551
|
Plotnikov D, Huang Y, Khawaja AP, Foster PJ, Zhu Z, Guggenheim JA, He M. High Blood Pressure and Intraocular Pressure: A Mendelian Randomization Study. Invest Ophthalmol Vis Sci 2022; 63:29. [PMID: 35762941 PMCID: PMC9251815 DOI: 10.1167/iovs.63.6.29] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose To test for causality with regard to the association between blood pressure (BP) and intraocular pressure (IOP) and glaucoma. Methods Single nucleotide polymorphisms (SNPs) associated with BP were identified in a genome-wide association study (GWAS) meta-analysis of 526,001 participants of European ancestry. These SNPs were used to assess the BP versus IOP relationship in a distinct sample (n = 70,832) whose corneal-compensated IOP (IOPcc) was measured. To evaluate the BP versus primary open-angle glaucoma (POAG) relationship, additional Mendelian randomization (MR) analyses were conducted using published GWAS summary statistics. Results Observational analysis revealed a linear relationship between BP traits and IOPcc, with a +0.28 mm Hg increase in IOPcc per 10-mm Hg increase in systolic BP (95% confidence interval [CI], 0.26-0.29); for diastolic blood pressure (DBP) and pulse pressure (PP), these estimates were +0.41 mm Hg and +0.36 mm Hg, respectively. An inverse-variance weighted MR analysis did not support a causal relationship, as the estimated causal effect was +0.01 mm Hg IOPcc per 10-mm Hg increase in systolic blood pressure (SBP); +0.13 mm Hg IOPcc per 10-mm Hg increase in DBP; and +0.02 mm Hg IOPcc per 10-mm Hg increase in PP (all P > 0.05). With regard to the risk of POAG, MR analyse yielded causal effect estimate of odds ratio = 0.98 (95% CI, 0.92-1.04) per 10-mm Hg increase in SBP. Neither DBP nor PP demonstrated evidence of a causal effect on POAG. Conclusions A range of different MR analysis methods provided evidence, in general, that the causal effect of BP on IOP (and POAG) was modest, or even zero. However, interpretation was complicated by SNPs associated with BP potentially having pleiotropic effects on IOP.
Collapse
Affiliation(s)
- Denis Plotnikov
- Central Research Laboratory, Kazan State Medical University, Kazan, Russia
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, United Kingdom
| | - Yu Huang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Anthony P. Khawaja
- NIHR Moorfields Biomedical Research Centre and UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Paul J. Foster
- NIHR Moorfields Biomedical Research Centre and UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Zhuoting Zhu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jeremy A. Guggenheim
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, United Kingdom
| | - Mingguang He
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat Sen University, Guangzhou, China
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, University of Melbourne, Melbourne, Australia
| |
Collapse
|
552
|
A Review of Vascular Traits and Assessment Techniques, and Their Heritability. Artery Res 2022. [DOI: 10.1007/s44200-022-00016-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
AbstractVarious tools are available to assess atherosclerosis, arterial stiffening, and endothelial function. They offer utility in the assessment of hypertensive phenotypes, in cardiovascular risk prediction, and as surrogate endpoints in clinical trials. We explore the relative influence of participant genetics, with reference to large-scale genomic studies, population-based cohorts, and candidate gene studies. We find heritability estimates highest for carotid intima-media thickness (CIMT 35–65%), followed by pulse wave velocity as a measure of arterial stiffness (26–43%), and flow mediated dilatation as a surrogate for endothelial function (14–39%); data were lacking for peripheral artery tonometry. We furthermore examine genes and polymorphisms relevant to each technique. We conclude that CIMT and pulse wave velocity dominate the existing evidence base, with fewer published genomic linkages for measures of endothelial function. We finally make recommendations regarding planning and reporting of data relating to vascular assessment techniques, particularly when genomic data are also available, to facilitate integration of these tools into cardiovascular disease research.
Collapse
|
553
|
Huan T, Nguyen S, Colicino E, Ochoa‐Rosales C, Hill WD, Brody JA, Soerensen M, Zhang Y, Baldassari A, Elhadad MA, Toshiko T, Zheng Y, Domingo‐Relloso A, Lee DH, Ma J, Yao C, Liu C, Hwang S, Joehanes R, Fornage M, Bressler J, van Meurs JB, Debrabant B, Mengel‐From J, Hjelmborg J, Christensen K, Vokonas P, Schwartz J, Gahrib SA, Sotoodehnia N, Sitlani CM, Kunze S, Gieger C, Peters A, Waldenberger M, Deary IJ, Ferrucci L, Qu Y, Greenland P, Lloyd‐Jones DM, Hou L, Bandinelli S, Voortman T, Hermann B, Baccarelli A, Whitsel E, Pankow JS, Levy D. Integrative analysis of clinical and epigenetic biomarkers of mortality. Aging Cell 2022; 21:e13608. [PMID: 35546478 PMCID: PMC9197414 DOI: 10.1111/acel.13608] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 03/03/2022] [Accepted: 03/24/2022] [Indexed: 01/28/2023] Open
Abstract
DNA methylation (DNAm) has been reported to be associated with many diseases and with mortality. We hypothesized that the integration of DNAm with clinical risk factors would improve mortality prediction. We performed an epigenome-wide association study of whole blood DNAm in relation to mortality in 15 cohorts (n = 15,013). During a mean follow-up of 10 years, there were 4314 deaths from all causes including 1235 cardiovascular disease (CVD) deaths and 868 cancer deaths. Ancestry-stratified meta-analysis of all-cause mortality identified 163 CpGs in European ancestry (EA) and 17 in African ancestry (AA) participants at p < 1 × 10-7 , of which 41 (EA) and 16 (AA) were also associated with CVD death, and 15 (EA) and 9 (AA) with cancer death. We built DNAm-based prediction models for all-cause mortality that predicted mortality risk after adjusting for clinical risk factors. The mortality prediction model trained by integrating DNAm with clinical risk factors showed an improvement in prediction of cancer death with 5% increase in the C-index in a replication cohort, compared with the model including clinical risk factors alone. Mendelian randomization identified 15 putatively causal CpGs in relation to longevity, CVD, or cancer risk. For example, cg06885782 (in KCNQ4) was positively associated with risk for prostate cancer (Beta = 1.2, PMR = 4.1 × 10-4 ) and negatively associated with longevity (Beta = -1.9, PMR = 0.02). Pathway analysis revealed that genes associated with mortality-related CpGs are enriched for immune- and cancer-related pathways. We identified replicable DNAm signatures of mortality and demonstrated the potential utility of CpGs as informative biomarkers for prediction of mortality risk.
Collapse
Affiliation(s)
- Tianxiao Huan
- The Framingham Heart StudyFraminghamMassachusettsUSA
- The Population Sciences BranchDivision of Intramural ResearchNational Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
- Department of Ophthalmology and Visual SciencesUniversity of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - Steve Nguyen
- Division of Epidemiology & Community HealthSchool of Public HealthUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Elena Colicino
- Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Carolina Ochoa‐Rosales
- Department of EpidemiologyErasmus University Medical CenterRotterdamthe Netherlands
- Centro de Vida Saludable de la Universidad de ConcepciónConcepciónChile
| | - W. David Hill
- Department of PsychologyLothian Birth CohortsUniversity of EdinburghEdinburghUK
| | - Jennifer A. Brody
- Cardiovascular Health Research UnitDepartment of MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Mette Soerensen
- Department of Public HealthEpidemiology, Biostatistics and BiodemographyUniversity of Southern DenmarkOdense CDenmark
- Department of Clinical Biochemistry and PharmacologyCenter for Individualized Medicine in Arterial DiseasesOdense University HospitalOdense CDenmark
- Department of Clinical GeneticsOdense University HospitalOdense CDenmark
| | - Yan Zhang
- Division of Clinical Epidemiology & Aging ResearchGerman Cancer Rsrch Ctr (DKFZ)HeidelbergGermany
| | - Antoine Baldassari
- Department of EpidemiologyGillings School of Global Public HealthUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - Mohamed Ahmed Elhadad
- Research Unit of Molecular EpidemiologyHelmholtz Zentrum MünchenGerman Research Center for Environmental HealthNeuherbergGermany
- Institute of EpidemiologyHelmholtz Zentrum MünchenGerman Research Center for Environmental HealthNeuherbergGermany
- German Research Center for Cardiovascular Disease (DZHK)Partner Site Munich Heart AllianceMunichGermany
| | - Tanaka Toshiko
- Translational Gerontology BranchNational Institute on AgingBaltimoreMarylandUSA
| | - Yinan Zheng
- Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Arce Domingo‐Relloso
- Department of Chronic Diseases EpidemiologyNational Center for EpidemiologyCarlos III Health InstituteMadridSpain
- Department of Environmental Health SciencesColumbia University Mailman School of Public HealthNew YorkNew YorkUSA
- Department of Statistics and Operations ResearchUniversity of ValenciaValenciaSpain
| | - Dong Heon Lee
- The Framingham Heart StudyFraminghamMassachusettsUSA
- The Population Sciences BranchDivision of Intramural ResearchNational Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Jiantao Ma
- The Framingham Heart StudyFraminghamMassachusettsUSA
- The Population Sciences BranchDivision of Intramural ResearchNational Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
- Nutrition Epidemiology and Data ScienceFriedman School of Nutrition Science and PolicyTufts UniversityBostonMassachusettsUSA
| | - Chen Yao
- The Framingham Heart StudyFraminghamMassachusettsUSA
- The Population Sciences BranchDivision of Intramural ResearchNational Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Chunyu Liu
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Shih‐Jen Hwang
- The Framingham Heart StudyFraminghamMassachusettsUSA
- The Population Sciences BranchDivision of Intramural ResearchNational Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Roby Joehanes
- The Framingham Heart StudyFraminghamMassachusettsUSA
- The Population Sciences BranchDivision of Intramural ResearchNational Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Myriam Fornage
- Human Genetics CenterSchool of Public HealthUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Jan Bressler
- Department of Internal MedicineErasmusRotterdamthe Netherlands
| | | | - Birgit Debrabant
- Department of Public HealthEpidemiology, Biostatistics and BiodemographyUniversity of Southern DenmarkOdense CDenmark
| | - Jonas Mengel‐From
- Department of Public HealthEpidemiology, Biostatistics and BiodemographyUniversity of Southern DenmarkOdense CDenmark
- Department of Clinical GeneticsOdense University HospitalOdense CDenmark
| | - Jacob Hjelmborg
- Department of Public HealthEpidemiology, Biostatistics and BiodemographyUniversity of Southern DenmarkOdense CDenmark
| | - Kaare Christensen
- Department of Public HealthEpidemiology, Biostatistics and BiodemographyUniversity of Southern DenmarkOdense CDenmark
