51
|
Bledsoe X, Gamazon ER. NeuroimaGene: an R package for assessing the neurological correlates of genetically regulated gene expression. BMC Bioinformatics 2024; 25:325. [PMID: 39379815 PMCID: PMC11463069 DOI: 10.1186/s12859-024-05936-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/18/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND We present the NeuroimaGene resource as an R package designed to assist researchers in identifying genes and neurologic features relevant to psychiatric and neurological health. While recent studies have identified hundreds of genes as potential components of pathophysiology in neurologic and psychiatric disease, interpreting the physiological consequences of this variation is challenging. The integration of neuroimaging data with molecular findings is a step toward addressing this challenge. In addition to sharing associations with both molecular variation and clinical phenotypes, neuroimaging features are intrinsically informative of cognitive processes. NeuroimaGene provides a tool to understand how disease-associated genes relate to the intermediate structure of the brain. RESULTS We created NeuroimaGene, a user-friendly, open access R package now available for public use. Its primary function is to identify neuroimaging derived brain features that are impacted by genetically regulated expression of user-provided genes or gene sets. This resource can be used to (1) characterize individual genes or gene sets as relevant to the structure and function of the brain, (2) identify the region(s) of the brain or body in which expression of target gene(s) is neurologically relevant, (3) impute the brain features most impacted by user-defined gene sets such as those produced by cohort level gene association studies, and (4) generate publication level, modifiable visual plots of significant findings. We demonstrate the utility of the resource by identifying neurologic correlates of stroke-associated genes derived from pre-existing analyses. CONCLUSIONS Integrating neurologic data as an intermediate phenotype in the pathway from genes to brain-based diagnostic phenotypes increases the interpretability of molecular studies and enriches our understanding of disease pathophysiology. The NeuroimaGene R package is designed to assist in this process and is publicly available for use.
Collapse
Affiliation(s)
- Xavier Bledsoe
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Eric R Gamazon
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Memory & Alzheimer's Center, Nashville, TN, USA
| |
Collapse
|
52
|
Yang Y, Lorincz-Comi N, Zhu X. Estimation of a genetic Gaussian network using GWAS summary data. Biometrics 2024; 80:ujae148. [PMID: 39656744 DOI: 10.1093/biomtc/ujae148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 11/02/2024] [Accepted: 11/14/2024] [Indexed: 12/16/2024]
Abstract
A genetic Gaussian network of multiple phenotypes, constructed through the inverse matrix of the genetic correlation matrix, is informative for understanding the biological dependencies of the phenotypes. However, its estimation may be challenging because the genetic correlation estimates are biased due to estimation errors and idiosyncratic pleiotropy inherent in GWAS summary statistics. Here, we introduce a novel approach called estimation of genetic graph (EGG), which eliminates the estimation error bias and idiosyncratic pleiotropy bias with the same techniques used in multivariable Mendelian randomization. The genetic network estimated by EGG can be interpreted as shared common biological contributions between phenotypes, conditional on others. We use both simulations and real data to demonstrate the superior efficacy of our novel method in comparison with the traditional network estimators.
Collapse
Affiliation(s)
- Yihe Yang
- Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, United States
| | - Noah Lorincz-Comi
- Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, United States
| | - Xiaofeng Zhu
- Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, United States
| |
Collapse
|
53
|
Cárcel-Márquez J, Muiño E, Gallego-Fabrega C, Cullell N, Lledós M, Llucià-Carol L, Martín-Campos JM, Sobrino T, Campos F, Castillo J, Freijo M, Arenillas JF, Obach V, Álvarez-Sabín J, Molina CA, Ribó M, Jiménez-Conde J, Roquer J, Muñoz-Narbona L, Lopez-Cancio E, Millán M, Diaz-Navarro R, Vives-Bauza C, Serrano-Heras G, Segura T, Ibañez L, Heitsch L, Delgado P, Dhar R, Krupinski J, Prats-Sánchez L, Camps-Renom P, Guasch M, Ezcurra G, Blay N, Sumoy L, de Cid R, Montaner J, Cruchaga C, Lee JM, Martí-Fàbregas J, Férnandez-Cadenas I. Sex-Stratified Genome-Wide Association Study in the Spanish Population Identifies a Novel Locus for Lacunar Stroke. Stroke 2024; 55:2462-2471. [PMID: 39315829 DOI: 10.1161/strokeaha.124.047833] [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: 05/15/2024] [Revised: 08/07/2024] [Accepted: 08/14/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND Ischemic stroke (IS) represents a significant health burden globally, necessitating a better understanding of its genetic underpinnings to improve prevention and treatment strategies. Despite advances in IS genetics, studies focusing on the Spanish population and sex-stratified analyses are lacking. METHODS A case-control genome-wide association study was conducted with 9081 individuals (3493 IS cases and 5588 healthy controls). IS subtypes using Trial of ORG 10172 in Acute Stroke Treatment criteria were explored in a sex-stratified approach. Replication efforts involved the MEGASTROKE, GIGASTROKE, and the UK Biobank international cohorts. Post-genome-wide association study analysis included: in silico proteomic analysis, gene-based analysis, quantitative trait loci annotation, transcriptome-wide association analysis, and bioinformatic analysis using chromatin accessibility data. RESULTS Identified as associated with IS and its subtypes were 4 significant and independent loci. Replication confirmed 5p15.2 as a new locus associated with small-vessel occlusion stroke, with rs59970332-T as the lead variant (beta [SE], 0.13 [0.02]; P=4.34×10-8). Functional analyses revealed CTNND2 given proximity and its implication in pathways involved in vascular integrity and angiogenesis. Integration of Hi-C data identified additional potentially modulated genes, and in silico proteomic analysis suggested a distinctive blood proteome profile associated with the lead variant. Gene-set enrichment analyses highlighted pathways consistent with small-vessel disease pathogenesis. Gene-based associations with known stroke-related genes such as F2 and FGG were also observed, reinforcing the relevance of our findings. CONCLUSIONS We found CTNND2 as a potential key molecule in small-vessel occlusion stroke risk, and predominantly in males. This study sheds light on the genetic architecture of IS in the Spanish population, providing novel insights into sex-specific associations and potential molecular mechanisms. Further research, including replication in larger cohorts, is essential for a comprehensive understanding of these findings and for their translation to clinical practice.
Collapse
Affiliation(s)
- Jara Cárcel-Márquez
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca Sant Pau, Barcelona, Spain (J.C.-M., E.M., C.G.-F., N.C., M.L., L.L.-C., J.M.M.-C., I.F.-C.)
| | - Elena Muiño
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca Sant Pau, Barcelona, Spain (J.C.-M., E.M., C.G.-F., N.C., M.L., L.L.-C., J.M.M.-C., I.F.-C.)
- Epilepsy Unit (E.M.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Cristina Gallego-Fabrega
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca Sant Pau, Barcelona, Spain (J.C.-M., E.M., C.G.-F., N.C., M.L., L.L.-C., J.M.M.-C., I.F.-C.)
| | - Natalia Cullell
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca Sant Pau, Barcelona, Spain (J.C.-M., E.M., C.G.-F., N.C., M.L., L.L.-C., J.M.M.-C., I.F.-C.)
- Stroke Pharmacogenomics and Genetics Laboratory, Fundación Docència I Recerca Mútua Terrassa, Hospital Mútua Terrassa, Spain (N.C.)
| | - Miquel Lledós
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca Sant Pau, Barcelona, Spain (J.C.-M., E.M., C.G.-F., N.C., M.L., L.L.-C., J.M.M.-C., I.F.-C.)
| | - Laia Llucià-Carol
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca Sant Pau, Barcelona, Spain (J.C.-M., E.M., C.G.-F., N.C., M.L., L.L.-C., J.M.M.-C., I.F.-C.)
| | - Jesús M Martín-Campos
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca Sant Pau, Barcelona, Spain (J.C.-M., E.M., C.G.-F., N.C., M.L., L.L.-C., J.M.M.-C., I.F.-C.)
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (T. Sobrino, F.C., J.C.), La Coruña, Spain
| | - Francisco Campos
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (T. Sobrino, F.C., J.C.), La Coruña, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain (F.C.)
| | - José Castillo
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (T. Sobrino, F.C., J.C.), La Coruña, Spain
| | - Marimar Freijo
- Biocruces-Bizkaia Health Research Institute, Department of Neurology, Bilbao, Spain (M.F.)
| | | | - Victor Obach
- Department of Neurology, Hospital Clínic de Barcelona, IDIBAPS, Spain (V.O.)
| | - José Álvarez-Sabín
- Stroke Unit, Department of Neurology, Hospital Universitari Vall d'Hebron, Barcelona, Spain (J.A.-S., C.A.M., M.R.)
| | - Carlos A Molina
- Stroke Unit, Department of Neurology, Hospital Universitari Vall d'Hebron, Barcelona, Spain (J.A.-S., C.A.M., M.R.)
| | - Marc Ribó
- Stroke Unit, Department of Neurology, Hospital Universitari Vall d'Hebron, Barcelona, Spain (J.A.-S., C.A.M., M.R.)
| | - Jordi Jiménez-Conde
- Department of Neurology, IMIM-Hospital del Mar; Neurovascular Research Group, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain (J.J.-C., J.R.)
| | - Jaume Roquer
- Department of Neurology, IMIM-Hospital del Mar; Neurovascular Research Group, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain (J.J.-C., J.R.)
| | - Lucia Muñoz-Narbona
- Department of Neurosciences, Hospital Germans Trias I Pujol (L.M.-N., M.M.), Universitat Autònoma de Barcelona, Spain
| | - Elena Lopez-Cancio
- Departament of Neurology, University Hospital Central de Asturias, Spain (E.L.-C.)
| | - Mònica Millán
- Department of Neurosciences, Hospital Germans Trias I Pujol (L.M.-N., M.M.), Universitat Autònoma de Barcelona, Spain
| | - Rosa Diaz-Navarro
- Department of Neurology, Son Espases University Hospital, Illes Balears Health Research Institute, Spain (R.D.-N., C.V.-B.)
| | - Cristòfol Vives-Bauza
- Department of Neurology, Son Espases University Hospital, Illes Balears Health Research Institute, Spain (R.D.-N., C.V.-B.)
| | - Gemma Serrano-Heras
- Department of Neurology, University Hospital of Albacete, Spain (G.S.-H., T. Segura)
| | - Tomás Segura
- Department of Neurology, University Hospital of Albacete, Spain (G.S.-H., T. Segura)
| | - Laura Ibañez
- Department of Psychiatry (L.I., C.C.), Washington University School of Medicine, St. Louis, MO
- Department of Neurology (L.I., L.H., R.D., J.-M.L.), Washington University School of Medicine, St. Louis, MO
- Neurogenomics and Informatics Center at Washington University in St. Louis, MO (L.I., C.C.)
| | - Laura Heitsch
- Department of Neurology (L.I., L.H., R.D., J.-M.L.), Washington University School of Medicine, St. Louis, MO
- Department of Emergency Medicine (L.H.), Washington University School of Medicine, St. Louis, MO
| | - Pilar Delgado
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (P.D.), Universitat Autònoma de Barcelona, Spain
| | - Rajat Dhar
- Department of Neurology (L.I., L.H., R.D., J.-M.L.), Washington University School of Medicine, St. Louis, MO
| | - Jerzy Krupinski
- Neurology Service, Hospital Universitari Mútua Terrassa, Spain (J.K.)
| | - Luis Prats-Sánchez
- Stroke Unit (L.P.-S., P.C.-R., M.G., G.E., J.M.-F.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Pol Camps-Renom
- Stroke Unit (L.P.-S., P.C.-R., M.G., G.E., J.M.-F.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Marina Guasch
- Stroke Unit (L.P.-S., P.C.-R., M.G., G.E., J.M.-F.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Garbiñe Ezcurra
- Stroke Unit (L.P.-S., P.C.-R., M.G., G.E., J.M.-F.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Natalia Blay
- GenomesForLife-GCAT Lab (N.B., R.d.C.), Germans Trias i Pujol Research Institute, Barcelona, Spain
| | - Lauro Sumoy
- High Content Genomics and Bioinformatics Unit (L.S.), Germans Trias i Pujol Research Institute, Barcelona, Spain
| | - Rafael de Cid
- GenomesForLife-GCAT Lab (N.B., R.d.C.), Germans Trias i Pujol Research Institute, Barcelona, Spain
| | - Joan Montaner
- Institute de Biomedicine of Seville, IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville and Department of Neurology, Hospital Universitario Virgen Macarena, Spain (J.M.)
| | - Carlos Cruchaga
- Department of Psychiatry (L.I., C.C.), Washington University School of Medicine, St. Louis, MO
- Neurogenomics and Informatics Center at Washington University in St. Louis, MO (L.I., C.C.)
| | - Jin-Moo Lee
- Department of Neurology (L.I., L.H., R.D., J.-M.L.), Washington University School of Medicine, St. Louis, MO
| | - Joan Martí-Fàbregas
- Stroke Unit (L.P.-S., P.C.-R., M.G., G.E., J.M.-F.), Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Israel Férnandez-Cadenas
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca Sant Pau, Barcelona, Spain (J.C.-M., E.M., C.G.-F., N.C., M.L., L.L.-C., J.M.M.-C., I.F.-C.)
| |
Collapse
|
54
|
Rundek T, Romano JG. Brain Health. Stroke 2024; 55:2532-2535. [PMID: 38551086 PMCID: PMC11421983 DOI: 10.1161/strokeaha.124.044292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Affiliation(s)
- Tatjana Rundek
- Department of Neurology (T.R., J.G.R.), Miller School of Medicine, University of Miami, FL
- Evelyn F. McKnight Brain Institute (T.R.), Miller School of Medicine, University of Miami, FL
| | - Jose G Romano
- Department of Neurology (T.R., J.G.R.), Miller School of Medicine, University of Miami, FL
| |
Collapse
|
55
|
Zhong J, Zhang P, Dong Y, Xu Y, Huang H, Ye R, Liu X, Sun W. Well-Being and Cardiovascular Health: Insights From the UK Biobank Study. J Am Heart Assoc 2024; 13:e035225. [PMID: 39291465 DOI: 10.1161/jaha.124.035225] [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: 02/27/2024] [Accepted: 07/15/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Cardiovascular diseases (CVDs) are a leading global health concern. Emerging evidence suggests a potential protective role of well-being in reducing CVD risk. METHODS AND RESULTS We conducted a cohort analysis using the UK Biobank data set, encompassing 121 317 participants. We assessed the well-being of participants using a well-being index derived from baseline questionnaires. Well-being categories were derived by latent class analysis using general happiness and satisfaction with family, friendships, health, and finance situations. The relationship between well-being and 4 major CVDs was analyzed using Cox proportional hazards models and Mendelian randomization. The study also examined the impacts of well-being on lifestyle factors and inflammatory markers, and its mediating role in the well-being-CVD relationship. Higher well-being was associated with a significantly reduced risk of various CVDs. Latent class analysis identified 4 distinct well-being groups (low, variable, moderate-to-high, and high satisfaction), with higher satisfaction levels generally associated with lower risk of CVDs. Mendelian randomization suggested potential causal relationships between well-being and reduced risk of CVDs. Participants with greater well-being demonstrated healthier behaviors and lower levels of inflammatory markers. Mediation analysis indicated that lifestyle and inflammatory markers partially mediated the relationship between well-being and CVDs. CONCLUSIONS This study demonstrates a robust inverse association between well-being and the risks of CVDs, suggesting that enhancing well-being may be a viable strategy for CVD prevention. The role of lifestyle factors and inflammation as a mediator provides insight into possible biological pathways linking psychological states and cardiovascular health.
