Wilcoxon rank-sum test was used. The association of AMD, EMD, and the GRS was evaluated using logistic regression models adjusted for age and sex. Individual associations of common risk variants for AMD with EMD were explored. Results EMD were found in 755 eyes: 252 (14.4%) with AMD and 503 (28.7%) without. In total, 122 eyes (7.0%) had only AMD, and 876 (50.0%) were controls. EMD were strongly associated with AMD (odds ratio [OR], 3.333; 95% confidence interval [CI], 2.356-4.623; P < 0.001). The GRS was associated with an increased risk of AMD (OR, 1.416; 95% CI, 1.218-1.646; P < 0.001) but not with EMD. Individually, the common risk variants ARMS2 rs10490924 (P = 0.042), C3 rs2230199 (P = 0.042), and CETP rs5817082 (P = 0.042) were associated with EMD, after adjustment for AMD, sex, and age. Conclusions We found a strong association between EMD and AMD, suggesting a common pathogenesis. The GRS for AMD was not associated with EMD, but a partially overlapping genetic basis was suggested when assessing individual risk variants. We propose that EMD per se do not represent an increase in the global genetic risk for AMD.
Collapse
Affiliation(s)
- Cláudia Farinha
- AIBILI—Association for Innovation and Biomedical Research on Light and Image, Coimbra, Portugal
- Ophthalmology Department, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research. Faculty of Medicine (iCBR-FMUC), Coimbra, Portugal
| | - Patrícia Barreto
- AIBILI—Association for Innovation and Biomedical Research on Light and Image, Coimbra, Portugal
| | - Rita Coimbra
- AIBILI—Association for Innovation and Biomedical Research on Light and Image, Coimbra, Portugal
- Department of Mathematics, University of Aveiro, Aveiro, Portugal
| | | | - Inês Figueiredo
- AIBILI—Association for Innovation and Biomedical Research on Light and Image, Coimbra, Portugal
- Ophthalmology Department, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
| | - Maria Luz Cachulo
- AIBILI—Association for Innovation and Biomedical Research on Light and Image, Coimbra, Portugal
- Ophthalmology Department, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research. Faculty of Medicine (iCBR-FMUC), Coimbra, Portugal
| | - José Cunha-Vaz
- AIBILI—Association for Innovation and Biomedical Research on Light and Image, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research. Faculty of Medicine (iCBR-FMUC), Coimbra, Portugal
| | - Rufino Silva
- AIBILI—Association for Innovation and Biomedical Research on Light and Image, Coimbra, Portugal
- Ophthalmology Department, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research. Faculty of Medicine (iCBR-FMUC), Coimbra, Portugal
| |
Collapse
|
31
|
Zhang X, Zhu Z, Huang Y, Shang X, O'Brien TJ, Kwan P, Ha J, Wang W, Liu S, Zhang X, Kiburg K, Bao Y, Wang J, Yu H, He M, Zhang L. Shared genetic aetiology of Alzheimer's disease and age-related macular degeneration by APOC1 and APOE genes. BMJ Neurol Open 2024; 6:e000570. [PMID: 38646507 PMCID: PMC11029327 DOI: 10.1136/bmjno-2023-000570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 04/04/2024] [Indexed: 04/23/2024] Open
Abstract
Background Alzheimer's disease (AD) and age-related macular degeneration (AMD) share similar pathological features, suggesting common genetic aetiologies between the two. Investigating gene associations between AD and AMD may provide useful insights into the underlying pathogenesis and inform integrated prevention and treatment for both diseases. Methods A stratified quantile-quantile (QQ) plot was constructed to detect the pleiotropy among AD and AMD based on genome-wide association studies data from 17 008 patients with AD and 30 178 patients with AMD. A Bayesian conditional false discovery rate-based (cFDR) method was used to identify pleiotropic genes. UK Biobank was used to verify the pleiotropy analysis. Biological network and enrichment analysis were conducted to explain the biological reason for pleiotropy phenomena. A diagnostic test based on gene expression data was used to predict biomarkers for AD and AMD based on pleiotropic genes and their regulators. Results Significant pleiotropy was found between AD and AMD (significant leftward shift on QQ plots). APOC1 and APOE were identified as pleiotropic genes for AD-AMD (cFDR <0.01). Network analysis revealed that APOC1 and APOE occupied borderline positions on the gene co-expression networks. Both APOC1 and APOE genes were enriched on the herpes simplex virus 1 infection pathway. Further, machine learning-based diagnostic tests identified that APOC1, APOE (areas under the curve (AUCs) >0.65) and their upstream regulators, especially ZNF131, ADNP2 and HINFP, could be potential biomarkers for both AD and AMD (AUCs >0.8). Conclusion In this study, we confirmed the genetic pleiotropy between AD and AMD and identified APOC1 and APOE as pleiotropic genes. Further, the integration of multiomics data identified ZNF131, ADNP2 and HINFP as novel diagnostic biomarkers for AD and AMD.
Collapse
Affiliation(s)
- Xueli Zhang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Zhuoting Zhu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yu Huang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xianwen Shang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Terence J O'Brien
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Patrick Kwan
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Jason Ha
- Alfred Health, Melbourne, Victoria, Australia
| | - Wei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Shunming Liu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiayin Zhang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Katerina Kiburg
- Centre for Eye Research, University of Melbourne, East Melbourne, Victoria, Australia
| | - Yining Bao
- China-Australia Joint Research Center for Infectious Diseases, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Jing Wang
- China-Australia Joint Research Center for Infectious Diseases, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Honghua Yu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Mingguang He
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Lei Zhang
- Clinical Medical Research Center, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
- Artificial Intelligence and Modelling in Epidemiology Program, Melbourne Sexual Health Centre, Alfred Health, Melbourne, VIC, Australia
| |
Collapse
|
32
|
Zhang M, Duan L, Feng Y. Causal association between rheumatoid arthritis and an increased risk of age-related macular degeneration: A Mendelian randomization study. Medicine (Baltimore) 2024; 103:e37753. [PMID: 38608102 PMCID: PMC11018156 DOI: 10.1097/md.0000000000037753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/08/2024] [Indexed: 04/14/2024] Open
Abstract
This study's goal is to evaluate if there is a causal connection between rheumatoid arthritis (RA) and age-related macular degeneration (AMD), despite past epidemiological studies suggesting an association between the 2 disorders. The impact of RA on AMD is still unknown. Mendelian randomization (MR) was utilized in this study to assess the two-sample causal relationship between RA and AMD. Summary data from GWAS for RA and AMD in individuals with all European ancestries were gathered using the IEU GWAS database. The GWAS summary statistics of RA (14,361 RA patients and 43,923 healthy controls) and AMD (14,034 AMD patients and 91,214 controls participated) were obtained from the IEU GWAS database. After identifying suitable instrumental variables in line with the 3 MR assumptions, we conducted MR using the Mendelian randomization-Egger (MR-Egger), weighted median, and inverse variance weighting techniques. The MR-Egger intercept and MR-Polyvalent Residuals and Outliers methods were used to investigate the effects of horizontal pleiotropy. The leave-one-out strategy was used to prevent bias caused by certain single nucleotide polymorphisms. Sensitivity analysis was used to detect the heterogeneity. Using 50 single nucleotide polymorphisms as instrumental variables, this study examined the relationship between RA and AMD and discovered that RA increased the risk of AMD (inverse variance weighting odds ratio [OR] = 1.056, 95% confidence interval [CI] = 1.02-1.09, P = 5.44E-04; weighted median OR = 1.085, 95% CI = 1.04-1.14, P = 4.05E-04; MR-Egger OR = 1.074, 95% CI = 1.01-1.14, P = 2.18E-2). The current investigation demonstrated a causal link between AMD and RA. RA increased the risk of AMD. It is advised that future research concentrate on the processes underlying the relationship between RA and AMD.