- Department of Clinical GeneticsOdense University HospitalOdense CDenmark
| | - Pantel Vokonas
- Veterans AffairsNormative Aging StudyBostonMassachusettsUSA
- Veterans AffairsBoston Healthcare SystemBostonMassachusettsUSA
- Boston University School of Public HealthBostonMassachusettsUSA
| | - Joel Schwartz
- Departments of Environmental Health and EpidemiologyHarvard TH Chan School of Public HealthBostonMassachusettsUSA
| | - Sina A. Gahrib
- Cardiovascular Health Research UnitDepartment of MedicineUniversity of WashingtonSeattleWashingtonUSA
- Department of PsychologyUniv of EdinburghEdinburghUK
| | - Nona Sotoodehnia
- Cardiovascular Health Research UnitDepartment of MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Colleen M. Sitlani
- Cardiovascular Health Research UnitDepartment of MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Sonja Kunze
- Research Unit of Molecular EpidemiologyHelmholtz Zentrum MünchenGerman Research Center for Environmental HealthNeuherbergGermany
- Institute of EpidemiologyHelmholtz Zentrum MünchenGerman Research Center for Environmental HealthNeuherbergGermany
| | - Christian Gieger
- Research Unit of Molecular EpidemiologyHelmholtz Zentrum MünchenGerman Research Center for Environmental HealthNeuherbergGermany
- Institute of EpidemiologyHelmholtz Zentrum MünchenGerman Research Center for Environmental HealthNeuherbergGermany
- German Research Center for Cardiovascular Disease (DZHK)Partner Site Munich Heart AllianceMunichGermany
| | - Annette Peters
- Institute of EpidemiologyHelmholtz Zentrum MünchenGerman Research Center for Environmental HealthNeuherbergGermany
- German Research Center for Cardiovascular Disease (DZHK)Partner Site Munich Heart AllianceMunichGermany
- German Center for Diabetes Research (DZD)München‐Neuherberg, NeuherbergGermany
- Institute of Medical Information Sciences, Biometry and EpidemiologyLudwig‐Maximilians‐UniversityMunichGermany
| | - Melanie Waldenberger
- Research Unit of Molecular EpidemiologyHelmholtz Zentrum MünchenGerman Research Center for Environmental HealthNeuherbergGermany
- Institute of EpidemiologyHelmholtz Zentrum MünchenGerman Research Center for Environmental HealthNeuherbergGermany
- German Research Center for Cardiovascular Disease (DZHK)Partner Site Munich Heart AllianceMunichGermany
| | - Ian J. Deary
- Division of PulmonaryCritical Care and Sleep MedicineCenter for Lung BiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Luigi Ferrucci
- Translational Gerontology BranchNational Institute on AgingBaltimoreMarylandUSA
| | - Yishu Qu
- Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Philip Greenland
- Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Donald M. Lloyd‐Jones
- Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Lifang Hou
- Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | | | - Trudy Voortman
- Department of EpidemiologyErasmus University Medical CenterRotterdamthe Netherlands
| | - Brenner Hermann
- Division of Clinical Epidemiology & Aging ResearchGerman Cancer Rsrch Ctr (DKFZ)HeidelbergGermany
- Network Aging Research (NAR)University of HeidelbergHeidelbergGermany
| | - Andrea Baccarelli
- Precision Medicine ProgramDepartment of Environmental Health SciencesMailman School of Public HealthColumbia UniversityNew YorkNew YorkUSA
| | - Eric Whitsel
- Department of EpidemiologyGillings School of Global Public HealthUniversity of North CarolinaChapel HillNorth CarolinaUSA
- Department of MedicineSchool of MedicineUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - James S. Pankow
- Division of Epidemiology & Community HealthSchool of Public HealthUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Daniel Levy
- The Framingham Heart StudyFraminghamMassachusettsUSA
- The Population Sciences BranchDivision of Intramural ResearchNational Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
554
|
Liu C, Joehanes R, Ma J, Wang Y, Sun X, Keshawarz A, Sooda M, Huan T, Hwang SJ, Bui H, Tejada B, Munson PJ, Cumhur D, Heard-Costa NL, Pitsillides AN, Peloso GM, Feolo M, Sharopova N, Vasan RS, Levy D. Whole Genome DNA and RNA Sequencing of Whole Blood Elucidates the Genetic Architecture of Gene Expression Underlying a Wide Range of Diseases. RESEARCH SQUARE 2022:rs.3.rs-1598646. [PMID: 35664994 PMCID: PMC9164515 DOI: 10.21203/rs.3.rs-1598646/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
To create a scientific resource of expression quantitative trail loci (eQTL), we conducted a genome-wide association study (GWAS) using genotypes obtained from whole genome sequencing (WGS) of DNA and gene expression levels from RNA sequencing (RNA-seq) of whole blood in 2622 participants in Framingham Heart Study. We identified 6,778,286 cis -eQTL variant-gene transcript (eGene) pairs at p < 5x10 - 8 (2,855,111 unique cis -eQTL variants and 15,982 unique eGenes) and 1,469,754 trans -eQTL variant-eGene pairs at p < 1e-12 (526,056 unique trans -eQTL variants and 7,233 unique eGenes). In addition, 442,379 cis -eQTL variants were associated with expression of 1518 long non-protein coding RNAs (lncRNAs). Gene Ontology (GO) analyses revealed that the top GO terms for cis- eGenes are enriched for immune functions (FDR < 0.05). The cis -eQTL variants are enriched for SNPs reported to be associated with 815 traits in prior GWAS, including cardiovascular disease risk factors. As proof of concept, we used this eQTL resource in conjunction with genetic variants from public GWAS databases in causal inference testing (e.g., COVID-19 severity). After Bonferroni correction, Mendelian randomization analyses identified putative causal associations of 60 eGenes with systolic blood pressure, 13 genes with coronary artery disease, and seven genes with COVID-19 severity. This study created a comprehensive eQTL resource via BioData Catalyst that will be made available to the scientific community. This will advance understanding of the genetic architecture of gene expression underlying a wide range of diseases.
Collapse
|
555
|
Krga I, Corral-Jara KF, Barber-Chamoux N, Dubray C, Morand C, Milenkovic D. Grapefruit Juice Flavanones Modulate the Expression of Genes Regulating Inflammation, Cell Interactions and Vascular Function in Peripheral Blood Mononuclear Cells of Postmenopausal Women. Front Nutr 2022; 9:907595. [PMID: 35694160 PMCID: PMC9178201 DOI: 10.3389/fnut.2022.907595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/26/2022] [Indexed: 12/04/2022] Open
Abstract
Grapefruit is a rich source of flavanones, phytochemicals suggested excreting vasculoprotective effects. We previously showed that flavanones in grapefruit juice (GFJ) reduced postmenopausal women’s pulse-wave velocity (PWV), a measure of arterial stiffness. However, mechanisms of flavanone action in humans are largely unknown. This study aimed to decipher molecular mechanisms of flavanones by multi-omics analysis in PBMCs of volunteers consuming GFJ and flavanone-free control drink for 6 months. Modulated genes and microRNAs (miRNAs) were identified using microarrays. Bioinformatics analyses assessed their functions, interactions and correlations with previously observed changes in PWV. GFJ modified gene and miRNA expressions. Integrated analysis of modulated genes and miRNA-target genes suggests regulation of inflammation, immune response, cell interaction and mobility. Bioinformatics identified putative mediators of the observed nutrigenomic effect (STAT3, NF-κB) and molecular docking demonstrated potential binding of flavanone metabolites to transcription factors and cell-signaling proteins. We also observed 34 significant correlations between changes in gene expression and PWV. Moreover, global gene expression was negatively correlated with gene expression profiles in arterial stiffness and hypertension. This study revealed molecular mechanisms underlying vasculoprotective effects of flavanones, including interactions with transcription factors and gene and miRNA expression changes that inversely correlate with gene expression profiles associated with cardiovascular risk factors.
Collapse
Affiliation(s)
- Irena Krga
- Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | | | | | - Claude Dubray
- Institut National de la Santé et de la Recherche Médicale (INSERM), CIC 501, UMR 766, Clermont-Ferrand, France
| | - Christine Morand
- Université Clermont Auvergne, INRAE, UNH, Clermont-Ferrand, France
| | - Dragan Milenkovic
- Université Clermont Auvergne, INRAE, UNH, Clermont-Ferrand, France
- Department of Nutrition, College of Agricultural and Environmental Sciences, University of California, Davis, Davis, CA, United States
- *Correspondence: Dragan Milenkovic,
| |
Collapse
|
556
|
Huang RDL, Nguyen XMT, Peloso GM, Trinder M, Posner DC, Aragam KG, Ho YL, Lynch JA, Damrauer SM, Chang KM, Tsao PS, Natarajan P, Assimes T, Gaziano JM, Djousse L, Cho K, Wilson PWF, Huffman JE, O’Donnell CJ, on behalf of the Veterans Affairs’ Million Veteran Program. Genome-wide and phenome-wide analysis of ideal cardiovascular health in the VA Million Veteran Program. PLoS One 2022; 17:e0267900. [PMID: 35613103 PMCID: PMC9132265 DOI: 10.1371/journal.pone.0267900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 04/18/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Genetic studies may help identify causal pathways; therefore, we sought to identify genetic determinants of ideal CVH and their association with CVD outcomes in the multi-population Veteran Administration Million Veteran Program. METHODS An ideal health score (IHS) was calculated from 3 clinical factors (blood pressure, total cholesterol, and blood glucose levels) and 3 behavioral factors (smoking status, physical activity, and BMI), ascertained at baseline. Multi-population genome-wide association study (GWAS) was performed on IHS and binary ideal health using linear and logistic regression, respectively. Using the genome-wide significant SNPs from the IHS GWAS, we created a weighted IHS polygenic risk score (PRSIHS) which was used (i) to conduct a phenome-wide association study (PheWAS) of associations between PRSIHS and ICD-9 phenotypes and (ii) to further test for associations with mortality and selected CVD outcomes using logistic and Cox regression and, as an instrumental variable, in Mendelian Randomization. RESULTS The discovery and replication cohorts consisted of 142,404 (119,129 European American (EUR); 16,495 African American (AFR)), and 45,766 (37,646 EUR; 5,366 AFR) participants, respectively. The mean age was 65.8 years (SD = 11.2) and 92.7% were male. Overall, 4.2% exhibited ideal CVH based on the clinical and behavioral factors. In the multi-population meta-analysis, variants at 17 loci were associated with IHS and each had known GWAS associations with multiple components of the IHS. PheWAS analysis in 456,026 participants showed that increased PRSIHS was associated with a lower odds ratio for many CVD outcomes and risk factors. Both IHS and PRSIHS measures of ideal CVH were associated with significantly less CVD outcomes and CVD mortality. CONCLUSION A set of high interest genetic variants contribute to the presence of ideal CVH in a multi-ethnic cohort of US Veterans. Genetically influenced ideal CVH is associated with lower odds of CVD outcomes and mortality.