Collapse
Affiliation(s)
- Jinghui Zhong
- Department of Neurology, Division of Life Sciences and Medicine, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC University of Science and Technology of China Hefei Anhui China
| | - Pan Zhang
- Department of Neurology, Division of Life Sciences and Medicine, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC University of Science and Technology of China Hefei Anhui China
| | - Yiran Dong
- Department of Neurology, Division of Life Sciences and Medicine, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC University of Science and Technology of China Hefei Anhui China
| | - Yingjie Xu
- Department of Neurology, Division of Life Sciences and Medicine, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC University of Science and Technology of China Hefei Anhui China
| | - Hongmei Huang
- Department of Neurology, Division of Life Sciences and Medicine, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC University of Science and Technology of China Hefei Anhui China
| | - Ruidong Ye
- Department of Neurology, Jinling Hospital, Affiliated Hospital of Medical School Nanjing University Nanjing China
| | - Xinfeng Liu
- Department of Neurology, Division of Life Sciences and Medicine, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC University of Science and Technology of China Hefei Anhui China
- Department of Neurology, Jinling Hospital, Affiliated Hospital of Medical School Nanjing University Nanjing China
| | - Wen Sun
- Department of Neurology, Division of Life Sciences and Medicine, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC University of Science and Technology of China Hefei Anhui China
| |
Collapse
|
56
|
Zhang W, Zhu J, Wu X, Feng T, Liao W, Li X, Chen J, Zhang L, Xiao C, Cui H, Yang C, Yan P, Wang Y, Tang M, Chen L, Liu Y, Zou Y, Wu X, Zhang L, Yang C, Yao Y, Li J, Liu Z, Jiang X, Zhang B. Phenotypic and genetic effect of carotid intima-media thickness on the risk of stroke. Hum Genet 2024; 143:1131-1143. [PMID: 38578439 DOI: 10.1007/s00439-024-02666-1] [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: 10/16/2023] [Accepted: 03/05/2024] [Indexed: 04/06/2024]
Abstract
While carotid intima-media thickness (cIMT) as a noninvasive surrogate measure of atherosclerosis is widely considered a risk factor for stroke, the intrinsic link underlying cIMT and stroke has not been fully understood. We aimed to evaluate the clinical value of cIMT in stroke through the investigation of phenotypic and genetic relationships between cIMT and stroke. We evaluated phenotypic associations using observational data from UK Biobank (N = 21,526). We then investigated genetic relationships leveraging genomic data conducted in predominantly European ancestry for cIMT (N = 45,185) and any stroke (AS, Ncase/Ncontrol=40,585/406,111). Observational analyses suggested an increased hazard of stroke per one standard deviation increase in cIMT (cIMTmax-AS: hazard ratio (HR) = 1.39, 95%CI = 1.09-1.79; cIMTmean-AS: HR = 1.39, 95%CI = 1.09-1.78; cIMTmin-AS: HR = 1.32, 95%CI = 1.04-1.68). A positive global genetic correlation was observed (cIMTmax-AS: [Formula: see text]=0.23, P=9.44 × 10-5; cIMTmean-AS: [Formula: see text]=0.21, P=3.00 × 10-4; cIMTmin-AS: [Formula: see text]=0.16, P=6.30 × 10-3). This was further substantiated by five shared independent loci and 15 shared expression-trait associations. Mendelian randomization analyses suggested no causal effect of cIMT on stroke (cIMTmax-AS: odds ratio (OR)=1.12, 95%CI=0.97-1.28; cIMTmean-AS: OR=1.09, 95%CI=0.93-1.26; cIMTmin-AS: OR=1.03, 95%CI = 0.90-1.17). A putative association was observed for genetically predicted stroke on cIMT (AS-cIMTmax: beta=0.07, 95%CI = 0.01-0.13; AS-cIMTmean: beta=0.08, 95%CI = 0.01-0.15; AS-cIMTmin: beta = 0.08, 95%CI = 0.01-0.16) in the reverse direction MR, which attenuated to non-significant in sensitivity analysis. Our work does not find evidence supporting causal associations between cIMT and stroke. The pronounced cIMT-stroke association is intrinsic, and mostly attributed to shared genetic components. The clinical value of cIMT as a surrogate marker for stroke risk in the general population is likely limited.
Collapse
Affiliation(s)
- Wenqiang Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Jingwei Zhu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Xuan Wu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Tianle Feng
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Wei Liao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Xuan Li
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Jianci Chen
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Li Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Chenghan Xiao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
- Department of Maternal, Child and Adolescent Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Huijie Cui
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Chao Yang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Peijing Yan
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Yutong Wang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Mingshuang Tang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Lin Chen
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Yunjie Liu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Yanqiu Zou
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Xueyao Wu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Ling Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
- Department of Iatrical Polymer Material and Artificial Apparatus, School of Polymer Science and Engineering, Sichuan University, Chengdu, China
| | - Chunxia Yang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Yuqin Yao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
- Department of Occupational and Environmental Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Jiayuan Li
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Zhenmi Liu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
- Department of Maternal, Child and Adolescent Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xia Jiang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China.
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
- Department of Clinical Neuroscience, Karolinskaa Institutet, Stockholm, Sweden.
| | - Ben Zhang
- Hainan General Hospital and Hainan Affiliated Hospital, Hainan Medical University, Haikou, China; West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
57
|
Cavaillès C, Andrews SJ, Leng Y, Chatterjee A, Daghlas I, Yaffe K. Causal Associations of Sleep Apnea With Alzheimer Disease and Cardiovascular Disease: A Bidirectional Mendelian Randomization Analysis. J Am Heart Assoc 2024; 13:e033850. [PMID: 39258525 DOI: 10.1161/jaha.123.033850] [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: 12/04/2023] [Accepted: 06/26/2024] [Indexed: 09/12/2024]
Abstract
BACKGROUND Sleep apnea (SA) has been linked to an increased risk of dementia in numerous observational studies; whether this is driven by neurodegenerative, vascular, or other mechanisms is not clear. We sought to examine the bidirectional causal relationships between SA, Alzheimer disease (AD), coronary artery disease (CAD), and ischemic stroke using Mendelian randomization. METHODS AND RESULTS Using summary statistics from 4 recent, large genome-wide association studies of SA (n=523 366), AD (n=94 437), CAD (n=1 165 690), and stroke (n=1 308 460), we conducted bidirectional 2-sample Mendelian randomization analyses. Our primary analytic method was fixed-effects inverse variance-weighted (IVW) Mendelian randomization; diagnostics tests and sensitivity analyses were conducted to verify the robustness of the results. We identified a significant causal effect of SA on the risk of CAD (odds ratio [ORIVW]=1.35 per log-odds increase in SA liability [95% CI=1.25-1.47]) and stroke (ORIVW=1.13 [95% CI=1.01-1.25]). These associations were somewhat attenuated after excluding single-nucleotide polymorphisms associated with body mass index (ORIVW=1.26 [95% CI=1.15-1.39] for CAD risk; ORIVW=1.08 [95% CI=0.96-1.22] for stroke risk). SA was not causally associated with a higher risk of AD (ORIVW=1.14 [95% CI=0.91-1.43]). We did not find causal effects of AD, CAD, or stroke on risk of SA. CONCLUSIONS These results suggest that SA increased the risk of CAD, and the identified causal association with stroke risk may be confounded by body mass index. Moreover, no causal effect of SA on AD risk was found. Future studies are warranted to investigate cardiovascular pathways between sleep disorders, including SA, and dementia.
Collapse
Affiliation(s)
- Clémence Cavaillès
- Department of Psychiatry and Behavioral Sciences University of California San Francisco San Francisco CA
| | - Shea J Andrews
- Department of Psychiatry and Behavioral Sciences University of California San Francisco San Francisco CA
| | - Yue Leng
- Department of Psychiatry and Behavioral Sciences University of California San Francisco San Francisco CA
| | | | - Iyas Daghlas
- Department of Neurology University of California San Francisco San Francisco CA
| | - Kristine Yaffe
- Department of Psychiatry and Behavioral Sciences University of California San Francisco San Francisco CA
- San Francisco Veterans Affairs Health Care System San Francisco CA
- Department of Neurology University of California San Francisco San Francisco CA
- Department of Epidemiology University of California San Francisco San Francisco CA
| |
Collapse
|
58
|
Wolford BN, Åsvold BO. Bidirectional Mendelian Randomization to Elucidate the Relationship Between Healthy Sleep, Brains, and Hearts. J Am Heart Assoc 2024; 13:e037394. [PMID: 39258560 DOI: 10.1161/jaha.124.037394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 08/12/2024] [Indexed: 09/12/2024]
Affiliation(s)
- Brooke N Wolford
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing Norwegian University of Science and Technology Trondheim Norway
| | - Bjørn O Åsvold
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing Norwegian University of Science and Technology Trondheim Norway
- Department of Endocrinology, Clinic of Medicine, St. Olav's Hospital Trondheim University Hospital Trondheim Norway
| |
Collapse
|
59
|
Kelemen M, Danesh J, Di Angelantonio E, Inouye M, O'Sullivan J, Pennells L, Roychowdhury T, Sweeting MJ, Wood AM, Harrison S, Kim LG. Evaluating the cost-effectiveness of polygenic risk score-stratified screening for abdominal aortic aneurysm. Nat Commun 2024; 15:8063. [PMID: 39277617 PMCID: PMC11401842 DOI: 10.1038/s41467-024-52452-w] [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: 03/01/2024] [Accepted: 09/09/2024] [Indexed: 09/17/2024] Open
Abstract
As the heritability of abdominal aortic aneurysm (AAA) is high and AAA partially shares genetic architecture with other cardiovascular diseases, genetic information could help inform AAA screening strategies. Exploiting pleiotropy and meta-analysing summary data from large studies, we construct a polygenic risk score (PRS) for AAA. Leveraging related traits improves PRS performance (R2) by 22.7%, relative to using AAA alone. Compared with the low PRS tertile, intermediate and high tertiles have hazard ratios for AAA of 2.13 (95%CI 1.61, 2.82) and 3.70 (95%CI 2.86, 4.80) respectively, adjusted for clinical risk factors. Using simulation modelling, we compare PRS- and smoking-stratified screening with inviting men at age 65 and not inviting women (current UK strategy). In a futuristic scenario where genomic information is available, our modelling suggests inviting male current smokers with high PRS earlier than 65 and screening female smokers with high/intermediate PRS at 65 and 70 respectively, may improve cost-effectiveness.
Collapse
Affiliation(s)
- M Kelemen
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - J Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - E Di Angelantonio
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - M Inouye
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Baker Heart & Diabetes Institute, Melbourne, Australia
| | - J O'Sullivan
- Division of Cardiology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - L Pennells
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - T Roychowdhury
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - M J Sweeting
- Department of Population Health Sciences, University of Leicester, Leicester, UK
| | - A M Wood
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | | | - L G Kim
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
60
|
Rodriguez-Flores JL, Khalid S, Parikshak N, Rasheed A, Ye B, Kapoor M, Backman J, Sepehrband F, Gioia SAD, Gelfman S, De T, Banerjee N, Sharma D, Martinez H, Castaneda S, D'Ambrosio D, Zhang XA, Xun P, Tsai E, Tsai IC, Khan MZ, Jahanzaib M, Mian MR, Liaqat MB, Mahmood K, Salam TU, Hussain M, Iqbal J, Aslam F, Cantor MN, Tzoneva G, Overton J, Marchini J, Reid JG, Baras A, Verweij N, Lotta LA, Coppola G, Karalis K, Economides A, Fazio S, Liedtke W, Danesh J, Kamal A, Frossard P, Coleman T, Shuldiner AR, Saleheen D. NOTCH3 p.Arg1231Cys is markedly enriched in South Asians and associated with stroke. Nat Commun 2024; 15:8029. [PMID: 39271666 PMCID: PMC11399414 DOI: 10.1038/s41467-024-51819-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 08/19/2024] [Indexed: 09/15/2024] Open
Abstract
The genetic factors of stroke in South Asians are largely unexplored. Exome-wide sequencing and association analysis (ExWAS) in 75 K Pakistanis identified NM_000435.3(NOTCH3):c.3691 C > T, encoding the missense amino acid substitution p.Arg1231Cys, enriched in South Asians (alternate allele frequency = 0.58% compared to 0.019% in Western Europeans), and associated with subcortical hemorrhagic stroke [odds ratio (OR) = 3.39, 95% confidence interval (CI) = [2.26, 5.10], p = 3.87 × 10-9), and all strokes (OR [CI] = 2.30 [1.77, 3.01], p = 7.79 × 10-10). NOTCH3 p.Arg231Cys was strongly associated with white matter hyperintensity on MRI in United Kingdom Biobank (UKB) participants (effect [95% CI] in SD units = 1.1 [0.61, 1.5], p = 3.0 × 10-6). The variant is attributable for approximately 2.0% of hemorrhagic strokes and 1.1% of all strokes in South Asians. These findings highlight the value of diversity in genetic studies and have major implications for genomic medicine and therapeutic development in South Asian populations.
Collapse
Affiliation(s)
| | - Shareef Khalid
- Columbia University, New York, NY, USA
- Center for Non-Communicable Diseases, Karachi, Pakistan
| | | | - Asif Rasheed
- Center for Non-Communicable Diseases, Karachi, Pakistan
| | - Bin Ye
- Regeneron Genetics Center, Tarrytown, NY, USA
| | | | | | | | | | | | - Tanima De
- Regeneron Genetics Center, Tarrytown, NY, USA
| | | | | | | | | | | | | | | | - Ellen Tsai
- University of California at Los Angeles, Los Angeles, CA, USA
| | - I-Chun Tsai
- Regeneron Pharmaceuticals Inc, Tarrytown, NY, USA
| | | | | | | | | | - Khalid Mahmood
- Dow University of Health Sciences and Civil Hospital, Karachi, Pakistan
| | | | | | - Javed Iqbal
- Department of Neurology, Allied Hospital, Faisalabad, Pakistan
| | - Faizan Aslam
- Department of Neurology, Aziz Fatima Hospital, Faisalabad, Pakistan
| | | | | | | | | | | | - Aris Baras
- Regeneron Genetics Center, Tarrytown, NY, USA
| | | | | | | | | | | | - Sergio Fazio
- Regeneron Pharmaceuticals Inc, Tarrytown, NY, USA
| | | | - John Danesh
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Ayeesha Kamal
- Section of Neurology, Department of Medicine, Aga Khan University, Karachi, Pakistan
| | | | | | | | - Danish Saleheen
- Columbia University, New York, NY, USA.
- Center for Non-Communicable Diseases, Karachi, Pakistan.
| |
Collapse
|
61
|
Samani NJ, Beeston E, Greengrass C, Riveros-McKay F, Debiec R, Lawday D, Wang Q, Budgeon CA, Braund PS, Bramley R, Kharodia S, Newton M, Marshall A, Krzeminski A, Zafar A, Chahal A, Heer A, Khunti K, Joshi N, Lakhani M, Farooqi A, Plagnol V, Donnelly P, Weale ME, Nelson CP. Polygenic risk score adds to a clinical risk score in the prediction of cardiovascular disease in a clinical setting. Eur Heart J 2024; 45:3152-3160. [PMID: 38848106 PMCID: PMC11379490 DOI: 10.1093/eurheartj/ehae342] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 04/10/2024] [Accepted: 05/16/2024] [Indexed: 09/08/2024] Open
Abstract
BACKGROUND AND AIMS A cardiovascular disease polygenic risk score (CVD-PRS) can stratify individuals into different categories of cardiovascular risk, but whether the addition of a CVD-PRS to clinical risk scores improves the identification of individuals at increased risk in a real-world clinical setting is unknown. METHODS The Genetics and the Vascular Health Check Study (GENVASC) was embedded within the UK National Health Service Health Check (NHSHC) programme which invites individuals between 40-74 years of age without known CVD to attend an assessment in a UK general practice where CVD risk factors are measured and a CVD risk score (QRISK2) is calculated. Between 2012-2020, 44,141 individuals (55.7% females, 15.8% non-white) who attended an NHSHC in 147 participating practices across two counties in England were recruited and followed. When 195 individuals (cases) had suffered a major CVD event (CVD death, myocardial infarction or acute coronary syndrome, coronary revascularisation, stroke), 396 propensity-matched controls with a similar risk profile were identified, and a nested case-control genetic study undertaken to see if the addition of a CVD-PRS to QRISK2 in the form of an integrated risk tool (IRT) combined with QRISK2 would have identified more individuals at the time of their NHSHC as at high risk (QRISK2 10-year CVD risk of ≥10%), compared with QRISK2 alone. RESULTS The distribution of the standardised CVD-PRS was significantly different in cases compared with controls (cases mean score .32; controls, -.18, P = 8.28×10-9). QRISK2 identified 61.5% (95% confidence interval [CI]: 54.3%-68.4%) of individuals who subsequently developed a major CVD event as being at high risk at their NHSHC, while the combination of QRISK2 and IRT identified 68.7% (95% CI: 61.7%-75.2%), a relative increase of 11.7% (P = 1×10-4). The odds ratio (OR) of being up-classified was 2.41 (95% CI: 1.03-5.64, P = .031) for cases compared with controls. In individuals aged 40-54 years, QRISK2 identified 26.0% (95% CI: 16.5%-37.6%) of those who developed a major CVD event, while the combination of QRISK2 and IRT identified 38.4% (95% CI: 27.2%-50.5%), indicating a stronger relative increase of 47.7% in the younger age group (P = .001). The combination of QRISK2 and IRT increased the proportion of additional cases identified similarly in women as in men, and in non-white ethnicities compared with white ethnicity. The findings were similar when the CVD-PRS was added to the atherosclerotic cardiovascular disease pooled cohort equations (ASCVD-PCE) or SCORE2 clinical scores. CONCLUSIONS In a clinical setting, the addition of genetic information to clinical risk assessment significantly improved the identification of individuals who went on to have a major CVD event as being at high risk, especially among younger individuals. The findings provide important real-world evidence of the potential value of implementing a CVD-PRS into health systems.