Collapse
Affiliation(s)
- Mengzhu Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lincheng Duan
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yue Feng
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
33
|
Sendecki A, Ledwoń D, Tuszy A, Nycz J, Wąsowska A, Boguszewska-Chachulska A, Wylęgała A, Mitas AW, Wylęgała E, Teper S. Association of Genetic Risk for Age-Related Macular Degeneration with Morphological Features of the Retinal Microvascular Network. Diagnostics (Basel) 2024; 14:770. [PMID: 38611684 PMCID: PMC11011905 DOI: 10.3390/diagnostics14070770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 03/28/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Age-related macular degeneration (AMD) is a multifactorial disease encompassing a complex interaction between aging, environmental risk factors, and genetic susceptibility. The study aimed to determine whether there is a relationship between the polygenic risk score (PRS) in patients with AMD and the characteristics of the retinal vascular network visualized by optical coherence tomography angiography (OCTA). METHODS 235 patients with AMD and 97 healthy controls were included. We used data from a previous AMD PRS study with the same group. The vascular features from different retina layers were compared between the control group and the patients with AMD. The association between features and PRS was then analyzed using univariate and multivariate approaches. RESULTS Significant differences between the control group and AMD patients were found in the vessel diameter distribution (variance: p = 0.0193, skewness: p = 0.0457) and fractal dimension distribution (mean: p = 0.0024, variance: p = 0.0123). Both univariate and multivariate analyses showed no direct and significant association between the characteristics of the vascular network and AMD PRS. CONCLUSIONS The vascular features of the retina do not constitute a biomarker of the risk of AMD. We have not identified a genotype-phenotype relationship, and the expression of AMD-related genes is perhaps not associated with the characteristics of the retinal vascular network.
Collapse
Affiliation(s)
- Adam Sendecki
- Chair and Clinical Department of Ophthalmology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-752 Katowice, Poland; (A.S.); (A.W.); (A.W.); (E.W.); (S.T.)
| | - Daniel Ledwoń
- Faculty of Biomedical Engineering, Silesian University of Technology, 41-800 Zabrze, Poland; (A.T.); (A.W.M.)
| | - Aleksandra Tuszy
- Faculty of Biomedical Engineering, Silesian University of Technology, 41-800 Zabrze, Poland; (A.T.); (A.W.M.)
| | - Julia Nycz
- Institute of Biomedical Engineering and Informatics, Technische Universität Ilmenau, 98693 Ilmenau, Germany;
| | - Anna Wąsowska
- Chair and Clinical Department of Ophthalmology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-752 Katowice, Poland; (A.S.); (A.W.); (A.W.); (E.W.); (S.T.)
- Genomed S.A., 02-971 Warszawa, Poland;
| | | | - Adam Wylęgała
- Chair and Clinical Department of Ophthalmology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-752 Katowice, Poland; (A.S.); (A.W.); (A.W.); (E.W.); (S.T.)
| | - Andrzej W. Mitas
- Faculty of Biomedical Engineering, Silesian University of Technology, 41-800 Zabrze, Poland; (A.T.); (A.W.M.)
| | - Edward Wylęgała
- Chair and Clinical Department of Ophthalmology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-752 Katowice, Poland; (A.S.); (A.W.); (A.W.); (E.W.); (S.T.)
| | - Sławomir Teper
- Chair and Clinical Department of Ophthalmology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-752 Katowice, Poland; (A.S.); (A.W.); (A.W.); (E.W.); (S.T.)
| |
Collapse
|
34
|
Yoon BW, Lee Y, Seo JH. Potential Causal Association between C-Reactive Protein Levels in Age-Related Macular Degeneration: A Two-Sample Mendelian Randomization Study. Biomedicines 2024; 12:807. [PMID: 38672162 PMCID: PMC11047998 DOI: 10.3390/biomedicines12040807] [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: 12/20/2023] [Revised: 03/31/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Researchers have proposed a possible correlation between age-related macular degeneration (AMD) and inflammation or C-reactive protein (CRP) levels. We investigated the potential causal relationship between CRP levels and AMD. Single-nucleotide polymorphisms (SNPs) associated with CRP exposure were selected as the instrumental variables (IVs) with significance (p < 5 × 10-8) from the genome-wide association study (GWAS) meta-analysis data of Biobank Japan and the UK Biobank. GWAS data for AMD were obtained from 11 International AMD Genomics Consortium studies. An evaluation of causal estimates, utilizing the inverse-variance-weighted (IVW), weighted-median, MR-Egger, MR-Pleiotropy-Residual-Sum, and Outlier tests, was conducted in a two-sample Mendelian randomization (MR) study. We observed significant causal associations between CRP levels and AMD (odds ratio [OR] = 1.13, 95% CI = [1.02-1.24], and p = 0.014 in IVW; OR = 1.18, 95% CI = [1.00-1.38], and p = 0.044 in weight median; OR = 1.31, 95% CI = [1.13-1.52], and p < 0.001 in MR-Egger). The causal relationship between CRP and AMD warrants further research to address the significance of inflammation as a risk factor for AMD.