Collapse
Affiliation(s)
- Rose D. L. Huang
- Center for Population Genomics, Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, Massachusetts, United States of America
| | - Xuan-Mai T. Nguyen
- MAVERIC, VA Boston Healthcare System, Boston, Massachusetts, United States of America
- Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Champaign, Illinois, United States of America
| | - Gina M. Peloso
- Center for Population Genomics, Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, Massachusetts, United States of America
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Mark Trinder
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Daniel C. Posner
- MAVERIC, VA Boston Healthcare System, Boston, Massachusetts, United States of America
| | - Krishna G. Aragam
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Yuk-Lam Ho
- MAVERIC, VA Boston Healthcare System, Boston, Massachusetts, United States of America
| | - Julie A. Lynch
- VA Informatics & Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, Utah, United States of America
- College of Nursing & Health Sciences, University of Massachusetts Boston, Boston, Massachusetts, United States of America
| | - Scott M. Damrauer
- Corporal Michael Crescenz VA Medical Center, Philadelphia, Pennsylvania, United States of America
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kyong-Mi Chang
- Corporal Michael Crescenz VA Medical Center, Philadelphia, Pennsylvania, United States of America
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Philip S. Tsao
- VA Palo Alto Health Care System, Palo Alto, California, United States of America
- Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - Pradeep Natarajan
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Themistocles Assimes
- VA Palo Alto Health Care System, Palo Alto, California, United States of America
- Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - J. Michael Gaziano
- MAVERIC, VA Boston Healthcare System, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Aging, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Luc Djousse
- MAVERIC, VA Boston Healthcare System, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Aging, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Kelly Cho
- MAVERIC, VA Boston Healthcare System, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Aging, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Peter W. F. Wilson
- Atlanta VA Medical Center, Decatur, Georgia, United States of America
- Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Jennifer E. Huffman
- Center for Population Genomics, Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, Massachusetts, United States of America
| | - Christopher J. O’Donnell
- Center for Population Genomics, Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Heart & Vascular Center, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | | |
Collapse
|
557
|
Chen D, Zhang Y, Yidilisi A, Xu Y, Dong Q, Jiang J. Causal Associations Between Circulating Adipokines and Cardiovascular Disease: A Mendelian Randomization Study. J Clin Endocrinol Metab 2022; 107:e2572-e2580. [PMID: 35134201 PMCID: PMC9113792 DOI: 10.1210/clinem/dgac048] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Observational studies have suggested associations between adipokines and cardiovascular disease (CVD), but the roles of certain adipokines remain controversial, and these associations have not yet been ascertained causally. OBJECTIVE To investigate whether circulating adipokines causally affect the risk of CVD using 2-sample Mendelian randomization (MR). METHODS Independent genetic variants strongly associated with adiponectin, resistin, chemerin, and retinol binding protein 4 (RBP4) were selected from public genome-wide association studies. Summary-level statistics for CVD, including coronary artery disease (CAD), myocardial infarction, atrial fibrillation (AF), heart failure (HF), and stroke and its subtypes were collected. The inverse-variance weighted and Wald ratio methods were used for the MR estimates. The MR pleiotropy residual sum and outlier, weighted median, MR-Egger, leave-one-out analysis, MR Steiger, and colocalization analyses were used in the sensitivity analysis. RESULTS Genetically predicted resistin levels were positively associated with AF risk (odds ratio [OR] 1.09; 95% confidence interval [CI], 1.04-1.13; P = 4.1 × 10-5), which was attenuated to null after adjusting for blood pressure. We observed suggestive associations between higher genetically predicted chemerin levels and an increased risk of CAD (OR 1.27; 95% CI, 1.01-1.60; P = 0.040), higher genetically predicted RBP4 levels and an increased risk of HF (OR 1.14; 95% CI, 1.02-1.27; P = 0.024). There was no causal association between genetically predicted adiponectin levels and CVD risk. CONCLUSIONS Our findings reveal the causal association between resistin and AF, probably acting through blood pressure, and suggest potential causal associations between chemerin and CAD, RBP4, and HF.
Collapse
Affiliation(s)
- Delong Chen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuxuan Zhang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Abuduwufuer Yidilisi
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Xu
- Department of Cardiology, Ningbo First Hospital, Ningbo, China
| | - Qichao Dong
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Jiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
558
|
Quintanilha JCF, Etheridge AS, Graynor BJ, Larson NB, Crona DJ, Mitchell BD, Innocenti F. Polygenic Risk Scores for Blood Pressure to Assess the Risk of Severe Bevacizumab-Induced Hypertension in Cancer Patients (Alliance). Clin Pharmacol Ther 2022; 112:364-371. [PMID: 35527502 PMCID: PMC9296545 DOI: 10.1002/cpt.2635] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 05/01/2022] [Indexed: 11/10/2022]
Abstract
Hypertension is a common bevacizumab-induced toxicity. No markers are available to predict patients at risk of developing hypertension. We hypothesized that genetic risk of essential hypertension, as measured by a blood pressure polygenic risk score (PRS), would be associated with risk of severe bevacizumab-induced hypertension. PRSs were calculated for 1,027 bevacizumab-treated patients of European descent with cancer from four clinical trials (Alliance for Clinical Trials in Oncology (Alliance) / Cancer and Leukemia Group B (CALGB) 80303, 40503, 90401, 40502) using summary systolic blood pressure (SBP) and diastolic blood pressure (DBP) genome-wide association results obtained from 757,601 individuals of European descent. The association between PRS and grade 3 bevacizumab-induced hypertension (Common Toxicity Criteria for Adverse Events version 3) in each trial was performed by multivariable logistic regression. Fixed-effect meta-analyses odds ratios (ORs) per standard deviation (SD) of the association of PRS (quantitative) and hypertension across trials were estimated by inverse-variance weighting. PRSs were additionally stratified into quintiles, with the bottom quintile as the referent group. The OR of the association between hypertension and each quintile vs. the referent group was determined by logistic regression. The most significant PRS (quantitative)-hypertension association included up to 67 single-nucleotide variants (SNPs) associated with SBP (P = 0.0077, OR per SD = 1.31, 95% confidence interval (CI), 1.07-1.60), and up to 53 SNPs associated with DBP (P = 0.0209, OR per SD = 1.27, 95% CI, 1.04-1.56). Patients in the top quintile had a higher risk of developing bevacizumab-induced hypertension compared with patients in the bottom quintile using SNPs associated with SBP (P = 4.75 × 10-4 , OR = 3.72, 95% CI, 1.84-8.16) and DBP (P = 0.076, OR = 1.83, 95% CI, 0.95-3.64). Genetic variants associated with essential hypertension, mainly SBP, increase the risk of severe bevacizumab-induced hypertension.
Collapse
Affiliation(s)
- Julia C F Quintanilha
- University of North Carolina Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Amy S Etheridge
- University of North Carolina Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Brady J Graynor
- School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Nicholas B Larson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Daniel J Crona
- University of North Carolina Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | | |
Collapse
|
559
|
Tapela NM, Collister J, Liu X, Clifton L, Stiby A, Murgia F, Hopewell JC, Hunter DJ. Are polygenic risk scores for systolic blood pressure and LDL-cholesterol associated with treatment effectiveness, and clinical outcomes among those on treatment? Eur J Prev Cardiol 2022; 29:925-937. [PMID: 34864974 DOI: 10.1093/eurjpc/zwab192] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 10/25/2021] [Indexed: 12/27/2022]
Abstract
AIMS Many studies have investigated associations between polygenic risk scores (PRS) and the incidence of cardiovascular disease (CVD); few have examined whether risk factor-related PRS predict CVD outcomes among adults treated with risk-modifying therapies. We assessed whether PRS for systolic blood pressure (PRSSBP) and for low-density lipoprotein cholesterol (PRSLDL-C) were associated with achieving SBP and LDL-C-related targets, and with major adverse cardiovascular events (MACE: non-fatal stroke or myocardial infarction, CVD death, and revascularization procedures). METHODS AND RESULTS Using observational data from the UK Biobank (UKB), we calculated PRSSBP and PRSLDL-C and constructed two sub-cohorts of unrelated adults of White British ancestry aged 40-69 years and with no history of CVD, who reported taking medications used in the treatment of hypertension or hypercholesterolaemia. Treatment effectiveness in achieving adequate risk factor control was ascertained using on-treatment blood pressure (BP) or LDL-C levels measured at enrolment (uncontrolled hypertension: BP ≥ 140/90 mmHg; uncontrolled hypercholesterolaemia: LDL-C ≥ 3 mmol/L). We conducted multivariable logistic and Cox regression modelling for incident events, adjusting for socioeconomic characteristics, and CVD risk factors. There were 55 439 participants using BP lowering therapies (51.0% male, mean age 61.0 years, median follow-up 11.5 years) and 33 787 using LDL-C lowering therapies (58.5% male, mean age 61.7 years, median follow-up 11.4 years). PRSSBP was associated with uncontrolled hypertension (odds ratio 1.70; 95% confidence interval: 1.60-1.80) top vs. bottom quintile, equivalent to a 5.4 mmHg difference in SBP, and with MACE [hazard ratio (HR) 1.13; 1.04-1.23]. PRSLDL-C was associated with uncontrolled hypercholesterolaemia (HR 2.78; 2.58-3.00) but was not associated with subsequent MACE. CONCLUSION We extend previous findings in the UKB cohort to examine PRSSBP and PRSLDL-C with treatment effectiveness. Our results indicate that both PRSSBP and PRSLDL-C can help identify individuals who, despite being on treatment, have inadequately controlled SBP and LDL-C, and for SBP are at higher risk for CVD events. This extends the potential role of PRS in clinical practice from identifying patients who may need these interventions to identifying patients who may need more intensive intervention.
Collapse
Affiliation(s)
- Neo M Tapela
- Nuffield Department of Population Health, University of Oxford Big Data Institute, Old Road Campus, Headington, Oxford, OX3 7LF, UK
- Botswana Harvard AIDS Institute Partnership, Princess Marina Hospital, Plot No. 1836, Northring Road, Gaborone, Botswana
- Department of Medicine, Division of Global Health Equity, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Jennifer Collister
- Nuffield Department of Population Health, University of Oxford Big Data Institute, Old Road Campus, Headington, Oxford, OX3 7LF, UK
| | - Xiaonan Liu
- Nuffield Department of Population Health, University of Oxford Big Data Institute, Old Road Campus, Headington, Oxford, OX3 7LF, UK
| | - Lei Clifton
- Nuffield Department of Population Health, University of Oxford Big Data Institute, Old Road Campus, Headington, Oxford, OX3 7LF, UK
| | - Alexander Stiby
- Nuffield Department of Population Health, University of Oxford Big Data Institute, Old Road Campus, Headington, Oxford, OX3 7LF, UK
| | - Federico Murgia
- Nuffield Department of Population Health, University of Oxford Big Data Institute, Old Road Campus, Headington, Oxford, OX3 7LF, UK
| | - Jemma C Hopewell
- Nuffield Department of Population Health, University of Oxford Big Data Institute, Old Road Campus, Headington, Oxford, OX3 7LF, UK
| | - David J Hunter
- Nuffield Department of Population Health, University of Oxford Big Data Institute, Old Road Campus, Headington, Oxford, OX3 7LF, UK
- Department of Epidemiology, Harvard TH Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA
| |
Collapse
|
560
|
Abstract
Behavior genetics is a controversial science. For decades, scholars have sought to understand the role of heredity in human behavior and life-course outcomes. Recently, technological advances and the rapid expansion of genomic databases have facilitated the discovery of genes associated with human phenotypes such as educational attainment and substance use disorders. To maximize the potential of this flourishing science, and to minimize potential harms, careful analysis of what it would mean for genes to be causes of human behavior is needed. In this paper, we advance a framework for identifying instances of genetic causes, interpreting those causal relationships, and applying them to advance causal knowledge more generally in the social sciences. Central to thinking about genes as causes is counterfactual reasoning, the cornerstone of causal thinking in statistics, medicine, and philosophy. We argue that within-family genetic effects represent the product of a counterfactual comparison in the same way as average treatment effects (ATEs) from randomized controlled trials (RCTs). Both ATEs from RCTs and within-family genetic effects are shallow causes: They operate within intricate causal systems (non-unitary), produce heterogeneous effects across individuals (non-uniform), and are not mechanistically informative (non-explanatory). Despite these limitations, shallow causal knowledge can be used to improve understanding of the etiology of human behavior and to explore sources of heterogeneity and fade-out in treatment effects.