Collapse
Affiliation(s)
- Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Emma Beeston
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Chris Greengrass
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | | | - Radoslaw Debiec
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Daniel Lawday
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Qingning Wang
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Charley A Budgeon
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- School of Population and Global Health, University of Western Australia, Perth WA 6009, Australia
| | - Peter S Braund
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Richard Bramley
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Shireen Kharodia
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Michelle Newton
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Andrea Marshall
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | | | - Azhar Zafar
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester LE5 4PW, UK
- Diabetes and Cardiovascular Medicine General Practice Alliance Federation Research and Training Academy, Northampton NN2 6AL, UK
| | - Anuj Chahal
- South Leicestershire Medical Group, Kibworth Beauchamp LE8 0LG, UK
| | | | - Kamlesh Khunti
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester LE5 4PW, UK
| | - Nitin Joshi
- Willowbrook Medical Centre, Leicester LE5 2NL, UK
| | - Mayur Lakhani
- Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Azhar Farooqi
- Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Vincent Plagnol
- Genomics plc, King Charles House, Park End Street, Oxford OX1 1 JD, UK
| | - Peter Donnelly
- Genomics plc, King Charles House, Park End Street, Oxford OX1 1 JD, UK
| | - Michael E Weale
- Genomics plc, King Charles House, Park End Street, Oxford OX1 1 JD, UK
| | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| |
Collapse
|
62
|
Huang R, Kong X, Geng R, Wu J, Chen T, Li J, Li C, Wu Y, You D, Zhao Y, Zhong Z, Ni S, Bai J. Joint and interactive associations of body mass index and genetic factors with cardiovascular disease: a prospective study in UK Biobank. BMC Public Health 2024; 24:2371. [PMID: 39223569 PMCID: PMC11367834 DOI: 10.1186/s12889-024-19916-6] [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: 01/15/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Both body mass index (BMI) and genetic factors independently contribute to cardiovascular disease (CVD). However, it is unclear whether genetic risk modifies the association between BMI and the risk of incident CVD. This study aimed to investigate whether BMI categories and genetic risk jointly and interactively contribute to incident CVD events, including hypertension (HTN), atrial fibrillation (AF), coronary heart disease (CHD), stroke, and heart failure (HF). METHODS A total of 496,851 participants from the UK Biobank with one or more new-onset CVD events were included in the analyses. BMI was categorized as normal weight (< 25.0 kg/m2), overweight (25.0-29.9 kg/m2), and obesity (≥ 30.0 kg/m2). Genetic risk for each outcome was defined as low (lowest tertile), intermediate (second tertile), and high (highest tertile) using polygenic risk score. The joint associations of BMI categories and genetic risk with incident CVD were investigated using Cox proportional hazard models. Additionally, additive interactions were evaluated. RESULTS Among the 496,851 participants, 270,726 (54.5%) were female, with a mean (SD) age was 56.5 (8.1) years. Over a median follow-up (IQR) of 12.4 (11.5-13.1) years, 102,131 (22.9%) participants developed HTN, 26,301 (5.4%) developed AF, 32,222 (6.9%) developed CHD, 10,684 (2.2%) developed stroke, and 13,304 (2.7%) developed HF. Compared with the normal weight with low genetic risk, the obesity with high genetic risk had the highest risk of CVD: HTN (HR: 3.96; 95%CI: 3.84-4.09), AF (HR: 3.60; 95%CI: 3.38-3.83), CHD (HR: 2.76; 95%CI: 2.61-2.91), stroke (HR: 1.44; 95%CI: 1.31-1.57), and HF (HR: 2.47; 95%CI: 2.27-2.69). There were significant additive interactions between BMI categories and genetic risk for HTN, AF, and CHD, with relative excess risk of 0.53 (95%CI: 0.43-0.62), 0.67 (95%CI: 0.51-0.83), and 0.37 (95%CI: 0.25-0.49), respectively. CONCLUSIONS BMI and genetic factors jointly and interactively contribute to incident CVD, especially among participants with high genetic risk. These findings have public health implications for identifying populations more likely to have cardiovascular benefit from weight loss interventions.
Collapse
Affiliation(s)
- Ruyu Huang
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Xinxin Kong
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Rui Geng
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Jingwei Wu
- Department of Epidemiology and Biostatistics, College of Public Health, Temple University, Philadelphia, PA, 19122, USA
| | - Tao Chen
- Center for Health Economics, University of York, York, YO105DD, UK
| | - Jiong Li
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Chunjian Li
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yaqian Wu
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Dongfang You
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yang Zhao
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Zihang Zhong
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Senmiao Ni
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Jianling Bai
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
63
|
Ball NJ, Ghimire S, Follain G, Pajari AO, Wurzinger D, Vaitkevičiūtė M, Cowell AR, Berki B, Ivaska J, Paatero I, Goult BT, Jacquemet G. TLNRD1 is a CCM complex component and regulates endothelial barrier integrity. J Cell Biol 2024; 223:e202310030. [PMID: 39013281 PMCID: PMC11252447 DOI: 10.1083/jcb.202310030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 04/08/2024] [Accepted: 05/23/2024] [Indexed: 07/18/2024] Open
Abstract
We previously identified talin rod domain-containing protein 1 (TLNRD1) as a potent actin-bundling protein in vitro. Here, we report that TLNRD1 is expressed in the vasculature in vivo. Its depletion leads to vascular abnormalities in vivo and modulation of endothelial cell monolayer integrity in vitro. We demonstrate that TLNRD1 is a component of the cerebral cavernous malformations (CCM) complex through its direct interaction with CCM2, which is mediated by a hydrophobic C-terminal helix in CCM2 that attaches to a hydrophobic groove on the four-helix domain of TLNRD1. Disruption of this binding interface leads to CCM2 and TLNRD1 accumulation in the nucleus and actin fibers. Our findings indicate that CCM2 controls TLNRD1 localization to the cytoplasm and inhibits its actin-bundling activity and that the CCM2-TLNRD1 interaction impacts endothelial actin stress fiber and focal adhesion formation. Based on these results, we propose a new pathway by which the CCM complex modulates the actin cytoskeleton and vascular integrity.
Collapse
Affiliation(s)
- Neil J. Ball
- School of Biosciences, University of Kent, Canterbury, UK
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Sujan Ghimire
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
| | - Gautier Follain
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Ada O. Pajari
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
| | - Diana Wurzinger
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
| | - Monika Vaitkevičiūtė
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
| | | | - Bence Berki
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- Department of Life Technologies, University of Turku, Turku, Finland
- Western Finnish Cancer Center (FICAN West), University of Turku, Turku, Finland
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
- InFLAMES Research Flagship Center, University of Turku and Åbo Akademi University, Turku, Finland
| | - Ilkka Paatero
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Benjamin T. Goult
- School of Biosciences, University of Kent, Canterbury, UK
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Guillaume Jacquemet
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku and Åbo Akademi University, Turku, Finland
- Turku Bioimaging, University of Turku and Åbo Akademi University, Turku, Finland
| |
Collapse
|
64
|
Ruan W, Zhou X, Liu H, Wang T, Zhang G, Lin K. Causal role of circulating inflammatory cytokines in cardiac diseases, structure and function. Heart Lung 2024; 67:70-79. [PMID: 38714139 DOI: 10.1016/j.hrtlng.2024.04.018] [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: 01/28/2024] [Revised: 03/25/2024] [Accepted: 04/27/2024] [Indexed: 05/09/2024]
Abstract
BACKGROUND Inflammation is implicated in cardiovascular disease (CVD) pathogenesis, but causal roles of specific circulating inflammatory cytokines remain unclear. Mendelian randomization (MR) studies are well-poised to provide etiological insights beyond constraints of conventional research. METHODS We conducted a large-scale MR study to investigate potential causal relationships of 91 inflammatory proteins with CVD outcomes and cardiac remodeling using summary-level genetic data. Outcomes included coronary artery disease, myocardial infarction, stroke, atrial fibrillation, heart failure, abdominal aortic aneurysm, deep vein thrombosis of lower extremities, pulmonary embolism, cardiac structure and functional parameters. Inverse-variance weighted analysis was undertaken as the primary analysis, with several sensitivity analyses applied. RESULTS Hepatocyte growth factor (HGF) demonstrated a causal relationship with increased susceptibility to both any stroke (OR 1.111; 95 % CI 1.044 - 1.183; P = 9.50e-04) and ischemic stroke (OR 1.121; 95 % CI 1.047 - 1.200; P = 1.04e-03). Programmed cell death 1 ligand 1 (PD-L1) was negatively associated with atrial fibrillation risk (OR 0.936, 95 % CI 0.901 - 0.973; P = 7.69e-04). CCL20, CDCP1, Flt3L and IL-10RA were identified as causal coronary artery disease risk factors, while LIF and ST1A1 had protective effects. IL-4 and LIF-R demonstrated causal links with right heart functional changes. CONCLUSIONS Our MR study nominates specific circulating inflammatory cytokines as potential targets for CVD treatment and prevention. Further research into mechanisms and clinical translation are warranted.
Collapse
Affiliation(s)
- Weiqiang Ruan
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, #37 Guoxue Alley, Wuhou District, Chengdu, Sichuan Province 610041, PR China
| | - Xiaoqin Zhou
- Research Center of Clinical Epidemiology and Evidence-Based Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China; Center of Biostatistics, Design, Measurement and Evaluation (CBDME), Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Huizhen Liu
- Center of Biostatistics, Design, Measurement and Evaluation (CBDME), Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Ting Wang
- Center of Biostatistics, Design, Measurement and Evaluation (CBDME), Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Guiying Zhang
- Research Center of Clinical Epidemiology and Evidence-Based Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Ke Lin
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, #37 Guoxue Alley, Wuhou District, Chengdu, Sichuan Province 610041, PR China.
| |
Collapse
|
65
|
A genome-wide association meta-analysis of all-cause and vascular dementia. Alzheimers Dement 2024; 20:5973-5995. [PMID: 39046104 PMCID: PMC11497727 DOI: 10.1002/alz.14115] [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: 06/09/2023] [Revised: 04/30/2024] [Accepted: 05/20/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION Dementia is a multifactorial disease with Alzheimer's disease (AD) and vascular dementia (VaD) pathologies making the largest contributions. Yet, most genome-wide association studies (GWAS) focus on AD. METHODS We conducted a GWAS of all-cause dementia (ACD) and examined the genetic overlap with VaD. Our dataset includes 800,597 individuals, with 46,902 and 8702 cases of ACD and VaD, respectively. Known AD loci for ACD and VaD were replicated. Bioinformatic analyses prioritized genes that are likely functionally relevant and shared with closely related traits and risk factors. RESULTS For ACD, novel loci identified were associated with energy transport (SEMA4D), neuronal excitability (ANO3), amyloid deposition in the brain (RBFOX1), and magnetic resonance imaging markers of small vessel disease (SVD; HBEGF). Novel VaD loci were associated with hypertension, diabetes, and neuron maintenance (SPRY2, FOXA2, AJAP1, and PSMA3). DISCUSSION Our study identified genetic risks underlying ACD, demonstrating overlap with neurodegenerative processes, vascular risk factors, and cerebral SVD. HIGHLIGHTS We conducted the largest genome-wide association study of all-cause dementia (ACD) and vascular dementia (VaD). Known genetic variants associated with AD were replicated for ACD and VaD. Functional analyses identified novel loci for ACD and VaD. Genetic risks of ACD overlapped with neurodegeneration, vascular risk factors, and cerebral small vessel disease.
Collapse
|
66
|
Kang DS, Yang PS, Kim D, Jang E, Yu HT, Kim TH, Sung JH, Pak HN, Lee MH, Lip GYH, Joung B. Racial Differences in Ischemic and Hemorrhagic Stroke: An Ecological Epidemiological Study. Thromb Haemost 2024; 124:883-892. [PMID: 38423097 DOI: 10.1055/a-2278-8769] [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] [Indexed: 03/02/2024]
Abstract
BACKGROUND This study aimed to evaluate racial differences in the incidence of stroke by conducting an ecological epidemiological study using UK Biobank and Korean nationwide data. METHODS This study used individual data from the Korean National Health Insurance Service-Health Screening and UK Biobank, which included participants who underwent health examinations between 2006 and 2010. We included 112,750 East Asians (50.7% men, mean age: 52.6 years) and 210,995 Caucasians (44.7% men, mean age: 55.0 years) who were not diagnosed with atrial fibrillation, cardiovascular diseases, chronic kidney disease, chronic obstructive pulmonary disease, or cancer. The primary outcome was defined as a composite of ischemic and hemorrhagic stroke. RESULTS East Asians tended to have a lower body mass index (23.7 vs. 26.4 kg/m2, p < 0.001) and a higher proportion of participants who did not engage in moderate-to-vigorous physical activity (49.6% vs. 10.7%, p < 0.001) than Caucasians. During the follow-up, East Asians had higher 5-year incidence rates (presented as per 1,000 person-years) for primary outcome (1.73 vs. 0.50; IR ratio [IRR]: 3.48, 95% confidence interval [CI]: 3.13-3.88), ischemic stroke (1.23 vs. 0.33; IRR: 3.70, 95% CI: 3.25-4.21), hemorrhagic stroke (0.56 vs. 0.18; IRR: 3.20, 95% CI: 2.67-3.84), and atrial fibrillation-related stroke (0.19 vs. 0.09; IRR: 2.04, 95% CI: 1.55-2.68). CONCLUSION Based on this ecological epidemiological study, racial differences in stroke incidence were robust to a variety of statistical analyses, regardless of the subtype. This suggests the need for region-specific approaches to stroke prevention.
Collapse
Affiliation(s)
- Dong-Seon Kang
- Division of Cardiology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Pil-Sung Yang
- Division of Cardiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Daehoon Kim
- Division of Cardiology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eunsun Jang
- Division of Cardiology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hee Tae Yu
- Division of Cardiology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Tae-Hoon Kim
- Division of Cardiology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jung-Hoon Sung
- Division of Cardiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Hui-Nam Pak
- Division of Cardiology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Moon-Hyoung Lee
- Division of Cardiology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart and Chest Hospital, Liverpool, United Kingdom
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Boyoung Joung
- Division of Cardiology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
67
|
Adeniji O, Olowoyo P, Akinyemi R, Fisher M, Owolabi M. Advances in Primordial, Primary, and Secondary Prevention of Stroke in Diverse Populations. Stroke 2024; 55:2359-2365. [PMID: 38445485 DOI: 10.1161/strokeaha.123.044231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/22/2024] [Accepted: 01/30/2024] [Indexed: 03/07/2024]
Affiliation(s)
- Olaleye Adeniji
- Department of Neurology, University College Hospital, Ibadan, Nigeria (O.A., R.A., M.O.)
| | - Paul Olowoyo
- Neurology Unit, Department of Medicine, Afe Babalola University Ado Ekiti, Nigeria (P.O.)
| | - Rufus Akinyemi
- Department of Neurology, University College Hospital, Ibadan, Nigeria (O.A., R.A., M.O.)
- Center for Genomic and Precision Medicine, University of Ibadan, Nigeria (R.A., M.O.)
| | - Marc Fisher
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (M.F.)
| | - Mayowa Owolabi
- Department of Neurology, University College Hospital, Ibadan, Nigeria (O.A., R.A., M.O.)
- Center for Genomic and Precision Medicine, University of Ibadan, Nigeria (R.A., M.O.)
| |
Collapse
|
68
|
Zhu C, Liu J, Lin J, Xu J, Yu E. Investigating the effects of Ginkgo biloba leaf extract on cognitive function in Alzheimer's disease. CNS Neurosci Ther 2024; 30:e14914. [PMID: 39238068 PMCID: PMC11377177 DOI: 10.1111/cns.14914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 07/18/2024] [Accepted: 07/29/2024] [Indexed: 09/07/2024] Open
Abstract
AIMS Alzheimer's disease (AD) is a neurodegenerative disorder with limited treatment options. This study aimed to investigate the therapeutic effects of Ginkgo biloba leaf extract (GBE) on AD and explore its potential mechanisms of action. METHODS Key chemical components of GBE, including quercetin, luteolin, and kaempferol, were identified using network pharmacology methods. Bioinformatics analysis revealed their potential roles in AD through modulation of the PI3K/AKT/NF-κB signaling pathway. RESULTS Mouse experiments demonstrated that GBE improved cognitive function, enhanced neuronal morphology, and reduced serum inflammatory factors. Additionally, GBE modulated the expression of relevant proteins and mRNA. CONCLUSION GBE shows promise as a potential treatment for AD. Its beneficial effects on cognitive function, neuronal morphology, and inflammation may be attributed to its modulation of the PI3K/AKT/NF-κB signaling pathway. These findings provide experimental evidence for the application of Ginkgo biloba leaf in AD treatment and highlight its potential mechanisms of action.