Collapse
Affiliation(s)
- Byung Woo Yoon
- Department of Internal Medicine, Chung-Ang University Gwangmyung Hospital, Gwangmyung 14353, Republic of Korea;
- College of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Young Lee
- Department of Applied Statistics, Chung-Ang University, Seoul 06974, Republic of Korea;
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul 05368, Republic of Korea
| | - Je Hyun Seo
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul 05368, Republic of Korea
| |
Collapse
|
35
|
Wang L, Wei W, Zhao Y, Chen S, Wu D, Tu M. Causal associations of refractive error and early age-related macular degeneration: A Mendelian randomization study. Exp Eye Res 2024; 241:109850. [PMID: 38423204 DOI: 10.1016/j.exer.2024.109850] [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: 06/18/2023] [Revised: 02/19/2024] [Accepted: 02/26/2024] [Indexed: 03/02/2024]
Abstract
This study aims to determine the risk associated with early age-related macular degeneration (AMD) due to refractive errors (RE) using an analysis of genome-wide association study (GWAS) data through the two-sample Mendelian randomization approach. Single-nucleotide polymorphisms (SNPs) linked to refractive errors (RE) were obtained from numerous GWAS studies involving individuals of European descent. The data for early AMD was obtained from a diverse, multiethnic GWAS meta-analysis that included 105,248 participants (14,034 cases and 91,214 controls). The primary outcome measure focused on the rise in early AMD risk corresponding to a 1-diopter alteration in spherical power and cylindrical power. In the main Mendelian randomization analysis, inverse-variance weighting (IVW) methods were applied for the evaluation. Mendelian Randomization (MR) study revealed a substantial impact of refractive error (RE) on early AMD risk, with a 1-diopter increase in hypermetropia being related to a 1.16 odds ratio (OR) for a greater risk of early AMD (95% CI, 1.10-1.23; P < 0.01). This conclusion was further supported by four supplementary approaches, namely, Weighted mode, Weighted-median, Simple mode, and MR-Egger. The results suggest a heightened risk of early AMD correlated with hyperopia, necessitating further research to thoroughly elucidate this potential causal relationship.
Collapse
Affiliation(s)
- Lingling Wang
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Wenlong Wei
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - YongJi Zhao
- The Affiliated Eye Hospital of Nanchang University, Nanchang, 330006, China
| | - Sixi Chen
- Clinical Laboratory Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Dongjing Wu
- Clinical Laboratory Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Mengjun Tu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
36
|
Seo JH, Lee Y. Causal Associations of Glaucoma and Age-Related Macular Degeneration with Cataract: A Bidirectional Two-Sample Mendelian Randomisation Study. Genes (Basel) 2024; 15:413. [PMID: 38674349 PMCID: PMC11049509 DOI: 10.3390/genes15040413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 03/21/2024] [Accepted: 03/24/2024] [Indexed: 04/28/2024] Open
Abstract
Common age-related eye disorders include glaucoma, cataract, and age-related macular degeneration (AMD); however, little is known about their relationship with age. This study investigated the potential causal relationship between glaucoma and AMD with cataract using genetic data from multi-ethnic populations. Single-nucleotide polymorphisms (SNPs) associated with exposure to cataract were selected as instrumental variables (IVs) from genome-wide association studies using meta-analysis data from BioBank Japan and UK Biobank. A bidirectional two-sample Mendelian randomisation (MR) study was conducted to assess the causal estimates using inverse variance weighted, MR-Egger, and MR pleiotropy residual sum and outlier tests. SNPs with (p < 5.0 × 10-8) were selected as IVs for cataract, primary open-angle glaucoma, and AMD. We found no causal effects of cataract on glaucoma or AMD (all p > 0.05). Furthermore, there were no causal effects of AMD on cataract (odds ratio [OR] = 1.02, p = 0.400). However, glaucoma had a substantial causal effect on cataract (OR = 1.14, p = 0.020). Our study found no evidence for a causal relationship of cataract on glaucoma or AMD and a casual effect of AMD on cataract. Nonetheless, glaucoma demonstrates a causal link with cataract formation, indicating the need for future investigations of age-related eye diseases.
Collapse
Affiliation(s)
- Je Hyun Seo
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul 05368, Republic of Korea;
| | - Young Lee
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul 05368, Republic of Korea;
- Department of Applied Statistics, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
37
|
Shelton DA, Gefke I, Summers V, Kim YK, Yu H, Getz Y, Ferdous S, Donaldson K, Liao K, Papania JT, Chrenek MA, Boatright JH, Nickerson JM. Age-Related RPE changes in Wildtype C57BL/6J Mice between 2 and 32 Months. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.30.574142. [PMID: 38352604 PMCID: PMC10862734 DOI: 10.1101/2024.01.30.574142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Purpose This study provides a systematic evaluation of age-related changes in RPE cell structure and function using a morphometric approach. We aim to better capture nuanced predictive changes in cell heterogeneity that reflect loss of RPE integrity during normal aging. Using C57BL6/J mice ranging from P60-P730, we sought to evaluate how regional changes in RPE shape reflect incremental losses in RPE cell function with advancing age. We hypothesize that tracking global morphological changes in RPE is predictive of functional defects over time. Methods We tested three groups of C57BL/6J mice (young: P60-180; Middle-aged: P365-729; aged: 730+) for function and structural defects using electroretinograms, immunofluorescence, and phagocytosis assays. Results The largest changes in RPE morphology were evident between the young and aged groups, while the middle-aged group exhibited smaller but notable region-specific differences. We observed a 1.9-fold increase in cytoplasmic alpha-catenin expression specifically in the central-medial region of the eye between the young and aged group. There was an 8-fold increase in subretinal, IBA-1-positive immune cell recruitment and a significant decrease in visual function in aged mice compared to young mice. Functional defects in the RPE corroborated by changes in RPE phagocytotic capacity. Conclusions The marked increase of cytoplasmic alpha-catenin expression and subretinal immune cell deposition, and decreased visual output coincide with regional changes in RPE cell morphometrics when stratified by age. These cumulative changes in the RPE morphology showed predictive regional patterns of stress associated with loss of RPE integrity.
