1
|
Ortín Vela S, Beyeler MJ, Trofimova O, Iuliani I, Vargas Quiros JD, de Vries VA, Meloni I, Elwakil A, Hoogewoud F, Liefers B, Presby D, Ramdas WD, Tomasoni M, Schlingemann R, Klaver CCW, Bergmann S. Phenotypic and genetic characteristics of retinal vascular parameters and their association with diseases. Nat Commun 2024; 15:9593. [PMID: 39505872 PMCID: PMC11542103 DOI: 10.1038/s41467-024-52334-1] [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: 10/05/2023] [Accepted: 09/03/2024] [Indexed: 11/08/2024] Open
Abstract
Fundus images allow for non-invasive assessment of the retinal vasculature whose features provide important information on health. Using a fully automated image processing pipeline, we extract 17 different morphological vascular phenotypes, including median vessels diameter, diameter variability, main temporal angles, vascular density, central retinal equivalents, the number of bifurcations, and tortuosity, from over 130,000 fundus images of close to 72,000 UK Biobank subjects. We perform genome-wide association studies of these phenotypes. From this, we estimate their heritabilities, ranging between 5 and 25%, and genetic cross-phenotype correlations, which mostly mirror the corresponding phenotypic correlations, but tend to be slightly larger. Projecting our genetic association signals onto genes and pathways reveals remarkably low overlap suggesting largely decoupled mechanisms modulating the different phenotypes. We find that diameter variability, especially for the veins, associates with diseases including heart attack, pulmonary embolism, and age of death. Mendelian Randomization analysis suggests a causal influence of blood pressure and body mass index on retinal vessel morphology, among other results. We validate key findings in two independent smaller cohorts. Our analyses provide evidence that large-scale analysis of image-derived vascular phenotypes has sufficient power for obtaining functional and causal insights into the processes modulating the retinal vasculature.
Collapse
Affiliation(s)
- Sofía Ortín Vela
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | - Michael J Beyeler
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | - Olga Trofimova
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Ilaria Iuliani
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Jose D Vargas Quiros
- Department of Ophthalmology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Victor A de Vries
- Department of Ophthalmology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ilenia Meloni
- Department of Ophthalmology, University of Lausanne, Fondation Asile des Aveugles, Jules Gonin Eye Hospital, Lausanne, Switzerland
- Platform for Research in Ocular Imaging, Department of Ophthalmology, University of Lausanne, Fondation Asile des Aveugles, Jules Gonin Eye Hospital, Lausanne, Switzerland
| | - Adham Elwakil
- Department of Ophthalmology, University of Lausanne, Fondation Asile des Aveugles, Jules Gonin Eye Hospital, Lausanne, Switzerland
- Platform for Research in Ocular Imaging, Department of Ophthalmology, University of Lausanne, Fondation Asile des Aveugles, Jules Gonin Eye Hospital, Lausanne, Switzerland
| | - Florence Hoogewoud
- Department of Ophthalmology, University of Lausanne, Fondation Asile des Aveugles, Jules Gonin Eye Hospital, Lausanne, Switzerland
| | - Bart Liefers
- Department of Ophthalmology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - David Presby
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Wishal D Ramdas
- Department of Ophthalmology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Mattia Tomasoni
- Department of Ophthalmology, University of Lausanne, Fondation Asile des Aveugles, Jules Gonin Eye Hospital, Lausanne, Switzerland
- Platform for Research in Ocular Imaging, Department of Ophthalmology, University of Lausanne, Fondation Asile des Aveugles, Jules Gonin Eye Hospital, Lausanne, Switzerland
| | - Reinier Schlingemann
- Department of Ophthalmology, University of Lausanne, Fondation Asile des Aveugles, Jules Gonin Eye Hospital, Lausanne, Switzerland
- Department of Ophthalmology, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - Caroline C W Klaver
- Department of Ophthalmology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute of Molecular and Clinical Ophthalmology, University of Basel, Basel, Switzerland
| | - Sven Bergmann
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
- Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
2
|
Chua J, Tan B, Wong D, Garhöfer G, Liew XW, Popa-Cherecheanu A, Loong Chin CW, Milea D, Li-Hsian Chen C, Schmetterer L. Optical coherence tomography angiography of the retina and choroid in systemic diseases. Prog Retin Eye Res 2024; 103:101292. [PMID: 39218142 DOI: 10.1016/j.preteyeres.2024.101292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Optical coherence tomography angiography (OCTA) has transformed ocular vascular imaging, revealing microvascular changes linked to various systemic diseases. This review explores its applications in diabetes, hypertension, cardiovascular diseases, and neurodegenerative diseases. While OCTA provides a valuable window into the body's microvasculature, interpreting the findings can be complex. Additionally, challenges exist due to the relative non-specificity of its findings where changes observed in OCTA might not be unique to a specific disease, variations between OCTA machines, the lack of a standardized normative database for comparison, and potential image artifacts. Despite these limitations, OCTA holds immense potential for the future. The review highlights promising advancements like quantitative analysis of OCTA images, integration of artificial intelligence for faster and more accurate interpretation, and multi-modal imaging combining OCTA with other techniques for a more comprehensive characterization of the ocular vasculature. Furthermore, OCTA's potential future role in personalized medicine, enabling tailored treatment plans based on individual OCTA findings, community screening programs for early disease detection, and longitudinal studies tracking disease progression over time is also discussed. In conclusion, OCTA presents a significant opportunity to improve our understanding and management of systemic diseases. Addressing current limitations and pursuing these exciting future directions can solidify OCTA as an indispensable tool for diagnosis, monitoring disease progression, and potentially guiding treatment decisions across various systemic health conditions.
Collapse
Affiliation(s)
- Jacqueline Chua
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Academic Clinical Program, Duke-NUS Medical School, National University of Singapore, Singapore
| | - Bingyao Tan
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; SERI-NTU Advanced Ocular Engineering (STANCE), Singapore, Singapore
| | - Damon Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Academic Clinical Program, Duke-NUS Medical School, National University of Singapore, Singapore; SERI-NTU Advanced Ocular Engineering (STANCE), Singapore, Singapore; School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore; Institute of Molecular and Clinical Ophthalmology, Basel, Switzerland
| | - Gerhard Garhöfer
- Department of Clinical Pharmacology, Medical University Vienna, Vienna, Austria
| | - Xin Wei Liew
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Alina Popa-Cherecheanu
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Emergency University Hospital, Department of Ophthalmology, Bucharest, Romania
| | - Calvin Woon Loong Chin
- Academic Clinical Program, Duke-NUS Medical School, National University of Singapore, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Dan Milea
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Fondation Ophtalmologique Adolphe De Rothschild, Paris, France
| | - Christopher Li-Hsian Chen
- Memory Aging and Cognition Centre, Departments of Pharmacology and Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Leopold Schmetterer
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Academic Clinical Program, Duke-NUS Medical School, National University of Singapore, Singapore; SERI-NTU Advanced Ocular Engineering (STANCE), Singapore, Singapore; School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore; Institute of Molecular and Clinical Ophthalmology, Basel, Switzerland; Department of Clinical Pharmacology, Medical University Vienna, Vienna, Austria; Fondation Ophtalmologique Adolphe De Rothschild, Paris, France; Center for Medical Physics and Biomedical Engineering, Medical University Vienna, Vienna, Austria.
| |
Collapse
|
3
|
Lishinsky-Fischer N, Misgav K, Chowers I, Tiosano L, Shwartz Y, Levy J. Association of subretinal drusenoid deposits and cardiovascular disease. Sci Rep 2024; 14:25569. [PMID: 39462007 PMCID: PMC11513000 DOI: 10.1038/s41598-024-76342-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
The presence of subretinal drusenoid deposits (SDDs) in patients with age-related macular degeneration (AMD) appears to be correlated with cardiovascular disease (CVD) and may serve as a useful tool for predicting certain forms of CVD. Here, we tested this hypothesis by examining whether patients with AMD with SDDs are more likely to have a cardiovascular-related condition and/or undergo a cardiovascular procedure. This is a retrospective cohort study. We included a total of 597 patients with AMD either with SDDs (n = 332) or without SDDs (n = 265). All patients underwent spectral-domain optical coherence tomography (SD-OCT). The SD-OCT scans were annotated by masked, experienced graders. We also extracted data from the patients' electronic medical records (EMRs), including patient demographics, cardiovascular diagnoses, and CVD-related procedures based on ICD-9 codes. AMD patients with SDDs were more likely to be diagnosed with CVD or undergo a cardiovascular procedure compared to AMD patients without SDDs, particularly percutaneous transluminal coronary angioplasty (PTCA; OR 2.73, 95% CI [1.21, 6.13], p = 0.02). Multivariate analysis confirmed the association between the presence of SDDs and a background of PTCA in the presence of other covariates. These data suggest that the presence of SDDs in patients with AMD correlates with certain severe cardiovascular conditions; SDDs and CVD may share common pathogenic pathways.
Collapse
Affiliation(s)
| | - Kinneret Misgav
- Data Research Unit, Hadassah Research Fund, Hadassah Medical Center, Jerusalem, Israel
| | - Itay Chowers
- Ophthalmology Department, Hadassah Medical Center, Jerusalem, Israel
| | - Liran Tiosano
- Ophthalmology Department, Hadassah Medical Center, Jerusalem, Israel
| | - Yahel Shwartz
- Ophthalmology Department, Hadassah Medical Center, Jerusalem, Israel
| | - Jaime Levy
- Ophthalmology Department, Hadassah Medical Center, Jerusalem, Israel.
- Ophthalmology Department, Faculty of Medicine, Hadassah Medical Center, Hebrew University, Kiryat Hadassah, POB 12000, Jerusalem, 91120, Israel.
| |
Collapse
|
4
|
Huang Y, Plotnikov D, Wang H, Shi D, Li C, Zhang X, Zhang X, Tang S, Shang X, Hu Y, Yu H, Zhang H, Guggenheim JA, He M. GWAS-by-subtraction reveals an IOP-independent component of primary open angle glaucoma. Nat Commun 2024; 15:8962. [PMID: 39419966 PMCID: PMC11487129 DOI: 10.1038/s41467-024-53331-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
The etiology of primary open angle glaucoma is constituted by both intraocular pressure-dependent and intraocular pressure-independent mechanisms. However, GWASs of traits affecting primary open angle glaucoma through mechanisms independent of intraocular pressure remains limited. Here, we address this gap by subtracting the genetic effects of a GWAS for intraocular pressure from a GWAS for primary open angle glaucoma to reveal the genetic contribution to primary open angle glaucoma via intraocular pressure-independent mechanisms. Seventeen independent genome-wide significant SNPs were associated with the intraocular pressure-independent component of primary open angle glaucoma. Of these, 7 are located outside known normal tension glaucoma loci, 11 are located outside known intraocular pressure loci, and 2 are novel primary open angle glaucoma loci. The intraocular pressure-independent genetic component of primary open angle glaucoma is associated with glaucoma endophenotypes, while the intraocular pressure-dependent component is associated with blood pressure and vascular permeability. A genetic risk score for the intraocular pressure-independent component of primary open angle glaucoma is associated with 26 different retinal micro-vascular features, which contrasts with the genetic risk score for the intraocular pressure-dependent component. Increased understanding of these intraocular pressure-dependent and intraocular pressure-independent components provides insights into the pathogenesis of glaucoma.
Collapse
Affiliation(s)
- Yu Huang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
- Division of Population Health and Genomics, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK.
| | - Denis Plotnikov
- Central Research Laboratory, Kazan State Medical University, Kazan, Russia
- School of Optometry & Vision Sciences, Cardiff University, Cardiff, UK
| | - Huan Wang
- Division of Population Health and Genomics, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Danli Shi
- Experimental Ophthalmology, The Hong Kong Polytechnic University, Hong Kong, People's Republic of China
| | - Cong Li
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Xueli Zhang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Xiayin Zhang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Shulin Tang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Xianwen Shang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- Centre for Eye Research Australia, Melbourne, VIC, 3002, Australia
| | - Yijun Hu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Honghua Yu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Hongyang Zhang
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, 510080, China.
| | | | - Mingguang He
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- Experimental Ophthalmology, The Hong Kong Polytechnic University, Hong Kong, People's Republic of China.
| |
Collapse
|
5
|
Rovelt J, Freiberg J, Azuara-Blanco A, Virgili G, Kolko M. The Multifaceted Non-Genetic Risk Factors for Primary Open-Angle Glaucoma - An Overview of Systematic Reviews. Ophthalmol Glaucoma 2024:S2589-4196(24)00182-0. [PMID: 39419202 DOI: 10.1016/j.ogla.2024.10.004] [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: 03/31/2024] [Revised: 10/01/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
TOPIC A synthesis of the current knowledge on risk factors for primary open-angle glaucoma (POAG). METHOD We conducted a systematic search of PubMed, Embase, Web of Science, and the Cochrane Library to identify relevant systematic reviews (SR) published in English after 2012. The inclusion criteria focused on SRs investigating risk factors for POAG. We registered the study prospectively in the PROSPERO database (CRD42022351372) and used Covidence and the Risk Of Bias In Systematic reviews (ROBIS) tool to manage article selection and assess the risk of bias in the included SRs. Data was extracted independently by two authors. RESULTS After removing duplicate SRs, we assessed 2,542 SRs. Of these, 2,396 were determined to be irrelevant, leaving 138 for a full text review. Following this, 78 were excluded with reasons, resulting in 57 SRs. Of these, 30 had a low risk of bias. In our bias assessment, SRs categorized as high risk of bias were characterized by 1) lack of sufficient detail in the bias assessment of the SR and 2) insufficient information or missing calculations of heterogeneity among the included studies. In our study, we identified 22 risk factors associated with POAG. The SRs covered a wide range of risk factors for POAG. Among these, the strongest associations with glaucoma, based on effect size, were observed in two SRs related to obstructive sleep apnea (OSA), with a pooled odds ratio (OR) of 3.66 (95% CI: 1.70-7.90) and an adjusted OR of 2.46 (95% CI: 1.32-4.59). Similarly, two SRs investigating Helicobacter pylori (H. pylori) infections showed significant associations, with pooled ORs of 2.08 (95% CI: 1.48-2.93) and 2.08 (95% CI: 1.42-3.04), respectively. CONCLUSION This article summarizes the current knowledge on risk factors for POAG from published SRs. Our findings highlight the complexity of the disease and the nature of the factors that may affect various populations. Among the reported associations with low risk of bias, we found the highest effect estimates for OSA and H. pylori infections in relation to POAG. Our review helps advance understanding, guide future research and inform strategies for the prevention and treatment of POAG.
Collapse
Affiliation(s)
- Jens Rovelt
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Josefine Freiberg
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | | | - Gianni Virgili
- Department of Neurosciences, Psychology Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Miriam Kolko
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark; Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, Glostrup, Denmark.
| |
Collapse
|
6
|
Modaresinejad M, Yang X, Mohammad Nezhady MA, Zhu T, Bajon E, Hou X, Tahiri H, Hardy P, Rivera JC, Lachapelle P, Chemtob S. Endoplasmic Reticulum Stress Delays Choroid Development in the HCAR1 Knockout Mouse. THE AMERICAN JOURNAL OF PATHOLOGY 2024:S0002-9440(24)00354-7. [PMID: 39332673 DOI: 10.1016/j.ajpath.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 08/20/2024] [Accepted: 09/03/2024] [Indexed: 09/29/2024]
Abstract
The subretina, composed of the choroid and the retinal pigment epithelium (RPE), bears a critical role in proper vision. In addition to phagocytosis of photoreceptor debris, the RPE shuttles oxygen and nutrients to the neuroretina. For their own energy production, RPE cells mainly rely on lactate, a major by-product of glycolysis. Lactate, in turn, is believed to convey most of its biological effects via the hydroxycarboxylic acid receptor 1 (HCAR1). Here, the lactate-specific receptor, HCAR1, is found to be exclusively expressed in the RPE cells within the subretina, and Hcar1-/- mice exhibit a substantially thinner choroidal vasculature during development. Notably, the angiogenic properties of lactate on the choroid are impacted by the absence of Hcar1. HCAR1-deficient mice exhibit elevated endoplasmic reticulum stress along with eukaryotic initiation factor 2α phosphorylation, a significant decrease in the global protein translation rate, and a lower proliferation rate of choroidal vasculature. Strikingly, inhibition of the integrated stress response using an inhibitor that reverses the effect of eukaryotic initiation factor 2α phosphorylation restores protein translation and rescues choroidal thinning. These results provide evidence that lactate signalling via HCAR1 is important for choroidal development/angiogenesis and highlight the importance of this receptor in establishing mature vision.