Collapse
Affiliation(s)
- James W Madole
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
- VA Puget Sound Health Care System, Seattle, WA, USA
| | - K Paige Harden
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
561
|
Li Y, Song L, Rong W, Zhang Y, Yao X, Fang X, Li Y, Du J. Exome risk score for predicting susceptibility to and severity of isolated thoracic aortic aneurysm. Hum Mol Genet 2022; 31:3672-3682. [PMID: 35512356 DOI: 10.1093/hmg/ddac099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/01/2022] [Accepted: 04/25/2022] [Indexed: 11/14/2022] Open
Abstract
Isolated thoracic aortic aneurysms (TAAs) are asymptomatic before dissection or rupture and heterogeneous in clinical phenotype. It is urgent need but difficult to identify individuals at high risk to enable enhanced screening or preventive therapies. Because TAAs have a genetic component, one possible approach is to stratify individuals based on inherited DNA variations. Here, we constructed an integrated exome risk score (ERS) based on both common and rare variants found in whole exome sequencing through a machine-learning framework in discovery population consisting of 551 cases and 1071 controls. We evaluated the performance of the ERS in an independent population including 151 cases and 779 controls with a raw odds ratio (OR) per 1 standard deviation (SD) = 1.95 and area under the receiver operating characteristic curve (AUC) = 0.680. When adjusted by gender and the first four principal components, OR per SD = 1.68 and AUC reached 0.783. Individuals in the top 20% of ERS distribution had an OR of 3.20 compared with others. Finally, we found that individuals with top 20% ERS developed TAA at a younger age (P = 0.002) and with a larger diameter (P = 0.016) compared with lower ERS, and were more likely to suffer from aortic root aneurysms (P = 0.009). Our analysis provides a global view of the genetic components of isolated TAA. The exome score developed and evaluated here is the first polygenic risk score for TAA and is a promising predictor of disease risk and severity, which will facilitate the implementation of the risk-reduction strategies.
Collapse
Affiliation(s)
- Yang Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.,Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing 100029, China.,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100029, China
| | - Li Song
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | - Wei Rong
- Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.,Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing 100029, China.,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100029, China
| | | | - Xiaoming Yao
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | | | - Yulin Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.,Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing 100029, China.,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100029, China
| | - Jie Du
- Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.,Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing 100029, China.,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100029, China
| |
Collapse
|
562
|
Liu C, Joehanes R, Ma J, Wang Y, Sun X, Keshawarz A, Sooda M, Huan T, Hwang SJ, Bui H, Tejada B, Munson PJ, Cumhur D, Heard-Costa NL, Pitsillides AN, Peloso GM, Feolo M, Sharopova N, Vasan RS, Levy D. Whole Genome DNA and RNA Sequencing of Whole Blood Elucidates the Genetic Architecture of Gene Expression Underlying a Wide Range of Diseases. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.04.13.22273841. [PMID: 35547845 PMCID: PMC9094109 DOI: 10.1101/2022.04.13.22273841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
To create a scientific resource of expression quantitative trail loci (eQTL), we conducted a genome-wide association study (GWAS) using genotypes obtained from whole genome sequencing (WGS) of DNA and gene expression levels from RNA sequencing (RNA-seq) of whole blood in 2622 participants in Framingham Heart Study. We identified 6,778,286 cis -eQTL variant-gene transcript (eGene) pairs at p <5×10 -8 (2,855,111 unique cis -eQTL variants and 15,982 unique eGenes) and 1,469,754 trans -eQTL variant-eGene pairs at p <1e-12 (526,056 unique trans -eQTL variants and 7,233 unique eGenes). In addition, 442,379 cis -eQTL variants were associated with expression of 1518 long non-protein coding RNAs (lncRNAs). Gene Ontology (GO) analyses revealed that the top GO terms for cis- eGenes are enriched for immune functions (FDR <0.05). The cis -eQTL variants are enriched for SNPs reported to be associated with 815 traits in prior GWAS, including cardiovascular disease risk factors. As proof of concept, we used this eQTL resource in conjunction with genetic variants from public GWAS databases in causal inference testing (e.g., COVID-19 severity). After Bonferroni correction, Mendelian randomization analyses identified putative causal associations of 60 eGenes with systolic blood pressure, 13 genes with coronary artery disease, and seven genes with COVID-19 severity. This study created a comprehensive eQTL resource via BioData Catalyst that will be made available to the scientific community. This will advance understanding of the genetic architecture of gene expression underlying a wide range of diseases.
Collapse
Affiliation(s)
- Chunyu Liu
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Roby Joehanes
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jiantao Ma
- Nutrition Epidemiology and Data Science, Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Yuxuan Wang
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Xianbang Sun
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Amena Keshawarz
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Meera Sooda
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tianxiao Huan
- University of Massachusetts Medical School, Worcester, MA, USA
| | - Shih-Jen Hwang
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Helena Bui
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Brandon Tejada
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Peter J Munson
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Nancy L Heard-Costa
- Framingham Heart Study, Framingham, MA, USA
- Departments of Medicine and Epidemiology, Boston University Schools of Medicine and Public Health, Boston, MA, USA
| | | | - Gina M Peloso
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Michael Feolo
- University of Massachusetts Medical School, Worcester, MA, USA
| | | | - Ramachandran S Vasan
- Framingham Heart Study, Framingham, MA, USA
- Departments of Medicine and Epidemiology, Boston University Schools of Medicine and Public Health, Boston, MA, USA
| | - Daniel Levy
- Framingham Heart Study, Framingham, MA, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
563
|
Korte N, Ilkan Z, Pearson CL, Pfeiffer T, Singhal P, Rock JR, Sethi H, Gill D, Attwell D, Tammaro P. The Ca2+-gated channel TMEM16A amplifies capillary pericyte contraction and reduces cerebral blood flow after ischemia. J Clin Invest 2022; 132:e154118. [PMID: 35316222 PMCID: PMC9057602 DOI: 10.1172/jci154118] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 03/16/2022] [Indexed: 11/26/2022] Open
Abstract
Pericyte-mediated capillary constriction decreases cerebral blood flow in stroke after an occluded artery is unblocked. The determinants of pericyte tone are poorly understood. We show that a small rise in cytoplasmic Ca2+ concentration ([Ca2+]i) in pericytes activated chloride efflux through the Ca2+-gated anion channel TMEM16A, thus depolarizing the cell and opening voltage-gated calcium channels. This mechanism strongly amplified the pericyte [Ca2+]i rise and capillary constriction evoked by contractile agonists and ischemia. In a rodent stroke model, TMEM16A inhibition slowed the ischemia-evoked pericyte [Ca2+]i rise, capillary constriction, and pericyte death; reduced neutrophil stalling; and improved cerebrovascular reperfusion. Genetic analysis implicated altered TMEM16A expression in poor patient recovery from ischemic stroke. Thus, pericyte TMEM16A is a crucial regulator of cerebral capillary function and a potential therapeutic target for stroke and possibly other disorders of impaired microvascular flow, such as Alzheimer's disease and vascular dementia.
Collapse
Affiliation(s)
- Nils Korte
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, United Kingdom
| | - Zeki Ilkan
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Claire L. Pearson
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Thomas Pfeiffer
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, United Kingdom
| | - Prabhav Singhal
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, United Kingdom
| | - Jason R. Rock
- Center for Regenerative Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Huma Sethi
- Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, St Mary’s Hospital, Imperial College London, London, United Kingdom
| | - David Attwell
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, United Kingdom
| | - Paolo Tammaro
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
564
|
Xue H, Pan W. Robust inference of bi-directional causal relationships in presence of correlated pleiotropy with GWAS summary data. PLoS Genet 2022; 18:e1010205. [PMID: 35576237 PMCID: PMC9135345 DOI: 10.1371/journal.pgen.1010205] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 05/26/2022] [Accepted: 04/15/2022] [Indexed: 11/25/2022] Open
Abstract
To infer a causal relationship between two traits, several correlation-based causal direction (CD) methods have been proposed with the use of SNPs as instrumental variables (IVs) based on GWAS summary data for the two traits; however, none of the existing CD methods can deal with SNPs with correlated pleiotropy. Alternatively, reciprocal Mendelian randomization (MR) can be applied, which however may perform poorly in the presence of (unknown) invalid IVs, especially for bi-directional causal relationships. In this paper, first, we propose a CD method that performs better than existing CD methods regardless of the presence of correlated pleiotropy. Second, along with a simple but yet effective IV screening rule, we propose applying a closely related and state-of-the-art MR method in reciprocal MR, showing its almost identical performance to that of the new CD method when their model assumptions hold; however, if the modeling assumptions are violated, the new CD method is expected to better control type I errors. Notably bi-directional causal relationships impose some unique challenges beyond those for uni-directional ones, and thus requiring special treatments. For example, we point out for the first time several scenarios where a bi-directional relationship, but not a uni-directional one, can unexpectedly cause the violation of some weak modeling assumptions commonly required by many robust MR methods. We also offer some numerical support and a modeling justification for the application of our new methods (and more generally MR) to binary traits. Finally we applied the proposed methods to 12 risk factors and 4 common diseases, confirming mostly well-known uni-directional causal relationships, while identifying some novel and plausible bi-directional ones such as between body mass index and type 2 diabetes (T2D), and between diastolic blood pressure and stroke.
Collapse
Affiliation(s)
- Haoran Xue
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Wei Pan
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
565
|
Cai J, He L, Wang H, Rong X, Chen M, Shen Q, Li X, Li M, Peng Y. Genetic liability for prescription opioid use and risk of cardiovascular diseases: a multivariable Mendelian randomization study. Addiction 2022; 117:1382-1391. [PMID: 34859517 DOI: 10.1111/add.15767] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 11/08/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Observational studies have yielded conflicting results on the association of prescription opioid use (POU) with the risk of cardiovascular diseases (CVD). Residual confounding and potential reverse causality are inevitable in such conventional observational studies. This study used Mendelian randomization (MR) design to estimate the causal effect of POU on the risk of CVD, including coronary heart disease (CHD), myocardial infarction (MI) and ischemic stroke (IS), as well as their common risk factors. DESIGN We estimated the causal effect of genetic liability for POU on CVD in a two-sample MR framework. Complementary sensitivity analyses were conducted to test the robustness of our results. SETTING Genome-wide association studies (GWAS) that were based on predominantly European ancestry. PARTICIPANTS The sample sizes of the GWAS used in this study ranged from 69 033 to 757 601 participants. MEASUREMENTS Genetic variants predictive of the POU and their corresponding summary-level information in the outcomes were retrieved and extracted from the respective GWAS. FINDINGS Using univariable MR, we found evidence for a causal effect of genetic liability for POU on an increased risk of CHD [odds ratio (OR) = 1.09; 95% confidence interval (CI) = 1.02-1.16; P = 0.008] and MI (OR = 1.13; 95% CI = 1.04-1.22; P = 0.002). In multivariable MR, the association remained after accounting for comorbid pain conditions, but was attenuated with adjustment for potential mediators, including body mass index (BMI), waist circumference (WC) and type 2 diabetes (T2D). CONCLUSION Mendelian randomization estimates provide robust evidence for the causal effects of genetic liability for prescription opioid use on an increased risk of coronary heart disease and myocardial infarction, which might be mediated by obesity-related traits.
Collapse
Affiliation(s)
- Jiahao Cai
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lei He
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongxuan Wang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoming Rong
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ming Chen
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qingyu Shen
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiangpen Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mei Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ying Peng
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
566
|
Lin Z, Pan I, Pan W. A practical problem with Egger regression in Mendelian randomization. PLoS Genet 2022; 18:e1010166. [PMID: 35507585 PMCID: PMC9109933 DOI: 10.1371/journal.pgen.1010166] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 05/16/2022] [Accepted: 03/25/2022] [Indexed: 11/19/2022] Open
Abstract
Mendelian randomization (MR) is an instrumental variable (IV) method using genetic variants such as single nucleotide polymorphisms (SNPs) as IVs to disentangle the causal relationship between an exposure and an outcome. Since any causal conclusion critically depends on the three valid IV assumptions, which will likely be violated in practice, MR methods robust to the IV assumptions are greatly needed. As such a method, Egger regression stands out as one of the most widely used due to its easy use and perceived robustness. Although Egger regression is claimed to be robust to directional pleiotropy under the instrument strength independent of direct effect (InSIDE) assumption, it is known to be dependent on the orientations/coding schemes of SNPs (i.e. which allele of an SNP is selected as the reference group). The current practice, as recommended as the default setting in some popular MR software packages, is to orientate the SNPs to be all positively associated with the exposure, which however, to our knowledge, has not been fully studied to assess its robustness and potential impact. We use both numerical examples (with both real data and simulated data) and analytical results to demonstrate the practical problem of Egger regression with respect to its heavy dependence on the SNP orientations. Under the assumption that InSIDE holds for some specific (and unknown) coding scheme of the SNPs, we analytically show that other coding schemes would in general lead to the violation of InSIDE. Other related MR and IV regression methods may suffer from the same problem. Cautions should be taken when applying Egger regression (and related MR and IV regression methods) in practice.