Collapse
Affiliation(s)
- Cheng Zhu
- School of Mental Health, Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, China
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Jie Liu
- The Second People's Hospital of Chuzhou Sleep Disorders Department, Chuzhou, China
| | - Jixin Lin
- Second Clinical Medicine Faculty, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiaxi Xu
- General Psychiatric Department, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Enyan Yu
- Clinical Psychology Department, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
69
|
Fry H, Mazidi M, Kartsonaki C, Clarke R, Walters RG, Chen Z, Millwood IY. The Role of Furin and Its Therapeutic Potential in Cardiovascular Disease Risk. Int J Mol Sci 2024; 25:9237. [PMID: 39273186 PMCID: PMC11394739 DOI: 10.3390/ijms25179237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 09/15/2024] Open
Abstract
Furin is an important proteolytic enzyme, converting several proteins from inactive precursors to their active forms. Recently, proteo-genomic analyses in European and East Asian populations suggested a causal association of furin with ischaemic heart disease, and there is growing interest in its role in cardiovascular disease (CVD) aetiology. In this narrative review, we present a critical appraisal of evidence from population studies to assess furin's role in CVD risk and potential as a drug target for CVD. Whilst most observational studies report positive associations between furin expression and CVD risk, some studies report opposing effects, which may reflect the complex biological roles of furin and its substrates. Genetic variation in FURIN is also associated with CVD and its risk factors. We found no evidence of current clinical development of furin as a drug target for CVD, although several phase 1 and 2 clinical trials of furin inhibitors as a type of cancer immunotherapy have been completed. The growing field of proteo-genomics in large-scale population studies may inform the future development of furin and other potential drug targets to improve the treatment and prevention of CVD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Iona Y. Millwood
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK; (H.F.); (M.M.); (C.K.); (R.C.); (R.G.W.); (Z.C.)
| |
Collapse
|
70
|
Wu S, Meena D, Yarmolinsky J, Gill D, Smith A, Dib MJ, Chauhan G, Rohatgi A, Dehghan A, Tzoulaki I. Mendelian Randomization and Bayesian Colocalization Analysis Implicate Glycoprotein VI as a Potential Drug Target for Cardioembolic Stroke in South Asian Populations. J Am Heart Assoc 2024; 13:e035008. [PMID: 39119976 DOI: 10.1161/jaha.124.035008] [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: 04/04/2024] [Accepted: 06/20/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND Circulating plasma proteins are clinically useful biomarkers for stroke risk. We examined the causal links between plasma proteins and stroke risk in individuals of South Asian ancestry. METHODS AND RESULTS We applied proteome-wide Mendelian randomization and colocalization approaches to understand causality of 2922 plasma proteins on stroke risk in individuals of South Asian ancestry. We obtained genetic instruments (proxies) for plasma proteins from the UK Biobank (N=920). Genome-wide association studies summary data for strokes (N≤11 312) were sourced from GIGASTROKE consortium. Our primary approach involved the Wald ratio or inverse-variance-weighted methods, with statistical significance set at false discovery rate <0.1. Additionally, a Bayesian colocalization approach assessed shared causal variants among proteome, transcriptome, and stroke phenotypes to minimize bias from linkage disequilibrium. We found evidence of a potential causal effect of plasma GP6 (glycoprotein VI) levels on cardioembolic stroke (odds ratio [OR]Wald ratio=2.53 [95% CI, 1.59-4.03]; P=9.2×10-5, false discovery rate=0.059). Generalized Mendelian randomization accounting for correlated single nucleotide polymorphisms (SNPs), with the P value threshold at P<5×10-8 and clumped at r2=0.3, showed consistent direction of effect of GP6 on cardioembolic stroke (ORgeneralized inverse-variance-weighted=2.21 [95% CI, 1.46-3.33]; P=1.6×10-4). Colocalization analysis indicated that plasma GP6 levels colocalize with cardioembolic stroke (posterior probability=91.4%). Multitrait colocalization combining transcriptome, proteome, and cardioembolic stroke showed moderate to strong evidence that these 2 traits colocalize with GP6 expression in the coronary artery and brain tissues (multitrait posterior probability>50%). The potential causal effect of GP6 on cardioembolic stroke was not significant in European populations (ORinverse-variance-weighted=1.08 [95% CI, 0.93-1.26]; P=0.29). CONCLUSIONS Our joint Mendelian randomization and colocalization analyses suggest that genetically predicted GP6 is potentially causally associated with cardioembolic stroke risk in individuals of South Asian ancestry. As genetic data on individuals of South Asian ancestry increase, future Mendelian randomization studies with larger sample size for plasma GP6 levels should be implemented to further validate our findings. Additionally, clinical studies will be necessary to verify GP6 as a therapeutic target for cardioembolic stroke in South Asians.
Collapse
Affiliation(s)
- Siwei Wu
- Department of Epidemiology and Biostatistics School of Public Health, Imperial College London London United Kingdom
| | - Devendra Meena
- Department of Epidemiology and Biostatistics School of Public Health, Imperial College London London United Kingdom
| | - James Yarmolinsky
- Department of Epidemiology and Biostatistics School of Public Health, Imperial College London London United Kingdom
| | - Dipender Gill
- Department of Epidemiology and Biostatistics School of Public Health, Imperial College London London United Kingdom
| | - Alexander Smith
- Department of Epidemiology and Biostatistics School of Public Health, Imperial College London London United Kingdom
| | - Marie-Joe Dib
- Division of Cardiovascular Medicine Hospital of the University of Pennsylvania Philadelphia PA USA
| | - Ganesh Chauhan
- Department of Genetics & Genomics Rajendra Institute of Medical Sciences (RIMS) Ranchi India
| | - Anand Rohatgi
- Department of Medicine, Division of Cardiology University of Texas Southwestern Medical Center Dallas TX USA
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics School of Public Health, Imperial College London London United Kingdom
- Dementia Research Institute, Imperial College London London United Kingdom
| | - Ioanna Tzoulaki
- Department of Epidemiology and Biostatistics School of Public Health, Imperial College London London United Kingdom
- Dementia Research Institute, Imperial College London London United Kingdom
- Biomedical Research Foundation Academy of Athens Athens Greece
| |
Collapse
|
71
|
Chen X, Li A, Zhou W, Yao L. No genetic association between iron deficiency anemia and ischemic stroke and its subtypes: a bidirectional two-sample Mendelian randomization study. Front Neurol 2024; 15:1408758. [PMID: 39228510 PMCID: PMC11369898 DOI: 10.3389/fneur.2024.1408758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/08/2024] [Indexed: 09/05/2024] Open
Abstract
Background Observational researches have suggested a connection between iron deficiency anemia (IDA) and an increased likelihood of ischemic stroke (IS), yet establishing causality is challenging owing to the inherent limitations of such studies, including their vulnerability to confounding factors and the potential for reverse causation. This study employs a bidirectional two-sample Mendelian randomization (MR) approach to assess the causal linkage between IDA and IS and its subtypes. Methods Identifiable single nucleotide polymorphisms (SNPs) with significant links to either IDA or IS and its subtypes were employed as instrumental variables (IVs). The relationship between IDA and any IS, small vessel stroke (SVS), cardioembolic stroke (CES), and large artery stroke (LAS), was quantified using the inverse variance weighted (IVW) method. Complementary analyses utilizing MR-Egger and weighted median methods further supplemented the IVW findings. Moreover, the leave-one-out analysis, MR-Egger intercept test, MR-PRESSO global test, and Cochrane's Q test were conducted for sensitivity analyses. Results This study revealed no correlation between IDA and any IS (IVW method: OR [95% CI] = 0.977 [0.863-1.106]; p = 0.716), LAS (OR [95% CI] = 1.158 [0.771-1.740]; p = 0.479), CES (OR [95% CI] = 1.065 [0.882-1.285]; p = 0.512), or SVS (OR [95% CI] = 1.138 [0.865-1.498]; p = 0.357). Conducting a reverse MR analysis, it was determined that there is no causal connection between any IS, LAS, CES, SVS, and IDA (all p > 0.05). Sensitivity analysis indicated that heterogeneity was not significant and no evidence of horizontal pleiotropy was detected. Conclusion This MR study suggested no causal effect of IDA on IS, LAS, CES, and SVS. Through reverse MR analyses, it was determined that IS and its subtypes did not exert a causal impact on IDA.
Collapse
Affiliation(s)
- Xingyu Chen
- Department of Neurology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Aiping Li
- Department of Neurology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Wensheng Zhou
- Department of Neurology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Liping Yao
- Department of Neurology, The Third Hospital of Changsha, Changsha, China
| |
Collapse
|
72
|
Zhang Q, Shen C, Zhang L, Wang M. Causal Relationship between Chronic Hepatitis B and Stroke in East Asians: A Mendelian Randomization Study. J Cardiovasc Dev Dis 2024; 11:247. [PMID: 39195155 DOI: 10.3390/jcdd11080247] [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: 07/01/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 08/29/2024] Open
Abstract
Both chronic hepatitis B (CHB) and stroke contribute to a high burden of disease in the majority of low- and middle-income countries. Epidemiological studies yield conflicting results on the association between CHB and stroke, and the causal relationship remains inconclusive. This study aimed to assess the causal effects of CHB on stroke and its subtypes in East Asians by Mendelian randomization (MR) analysis. Variants associated with CHB were obtained from a genome-wide association study (GWAS) of Chinese samples as instrumental variables. The summary statistics for stroke in East Asians were derived from the largest published GWAS to date. Two-sample MR analyses were implemented to evaluate the causal effects of CHB on stroke and its subtypes by using the canonical inverse variance weighting method and other supplementary approaches. We observed an association between genetic predisposition to CHB and a decreased risk of large-artery atherosclerotic stroke (odds ratio = 0.872, 95% confidence interval = 0.786-0.967, p = 0.010). The causal effects of CHB on other stroke outcomes were not statistically significant. Evidence for heterogeneity and horizontal pleiotropy were not found in our analyses. This study provides genetic evidence for a negative association between CHB and stroke in East Asians, which helps improve our understanding of the etiology of stroke.
Collapse
Affiliation(s)
- Qi Zhang
- School of Information and Electronic Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China
| | - Cancong Shen
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China
| | - Lei Zhang
- School of Information and Electronic Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China
| | - Maiqiu Wang
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China
| |
Collapse
|
73
|
Inoue K, Naito T, Fuji R, Sonehara K, Yamamoto K, Baba R, Kodama T, Otagaki Y, Okada A, Itcho K, Kobuke K, Ohno H, Morisaki T, Hattori N, Goto A, Nishikawa T, Oki K, Okada Y. Primary Aldosteronism and Risk of Cardiovascular Outcomes: Genome-Wide Association and Mendelian Randomization Study. J Am Heart Assoc 2024; 13:e034180. [PMID: 39101507 DOI: 10.1161/jaha.123.034180] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/02/2024] [Indexed: 08/06/2024]
Abstract
BACKGROUND Observational studies have reported associations between primary aldosteronism (PA) and cardiovascular outcomes, including coronary artery diseases (CAD), congestive heart failure (CHF), and stroke. However, establishing causality remains a challenge due to the lack of randomized controlled trial data on this topic. We thus aimed to investigate the causal relationship between PA and the risk of developing CAD, CHF, and stroke. METHODS AND RESULTS Cross-ancestry meta-analysis of genome-wide association studies combining East Asian and European ancestry (1560 PA cases and 742 139 controls) was conducted to identify single-nucleotide variants that are associated with PA. Then, using the identified genetic variants as instrumental variables, we conducted the 2-sample Mendelian randomization analysis to investigate the causal relationship between PA and incident CAD, CHF, and stroke among both East Asian and European ancestry. Summary association results were extracted from large genome-wide association studies consortia. Our cross-ancestry meta-analysis of East Asian and European populations identified 7 genetic loci significantly associated with the risk of PA, for which the genes nearest to the lead variants were CASZ1, WNT2B, HOTTIP, LSP1, TBX3, RXFP2, and NDP. Among the East Asian population, the pooled odds ratio estimates using these 7 genetic instruments of PA were 1.07 (95% CI, 1.03-1.11) for CAD, 1.10 (95% CI, 1.01-1.20) for CHF, and 1.13 (95% CI, 1.09-1.18) for stroke. The results were consistent among the European population. CONCLUSIONS Our 2-sample Mendelian randomization study revealed that PA had increased risks of CAD, CHF, and stroke. These findings highlight that early and active screening of PA is critical to prevent future cardiovascular events.
Collapse
Affiliation(s)
- Kosuke Inoue
- Department of Social Epidemiology, Graduate School of Medicine Kyoto University Kyoto Japan
- Hakubi Center for Advanced Research Kyoto University Kyoto Japan
| | - Tatsuhiko Naito
- Department of Statistical Genetics Osaka University Graduate School of Medicine Suita Japan
- Laboratory for Systems Genetics RIKEN Center for Integrative Medical Sciences Yokohama Kanagawa Japan
| | - Ryosuke Fuji
- Institute for Biomedicine (Affiliated to the University of Lübeck) Eurac Research Bolzano Italy
- Department of Preventive Medical Science Fujita Health University School of Medical Sciences Toyoake Japan
| | - Kyuto Sonehara
- Department of Statistical Genetics Osaka University Graduate School of Medicine Suita Japan
- Laboratory for Systems Genetics RIKEN Center for Integrative Medical Sciences Yokohama Kanagawa Japan
- Department of Genome Informatics, Graduate School of Medicine The University of Tokyo Tokyo Japan
| | - Kenichi Yamamoto
- Department of Statistical Genetics Osaka University Graduate School of Medicine Suita Japan
| | - Ryuta Baba
- Department of Molecular and Internal Medicine Hiroshima University Hiroshima Japan
| | - Takaya Kodama
- Department of Molecular and Internal Medicine Hiroshima University Hiroshima Japan
| | - Yu Otagaki
- Department of Molecular and Internal Medicine Hiroshima University Hiroshima Japan
| | - Akira Okada
- Department of Molecular and Internal Medicine Hiroshima University Hiroshima Japan
| | - Kiyotaka Itcho
- Department of Molecular and Internal Medicine Hiroshima University Hiroshima Japan
| | - Kazuhiro Kobuke
- Department of Molecular and Internal Medicine Hiroshima University Hiroshima Japan
| | - Haruya Ohno
- Department of Molecular and Internal Medicine Hiroshima University Hiroshima Japan
| | - Takayuki Morisaki
- Division of Molecular Pathology, Institute of Medical Science The University of Tokyo Tokyo Japan
- Department of Internal Medicine, Institute of Medical Science The University of Tokyo Hospital Tokyo Japan
| | - Noboru Hattori
- Department of Molecular and Internal Medicine Hiroshima University Hiroshima Japan
| | - Atsushi Goto
- Department of Public Health Yokohama City University Yokohama Japan
| | - Tetsuo Nishikawa
- Endocrinology and Diabetes Center Yokohama Rosai Hospital Yokohama Japan
| | - Kenji Oki
- Department of Molecular and Internal Medicine Hiroshima University Hiroshima Japan
| | - Yukinori Okada
- Department of Statistical Genetics Osaka University Graduate School of Medicine Suita Japan
- Laboratory for Systems Genetics RIKEN Center for Integrative Medical Sciences Yokohama Kanagawa Japan
- Department of Genome Informatics, Graduate School of Medicine The University of Tokyo Tokyo Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives Osaka University Suita Japan
| |
Collapse
|
74
|
Li C, Malloy M, Ture SK, Nieves-Lopez B, Thibord F, Johnson AD, Morrell CN. G protein-coupled receptor kinase 5 regulates thrombin signaling in platelets. Res Pract Thromb Haemost 2024; 8:102556. [PMID: 39309233 PMCID: PMC11415800 DOI: 10.1016/j.rpth.2024.102556] [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: 04/17/2024] [Revised: 06/28/2024] [Accepted: 08/16/2024] [Indexed: 09/25/2024] Open
Abstract
Background Our prior genome-wide association study of thrombin-induced platelet aggregation identified a G protein-coupled receptor kinase 5 (GRK5) noncoding variant (rs10886430-G) that is strongly associated with increased platelet reactivity to thrombin. This variant predisposes to increased risk of stroke, pulmonary embolism, and venous thromboembolism. Objectives To determine role of platelet specific GRK5 in platelet responses to agonists and injury. Methods Platelets from GRK5 mutant mice have been shown to have increased thrombin sensitivity, indicating that GRK5 may be a negative regulator of platelet activation. However, this has not been studied in a platelet-specific manner. We therefore used platelet-specific GRK5 mutant mice and models of thrombosis and pulmonary embolism. Results We now demonstrate that mice lacking GRK5 specifically in platelets had a mild increase in thrombin responses in vitro and a shortened time to arterial thrombosis in vivo. In addition, platelet GRK5 mutant mice had increased thrombin but not collagen-induced thrombus burden in a mouse model of pulmonary embolism. Conclusion These data indicate that platelet GRK5 has a significant role in limiting platelet responses to thrombin.
Collapse
Affiliation(s)
- Chen Li
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Michael Malloy
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Sara K. Ture
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Benjamin Nieves-Lopez
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| | - Florian Thibord
- Population Sciences Branch, National Heart, Lung and Blood Institute, Framingham, Massachusetts, USA
| | - Andrew D. Johnson
- Population Sciences Branch, National Heart, Lung and Blood Institute, Framingham, Massachusetts, USA
| | - Craig N. Morrell
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
75
|
Pathan N, Kharod MK, Nawab S, Di Scipio M, Paré G, Chong M. Genetic Determinants of Vascular Dementia. Can J Cardiol 2024; 40:1412-1423. [PMID: 38579965 DOI: 10.1016/j.cjca.2024.03.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/20/2024] [Accepted: 03/29/2024] [Indexed: 04/07/2024] Open
Abstract
Vascular dementia (VaD) is a prevalent form of cognitive impairment with underlying vascular etiology. In this review, we examine recent genetic advancements in our understanding of VaD, encompassing a range of methodologies including genome-wide association studies, polygenic risk scores, heritability estimates, and family studies for monogenic disorders revealing the complex and heterogeneous nature of the disease. We report well known genetic associations and highlight potential pathways and mechanisms implicated in VaD and its pathological risk factors, including stroke, cerebral small vessel disease, and cerebral amyloid angiopathy. Moreover, we discuss important modifiable risk factors such as hypertension, diabetes, and dyslipidemia, emphasizing the importance of a multifactorial approach in prevention, treatment, and understanding the genetic basis of VaD. Last, we outline several areas of scientific advancements to improve clinical care, highlighting that large-scale collaborative efforts, together with an integromics approach can enhance the robustness of genetic discoveries. Indeed, understanding the genetics of VaD and its pathophysiological risk factors hold the potential to redefine VaD on the basis of molecular mechanisms and to generate novel diagnostic, prognostic, and therapeutic tools.