Collapse
Affiliation(s)
- Monir Modaresinejad
- Program in Biomedical Science, Faculty of Medicine, Université de Montréal, Montreal, Quebec; Department of Pediatrics, Ophthalmology and Pharmacology, Centre de Recherche du CHU Sainte-Justine, Montréal, Quebec
| | - Xiaojuan Yang
- School of Optometry, Université de Montréal, Montreal, Quebec; Department of Pediatrics, Ophthalmology and Pharmacology, Centre de Recherche du CHU Sainte-Justine, Montréal, Quebec; Departments of Ophthalmology and Neurology-Neurosurgery, Research Institute of the McGill University Health Centre-Montreal Children's Hospital, Montreal, Quebec, Canada
| | - Mohammad Ali Mohammad Nezhady
- Department of Pediatrics, Ophthalmology and Pharmacology, Centre de Recherche du CHU Sainte-Justine, Montréal, Quebec; Program in Molecular Biology, Faculty of Medicine, Université de Montréal, Montreal, Quebec
| | - Tang Zhu
- Department of Pediatrics, Ophthalmology and Pharmacology, Centre de Recherche du CHU Sainte-Justine, Montréal, Quebec
| | - Emmanuel Bajon
- Department of Pediatrics, Ophthalmology and Pharmacology, Centre de Recherche du CHU Sainte-Justine, Montréal, Quebec
| | - Xin Hou
- Department of Pediatrics, Ophthalmology and Pharmacology, Centre de Recherche du CHU Sainte-Justine, Montréal, Quebec
| | - Houda Tahiri
- Department of Pediatrics, Ophthalmology and Pharmacology, Centre de Recherche du CHU Sainte-Justine, Montréal, Quebec
| | - Pierre Hardy
- Department of Pediatrics, Ophthalmology and Pharmacology, Centre de Recherche du CHU Sainte-Justine, Montréal, Quebec
| | - José Carlos Rivera
- Department of Pediatrics, Ophthalmology and Pharmacology, Centre de Recherche du CHU Sainte-Justine, Montréal, Quebec
| | - Pierre Lachapelle
- Departments of Ophthalmology and Neurology-Neurosurgery, Research Institute of the McGill University Health Centre-Montreal Children's Hospital, Montreal, Quebec, Canada
| | - Sylvain Chemtob
- Program in Biomedical Science, Faculty of Medicine, Université de Montréal, Montreal, Quebec; School of Optometry, Université de Montréal, Montreal, Quebec; Department of Pediatrics, Ophthalmology and Pharmacology, Centre de Recherche du CHU Sainte-Justine, Montréal, Quebec.
| |
Collapse
|
7
|
Abdin A, Abdin AD, Merone G, Aljundi W, Haring B, Abu Dail Y, Mahfoud F, Emrich I, Al Ghorani H, Böhm EW, Seitz B, Böhm M. Cardio-ocular syndrome: Retinal microvascular changes in acutely decompensated heart failure. Eur J Heart Fail 2024. [PMID: 39317947 DOI: 10.1002/ejhf.3474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/19/2024] [Accepted: 09/04/2024] [Indexed: 09/26/2024] Open
Abstract
AIMS To investigate the changes in retinal microvasculature by contemporary imaging techniques during episodes of acute decompensated heart failure (ADHF) and following recompensation compared to age-matched controls without known cardiac or retinal disease. METHODS AND RESULTS Adult patients hospitalized with a primary diagnosis of ADHF, regardless of left ventricular ejection fraction (LVEF) and treated with a minimum dose of 40 mg of intravenous furosemide or equivalent were included. Transthoracic echocardiography was conducted in all patients. Eye examinations were performed out within the initial 24 h after admission and after recompensation before discharge. All eyes underwent a general examination, including a best corrected visual acuity test, dilated fundoscopy, spectral-domain optical coherence tomography (OCT) as well as OCT angiography (OCT-A). In addition, 40 participants without documented cardiac or retinal diseases served as controls. Forty patients with ADHF (mean age 78.9 ± 8.8 years; 32% female) with a mean LVEF of 43 ± 12.8% were included. All patients were treated with intravenous diuretics for a median of 4.3 ± 2.8 days. There was a significant reduction in N-terminal pro-B-type natriuretic peptide from baseline up to discharge (10 396 [interquartile range 6410] vs. 6380 [interquartile range 3933] pg/ml, p ≤ 0.001) and inferior vena cava diameters (2.13 ± 0.4 vs. 1.63 ± 0.3 cm, p = 0.003). Compared to the control group, patients with ADHF showed on admission impaired visual acuity (0.15 ± 0.1 vs. 0.35 ± 0.1 logMAR, p < 0.001), reduced macular vessel density (18.0 ± 1.9 vs. 14.3 ± 3.6 mm/mm2, p < 0.001) and perfusion density (42.6 ± 3.2 vs. 35.2 ± 9.7%, p < 0.001). After recompensation, the mean overall vessel density and mean overall perfusion density were markedly increased at discharge (14.3 ± 3.6 vs. 19.7 ± 2.6 mm/mm2, p = 0.001, and 35.2 ± 9.7 vs. 39.2 ± 6.5%, p = 0.005, respectively). The mean diameter of the superior temporal retinal vein at admission was significantly larger compared to the control group (136 ± 19 vs. 124 ± 22 μm, p = 0.008) and decreased significantly to 122 ± 15 μm at discharge (p < 0.001). CONCLUSION This analysis revealed a remarkable reversible change in retinal microvasculature after ADHF. This could provide a valuable evidence for use of OCT-A in the assessment of overall microperfusion and haemodynamic status in patients with acute heart failure.
Collapse
Affiliation(s)
- Amr Abdin
- Internal Medicine Clinic III, Cardiology, Angiology and Intensive Care Medicine, Saarland University Hospital, Homburg, Germany
| | - Alaa Din Abdin
- Department of Ophthalmology, Saarland University Medical Center, Homburg, Germany
| | - Giuseppe Merone
- Internal Medicine Clinic III, Cardiology, Angiology and Intensive Care Medicine, Saarland University Hospital, Homburg, Germany
| | - Wissam Aljundi
- Department of Ophthalmology, Saarland University Medical Center, Homburg, Germany
| | - Bernhard Haring
- Internal Medicine Clinic III, Cardiology, Angiology and Intensive Care Medicine, Saarland University Hospital, Homburg, Germany
| | - Yaser Abu Dail
- Department of Ophthalmology, Saarland University Medical Center, Homburg, Germany
| | - Felix Mahfoud
- Internal Medicine Clinic III, Cardiology, Angiology and Intensive Care Medicine, Saarland University Hospital, Homburg, Germany
| | - Insa Emrich
- Internal Medicine Clinic III, Cardiology, Angiology and Intensive Care Medicine, Saarland University Hospital, Homburg, Germany
| | - Hussam Al Ghorani
- Internal Medicine Clinic III, Cardiology, Angiology and Intensive Care Medicine, Saarland University Hospital, Homburg, Germany
| | - Elsa Wilma Böhm
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Berthold Seitz
- Department of Ophthalmology, Saarland University Medical Center, Homburg, Germany
| | - Michael Böhm
- Internal Medicine Clinic III, Cardiology, Angiology and Intensive Care Medicine, Saarland University Hospital, Homburg, Germany
| |
Collapse
|
8
|
Repo O, Juonala M, Niinikoski H, Rovio S, Mykkänen J, Lagström H, Cheung CY, Yang D, Vaahtoranta-Lehtonen H, Jula A, Nevalainen J, Rönnemaa T, Viikari J, Raitakari O, Tapp R, Pahkala K. Randomized 20-year infancy-onset dietary intervention, life-long cardiovascular risk factors and retinal microvasculature. Eur Heart J 2024; 45:3072-3085. [PMID: 38995853 PMCID: PMC11365608 DOI: 10.1093/eurheartj/ehae423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 02/01/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND AND AIMS Retinal microvasculature characteristics predict cardiovascular morbidity and mortality. This study investigated associations of lifelong cardiovascular risk factors and effects of dietary intervention on retinal microvasculature in young adulthood. METHODS The cohort is derived from the longitudinal Special Turku Coronary Risk Factor Intervention Project study. The Special Turku Coronary Risk Factor Intervention Project is a 20-year infancy-onset randomized controlled dietary intervention study with frequent study visits and follow-up extending to age 26 years. The dietary intervention aimed at a heart-healthy diet. Fundus photographs were taken at the 26-year follow-up, and microvascular measures [arteriolar and venular diameters, tortuosity (simple and curvature) and fractal dimensions] were derived (n = 486). Cumulative exposure as the area under the curve for cardiovascular risk factors and dietary components was determined for the longest available time period (e.g. from age 7 months to 26 years). RESULTS The dietary intervention had a favourable effect on retinal microvasculature resulting in less tortuous arterioles and venules and increased arteriolar fractal dimension in the intervention group when compared with the control group. The intervention effects were found even when controlled for the cumulative cardiovascular risk factors. Reduced lifelong cumulative intake of saturated fats, main target of the intervention, was also associated with less tortuous venules. Several lifelong cumulative risk factors were independently associated with the retinal microvascular measures, e.g. cumulative systolic blood pressure with narrower arterioles. CONCLUSIONS Infancy-onset 20-year dietary intervention had favourable effects on the retinal microvasculature in young adulthood. Several lifelong cumulative cardiovascular risk factors were independently associated with retinal microvascular structure.
Collapse
Affiliation(s)
- Oskari Repo
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
- Centre for Population Health Research, Turku University Hospital, University of Turku, Turku, Finland
| | - Markus Juonala
- Department of Medicine, University of Turku, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Harri Niinikoski
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
- Centre for Population Health Research, Turku University Hospital, University of Turku, Turku, Finland
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital, University of Turku, Turku, Finland
| | - Suvi Rovio
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
- Centre for Population Health Research, Turku University Hospital, University of Turku, Turku, Finland
| | - Juha Mykkänen
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
- Centre for Population Health Research, Turku University Hospital, University of Turku, Turku, Finland
| | - Hanna Lagström
- Centre for Population Health Research, Turku University Hospital, University of Turku, Turku, Finland
- Department of Public Health, Turku University Hospital, University of Turku, Turku, Finland
- Research Services, Turku University Hospital, Turku, Finland
| | - Carol Y Cheung
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Dawei Yang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | | | - Antti Jula
- Department of Chronic Disease Prevention, Institute for Health and Welfare, Turku, Finland
| | - Jaakko Nevalainen
- Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Tapani Rönnemaa
- Department of Medicine, University of Turku, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Jorma Viikari
- Department of Medicine, University of Turku, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Olli Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
- Centre for Population Health Research, Turku University Hospital, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Robyn Tapp
- Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC, Australia
- Research Institute for Health and Wellbeing, Coventry University, Coventry, United Kingdom
| | - Katja Pahkala
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
- Centre for Population Health Research, Turku University Hospital, University of Turku, Turku, Finland
- Paavo Nurmi Centre and Unit for Health and Physical Activity, University of Turku, Turku, Finland
| |
Collapse
|
9
|
Araújo O, Hernández-Negrín H, Casaroli-Marano RP, Hernández-Rodríguez J, Adán A, Espinosa G, Pelegrín L, Cervera R. Factors associated with early hydroxychloroquine-induced retinal toxicity in patients with systemic lupus erythematosus. Graefes Arch Clin Exp Ophthalmol 2024; 262:2823-2832. [PMID: 38578332 DOI: 10.1007/s00417-024-06461-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 02/12/2024] [Accepted: 03/20/2024] [Indexed: 04/06/2024] Open
Abstract
PURPOSE Hydroxychloroquine is currently recommended for the treatment of systemic lupus erythematosus (SLE), but it can cause irreversible retinal toxicity. This study aimed to identify factors associated with early hydroxychloroquine-induced retinal toxicity in patients with SLE from a single centre for 20 years. METHODS SLE patients diagnosed between 1998 and 2017 and followed up for at least 1 year were included. Demographic, clinical, laboratory and therapeutic data were collected from the electronic medical records and retrospectively analysed. Early hydroxychloroquine-induced retinal toxicity was defined as the development of macular toxicity within the first 5 years of hydroxychloroquine treatment. RESULTS A total of 345 patients followed for a median of 15 years were analysed; 337 (97.7%) patients received hydroxychloroquine, 38 (11.3%) of them presented with retinal toxicity, and 10 (3%) developed early retinal toxicity. These patients had a mean treatment duration of 3.3 years with a mean cumulative dose of 241 g. Patients were diagnosed by visual field (VF) and fundoscopy, and two were also assessed using spectral domain optical coherence tomography (SD-OCT). The median (IQR) age of patients with early toxicity was 56 (51-66) years, and 80% were female. Factors independently associated with early hydroxychloroquine-induced retinal toxicity were lupus anticoagulant positivity (OR 4.2; 95% CI 1.2-15.5) and hypercholesterolaemia (OR 5.6; 95% CI 1.5-21.5). CONCLUSION Our results suggest that lupus anticoagulant positivity and hypercholesterolaemia among SLE patients may be risk factors for early hydroxychloroquine-induced retinal toxicity, regardless of the dose or duration of treatment.
Collapse
Affiliation(s)
- Olga Araújo
- Department of Autoimmune Diseases, Member of the European Reference Centres (ERN) Re-CONNET and RITA, Reference Centre for Systemic Autoimmune Diseases, Vasculitis and Autoinflammatory Diseases of the Catalan and Spanish Health Systems, Hospital Clínic de Barcelona. Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Villarroel 170, 08036, Barcelona, Catalonia, Spain
| | - Halbert Hernández-Negrín
- Department of Autoimmune Diseases, Member of the European Reference Centres (ERN) Re-CONNET and RITA, Reference Centre for Systemic Autoimmune Diseases, Vasculitis and Autoinflammatory Diseases of the Catalan and Spanish Health Systems, Hospital Clínic de Barcelona. Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Villarroel 170, 08036, Barcelona, Catalonia, Spain
| | - Ricardo P Casaroli-Marano
- Department of Ophthalmology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Catalonia, Spain
| | - José Hernández-Rodríguez
- Department of Autoimmune Diseases, Member of the European Reference Centres (ERN) Re-CONNET and RITA, Reference Centre for Systemic Autoimmune Diseases, Vasculitis and Autoinflammatory Diseases of the Catalan and Spanish Health Systems, Hospital Clínic de Barcelona. Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Villarroel 170, 08036, Barcelona, Catalonia, Spain
| | - Alfredo Adán
- Department of Ophthalmology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Catalonia, Spain
| | - Gerard Espinosa
- Department of Autoimmune Diseases, Member of the European Reference Centres (ERN) Re-CONNET and RITA, Reference Centre for Systemic Autoimmune Diseases, Vasculitis and Autoinflammatory Diseases of the Catalan and Spanish Health Systems, Hospital Clínic de Barcelona. Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Villarroel 170, 08036, Barcelona, Catalonia, Spain.
| | - Laura Pelegrín
- Department of Ophthalmology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Catalonia, Spain
| | - Ricard Cervera
- Department of Autoimmune Diseases, Member of the European Reference Centres (ERN) Re-CONNET and RITA, Reference Centre for Systemic Autoimmune Diseases, Vasculitis and Autoinflammatory Diseases of the Catalan and Spanish Health Systems, Hospital Clínic de Barcelona. Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Villarroel 170, 08036, Barcelona, Catalonia, Spain
| |
Collapse
|
10
|
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
|
11
|
Feng J, Xie F, Wu Z, Wu Y. Age-related macular degeneration and cardiovascular disease in US population: an observational study. Acta Cardiol 2024; 79:665-671. [PMID: 38126346 DOI: 10.1080/00015385.2023.2295103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/26/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND As far as we know, age-related macular degeneration (AMD) has become one of the predominant causes of visual impairments. Previous studies have revealed that AMD and many cardiovascular diseases (CVDs) share the same pathologic and genotypic factors, making the connection between AMD and CVD a hot topic. However, the conclusions of the available studies on the relationship between them are somewhat divergent. METHODS We screened 5523 eligible participants from the National Health and Nutrition Examination Survey (NHANES) database from 2005 through 2008 for an observational clinical study design. Binary logistic regression modelling was used to estimate the relations between AMD and various CVDs with and without adjustment for demographics, health status, and behaviours related to health. RESULTS Binary logistic regression analyses showed that AMD was able to increase the risk of CVDs in patients both unadjusted and after adjusting for confounding variables. CONCLUSIONS Within this study, preventing the development of AMD might cut down the incidence of several CVDs, in particular, significantly lowering the stroke risk. These findings indicate that interventions to prevent AMD may also help to prevent CVDs. In general, late AMD has a more severe impact on the risk of CVDs compared with early AMD. These results could help clinical ophthalmology and cardiovascular medicine in their clinical education and interventions.
Collapse
Affiliation(s)
- Jie Feng
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Feng Xie
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhijian Wu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
12
|
Yang Z, Zhang J, Zheng Y. Associations Between Life's Essential 8 and Major Ocular Diseases in the American Middle-Aged and Elderly Population. Am J Ophthalmol 2024; 268:76-85. [PMID: 39089359 DOI: 10.1016/j.ajo.2024.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/15/2024] [Accepted: 07/21/2024] [Indexed: 08/03/2024]
Abstract
PURPOSE To explore the correlation between cardiovascular health (CVH) and ocular diseases, given their shared risk factors and biological mechanisms, this study utilizes the newly updated Life's Essential 8 (LE8) algorithm. DESIGN A cross-sectional study. METHODS This analysis, conducted from February 15 to April 1, 2024, in Changchun, includes data from 4146 participants aged 40 and above, drawn from the National Health and Nutrition Examination Survey database (2005-2008). It covers information on visual health status, dietary habits through interviews, and professional ophthalmological examinations. Participants' CVH status was assessed using the LE8 algorithm, and relationships with major ocular diseases such as retinopathy, cataracts, diabetic retinopathy, glaucoma, and age-related macular degeneration were explored through weighted logistic regression analysis, restricted cubic splines, stratified analysis, and sensitivity analysis. RESULTS After multivariable adjustment, lower LE8 scores showed a significant positive relationship with any ocular disease (odds ratio [OR]: 2.03, 95% confidence interval [CI]: 1.39-2.96, P = .001), any objectively determined ocular disease (OR: 2.24, 95% CI: 1.48-3.38, P < .001), retinopathy (OR: 2.88, 95% CI: 1.89-4.41, P < .001), diabetic retinopathy (OR: 10.23, 95% CI: 3.11-33.61, P < .001), and glaucoma (OR: 2.76, 95% CI: 1.47-5.21, P = .003), with all trends significant (all P < .01). Additionally, lower scores in the behavioral subdomain were significantly correlated with an elevated risk of cataracts (OR: 1.45, 95% CI: 1.03-2.04). Subgroup analyses revealed more pronounced negative correlations between LE8 and retinopathy among females and those suffering from chronic kidney disease. CONCLUSIONS A low CVH score was linked to an increased likelihood of ocular diseases in a US-populated-based study. This correlation supports the potential benefits of enhancing cardiovascular wellness to mitigate the development of ocular conditions.