Collapse
Affiliation(s)
- Debresha A. Shelton
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Isabelle Gefke
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Vivian Summers
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Yong-Kyu Kim
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Department of Ophthalmology, Hallym University College of Medicine, Kangdong Sacred Heart Hospital, Seoul, South Korea
| | - Hanyi Yu
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Department of Computer Science, Emory University, Atlanta, Georgia, United States
| | - Yana Getz
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Salma Ferdous
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States
| | - Kevin Donaldson
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Kristie Liao
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Jack T. Papania
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Micah A. Chrenek
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Jeffrey H. Boatright
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Decatur, Georgia, United States
| | - John M. Nickerson
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| |
Collapse
|
38
|
Laudenberg N, Kinuthia UM, Langmann T. Microglia depletion/repopulation does not affect light-induced retinal degeneration in mice. Front Immunol 2024; 14:1345382. [PMID: 38288111 PMCID: PMC10822957 DOI: 10.3389/fimmu.2023.1345382] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 12/28/2023] [Indexed: 01/31/2024] Open
Abstract
Reactive microglia are a hallmark of age-related retinal degenerative diseases including age-related macular degeneration (AMD). These cells are capable of secreting neurotoxic substances that may aggravate inflammation that leads to loss of photoreceptors and impaired vision. Despite their role in driving detrimental inflammation, microglia also play supporting roles in the retina as they are a crucial cellular component of the regulatory innate immune system. In this study, we used the colony stimulating factor 1 receptor (CSF1R)-antagonist PLX3397 to investigate the effects of microglia depletion and repopulation in a mouse model of acute retinal degeneration that mimics some aspects of dry AMD. Our main goal was to investigate whether microglia depletion and repopulation affects the outcome of light-induced retinal degeneration. We found that microglia depletion effectively decreased the expression of several key pro-inflammatory factors but was unable to influence the extent of retinal degeneration as determined by optical coherence tomography (OCT) and histology. Interestingly, we found prominent cell debris accumulation in the outer retina under conditions of microglia depletion, presumably due to the lack of efficient phagocytosis that could not be compensated by the retinal pigment epithelium. Moreover, our in vivo experiments showed that renewal of retinal microglia by repopulation did also not prevent rapid microglia activation or preserve photoreceptor death under conditions of light damage. We conclude that microglia ablation strongly reduces the expression of pro-inflammatory factors but cannot prevent photoreceptor loss in the light-damage paradigm of retinal degeneration.
Collapse
Affiliation(s)
- Nils Laudenberg
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Urbanus Muthai Kinuthia
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| |
Collapse
|
39
|
Dabravolski S, Orekhov NA, Melnichenko A, Sukhorukov VN, Popov MA, Orekhov A. Cholesteryl Ester Transfer Protein (CETP) Variations in Relation to Lipid Profiles and Cardiovascular Diseases: An Update. Curr Pharm Des 2024; 30:742-756. [PMID: 38425105 DOI: 10.2174/0113816128284695240219093612] [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: 10/26/2023] [Revised: 01/02/2024] [Accepted: 01/11/2024] [Indexed: 03/02/2024]
Abstract
Lipid metabolism plays an essential role in the pathogenesis of cardiovascular and metabolic diseases. Cholesteryl ester transfer protein (CETP) is a crucial glycoprotein involved in lipid metabolism by transferring cholesteryl esters (CE) and triglycerides (TG) between plasma lipoproteins. CETP activity results in reduced HDL-C and increased VLDL- and LDL-C concentrations, thus increasing the risk of cardiovascular and metabolic diseases. In this review, we discuss the structure of CETP and its mechanism of action. Furthermore, we focus on recent experiments on animal CETP-expressing models, deciphering the regulation and functions of CETP in various genetic backgrounds and interaction with different external factors. Finally, we discuss recent publications revealing the association of CETP single nucleotide polymorphisms (SNPs) with the risk of cardiovascular and metabolic diseases, lifestyle factors, diet and therapeutic interventions. While CETP SNPs can be used as effective diagnostic markers, diet, lifestyle, gender and ethnic specificity should also be considered for effective treatment.
Collapse
Affiliation(s)
- Siarhei Dabravolski
- Department of Biotechnology Engineering, ORT Braude College, Braude Academic College of Engineering, Karmiel, Israel
| | - Nikolay A Orekhov
- Laboratory of Angiopatology, Research Institute of General Pathology and Pathophysiology, The Russian Academy of Medical Sciences, Moscow, Russian Federation
| | - Alexandra Melnichenko
- Laboratory of Angiopatology, Research Institute of General Pathology and Pathophysiology, The Russian Academy of Medical Sciences, Moscow, Russian Federation
| | - Vasily N Sukhorukov
- Laboratory of Angiopatology, Research Institute of General Pathology and Pathophysiology, The Russian Academy of Medical Sciences, Moscow, Russian Federation
| | - Mikhail A Popov
- Laboratory of Angiopatology, Research Institute of General Pathology and Pathophysiology, The Russian Academy of Medical Sciences, Moscow, Russian Federation
| | - Alexander Orekhov
- Laboratory of Angiopatology, Research Institute of General Pathology and Pathophysiology, The Russian Academy of Medical Sciences, Moscow, Russian Federation
| |
Collapse
|
40
|
He W, Han X, Ong JS, Wu Y, Hewitt AW, Mackey DA, Gharahkhani P, MacGregor S. Genome-Wide Meta-analysis Identifies Risk Loci and Improves Disease Prediction of Age-Related Macular Degeneration. Ophthalmology 2024; 131:16-29. [PMID: 37634759 DOI: 10.1016/j.ophtha.2023.08.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 07/22/2023] [Accepted: 08/15/2023] [Indexed: 08/29/2023] Open
Abstract
PURPOSE To identify age-related macular degeneration (AMD) risk loci and to establish a polygenic prediction model. DESIGN Genome-wide association study (GWAS) and polygenic risk score (PRS) construction. PARTICIPANTS We included 64 885 European patients with AMD and 568 740 control participants (with overlapped samples) in the UK Biobank, Genetic Epidemiology Research on Aging (GERA), International AMD Consortium, FinnGen, and published early AMD GWASs in meta-analyses, as well as 733 European patients with AMD and 20 487 control participants from the Canadian Longitudinal Study on Aging (CLSA) and non-Europeans from the UK Biobank and GERA for polygenic risk score validation. METHODS A multitrait meta-analysis of GWASs comprised 64 885 patients with AMD and 568 740 control participants; the multitrait approach accounted for sample overlap. We constructed a PRS for AMD based on both previously reported as well as unreported AMD loci. We applied the PRS to nonoverlapping data from the CLSA. MAIN OUTCOME MEASURES We identified several single nucleotide polymorphisms associated with AMD and established a PRS for AMD risk prediction. RESULTS We identified 63 AMD risk loci alongside the well-established AMD loci CFH and ARMS2, including 9 loci that were not reported in previous GWASs, some of which previously were linked to other eye diseases such as glaucoma (e.g., HIC1). We applied our PRS to nonoverlapping data from the CLSA. A new PRS was constructed using the PRS method, PRS-CS, and significantly improved the prediction accuracy of AMD risk compared with PRSs from previously published datasets. We further showed that even people who carry all the well-known AMD risk alleles at CFH and ARMS2 vary considerably in their AMD risk (ranging from close to 0 in individuals with low PRS to > 50% in individuals with high PRS). Although our PRS was derived in individuals of European ancestry, the PRS shows potential for predicting risk in people of East Asian, South Asian, and Latino ancestry. CONCLUSIONS Our findings improve the knowledge of the genetic architecture of AMD and help achieve better accuracy in AMD prediction. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Weixiong He
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia.