Collapse
Affiliation(s)
- Nazia Pathan
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada; Department of Pathology and Molecular Medicine, McMaster University, Michael G. DeGroote School of Medicine, Hamilton, Ontario, Canada
| | - Muskaan Kaur Kharod
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
| | - Sajjha Nawab
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
| | - Matteo Di Scipio
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada; Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Guillaume Paré
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada; Department of Pathology and Molecular Medicine, McMaster University, Michael G. DeGroote School of Medicine, Hamilton, Ontario, Canada; Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada; Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada.
| | - Michael Chong
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada; Department of Pathology and Molecular Medicine, McMaster University, Michael G. DeGroote School of Medicine, Hamilton, Ontario, Canada; Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada.
| |
Collapse
|
76
|
Debette S, Paré G. Stroke Genetics, Genomics, and Precision Medicine. Stroke 2024; 55:2163-2168. [PMID: 38511336 DOI: 10.1161/strokeaha.123.044212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Affiliation(s)
- Stéphanie Debette
- University of Bordeaux, INSERM, Bordeaux Population Health, France (S.D.)
- Department of Neurology, Institute for Neurodegenerative Diseases, Bordeaux University Hospital, France (S.D.)
| | - Guillaume Paré
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada (G.P.)
| |
Collapse
|
77
|
Li P, Liu M, He WM. Integrated Transcriptomic Analysis Reveals Reciprocal Interactions between SARS-CoV-2 Infection and Multi-Organ Dysfunction, Especially the Correlation of Renal Failure and COVID-19. Life (Basel) 2024; 14:960. [PMID: 39202702 PMCID: PMC11355357 DOI: 10.3390/life14080960] [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: 06/11/2024] [Revised: 07/22/2024] [Accepted: 07/29/2024] [Indexed: 09/03/2024] Open
Abstract
The COVID-19 pandemic, which is caused by the SARS-CoV-2 virus, has resulted in extensive health challenges globally. While SARS-CoV-2 primarily targets the respiratory system, clinical studies have revealed that it could also affect multiple organs, including the heart, kidneys, liver, and brain, leading to severe complications. To unravel the intricate molecular interactions between the virus and host tissues, we performed an integrated transcriptomic analysis to investigate the effects of SARS-CoV-2 on various organs, with a particular focus on the relationship between renal failure and COVID-19. A comparative analysis showed that SARS-CoV-2 triggers a systemic immune response in the brain, heart, and kidney tissues, characterized by significant upregulation of cytokine and chemokine secretion, along with enhanced migration of lymphocytes and leukocytes. A weighted gene co-expression network analysis demonstrated that SARS-CoV-2 could also induce tissue-specific transcriptional profiling. More importantly, single-cell sequencing revealed that COVID-19 patients with renal failure exhibited lower metabolic activity in lung epithelial and B cells, with reduced ligand-receptor interactions, especially CD226 and ICAM, suggesting a compromised immune response. A trajectory analysis revealed that COVID-19 patients with renal failure exhibited less mature alveolar type 1 cells. Furthermore, these patients showed potential fibrosis in the hearts, liver, and lung increased extracellular matrix remodeling activities. However, there was no significant metabolic dysregulation in the liver of COVID-19 patients with renal failure. Candidate drugs prediction by Drug Signatures database and LINCS L1000 Antibody Perturbations Database underscored the importance of considering multi-organ effects in COVID-19 management and highlight potential therapeutic strategies, including targeting viral entry and replication, controlling tissue fibrosis, and alleviating inflammation.
Collapse
Affiliation(s)
- Pai Li
- Capricorn Partner, 3000 Leuven, Belgium
| | - Meng Liu
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Wei-Ming He
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| |
Collapse
|
78
|
Karaaslanli A, Aşir F, Gürsoy GT, Tuncer MC. Biochanin A restored the blood-brain barrier in cerebral ischemia-reperfusion in rats. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2024; 70:e20240025. [PMID: 39045961 PMCID: PMC11288263 DOI: 10.1590/1806-9282.20240025] [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: 02/11/2024] [Accepted: 02/12/2024] [Indexed: 07/25/2024]
Abstract
OBJECTIVE Blood-brain barrier is a protective layer that regulates the influx and efflux of biological materials for cerebral tissue. The aim of this study was to investigate the effects of Biochanin A on cerebral histopathology and blood-brain barrier immunohistochemically. METHODS A total of 24 rats were assigned to three groups: sham, ischemia-reperfusion, and ischemia-reperfusion+Biochanin A. Ischemia-reperfusion was performed by occluding the left carotid artery for 2/24 h. Notably, 20 mg/kg Biochanin A was administered to rats for 7 days after ischemia-reperfusion. Blood was collected for malondialdehyde and total oxidant/antioxidant status analysis. Cerebral tissues were processed for histopathology and further for immunohistochemical analysis. RESULTS Malondialdehyde content with total oxidant status value was significantly increased and total antioxidant status values were significantly decreased in the ischemia-reperfusion group compared with the sham group. Biochanin A treatment significantly improved scores in the ischemia-reperfusion+Biochanin A group. The normal histological appearance was recorded in the cerebral sections of the sham group. Degenerated neurons and vascular structures with disrupted integrity of the cerebral cortex were observed after ischemia-reperfusion. Biochanin A alleviated the histopathology in the cerebrum in the ischemia-reperfusion+Biochanin A group. Ischemia-reperfusion injury decreased the expression of blood-brain barrier in the ischemia-reperfusion group compared to the sham group. Administration of Biochanin A upregulated the blood-brain barrier immunoreactivity in the cerebrum by restoring blood-brain barrier. CONCLUSION Cerebral ischemia-reperfusion caused an increase in oxidative stress and pathological lesions in the cerebrum. Biochanin A treatment restored the adverse effects of ischemia-reperfusion injury by restoring blood-brain barrier.
Collapse
Affiliation(s)
| | - Fırat Aşir
- Dicle University, Faculty of Medicine, Department of Histology and Embryology – Diyarbkır, Turkey
| | | | - Mehmet Cudi Tuncer
- Dicle University, Faculty of Medicine, Department of Anatomy – Diyarbakir, Turkey
| |
Collapse
|
79
|
Daghlas I, Gill D. Mechanisms of Hypercoagulability Driving Stroke Risk in Obesity: A Mendelian Randomization Study. Neurology 2024; 103:e209431. [PMID: 38861688 DOI: 10.1212/wnl.0000000000209431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Obesity is hypothesized to induce a hypercoagulable state that increases stroke risk. The molecular mechanisms underlying this association are largely uncharacterized. We aimed to apply mendelian randomization to identify whether the association of genetically proxied body mass index (BMI) with cardioembolic stroke risk is mediated by changes in levels of circulating coagulation factors. METHODS Genetic proxies for BMI and levels of circulating coagulation factors were obtained, respectively, from the Genetic Investigation of ANthropometric Traits consortium (n = 694,649) and deCODE cohort (n = 35,559). Genetic associations with cardioembolic stroke risk were obtained from the GIGASTROKE consortium (10,804 cases and 1,234,804 controls). We performed a two-sample mendelian randomization analysis testing the association of genetically proxied BMI with cardioembolic stroke risk, genetically proxied BMI with levels of coagulation factors, and genetically proxied levels of coagulation factors with cardioembolic stroke risk. These estimates were carried forward to mediation and sensitivity analyses. RESULTS A 1-SD increase in genetically proxied BMI associated with increased cardioembolic stroke risk (OR of cardioembolic stroke per 1-SD of BMI 1.20, 95% CI 1.08-1.33, p = 8.65 × 10-4) with similar findings in statistical sensitivity analyses more robust to the inclusion of pleiotropic variants. Genetically proxied BMI was further associated with increased levels of Factor VII, Factor Xa, Factor XI, and Protein S (all p < 5.9 × 10-6). Of these factors, genetically proxied levels of Factor XI were associated with cardioembolic stroke risk (OR of cardioembolic stroke per 1-SD increase in Factor XI levels 1.32, 1.19-1.46, p = 6.18 × 10-8). The mediated effect of genetically proxied BMI through Factor XI accounted for 26% (6%-49%) of the total effect of BMI on cardioembolic stroke. DISCUSSION Human genetic data support increased levels of Factor XI as a mechanistic explanation for how obesity increases cardioembolic stroke risk. The clinical relevance of this association warrants further investigation within ongoing clinical trials of Factor XI inhibition.
Collapse
Affiliation(s)
- Iyas Daghlas
- From the Department of Neurology (I.D.), University of California, San Francisco; and Department of Epidemiology and Biostatistics (D.G.), School of Public Health, Imperial College London, United Kingdom
| | - Dipender Gill
- From the Department of Neurology (I.D.), University of California, San Francisco; and Department of Epidemiology and Biostatistics (D.G.), School of Public Health, Imperial College London, United Kingdom
| |
Collapse
|
80
|
Ribeuz HL, Willer ASM, Chevalier B, Sancho M, Masson B, Eyries M, Jung V, Guerrera IC, Dutheil M, Jekmek KE, Laubry L, Carpentier G, Perez-Vizcaino F, Tu L, Guignabert C, Chaumais MC, Péchoux C, Humbert M, Hinzpeter A, Mercier O, Capuano V, Montani D, Antigny F. Role of KCNK3 Dysfunction in Dasatinib-associated Pulmonary Arterial Hypertension and Endothelial Cell Dysfunction. Am J Respir Cell Mol Biol 2024; 71:95-109. [PMID: 38546978 DOI: 10.1165/rcmb.2023-0185oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 03/27/2024] [Indexed: 07/02/2024] Open
Abstract
Pulmonary arterial (PA) hypertension (PAH) is a severe cardiopulmonary disease that may be triggered by exposure to drugs such as dasatinib or facilitated by genetic predispositions. The incidence of dasatinib-associated PAH is estimated at 0.45%, suggesting individual predispositions. The mechanisms of dasatinib-associated PAH are still incomplete. We discovered a KCNK3 gene (Potassium channel subfamily K member 3; coding for outward K+ channel) variant in a patient with dasatinib-associated PAH and investigated the impact of this variant on KCNK3 function. Additionally, we assessed the effects of dasatinib exposure on KCNK3 expression. In control human PA smooth muscle cells (hPASMCs) and human pulmonary endothelial cells (hPECs), we evaluated the consequences of KCNK3 knockdown on cell migration, mitochondrial membrane potential, ATP production, and in vitro tube formation. Using mass spectrometry, we determined the KCNK3 interactome. Patch-clamp experiments revealed that the KCNK3 variant represents a loss-of-function variant. Dasatinib contributed to PA constriction by decreasing KCNK3 function and expression. In control hPASMCs, KCNK3 knockdown promotes mitochondrial membrane depolarization and glycolytic shift. Dasatinib exposure or KCNK3 knockdown reduced the number of caveolae in hPECs. Moreover, KCNK3 knockdown in control hPECs reduced migration, proliferation, and in vitro tubulogenesis. Using proximity labeling and mass spectrometry, we identified the KCNK3 interactome, revealing that KCNK3 interacts with various proteins across different cellular compartments. We identified a novel pathogenic variant in KCNK3 and showed that dasatinib downregulates KCNK3, emphasizing the relationship between dasatinib-associated PAH and KCNK3 dysfunction. We demonstrated that a loss of KCNK3-dependent signaling contributes to endothelial dysfunction in PAH and glycolytic switch of hPASMCs.
Collapse
Affiliation(s)
- Hélène Le Ribeuz
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Anaïs Saint-Martin Willer
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Benoit Chevalier
- Paris Cité University, CNRS, INSERM, Institut Necker Enfants Malades-INEM, Paris, France
| | - Maria Sancho
- Department of Physiology and
- Department of Pharmacology, University of Vermont, Burlington, Vermont
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Bastien Masson
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Mélanie Eyries
- Genetics Department, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Vincent Jung
- INSERM US24/CNRS UAR3633, Proteomic Platform Necker, Université Paris Cité-Federative Research Structure Necker, Paris, France
| | - Ida Chiara Guerrera
- INSERM US24/CNRS UAR3633, Proteomic Platform Necker, Université Paris Cité-Federative Research Structure Necker, Paris, France
| | - Mary Dutheil
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Kristelle El Jekmek
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Loann Laubry
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Gilles Carpentier
- Gly-CRRET Research Unit 4397, Paris-Est Créteil University, Créteil, France
| | - Francisco Perez-Vizcaino
- Department of Pharmacology and Toxicology, Faculty of Medicine, University Complutense of Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Ly Tu
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Christophe Guignabert
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Marie-Camille Chaumais
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
- Paris-Saclay University, Faculty of Pharmacy, Orsay, France
- Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | - Christine Péchoux
- Paris-Saclay University, INRAE, AgroparisTech, GABI, Jouy-en-Josas, France
| | - Marc Humbert
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
- Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | - Alexandre Hinzpeter
- Paris Cité University, CNRS, INSERM, Institut Necker Enfants Malades-INEM, Paris, France
| | - Olaf Mercier
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
- Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Groupe Hospitalier Paris Saint-Joseph-Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - Véronique Capuano
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | - David Montani
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
- Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | - Fabrice Antigny
- Paris-Saclay University, Faculty of Medecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Le Plessis-Robinson, France
| |
Collapse
|
81
|
Xue B, Jian X, Peng L, Wu C, Fahira A, Syed AAS, Xia D, Wang B, Niu M, Jiang Y, Ding Y, Gao C, Zhao X, Zhang Q, Shi Y, Li Z. Dissecting the genetic and causal relationship between sleep-related traits and common brain disorders. Sleep Med 2024; 119:201-209. [PMID: 38703603 DOI: 10.1016/j.sleep.2024.04.032] [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: 02/28/2024] [Revised: 04/14/2024] [Accepted: 04/24/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND There is a profound connection between abnormal sleep patterns and brain disorders, suggesting a shared influential association. However, the shared genetic basis and potential causal relationships between sleep-related traits and brain disorders are yet to be fully elucidated. METHODS Utilizing linkage disequilibrium score regression (LDSC) and bidirectional two-sample univariable Mendelian Randomization (UVMR) analyses with large-scale GWAS datasets, we investigated the genetic correlations and causal associations across six sleep traits and 24 prevalent brain disorders. Additionally, a multivariable Mendelian Randomization (MVMR) analysis evaluated the cumulative effects of various sleep traits on each brain disorder, complemented by genetic loci characterization to pinpoint pertinent genes and pathways. RESULTS LDSC analysis identified significant genetic correlations in 66 out of 144 (45.8 %) pairs between sleep-related traits and brain disorders, with the most pronounced correlations observed in psychiatric disorders (66 %, 48/72). UVMR analysis identified 29 causal relationships (FDR<0.05) between sleep traits and brain disorders, with 19 associations newly discovered according to our knowledge. Notably, major depression, attention-deficit/hyperactivity disorder, bipolar disorder, cannabis use disorder, and anorexia nervosa showed bidirectional causal relations with sleep traits, especially insomnia's marked influence on major depression (IVW beta 0.468, FDR = 5.24E-09). MVMR analysis revealed a nuanced interplay among various sleep traits and their impact on brain disorders. Genetic loci characterization underscored potential genes, such as HOXB2, while further enrichment analyses illuminated the importance of synaptic processes in these relationships. CONCLUSIONS This study provides compelling evidence for the causal relationships and shared genetic backgrounds between common sleep-related traits and brain disorders.