Collapse
Affiliation(s)
- Ziling Yang
- From the Department of Ophthalmology (Z.Y., Y.Z.), The Second Hospital of Jilin University, Jilin University, Changchun, China
| | - Jinming Zhang
- The First Hospital of Jilin University, Jilin University (J.Z.), Changchun, China
| | - Yajuan Zheng
- From the Department of Ophthalmology (Z.Y., Y.Z.), The Second Hospital of Jilin University, Jilin University, Changchun, China.
| |
Collapse
|
13
|
Ma Y, Zhang Y, Zhang HY, Zhao Y, Li XM, Jiang YF, Yao MD, Jiang Q, Yan B. Dual anti-angiogenic and anti-inflammatory action of tRNA-Cys-5-0007 in ocular vascular disease. J Transl Med 2024; 22:562. [PMID: 38867291 PMCID: PMC11167814 DOI: 10.1186/s12967-024-05338-w] [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: 01/11/2024] [Accepted: 05/22/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Intravitreal injections of angiogenesis inhibitors have proved efficacious in the majority of patients with ocular angiogenesis. However, one-fourth of all treated patients fail to derive benefits from intravitreal injections. tRNA-derived small RNA (tsRNA) emerges as a crucial class of non-coding RNA molecules, orchestrating key roles in the progression of human diseases by modulating multiple targets. Through our prior sequencing analyses and bioinformatics predictions, tRNA-Cys-5-0007 has shown as a potential regulator of ocular angiogenesis. This study endeavors to elucidate the precise role of tRNA-Cys-5-0007 in the context of ocular angiogenesis. METHODS Quantitative reverse transcription PCR (qRT-PCR) assays were employed to detect tRNA-Cys-5-0007expression. EdU assays, sprouting assays, transwell assays, and Matrigel assays were conducted to elucidate the involvement of tRNA-Cys-5-0007 in endothelial angiogenic effects. STZ-induced diabetic model, OIR model, and laser-induced CNV model were utilized to replicate the pivotal features of ocular vascular diseases and evaluate the influence of tRNA-Cys-5-0007 on ocular angiogenesis and inflammatory responses. Bioinformatics analysis, luciferase activity assays, RNA pull-down assays, and in vitro studies were employed to elucidate the anti-angiogenic mechanism of tRNA-Cys-5-0007. Exosomal formulation was employed to enhance the synergistic anti-angiogenic and anti-inflammatory efficacy of tRNA-Cys-5-0007. RESULTS tRNA-Cys-5-0007 expression was down-regulated under angiogenic conditions. Conversely, tRNA-Cys-5-0007 overexpression exhibited anti-angiogenic effects in retinal endothelial cells, as evidenced by reduced proliferation, sprouting, migration, and tube formation abilities. In diabetic, laser-induced CNV, and OIR models, tRNA-Cys-5-0007 overexpression led to decreased ocular vessel leakage, inhibited angiogenesis, and reduced ocular inflammation. Mechanistically, these effects were attributed to the targeting of vascular endothelial growth factor A (VEGFA) and TGF-β1 by tRNA-Cys-5-0007. The utilization of an exosomal formulation further potentiated the synergistic anti-angiogenic and anti-inflammatory efficacy of tRNA-Cys-5-0007. CONCLUSIONS Concurrent targeting of tRNA-Cys-5-0007 for anti-angiogenic and anti-inflammatory therapy holds promise for enhancing the effectiveness of current anti-angiogenic therapy.
Collapse
Affiliation(s)
- Yan Ma
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, 210000, China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210000, China
| | - Ying Zhang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210000, China
| | - Hui-Ying Zhang
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, 210000, China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210000, China
| | - Ya Zhao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Xiu-Miao Li
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210000, China
| | - Yi-Fei Jiang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210000, China
| | - Mu-Di Yao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| | - Qin Jiang
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, 210000, China.
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210000, China.
| | - Biao Yan
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
14
|
Liu X, Tan H, Wang W, Chen Z. Deep learning based retinal vessel segmentation and hypertensive retinopathy quantification using heterogeneous features cross-attention neural network. Front Med (Lausanne) 2024; 11:1377479. [PMID: 38841586 PMCID: PMC11150614 DOI: 10.3389/fmed.2024.1377479] [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: 01/27/2024] [Accepted: 05/09/2024] [Indexed: 06/07/2024] Open
Abstract
Retinal vessels play a pivotal role as biomarkers in the detection of retinal diseases, including hypertensive retinopathy. The manual identification of these retinal vessels is both resource-intensive and time-consuming. The fidelity of vessel segmentation in automated methods directly depends on the fundus images' quality. In instances of sub-optimal image quality, applying deep learning-based methodologies emerges as a more effective approach for precise segmentation. We propose a heterogeneous neural network combining the benefit of local semantic information extraction of convolutional neural network and long-range spatial features mining of transformer network structures. Such cross-attention network structure boosts the model's ability to tackle vessel structures in the retinal images. Experiments on four publicly available datasets demonstrate our model's superior performance on vessel segmentation and the big potential of hypertensive retinopathy quantification.
Collapse
Affiliation(s)
- Xinghui Liu
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
- Department of Cardiovascular Medicine, Guizhou Provincial People's Hospital, Guiyang, China
| | - Hongwen Tan
- Department of Cardiovascular Medicine, Guizhou Provincial People's Hospital, Guiyang, China
| | - Wu Wang
- Electrical Engineering College, Guizhou University, Guiyang, China
| | - Zhangrong Chen
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
- Department of Cardiovascular Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
15
|
Iorga RE, Costin D, Munteanu-Dănulescu RS, Rezuș E, Moraru AD. Non-Invasive Retinal Vessel Analysis as a Predictor for Cardiovascular Disease. J Pers Med 2024; 14:501. [PMID: 38793083 PMCID: PMC11122007 DOI: 10.3390/jpm14050501] [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: 04/19/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Cardiovascular disease (CVD) is the most frequent cause of death worldwide. The alterations in the microcirculation may predict the cardiovascular mortality. The retinal vasculature can be used as a model to study vascular alterations associated with cardiovascular disease. In order to quantify microvascular changes in a non-invasive way, fundus images can be taken and analysed. The central retinal arteriolar (CRAE), the venular (CRVE) diameter and the arteriolar-to-venular diameter ratio (AVR) can be used as biomarkers to predict the cardiovascular mortality. A narrower CRAE, wider CRVE and a lower AVR have been associated with increased cardiovascular events. Dynamic retinal vessel analysis (DRVA) allows the quantification of retinal changes using digital image sequences in response to visual stimulation with flicker light. This article is not just a review of the current literature, it also aims to discuss the methodological benefits and to identify research gaps. It highlights the potential use of microvascular biomarkers for screening and treatment monitoring of cardiovascular disease. Artificial intelligence (AI), such as Quantitative Analysis of Retinal vessel Topology and size (QUARTZ), and SIVA-deep learning system (SIVA-DLS), seems efficient in extracting information from fundus photographs and has the advantage of increasing diagnosis accuracy and improving patient care by complementing the role of physicians. Retinal vascular imaging using AI may help identify the cardiovascular risk, and is an important tool in primary cardiovascular disease prevention. Further research should explore the potential clinical application of retinal microvascular biomarkers, in order to assess systemic vascular health status, and to predict cardiovascular events.
Collapse
Affiliation(s)
- Raluca Eugenia Iorga
- Department of Surgery II, Discipline of Ophthalmology, “Grigore T. Popa” University of Medicine and Pharmacy, Strada Universitatii No. 16, 700115 Iași, Romania; (R.E.I.); (A.D.M.)
| | - Damiana Costin
- Doctoral School, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | | | - Elena Rezuș
- Department of Internal Medicine II, Discipline of Reumathology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania;
| | - Andreea Dana Moraru
- Department of Surgery II, Discipline of Ophthalmology, “Grigore T. Popa” University of Medicine and Pharmacy, Strada Universitatii No. 16, 700115 Iași, Romania; (R.E.I.); (A.D.M.)
| |
Collapse
|
16
|
Chang CJ, Fan YH, Chiu YC, Cheng WM. Cold hypersensitivity in the hands and feet is associated with erectile dysfunction in young Taiwanese men. Sci Rep 2024; 14:10577. [PMID: 38719920 PMCID: PMC11078973 DOI: 10.1038/s41598-024-60260-x] [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: 04/20/2024] [Indexed: 05/12/2024] Open
Abstract
Cold hypersensitivity in the hands and feet (CHHF) is a protective or predisposing factor for many diseases; however, the relationship between CHHF and erectile dysfunction (ED) remains unclear. We aimed to investigate associations between CHHF and ED among young men of Southeast Asian origin. In this cross-sectional study, sexually active Taiwanese men aged 20-40 years were enrolled via an online questionnaire comprising general demographic information, comorbidities, subjective thermal sensations of their hands and feet in the past 6 months, and their erectile function using the International Index of Erectile Function-5 (IIEF-5). Participants who reported cold sensation of hands and feet were classified to have CHHF; those with IIEF-5 score ≤ 21 were considered to have ED. Total 54.2% and 27.9% of participants had ED and CHHF, respectively. Men with CHHF were significantly younger, had lower body mass index and IIEF-5 scores (p < 0.001), and a lower prevalence of diabetes mellitus (p = 0.033) along with higher prevalence of ED, psychiatric disorders, and insomnia (p < 0.001). After adjusting for predisposing factors of ED, CHHF (odds ratio 1.410, 95% confidence interval 1.159-1.714; p = 0.001) remained an independent predictor of ED. Thus, CHHF is independently associated with ED, affecting more than a quarter of young Taiwanese men. Autonomic dysregulation and subclinical endothelial dysfunction may be common pathophysiologies of CHHF and ED.
Collapse
Affiliation(s)
- Che-Jui Chang
- Division of Urology, Department of Surgery, Zhongxiao Branch, Taipei City Hospital, Taipei, Taiwan
| | - Yu-Hua Fan
- Department of Urology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Shu-Tien Urological Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Chun Chiu
- Division of Urology, Department of Surgery, Zhongxiao Branch, Taipei City Hospital, Taipei, Taiwan
- Department of Urology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Shu-Tien Urological Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Exercise and Health Sciences, University of Taipei, Taipei, Taiwan
| | - Wei-Ming Cheng
- Division of Urology, Department of Surgery, Zhongxiao Branch, Taipei City Hospital, Taipei, Taiwan.
- Department of Urology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
17
|
Yuan Y, Dong M, Wen S, Yuan X, Zhou L. Retinal microcirculation: A window into systemic circulation and metabolic disease. Exp Eye Res 2024; 242:109885. [PMID: 38574944 DOI: 10.1016/j.exer.2024.109885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/06/2024]
Abstract
The retinal microcirculation system constitutes a unique terminal vessel bed of the systemic circulation, and its perfusion status is directly associated with the neural function of the retina. This vascular network, essential for nourishing various layers of the retina, comprises two primary microcirculation systems: the retinal microcirculation and the choroidal microcirculation, with each system supplying blood to distinct retinal layers and maintaining the associated neural function. The blood flow of those capillaries is regulated via different mechanisms. However, a range of internal and external factors can disrupt the normal architecture and blood flow within the retinal microcirculation, leading to several retinal pathologies, including diabetic retinopathy, macular edema, and vascular occlusions. Metabolic disturbances such as hyperglycemia, hypertension, and dyslipidemia are known to modify retinal microcirculation through various pathways. These alterations are observable in chronic metabolic conditions like diabetes, coronary artery disease, and cerebral microvascular disease due to advances in non-invasive or minimally invasive retinal imaging techniques. Thus, examination of the retinal microcirculation can provide insights into the progression of numerous chronic metabolic disorders. This review discusses the anatomy, physiology and pathophysiology of the retinal microvascular system, with a particular emphasis on the connections between retinal microcirculation and systemic circulation in both healthy states and in the context of prevalent chronic metabolic diseases.
Collapse
Affiliation(s)
- Yue Yuan
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China.
| | - Meiyuan Dong
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China; Graduate School of Hebei Medical University, Shijiazhuang, China.
| | - Song Wen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China.
| | - Xinlu Yuan
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China.
| | - Ligang Zhou
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China; Graduate School of Hebei Medical University, Shijiazhuang, China; Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Shanghai, China.
| |
Collapse
|
18
|
Kang N, Wang M, Pang C, Lan R, Li B, Guan J, Wang H. Cross-patch feature interactive net with edge refinement for retinal vessel segmentation. Comput Biol Med 2024; 174:108443. [PMID: 38608328 DOI: 10.1016/j.compbiomed.2024.108443] [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: 01/03/2024] [Revised: 03/14/2024] [Accepted: 04/07/2024] [Indexed: 04/14/2024]
Abstract
Retinal vessel segmentation based on deep learning is an important auxiliary method for assisting clinical doctors in diagnosing retinal diseases. However, existing methods often produce mis-segmentation when dealing with low contrast images and thin blood vessels, which affects the continuity and integrity of the vessel skeleton. In addition, existing deep learning methods tend to lose a lot of detailed information during training, which affects the accuracy of segmentation. To address these issues, we propose a novel dual-decoder based Cross-patch Feature Interactive Net with Edge Refinement (CFI-Net) for end-to-end retinal vessel segmentation. In the encoder part, a joint refinement down-sampling method (JRDM) is proposed to compress feature information in the process of reducing image size, so as to reduce the loss of thin vessels and vessel edge information during the encoding process. In the decoder part, we adopt a dual-path model based on edge detection, and propose a Cross-patch Interactive Attention Mechanism (CIAM) in the main path to enhancing multi-scale spatial channel features and transferring cross-spatial information. Consequently, it improve the network's ability to segment complete and continuous vessel skeletons, reducing vessel segmentation fractures. Finally, the Adaptive Spatial Context Guide Method (ASCGM) is proposed to fuse the prediction results of the two decoder paths, which enhances segmentation details while removing part of the background noise. We evaluated our model on two retinal image datasets and one coronary angiography dataset, achieving outstanding performance in segmentation comprehensive assessment metrics such as AUC and CAL. The experimental results showed that the proposed CFI-Net has superior segmentation performance compared with other existing methods, especially for thin vessels and vessel edges. The code is available at https://github.com/kita0420/CFI-Net.
Collapse
Affiliation(s)
- Ning Kang
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, 541004, China
| | - Maofa Wang
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, 541004, China
| | - Cheng Pang
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, 541004, China; Guangxi Key Laboratory of Image and Graphic Intelligent Processing, Guilin, 541004, China
| | - Rushi Lan
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, 541004, China; Guangxi Key Laboratory of Image and Graphic Intelligent Processing, Guilin, 541004, China
| | - Bingbing Li
- Department of Pathology, Ganzhou Municipal Hospital, Ganzhou, 341000, China
| | - Junlin Guan
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, 541004, China.
| | - Huadeng Wang
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, 541004, China; Guangxi Key Laboratory of Image and Graphic Intelligent Processing, Guilin, 541004, China
| |
Collapse
|
19
|
wang J, Weng H, Qian Y, Wang Y, Wang L, Wang X, Zhang P, Wang Z. The impact of serum BNP on retinal perfusion assessed by an AI-based denoising optical coherence tomography angiography in CHD patients. Heliyon 2024; 10:e29305. [PMID: 38655359 PMCID: PMC11035033 DOI: 10.1016/j.heliyon.2024.e29305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/26/2024] Open
Abstract
Background To investigate the correlation between retinal vessel density (VD) parameters with serum B-type natriuretic peptide (BNP) in patients with coronary heart disease (CHD) using novel optical coherence tomography angiography (OCTA) denoising images based on artificial intelligence (AI). Methods OCTA images of the optic nerve and macular area were obtained using a Canon-HS100 OCT device in 176 patients with CHD. Baseline information and blood test results were recorded. Results Retinal VD parameters of the macular and optic nerves on OCTA were significantly decreased in patients with CHD after denoising. Retinal VD of the superficial capillary plexus (SCP), deep capillary plexus (DCP) and radial peripapillary capillary (RPC) was strongly correlated with serum BNP levels in patients with CHD. Significant differences were noted in retinal thickness and retinal VD (SCP, DCP and RPC) between the increased BNP and normal BNP groups in patients with CHD. Conclusion Deep learning denoising can remove background noise and smooth rough vessel surfaces. SCP,DCP and RPC may be potential clinical markers of cardiac function in patients with CHD. Denoising shows great potential for improving the sensitivity of OCTA images as a biomarker for CHD progression.
Collapse
Affiliation(s)
- Jin wang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Huan Weng
- Department of Ophthalmology, Huashan Hospital of Fudan University, Shanghai, China
| | - Yiwen Qian
- Department of Ophthalmology, Huashan Hospital of Fudan University, Shanghai, China
| | - Yuceng Wang
- Department of Ophthalmology, Huashan Hospital of Fudan University, Shanghai, China
| | - Luoziyi Wang
- Department of Ophthalmology, Huashan Hospital of Fudan University, Shanghai, China
| | - Xin Wang
- Department of Ophthalmology, Huashan Hospital of Fudan University, Shanghai, China
| | - Pei Zhang
- Department of Ophthalmology, Huashan Hospital of Fudan University, Shanghai, China
| | - Zhiliang Wang
- Department of Ophthalmology, Huashan Hospital of Fudan University, Shanghai, China
| |
Collapse
|
20
|
Charles LE, Gu JK, Violanti JM. Impact of Shiftwork on Retinal Vasculature Diameters over a 5-Year Period: A Preliminary Investigation Using the BCOPS Study Data. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:439. [PMID: 38673350 PMCID: PMC11050094 DOI: 10.3390/ijerph21040439] [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: 12/19/2023] [Revised: 03/06/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024]
Abstract
Our aim was to investigate the impact of shiftwork on changes in central retinal arteriolar equivalent (CRAE), a measure of arteriolar width, and central retinal venular equivalent (CRVE), a measure of venular width, over five years. The participants were 117 officers (72.7% men) examined at the first (2011-2014) and second (2015-2019) follow-up examinations in the Buffalo Cardio-Metabolic Occupational Police Stress study. Shiftwork data were obtained from the City of Buffalo, NY payroll records. Retinal diameters were measured using a standardized protocol. ANCOVA was used to compare mean change in CRAE and CRVE between the two examinations across shiftwork categories. Among men only, those who worked ≥70% hours on day shifts had a larger decrease in mean CRAE (-7.13 µm ± 2.51) compared to those who worked <70% day (-0.08 ± 0.96; p = 0.011). Among patrol officers, those who worked ≥70% day had a larger decrease in CRAE compared to those who worked <70% day (p = 0.015). Also, officers who worked ≥70% day had an increase in mean CRVE (µm) (4.56 ± 2.56) compared to those who worked <70% (-2.32 ± 1.32; p = 0.027). Over the five-year period, we observed adverse changes in arteriolar and venular diameters among officers who worked ≥70% on day shifts. The results should be interpreted with caution due to the small sample sizes.