| | - Xikun Han
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Jue-Sheng Ong
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Yeda Wu
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Alex W Hewitt
- Centre for Eye Research Australia, University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, Victorian, Australia; School of Medicine, Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - David A Mackey
- Lions Eye Institute, Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Western Australia, Australia
| | - Puya Gharahkhani
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Stuart MacGregor
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
41
|
DiCesare SM, Ortega AJ, Collier GE, Daniel S, Thompson KN, McCoy MK, Posner BA, Hulleman JD. GSK3 inhibition reduces ECM production and prevents age-related macular degeneration-like pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.14.571757. [PMID: 38168310 PMCID: PMC10760106 DOI: 10.1101/2023.12.14.571757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Malattia Leventinese/Doyne Honeycomb Retinal Dystrophy (ML/DHRD) is an age-related macular degeneration (AMD)-like retinal dystrophy caused by an autosomal dominant R345W mutation in the secreted glycoprotein, fibulin-3 (F3). To identify new small molecules that reduce F3 production from retinal pigmented epithelium (RPE) cells, we knocked-in a luminescent peptide tag (HiBiT) into the endogenous F3 locus which enabled simple, sensitive, and high throughput detection of the protein. The GSK3 inhibitor, CHIR99021 (CHIR), significantly reduced F3 burden (expression, secretion, and intracellular levels) in immortalized RPE and non-RPE cells. Low-level, long-term CHIR treatment promoted remodeling of the RPE extracellular matrix (ECM), reducing sub-RPE deposit-associated proteins (e.g., amelotin, complement component 3, collagen IV, and fibronectin), while increasing RPE differentiation factors (e.g., tyrosinase, and pigment epithelium derived factor). In vivo, treatment of 8 mo R345W+/+ knockin mice with CHIR (25 mg/kg i.p., 1 mo) was well tolerated and significantly reduced R345W F3-associated AMD-like basal laminar deposit number and size, thereby preventing the main pathological feature in these mice. This is the first demonstration of small molecule-based prevention of AMD-like pathology in ML/DHRD mice and may herald a rejuvenation of interest in GSK3 inhibition for the treatment of neurodegenerative diseases, including, potentially AMD itself.
Collapse
Affiliation(s)
- Sophia M. DiCesare
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, 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
| | - Steffi Daniel
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6 St. SE, Minneapolis, Minnesota, 55455, United States
| | - Krista N. Thompson
- 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, United States
| | - Bruce A. Posner
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, United States
| | - John D. Hulleman
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6 St. SE, Minneapolis, Minnesota, 55455, United States
| |
Collapse
|
42
|
Manikandan SK, Logan A, Cerrada-Gimenez M, Fitzhenry L, Coffey L, Kaja S, Rani S. Immune System, Inflammation and Autoantigens in Wet Age-Related Macular Degeneration: Pathological Significance and Therapeutic Importance. Life (Basel) 2023; 13:2236. [PMID: 38137838 PMCID: PMC10744676 DOI: 10.3390/life13122236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/06/2023] [Accepted: 11/13/2023] [Indexed: 12/24/2023] Open
Abstract
Wet age-related macular degeneration (wAMD) is a chronic inflammation-associated neurodegenerative disease affecting the posterior part of the eye in the aging population. Aging results in the reduced functionality of cells and tissues, including the cells of the retina. Initiators of a chronic inflammatory and pathologic state in wAMD may be a result of the accumulation of inevitable metabolic injuries associated with the maintenance of tissue homeostasis from a young age to over 50. Apart from this, risk factors like smoking, genetic predisposition, and failure to repair the injuries that occur, alongside attempts to rescue the hypoxic outer retina may also contribute to the pathogenesis. Aging of the immune system (immunosenescence) and a compromised outer blood retinal barrier (BRB) result in the exposure of the privileged milieu of the retina to the systemic immune system, further increasing the severity of the disease. When immune-privileged sites like the retina are under pathological stress, certain age- and disease-related conditions may necessitate assistance from cells distant from the resident ones to help restore the functionality of the tissue. As a necessary part of tissue repair, inflammation is a major response to disease and recruits immune cells to the site of damage. We suspect that the specific reparative inflammatory responses are controlled by an autoantigen-T cell-mediated mechanism, a process that may be hindered in wAMD.
Collapse
Affiliation(s)
- Sreeraj Kuruppilakath Manikandan
- Ocular Therapeutics Research Group, Pharmaceutical and Molecular Biotechnology Research Centre, South East Technological University, Waterford Campus, X91 K0EK Waterford, Ireland; (S.K.M.); (L.F.)
| | - Ann Logan
- Department of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7HL, UK;
| | | | - Laurence Fitzhenry
- Ocular Therapeutics Research Group, Pharmaceutical and Molecular Biotechnology Research Centre, South East Technological University, Waterford Campus, X91 K0EK Waterford, Ireland; (S.K.M.); (L.F.)
| | - Lee Coffey
- Ocular Therapeutics Research Group, Pharmaceutical and Molecular Biotechnology Research Centre, South East Technological University, Waterford Campus, X91 K0EK Waterford, Ireland; (S.K.M.); (L.F.)
| | - Simon Kaja
- Departments of Ophthalmology, Molecular Pharmacology & Neuroscience, Loyola University Chicago, Maywood, IL 60153, USA
| | - Sweta Rani
- Ocular Therapeutics Research Group, Pharmaceutical and Molecular Biotechnology Research Centre, South East Technological University, Waterford Campus, X91 K0EK Waterford, Ireland; (S.K.M.); (L.F.)