Collapse
Affiliation(s)
- Baiqiang Xue
- The Affiliated Hospital of Qingdao University, The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, 266003, China; School of Public Health, Qingdao University, Qingdao, China
| | - Xuemin Jian
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Lixia Peng
- The Affiliated Hospital of Qingdao University, The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, 266003, China; School of Pharmacy, Qingdao University, Qingdao, 266003, China
| | - Chuanhong Wu
- The Affiliated Hospital of Qingdao University, The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, 266003, China; School of Basic Medicine, Qingdao University, Qingdao, 266003, China
| | - Aamir Fahira
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Ali Alamdar Shah Syed
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Disong Xia
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Baokun Wang
- The Affiliated Hospital of Qingdao University, The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, 266003, China; School of Pharmacy, Qingdao University, Qingdao, 266003, China
| | - Mingming Niu
- The Affiliated Hospital of Qingdao University, The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, 266003, China; School of Public Health, Qingdao University, Qingdao, China
| | - Yajie Jiang
- The Affiliated Hospital of Qingdao University, The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, 266003, China; School of Public Health, Qingdao University, Qingdao, China
| | - Yonghe Ding
- The Affiliated Hospital of Qingdao University, The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, 266003, China; School of Public Health, Qingdao University, Qingdao, China
| | - Chengwen Gao
- The Affiliated Hospital of Qingdao University, The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, 266003, China
| | - Xiangzhong Zhao
- The Affiliated Hospital of Qingdao University, The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, 266003, China
| | - Qian Zhang
- The Affiliated Hospital of Qingdao University, The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, 266003, China
| | - Yongyong Shi
- The Affiliated Hospital of Qingdao University, The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, 266003, China; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030, China; School of Basic Medicine, Qingdao University, Qingdao, 266003, China; Shanghai Clinical Research Center for Mental Health, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Shandong Provincial Key Laboratory of Metabolic Disease & the Metabolic Disease Institute of Qingdao University, Qingdao, 266003, China; Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, 200030, China; Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, 200030, China; Department of Psychiatry, the First Teaching Hospital of Xinjiang Medical University, Urumqi, 830054, China; Changning Mental Health Center, Shanghai, 200042, China.
| | - Zhiqiang Li
- The Affiliated Hospital of Qingdao University, The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, 266003, China; School of Public Health, Qingdao University, Qingdao, China; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030, China; School of Pharmacy, Qingdao University, Qingdao, 266003, China; School of Basic Medicine, Qingdao University, Qingdao, 266003, China; Shanghai Clinical Research Center for Mental Health, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Shandong Provincial Key Laboratory of Metabolic Disease & the Metabolic Disease Institute of Qingdao University, Qingdao, 266003, China; Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, 200030, China.
| |
Collapse
|
82
|
Soremekun O, Mhlongwe T, Kirabo G, Akinyele C, Chikowore T, Fatumo S, Gill D. Genetically Proxied Lipid-Lowering Drug Target Perturbation and Ischemic Stroke Risk in European and African Ancestry Individuals: Mendelian Randomization Study. Stroke 2024; 55:e185-e186. [PMID: 38860386 PMCID: PMC11198944 DOI: 10.1161/strokeaha.123.045261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Affiliation(s)
- Opeyemi Soremekun
- The African Computational Genomics Research Group, Medical Research Council/Uganda Virus Research Institute, and London School of Hygiene and Tropical Medicine (MRC/UVRI & LSHTM), Entebbe, Uganda (O.S., G.K., S.F.)
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom (O.S.)
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa (O.S., T.M.)
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom (O.S, D.G.)
| | - Thobeka Mhlongwe
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa (O.S., T.M.)
| | - Gloria Kirabo
- The African Computational Genomics Research Group, Medical Research Council/Uganda Virus Research Institute, and London School of Hygiene and Tropical Medicine (MRC/UVRI & LSHTM), Entebbe, Uganda (O.S., G.K., S.F.)
| | | | - Tinashe Chikowore
- MRC/Wits Developmental Pathways for Health Research Unit, Department of Pediatrics, Faculty of Health Sciences (T.C.), University of the Witwatersrand, Johannesburg, South Africa
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences (T.C.), University of the Witwatersrand, Johannesburg, South Africa
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA (T.C.)
- Harvard Medical School, Boston, MA (T.C.)
| | - Segun Fatumo
- The African Computational Genomics Research Group, Medical Research Council/Uganda Virus Research Institute, and London School of Hygiene and Tropical Medicine (MRC/UVRI & LSHTM), Entebbe, Uganda (O.S., G.K., S.F.)
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine London, United Kingdom (S.F.)
- Precision Healthcare University Research Institute Queen Mary University of London, United Kingdom (S.F.)
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom (O.S, D.G.)
| |
Collapse
|
83
|
Hilkens NA, Casolla B, Leung TW, de Leeuw FE. Stroke. Lancet 2024; 403:2820-2836. [PMID: 38759664 DOI: 10.1016/s0140-6736(24)00642-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/19/2024] [Accepted: 03/26/2024] [Indexed: 05/19/2024]
Abstract
Stroke affects up to one in five people during their lifetime in some high-income countries, and up to almost one in two in low-income countries. Globally, it is the second leading cause of death. Clinically, the disease is characterised by sudden neurological deficits. Vascular aetiologies contribute to the most common causes of ischaemic stroke, including large artery disease, cardioembolism, and small vessel disease. Small vessel disease is also the most frequent cause of intracerebral haemorrhage, followed by macrovascular causes. For acute ischaemic stroke, multimodal CT or MRI reveal infarct core, ischaemic penumbra, and site of vascular occlusion. For intracerebral haemorrhage, neuroimaging identifies early radiological markers of haematoma expansion and probable underlying cause. For intravenous thrombolysis in ischaemic stroke, tenecteplase is now a safe and effective alternative to alteplase. In patients with strokes caused by large vessel occlusion, the indications for endovascular thrombectomy have been extended to include larger core infarcts and basilar artery occlusion, and the treatment time window has increased to up to 24 h from stroke onset. Regarding intracerebral haemorrhage, prompt delivery of bundled care consisting of immediate anticoagulation reversal, simultaneous blood pressure lowering, and prespecified stroke unit protocols can improve clinical outcomes. Guided by underlying stroke mechanisms, secondary prevention encompasses pharmacological, vascular, or endovascular interventions and lifestyle modifications.
Collapse
Affiliation(s)
- Nina A Hilkens
- Department of Neurology, Radboud University Nijmegen Medical Center, Nijmegen, Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
| | - Barbara Casolla
- Université Nice Cote d'Azur UR2CA-URRIS, Stroke Unit, CHU Pasteur 2, Nice, France
| | - Thomas W Leung
- Division of Neurology, Department of Medicine and Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| | - Frank-Erik de Leeuw
- Department of Neurology, Radboud University Nijmegen Medical Center, Nijmegen, Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands.
| |
Collapse
|
84
|
Dunca D, Chopade S, Gordillo-Marañón M, Hingorani AD, Kuchenbaecker K, Finan C, Schmidt AF. Comparing the effects of CETP in East Asian and European ancestries: a Mendelian randomization study. Nat Commun 2024; 15:5302. [PMID: 38906890 PMCID: PMC11192935 DOI: 10.1038/s41467-024-49109-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 05/24/2024] [Indexed: 06/23/2024] Open
Abstract
CETP inhibitors are a class of lipid-lowering drugs in development for treatment of coronary heart disease (CHD). Genetic studies in East Asian ancestry have interpreted the lack of CETP signal with low-density lipoprotein cholesterol (LDL-C) and lack of drug target Mendelian randomization (MR) effect on CHD as evidence that CETP inhibitors might not be effective in East Asian participants. Capitalizing on recent increases in sample size of East Asian genetic studies, we conducted a drug target MR analysis, scaled to a standard deviation increase in high-density lipoprotein cholesterol. Despite finding evidence for possible neutral effects of lower CETP levels on LDL-C, systolic blood pressure and pulse pressure in East Asians (interaction p-values < 1.6 × 10-3), effects on cardiovascular outcomes were similarly protective in both ancestry groups. In conclusion, on-target inhibition of CETP is anticipated to decrease cardiovascular disease in individuals of both European and East Asian ancestries.
Collapse
Affiliation(s)
- Diana Dunca
- Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, United Kingdom.
- UCL Genetics Institute, University College London, London, UK.
| | - Sandesh Chopade
- Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, United Kingdom
| | - María Gordillo-Marañón
- Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, United Kingdom
| | - Aroon D Hingorani
- Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, United Kingdom
- UCL British Heart Foundation Research Accelerator, London, UK
- Health Data Research UK, London, UK
| | - Karoline Kuchenbaecker
- UCL Genetics Institute, University College London, London, UK
- Division of Psychiatry, University College London, London, UK
| | - Chris Finan
- Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, United Kingdom
- UCL British Heart Foundation Research Accelerator, London, UK
- Health Data Research UK, London, UK
| | - Amand F Schmidt
- Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, United Kingdom
- UCL British Heart Foundation Research Accelerator, London, UK
- Department of Cardiology, Amsterdam UMC Heart Center, Amsterdam, The Netherlands
| |
Collapse
|
85
|
Bergstedt J, Pasman JA, Ma Z, Harder A, Yao S, Parker N, Treur JL, Smit DJA, Frei O, Shadrin AA, Meijsen JJ, Shen Q, Hägg S, Tornvall P, Buil A, Werge T, Hjerling-Leffler J, Als TD, Børglum AD, Lewis CM, McIntosh AM, Valdimarsdóttir UA, Andreassen OA, Sullivan PF, Lu Y, Fang F. Distinct biological signature and modifiable risk factors underlie the comorbidity between major depressive disorder and cardiovascular disease. NATURE CARDIOVASCULAR RESEARCH 2024; 3:754-769. [PMID: 38898929 PMCID: PMC11182748 DOI: 10.1038/s44161-024-00488-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 05/08/2024] [Indexed: 06/21/2024]
Abstract
Major depressive disorder (MDD) and cardiovascular disease (CVD) are often comorbid, resulting in excess morbidity and mortality. Here we show that CVDs share most of their genetic risk factors with MDD. Multivariate genome-wide association analysis of shared genetic liability between MDD and atherosclerotic CVD revealed seven loci and distinct patterns of tissue and brain cell-type enrichments, suggesting the involvement of the thalamus. Part of the genetic overlap was explained by shared inflammatory, metabolic and psychosocial or lifestyle risk factors. Our data indicated causal effects of genetic liability to MDD on CVD risk, but not from most CVDs to MDD, and showed that the causal effects were partly explained by metabolic and psychosocial or lifestyle factors. The distinct signature of MDD-atherosclerotic CVD comorbidity suggests an immunometabolic subtype of MDD that is more strongly associated with CVD than overall MDD. In summary, we identified biological mechanisms underlying MDD-CVD comorbidity and modifiable risk factors for prevention of CVD in individuals with MDD.
Collapse
Affiliation(s)
- Jacob Bergstedt
- Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Joëlle A. Pasman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Ziyan Ma
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Arvid Harder
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Shuyang Yao
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Nadine Parker
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Jorien L. Treur
- Genetic Epidemiology, Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Dirk J. A. Smit
- Genetic Epidemiology, Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Oleksandr Frei
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Centre for Bioinformatics, Department of Informatics, University of Oslo, Oslo, Norway
| | - Alexey A. Shadrin
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Joeri J. Meijsen
- Institute of Biological Psychiatry, Mental Health Center Sct. Hans, Mental Health Services Copenhagen, Roskilde, Denmark
| | - Qing Shen
- Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Clinical Research Center for Mental Disorders, Shanghai Pudong New Area Mental Health Center, Tongji University School of Medicine, Shanghai, China
- Institute for Advanced Study, Tongji University, Shanghai, China
| | - Sara Hägg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Per Tornvall
- Department of Clinical Science and Education Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Alfonso Buil
- Institute of Biological Psychiatry, Mental Health Center Sct. Hans, Mental Health Services Copenhagen, Roskilde, Denmark
| | - Thomas Werge
- Institute of Biological Psychiatry, Mental Health Center Sct. Hans, Mental Health Services Copenhagen, Roskilde, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Hjerling-Leffler
- Department Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Thomas D. Als
- Department of Molecular Medicine (MOMA), Molecular Diagnostic Laboratory, Aarhus University Hospital, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, Aarhus, Denmark
| | - Anders D. Børglum
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, Aarhus, Denmark
| | - Cathryn M. Lewis
- Social, Genetic and Developmental Psychiatry Centre, King’s College London, London, UK
- Department of Medical and Molecular Genetics, King’s College London, London, UK
| | - Andrew M. McIntosh
- Centre for Clinical Brain Sciences, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK
- Centre for Genomics and Experimental Medicine, University of Edinburgh, Edinburgh, UK
| | - Unnur A. Valdimarsdóttir
- Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Centre of Public Health Sciences, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- Department of Epidemiology, Harvard TH Chan School of Public Health, Harvard University, Boston, MA USA
| | - Ole A. Andreassen
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Patrick F. Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Yi Lu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Fang Fang
- Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
86
|
Kojima N, Koido M, He Y, Shimmori Y, Hachiya T, Japan B, Debette S, Kamatani Y. Recurrent stroke prediction by applying a stroke polygenic risk score in the Japanese population. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.17.24309034. [PMID: 39371120 PMCID: PMC11451717 DOI: 10.1101/2024.06.17.24309034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Background Recently, various polygenic risk score (PRS)-based methods were developed to improve stroke prediction. However, current PRSs (including cross-ancestry PRS) poorly predict recurrent stroke. Here, we aimed to determine whether the best PRS for Japanese individuals can also predict stroke recurrence in this population by extensively comparing the methods and maximizing the predictive performance for stroke onset. Methods We used data from the BioBank Japan (BBJ) 1st cohort (n=179,938) to derive and optimize the PRSs using a 10-fold cross-validation. We integrated the optimized PRSs for multiple traits, such as vascular risk factors and stroke subtypes to generate a single PRS using the meta-scoring approach (metaGRS). We used an independent BBJ 2nd cohort (n=41,929) as a test sample to evaluate the association of the metaGRS with stroke and recurrent stroke. Results We analyzed recurrent stroke cases (n=174) and non-recurrent stroke controls (n=1,153) among subjects within the BBJ 2nd cohort. After adjusting for known risk factors, metaGRS was associated with stroke recurrence (adjusted OR per SD 1.18 [95% CI: 1.00-1.39, p=0.044]), although no significant correlation was observed with the published PRSs. We administered three distinct tests to consider the potential index event bias; however, the outcomes derived from these examinations did not provide any significant indication of the influence of index event bias. The high metaGRS group without a history of hypertension had a higher risk of stroke recurrence than that of the low metaGRS group (adjusted OR 2.24 [95% CI: 1.07-4.66, p=0.032]). However, this association was weak in the hypertension group (adjusted OR 1.21 [95% CI: 0.69-2.13, p=0.50]). Conclusions The metaGRS developed in a Japanese cohort predicted stroke recurrence in an independent cohort of patients. In particular, it predicted an increased risk of recurrence among stroke patients without hypertension. These findings provide clues for additional genetic risk stratification and help in developing personalized strategies for stroke recurrence prevention.
Collapse
Affiliation(s)
- Naoki Kojima
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Masaru Koido
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Yunye He
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuka Shimmori
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Tsuyoshi Hachiya
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | | | - Stéphanie Debette
- Bordeaux Population Health Research Center, University of Bordeaux, Inserm, UMR 1219, Bordeaux, France
- Department of Neurology, Institute for Neurodegenerative Diseases, CHU de Bordeaux, Bordeaux, France
| | - Yoichiro Kamatani
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
87
|
Xu W, Liang X, Chen L, Hong W, Hu X. Biobanks in chronic disease management: A comprehensive review of strategies, challenges, and future directions. Heliyon 2024; 10:e32063. [PMID: 38868047 PMCID: PMC11168399 DOI: 10.1016/j.heliyon.2024.e32063] [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: 12/22/2023] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/14/2024] Open
Abstract
Biobanks, through the collection and storage of patient blood, tissue, genomic, and other biological samples, provide unique and rich resources for the research and management of chronic diseases such as cardiovascular diseases, diabetes, and cancer. These samples contain valuable cellular and molecular level information that can be utilized to decipher the pathogenesis of diseases, guide the development of novel diagnostic technologies, treatment methods, and personalized medical strategies. This article first outlines the historical evolution of biobanks, their classification, and the impact of technological advancements. Subsequently, it elaborates on the significant role of biobanks in revealing molecular biomarkers of chronic diseases, promoting the translation of basic research to clinical applications, and achieving individualized treatment and management. Additionally, challenges such as standardization of sample processing, information privacy, and security are discussed. Finally, from the perspectives of policy support, regulatory improvement, and public participation, this article provides a forecast on the future development directions of biobanks and strategies to address challenges, aiming to safeguard and enhance their unique advantages in supporting chronic disease prevention and treatment.
Collapse
Affiliation(s)
- Wanna Xu
- Shenzhen Center for Chronic Disease Control, Shenzhen Institute of Dermatology, Shenzhen, 518020, China
| | - Xiongshun Liang
- Shenzhen Center for Chronic Disease Control, Shenzhen Institute of Dermatology, Shenzhen, 518020, China
| | - Lin Chen
- Shenzhen Center for Chronic Disease Control, Shenzhen Institute of Dermatology, Shenzhen, 518020, China
| | - Wenxu Hong
- Shenzhen Center for Chronic Disease Control, Shenzhen Institute of Dermatology, Shenzhen, 518020, China
| | - Xuqiao Hu
- Shenzhen Center for Chronic Disease Control, Shenzhen Institute of Dermatology, Shenzhen, 518020, China
- Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| |
Collapse
|
88
|
Li H, Zhang H, Hua W, Liu H, Zhang B, Dong H, Liu J, Zhou Y, Yang P, Jing M. Causal relationship between gut microbiota and functional outcomes after ischemic stroke: A comprehensive Mendelian randomization study. J Stroke Cerebrovasc Dis 2024; 33:107814. [PMID: 38880364 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/22/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024] Open
Abstract
AIMS To investigate the association of the genetic predisposition of specific gut microbiotas with the clinical outcome of ischemic stroke. METHODS We leveraged publicly available genome-wide association study (GWAS) data to perform Mendelian randomization (MR) analysis. The gut microbiota-related GWAS data from 18,340 individuals from the international consortium MiBioGen was used. The summary data for functional outcomes after ischemic stroke was obtained from the Genetics of Ischemic Stroke Functional Outcome (GISCOME) network meta-analysis. The primary outcomes were judged by the modified Rankin Scale (mRS). The principal analyses were conducted using the inverse-variance weighted (IVW) MR method. The Cochran's Q test, weighted median, MR-Egger regression, leave-one-SNP-out analysis, MR-Pleiotropy Residual Sum, and Outlier methods were adopted as sensitivity analyses. Furthermore, we performed bi-directional MR analysis and the MR Steiger directionality test to examine the direction of the causal relations. RESULTS The results demonstrated that the genetic predisposition of genus Lactococcus, genus Ruminococcaceae NK4A214 group, family Peptostreptococcaceae, and genus Odoribacter was positively associated with favorable functional outcome after ischemic stroke. Genus Collinsella, genus Ruminococcaceae UCG005, genus Akkermansia, genus Eubacterium oxidoreducens group, and family Verrucomicrobiaceae were identified to be associated with worse functional outcomes after ischemic stroke. Our results showed no evidence of heterogeneity, directional pleiotropic effects, or collider bias, and the sensitivity of our analysis was acceptable. CONCLUSION The genetic predisposition of different gut microbiotas was associated with the clinical outcome of ischemic stroke. Microbiota adjustment was a promising method to improve the clinical outcome of ischemic stroke.