Collapse
Affiliation(s)
- Luenda E. Charles
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV 26505-2888, USA;
| | - Ja K. Gu
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV 26505-2888, USA;
| | - John M. Violanti
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, State University of New York at Buffalo, Buffalo, NY 14214-8001, USA;
| |
Collapse
|
21
|
Prem Senthil M, Kurban C, Thuy Nguyen N, Nguyen AP, Chakraborty R, Delaney C, Clark R, Anand S, Bhardwaj H. Role of noninvasive ocular imaging as a biomarker in peripheral artery disease (PAD): A systematic review. Vasc Med 2024; 29:215-222. [PMID: 38054219 PMCID: PMC11010562 DOI: 10.1177/1358863x231210866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
This study aimed to review the current literature exploring the utility of noninvasive ocular imaging for the diagnosis of peripheral artery disease (PAD). Our search was conducted in early April 2022 and included the databases Medline, Scopus, Embase, Cochrane, and others. Five articles were included in the final review. Of the five studies that used ocular imaging in PAD, two studies used retinal color fundus photography, one used optical coherence tomography (OCT), and two used optical coherence tomography angiography (OCTA) to assess the ocular changes in PAD. PAD was associated with both structural and functional changes in the retina. Structural alterations around the optic disc and temporal retinal vascular arcades were seen in color fundus photography of patients with PAD compared to healthy individuals. The presence of retinal hemorrhages, exudates, and microaneurysms in color fundus photography was associated with an increased future risk of PAD, especially the severe form of the disease. The retinal nerve fiber layer (RNFL) was significantly thinner in the nasal quadrant in patients with PAD compared to age-matched healthy individuals in OCT. Similarly, the choroidal thickness in the subfoveal region was significantly thinner in patients with PAD compared to controls. Patients with PAD also had a significant reduction in the retinal and choroidal circulation in OCTA compared to healthy controls. As PAD causes thinning and ischemic changes in retinal vessels, examination of the retinal vessels using retinal imaging techniques can provide useful information about early microvascular damage in PAD. Ocular imaging could potentially serve as a biomarker for PAD. PROSPERO ID: CRD42022310637.
Collapse
Affiliation(s)
- Mallika Prem Senthil
- College of Nursing and Health Sciences, Caring Futures Institute, Flinders University, Adelaide, SA, Australia
| | - Chroran Kurban
- College of Nursing and Health Sciences, Caring Futures Institute, Flinders University, Adelaide, SA, Australia
| | - Ngoc Thuy Nguyen
- College of Nursing and Health Sciences, Caring Futures Institute, Flinders University, Adelaide, SA, Australia
| | - Anh-Phuong Nguyen
- College of Nursing and Health Sciences, Caring Futures Institute, Flinders University, Adelaide, SA, Australia
| | - Ranjay Chakraborty
- College of Nursing and Health Sciences, Caring Futures Institute, Flinders University, Adelaide, SA, Australia
| | - Christopher Delaney
- Department of Vascular and Endovascular Surgery, Flinders Medical Centre, Adelaide, SA, Australia
| | - Robyn Clark
- College of Nursing and Health Sciences, Caring Futures Institute, Flinders University, Adelaide, SA, Australia
| | - Saumya Anand
- College of Nursing and Health Sciences, Caring Futures Institute, Flinders University, Adelaide, SA, Australia
| | - Heena Bhardwaj
- College of Nursing and Health Sciences, Caring Futures Institute, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
22
|
De Ciuceis C, Rosei CA, Malerba P, Rossini C, Nardin M, Chiarini G, Famà F, Lemoli M, Baresi M, Petelca A, Bortoluzzi C, Porteri E, Salvetti M, Muiesan ML, Rosei EA, Rizzoni D. Prognostic significance of the wall to lumen ratio of retinal arterioles evaluated by adaptive optics. Eur J Intern Med 2024; 122:86-92. [PMID: 37914655 DOI: 10.1016/j.ejim.2023.10.035] [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: 07/14/2023] [Revised: 10/16/2023] [Accepted: 10/27/2023] [Indexed: 11/03/2023]
Abstract
OBJECTIVE Microvascular structural alterations may be considered an important form of hypertension-mediated organ damage. An increased media-to-lumen ratio of subcutaneous small arteries evaluated with locally invasive techniques (micromyography) predicts the development of cardiovascular (CV) events. However, it is not known whether retinal arteriole structural alterations evaluated with a noninvasive approach (Adaptive Optics) may have a prognostic significance. DESIGN AND METHODS Two-hundred and thirty-seven subjects (mean age 58.7 ± 16.1 years, age range 13-89 years; 116 males) were included in the study: 65 normotensive subjects (27.4 %) and 172 patients with essential hypertension or primary aldosteronism (72.6 %). All subjects underwent a non-invasive evaluation of retinal arteriolar wall-to-lumen ratio (WLR) by Adaptive Optics. Subjects were re-evaluated after an average follow-up time of 4.55 years in order to assess the occurrence of clinical events (non CV and/or CV death or events). RESULTS Fifty-four events occurred in the study population:26 were cardio-cerebrovascular events (ischemic or hemorragic stroke, atrial fibrillation, heart failure, coronary artery disease, peripheral artery disease, cardiac valvular disease) while the remaining were deaths for any cause, or neoplastic diseases. Subjects with events were older and had a WLR of retinal arterioles significantly greater than those without events. The event-free survival was significantly worse in those with a baseline WLR above the median value of the population (0.28) according to Kaplan-Mayer survival curves and multivariate analysis (Cox's proportional hazard model). The evidence was confirmed after restricting the analysis to CV events. CONCLUSIONS Structural alterations of retinal arterioles evaluated by Adaptive Optics may predict total and CV events.
Collapse
Affiliation(s)
- Carolina De Ciuceis
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia 25123, Italy.
| | - Claudia Agabiti Rosei
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia 25123, Italy
| | - Paolo Malerba
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia 25123, Italy
| | - Claudia Rossini
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia 25123, Italy
| | - Matteo Nardin
- Third Division of Medicine, ASST Spedali Civili di Brescia, Italy
| | - Giulia Chiarini
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia 25123, Italy
| | - Francesca Famà
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia 25123, Italy
| | - Matteo Lemoli
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia 25123, Italy
| | - Mattia Baresi
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia 25123, Italy
| | - Alina Petelca
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia 25123, Italy
| | - Chiara Bortoluzzi
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia 25123, Italy
| | - Enzo Porteri
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia 25123, Italy
| | - Massimo Salvetti
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia 25123, Italy
| | - Maria Lorenza Muiesan
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia 25123, Italy
| | - Enrico Agabiti Rosei
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia 25123, Italy
| | - Damiano Rizzoni
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia 25123, Italy; Division of Medicine, ASST Spedali Civili di Brescia, Montichiari, Brescia, Italy
| |
Collapse
|
23
|
Żmijewska MA, Wawrzyniak ZM, Janiszewski M, Zaleska-Żmijewska A. Retinal Microcirculation Measurements in Response to Endurance Exercises Analysed by Adaptive Optics Retinal Camera. Diagnostics (Basel) 2024; 14:710. [PMID: 38611623 PMCID: PMC11012106 DOI: 10.3390/diagnostics14070710] [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: 03/05/2024] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
This study aimed to precisely investigate the effects of intensive physical exercise on retinal microvascular regulation in healthy volunteers through adaptive optics retinal camera (AO) measurement. We included healthy volunteers (11 men and 14 women) aged 20.6 ± 0.9. The heart rate (HR) and systolic and diastolic blood pressures (SBP, DBP) were recorded before and after a submaximal physical exertion of continuously riding a training ergometer. The superior temporal retinal artery measurements were captured using the AO-rtx1TM (Imagine Eyes, Orsay, France) without pupil dilation. We compared measures of vessel diameter (VD), lumen diameter (LD), two walls (Wall 1, 2), wall-to-lumen ratio (WLR), and wall cross-sectional analysis (WCSA) before and immediately after the cessation of exercise. Cardiovascular parameter results: After exercise, SBP, DBP, and HR changed significantly from 130.2 ± 13.2 to 159.7 ± 15.6 mm Hg, 81.2 ± 6.3 to 77.1 ± 8.2 mm Hg, and 80.8 ± 16.1 to 175.0 ± 6.2 bpm, respectively (p < 0.002). Retinal microcirculation analysis showed no significant decrease in LD, Wall 1 after exercise: from 96.0 ± 6.8 to 94.9 ± 6.7 (p = 0.258), from 11.0 ± 1.5 to 10.4 ± 1.5 (p = 0.107), respectively, and significant reduction in VD from 118.5 ± 8.3 to 115.9 ± 8.3 (p = 0.047), Wall 2 from 11.5 ± 1.0 to 10.7 ± 1.3 (p = 0.017), WLR from 0.234 ± 0.02 to 0.222 ± 0.010 (p = 0.046), WCSA from 3802.8 ± 577.6 to 3512.3 ± 535.3 (p = 0.016). The AO is a promising technique for investigating the effects of exercise on microcirculation, allowing for the tracking of changes throughout the observation. Intensive dynamic physical exertion increases blood pressure and heart rate and causes the vasoconstriction of small retinal arterioles due to the autoregulation mechanism.
Collapse
Affiliation(s)
- Maria Anna Żmijewska
- Faculty of Medicine, Student Scientific Society “Eye”, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Zbigniew M. Wawrzyniak
- Faculty of Electronics and Information Technology, Warsaw University of Technology, 00-065 Warsaw, Poland
| | - Maciej Janiszewski
- Faculty of Medicine and Dentistry, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Anna Zaleska-Żmijewska
- Department of Ophthalmology, Public Ophthalmic Clinical Hospital (SPKSO), Medical University of Warsaw, 00-576 Warsaw, Poland;
| |
Collapse
|
24
|
Buthelezi LM, Munsamy AJ, Mashige KP. Inflammatory mechanisms contributing to retinal alterations in HIV infection and long-term ART. South Afr J HIV Med 2024; 25:1548. [PMID: 38628910 PMCID: PMC11019112 DOI: 10.4102/sajhivmed.v25i1.1548] [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: 10/30/2023] [Accepted: 01/24/2024] [Indexed: 04/19/2024] Open
Abstract
People living with HIV (PLWH) may face an increased risk of eye complications associated with ageing, chronic inflammation, and the toxicity arising from long-term antiretroviral therapy (ART). This review aims to understand how inflammatory pathways contribute to retinal alterations observed in PLWH on long-term ART. This review was conducted using four electronic database searches, namely Scopus, Hinari, Google Scholar, and PubMed; from 1996 (when ART became available) until January 2022, without language restriction. Sources from clinical trials, meta-analyses, randomised controlled trials, and systematic reviews were used. Dysregulated para-inflammation (chronic inflammation) damages the blood-retina barrier, resulting in the altered retinal immune privilege and leading to the development of retinal and blood vessel changes. There is an interplay between the effects of the disease versus ART. ART causes mitochondrial toxicity, which affects the retinal ganglion cells and retinal pigment epithelium (RPE) due to oxidative stress. Infection by HIV also affects retinal microglia, which contributes to RPE damage. Both of these mechanisms affect the blood vessels. Assessing the integrity of the inner and outer blood-retina barrier is a pivotal point in pinpointing the pathogenesis of inner retinal alterations. Optical coherence tomography is a valuable tool to assess these changes. There is a paucity of research to understand how these structural changes may affect visual function, such as contrast sensitivity and colour vision.
Collapse
Affiliation(s)
- Lungile M Buthelezi
- Department of Optometry, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Alvin J Munsamy
- Department of Optometry, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Khathutshelo P Mashige
- Department of Optometry, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
25
|
Richer E, Solano MM, Cheriet F, Lesk MR, Costantino S. Denoising OCT videos based on temporal redundancy. Sci Rep 2024; 14:6605. [PMID: 38503804 PMCID: PMC10951312 DOI: 10.1038/s41598-024-56935-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 03/12/2024] [Indexed: 03/21/2024] Open
Abstract
The identification of eye diseases and their progression often relies on a clear visualization of the anatomy and on different metrics extracted from Optical Coherence Tomography (OCT) B-scans. However, speckle noise hinders the quality of rapid OCT imaging, hampering the extraction and reliability of biomarkers that require time series. By synchronizing the acquisition of OCT images with the timing of the cardiac pulse, we transform a low-quality OCT video into a clear version by phase-wrapping each frame to the heart pulsation and averaging frames that correspond to the same instant in the cardiac cycle. Here, we compare the performance of our one-cycle denoising strategy with a deep-learning architecture, Noise2Noise, as well as classical denoising methods such as BM3D and Non-Local Means (NLM). We systematically analyze different image quality descriptors as well as region-specific metrics to assess the denoising performance based on the anatomy of the eye. The one-cycle method achieves the highest denoising performance, increases image quality and preserves the high-resolution structures within the eye tissues. The proposed workflow can be readily implemented in a clinical setting.
Collapse
Affiliation(s)
- Emmanuelle Richer
- Department of Computer Engineering and Software Engineering, École Polytechnique de Montréal, Montreal, QC, H3T 1J4, Canada
- Maisonneuve-Rosemont Hospital Research Center, Montreal, QC, H1T 2M4, Canada
| | - Marissé Masís Solano
- Maisonneuve-Rosemont Hospital Research Center, Montreal, QC, H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC, H3T 1P1, Canada
| | - Farida Cheriet
- Department of Computer Engineering and Software Engineering, École Polytechnique de Montréal, Montreal, QC, H3T 1J4, Canada
| | - Mark R Lesk
- Maisonneuve-Rosemont Hospital Research Center, Montreal, QC, H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC, H3T 1P1, Canada
| | - Santiago Costantino
- Maisonneuve-Rosemont Hospital Research Center, Montreal, QC, H1T 2M4, Canada.
- Department of Ophthalmology, Université de Montréal, Montreal, QC, H3T 1P1, Canada.
| |
Collapse
|
26
|
Hervella ÁS, Ramos L, Rouco J, Novo J, Ortega M. Explainable artificial intelligence for the automated assessment of the retinal vascular tortuosity. Med Biol Eng Comput 2024; 62:865-881. [PMID: 38060101 PMCID: PMC10881731 DOI: 10.1007/s11517-023-02978-w] [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: 07/11/2023] [Accepted: 11/22/2023] [Indexed: 12/08/2023]
Abstract
Retinal vascular tortuosity is an excessive bending and twisting of the blood vessels in the retina that is associated with numerous health conditions. We propose a novel methodology for the automated assessment of the retinal vascular tortuosity from color fundus images. Our methodology takes into consideration several anatomical factors to weigh the importance of each individual blood vessel. First, we use deep neural networks to produce a robust extraction of the different anatomical structures. Then, the weighting coefficients that are required for the integration of the different anatomical factors are adjusted using evolutionary computation. Finally, the proposed methodology also provides visual representations that explain the contribution of each individual blood vessel to the predicted tortuosity, hence allowing us to understand the decisions of the model. We validate our proposal in a dataset of color fundus images providing a consensus ground truth as well as the annotations of five clinical experts. Our proposal outperforms previous automated methods and offers a performance that is comparable to that of the clinical experts. Therefore, our methodology demonstrates to be a viable alternative for the assessment of the retinal vascular tortuosity. This could facilitate the use of this biomarker in clinical practice and medical research.
Collapse
Affiliation(s)
- Álvaro S Hervella
- Centro de Investigación CITIC, Universidade da Coruña, A Coruña, Spain.
- Grupo VARPA, Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña, A Coruña, Spain.
| | - Lucía Ramos
- Centro de Investigación CITIC, Universidade da Coruña, A Coruña, Spain
- Grupo VARPA, Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña, A Coruña, Spain
| | - José Rouco
- Centro de Investigación CITIC, Universidade da Coruña, A Coruña, Spain
- Grupo VARPA, Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña, A Coruña, Spain
| | - Jorge Novo
- Centro de Investigación CITIC, Universidade da Coruña, A Coruña, Spain
- Grupo VARPA, Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña, A Coruña, Spain
| | - Marcos Ortega
- Centro de Investigación CITIC, Universidade da Coruña, A Coruña, Spain
- Grupo VARPA, Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña, A Coruña, Spain
| |
Collapse
|
27
|
Asiello JD, Kelley CM, Cannone K, McInnis L, Begin A, Dittrich M, Donovan E, Beatty K, Lam N, Taubert A, Wong J. Addressing the functional needs of left ventricular assist device candidates: Development and feasibility of an occupational therapy pre-operative evaluation. Heart Lung 2024; 64:198-207. [PMID: 38301417 DOI: 10.1016/j.hrtlng.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND Everyday living with a left ventricular assist device (LVAD) is complex, particularly for people with physical or cognitive impairments or limited social supports. There is a need for standardized pre-operative functional evaluations. OBJECTIVES Our objectives were to describe a pre-operative occupational therapy (OT) evaluation for LVAD candidates, assess its feasibility in routine care, and characterize functional needs. METHODS We retrospectively reviewed electronic medical records of pre-operative OT consultations for LVAD candidates over four years (n = 209). Occupational profile, vision, and sensation were operationalized from documentation narratives. Daily functioning was measured with Activity Measure for Post-Acute Care, grip strength with dynamometer, cognition with Montreal Cognitive Assessment and Allen Cognitive Level Screen-5, and LVAD self-management with a performance-based ordinal scale. RESULTS 89.5 % of consultations were completed, averaging 61.2 min (n = 187): 79.1 % (148/187) inpatient and 20.9 % (39/187) outpatient. Patients completed 87.7 % (164/187) to 100.0 % (187/187) of evaluation components. 21.9 % (41/187) of candidates lived alone. 6.4 % (12/187) and 7.0 % (13/185) had visual and sensory dysfunction. 57.4 % were independent with daily activities (104/181). 17.7 % (32/181) had impaired grip strength. 69.3 % (124/179) had impaired cognition, 29.7 % (51/172) with impaired functional cognition for everyday activities. 88.4 % (145/164) required physical or cueing assistance while practicing LVAD batteries management. OTs interpreted that 20.9 % (39/187) would likely require 24/7 post-operative support with LVAD self-care. CONCLUSION Pre-operative OT evaluations were feasible and emphasized complex functional needs. Assessing LVAD self-care abilities may inform candidacy and facilitate early interventions to optimize functioning. OT should be consulted within interprofessional teams for all LVAD candidates.