Collapse
Affiliation(s)
- Zhaotong Lin
- Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Isaac Pan
- Pomona College, Claremont, California, United States of America
| | - Wei Pan
- Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
567
|
Jung J, Kim H. Shared genetic etiology and antagonistic relationship of plasma renin activity and systolic blood pressure in a Korean cohorts. Genomics 2022; 114:110334. [PMID: 35278618 DOI: 10.1016/j.ygeno.2022.110334] [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: 11/08/2021] [Revised: 02/11/2022] [Accepted: 03/06/2022] [Indexed: 01/14/2023]
Abstract
Despite extensive studies on blood pressure, its genetic risk factors remain uncertain. Even one of the most researched blood pressure-related traits - renin - is not fully understood genetically. Here, we determine the genetic relationship and associated predisposition between blood pressure and baseline renin. In 8840 Korean individuals, we observed a strong negative genome-wide genetic correlation (rg = -0.484) between systolic blood pressure (SBP) and plasma renin activity (PRA), suggesting that antagonistic genetic signals explain the variance in the two traits. We found 51 significant pleiotropic SNPs affecting the two traits, which could contribute to the Renin-Angiotensin-Aldosterone System (RAAS). Our findings provide insight into studies on RAAS by identifying the genome-wide relationship and susceptibility loci of SBP and PRA.
Collapse
Affiliation(s)
- Jaehoon Jung
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 151-742, Republic of Korea; eGnome, 26 Beobwon-ro, Songpa-gu, Seoul 05836, Republic of Korea.
| | - Heebal Kim
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 151-742, Republic of Korea; eGnome, 26 Beobwon-ro, Songpa-gu, Seoul 05836, Republic of Korea; Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 151-742, Republic of Korea.
| |
Collapse
|
568
|
Yang F, Lu Y, Chen S, Wang K, Hu T, Cui H. Sex-specific effect of serum urate levels on coronary heart disease and myocardial infarction prevention: A Mendelian randomization study. Nutr Metab Cardiovasc Dis 2022; 32:1266-1274. [PMID: 35197211 DOI: 10.1016/j.numecd.2022.01.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND AIMS Observational studies have examined serum urate levels in relation to coronary heart disease (CHD) and myocardial infarction (MI). Whether these associations are causal remains controversial, due to confounding factors and reverse causality. We aim to investigate the causality of these associations using Mendelian randomization method. METHODS AND RESULTS Instrumental variables were obtained from the largest genome-wide association studies of serum urate (457,690 individuals) to date. Summary statistics were from CARDIoGRAMplusC4D consortium (60,801 CHD cases; 43,676 MI cases), FinnGen (21,012 CHD cases; 12,801 MI cases), UK Biobank (10,157 CHD cases; 7018 MI cases), and Biobank Japan (29,319 CHD cases). Inverse-variance weighted method was applied as the main results. Other statistical methods and reverse MR analysis were conducted in the supplementary analyses. Elevated genetically determined serum urate levels were associated with increased risks of CHD and MI. The association pattern remained for the datasets in FinnGen, the combined results of three independent data sources (CHD: odds ratio (OR), 1.10; 95%CI, 1.06-1.15; p = 4.2 × 10-6; MI: OR, 1.12; 95%CI, 1.07-1.18; p = 2.7 × 10-6), and East Asian population. Interestingly, sex-specific subgroup analyses revealed that these associations kept in men only, but not among women in individuals of European ancestry. No consistent evidence was found for the causal effect of CHD or MI on serum urate levels. CONCLUSION We provide consistent evidence for the causal effect of genetically predicted serum urate levels on CHD and MI, but not the reverse effect. Urate-lowering therapy may be of cardiovascular benefit in the prevention of CHD and MI, especially for men.
Collapse
Affiliation(s)
- Fangkun Yang
- Department of Cardiology, Ningbo Hospital of Zhejiang University (Ningbo First Hospital), School of Medicine, Zhejiang University, Ningbo, Zhejiang, China
| | - Yunlong Lu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Songzan Chen
- School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Kai Wang
- Eye Center of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Teng Hu
- School of Medicine, Ningbo University, Ningbo, China
| | - Hanbin Cui
- Cardiology Center, Ningbo First Hospital, Ningbo University, Ningbo, Zhejiang, China.
| |
Collapse
|
569
|
Rukh G, Ahmad S, Lind L, Schiöth HB. Evidence of a Causal Link Between the Well-Being Spectrum and the Risk of Myocardial Infarction: A Mendelian Randomization Study. Front Genet 2022; 13:842223. [PMID: 35571065 PMCID: PMC9096350 DOI: 10.3389/fgene.2022.842223] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/30/2022] [Indexed: 12/02/2022] Open
Abstract
Epidemiological studies have provided extensive evidence regarding the role of psychological risk factors in the pathogenesis of cardiovascular disease (CVD), but whether these associations are causal in nature is still unknown. We aimed to investigate whether the association between the wellbeing spectrum (WBS; derived from four psychological traits including life satisfaction, positive affect, neuroticism, and depressive symptoms) and CVD risk is causal. By employing a two-sample Mendelian randomization (MR) approach, the effect of the WBS on four CVD outcomes, including atrial fibrillation, heart failure, myocardial infarction, and ischemic stroke, was investigated. The genetically predicted WBS was associated with 38% lower risk for heart failure (odds ratio (OR): 0.62; 95% confidence interval [CI]: 0.50–0.78; P: 2.2 × 10−5) and 40% reduced risk of myocardial infarction (OR: 0.60; 95% CI: 0.47–0.78; P: 1.1 × 10−4). Of the WBS constituent traits, only depressive symptoms showed a positive causal association with heart failure and myocardial infarction. Neither WBS nor WBS constituent traits were associated with atrial fibrillation and ischemic stroke. In multivariable MR analyses, when genetic instruments for traditional CVD risk factors were also taken into consideration, the WBS was causally associated with a reduced risk for heart failure (OR: 0.72; 95% CI: 0.58–0.88; P: 0.001) and myocardial infarction (OR: 0.67; 95% CI: 0.52–0.86; P: 0.002). This study provides evidence that a higher WBS is causally associated with a decreased risk of developing CVD and, more specifically, myocardial infarction; moreover, the association is mainly driven by depressive symptoms. These results support current guidelines that suggest improving psychological wellbeing may help in reducing the burden of cardiovascular disease.
Collapse
Affiliation(s)
- Gull Rukh
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- *Correspondence: Gull Rukh,
| | - Shafqat Ahmad
- Molecular Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Preventive Medicine Division, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, United States
| | - Lars Lind
- Cardiovascular Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Helgi Birgir Schiöth
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
570
|
Drapkina OM, Kontsevaya AV, Kalinina AM, Avdeev SM, Agaltsov MV, Alexandrova LM, Antsiferova AA, Aronov DM, Akhmedzhanov NM, Balanova YA, Balakhonova TV, Berns SA, Bochkarev MV, Bochkareva EV, Bubnova MV, Budnevsky AV, Gambaryan MG, Gorbunov VM, Gorny BE, Gorshkov AY, Gumanova NG, Dadaeva VA, Drozdova LY, Egorov VA, Eliashevich SO, Ershova AI, Ivanova ES, Imaeva AE, Ipatov PV, Kaprin AD, Karamnova NS, Kobalava ZD, Konradi AO, Kopylova OV, Korostovtseva LS, Kotova MB, Kulikova MS, Lavrenova EA, Lischenko OV, Lopatina MV, Lukina YV, Lukyanov MM, Mayev IV, Mamedov MN, Markelova SV, Martsevich SY, Metelskaya VA, Meshkov AN, Milushkina OY, Mukaneeva DK, Myrzamatova AO, Nebieridze DV, Orlov DO, Poddubskaya EA, Popovich MV, Popovkina OE, Potievskaya VI, Prozorova GG, Rakovskaya YS, Rotar OP, Rybakov IA, Sviryaev YV, Skripnikova IA, Skoblina NA, Smirnova MI, Starinsky VV, Tolpygina SN, Usova EV, Khailova ZV, Shalnova SA, Shepel RN, Shishkova VN, Yavelov IS. 2022 Prevention of chronic non-communicable diseases in Of the Russian Federation. National guidelines. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2022; 21:3235. [DOI: 10.15829/1728-8800-2022-3235] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
571
|
Henry A, Gordillo-Marañón M, Finan C, Schmidt AF, Ferreira JP, Karra R, Sundström J, Lind L, Ärnlöv J, Zannad F, Mälarstig A, Hingorani AD, Lumbers RT, HERMES and SCALLOP Consortia. Therapeutic Targets for Heart Failure Identified Using Proteomics and Mendelian Randomization. Circulation 2022; 145:1205-1217. [PMID: 35300523 PMCID: PMC9010023 DOI: 10.1161/circulationaha.121.056663] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 02/09/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Heart failure (HF) is a highly prevalent disorder for which disease mechanisms are incompletely understood. The discovery of disease-associated proteins with causal genetic evidence provides an opportunity to identify new therapeutic targets. METHODS We investigated the observational and causal associations of 90 cardiovascular proteins, which were measured using affinity-based proteomic assays. First, we estimated the associations of 90 cardiovascular proteins with incident heart failure by means of a fixed-effect meta-analysis of 4 population-based studies, composed of a total of 3019 participants with 732 HF events. The causal effects of HF-associated proteins were then investigated by Mendelian randomization, using cis-protein quantitative loci genetic instruments identified from genomewide association studies in more than 30 000 individuals. To improve the precision of causal estimates, we implemented an Mendelian randomization model that accounted for linkage disequilibrium between instruments and tested the robustness of causal estimates through a multiverse sensitivity analysis that included up to 120 combinations of instrument selection parameters and Mendelian randomization models per protein. The druggability of candidate proteins was surveyed, and mechanism of action and potential on-target side effects were explored with cross-trait Mendelian randomization analysis. RESULTS Forty-four of ninety proteins were positively associated with risk of incident HF (P<6.0×10-4). Among these, 8 proteins had evidence of a causal association with HF that was robust to multiverse sensitivity analysis: higher CSF-1 (macrophage colony-stimulating factor 1), Gal-3 (galectin-3) and KIM-1 (kidney injury molecule 1) were positively associated with risk of HF, whereas higher ADM (adrenomedullin), CHI3L1 (chitinase-3-like protein 1), CTSL1 (cathepsin L1), FGF-23 (fibroblast growth factor 23), and MMP-12 (matrix metalloproteinase-12) were protective. Therapeutics targeting ADM and Gal-3 are currently under evaluation in clinical trials, and all the remaining proteins were considered druggable, except KIM-1. CONCLUSIONS We identified 44 circulating proteins that were associated with incident HF, of which 8 showed evidence of a causal relationship and 7 were druggable, including adrenomedullin, which represents a particularly promising drug target. Our approach demonstrates a tractable roadmap for the triangulation of population genomic and proteomic data for the prioritization of therapeutic targets for complex human diseases.