| |
Collapse
|
43
|
Li C, Lu P. Association of Gut Microbiota with Age-Related Macular Degeneration and Glaucoma: A Bidirectional Mendelian Randomization Study. Nutrients 2023; 15:4646. [PMID: 37960299 PMCID: PMC10650403 DOI: 10.3390/nu15214646] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 10/26/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
The objective of this study was to examine the correlation between gut microbiota and both age-related macular degeneration (AMD) and glaucoma. Mendelian randomization studies were conducted utilizing the data sourced from the genome-wide association study (GWAS) database for the gut microbiome, AMD, and glaucoma. Single nucleotide polymorphism (SNP) estimates were summarized through five Mendelian randomization (MR) methods. We utilized Cochran's Q statistic to evaluate the heterogeneity of the instrumental variables (IVs). Additionally, we employed a "leave-one-out" approach to verify the stability of our findings. Inverse variance weighted (IVW) suggests that Eubacterium (oxidoreducens group) and Parabacteroides had a protective effect on AMD. Both weighted median and IVW suggest that Lachnospiraceae (NK4A136 group) and Ruminococcaceae (UCG009) had a protective effect on AMD. However, both weighted median and IVW suggest that Dorea had a risk effect on AMD. Similarly, The IVW of Eubacterium (ventriosum group) showed a risk effect on AMD. The weighted median of Eubacterium (nodatum group), Lachnospiraceae (NC2004 group), and Roseburia had a risk effect on glaucoma. IVW suggested that Ruminococcaceae (UCG004) had a risk effect on glaucoma. Reverse MR analysis found a causal link between Eubacterium (nodatum group) and glaucoma. No causal relationships were found between AMD or glaucoma and the other mentioned bacterial groups. No significant heterogeneity or evidence of horizontal pleiotropy was detected. This study found that certain gut bacteria had protective effects on AMD, while others may be risk factors for AMD or glaucoma. Likewise, reverse MR found that glaucoma led to an increased abundance of certain gut bacteria. Further trials are needed to clarify the specific mechanisms involved.
Collapse
Affiliation(s)
| | - Peirong Lu
- The First Affiliated Hospital of Soochow University, Pinghai Road 899, Suzhou 215005, China;
| |
Collapse
|
44
|
Liu J, Copland DA, Clare AJ, Gorski M, Richards BT, Scott L, Theodoropoulou S, Greferath U, Cox K, Bell OH, Ou K, Powell JLB, Wu J, Robles LM, Li Y, Nicholson LB, Coffey PJ, Fletcher EL, Guymer R, Radeke MJ, Heid IM, Hageman GS, Chan YK, Dick AD. Replenishing Age-Related Decline of IRAK-M Expression in Retinal Pigment Epithelium Attenuates Outer Retinal Degeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.27.559733. [PMID: 37808640 PMCID: PMC10557650 DOI: 10.1101/2023.09.27.559733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Unchecked, chronic inflammation is a constitutive component of age-related diseases, including age-related macular degeneration (AMD). Here we identified interleukin-1 receptor-associated kinase (IRAK)-M as a key immunoregulator in retinal pigment epithelium (RPE) that declines with age. Rare genetic variants of IRAK-M increased the likelihood of AMD. IRAK-M expression in RPE declined with age or oxidative stress and was further reduced in AMD. IRAK-M-deficient mice exhibited increased incidence of outer retinal degeneration at earlier ages, which was further exacerbated by oxidative stressors. The absence of IRAK-M disrupted RPE cell homeostasis, including compromised mitochondrial function, cellular senescence, and aberrant cytokine production. IRAK-M overexpression protected RPE cells against oxidative or immune stressors. Subretinal delivery of AAV-expressing IRAK-M rescued light-induced outer retinal degeneration in wild-type mice and attenuated age-related spontaneous retinal degeneration in IRAK-M-deficient mice. Our data support that replenishment of IRAK-M expression may redress dysregulated pro-inflammatory processes in AMD, thereby treating degeneration.
Collapse
Affiliation(s)
- Jian Liu
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - David A. Copland
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Alison J. Clare
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Mathias Gorski
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Burt T. Richards
- Sharon Eccles Steele Center for Translational Medicine, John A. Moran Eye Center, University of Utah School of Medicine, Salt Lake City, Utah, United States
| | - Louis Scott
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Sofia Theodoropoulou
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Ursula Greferath
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| | - Katherine Cox
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Oliver H. Bell
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Kepeng Ou
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Jenna Le Brun Powell
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Jiahui Wu
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Luis Martinez Robles
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Yingxin Li
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Lindsay B. Nicholson
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Peter J. Coffey
- Institute of Ophthalmology, University College London, London, United Kingdom
| | - Erica L. Fletcher
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| | - Robyn Guymer
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, University of Melbourne, Melbourne, Australia
| | - Monte J. Radeke
- Neuroscience Research Institute, University of California, Santa Barbara, California, United States
| | - Iris M. Heid
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Gregory S. Hageman
- Sharon Eccles Steele Center for Translational Medicine, John A. Moran Eye Center, University of Utah School of Medicine, Salt Lake City, Utah, United States
| | - Ying Kai Chan
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, United States
| | - Andrew D. Dick
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
- Institute of Ophthalmology, University College London, London, United Kingdom
- National Institute for Health Research Biomedical Research Centre, Moorfields Eye Hospital, London, United Kingdom
| |
Collapse
|
45
|
Fan Q, Li H, Wang X, Tham YC, Teo KYC, Yasuda M, Lim WK, Kwan YP, Teo JX, Chen CJ, Chen LJ, Ahn J, Davila S, Miyake M, Tan P, Park KH, Pang CP, Khor CC, Wong TY, Yanagi Y, Cheung CMG, Cheng CY. Contribution of common and rare variants to Asian neovascular age-related macular degeneration subtypes. Nat Commun 2023; 14:5574. [PMID: 37696869 PMCID: PMC10495468 DOI: 10.1038/s41467-023-41256-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 08/28/2023] [Indexed: 09/13/2023] Open
Abstract
Neovascular age-related macular degeneration (nAMD), along with its clinical subtype known as polypoidal choroidal vasculopathy (PCV), are among the leading causes of vision loss in elderly Asians. In a genome-wide association study (GWAS) comprising 3,128 nAMD (1,555 PCV and 1,573 typical nAMD), and 5,493 controls of East Asian ancestry, we identify twelve loci, of which four are novel ([Formula: see text]). Substantial genetic sharing between PCV and typical nAMD is noted (rg = 0.666), whereas collagen extracellular matrix and fibrosis-related pathways are more pronounced for PCV. Whole-exome sequencing in 259 PCV patients revealed functional rare variants burden in collagen type I alpha 1 chain gene (COL1A1; [Formula: see text]) and potential enrichment of functional rare mutations at AMD-associated loci. At the GATA binding protein 5 (GATA5) locus, the most significant GWAS novel loci, the expressions of genes including laminin subunit alpha 5 (Lama5), mitochondrial ribosome associated GTPase 2 (Mtg2), and collagen type IX alpha 3 chain (Col9A3), are significantly induced during retinal angiogenesis and subretinal fibrosis in murine models. Furthermore, retinoic acid increased the expression of LAMA5 and MTG2 in vitro. Taken together, our data provide insights into the genetic basis of AMD pathogenesis in the Asian population.