Collapse
Affiliation(s)
- Jessica D Asiello
- Department of Occupational Therapy, MGH Institute of Health Professions, Charlestown Navy Yard, 36 1st Avenue, Boston, MA 02129, United States; Occupational Therapy Services, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, United States.
| | - Christina M Kelley
- Occupational Therapy Services, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, United States
| | - Kellie Cannone
- Department of Occupational Therapy, MGH Institute of Health Professions, Charlestown Navy Yard, 36 1st Avenue, Boston, MA 02129, United States
| | - Lauren McInnis
- Department of Occupational Therapy, MGH Institute of Health Professions, Charlestown Navy Yard, 36 1st Avenue, Boston, MA 02129, United States
| | - Abigail Begin
- Occupational Therapy Services, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, United States
| | - Marissa Dittrich
- Occupational Therapy Services, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, United States
| | - Erin Donovan
- Occupational Therapy Services, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, United States
| | - Kimberly Beatty
- Occupational Therapy Services, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, United States
| | - Nicole Lam
- Occupational Therapy Services, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, United States
| | - Alyssa Taubert
- Occupational Therapy Services, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, United States
| | - John Wong
- Department of Occupational Therapy, MGH Institute of Health Professions, Charlestown Navy Yard, 36 1st Avenue, Boston, MA 02129, United States; School of Nursing, MGH Institute of Health Professions, Charlestown Navy Yard, 36 1st Avenue, Boston, MA 02129, United States
| |
Collapse
|
28
|
Rossi VA, Laptseva N, Nebunu D, Haider T, Nägele MP, Ruschitzka F, Sudano I, Flammer AJ. Impaired retinal micro-vascular function in patients with atrial fibrillation. Int J Cardiol 2024; 398:131592. [PMID: 37979794 DOI: 10.1016/j.ijcard.2023.131592] [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: 10/02/2023] [Revised: 11/01/2023] [Accepted: 11/14/2023] [Indexed: 11/20/2023]
Abstract
BACKGROUND Cardiovascular (CV) risk factors and CV diseases, in particular heart failure, are strongly associated with impaired microvascular retinal endothelial function. Whether atrial fibrillation (AF) contributes to vascular dysfunction is not clear. Therefore, the aim of this study was to investigate the impact of AF on retinal microvascular function. METHODS In this study, vascular function was measured non-invasively with flicker-light induced vasodilatation of retinal arterioles (FIDart%). Patients with a history of AF and risk factors for heart failure (HF) or heart failure (n = 69; age 67.9 ± 9.2 years, 71% male, 35% HFrEF, 56% paroxysmal, 25% persistent, 19% permanent AF), as well as age, sex and ejection fraction matched patients with absent history of AF (n = 66; age 63.4 ± 10.6 years, 67% male, 47% HFrEF) were included. Patients with AF were further divided into those with paroxysmal AF (in sinus rhythm - AFSR: n = 38, age 71.4 ± 9.2, 73% male), and those with AF at the time of the study visit. RESULTS Retinal microvascular function was impaired in patients with AF compared to patients without AF (FIDart% 1.1% [0.3-2.8] vs. 2.7% [1.3-5.1], p < 0.001). Patients currently in AF have poorer retinal microvascular function (FIDart% 0.8% [0.1-1.9) compared to patients with a history of AF but currently in SR at the time of retinal function measurement (1.5% [0.6-4.9] p = 0.017). In patients with AF, impaired retinal vascular function was independently associated with larger left atrial volume (mean 49.8 ± 18.4), even after correction for confounding factors in different models (SCR = -0. 251 to -0.256, p = 0.035-0.01). CONCLUSIONS AF in patients with heart failure is associated with impaired vascular function, even if currently in sinus rhythm. The association of retinal microvascular dysfunction with left atrial volume, a surrogate for elevated cardiac filling pressures, may further highlight the important interplay between the vasculature and elevated filling pressures in the development of AF.
Collapse
Affiliation(s)
| | | | - Delia Nebunu
- University Heart Center, University Hospital of Zurich, Switzerland
| | - Thomas Haider
- University Heart Center, University Hospital of Zurich, Switzerland
| | | | - Frank Ruschitzka
- University Heart Center, University Hospital of Zurich, Switzerland; Center for Translational and Experimental Cardiology, Schlieren, Switzerland; University of Zurich, Zurich, Switzerland
| | - Isabella Sudano
- University Heart Center, University Hospital of Zurich, Switzerland; Center for Translational and Experimental Cardiology, Schlieren, Switzerland; University of Zurich, Zurich, Switzerland
| | - Andreas J Flammer
- University Heart Center, University Hospital of Zurich, Switzerland; Center for Translational and Experimental Cardiology, Schlieren, Switzerland; University of Zurich, Zurich, Switzerland.
| |
Collapse
|
29
|
Piantoni S, Regola F, Angeli F, Caproli A, Trovati A, Tomasi C, Chiarini G, Rossini C, Rosei CA, De Ciuceis C, Franceschini F, Muiesan ML, Rizzoni D, Airò P. Retinal microvascular alterations in patients with active rheumatoid arthritis without cardiovascular risk factors: the potential effects of T cell co-stimulation blockade. Front Med (Lausanne) 2024; 11:1247024. [PMID: 38420362 PMCID: PMC10899475 DOI: 10.3389/fmed.2024.1247024] [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: 06/25/2023] [Accepted: 01/05/2024] [Indexed: 03/02/2024] Open
Abstract
Background The evaluation of microvascular alterations might provide clinically useful information for patients with an increased cardiovascular (CV) risk, such as those with rheumatoid arthritis (RA), being the small artery remodeling the earliest form of target organ damage in primary CV diseases, such as arterial hypertension. The evaluation of retinal arterioles is a non-invasive technique aimed to identify an early microvascular damage, represented by the increase of the wall-to-lumen ratio (WLR) index. Abatacept (ABA), a T-cell co-stimulator blocker, is used to treat RA. A CV protective action was hypothesized for its peculiar mechanism of action in the modulation of T-cells, potentially involved in the pathogenesis of CV comorbidity. The study aimed to non-invasively investigate morphological characteristics of retinal arterioles in a cohort of RA patients treated with ABA. Materials and methods Seventeen RA patients [median (25th-75thpercentile) age = 58 (48-64) years, baseline 28-joint Disease Activity Score DAS28-C-reactive protein (DAS28-CRP) = 4.4 (3.9-4.6), body mass index (BMI) = 24.2 (23.4-26) kg/m2, rheumatoid factor positive:52.9%, anti-citrullinated peptide autoantibodies positive:76.5%] without known CV risk factors (arterial hypertension, diabetes, hypercholesterolemia, previous CV events, smoking) were evaluated by the adaptive optics imaging system of retinal arterioles before and every 6 months of therapy with ABA (T0, T6 and T12). Office blood pressure evaluation, 24-h ambulatory blood pressure monitoring and tissue-doppler echocardiography were also performed. Results A progressive significant reduction of the WLR of retinal arterioles was observed [T0 = 0.28 (0.25-0.30), T6 = 0.27 (0.24-0.31), T12 = 0.23 (0.23-0.26); p T0 vs. T6 = 0.414; p T6 vs. T12 = 0.02; p T0 vs. T12 = 0.009], without significant variations in other parameters. The T0-T12 reduction of WLR was correlated with that of DAS28-CRP (r:0.789; p = 0.005). Moreover, a significant reduction of diastolic office blood pressure and a trend for reduction of daily pressure measured by ambulatory monitoring were observed. Conclusion In a cohort of RA patients without known CV risk factors, a reduction of retinal microvascular alterations was demonstrated after treatment for 12 months with ABA, in parallel with the reduction of disease activity. These results might suggest the possibility of microvascular abnormalities regression induced by the immune system modulation.
Collapse
Affiliation(s)
- Silvia Piantoni
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Francesca Regola
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Fabrizio Angeli
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alessia Caproli
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Annalisa Trovati
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Cesare Tomasi
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Giulia Chiarini
- Internal Medicine Unit, ASST Spedali Civili, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Claudia Rossini
- Internal Medicine Unit, ASST Spedali Civili, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Claudia Agabiti Rosei
- Internal Medicine Unit, ASST Spedali Civili, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Carolina De Ciuceis
- Internal Medicine Unit, ASST Spedali Civili, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Franco Franceschini
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Maria Lorenza Muiesan
- Internal Medicine Unit, ASST Spedali Civili, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Damiano Rizzoni
- Internal Medicine Unit, ASST Spedali Civili, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Paolo Airò
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
30
|
Huang Y, Cheung CY, Li D, Tham YC, Sheng B, Cheng CY, Wang YX, Wong TY. AI-integrated ocular imaging for predicting cardiovascular disease: advancements and future outlook. Eye (Lond) 2024; 38:464-472. [PMID: 37709926 PMCID: PMC10858189 DOI: 10.1038/s41433-023-02724-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 07/26/2023] [Accepted: 08/25/2023] [Indexed: 09/16/2023] Open
Abstract
Cardiovascular disease (CVD) remains the leading cause of death worldwide. Assessing of CVD risk plays an essential role in identifying individuals at higher risk and enables the implementation of targeted intervention strategies, leading to improved CVD prevalence reduction and patient survival rates. The ocular vasculature, particularly the retinal vasculature, has emerged as a potential means for CVD risk stratification due to its anatomical similarities and physiological characteristics shared with other vital organs, such as the brain and heart. The integration of artificial intelligence (AI) into ocular imaging has the potential to overcome limitations associated with traditional semi-automated image analysis, including inefficiency and manual measurement errors. Furthermore, AI techniques may uncover novel and subtle features that contribute to the identification of ocular biomarkers associated with CVD. This review provides a comprehensive overview of advancements made in AI-based ocular image analysis for predicting CVD, including the prediction of CVD risk factors, the replacement of traditional CVD biomarkers (e.g., CT-scan measured coronary artery calcium score), and the prediction of symptomatic CVD events. The review covers a range of ocular imaging modalities, including colour fundus photography, optical coherence tomography, and optical coherence tomography angiography, and other types of images like external eye images. Additionally, the review addresses the current limitations of AI research in this field and discusses the challenges associated with translating AI algorithms into clinical practice.
Collapse
Affiliation(s)
- Yu Huang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Carol Y Cheung
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Dawei Li
- College of Future Technology, Peking University, Beijing, China
| | - Yih Chung Tham
- Centre for Innovation and Precision Eye Health and Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Center, Singapore, Singapore
| | - Bin Sheng
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ching Yu Cheng
- Centre for Innovation and Precision Eye Health and Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Center, Singapore, Singapore
| | - Ya Xing Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Center, Singapore, Singapore.
- Tsinghua Medicine, Tsinghua University, Beijing, China.
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Beijing, China.
| |
Collapse
|
31
|
Sideri AM, Mitsopoulou D, Kandarakis SA, Katsimpris A, Kanakis M, Karamaounas A, Brouzas D, Petrou P, Papakonstantinou E, Droutsas K, Giannopoulos G, Georgalas I. Optical Coherence Tomography Angiography Changes in Patients Diagnosed With Acute Coronary Syndrome: A Systematic Review and Meta-Analysis. Cureus 2024; 16:e54121. [PMID: 38487148 PMCID: PMC10939045 DOI: 10.7759/cureus.54121] [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] [Accepted: 02/13/2024] [Indexed: 03/17/2024] Open
Abstract
We conducted a systematic review and meta-analysis to assess the association between optical coherence tomography angiography (OCTA) parameters and acute coronary syndrome (ACS). Two independent reviewers searched the electronic databases (MEDLINE (Medical Literature Analysis and Retrieval System Online), Scopus, Embase (Excerpta Medica Database), Cochrane Library, ClinicalTrials.gov, and World Health Organization International Clinical Trials Registry Platform) from inception until April 2023. According to the inclusion criteria of this review, eligible were observational studies, randomized control trials, and registry/database studies that included the eyes of adult ACS patients and assessed OCTA parameters within the macula. The pooled standardized mean differences (SMD) between patients diagnosed with ACS and healthy controls with a confidence interval (CI) of 95% were calculated using the Hartung-Knapp-Sidik-Jonkman random-effects method. The heterogeneity was assessed by I2 and the Cochran Q and a random effects model was applied. Seven studies were eligible and included in our systematic review (n = 898), of which three were included in the meta-analysis (n = 341). The pooled SMD in the superficial vascular plexus (SVP), deep vascular plexus (DVP), and foveal avascular zone (FAZ) were -0.46 (95% CI: -0.94 to 0.01, p = 0.05, I2 = 0%, three studies), -0.10 (95% CI: -3.20 to 3.00, p = 0.75, I2 = 67%, two studies), and 0.43 (95% CI: -1.22 to 2.09, p = 0.38, I2 = 92%, three studies), respectively. Our findings suggest that there are no differences in OCTA metrics between ACS patients and healthy individuals.
Collapse
Affiliation(s)
- Anna Maria Sideri
- First Department of Ophthalmology, G. Gennimatas Hospital, National and Kapodistrian University of Athens, Athens, GRC
| | - Dimitra Mitsopoulou
- First Department of Ophthalmology, G. Gennimatas Hospital, National and Kapodistrian University of Athens, Athens, GRC
| | - Stylianos A Kandarakis
- First Department of Ophthalmology, G. Gennimatas Hospital, National and Kapodistrian University of Athens, Athens, GRC
| | | | - Menelaos Kanakis
- Ophthalmology, University Eye Clinic, Rion University Hospital, University of Patras, Patras, GRC
| | - Aristotelis Karamaounas
- First Department of Ophthalmology, G. Gennimatas Hospital, National and Kapodistrian University of Athens, Athens, GRC
| | - Dimitrios Brouzas
- Ophthalmology, National and Kapodistrian University of Athens, Athens, GRC
| | - Petros Petrou
- First Department of Ophthalmology, G. Gennimatas Hospital, National and Kapodistrian University of Athens, Athens, GRC
| | - Evangelia Papakonstantinou
- First Department of Ophthalmology, G. Gennimatas Hospital, National and Kapodistrian University of Athens, Athens, GRC
| | - Konstantinos Droutsas
- First Department of Ophthalmology, G. Gennimatas Hospital, National and Kapodistrian University of Athens, Athens, GRC
| | - Georgios Giannopoulos
- Third Department of Cardiology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, GRC
| | - Ilias Georgalas
- First Department of Ophthalmology, G. Gennimatas Hospital, National and Kapodistrian University of Athens, Athens, GRC
| |
Collapse
|
32
|
Ramesh PV, Morya AK, Aradhya AK, Pannerselvam P, Gopalakrishnan ST, Ramesh SV, Devadas AK, Krishna N. Unveiling the silent link: Normal-tension glaucoma's enigmatic bond with cardiac blood flow. World J Cardiol 2024; 16:10-15. [PMID: 38313391 PMCID: PMC10835469 DOI: 10.4330/wjc.v16.i1.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/14/2023] [Accepted: 01/03/2024] [Indexed: 01/19/2024] Open
Abstract
This comprehensive review embarks on a captivating journey into the complex relationship between cardiology and normal-tension glaucoma (NTG), a condition that continues to baffle clinicians and researchers alike. NTG, characterized by optic nerve damage and visual field loss despite normal intraocular pressure, has long puzzled clinicians. One emerging perspective suggests that alterations in ocular blood flow, particularly within the optic nerve head, may play a pivotal role in its pathogenesis. While NTG shares commonalities with its high-tension counterpart, its unique pathogenesis and potential ties to cardiovascular health make it a fascinating subject of exploration. It navigates through the complex web of vascular dysregulation, blood pressure and perfusion pressure, neurovascular coupling, and oxidative stress, seeking to uncover the hidden threads that tie the heart and eyes together in NTG. This review explores into the intricate mechanisms connecting cardiovascular factors to NTG, shedding light on how cardiac dynamics can influence ocular health, particularly in cases where intraocular pressure remains within the normal range. NTG's enigmatic nature, often characterized by seemingly contradictory risk factors and clinical profiles, underscores the need for a holistic approach to patient care. Drawing parallels to cardiac health, we examine into the shared vascular terrain connecting the heart and the eyes. Cardiovascular factors, including systemic blood flow, endothelial dysfunction, and microcirculatory anomalies, may exert a profound influence on ocular perfusion, impacting the delicate balance within the optic nerve head. By elucidating the subtle clues and potential associations between cardiology and NTG, this review invites clinicians to consider a broader perspective in their evaluation and management of this elusive condition. As the understanding of these connections evolves, so too may the prospects for early diagnosis and tailored interventions, ultimately enhancing the quality of life for those living with NTG.
Collapse
Affiliation(s)
- Prasanna Venkatesh Ramesh
- Department of Glaucoma and Research, Mahathma Eye Hospital Private Limited, Trichy 620017, Tamil Nadu, India.
| | - Arvind Kumar Morya
- Department of Ophthalmology, All India Institute of Medical Sciences, Hyderabad 508126, Telangana, India
| | - Ajanya K Aradhya
- Department of Optometry and Visual Science, Mahathma Eye Hospital Private Limited, Trichy 620017, Tamil Nadu, India
| | | | | | - Shruthy Vaishali Ramesh
- Department of Cataract and Refractive Surgery, Mahathma Eye Hospital Private Limited, Trichy 620017, Tamil Nadu, India
| | - Aji Kunnath Devadas
- Department of Optometry and Visual Science, Mahathma Eye Hospital Private Limited, Trichy 620017, Tamil Nadu, India
| | - Navaneeth Krishna
- Department of Optometry and Visual Science, Mahathma Eye Hospital Private Limited, Trichy 620017, Tamil Nadu, India
| |
Collapse
|
33
|
Bernstein IA, Fisher AC, Singh K, Wang SY. The Association Between Frailty and Visual Field Loss in US Adults. Am J Ophthalmol 2024; 257:38-45. [PMID: 37714282 PMCID: PMC10841077 DOI: 10.1016/j.ajo.2023.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/16/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023]
Abstract
PURPOSE To describe the association between visual field loss and frailty in a nationally representative cohort of US adults. DESIGN Retrospective cross-sectional study. METHODS The cohort included adults 40 years or older with complete eye examination data from the 2005-2006 and 2007-2008 National Health and Nutrition Examination Surveys (NHANES). Visual field loss (VFL) was determined by frequency doubling technology and a 2-2-1 algorithm. A 36-item deficit accumulation-based frailty index was used to divide subjects into 4 categories of increasing frailty severity. RESULTS Of the 4897 participants, 4402 (93.2%) had no VFL, 301 (4.1%) had unilateral VFL, and 194 (2.73%) had bilateral VFL. Within the sample, 2 subjects197 (53.1%) were categorized as non-frail, 1659 (31.3%) as vulnerable, 732 (11.3%) as mildly frail, and 312 (4.3%) as most frail. In multivariable models adjusted for demographics, visual acuity, and history of cataract surgery, subjects with unilateral VFL had higher adjusted odds of being in a more frail category (adjusted odds ratio [aOR], 2.07; 95% CI, 1.42-3.02) than subjects without VFL. Subjects with bilateral VFL also had higher odds of a more frail category compared to subjects without VFL (aOR, 1.74; 95% CI, 1.20-2.52). CONCLUSIONS In the 2005-2008 NHANES adult population, VFL is associated with higher odds of frailty, independent of central visual acuity loss. Frail individuals may be more susceptible to diseases that can cause VFL, and/or VFL may predispose to frailty. Additional studies are needed to determine the directionality of this relationship and to assess potential interventions.