Collapse
Affiliation(s)
- Albert Henry
- Institute of Cardiovascular Science (A.H., M.G.-M., C.F., A.F.S., A.D.H.), University College London, United Kingdom
- British Heart Foundation Research Accelerator (A.H., M.G.-M., C.F., A.F.S., A.D.H., R.T.L.), University College London, United Kingdom
- Institute of Health Informatics (A.H., R.T.L.), University College London, United Kingdom
| | - María Gordillo-Marañón
- Institute of Cardiovascular Science (A.H., M.G.-M., C.F., A.F.S., A.D.H.), University College London, United Kingdom
- British Heart Foundation Research Accelerator (A.H., M.G.-M., C.F., A.F.S., A.D.H., R.T.L.), University College London, United Kingdom
| | - Chris Finan
- Institute of Cardiovascular Science (A.H., M.G.-M., C.F., A.F.S., A.D.H.), University College London, United Kingdom
- British Heart Foundation Research Accelerator (A.H., M.G.-M., C.F., A.F.S., A.D.H., R.T.L.), University College London, United Kingdom
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, The Netherlands (C.F., A.F.S.)
| | - Amand F. Schmidt
- Institute of Cardiovascular Science (A.H., M.G.-M., C.F., A.F.S., A.D.H.), University College London, United Kingdom
- British Heart Foundation Research Accelerator (A.H., M.G.-M., C.F., A.F.S., A.D.H., R.T.L.), University College London, United Kingdom
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, The Netherlands (C.F., A.F.S.)
| | - João Pedro Ferreira
- Unidade de Investigação e Desenvolvimento Cardiovascular, Rede de Investigação em Saúde, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Portugal (J.P.F.)
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques - Plurithématique 14-33, and Inserm U1116, Centre Hospitalier Régional Universitaire, French Clinical Research Infrastructure Network, Investigation Network Initiative - Cardiovascular and Renal Clinical Trialists, Nancy, France (J.P.F., F.Z.)
| | - Ravi Karra
- Division of Cardiology, Department of Medicine (R.K.), Duke University Medical Center, Durham, NC
- Department of Pathology (R.K.), Duke University Medical Center, Durham, NC
| | - Johan Sundström
- Department of Medical Sciences, Uppsala University, Sweden (J.S., L.L.)
- The George Institute for Global Health, University of New South Wales, Sydney, Australia (J.S.)
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Sweden (J.S., L.L.)
| | - Johan Ärnlöv
- School of Health and Social Studies, Dalarna University, Falun, Sweden (J.Ä.)
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Huddinge, Sweden (J.Ä.)
| | - Faiez Zannad
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques - Plurithématique 14-33, and Inserm U1116, Centre Hospitalier Régional Universitaire, French Clinical Research Infrastructure Network, Investigation Network Initiative - Cardiovascular and Renal Clinical Trialists, Nancy, France (J.P.F., F.Z.)
| | - Anders Mälarstig
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Solna‚ Sweden (A.M.)
- Emerging Science and Innovation, Pfizer Worldwide Research, Development and Medical, Cambridge, MA (A.M.)
| | - Aroon D. Hingorani
- Institute of Cardiovascular Science (A.H., M.G.-M., C.F., A.F.S., A.D.H.), University College London, United Kingdom
- British Heart Foundation Research Accelerator (A.H., M.G.-M., C.F., A.F.S., A.D.H., R.T.L.), University College London, United Kingdom
| | - R. Thomas Lumbers
- British Heart Foundation Research Accelerator (A.H., M.G.-M., C.F., A.F.S., A.D.H., R.T.L.), University College London, United Kingdom
- Institute of Health Informatics (A.H., R.T.L.), University College London, United Kingdom
- Health Data Research UK London (R.T.L.), University College London, United Kingdom
| | - HERMES and SCALLOP Consortia
- Institute of Cardiovascular Science (A.H., M.G.-M., C.F., A.F.S., A.D.H.), University College London, United Kingdom
- British Heart Foundation Research Accelerator (A.H., M.G.-M., C.F., A.F.S., A.D.H., R.T.L.), University College London, United Kingdom
- Institute of Health Informatics (A.H., R.T.L.), University College London, United Kingdom
- Health Data Research UK London (R.T.L.), University College London, United Kingdom
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, The Netherlands (C.F., A.F.S.)
- Unidade de Investigação e Desenvolvimento Cardiovascular, Rede de Investigação em Saúde, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Portugal (J.P.F.)
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques - Plurithématique 14-33, and Inserm U1116, Centre Hospitalier Régional Universitaire, French Clinical Research Infrastructure Network, Investigation Network Initiative - Cardiovascular and Renal Clinical Trialists, Nancy, France (J.P.F., F.Z.)
- Division of Cardiology, Department of Medicine (R.K.), Duke University Medical Center, Durham, NC
- Department of Pathology (R.K.), Duke University Medical Center, Durham, NC
- Department of Medical Sciences, Uppsala University, Sweden (J.S., L.L.)
- The George Institute for Global Health, University of New South Wales, Sydney, Australia (J.S.)
- School of Health and Social Studies, Dalarna University, Falun, Sweden (J.Ä.)
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Huddinge, Sweden (J.Ä.)
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Solna‚ Sweden (A.M.)
- Emerging Science and Innovation, Pfizer Worldwide Research, Development and Medical, Cambridge, MA (A.M.)
| |
Collapse
|
572
|
Li Q, Yan S, Li Y, Kang H, Zhu H, Lv C. Mendelian Randomization Study of Heart Failure and Stroke Subtypes. Front Cardiovasc Med 2022; 9:844733. [PMID: 35463787 PMCID: PMC9021833 DOI: 10.3389/fcvm.2022.844733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 01/28/2022] [Indexed: 12/11/2022] Open
Abstract
Background Whether heart failure (HF) is an independent risk factor of ischemic stroke (IS) and hemorrhagic stroke remains controversial. We employed a multivariable Mendelian randomization (MR) to further investigate the causal effects of HF on the risk of stroke and stroke subtypes. Methods Genetically predicted HF was selected as an instrumental variable (IV) from published genome-wide association studies (GWAS) meta-analyses. Stroke data with different etiologies were extracted as outcome variables from another two GWAS meta-analyses. The random-effects inverse variance-weighted (IVW) model was applied as the main method, along with sensitivity analysis. Atrial fibrillation (AF), coronary heart disease (CHD), and systolic blood pressure (SBP) were controlled for mediating effects in multivariable MR. Results Genetically predicted HF was significantly associated with any IS [odds ratio (OR), 1.39; 95% CI, 1.12–1.74; p = 0.03], large artery stroke (LAS; OR, 1.84; 95% CI, 1.27–2.65; p = 0.001), and cardioembolic stroke (CES; OR, 1.73; 95% CI, 1.21–2.47; p = 0.003), but without small vessel stroke (SVS; OR, 1.1; 95% CI, 0.80–1.52; p = 0.56) and intracerebral hemorrhage (ICH; OR, 0.86; 95% CI, 0.41–1.83; p = 0.699) in univariable MR. However, these significant associations were attenuated to the null after adjusting for confounding factor in multivariable MR. Conclusion There was no direct causal association between HF and stroke in our study. The association between HF and IS can be driven by AF, CHD, and SBP.
Collapse
Affiliation(s)
- Quan Li
- Emergency Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Shijiao Yan
- School of Public Health, Hainan Medical University, Haikou, China
- Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, China
| | - Yan Li
- Emergency Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hai Kang
- Department of Emergency, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Huadong Zhu
- Emergency Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Huadong Zhu
| | - Chuanzhu Lv
- Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
- *Correspondence: Chuanzhu Lv
| |
Collapse
|
573
|
Rogne T, Liew Z, Hernáez Á, Brumpton BM, Magnus MC. Modifiable risk factors for ectopic pregnancy: a Mendelian randomization study. Am J Obstet Gynecol 2022; 227:339-341.e4. [PMID: 35390318 DOI: 10.1016/j.ajog.2022.03.063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/01/2022] [Accepted: 03/30/2022] [Indexed: 11/01/2022]
|
574
|
Parcha V, Pampana A, Bress AP, Irvin MR, Arora G, Arora P. Association of Polygenic Risk Score With Blood Pressure and Adverse Cardiovascular Outcomes in Individuals With Type II Diabetes: Insights From the ACCORD Trial. Hypertension 2022; 79:e100-e102. [PMID: 35369713 PMCID: PMC9010352 DOI: 10.1161/hypertensionaha.122.18976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Vibhu Parcha
- Division of Cardiovascular Disease (V.P., A.P., G.A., P.A.), University of Alabama at Birmingham
| | - Akhil Pampana
- Division of Cardiovascular Disease (V.P., A.P., G.A., P.A.), University of Alabama at Birmingham
| | - Adam P Bress
- Department of Population Health Sciences, University of Utah School of Medicine, Salt Lake City (A.B.)
| | - Marguerite R Irvin
- Department of Epidemiology, School of Public Health (M.R.I.), University of Alabama at Birmingham
| | - Garima Arora
- Division of Cardiovascular Disease (V.P., A.P., G.A., P.A.), University of Alabama at Birmingham
| | - Pankaj Arora
- Division of Cardiovascular Disease (V.P., A.P., G.A., P.A.), University of Alabama at Birmingham.,Section of Cardiology, Birmingham Veterans Affairs Medical Center, AL (P.A.)
| |
Collapse
|
575
|
Frerich S, Malik R, Georgakis MK, Sinner MF, Kittner SJ, Mitchell BD, Dichgans M. Cardiac Risk Factors for Stroke: A Comprehensive Mendelian Randomization Study. Stroke 2022; 53:e130-e135. [PMID: 34911345 PMCID: PMC10510836 DOI: 10.1161/strokeaha.121.036306] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/05/2021] [Accepted: 11/15/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Observational studies suggest an association of stroke with cardiac traits beyond atrial fibrillation, the leading source of cardioembolism. However, controversy remains regarding a causal role of these traits in stroke pathogenesis. Here, we leveraged genetic data to systematically assess associations between cardiac traits and stroke risk using a Mendelian Randomization framework. METHODS We studied 66 cardiac traits including cardiovascular diseases, magnetic resonance imaging-derived cardiac imaging, echocardiographic imaging, and electrocardiographic measures, as well as blood biomarkers in a 2-sample Mendelian Randomization approach. Genetic predisposition to each trait was explored for associations with risk of stroke and stroke subtypes in data from the MEGASTROKE consortium (40 585 cases/406 111 controls). Using multivariable Mendelian Randomization, we adjusted for potential pleiotropic or mediating effects relating to atrial fibrillation, coronary artery disease, and systolic blood pressure. RESULTS As expected, we observed strong independent associations between genetic predisposition to atrial fibrillation and cardioembolic stroke and between genetic predisposition to coronary artery disease as a proxy for atherosclerosis and large-artery stroke. Our data-driven analyses further indicated associations of genetic predisposition to both heart failure and lower resting heart rate with stroke. However, these associations were explained by atrial fibrillation, coronary artery disease, and systolic blood pressure in multivariable analyses. Genetically predicted P-wave terminal force in V1, an electrocardiographic marker for atrial cardiopathy, was inversely associated with large-artery stroke. CONCLUSIONS Available genetic data do not support substantial effects of cardiac traits on the risk of stroke beyond known clinical risk factors. Our findings highlight the need to carefully control for confounding and other potential biases in studies examining candidate cardiac risk factors for stroke.