Collapse
Affiliation(s)
- Qiao Fan
- Center for Quantitative Medicine, Duke-NUS Medical School, National University of Singapore, Singapore, Singapore.
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore.
| | - Hengtong Li
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Centre for Innovation and Precision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xiaomeng Wang
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore
- Center for Vision Research, Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Yih-Chung Tham
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Centre for Innovation and Precision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kelvin Yi Chong Teo
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Masayuki Yasuda
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Weng Khong Lim
- SingHealth Duke-NUS Institute of Precision Medicine, Singapore, Singapore
- SingHealth Duke-NUS Genomic Medicine Centre, Singapore, Singapore
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
- Laboratory of Genome Variation Analytics, Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Yuet Ping Kwan
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore
| | - Jing Xian Teo
- SingHealth Duke-NUS Institute of Precision Medicine, Singapore, Singapore
| | - Ching-Jou Chen
- Center for Vision Research, Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Li Jia Chen
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jeeyun Ahn
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Korea
| | - Sonia Davila
- SingHealth Duke-NUS Institute of Precision Medicine, Singapore, Singapore
| | - Masahiro Miyake
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Patrick Tan
- SingHealth Duke-NUS Institute of Precision Medicine, Singapore, Singapore
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
- Human Genetics, Genome Institute of Singapore, Singapore, Singapore
| | - Kyu Hyung Park
- Department of Ophthalmology, Seoul National University Hospital, Seoul, Korea
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Chiea Chuan Khor
- Human Genetics, Genome Institute of Singapore, Singapore, Singapore
| | - Tien Yin Wong
- Tsinghua Medicine, Tsinghua University, Beijing, China
| | - Yasuo Yanagi
- Department of Ophthalmology and Microtechnology, Yokohama City University, Yokohama, Japan
| | - Chui Ming Gemmy Cheung
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Ching-Yu Cheng
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore.
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore.
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Centre for Innovation and Precision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
46
|
Nordestgaard LT, Christoffersen M, Afzal S, Nordestgaard BG, Tybjærg-Hansen A, Frikke-Schmidt R. Genetic variants in the adenosine triphosphate-binding cassette transporter A1 and risk of age-related macular degeneration. Eur J Epidemiol 2023; 38:985-994. [PMID: 37335386 PMCID: PMC10501952 DOI: 10.1007/s10654-023-01021-4] [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: 12/14/2022] [Accepted: 05/25/2023] [Indexed: 06/21/2023]
Abstract
Genetic variants in ABCA1 are associated with higher concentrations of high-density lipoprotein (HDL) cholesterol. Higher HDL cholesterol concentrations are observationally and genetically associated with higher risk of age-related macular degeneration (AMD). However, whether amino acid-changing genetic variants in ABCA1 associated with high HDL cholesterol concentrations confer a higher risk of AMD in the general population is currently unknown. We tested this hypothesis. The study included 80,972 individuals (1,370 AMD cases) from the Copenhagen General Population Study (CGPS) and 9,584 individuals (142 AMD cases) from the Copenhagen City Heart Study (CCHS) with 10 to 18 years of follow-up. We created an HDL cholesterol weighted allele score based on amino acid-changing ABCA1 variants with a minor allele frequency above 0.001 and divided it into tertiles. The study included 55% women. Mean age was 58 years. The ABCA1 allele score for the third versus the first tertile was associated with HRs (95% confidence intervals (CIs)) of 1.30 (1.14-1.49) for all-cause AMD, 1.26 (1.06-1.50) for nonneovascular AMD, and 1.31 (1.12-1.53) for neovascular AMD in a multivariable adjusted model. On a continuous scale, higher concentrations of genetically determined HDL cholesterol were associated with higher risk of all-cause AMD, nonneovascular AMD, and neovascular AMD in an age- and sex adjusted model and in a multivariable adjusted model. In conclusion, amino acid-changing genetic variants in ABCA1 associated with higher HDL cholesterol concentrations were also associated with higher risk of AMD, suggesting a role for ABCA1 in AMD pathogenesis.
Collapse
Affiliation(s)
- Liv Tybjærg Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Mette Christoffersen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Shoaib Afzal
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Børge Grønne Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
- The Copenhagen City Heart Study, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Anne Tybjærg-Hansen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
- The Copenhagen City Heart Study, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
- The Copenhagen General Population Study, Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark.
| |
Collapse
|
47
|
Brandl C, Finger RP, Heid IM, Mauschitz MM. Age-Related Macular Degeneration in an Ageing Society - Current Epidemiological Research. Klin Monbl Augenheilkd 2023; 240:1052-1059. [PMID: 37666251 DOI: 10.1055/a-2105-1064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Epidemiological studies on age-related macular degeneration (AMD) provide crucial data on the frequency of early and late forms as well as associated risk factors. The increasing number of population-based cross-sectional and longitudinal cohort studies in Germany and Europe with published data is making prevalence and incidence estimators for AMD more robust, although they show mostly method-related fluctuations. This review article brings together the latest published epidemiological measures for AMD from Germany and Central as well as Western Europe. Based on this data and population figures for Germany and Europe, prevalence is projected, and future trends are forecasted. The epidemiological evidence for AMD-associated risk factors is also improving, especially through meta-analyses within large consortia with correspondingly high case numbers. This review article summarizes the latest findings and resulting recommendations for prevention approaches. Additionally, it discusses treatment options and future challenges.
Collapse
Affiliation(s)
- Caroline Brandl
- Universitäts-Augenklinik Regensburg, Universität Regensburg, Fakultät für Medizin, Deutschland
- Lehrstuhl für Genetische Epidemiologie, Universität Regensburg, Fakultät für Medizin, Deutschland
| | - Robert Patrick Finger
- Universitäts-Augenklinik, Universitätsmedizin Mannheim, Medizinische Fakultät Mannheim, Ruprecht-Karls-Universität Heidelberg, Mannheim, Deutschland
- Universitäts-Augenklinik Bonn, Universität Bonn, Deutschland
| | - Iris Maria Heid
- Lehrstuhl für Genetische Epidemiologie, Universität Regensburg, Fakultät für Medizin, Deutschland
| | | |
Collapse
|
48
|
Lei S, Liu Z, Li H. Sleep duration and age-related macular degeneration: a cross-sectional and Mendelian randomization study. Front Aging Neurosci 2023; 15:1247413. [PMID: 37674785 PMCID: PMC10477604 DOI: 10.3389/fnagi.2023.1247413] [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: 06/26/2023] [Accepted: 08/08/2023] [Indexed: 09/08/2023] Open
Abstract
Purpose To investigate the association between sleep duration and age-related macular degeneration (AMD). Design Cross-sectional study, bidirectional two-sample Mendelian randomization (MR). For cross-sectional analysis, we used survey data of 5,481 participants aged ≥40 years from the 2005 to 2008 National Health and Nutrition Examination Survey (NHANES). For MR analysis, we used sleep- and AMD-associated genome-wide association studies (GWAS) data involving large populations. Methods The association between sleep duration and AMD was assessed using logistic regression models. For MR analysis, the primary approach for MR analysis was the inverse-variance weighted (IVW) method. Results In cross-sectional analysis, after adjusting for multiple covariates, short sleep duration (SSD) was found to be associated with increased risk of early AMD [odds ratio (OR) = 1.364, P = 0.036). MR analysis supported the results of cross-sectional analysis: SSD increases the risk of early AMD (β = 0.102, IVW-P = 0.003). Conclusion Our findings provide the evidence supporting the association between sleep deficiency and higher risk of AMD. Further studies are required to confirm our findings and elucidate the mechanisms underlying this association.