Collapse
Affiliation(s)
- Isaac A Bernstein
- From the Department of Ophthalmology, Byers Eye Institute, Stanford University, Stanford, California, USA
| | - Ann Caroline Fisher
- From the Department of Ophthalmology, Byers Eye Institute, Stanford University, Stanford, California, USA
| | - Kuldev Singh
- From the Department of Ophthalmology, Byers Eye Institute, Stanford University, Stanford, California, USA
| | - Sophia Y Wang
- From the Department of Ophthalmology, Byers Eye Institute, Stanford University, Stanford, California, USA.
| |
Collapse
|
34
|
Şimdivar GHN, İncekalan TK, Gündüz A. Evaluation of retinal and peripapillary vessel density and subfoveal choroidal thickness changes in patients undergoing cardiopulmonary bypass: An OCTA study. Indian J Ophthalmol 2024; 72:S119-S124. [PMID: 38131553 PMCID: PMC10833158 DOI: 10.4103/ijo.ijo_2800_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/16/2022] [Accepted: 09/22/2023] [Indexed: 12/23/2023] Open
Abstract
PURPOSE We aimed to investigate hemodynamic changes in the retina, peripapillary area, and choroid in patients undergoing coronary artery bypass grafting (CABG). METHODS This prospective case series included 35 eyes of 35 patients. Vessel density (VD) of superficial capillary plexus, deep capillary plexus, radial peripapillary capillary, foveal avascular zone area, and subfoveal choroidal thickness changes were evaluated using optical coherence tomography angiography (OCTA) 1 day before and 2 weeks after surgery. The correlation of these parameters with aortic cross-clamp (ACC) duration and cardiopulmonary bypass (CPB) duration was evaluated. RESULTS There were no differences in the OCTA measurements of the patients after CABG compared to before surgery (P > 0.05). Inside-disc VD showed a negative correlation with both ACC (P = 0.036) and CPB duration (P = 0.048); peripapillary inferior hernia showed a negative correlation with both ACC (P = 0.042) and CPB duration (P = 0.027). Another negative correlation was observed between mean peripapillary VD and CPB duration (P = 0.039). CONCLUSION The CPB procedure had no permanent effect on retinal choroid and optic disc blood flow postoperatively. Prolonged ACC and CPB times seem to be important in terms of optic nerve blood flow. Clinicians should be alert for postoperative ocular complications after long surgeries.
Collapse
Affiliation(s)
- Göksu Hande Naz Şimdivar
- Department of Ophthalmology, University of Health Sciences Adana City Training and Research Hospital, Adana, Turkey
| | - Tuğba Kurumoğlu İncekalan
- Department of Ophthalmology, University of Health Sciences Adana City Training and Research Hospital, Adana, Turkey
| | - Akif Gündüz
- Department of Cardiovascular Surgery, University of Health Sciences Adana City Training and Research Hospital, Adana, Turkey
| |
Collapse
|
35
|
Müller C, Hauser C, Carrard J, Gugleta K, Hinrichs T, Schmidt-Trucksäss A, Hanssen H, Streese L. Effects of high-intensity interval training on retinal vessel diameters and oxygen saturation in patients with hypertension: A cross-sectional and randomized controlled trial. Microvasc Res 2024; 151:104616. [PMID: 37890716 DOI: 10.1016/j.mvr.2023.104616] [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: 06/20/2023] [Revised: 10/07/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023]
Abstract
INTRODUCTION Arterial hypertension is a global healthcare burden that affects macrovascular and microvascular structure and function and can promote vascular end-organ damage. This study aimed 1) to evaluate differences in microvascular health between normotensive individuals and patients with arterial hypertension and 2) to assess the effects of short-term high-intensity interval training (HIIT) on microvascular health in the subgroup with arterial hypertension as add-on treatment to antihypertensive medication. METHODS In the cross-sectional part, central retinal arteriolar (CRAE) and venular diameter equivalent (CRVE), arteriolar-to-venular diameter ratio (AVR), and retinal oxygen saturation (O2-saturation) were investigated in 19 normotensive healthy controls (mean age 56 ± 7 years) and 41 patients with arterial hypertension (mean age 59 ± 7 years). In the subsequent randomized controlled trial (RCT), patients with arterial hypertension were randomized to an intervention group (HIIT 3×/week) or a control group that received standard physical activity recommendations after baseline assessment. Assessments of retinal vessel biomarkers and patients` characteristics were repeated after the intervention period of 8 weeks. RESULTS In the cross-sectional part, individuals with normal blood pressure (BP) showed lower body mass index (BMI), body fat, 24 h systolic and diastolic BP, higher peak oxygen uptake, wider CRAE (174 ± 17 μm vs. 161 ± 17 μm, p = 0.009), and higher AVR (0.84 ± 0.05 vs. 0.79 ± 0.05, p = 0.003) compared to patients with hypertension. In the RCT, patients with arterial hypertension showed reduced BMI and fasting glucose levels after HIIT and control condition. In addition, the intervention group reduced body fat percentage (27.0 ± 5.5 vs. 25.8 ± 6.1, p = 0.023) and increased peak oxygen uptake (33.3 ± 5.7 vs. 36.7 ± 5.1, p < 0.001). No changes in BP were found in either group. The intervention group showed narrower CRVE (β -4.8 [95 % CI, -8.85, -0.81] p = 0.020) and higher AVR (0.03 [0.01, 0.04] p < 0.001) after eight weeks of HIIT compared to the control group. No statistically significant changes in retinal O2-saturation were found in either group. CONCLUSION Short-term HIIT proved to be an effective treatment to ameliorate hypertension-induced retinal microvascular abnormalities in patients with hypertension. Retinal vessel diameters may prove to be a sensitive biomarker to quantify treatment efficacy at the microvascular level, at the earliest possible stage in patients with hypertension.
Collapse
Affiliation(s)
- Cédric Müller
- Department of Sport, Exercise and Health, Medical Faculty, University of Basel, Basel, Switzerland
| | - Christoph Hauser
- Department of Sport, Exercise and Health, Medical Faculty, University of Basel, Basel, Switzerland
| | - Justin Carrard
- Department of Sport, Exercise and Health, Medical Faculty, University of Basel, Basel, Switzerland
| | - Konstantin Gugleta
- Department of Ophthalmology, University Hospital Basel, Basel, Switzerland
| | - Timo Hinrichs
- Department of Sport, Exercise and Health, Medical Faculty, University of Basel, Basel, Switzerland
| | - Arno Schmidt-Trucksäss
- Department of Sport, Exercise and Health, Medical Faculty, University of Basel, Basel, Switzerland; Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Henner Hanssen
- Department of Sport, Exercise and Health, Medical Faculty, University of Basel, Basel, Switzerland
| | - Lukas Streese
- Department of Sport, Exercise and Health, Medical Faculty, University of Basel, Basel, Switzerland; Faculty of Health Care, Niederrhein University of Applied Sciences, Krefeld, Germany.
| |
Collapse
|
36
|
Maugana VF, Kruger R, Kruger HS, Hanssen H, Smith W. Food intake in South African children and retinal microvascular health: The ExAMIN Youth SA study. Nutr Metab Cardiovasc Dis 2024; 34:188-197. [PMID: 37798229 DOI: 10.1016/j.numecd.2023.08.013] [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: 01/23/2023] [Revised: 08/15/2023] [Accepted: 08/19/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND AND AIMS Retinal arteriolar narrowing and venular widening are associated with increased cardiovascular risk, even at young ages. Whether diet contributes to early microvascular changes in children is not widely explored. We explored the associations of frequency of healthy and unhealthy food group intake with retinal vessel calibers in black and white children. METHODS AND RESULTS This study included school-aged (5-9 years) black (N = 433, 7.46 ± 0.98 years), and white (N = 403, 7.43 ± 0.82 years) children. Anthropometric and blood pressure measurements were taken, along with retinal vessel calibers (central retinal arteriolar (CRAE) and venular (CRVE) equivalents). Frequencies of food group intake were assessed using a food-frequency questionnaire. A factor analysis was performed to describe food group patters. Independent associations between retinal vessel calibers and frequencies of food group intake and food group patters were explored. In black children, cookies, cakes, and biscuits were associated with narrower arterioles (p < 0.05). In white children, cold sweetened beverages were associated with narrower arterioles (p = 0.02), whereas salty snacks were associated with narrower arterioles (p = 0.01) and wider venules (p < 0.05). Fruits were positively associated with CRAE (p = 0.03) in white children only. CONCLUSION A higher frequency of unhealthy food group consumption was associated with retinal arteriolar narrowing and venular widening in both black and white children. However, fruit intake was shown beneficial for retinal microvascular health in white children only. Our findings may highlight the importance of promoting healthy eating patterns from early childhood which may reduce the risk of premature cardiovascular disease development.
Collapse
Affiliation(s)
- Vuledzani F Maugana
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa
| | - Ruan Kruger
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa; SAMRC Extramural Unit for Hypertension and Cardiovascular Disease, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Herculina S Kruger
- SAMRC Extramural Unit for Hypertension and Cardiovascular Disease, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa; Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| | - Henner Hanssen
- Sports and Exercise Medicine, Department of Sport, Exercise and Health, University of Basel, Switzerland
| | - Wayne Smith
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa; SAMRC Extramural Unit for Hypertension and Cardiovascular Disease, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa.
| |
Collapse
|
37
|
Li C, Zhu Z, Yuan H, Hu Y, Xue Y, Zhong P, Huang M, Ren Y, Kuang Y, Zeng X, Yu H, Yang X. Association of preoperative retinal microcirculation and perioperative outcomes in patients undergoing congenital cardiac surgery. Orphanet J Rare Dis 2023; 18:385. [PMID: 38066637 PMCID: PMC10704768 DOI: 10.1186/s13023-023-02969-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Microcirculatory dysfunction is associated with increased morbidity and mortality in cardiac surgery patients. This study aimed to investigate the association between preoperative retinal microcirculation evaluated using optical coherence tomography angiography (OCTA) and perioperative outcomes in patients with congenital heart disease (CHD). METHODS This prospective, observational study was performed from May 2017 to January 2021. OCTA was used to automatically quantify the vessel density (VD) of the superficial capillary plexus, deep capillary plexus (DCP), and radial peripapillary capillary (RPC) preoperatively. The primary outcome was excessive postoperative bleeding, defined as bleeding volume > 75th percentile for 24-hour postoperative chest tube output. The secondary outcome was composite adverse outcomes, including one or more operative mortalities, early postoperative complications, and prolonged length of stay. The association between retinal VD and outcomes was assessed using Poisson regression. RESULTS In total, 173 CHD patients who underwent cardiac surgery were included (mean age, 26 years). Among them, 43 (24.9%) and 46 (26.6%) developed excessive postoperative bleeding and composite adverse outcomes, respectively. A lower VD of DCP (odds ratio [OR], 1.24; 95% confidence interval [CI], 1.08-1.43; P = 0.003) was independently associated with excessive postoperative bleeding, and a lower VD of RPC (OR, 1.97; 95% CI, 1.08-3.57; P = 0.027), and DCP (OR, 2.17; 95% CI, 1.08-4.37; P = 0.029) were independently associated with the postoperative composite adverse outcomes. CONCLUSION Preoperative retinal hypoperfusion was independently associated with an increased risk of perioperative adverse outcomes in patients with CHD, suggesting that retinal microcirculation evaluation could provide valuable information about the outcomes of cardiac surgery, thereby aiding physicians in tailoring individualized treatment.
Collapse
Affiliation(s)
- Cong Li
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhuoting Zhu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Haiyun Yuan
- Department of Cardiovascular Surgery, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yijun Hu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yunlian Xue
- Statistics Section, Information Management Department, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Pingting Zhong
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-Sen University, Guangzhou, China
| | - Manqing Huang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yun Ren
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Medical College, Shantou University, Shantou, China
| | - Yu Kuang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiaomin Zeng
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Honghua Yu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China.
| | - Xiaohong Yang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
38
|
Rossi VA, Nebunu D, Haider T, Laptseva N, Naegele MP, Ruschitzka F, Sudano I, Flammer AJ. Diverging role of epicardial adipose tissue across the entire heart failure spectrum. ESC Heart Fail 2023; 10:3419-3429. [PMID: 37697706 PMCID: PMC10682858 DOI: 10.1002/ehf2.14483] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 07/06/2023] [Indexed: 09/13/2023] Open
Abstract
AIMS Epicardial adipose tissue (EAT) is a metabolically highly active tissue modulating numerous pathophysiological processes. The aim of this study was to investigate the association between EAT thickness and endothelial function in patients with heart failure (HF) across the entire ejection fraction spectrum. METHODS AND RESULTS A total of 258 patients with HF with an ejection fraction across the entire spectrum [HF with reduced ejection fraction (HFrEF), n = 168, age 60.6 ± 11.2 years; HF with preserved ejection fraction (HFpEF), n = 50, mean age 65.1 ± 11.9 years; HF with mildly reduced ejection fraction (HFmrEF), n = 32, mean age 65 ± 12] were included. EAT was measured with transthoracic echocardiography. Vascular function was assessed with flicker-light-induced vasodilation of retinal arterioles (FIDart%) and flow-mediated dilatation (FMD%) in conduit arteries. Patients with HFrEF have less EAT compared with patients with HFpEF (4.2 ± 2 vs. 5.3 ± 2 mm, respectively, P < 0.001). Interestingly, EAT was significantly associated with impaired microvascular function (FIDart%; r = -0.213, P = 0.012) and FMD% (r = -0.186, P = 0.022), even after multivariate correction for confounding factors (age, body mass index, hypertension, and diabetes; standardized regression coefficient (SRC) = -0.184, P = 0.049 for FIDart% and SRC = -0.178, P = 0.043 for FMD%) in HFrEF but not in HFpEF. CONCLUSIONS Although less EAT is present in HFrEF than in HFpEF, only in HFrEF EAT is associated with vascular dysfunction. The diverging role of EAT in HF and its switch to a functionally deleterious tissue promoting HF progression provide the rationale to specifically target EAT, in particular in patients with reduced ejection fraction.
Collapse
Affiliation(s)
- Valentina A. Rossi
- Department of CardiologyUniversity Heart Centre, University Hospital of ZurichRaemistrasse 100Zurich8091Switzerland
- Centre for Translational and Experimental CardiologySchlierenSwitzerland
| | - Delia Nebunu
- Department of CardiologyUniversity Heart Centre, University Hospital of ZurichRaemistrasse 100Zurich8091Switzerland
| | - Thomas Haider
- Department of CardiologyUniversity Heart Centre, University Hospital of ZurichRaemistrasse 100Zurich8091Switzerland
| | - Natallia Laptseva
- Department of CardiologyUniversity Heart Centre, University Hospital of ZurichRaemistrasse 100Zurich8091Switzerland
- Centre for Translational and Experimental CardiologySchlierenSwitzerland
| | - Matthias P. Naegele
- Department of CardiologyUniversity Heart Centre, University Hospital of ZurichRaemistrasse 100Zurich8091Switzerland
| | - Frank Ruschitzka
- Department of CardiologyUniversity Heart Centre, University Hospital of ZurichRaemistrasse 100Zurich8091Switzerland
- Centre for Translational and Experimental CardiologySchlierenSwitzerland
- University of ZurichZurichSwitzerland
| | - Isabella Sudano
- Department of CardiologyUniversity Heart Centre, University Hospital of ZurichRaemistrasse 100Zurich8091Switzerland
- Centre for Translational and Experimental CardiologySchlierenSwitzerland
| | - Andreas J. Flammer
- Department of CardiologyUniversity Heart Centre, University Hospital of ZurichRaemistrasse 100Zurich8091Switzerland
- Centre for Translational and Experimental CardiologySchlierenSwitzerland
- University of ZurichZurichSwitzerland
| |
Collapse
|
39
|
Lim ZW, Chee ML, Soh ZD, Majithia S, Sahil T, Tan ST, Sabanayagam C, Wong TY, Cheng CY, Tham YC. Six-Year Incidence of Visual Impairment in a Multiethnic Asian Population: The Singapore Epidemiology of Eye Diseases Study. OPHTHALMOLOGY SCIENCE 2023; 3:100392. [PMID: 38025163 PMCID: PMC10630666 DOI: 10.1016/j.xops.2023.100392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 12/01/2023]
Abstract
Purpose To examine the 6-year incidence of visual impairment (VI) and identify risk factors associated with VI in a multiethnic Asian population. Design Prospective, population-based, cohort study. Participants Adults aged ≥ 40 years were recruited from the Singapore Epidemiology of Eye Diseases cohort study at baseline. Eligible subjects were re-examined after 6 years. Subjects included in the final analysis had a mean age of 56.1 ± 8.9 years, and 2801 (50.5%) were female. Methods All participants underwent standardized examination and interviewer-administered questionnaire at baseline. Incidences were standardized to the Singapore Population Census 2010. A Poisson binomial regression model was used to evaluate the associations between baseline factors and incident presenting VI. Main Outcome Measures Incident presenting VI was assessed at the 6-year follow-up visit. Visual impairment (presenting visual acuity < 20/40), low vision (presenting visual acuity < 20/40 but ≥ 20/200), and blindness (presenting visual acuity < 20/200) were defined based on United States definition. Results A total of 5551 subjects (2188 Chinese, 1837 Indians, and 1526 Malays) were evaluated, of whom 514 developed incident presenting VI over 6 years. Malays had a higher incidence of low vision and blindness (13.0%; 0.6%) than Indians (7.0%; 0.1%) and Chinese (7.7%; 0.2%). Among Malay individuals with VI at baseline, 52.8% remained visually impaired after 6 years, which was considerably higher than Chinese (32.4%) and Indians (37.2%). Older age (per decade; relative risk [RR] = 1.59), a history of cardiovascular disease (RR = 1.38), current smoking (RR = 1.31), smaller housing type (1- to 2-room public flat; RR = 2.01), and no formal education (RR = 1.63) at baseline were associated with a higher risk of incident VI (all P ≤ 0.027). Older age (> 60 years) contributed the highest population attributable risk to incident VI (27.1%), followed by lower monthly income (Singapore dollar < $2000; 26.4%) and smaller housing type (24.7%). Overall, undercorrected refractive error (49.1%) and cataract (82.6%) were leading causes for low vision and blindness, respectively. This was consistently observed across the 3 ethnicities. Conclusions In this multiethnic Asian population, Malays had a higher VI incidence compared to Indians and Chinese. Leading causes of VI are mostly treatable, suggesting that more efforts are needed to further mitigate preventable visual loss. Financial Disclosures The authors have no proprietary or commercial interest in any materials discussed in this article.