Collapse
Affiliation(s)
- Simon Frerich
- Institute for Stroke and Dementia Research (S.F., R.M., M.K.G., M.D.), University Hospital, LMU Munich, Germany
| | - Rainer Malik
- Institute for Stroke and Dementia Research (S.F., R.M., M.K.G., M.D.), University Hospital, LMU Munich, Germany
| | - Marios K. Georgakis
- Institute for Stroke and Dementia Research (S.F., R.M., M.K.G., M.D.), University Hospital, LMU Munich, Germany
| | - Moritz F. Sinner
- Department of Cardiology (M.F.S.), University Hospital, LMU Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site: Munich Heart Alliance, Germany (M.F.S.)
| | | | - Braxton D. Mitchell
- Department of Medicine (B.D.M.), University of Maryland School of Medicine and Baltimore VAMC
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (S.F., R.M., M.K.G., M.D.), University Hospital, LMU Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Germany (M.D.)
- German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany (M.D.)
| |
Collapse
|
576
|
Ward LD, Parker MM, Deaton AM, Tu HC, Flynn-Carroll AO, Hinkle G, Nioi P. Rare coding variants in DNA damage repair genes associated with timing of natural menopause. HGG ADVANCES 2022; 3:100079. [PMID: 35493704 PMCID: PMC9039695 DOI: 10.1016/j.xhgg.2021.100079] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 12/08/2021] [Indexed: 11/30/2022] Open
Abstract
The age of menopause is associated with fertility and disease risk, and its genetic control is of great interest. We use whole-exome sequences from 132,370 women in the UK Biobank to test for associations between rare damaging variants and age at natural menopause. Rare damaging variants in five genes are significantly associated with menopause: CHEK2 (p = 3.3 × 10−51), DCLRE1A (p = 8.4 × 10−13), and HELB (p = 5.7 × 10−7) with later menopause and TOP3A (p = 7.6 × 10−8) and CLPB (p = 8.1 × 10−7) with earlier menopause. Two additional genes are suggestive: RAD54L (p = 2.4 × 10−6) with later menopause and HROB (p = 2.9 × 10−6) with earlier menopause. In a follow-up analysis of repeated questionnaires in women who were initially premenopausal, CHEK2, TOP3A, and RAD54L genotypes are associated with subsequent menopause. Consistent with previous genome-wide association studies (GWASs), six of the seven genes are involved in the DNA damage repair pathway. Phenome-wide scans across 398,569 men and women revealed that in addition to known associations with cancers and blood cell counts, rare variants in CHEK2 are also associated with increased risk for uterine fibroids, polycystic ovary syndrome, and prostate hypertrophy; these associations are not shared with higher-penetrance breast cancer genes. Causal mediation analysis suggests that approximately 8% of the breast cancer risk conferred by CHEK2 pathogenic variants after menopause is mediated through delayed menopause.
Collapse
|
577
|
Xu X, Ye D, Liu B, Yang Y, Chen Y, Qian Y, Mao Y, Sun X. Assessing the impact of blood pressure in the development of inflammatory bowel disease. J Clin Hypertens (Greenwich) 2022; 24:566-572. [PMID: 35363426 PMCID: PMC9106082 DOI: 10.1111/jch.14477] [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: 01/20/2022] [Revised: 02/25/2022] [Accepted: 03/08/2022] [Indexed: 11/30/2022]
Abstract
The purpose of this study is to investigate the potential causal relationships between blood pressure and inflammatory bowel disease (IBD) by using the bidirectional Mendelian randomization (MR) approach. Summary‐level data for blood pressure was extracted from the hitherto largest genome‐wide study (GWAS) with 759 601 participants of European‐descent. We used 56 single nucleotide polymorphisms (SNPs) as instrumental variables (IVs) for blood pressure. Summary statistics for IBD were derived from a GWAS with an overall 59 957 participants of European ancestry, of which 109 IVs were selected. Several robust analytical methods, including inverse‐variance weighted (IVW) method, weighted‐median method, MR‐Egger regression, MR‐PRESSO test, maximum likelihood method, “leave‐one‐out” and multivariable MR analysis were used to evaluate the causal associations between blood pressure and IBD. Genetically predicted higher systolic blood pressure (SBP) was associated with an increased risk of IBD (odds ratio (OR) = 1.05, 95% confidence interval (CI):1.02–1.08, P = .001 by IVW). Subgroup analysis showed that higher SBP was positively associated with Crohn's disease (CD) (OR = 1.06, 95% CI:1.03–1.09, P = 9.18 × 10−5) and ulcerative colitis (UC) (OR = 1.05, 95% CI:1.01–1.09, P = .017) risk, respectively. In reverse‐direction MR analysis, the authors observed no evidence for the causal effect of IBD on blood pressure. Our findings suggested that high SBP was associated with an increased risk of IBD (for both UC and CD). Further studies are required to clarify the underlying mechanism of this causal association.
Collapse
Affiliation(s)
- Xia Xu
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ding Ye
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Bin Liu
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ying Yang
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ying Chen
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu Qian
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China.,Diseases & Population Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, China
| | - Yingying Mao
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaohui Sun
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
578
|
Deng AY. Genetics of Normotension Preventing Hypertension Leads to a Novel Physiological Paradigm. Rev Cardiovasc Med 2022; 23:119. [PMID: 39076226 PMCID: PMC11273859 DOI: 10.31083/j.rcm2304119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 12/05/2021] [Accepted: 12/22/2021] [Indexed: 07/31/2024] Open
Abstract
Possessing blood pressure in normal ranges is considered healthy, and does not warrant medical attention for obvious clinical reasons. However, to realize normotension and then maintain it even when confronted with a hypertensive threat must have its biological 'shield of armour'. While sensitivity to hypertension has been widely recognized and studied, inherent mechanisms that enable a physiological resistance to hypertension to occur have received little attention. Recent advances in normotension genetics have produced unexpected insights. A hypertension 'suppressor' likely inhabits the normotensive genome of inbred Lewis rats. This suppressor behaves as a 'master' control capable of functionally abrogating the effects of hypertension-promoting alleles from multiple quantitative trait loci. This conceptual advancement lays the foundation for uncovering an anti-hypertension gene. Discovering its identity will assist our attempts at developing innovative diagnostic and therapeutic strategies for circumventing and treating hypertension. This new domain of suppressing hypertension goes beyond the conventional pharmacological treatments of hypertension before symptoms appear. For this purpose, a valid theoretical basis and framework is needed that can interpret the experimental data and produce testable predictions for authenticating, enriching or amending the normotension paradigm in the future.
Collapse
Affiliation(s)
- Alan Y. Deng
- Research Centre, Centre hospitalier de l’Université de Montréal, Montréal, QC H2X 0A9, Canada
| |
Collapse
|
579
|
Yeung MW, Wang S, van de Vegte YJ, Borisov O, van Setten J, Snieder H, Verweij N, Said MA, van der Harst P. Twenty-Five Novel Loci for Carotid Intima-Media Thickness: A Genome-Wide Association Study in >45 000 Individuals and Meta-Analysis of >100 000 Individuals. Arterioscler Thromb Vasc Biol 2022; 42:484-501. [PMID: 34852643 PMCID: PMC8939707 DOI: 10.1161/atvbaha.121.317007] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 11/22/2021] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Carotid artery intima-media thickness (cIMT) is a widely accepted marker of subclinical atherosclerosis. Twenty susceptibility loci for cIMT were previously identified and the identification of additional susceptibility loci furthers our knowledge on the genetic architecture underlying atherosclerosis. APPROACH AND RESULTS We performed 3 genome-wide association studies in 45 185 participants from the UK Biobank study who underwent cIMT measurements and had data on minimum, mean, and maximum thickness. We replicated 15 known loci and identified 20 novel loci associated with cIMT at P<5×10-8. Seven novel loci (ZNF385D, ADAMTS9, EDNRA, HAND2, MYOCD, ITCH/EDEM2/MMP24, and MRTFA) were identified in all 3 phenotypes. An additional new locus (LOXL1) was identified in the meta-analysis of the 3 phenotypes. Sex interaction analysis revealed sex differences in 7 loci including a novel locus (SYNE3) in males. Meta-analysis of UK Biobank data with a previous meta-analysis led to identification of three novel loci (APOB, FIP1L1, and LOXL4). Transcriptome-wide association analyses implicated additional genes ARHGAP42, NDRG4, and KANK2. Gene set analysis showed an enrichment in extracellular organization and the PDGF (platelet-derived growth factor) signaling pathway. We found positive genetic correlations of cIMT with coronary artery disease rg=0.21 (P=1.4×10-7), peripheral artery disease rg=0.45 (P=5.3×10-5), and systolic blood pressure rg=0.30 (P=4.0×10-18). A negative genetic correlation between average of maximum cIMT and high-density lipoprotein was found rg=-0.12 (P=7.0×10-4). CONCLUSIONS Genome-wide association meta-analyses in >100 000 individuals identified 25 novel loci associated with cIMT providing insights into genes and tissue-specific regulatory mechanisms of proatherosclerotic processes. We found evidence for shared biological mechanisms with cardiovascular diseases.
Collapse
Affiliation(s)
- Ming Wai Yeung
- Department of Cardiology (M.W.Y., S.W., Y.J.v.d.V., N.V., M.A.S., P.v.d.H.), University of Groningen, University Medical Center Groningen, the Netherlands
| | - Siqi Wang
- Department of Cardiology (M.W.Y., S.W., Y.J.v.d.V., N.V., M.A.S., P.v.d.H.), University of Groningen, University Medical Center Groningen, the Netherlands
- Department of Epidemiology (S.W., H.S.), University of Groningen, University Medical Center Groningen, the Netherlands
- Division of Heart & Lungs, Department of Cardiology, University Medical Center Utrecht, University of Utrecht, the Netherlands (M.W.Y., J.v.S., P.v.d.H.)
| | - Yordi J. van de Vegte
- Department of Cardiology (M.W.Y., S.W., Y.J.v.d.V., N.V., M.A.S., P.v.d.H.), University of Groningen, University Medical Center Groningen, the Netherlands
| | - Oleg Borisov
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Germany (O.B.)
| | - Jessica van Setten
- Department of Epidemiology (S.W., H.S.), University of Groningen, University Medical Center Groningen, the Netherlands
| | - Harold Snieder
- Department of Epidemiology (S.W., H.S.), University of Groningen, University Medical Center Groningen, the Netherlands
| | - Niek Verweij
- Department of Cardiology (M.W.Y., S.W., Y.J.v.d.V., N.V., M.A.S., P.v.d.H.), University of Groningen, University Medical Center Groningen, the Netherlands
| | - M. Abdullah Said
- Department of Cardiology (M.W.Y., S.W., Y.J.v.d.V., N.V., M.A.S., P.v.d.H.), University of Groningen, University Medical Center Groningen, the Netherlands
| | - Pim van der Harst
- Department of Cardiology (M.W.Y., S.W., Y.J.v.d.V., N.V., M.A.S., P.v.d.H.), University of Groningen, University Medical Center Groningen, the Netherlands
- Division of Heart & Lungs, Department of Cardiology, University Medical Center Utrecht, University of Utrecht, the Netherlands (M.W.Y., J.v.S., P.v.d.H.)
| |
Collapse
|
580
|
Huang Y, Hui Q, Gwinn M, Hu YJ, Quyyumi AA, Vaccarino V, Sun YV. Interaction between genetics and smoking in determining risk of coronary artery diseases. Genet Epidemiol 2022; 46:199-212. [PMID: 35170807 PMCID: PMC9086149 DOI: 10.1002/gepi.22446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/18/2021] [Accepted: 01/20/2022] [Indexed: 12/15/2022]
Abstract
Coronary artery disease (CAD) is a preeminent cause of death, and smoking is a strong risk factor for CAD. Genetic factors contribute to the development of CAD, but the interplay between genetic predisposition and smoking history in CAD remains unclear. Using data from the UK Biobank, we constructed several genetic risk scores (GRSs) based on known CAD loci and assessed their interactions with smoking for the development of incident CAD in 307,147 participants of European ancestry who were free of CAD. We fitted Cox proportional hazard models and assessed gene-smoking interaction on both multiplicative and additive scales. Overall, we found no multiplicative interactions, but observed a synergistic additive interaction of GRS with both smoking status and pack-years of smoking, finding that the absolute CAD risk due to smoking was higher for those with high genetic risk. Trait-based sub-GRSs suggested smoking status and smoking intensity measured by pack-years might confer gene-smoking interaction effects with different intermediate risk factors for CAD. Our study results suggest that genetics could modify the effects of smoking on CAD and highlight the value of addressing gene-lifestyle interactions on both additive and multiplicative scales.