Collapse
Affiliation(s)
- Shizhen Lei
- Department of Ophthalmology, The First Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhouyang Liu
- Department of Neurology, The Third People’s Hospital of Chengdu, Chengdu, Sichuan, China
| | - Haihui Li
- Department of Ophthalmology, Yan’an People’s Hospital, Yan’an, Shaanxi, China
| |
Collapse
|
49
|
Borchert GA, Shamsnajafabadi H, Hu ML, De Silva SR, Downes SM, MacLaren RE, Xue K, Cehajic-Kapetanovic J. The Role of Inflammation in Age-Related Macular Degeneration-Therapeutic Landscapes in Geographic Atrophy. Cells 2023; 12:2092. [PMID: 37626902 PMCID: PMC10453093 DOI: 10.3390/cells12162092] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/28/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of vision loss and visual impairment in people over 50 years of age. In the current therapeutic landscape, intravitreal anti-vascular endothelial growth factor (anti-VEGF) therapies have been central to the management of neovascular AMD (also known as wet AMD), whereas treatments for geographic atrophy have lagged behind. Several therapeutic approaches are being developed for geographic atrophy with the goal of either slowing down disease progression or reversing sight loss. Such strategies target the inflammatory pathways, complement cascade, visual cycle or neuroprotective mechanisms to slow down the degeneration. In addition, retinal implants have been tried for vision restoration and stem cell therapies for potentially a dual purpose of slowing down the degeneration and restoring visual function. In particular, therapies focusing on the complement pathway have shown promising results with the FDA approved pegcetacoplan, a complement C3 inhibitor, and avacincaptad pegol, a complement C5 inhibitor. In this review, we discuss the mechanisms of inflammation in AMD and outline the therapeutic landscapes of atrophy AMD. Improved understanding of the various pathway components and their interplay in this complex neuroinflammatory degeneration will guide the development of current and future therapeutic options, such as optogenetic therapy.
Collapse
Affiliation(s)
- Grace A. Borchert
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, Oxford University, Oxford OX3 9DU, UK
| | - Hoda Shamsnajafabadi
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, Oxford University, Oxford OX3 9DU, UK
| | - Monica L. Hu
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, Oxford University, Oxford OX3 9DU, UK
| | - Samantha R. De Silva
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, Oxford University, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford University NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Susan M. Downes
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, Oxford University, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford University NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Robert E. MacLaren
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, Oxford University, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford University NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Kanmin Xue
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, Oxford University, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford University NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Jasmina Cehajic-Kapetanovic
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, Oxford University, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford University NHS Foundation Trust, Oxford OX3 9DU, UK
| |
Collapse
|
50
|
Li FF, Wang Y, Chen L, Chen C, Chen Q, Xiang L, Rao FQ, Shen LJ, Zheng QX, Yi Q, Huang XF. Causal effects of serum lipid biomarkers on early age-related macular degeneration using Mendelian randomization. GENES & NUTRITION 2023; 18:11. [PMID: 37479984 PMCID: PMC10362672 DOI: 10.1186/s12263-023-00730-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 06/18/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND Age-related macular degeneration (AMD) is one of the major causes of vision loss. Early AMD needs to be taken seriously, but the causal effects of lipid biomarkers on early AMD remain unclear. METHODS In this study, two-sample Mendelian randomization (MR) analysis was performed to systematically assess the causal relationships between seven serum lipid biomarkers (apolipoprotein A (ApoA), apolipoprotein B (ApoB), total cholesterol (CHOL), high-density lipoprotein cholesterol (HDL-C), direct low-density lipoprotein cholesterol (LDL-C), lipoprotein A [Lp(a)], and triglycerides (TG)) and risk of early AMD. In total, 14,034 cases and 91,214 controls of European ancestry were included in the analysis (number of SNPs = 11,304,110). RESULTS MR estimates revealed that a higher HDL-C level is strongly associated with increased risk of early AMD (OR = 1.25, 95% CI: 1.15-1.35, P = 2.61 × 10-8). In addition, level of ApoA is also positively associated with risk of early AMD (OR = 2.04, 95% CI: 1.50-2.77, P = 6.27 × 10-6). Conversely, higher levels of TG significantly decrease the risk of early AMD (OR = 0.77, 95% CI: 0.71-0.84, P = 5.02 × 10-10). Sensitivity analyses further supported these associations. Moreover, multivariable MR analyses, adjusted for the effects of correlated lipid biomarkers, yielded similar results. CONCLUSION This study identifies causal relationships between elevated circulating HDL-C/ApoA levels and increased risk of early AMD, in addition to finding that TG specifically reduces the risk of early AMD. These findings contribute to a better understanding of the role of lipid metabolism in drusen formation, particularly in early AMD development.
Collapse
Affiliation(s)
- Fen-Fen Li
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yuqin Wang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Lishuang Chen
- The Ningbo Eye Hospital, Wenzhou Medical University, Ningbo, China
| | - Chong Chen
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qi Chen
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Lue Xiang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Feng-Qin Rao
- School of Pharmaceutical Sciences of Wenzhou Medical University, Wenzhou, China
| | - Li-Jun Shen
- Center for Rehabilitation Medicine, Department of Ophthalmology, Zhejiang Provincial People's Hospital,Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Qin-Xiang Zheng
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
- The Ningbo Eye Hospital, Wenzhou Medical University, Ningbo, China.
| | - Quanyong Yi
- The Ningbo Eye Hospital, Wenzhou Medical University, Ningbo, 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, China.
| |
Collapse
|