Collapse
Affiliation(s)
- Zhi Wei Lim
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Miao-Li Chee
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Zhi Da Soh
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Shivani Majithia
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Thakur Sahil
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - See Teng Tan
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Charumathi Sabanayagam
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Eye Academic Clinical Program, Duke-NUS Medical School, Singapore
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Tsinghua Medicine, Tsinghua University, Beijing, China
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Beijing, China
| | - Ching-Yu Cheng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Eye Academic Clinical Program, Duke-NUS Medical School, Singapore
- Centre for Innovation and Precision Eye Health & Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yih-Chung Tham
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Eye Academic Clinical Program, Duke-NUS Medical School, Singapore
- Centre for Innovation and Precision Eye Health & Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
40
|
Tomasoni M, Beyeler MJ, Vela SO, Mounier N, Porcu E, Corre T, Krefl D, Button AL, Abouzeid H, Lazaros K, Bochud M, Schlingemann R, Bergin C, Bergmann S. Genome-wide Association Studies of Retinal Vessel Tortuosity Identify Numerous Novel Loci Revealing Genes and Pathways Associated With Ocular and Cardiometabolic Diseases. OPHTHALMOLOGY SCIENCE 2023; 3:100288. [PMID: 37131961 PMCID: PMC10149284 DOI: 10.1016/j.xops.2023.100288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 02/03/2023] [Accepted: 02/09/2023] [Indexed: 02/18/2023]
Abstract
Purpose To identify novel susceptibility loci for retinal vascular tortuosity, to better understand the molecular mechanisms modulating this trait, and reveal causal relationships with diseases and their risk factors. Design Genome-wide Association Studies (GWAS) of vascular tortuosity of retinal arteries and veins followed by replication meta-analysis and Mendelian randomization (MR). Participants We analyzed 116 639 fundus images of suitable quality from 63 662 participants from 3 cohorts, namely the UK Biobank (n = 62 751), the Swiss Kidney Project on Genes in Hypertension (n = 397), and OphtalmoLaus (n = 512). Methods Using a fully automated retina image processing pipeline to annotate vessels and a deep learning algorithm to determine the vessel type, we computed the median arterial, venous and combined vessel tortuosity measured by the distance factor (the length of a vessel segment over its chord length), as well as by 6 alternative measures that integrate over vessel curvature. We then performed the largest GWAS of these traits to date and assessed gene set enrichment using the novel high-precision statistical method PascalX. Main Outcome Measure We evaluated the genetic association of retinal tortuosity, measured by the distance factor. Results Higher retinal tortuosity was significantly associated with higher incidence of angina, myocardial infarction, stroke, deep vein thrombosis, and hypertension. We identified 175 significantly associated genetic loci in the UK Biobank; 173 of these were novel and 4 replicated in our second, much smaller, metacohort. We estimated heritability at ∼25% using linkage disequilibrium score regression. Vessel type specific GWAS revealed 116 loci for arteries and 63 for veins. Genes with significant association signals included COL4A2, ACTN4, LGALS4, LGALS7, LGALS7B, TNS1, MAP4K1, EIF3K, CAPN12, ECH1, and SYNPO2. These tortuosity genes were overexpressed in arteries and heart muscle and linked to pathways related to the structural properties of the vasculature. We demonstrated that retinal tortuosity loci served pleiotropic functions as cardiometabolic disease variants and risk factors. Concordantly, MR revealed causal effects between tortuosity, body mass index, and low-density lipoprotein. Conclusions Several alleles associated with retinal vessel tortuosity suggest a common genetic architecture of this trait with ocular diseases (glaucoma, myopia), cardiovascular diseases, and metabolic syndrome. Our results shed new light on the genetics of vascular diseases and their pathomechanisms and highlight how GWASs and heritability can be used to improve phenotype extraction from high-dimensional data, such as images. Financial Disclosures The author(s) have no proprietary or commercial interest in any materials discussed in this article.
Collapse
Affiliation(s)
- Mattia Tomasoni
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Jules-Gonin Eye Hospital, Lausanne, Switzerland
| | - Michael Johannes Beyeler
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Sofia Ortin Vela
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Ninon Mounier
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Eleonora Porcu
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Tanguy Corre
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Daniel Krefl
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Alexander Luke Button
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Hana Abouzeid
- Division of Ophthalmology, Geneva University Hospitals, Geneva, Switzerland
- Clinical Eye Research Center Memorial Adolphe de Rothschild, Geneva, Switzerland
| | | | - Murielle Bochud
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Reinier Schlingemann
- Jules-Gonin Eye Hospital, Lausanne, Switzerland
- Department of Ophthalmology, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | | | - Sven Bergmann
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
41
|
Dogan Z, Ileri C, Ozben B, Sunbul M, Tigen MK, Sahin O, Yesildag O. Evaluation of Arterial Stiffness and Subfoveal Choroidal Thickness in Patients with Coronary Slow Flow. ACTA CARDIOLOGICA SINICA 2023; 39:733-741. [PMID: 37720409 PMCID: PMC10499956 DOI: 10.6515/acs.202309_39(5).20230209a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 02/09/2023] [Indexed: 09/19/2023]
Abstract
Background Coronary slow flow may not only affect the coronary arteries, but it may also be a vascular problem affecting the rest of the arterial system. Objective The aim of this study was to determine peripheral arterial stiffness and the thickness of the choroid layer in patients with slow coronary flow. Methods Fifty consecutive patients (age, 54.3 ± 11.4 years, 38 male) with coronary slow flow and 25 consecutive patients (age, 50.5 ± 9.9 years, 16 male) with normal coronary arteries both documented by coronary angiography were included. Arterial stiffness parameters were measured noninvasively using a Mobil-O-Graph arteriography system. The choroidal thickness was assessed using the enhanced depth imaging optical coherence tomography method. Results The patients with coronary slow flow had significantly higher peripheral systolic blood pressure, peripheral pulse pressure, central pulse pressure, and pulse wave velocity (PWV) and significantly thinner choroidal thickness compared to the controls. Thrombolysis in myocardial infarction frame count was positively correlated with PWV (r: 0.237, p = 0.041) and negatively correlated with choroidal thickness (r: -0.249, p = 0.031). There was also a negative correlation between PWV and mean choroidal thickness (r: -0.565, p < 0.001). Linear regression analysis showed that coronary slow flow was an independent predictor of both PWV and choroidal thickness when adjusted by age and sex. Conclusions The acceleration of average peripheral arterial PWV with a thinning of choroidal thickness in patients with coronary slow flow may support the idea that this phenomenon may be a coronary presentation of a systemic microvascular disorder.
Collapse
Affiliation(s)
- Zekeriya Dogan
- Department of Cardiology, Marmara University School of Medicine
| | - Cigdem Ileri
- Department of Cardiology, Kosuyolu Heart Education and Research Hospital
| | - Beste Ozben
- Department of Cardiology, Marmara University School of Medicine
| | - Murat Sunbul
- Department of Cardiology, Marmara University School of Medicine
| | - M Kursat Tigen
- Department of Cardiology, Marmara University School of Medicine
| | - Ozlem Sahin
- Department of Ophthalmology, Marmara University School of Medicine, Istanbul, Turkey
| | - Osman Yesildag
- Department of Cardiology, Marmara University School of Medicine
| |
Collapse
|
42
|
Fu Y, Yusufu M, Wang Y, He M, Shi D, Wang R. Association of retinal microvascular density and complexity with incident coronary heart disease. Atherosclerosis 2023; 380:117196. [PMID: 37562159 DOI: 10.1016/j.atherosclerosis.2023.117196] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 06/20/2023] [Accepted: 07/26/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND AND AIMS The high mortality rate and huge disease burden of coronary heart disease (CHD) highlight the importance of its early detection and timely intervention. Given the non-invasive nature of fundus photography and recent development in the quantification of retinal microvascular parameters with deep learning techniques, our study aims to investigate the association between incident CHD and retinal microvascular parameters. METHODS UK Biobanks participants with gradable fundus images and without a history of diagnosed CHD at recruitment were included for analysis. A fully automated artificial intelligence system was used to extract quantitative measurements that represent the density and complexity of the retinal microvasculature, including fractal dimension (Df), number of vascular segments (NS), vascular skeleton density (VSD) and vascular area density (VAD). RESULTS A total of 57,947 participants (mean age 55.6 ± 8.1 years; 56% female) without a history of diagnosed CHD were included. During a median follow-up of 11.0 (interquartile range, 10.88 to 11.19) years, 3211 incident CHD events occurred. In multivariable Cox proportional hazards models, we found decreasing Df (adjusted HR = 0.80, 95% CI, 0.65-0.98, p = 0.033), lower NS of arteries (adjusted HR = 0.69, 95% CI, 0.54-0.88, p = 0.002) and venules (adjusted HR = 0.77, 95% CI, 0.61-0.97, p = 0.024), and reduced arterial VSD (adjusted HR = 0.72, 95% CI, 0.57-0.91, p = 0.007) and venous VSD (adjusted HR = 0.78, 95% CI, 0.62-0.98, p = 0.034) were related to an increased risk of incident CHD. CONCLUSIONS Our study revealed a significant association between retinal microvascular parameters and incident CHD. As the lower complexity and density of the retinal vascular network may indicate an increased risk of incident CHD, this may empower its prediction with the quantitative measurements of retinal structure.
Collapse
Affiliation(s)
- Yuechuan Fu
- Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Mayinuer Yusufu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia; Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia
| | - Yueye Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Mingguang He
- The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region of China.
| | - Danli Shi
- The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region of China.
| | - Ruobing Wang
- Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
43
|
Li H, Gao M, Song H, Wu X, Li G, Cui Y, Li Y, Xie Z, Ren Q, Zhang H. Predicting ischemic stroke risk from atrial fibrillation based on multi-spectral fundus images using deep learning. Front Cardiovasc Med 2023; 10:1185890. [PMID: 37600060 PMCID: PMC10434281 DOI: 10.3389/fcvm.2023.1185890] [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: 03/17/2023] [Accepted: 06/05/2023] [Indexed: 08/22/2023] Open
Abstract
Background Ischemic stroke (IS) is one of the most common serious secondary diseases of atrial fibrillation (AF) within 1 year after its occurrence, both of which have manifestations of ischemia and hypoxia of the small vessels in the early phase of the condition. The fundus is a collection of capillaries, while the retina responds differently to light of different wavelengths. Predicting the risk of IS occurring secondary to AF, based on subtle differences in fundus images of different wavelengths, is yet to be explored. This study was conducted to predict the risk of IS occurring secondary to AF based on multi-spectrum fundus images using deep learning. Methods A total of 150 AF participants without suffering from IS within 1 year after discharge and 100 IS participants with persistent arrhythmia symptoms or a history of AF diagnosis in the last year (defined as patients who would develop IS within 1 year after AF, based on fundus pathological manifestations generally prior to symptoms of the brain) were recruited. Fundus images at 548, 605, and 810 nm wavelengths were collected. Three classical deep neural network (DNN) models (Inception V3, ResNet50, SE50) were trained. Sociodemographic and selected routine clinical data were obtained. Results The accuracy of all DNNs with the single-spectral or multi-spectral combination images at the three wavelengths as input reached above 78%. The IS detection performance of DNNs with 605 nm spectral images as input was relatively more stable than with the other wavelengths. The multi-spectral combination models acquired a higher area under the curve (AUC) scores than the single-spectral models. Conclusions The probability of IS secondary to AF could be predicted based on multi-spectrum fundus images using deep learning, and combinations of multi-spectrum images improved the performance of DNNs. Acquiring different spectral fundus images is advantageous for the early prevention of cardiovascular and cerebrovascular diseases. The method in this study is a beneficial preliminary and initiative exploration for diseases that are difficult to predict the onset time such as IS.
Collapse
Affiliation(s)
- Hui Li
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, China
- Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen, China
- Shenzhen Bay Laboratory, Institute of Biomedical Engineering, Shenzhen, China
- National Biomedical Imaging Center, Peking University, Beijing, China
- Institute of Medical Technology, Peking University Health Science Center, Peking University, Beijing, China
| | - Mengdi Gao
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, China
- Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen, China
- Shenzhen Bay Laboratory, Institute of Biomedical Engineering, Shenzhen, China
- National Biomedical Imaging Center, Peking University, Beijing, China
- Institute of Medical Technology, Peking University Health Science Center, Peking University, Beijing, China
| | - Haiqing Song
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiao Wu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Gang Li
- Department of Cardiology, Beijing Yanhua Hospital, Beijing, China
| | - Yiwei Cui
- Department of Cardiology, Beijing Yanhua Hospital, Beijing, China
| | - Yang Li
- Department of Cardiology, Beijing Yanhua Hospital, Beijing, China
| | - Zhaoheng Xie
- Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen, China
- Shenzhen Bay Laboratory, Institute of Biomedical Engineering, Shenzhen, China
- National Biomedical Imaging Center, Peking University, Beijing, China
- Institute of Medical Technology, Peking University Health Science Center, Peking University, Beijing, China
| | - Qiushi Ren
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, China
- Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen, China
- Shenzhen Bay Laboratory, Institute of Biomedical Engineering, Shenzhen, China
- National Biomedical Imaging Center, Peking University, Beijing, China
- Institute of Medical Technology, Peking University Health Science Center, Peking University, Beijing, China
| | - Haitao Zhang
- Cardio-Metabolic Medicine Center, Fuwai Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
44
|
Sørhus E, Nakken CL, Donald CE, Ripley DM, Shiels HA, Meier S. Cardiac toxicity of phenanthrene depends on developmental stage in Atlantic cod (Gadus morhua). THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 881:163484. [PMID: 37068678 DOI: 10.1016/j.scitotenv.2023.163484] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/16/2023] [Accepted: 04/09/2023] [Indexed: 06/01/2023]
Abstract
Complex mixtures like crude oil, and single components such as Phenanthrene (Phe), induce cardiotoxicity by interfering with excitation-contraction coupling. However, recent work has demonstrated that the timing of pollutant exposure during embryogenesis greatly impacts the degree of cardiac dysfunction caused. Here, we aimed to clarify the temporal dependence of Phe toxicity and the downstream effects of cardiac dysfunction using Atlantic cod (Gadus morhua). Phe (nominal concentration, 1.12 μmol/L), or the L-type‑calcium channel blocker Nicardipine (Nic) (nominal concentration, 2 and 4 μmol/L), were individually applied to cod embryos either during cardiogenesis (early) or after the onset of cardiac function (late). Phe toxicity was highly dependent on the timing of exposure. Exposure after the onset of cardiac function (i.e. late) caused more severe cardiac and extracardiac abnormalities at 3 days post hatching (dph) than early exposure. Late Phe exposure resulted in a smaller ventricle, eliminated ventricular contraction, and reduced atrial contraction. In contrast, early Phe exposure did not have an effect on cardiac development and function. This temporal difference was not as evident in the Nic treatment. Early Nic exposure created similar morphological phenotypes to the late Phe exposure. The two treatments (early Nic and late Phe) also shared a cardiofunctional phenotype, comprised of eliminated ventricular, and reduced atrial, contraction. These data suggest that extracardiac abnormalities, such as the craniofacial deformities seen after late embryonic exposure to cardiotoxic oil components and mixtures, are mostly downstream effects of cardiac dysfunction.
Collapse
Affiliation(s)
- Elin Sørhus
- Institute of Marine Research, Department of Marine Toxicology, Bergen, Norway.
| | | | - Carey E Donald
- Institute of Marine Research, Department of Marine Toxicology, Bergen, Norway
| | - Daniel M Ripley
- University of Manchester, Division of Cardiovascular Sciences, United Kingdom of Great Britain and Northern Ireland
| | - Holly A Shiels
- University of Manchester, Division of Cardiovascular Sciences, United Kingdom of Great Britain and Northern Ireland
| | - Sonnich Meier
- Institute of Marine Research, Department of Marine Toxicology, Bergen, Norway
| |
Collapse
|
45
|
Hu W, Yii FSL, Chen R, Zhang X, Shang X, Kiburg K, Woods E, Vingrys A, Zhang L, Zhu Z, He M. A Systematic Review and Meta-Analysis of Applying Deep Learning in the Prediction of the Risk of Cardiovascular Diseases From Retinal Images. Transl Vis Sci Technol 2023; 12:14. [PMID: 37440249 PMCID: PMC10353749 DOI: 10.1167/tvst.12.7.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/08/2023] [Indexed: 07/14/2023] Open
Abstract
Purpose The purpose of this study was to perform a systematic review and meta-analysis to synthesize evidence from studies using deep learning (DL) to predict cardiovascular disease (CVD) risk from retinal images. Methods A systematic literature search was performed in MEDLINE, Scopus, and Web of Science up to June 2022. We extracted data pertaining to predicted outcomes, model development, and validation and model performance metrics. Included studies were graded using the Quality Assessment of Diagnostic Accuracies Studies 2 tool. Model performance was pooled across eligible studies using a random-effects meta-analysis model. Results A total of 26 studies were included in the analysis. There were 42 CVD risk-related outcomes predicted from retinal images were identified, including 33 CVD risk factors, 4 cardiac imaging biomarkers, 2 CVD risk scores, the presence of CVD, and incident CVD. Three studies that aimed to predict the development of future CVD events reported an area under the receiver operating curve (AUROC) between 0.68 and 0.81. Models that used retinal images as input data had a pooled mean absolute error of 3.19 years (95% confidence interval [CI] = 2.95-3.43) for age prediction; a pooled AUROC of 0.96 (95% CI = 0.95-0.97) for gender classification; a pooled AUROC of 0.80 (95% CI = 0.73-0.86) for diabetes detection; and a pooled AUROC of 0.86 (95% CI = 0.81-0.92) for the detection of chronic kidney disease. We observed a high level of heterogeneity and variation in study designs. Conclusions Although DL models appear to have reasonably good performance when it comes to predicting CVD risk, further work is necessary to evaluate the real-world applicability and predictive accuracy. Translational Relevance DL-based CVD risk assessment from retinal images holds great promise to be translated to clinical practice as a novel approach for CVD risk assessment, given its simple, quick, and noninvasive nature.