Collapse
Affiliation(s)
- He Li
- Emergency Department, PLA Naval Medical Center, Shanghai, China; Neurovascular Center, Naval Medical University Changhai hospital, Shanghai, China
| | - Haojun Zhang
- Emergency Department, PLA Naval Medical Center, Shanghai, China
| | - Weilong Hua
- Neurovascular Center, Naval Medical University Changhai hospital, Shanghai, China
| | - Hanchen Liu
- Neurovascular Center, Naval Medical University Changhai hospital, Shanghai, China
| | - Boyu Zhang
- Emergency Department, PLA Naval Medical Center, Shanghai, China
| | - Hui Dong
- Emergency Department, PLA Naval Medical Center, Shanghai, China
| | - Jianmin Liu
- Neurovascular Center, Naval Medical University Changhai hospital, Shanghai, China
| | - Yu Zhou
- Neurovascular Center, Naval Medical University Changhai hospital, Shanghai, China.
| | - Pengfei Yang
- Emergency Department, PLA Naval Medical Center, Shanghai, China.
| | - Mei Jing
- Neurovascular Center, Naval Medical University Changhai hospital, Shanghai, China.
| |
Collapse
|
89
|
Antikainen AA, Haukka JK, Kumar A, Syreeni A, Hägg-Holmberg S, Ylinen A, Kilpeläinen E, Kytölä A, Palotie A, Putaala J, Thorn LM, Harjutsalo V, Groop PH, Sandholm N. Whole-genome sequencing identifies variants in ANK1, LRRN1, HAS1, and other genes and regulatory regions for stroke in type 1 diabetes. Sci Rep 2024; 14:13453. [PMID: 38862513 PMCID: PMC11166668 DOI: 10.1038/s41598-024-61840-7] [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: 09/25/2023] [Accepted: 05/10/2024] [Indexed: 06/13/2024] Open
Abstract
Individuals with type 1 diabetes (T1D) carry a markedly increased risk of stroke, with distinct clinical and neuroimaging characteristics as compared to those without diabetes. Using whole-exome or whole-genome sequencing of 1,051 individuals with T1D, we aimed to find rare and low-frequency genomic variants associated with stroke in T1D. We analysed the genome comprehensively with single-variant analyses, gene aggregate analyses, and aggregate analyses on genomic windows, enhancers and promoters. In addition, we attempted replication in T1D using a genome-wide association study (N = 3,945) and direct genotyping (N = 3,263), and in the general population from the large-scale population-wide FinnGen project and UK Biobank summary statistics. We identified a rare missense variant on SREBF1 exome-wide significantly associated with stroke (rs114001633, p.Pro227Leu, p-value = 7.30 × 10-8), which replicated for hemorrhagic stroke in T1D. Using gene aggregate analysis, we identified exome-wide significant genes: ANK1 and LRRN1 displayed replication evidence in T1D, and LRRN1, HAS1 and UACA in the general population (UK Biobank). Furthermore, we performed sliding-window analyses and identified 14 genome-wide significant windows for stroke on 4q33-34.1, of which two replicated in T1D, and a suggestive genomic window on LINC01500, which replicated in T1D. Finally, we identified a suggestively stroke-associated TRPM2-AS promoter (p-value = 5.78 × 10-6) with borderline significant replication in T1D, which we validated with an in vitro cell-based assay. Due to the rarity of the identified genetic variants, future replication of the genomic regions represented here is required with sequencing of individuals with T1D. Nevertheless, we here report the first genome-wide analysis on stroke in individuals with diabetes.
Collapse
Affiliation(s)
- Anni A Antikainen
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jani K Haukka
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anmol Kumar
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anna Syreeni
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Stefanie Hägg-Holmberg
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anni Ylinen
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Elina Kilpeläinen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Anastasia Kytölä
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Analytic and Translational Genetics Unit, Department of Medicine, Department of Neurology and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- The Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jukka Putaala
- Neurology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Lena M Thorn
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Valma Harjutsalo
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia.
| | - Niina Sandholm
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
90
|
Wu J, Wang Y, Vlasschaert C, Lali R, Feiner J, Gaheer P, Yang S, Perrot N, Chong M, Paré G, Lanktree MB. Kidney Volume and Risk of Incident Kidney Outcomes. J Am Soc Nephrol 2024; 35:00001751-990000000-00349. [PMID: 38857205 PMCID: PMC11387033 DOI: 10.1681/asn.0000000000000419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Low total kidney volume (TKV) is a risk factor for chronic kidney disease (CKD). However, evaluations of nonlinear relationships, incident events, causal inference, and prognostic utility beyond traditional biomarkers are lacking. METHODS TKV, height-adjusted TKV, and body surface area-adjusted TKV (BSA-TKV) of 34,595 White British ancestry participants were derived from the UK Biobank. Association with incident CKD, acute kidney injury (AKI), and cardiovascular events were assessed with Cox proportional hazard models. Prognostic thresholds for CKD risk stratification were identified using a modified Mazumdar method with bootstrap resampling. Two-sample Mendelian randomization was performed to assess the bidirectional association of genetically predicted TKV with kidney and cardiovascular traits. RESULTS Adjusted for eGFR and albuminuria, a lower TKV of 10 mL was associated with a 6% higher risk of incident CKD (hazard ratio [HR] 1.06, 95% confidence interval [CI] 1.03 to 1.08, P = 5.8 x 10-6) in contrast to no association with incident AKI (HR 1.00, 95% CI 0.98 to 1.02, P = 0.66). Comparison of nested models demonstrated improved accuracy over the CKD Prognosis Consortium Incident CKD Risk Score with the addition of BSA-TKV or prognostic thresholds at 119 (10th percentile) and 145 mL/m2 (50th percentile). In Mendelian randomization, a lower genetically predicted TKV by 10 mL was associated with 10% higher CKD risk (odds ratio [OR] 1.10, 95% CI 1.06 to 1.14, P = 1.3 x 10-7). Reciprocally, an elevated risk of genetically predicted CKD by 2-fold was associated with a lower TKV by 7.88 mL (95% CI -9.81 to -5.95, P = 1.2 x 10-15). There were no significant observational or Mendelian randomization associations of TKV with cardiovascular complications. CONCLUSIONS Kidney volume was associated with incident CKD independent of traditional risk factors including baseline eGFR and albuminuria. Mendelian randomization demonstrated a bidirectional relationship between kidney volume and CKD.
Collapse
Affiliation(s)
- Jianhan Wu
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada
| | - Yifan Wang
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada
| | | | - Ricky Lali
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada
| | - James Feiner
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada
| | - Pukhraj Gaheer
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada
| | - Serena Yang
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada
| | - Nicolas Perrot
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada
| | - Michael Chong
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Guillaume Paré
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Matthew B Lanktree
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada
- Division of Nephrology, St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| |
Collapse
|
91
|
Jiménez-Balado J, Fernández-Pérez I, Gallego-Fábrega C, Lazcano U, Soriano-Tárraga C, Vallverdú-Prats M, Mola-Caminal M, Rey-Álvarez L, Macias-Gómez A, Suárez-Pérez A, Giralt-Steinhauer E, Rodríguez-Campello A, Cuadrado-Godia E, Ois Á, Esteller M, Roquer J, Fernández-Cadenas I, Jiménez-Conde J. DNA methylation and stroke prognosis: an epigenome-wide association study. Clin Epigenetics 2024; 16:75. [PMID: 38845005 PMCID: PMC11155152 DOI: 10.1186/s13148-024-01690-2] [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: 03/28/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND AND AIMS Stroke is the leading cause of adult-onset disability. Although clinical factors influence stroke outcome, there is a significant variability among individuals that may be attributed to genetics and epigenetics, including DNA methylation (DNAm). We aimed to study the association between DNAm and stroke prognosis. METHODS AND RESULTS To that aim, we conducted a two-phase study (discovery-replication and meta-analysis) in Caucasian patients with ischemic stroke from two independent centers (BasicMar [discovery, N = 316] and St. Pau [replication, N = 92]). Functional outcome was assessed using the modified Rankin Scale (mRS) at three months after stroke, being poor outcome defined as mRS > 2. DNAm was determined using the 450K and EPIC BeadChips in whole-blood samples collected within the first 24 h. We searched for differentially methylated positions (DMPs) in 370,344 CpGs, and candidates below p-value < 10-5 were subsequently tested in the replication cohort. We then meta-analyzed DMP results from both cohorts and used them to identify differentially methylated regions (DMRs). After doing the epigenome-wide association study, we found 29 DMPs at p-value < 10-5 and one of them was replicated: cg24391982, annotated to thrombospondin-2 (THBS2) gene (p-valuediscovery = 1.54·10-6; p-valuereplication = 9.17·10-4; p-valuemeta-analysis = 6.39·10-9). Besides, four DMRs were identified in patients with poor outcome annotated to zinc finger protein 57 homolog (ZFP57), Arachidonate 12-Lipoxygenase 12S Type (ALOX12), ABI Family Member 3 (ABI3) and Allantoicase (ALLC) genes (p-value < 1·10-9 in all cases). DISCUSSION Patients with poor outcome showed a DMP at THBS2 and four DMRs annotated to ZFP57, ALOX12, ABI3 and ALLC genes. This suggests an association between stroke outcome and DNAm, which may help identify new stroke recovery mechanisms.
Collapse
Affiliation(s)
- Joan Jiménez-Balado
- Neurovascular Research Group, Department of Neurology, Hospital del Mar Research Institute, C/ del Dr. Aiguader, 88, 08003, Barcelona, Spain.
| | - Isabel Fernández-Pérez
- Neurovascular Research Group, Department of Neurology, Hospital del Mar Research Institute, C/ del Dr. Aiguader, 88, 08003, Barcelona, Spain
- Medicine Department, Autonomous University of Barcelona, Barcelona, Spain
| | | | - Uxue Lazcano
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Biscaia, Spain
| | - Carolina Soriano-Tárraga
- Department of Psychiatry, NeuroGenomics and Informatics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Marta Vallverdú-Prats
- Neurovascular Research Group, Department of Neurology, Hospital del Mar Research Institute, C/ del Dr. Aiguader, 88, 08003, Barcelona, Spain
| | - Marina Mola-Caminal
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Lucía Rey-Álvarez
- Neurovascular Research Group, Department of Neurology, Hospital del Mar Research Institute, C/ del Dr. Aiguader, 88, 08003, Barcelona, Spain
| | - Adrià Macias-Gómez
- Neurovascular Research Group, Department of Neurology, Hospital del Mar Research Institute, C/ del Dr. Aiguader, 88, 08003, Barcelona, Spain
| | - Antoni Suárez-Pérez
- Neurovascular Research Group, Department of Neurology, Hospital del Mar Research Institute, C/ del Dr. Aiguader, 88, 08003, Barcelona, Spain
| | - Eva Giralt-Steinhauer
- Neurovascular Research Group, Department of Neurology, Hospital del Mar Research Institute, C/ del Dr. Aiguader, 88, 08003, Barcelona, Spain
| | - Ana Rodríguez-Campello
- Neurovascular Research Group, Department of Neurology, Hospital del Mar Research Institute, C/ del Dr. Aiguader, 88, 08003, Barcelona, Spain
- Medicine Department, DCEXS-Universitat Pompeu Fabra (UPF), 08002, Barcelona, Spain
| | - Elisa Cuadrado-Godia
- Neurovascular Research Group, Department of Neurology, Hospital del Mar Research Institute, C/ del Dr. Aiguader, 88, 08003, Barcelona, Spain
- Medicine Department, DCEXS-Universitat Pompeu Fabra (UPF), 08002, Barcelona, Spain
| | - Ángel Ois
- Neurovascular Research Group, Department of Neurology, Hospital del Mar Research Institute, C/ del Dr. Aiguader, 88, 08003, Barcelona, Spain
- Medicine Department, DCEXS-Universitat Pompeu Fabra (UPF), 08002, Barcelona, Spain
| | - Manel Esteller
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain
- Centro de Investigacion Biomedica en Red Cancer (CIBERONC), Madrid, Spain
- Institucio Catalana de Recerca I Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Catalonia, Spain
| | - Jaume Roquer
- Neurovascular Research Group, Department of Neurology, Hospital del Mar Research Institute, C/ del Dr. Aiguader, 88, 08003, Barcelona, Spain
| | | | - Jordi Jiménez-Conde
- Neurovascular Research Group, Department of Neurology, Hospital del Mar Research Institute, C/ del Dr. Aiguader, 88, 08003, Barcelona, Spain.
- Medicine Department, DCEXS-Universitat Pompeu Fabra (UPF), 08002, Barcelona, Spain.
| |
Collapse
|
92
|
Malik R, Beaufort N, Li J, Tanaka K, Georgakis MK, He Y, Koido M, Terao C, Japan B, Anderson CD, Kamatani Y, Zand R, Dichgans M. Genetically proxied HTRA1 protease activity and circulating levels independently predict risk of ischemic stroke and coronary artery disease. NATURE CARDIOVASCULAR RESEARCH 2024; 3:701-713. [PMID: 39196222 DOI: 10.1038/s44161-024-00475-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 04/23/2024] [Indexed: 08/29/2024]
Abstract
Genetic variants in HTRA1 are associated with stroke risk. However, the mechanisms mediating this remain largely unknown, as does the full spectrum of phenotypes associated with genetic variation in HTRA1. Here we show that rare HTRA1 variants are linked to ischemic stroke in the UK Biobank and BioBank Japan. Integrating data from biochemical experiments, we next show that variants causing loss of protease function associated with ischemic stroke, coronary artery disease and skeletal traits in the UK Biobank and MyCode cohorts. Moreover, a common variant modulating circulating HTRA1 mRNA and protein levels enhances the risk of ischemic stroke and coronary artery disease while lowering the risk of migraine and macular dystrophy in genome-wide association study, UK Biobank, MyCode and BioBank Japan data. We found no interaction between proxied HTRA1 activity and levels. Our findings demonstrate the role of HTRA1 for cardiovascular diseases and identify two mechanisms as potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Rainer Malik
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Nathalie Beaufort
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Jiang Li
- Department of Molecular and Functional Genomics, Geisinger Health System, Danville, PA, USA
| | - Koki Tanaka
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Marios K Georgakis
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Yunye He
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Masaru Koido
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - BioBank Japan
- Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Christopher D Anderson
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Boston, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Yoichiro Kamatani
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Ramin Zand
- Department of Neurology, Pennsylvania State University, Hershey, PA, USA
- Department of Neurology, Neuroscience Institute, Geisinger Health System, Danville, PA, USA
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University of Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
- German Center for Cardiovascular Research (DZHK), Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
93
|
Debette S, Ihara M. Redefining common and rare HTRA1 variants as risk factors for polyvascular disease. NATURE CARDIOVASCULAR RESEARCH 2024; 3:619-621. [PMID: 39196235 DOI: 10.1038/s44161-024-00492-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Affiliation(s)
- Stéphanie Debette
- Bordeaux Population Health Research Center, University of Bordeaux, INSERM, UMR 1219, Bordeaux, France.
- Department of Neurology, Institute for Neurodegenerative Diseases, Bordeaux University Hospital, Bordeaux, France.