Collapse
Affiliation(s)
- Wenyi Hu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia
| | - Fabian S. L. Yii
- Centre for Clinical Brain Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
- Curle Ophthalmology Laboratory, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Ruiye Chen
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia
| | - Xinyu Zhang
- Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xianwen Shang
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
| | - Katerina Kiburg
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
| | - Ekaterina Woods
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
| | - Algis Vingrys
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
- Department of Optometry and Vision Sciences, The University of Melbourne, Melbourne, Australia
| | - Lei Zhang
- Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
| | - Zhuoting Zhu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
| | - Mingguang He
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia
| |
Collapse
|
46
|
Yu M, Sun X, Zeng F, Gao X, Li Z, Yuan G, Wang T. The short-term effects of blood donation on the ocular parameters including blood flow of the retina and choroid in healthy people using OCT- angiography. BMC Ophthalmol 2023; 23:259. [PMID: 37303035 DOI: 10.1186/s12886-023-03002-3] [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: 01/15/2023] [Accepted: 05/30/2023] [Indexed: 06/13/2023] Open
Abstract
BACKGROUND To investigate the short-term effects of blood donation on the morphology and blood flow of the retina and choroid in healthy people using optical coherence tomography angiography (OCTA). METHODS Twenty-eight healthy blood donors (56 eyes) who participated in the 200 ml voluntary blood donation between March 2, 2021 and January 20, 2022 were included. The best corrected visual acuity (BCVA), systolic (SBP) and diastolic blood pressure (DBP), intraocular pressure (IOP), subfoveal choroid thickness (SFCT), retinal thickness (RT), retinal superficial vascular density (SVD), deep vascular density (DVD) and foveal avascular were a (FAZ) were measured and statistically analysed 10 min before, 30 min and 24 h after the blood donation. RESULTS The 200 ml blood donation could cause significant IOP reduction at 24 h (P = 0.006), which was negatively correlated with SBP (r = -0.268, P = 0.046), while SBP, DBP, or ocular perfusion pressure were not affected (> 0.05). Moreover, no significant difference existed in the OCT and OCTA indexes, including SFCT, RT, SVD, DVD, and FAZ, before and after the 200 ml blood donation (P > 0.05). The visual acuity was not affected either (P > 0.05). CONCLUSIONS The 200 ml blood donation was noted to be associated with statistically significant IOP reduction at 24 h, while SBP, DBP, or OPP was not affected. The blood flow of the retina and choroid or the visual acuity did not change significantly after the blood donation. Larger studies with different volumes of blood donation were needed to further analysis the effect of blood donation on ocular parameters.
Collapse
Affiliation(s)
- Mengmeng Yu
- Department of Ophthalmology, Eye Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Jinan, China
- School of Ophthalmology, Shandong First Medical University, Jinan, China
| | - Xiaolei Sun
- Department of Ophthalmology, Eye Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Jinan, China
- School of Ophthalmology, Shandong First Medical University, Jinan, China
| | - Fanxing Zeng
- Department of Ophthalmology, Eye Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Jinan, China
- School of Ophthalmology, Shandong First Medical University, Jinan, China
| | - Xiang Gao
- Department of Ophthalmology, Eye Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Jinan, China
- School of Ophthalmology, Shandong First Medical University, Jinan, China
| | - Zhenzhen Li
- Department of Ophthalmology, Eye Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Jinan, China
- School of Ophthalmology, Shandong First Medical University, Jinan, China
| | - Gongqiang Yuan
- Department of Ophthalmology, Eye Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Jinan, China.
- School of Ophthalmology, Shandong First Medical University, Jinan, China.
| | - Ting Wang
- Department of Ophthalmology, Eye Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Jinan, China.
- School of Ophthalmology, Shandong First Medical University, Jinan, China.
| |
Collapse
|
47
|
Yang S, Zhu Z, Yuan Y, Chen S, Shang X, Bulloch G, He M, Wang W. Analysis of Plasma Metabolic Profile on Ganglion Cell-Inner Plexiform Layer Thickness With Mortality and Common Diseases. JAMA Netw Open 2023; 6:e2313220. [PMID: 37191963 DOI: 10.1001/jamanetworkopen.2023.13220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/17/2023] Open
Abstract
Importance The neural retina is considered a unique window to systemic health, but its biological link with systemic health remains unknown. Objective To investigate the independent associations of retinal ganglion cell-inner plexiform layer thickness (GCIPLT) metabolic profiles with rates of mortality and morbidity of common diseases. Design, Setting, and Participants This cohort study evaluated UK Biobank participants enrolled between 2006 and 2010, and prospectively followed them up for multidisease diagnosis and mortality. Additional participants from the Guangzhou Diabetes Eye Study (GDES) underwent optical coherence tomography scanning and metabolomic profiling and were included for validation. Main Outcomes and Measures Systematic analysis of circulating plasma metabolites to identify GCIPLT metabolic profiles; prospective associations of these profiles with mortality and morbidity of 6 common diseases with their incremental discriminative value and clinical utility. Results Among 93 838 community-based participants (51 182 [54.5%] women), the mean (SD) age was 56.7 (8.1) years and mean (SD) follow-up was 12.3 (0.8) years. Of 249 metabolic metrics, 37 were independently associated with GCIPLT, including 8 positive and 29 negative associations, and most were associated with the rates of future mortality and common diseases. These metabolic profiles significantly improved the models for discriminating type 2 diabetes over clinical indicators (C statistic: 0.862; 95% CI, 0.852-0.872 vs clinical indicators only, 0.803; 95% CI, 0.792-0.814; P < .001), myocardial infarction (0.792; 95% CI, 0.775-0.808 vs 0.768; 95% CI, 0.751-0.786; P < .001), heart failure (0.803; 95% CI, 0.786-0.820 vs 0.790; 95% CI, 0.773-0.807; P < .001), stroke (0.739; 95% CI, 0.714-0.764 vs 0.719; 95% CI, 0.693-0.745; P < .001), all-cause mortality (0.747; 95% CI, 0.734-0.760 vs 0.724; 95% CI, 0.711-0.738; P < .001), and cardiovascular disease mortality (0.790; 95% CI, 0.767-0.812 vs 0.763; 95% CI, 0.739-0.788; P < .001). Additionally, the potential of GCIPLT metabolic profiles for risk stratification of cardiovascular diseases were further confirmed in the GDES cohort using a different metabolomic approach. Conclusions and Relevance In this prospective study of multinational participants, GCIPLT-associated metabolites demonstrated the potential to inform mortality and morbidity risks. Incorporating information on these profiles may facilitate individualized risk stratification for these health outcomes.
Collapse
Affiliation(s)
- Shaopeng Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Zhuoting Zhu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Victoria, Australia
| | - Yixiong Yuan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Shida Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Xianwen Shang
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Victoria, Australia
- Department of Ophthalmology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Gabriella Bulloch
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Victoria, Australia
| | - Mingguang He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Victoria, Australia
| | - Wei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| |
Collapse
|
48
|
Khalilipur E, Mahdizad Z, Molazadeh N, Faghihi H, Naderi N, Mehrabi Bahar M, Firouzi A, Sadeghipour P, Maleki M, Soltani Shahgoli S, Khalili Pour E, Riazi-Esfahani H. Microvascular and structural analysis of the retina and choroid in heart failure patients with reduced ejection fraction. Sci Rep 2023; 13:5467. [PMID: 37015968 PMCID: PMC10073248 DOI: 10.1038/s41598-023-32751-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 04/01/2023] [Indexed: 04/06/2023] Open
Abstract
This cross-sectional study was designed to assess alterations of choroidal and retinal microvasculature in patients with Heart Failure with Reduced Ejection Fraction (HFrEF) and compare them with a normal age and sex-matched population. Fifty-two eyes of 26 patients with HFrEF (left ventricular ejection fraction [LVEF] < 40%) and 64 eyes of 32 healthy individuals were considered as the patient and the control groups, respectively. We found no statistically significant differences in age-adjusted mean central macular thickness (CMT), superficial or deep retinal capillary plexus vascular densities, and choriocapillaris flow (CC flow) density between the HFrEF group and the normal controls, with the exception of the parafoveal mean superficial capillary plexus vascular density (P = 0.023), which remained statistically significant after adjusting for age (P = 0.034). The patients with HFrEF had a significantly lower subfoveal choroidal thickness (SFCT) than the normal subjects (264 ± 82 vs 313 ± 72; P = 0.009), and the difference was still statistically significant after age adjustment (P = 0.026). Although choroidal vascularity index (CVI) was lower in the HFrEF group than in the control group, the difference was not statistically significant before and after age adjustment (73.45 ± 6.67 vs 75.77 ± 5.92; P = 0.118 and P = 0.096, respectively). In conclusion, in patients with HFrEF, we observed a reduction in parafoveal retinal VD in the superficial capillary plexus, as well as SFCT, but no significant change in CVI, CMT, or CC flow density.
Collapse
Affiliation(s)
- Ehsan Khalilipur
- Cardiovascular Intervention Research Center, Rajaie Cardiovascular, Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Mahdizad
- Retina Ward, Farabi Eye Hospital, Tehran University of Medical Sciences, Qazvin Square, South Karegar Street, Tehran, 1336616351, Iran
| | - Negin Molazadeh
- Cardiovascular Intervention Research Center, Rajaie Cardiovascular, Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hooshang Faghihi
- Retina Ward, Farabi Eye Hospital, Tehran University of Medical Sciences, Qazvin Square, South Karegar Street, Tehran, 1336616351, Iran
| | - Nasim Naderi
- Cardiovascular Intervention Research Center, Rajaie Cardiovascular, Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Mehrabi Bahar
- Retina Ward, Farabi Eye Hospital, Tehran University of Medical Sciences, Qazvin Square, South Karegar Street, Tehran, 1336616351, Iran
| | - Ata Firouzi
- Cardiovascular Intervention Research Center, Rajaie Cardiovascular, Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Parham Sadeghipour
- Cardiovascular Intervention Research Center, Rajaie Cardiovascular, Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Maleki
- Cardiovascular Intervention Research Center, Rajaie Cardiovascular, Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sahel Soltani Shahgoli
- Retina Ward, Farabi Eye Hospital, Tehran University of Medical Sciences, Qazvin Square, South Karegar Street, Tehran, 1336616351, Iran
| | - Elias Khalili Pour
- Retina Ward, Farabi Eye Hospital, Tehran University of Medical Sciences, Qazvin Square, South Karegar Street, Tehran, 1336616351, Iran.
| | - Hamid Riazi-Esfahani
- Retina Ward, Farabi Eye Hospital, Tehran University of Medical Sciences, Qazvin Square, South Karegar Street, Tehran, 1336616351, Iran
| |
Collapse
|
49
|
Zhu Z, Shi D, Guankai P, Tan Z, Shang X, Hu W, Liao H, Zhang X, Huang Y, Yu H, Meng W, Wang W, Ge Z, Yang X, He M. Retinal age gap as a predictive biomarker for mortality risk. Br J Ophthalmol 2023; 107:547-554. [PMID: 35042683 DOI: 10.1136/bjophthalmol-2021-319807] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 10/27/2021] [Indexed: 01/09/2023]
Abstract
AIM To develop a deep learning (DL) model that predicts age from fundus images (retinal age) and to investigate the association between retinal age gap (retinal age predicted by DL model minus chronological age) and mortality risk. METHODS A total of 80 169 fundus images taken from 46 969 participants in the UK Biobank with reasonable quality were included in this study. Of these, 19 200 fundus images from 11 052 participants without prior medical history at the baseline examination were used to train and validate the DL model for age prediction using fivefold cross-validation. A total of 35 913 of the remaining 35 917 participants had available mortality data and were used to investigate the association between retinal age gap and mortality. RESULTS The DL model achieved a strong correlation of 0.81 (p<0·001) between retinal age and chronological age, and an overall mean absolute error of 3.55 years. Cox regression models showed that each 1 year increase in the retinal age gap was associated with a 2% increase in risk of all-cause mortality (hazard ratio (HR)=1.02, 95% CI 1.00 to 1.03, p=0.020) and a 3% increase in risk of cause-specific mortality attributable to non-cardiovascular and non-cancer disease (HR=1.03, 95% CI 1.00 to 1.05, p=0.041) after multivariable adjustments. No significant association was identified between retinal age gap and cardiovascular- or cancer-related mortality. CONCLUSIONS Our findings indicate that retinal age gap might be a potential biomarker of ageing that is closely related to risk of mortality, implying the potential of retinal image as a screening tool for risk stratification and delivery of tailored interventions.
Collapse
Affiliation(s)
- Zhuoting Zhu
- Department of Ophthalmology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Danli Shi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Peng Guankai
- Guangzhou Vision Tech Medical Technology Co., Ltd, GuangZhou, China
| | - Zachary Tan
- Centre for Eye Research Australia; Ophthalmology, University of Melbourne, East Melbourne, Victoria, Australia
| | - Xianwen Shang
- Department of Ophthalmology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Wenyi Hu
- Centre for Eye Research Australia; Ophthalmology, University of Melbourne, East Melbourne, Victoria, Australia
| | - Huan Liao
- Neural Regeneration Group, Institute of Reconstructive Neurobiology, University of Bonn, Bonn, Germany
| | - Xueli Zhang
- Department of Ophthalmology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Yu Huang
- Department of Ophthalmology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Honghua Yu
- Department of Ophthalmology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Wei Meng
- Guangzhou Vision Tech Medical Technology Co., Ltd, GuangZhou, China
| | - Wei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Zongyuan Ge
- Monash e-Research Centre, Monash University, Melbourne, Victoria, Australia
- Monash Medical AI Group, Monash University, Melbourne, Victoria, Australia
| | - Xiaohong Yang
- Department of Ophthalmology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Mingguang He
- Department of Ophthalmology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Centre for Eye Research Australia; Ophthalmology, University of Melbourne, East Melbourne, Victoria, Australia
| |
Collapse
|
50
|
Zhu Y, Li L, Han B, Sun X, Chen R, Lei Y, Kan H. Low ambient temperature and temperature drop as novel risk factors of acute glaucoma: a case-crossover study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:56513-56521. [PMID: 36920607 DOI: 10.1007/s11356-023-26235-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/27/2023] [Indexed: 06/18/2023]
Abstract
The prevalence of glaucoma has seasonal variation in population, but the role of ambient temperature and its variation remains unclear in this seasonal trend. So, we conducted a time-stratified case-crossover study to examine the association of ambient temperature and temperature change between neighboring days (TCN) with the risk of acute glaucoma. Data on meteorological parameters and glaucoma outpatient visit between 2015 and 2021 covered all districts of Shanghai. Conditional logistic regression with distributed lag nonlinear model was applied to estimate the association of temperature or TCN with the risk of acute glaucoma. A total of 7,746 patients diagnosed with acute primary angle-closure glaucoma (APACG) were included in this analysis. We observed a significant increase in the risk of acute glaucoma with cold temperature and temperature drop. Compared with the referent temperature (32℃), moderate low (12 °C) and extreme low (4 °C) temperature exposures were associated with higher risk of acute glaucoma outpatient visit, with the highest cumulative OR of 1.46 (95% CI: 1.11, 1.91) and 1.50 (95% CI: 1.09, 2.06) over lag 0-2 days. Temperature drop (TCN = - 4 °C) also increases the risk of acute glaucoma (OR = 1.34, 95% CI: 1.07, 1.67) over lag 0-7 days, comparing with no temperature change. Patients of female and above age 65 were more vulnerable to cold exposure and temperature drop. This case-crossover study provided novel and robust individual-level evidence that low ambient temperature and temperature drop significantly increase the acute glaucoma risk. The findings provide protective strategies for glaucoma patient, especially for female and the old, under cold exposure and sudden temperature decline.
Collapse
Affiliation(s)
- Yixiang Zhu
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education, NHC Key Lab of Health Technology Assessment, IRDR ICoE On Risk Interconnectivity and Governance On Weather/Climate Extremes Impact and Public Health, Fudan University, Shanghai, China
| | - Liping Li
- Department of Ophthalmology & Visual Science, Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, China
- NHC Key Laboratory of Myopia, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai, 200031, China
| | - Binze Han
- Department of Ophthalmology & Visual Science, Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, China
- NHC Key Laboratory of Myopia, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai, 200031, China
| | - Xinghuai Sun
- Department of Ophthalmology & Visual Science, Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, China
- NHC Key Laboratory of Myopia, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai, 200031, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Renjie Chen
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education, NHC Key Lab of Health Technology Assessment, IRDR ICoE On Risk Interconnectivity and Governance On Weather/Climate Extremes Impact and Public Health, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Meteorology and Health, Shanghai Typhoon Institute/CMA, Shanghai, 200030, China
| | - Yuan Lei
- Department of Ophthalmology & Visual Science, Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, China.
- NHC Key Laboratory of Myopia, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai, 200031, China.
| | - Haidong Kan
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education, NHC Key Lab of Health Technology Assessment, IRDR ICoE On Risk Interconnectivity and Governance On Weather/Climate Extremes Impact and Public Health, Fudan University, Shanghai, China
- Children's Hospital of Fudan University, National Center for Children's Health, Shanghai, China
| |
Collapse
|