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| |
Collapse
|
94
|
Frei O, Hindley G, Shadrin AA, van der Meer D, Akdeniz BC, Hagen E, Cheng W, O'Connell KS, Bahrami S, Parker N, Smeland OB, Holland D, de Leeuw C, Posthuma D, Andreassen OA, Dale AM. Improved functional mapping of complex trait heritability with GSA-MiXeR implicates biologically specific gene sets. Nat Genet 2024; 56:1310-1318. [PMID: 38831010 PMCID: PMC11759099 DOI: 10.1038/s41588-024-01771-1] [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: 12/02/2022] [Accepted: 04/24/2024] [Indexed: 06/05/2024]
Abstract
While genome-wide association studies are increasingly successful in discovering genomic loci associated with complex human traits and disorders, the biological interpretation of these findings remains challenging. Here we developed the GSA-MiXeR analytical tool for gene set analysis (GSA), which fits a model for the heritability of individual genes, accounting for linkage disequilibrium across variants and allowing the quantification of partitioned heritability and fold enrichment for small gene sets. We validated the method using extensive simulations and sensitivity analyses. When applied to a diverse selection of complex traits and disorders, including schizophrenia, GSA-MiXeR prioritizes gene sets with greater biological specificity compared to standard GSA approaches, implicating voltage-gated calcium channel function and dopaminergic signaling for schizophrenia. Such biologically relevant gene sets, often with fewer than ten genes, are more likely to provide insights into the pathobiology of complex diseases and highlight potential drug targets.
Collapse
Affiliation(s)
- Oleksandr Frei
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Centre for Bioinformatics, Department of Informatics, University of Oslo, Oslo, Norway.
| | - Guy Hindley
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Alexey A Shadrin
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Dennis van der Meer
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Bayram C Akdeniz
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Centre for Bioinformatics, Department of Informatics, University of Oslo, Oslo, Norway
| | - Espen Hagen
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Weiqiu Cheng
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kevin S O'Connell
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Shahram Bahrami
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nadine Parker
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Olav B Smeland
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Dominic Holland
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA, USA
| | - Christiaan de Leeuw
- Department of Complex Trait Genetics, Centre for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands
| | - Danielle Posthuma
- Department of Complex Trait Genetics, Centre for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands
| | - Ole A Andreassen
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anders M Dale
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
95
|
Huang S, Joshi A, Shi Z, Wei J, Tran H, Zheng SL, Duggan D, Ashworth A, Billings L, Helfand BT, Qamar A, Bulwa Z, Tafur A, Xu J. Combined polygenic scores for ischemic stroke risk factors aid risk assessment of ischemic stroke. Int J Cardiol 2024; 404:131990. [PMID: 38521508 DOI: 10.1016/j.ijcard.2024.131990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/01/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND Current risk assessment for ischemic stroke (IS) is limited to clinical variables. We hypothesize that polygenic scores (PGS) of IS (PGSIS) and IS-associated diseases such as atrial fibrillation (AF), venous thromboembolism (VTE), coronary artery disease (CAD), hypertension (HTN), and Type 2 diabetes (T2D) may improve the performance of IS risk assessment. METHODS Incident IS was followed for 479,476 participants in the UK Biobank who did not have an IS diagnosis prior to the recruitment. Lifestyle variables (obesity, smoking and alcohol) at the time of study recruitment, clinical diagnoses of IS-associated diseases, PGSIS, and five PGSs for IS-associated diseases were tested using the Cox proportional-hazards model. Predictive performance was assessed using the C-statistic and net reclassification index (NRI). RESULTS During a median average 12.5-year follow-up, 8374 subjects were diagnosed with IS. Known clinical variables (age, gender, clinical diagnoses of IS-associated diseases, obesity, and smoking) and PGSIS were all independently associated with IS (P < 0.001). In addition, PGSIS and each PGS for IS-associated diseases was also independently associated with IS (P < 0.001). Compared to the clinical model, a joint clinical/PGS model improved the C-statistic for predicting IS from 0.71 to 0.73 (P < 0.001) and significantly reclassified IS risk (NRI = 0.017, P < 0.001), and 6.48% of subjects were upgraded from low to high risk. CONCLUSIONS Adding PGSs of IS and IS-associated diseases to known clinical risk factors statistically improved risk assessment for IS, demonstrating the supplementary value of inherited susceptibility measurement . However, its clinical utility is likely limited due to modest improvements in predictive values.
Collapse
Affiliation(s)
- Sarah Huang
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, IL, USA
| | - Abhishek Joshi
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, IL, USA
| | - Zhuqing Shi
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, IL, USA
| | - Jun Wei
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, IL, USA
| | - Huy Tran
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, IL, USA
| | - S Lilly Zheng
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, IL, USA
| | - David Duggan
- Affiliate of City of Hope, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Annabelle Ashworth
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, IL, USA
| | - Liana Billings
- Department of Medicine, NorthShore University HealthSystem, Evanston, IL, USA; University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - Brian T Helfand
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, IL, USA; University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - Arman Qamar
- Cardiovascular Institute, NorthShore University HealthSystem, Evanston, IL, USA
| | - Zachary Bulwa
- Department of Neurology, NorthShore University HealthSystem, Evanston, IL, USA
| | - Alfonso Tafur
- Cardiovascular Institute, NorthShore University HealthSystem, Evanston, IL, USA
| | - Jianfeng Xu
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, IL, USA; University of Chicago Pritzker School of Medicine, Chicago, IL, USA.
| |
Collapse
|
96
|
Li W, Shi J, Yu Z, Garcia-Gabilondo M, Held A, Huang L, Deng W, Ning M, Ji X, Rosell A, Wainger BJ, Lo EH. SLC22A17 as a Cell Death-Linked Regulator of Tight Junctions in Cerebral Ischemia. Stroke 2024; 55:1650-1659. [PMID: 38738428 DOI: 10.1161/strokeaha.124.046736] [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: 01/31/2024] [Accepted: 04/17/2024] [Indexed: 05/14/2024]
Abstract
BACKGROUND Beyond neuronal injury, cell death pathways may also contribute to vascular injury after stroke. We examined protein networks linked to major cell death pathways and identified SLC22A17 (solute carrier family 22 member 17) as a novel mediator that regulates endothelial tight junctions after ischemia and inflammatory stress. METHODS Protein-protein interactions and brain enrichment analyses were performed using STRING, Cytoscape, and a human tissue-specific expression RNA-seq database. In vivo experiments were performed using mouse models of transient focal cerebral ischemia. Human stroke brain tissues were used to detect SLC22A17 by immunostaining. In vitro experiments were performed using human brain endothelial cultures subjected to inflammatory stress. Immunostaining and Western blot were used to assess responses in SLC22A17 and endothelial tight junctional proteins. Water content, dextran permeability, and electrical resistance assays were used to assess edema and blood-brain barrier (BBB) integrity. Gain and loss-of-function studies were performed using lentiviral overexpression of SLC22A17 or short interfering RNA against SLC22A17, respectively. RESULTS Protein-protein interaction analysis showed that core proteins from apoptosis, necroptosis, ferroptosis, and autophagy cell death pathways were closely linked. Among the 20 proteins identified in the network, the iron-handling solute carrier SLC22A17 emerged as the mediator enriched in the brain. After cerebral ischemia in vivo, endothelial expression of SLC22A17 increases in both human and mouse brains along with BBB leakage. In human brain endothelial cultures, short interfering RNA against SLC22A17 prevents TNF-α (tumor necrosis factor alpha)-induced ferroptosis and downregulation in tight junction proteins and disruption in transcellular permeability. Notably, SLC22A17 could repress the transcription of tight junctional genes. Finally, short interfering RNA against SLC22A17 ameliorates BBB leakage in a mouse model of focal cerebral ischemia. CONCLUSIONS Using a combination of cell culture, human stroke samples, and mouse models, our data suggest that SLC22A17 may play a role in the control of BBB function after cerebral ischemia. These findings may offer a novel mechanism and target for ameliorating BBB injury and edema after stroke.
Collapse
Affiliation(s)
- Wenlu Li
- Departments of Radiology and Neurology, Neuroprotection Research Laboratories (W.L., J.S., Z.Y., L.H., W.D., M.N., E.H.L.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Jingfei Shi
- Departments of Radiology and Neurology, Neuroprotection Research Laboratories (W.L., J.S., Z.Y., L.H., W.D., M.N., E.H.L.), Massachusetts General Hospital, Harvard Medical School, Boston
- Cerebrovascular Research Institute, Xuanwu Hospital, Capital Medical University, Beijing, China (J.S., X.J.)
| | - Zhanyang Yu
- Departments of Radiology and Neurology, Neuroprotection Research Laboratories (W.L., J.S., Z.Y., L.H., W.D., M.N., E.H.L.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Miguel Garcia-Gabilondo
- Neurovascular Research Laboratory, Vall d'Hebron Institut de Recerca, Universitat Autónoma de Barcelona, Spain (M.G.-G., A.R.)
| | - Aaron Held
- Department of Neurology, Sean M. Healey and AMG Center for ALS (A.H., B.J.W.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Lena Huang
- Departments of Radiology and Neurology, Neuroprotection Research Laboratories (W.L., J.S., Z.Y., L.H., W.D., M.N., E.H.L.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Wenjun Deng
- Departments of Radiology and Neurology, Neuroprotection Research Laboratories (W.L., J.S., Z.Y., L.H., W.D., M.N., E.H.L.), Massachusetts General Hospital, Harvard Medical School, Boston
- Department of Neurology, Clinical Proteomics Research Center (W.D., M.N.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Mingming Ning
- Departments of Radiology and Neurology, Neuroprotection Research Laboratories (W.L., J.S., Z.Y., L.H., W.D., M.N., E.H.L.), Massachusetts General Hospital, Harvard Medical School, Boston
- Department of Neurology, Clinical Proteomics Research Center (W.D., M.N.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Xunming Ji
- Cerebrovascular Research Institute, Xuanwu Hospital, Capital Medical University, Beijing, China (J.S., X.J.)
| | - Anna Rosell
- Neurovascular Research Laboratory, Vall d'Hebron Institut de Recerca, Universitat Autónoma de Barcelona, Spain (M.G.-G., A.R.)
| | - Brian J Wainger
- Department of Neurology, Sean M. Healey and AMG Center for ALS (A.H., B.J.W.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Eng H Lo
- Departments of Radiology and Neurology, Neuroprotection Research Laboratories (W.L., J.S., Z.Y., L.H., W.D., M.N., E.H.L.), Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
97
|
Genetic ancestry contributes to gene expression in the brain. Nat Neurosci 2024; 27:1042-1043. [PMID: 38769150 DOI: 10.1038/s41593-024-01645-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
|
98
|
Zhou T, Zhang L, He L, Lan Y, Ding L, Li L, Wang Z. GSK-126 Attenuates Cell Apoptosis in Ischemic Brain Injury by Modulating the EZH2-H3K27me3-Bcl2l1 Axis. Mol Neurobiol 2024; 61:3369-3383. [PMID: 37989985 DOI: 10.1007/s12035-023-03808-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 11/13/2023] [Indexed: 11/23/2023]
Abstract
Whether epigenetic modifications participate in the cell apoptosis after ischemic stroke remains unclear. Histone 3 tri-methylation at lysine 27 (H3K27me3) is a histone modification that leads to gene silencing and is involved in the pathogenesis of ischemic stroke. Since the expression of many antiapoptotic genes is inhibited in the ischemic brains, here we aimed to offer an epigenetic solution to cell apoptosis after stroke by reversing H3K27me3 levels after ischemia. GSK-126, a specific inhibitor of enhancer of zeste homolog 2 (EZH2), significantly decreased H3K27me3 levels and inhibited middle cerebral artery occlusion (MCAO) induced and oxygen glucose deprivation (OGD) induced cell apoptosis. Moreover, GSK-126 attenuated the apoptosis caused by oxidative stress, excitotoxicity, and excessive inflammatory responses in vitro. The role of H3K27me3 in regulating of the expression of the antiapoptotic molecule B cell lymphoma-2 like 1 (Bcl2l1) explained the antiapoptotic effect of GSK-126. In conclusion, we found that GSK-126 could effectively protect brain cells from apoptosis after cerebral ischemia, and this role of GSK-126 is closely related to an axis that regulates Bcl2l1 expression, beginning with the regulation of EZH2-dependent H3K27me3 modification.
Collapse
Affiliation(s)
- Tai Zhou
- Department of Pathophysiology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Lei Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Li He
- Department of Pathophysiology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Yan Lan
- Department of Pathophysiology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Lei Ding
- Department of Pathophysiology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Li Li
- Department of Pathophysiology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China.
- Laboratory of Clinical and Experimental Pathology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China.
| | - Zhongcheng Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China.
- Laboratory of Clinical and Experimental Pathology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
99
|
Zhang X, Hu X, Fang S, Li J, Liu Z, Xie W, Xu R, Dmytriw AA, Yang K, Ma Y, Jiao L, Wang T. Vascular Endothelial Growth Factor and Ischemic Stroke Risk: A Mendelian Randomization Study. Neurol Ther 2024; 13:727-737. [PMID: 38619804 PMCID: PMC11136897 DOI: 10.1007/s40120-024-00601-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/07/2024] [Indexed: 04/16/2024] Open
Abstract
INTRODUCTION Previous studies have reported controversial relationships between circulating vascular endothelial growth factors (VEGF) and ischemic stroke (IS). This study aims to demonstrate the causal effect between VEGF and IS using Mendelian randomization (MR). METHODS Summary statistics data from two large-scale genome-wide association studies (GWAS) for 16,112 patients with measured VEGF levels and 40,585 patients with IS were downloaded from public databases and included in this study. A published calculator was adopted for MR power calculation. The primary outcome was any ischemic stroke, and the secondary outcomes were large-artery stroke, cardioembolic stroke, and small-vessel stroke. We used the inverse variance-weighted (IVW) method for primary analysis, supplemented by MR-Egger regression and the weighted median method. RESULTS Nine SNPs were included to represent serum VEGF levels. The IVW method revealed no strong causal association between VEGF and any ischemic stroke (odds ratio [OR] 1.01, 95% CI 0.99-1.04, p = 0.39), cardioembolic stroke (OR 1.04, 95% CI 0.97-1.12, p = 0.28), large-artery stroke (OR 1.02, 95% CI 0.95-1.09, p = 0.62), and small-vessel stroke (OR 0.98, 95% CI 0.91-1.04, p = 0.46). These findings remained robust in sensitivity analyses. MR-Egger regression suggested no horizontal pleiotropy. CONCLUSIONS This Mendelian randomization study found no relationship between genetically predisposed serum VEGF levels and risks of IS or its subtypes.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
- China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
| | - Xinzhi Hu
- Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Shiyuan Fang
- Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
- Department of Neurology, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Jiayao Li
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
- China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
| | - Zhichao Liu
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - Weidun Xie
- Department of Computer Science, City University of Hong Kong, Kowloon, 999077, Hong Kong SAR
| | - Ran Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
- China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
| | - Adam A Dmytriw
- Neuroendovascular Program, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Kun Yang
- Department of Neurosurgery, Tai'an Central Hospital, 29 Longtan Road, Tai'an, 271000, Shandong, China
| | - Yan Ma
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
- China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China.
- China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China.
- Department of Interventional Neuroradiology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District Beijing, 100053, China.
| | - Tao Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China.
- China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China.
| |
Collapse
|
100
|
Seo Y, Bae H, Lee C. Bayesian colocalization of GWAS and eQTL signals reveals cell type-specific genes and regulatory variants for susceptibility to subtypes of ischemic stroke. Comput Biol Chem 2024; 110:108086. [PMID: 38744227 DOI: 10.1016/j.compbiolchem.2024.108086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/09/2024] [Accepted: 04/23/2024] [Indexed: 05/16/2024]
Abstract
A colocalization analysis of genome-wide association study (GWAS) signals and expression quantitative trait loci (eQTL) was conducted to pinpoint target genes and their regulatory nucleotide variants for subtypes of ischemic stroke. We utilized GWAS data from prominent meta-analysis consortia (MEGASTROKE and GIGASTROKE) and single-cell eQTL data in brain and blood tissues to enhance accuracy and minimize noise inherent in bulk RNA-seq. Employing Bayesian colocalization methods, we identified ten shared loci between GWAS and eQTL signals, targeting five eGenes. Specifically, RAPH1 and ICA1L were discovered for small vessel stroke (SVS), whereas SCYL3, CAV1, and CAV2 were for cardioembolic stroke (CS). However, no findings have been made for large artery stroke. The exploration and subsequent functional analysis of causal variants within the colocalized regions revealed their regulatory roles, particularly as enhancer variants (e.g., rs144505847 and rs72932755 targeting ICA1L; rs629234 targeting SCYL3; rs3807989 targeting CAV1 and CAV2). Notably, our study unveiled that all eQTL for CS were identified in oligodendrocytes, while those for SVS were across excitatory neurons, astrocytes, and oligodendrocyte precursor cells. This underscores the heterogeneous tissue-specific genetic factors by subtypes of ischemic stroke. The study emphasizes the need for intensive research efforts to discover causative genes and variants, unravelling the cell type-specific genetic architecture of ischemic stroke subtypes. This knowledge is crucial for advancing our understanding of the underlying pathophysiology and paving the way for precision neurology applications.
Collapse
Affiliation(s)
- Yunji Seo
- Department of Bioinformatics and Life Science, Soongsil University, 369 Sangdo-ro, Dongjak-gu, Seoul 06978, South Korea
| | - Hojin Bae
- Department of Bioinformatics and Life Science, Soongsil University, 369 Sangdo-ro, Dongjak-gu, Seoul 06978, South Korea
| | - Chaeyoung Lee
- Department of Bioinformatics and Life Science, Soongsil University, 369 Sangdo-ro, Dongjak-gu, Seoul 06978, South Korea.
| |
Collapse
|