Collapse
Affiliation(s)
- Yunfeng Huang
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Qin Hui
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Marta Gwinn
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Yi-Juan Hu
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Arshed A Quyyumi
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Viola Vaccarino
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Yan V Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA,Department of Biomedical Informatics, School of Medicine, Emory University, Atlanta, GA, USA
| |
Collapse
|
581
|
Fang S, Wu J, Reho JJ, Lu KT, Brozoski DT, Kumar G, Werthman AM, Silva SD, Muskus Veitia PC, Wackman KK, Mathison AJ, Teng BQ, Lin CW, Quelle FW, Sigmund CD. RhoBTB1 reverses established arterial stiffness in angiotensin-II hypertension by promoting actin depolymerization. JCI Insight 2022; 7:158043. [PMID: 35358093 PMCID: PMC9090250 DOI: 10.1172/jci.insight.158043] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/30/2022] [Indexed: 11/17/2022] Open
Abstract
Arterial stiffness predicts cardiovascular disease and all-cause mortality, but its treatment remains challenging. Mice treated with angiotensin II (Ang II) develop hypertension, arterial stiffness, vascular dysfunction, and a downregulation of Rho-related BTB domain–containing protein 1 (RhoBTB1) in the vasculature. RhoBTB1 is associated with blood pressure regulation, but its function is poorly understood. We tested the hypothesis that restoring RhoBTB1 can attenuate arterial stiffness, hypertension, and vascular dysfunction in Ang II–treated mice. Genetic complementation of RhoBTB1 in the vasculature was achieved using mice expressing a tamoxifen-inducible, smooth muscle–specific RhoBTB1 transgene. RhoBTB1 restoration efficiently and rapidly alleviated arterial stiffness but not hypertension or vascular dysfunction. Mechanistic studies revealed that RhoBTB1 had no substantial effect on several classical arterial stiffness contributors, such as collagen deposition, elastin content, and vascular smooth muscle remodeling. Instead, Ang II increased actin polymerization in the aorta, which was reversed by RhoBTB1. Changes in the levels of 2 regulators of actin polymerization, cofilin and vasodilator-stimulated phosphoprotein, in response to RhoBTB1 were consistent with an actin depolymerization mechanism. Our study reveals an important function of RhoBTB1, demonstrates its vital role in antagonizing established arterial stiffness, and further supports a functional and mechanistic separation among hypertension, vascular dysfunction, and arterial stiffness.
Collapse
Affiliation(s)
- Shi Fang
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| | - Jing Wu
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| | - John J Reho
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| | - Ko-Ting Lu
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| | - Daniel T Brozoski
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| | - Gaurav Kumar
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| | - Alec M Werthman
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| | - Sebastiao Donato Silva
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| | - Patricia C Muskus Veitia
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| | - Kelsey K Wackman
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| | - Angela J Mathison
- Department of Surgery and the Genomic Sciences and Precision Medicine Cente, Medical College of Wisconsin, Milwawkee, United States of America
| | - Bi Qing Teng
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, United States of America
| | - Chien-Wei Lin
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, United States of America
| | - Frederick W Quelle
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, United States of America
| | - Curt D Sigmund
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| |
Collapse
|
582
|
Lee SB, Park B, Hong KW, Jung DH. Genome-Wide Association of New-Onset Hypertension According to Renin Concentration: The Korean Genome and Epidemiology Cohort Study. J Cardiovasc Dev Dis 2022; 9:jcdd9040104. [PMID: 35448080 PMCID: PMC9025963 DOI: 10.3390/jcdd9040104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/21/2022] [Accepted: 03/29/2022] [Indexed: 11/16/2022] Open
Abstract
The renin-angiotensin system (RAS) is a crucial regulator of vascular resistance and blood volume in the body. This study aimed to examine the genetic predisposition of the plasma renin concentration influencing future hypertension incidence. Based on the Korean Genome and Epidemiology Cohort dataset, 5211 normotensive individuals at enrollment were observed over 12 years, categorized into the low-renin and high-renin groups. We conducted genome-wide association studies for the total, low-renin, and high-renin groups. Among the significant SNPs, the lead SNPs of each locus were focused on for further interpretation. The effect of genotypes was determined by logistic regression analysis between controls and new-onset hypertension, after adjusting for potential confounding variables. During a mean follow-up period of 7.6 years, 1704 participants (32.7%) developed hypertension. The low-renin group showed more incidence rates of new-onset hypertension (35.3%) than the high-renin group (26.5%). Among 153 SNPs in renin-related gene regions, two SNPs (rs11726091 and rs8137145) showed an association in the high-renin group, four SNPs (rs17038966, rs145286444, rs2118663, and rs12336898) in the low-renin group, and three SNPs (rs1938859, rs7968218, and rs117246401) in the total population. Most significantly, the low-renin SNP rs12336898 in the SPTAN1 gene, closely related to vascular wall remodeling, was associated with the development of hypertension (p-value = 1.3 × 10−6). We found the candidate genetic polymorphisms according to blood renin concentration. Our results might be a valuable indicator for hypertension risk prediction and preventive measure, considering renin concentration with genetic susceptibility.
Collapse
Affiliation(s)
- Sung-Bum Lee
- Severance Check-up, Yonsei University Health System, Yongin-si 16995, Korea;
- Department of Medicine, Graduate School, Yonsei University Wonju College of Medicine, Wonju-si 26426, Korea
| | - Byoungjin Park
- Department of Family Medicine, Yongin Severance Hosptal, Yongin-si 16995, Korea;
| | - Kyung-Won Hong
- Healthcare R&D Division, Theragen Bio Co., Ltd., Ganggyo-ro 145, Suwon-si 16229, Korea
- Correspondence: (K.-W.H.); (D.-H.J.)
| | - Dong-Hyuk Jung
- Department of Family Medicine, Yongin Severance Hosptal, Yongin-si 16995, Korea;
- Department of Family Medicine, Yonsei University College of Medicine, Seoul 03722, Korea
- Correspondence: (K.-W.H.); (D.-H.J.)
| |
Collapse
|
583
|
Chai T, Tian M, Yang X, Qiu Z, Lin X, Chen L. Association of Circulating Cathepsin B Levels With Blood Pressure and Aortic Dilation. Front Cardiovasc Med 2022; 9:762468. [PMID: 35425820 PMCID: PMC9001941 DOI: 10.3389/fcvm.2022.762468] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 03/07/2022] [Indexed: 12/04/2022] Open
Abstract
Hypertension is a key risk factor for spontaneous coronary artery dissection (SCAD) and aortic dilation. Circulating proteins play key roles in a range of biological processes and represent a major source of druggable targets. The aim of this study was to identify circulating proteins that were associated with blood pressure (BP), SCAD and aortic dilation. We identified shared genetic variants of BP and SCAD in genome-wide association studies, searched for circulating protein affected by these variants and examined the association of circulating protein levels with BP, aortic aneurysm and dissection (AAD) and aortic diameters by integrating data from circulating protein quantitative trait loci (pQTL) studies and genome wide association study (GWAS) in individuals from the UK Biobank using two-sample Mendelian randomization analysis methods. Single nucleotide polymorphisms (SNPs) in JAG1, ERI1, ULK4, THSD4, CMIP, COL4A2, FBN1, FAM76B, FGGY, NUS1, and HNF4G, which were related to extracellular matrix components, were associated with both BP and SCAD. We found 49 significant pQTL signals among these SNPs. The regulated proteins were encoded by MMP10, IL6R, FIGF, MMP1, CTSB, IGHG1, DSG2, TTC17, RETN, POMC, SCARF2, RELT, and GALNT16, which were enriched in biological processes such as collagen metabolic process and multicellular organism metabolic process. Causal associations between BP and AAD and aortic diameters were detected. Significant associations between circulating levels of cathepsin B, a well-known prorenin processing enzyme, and BP and aortic diameters were identified by using several Mendelian randomization analysis methods and were validated by independent data.
Collapse
Affiliation(s)
- Tianci Chai
- Department of Cardiovasclar Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
- Department of Anesthesiology, Xinyi People’s Hospital, Xuzhou, China
| | - Mengyue Tian
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xiaojie Yang
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhihuang Qiu
- Department of Cardiovasclar Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| | - Xinjian Lin
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Liangwan Chen
- Department of Cardiovasclar Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
- *Correspondence: Liangwan Chen,
| |
Collapse
|
584
|
Guo J, Guo X, Sun Y, Li Z, Jia P. Application of omics in hypertension and resistant hypertension. Hypertens Res 2022; 45:775-788. [PMID: 35264783 DOI: 10.1038/s41440-022-00885-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/11/2022] [Accepted: 01/29/2022] [Indexed: 12/12/2022]
Abstract
Hypertension is a major modifiable risk factor that affects the global health burden. Despite the availability of multiple antihypertensive drugs, blood pressure is often not optimally controlled. The prevalence of true resistant hypertension in treated hypertensive patients is ~2-20%, and these patients are at higher risk for adverse events and poor clinical outcomes. Therefore, an in-depth dissection of the pathophysiological mechanisms of hypertension and resistant hypertension is needed to identify more effective targets for regulating blood pressure. Omics technologies, such as genomics, transcriptomics, proteomics, metabolomics, and microbiomics, can accurately present the characteristics of organisms at varying molecular levels. Integrative omics can further reveal the network of interactions between molecular levels and provide a complete dynamic view of the organism. In this review, we describe the applications, progress, and challenges of omics technologies in hypertension. Specifically, we discuss the application of omics in resistant hypertension. We believe that omics approaches will produce a better understanding of the pathogenesis of hypertension and resistant hypertension and improve diagnostic and therapeutic strategies, thus increasing rates of blood pressure control and reducing the public health burden of hypertension.
Collapse
Affiliation(s)
- Jiuqi Guo
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Xiaofan Guo
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Yingxian Sun
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Zhao Li
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, 110001, China.
| | - Pengyu Jia
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
585
|
Zhang H, Mo X, Wang A, Peng H, Guo D, Zhong C, Zhu Z, Xu T, Zhang Y. Association of DNA Methylation in Blood Pressure-Related Genes With Ischemic Stroke Risk and Prognosis. Front Cardiovasc Med 2022; 9:796245. [PMID: 35345488 PMCID: PMC8957103 DOI: 10.3389/fcvm.2022.796245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 01/31/2022] [Indexed: 12/16/2022] Open
Abstract
BackgroundA genome-wide association study identified 12 genetic loci influencing blood pressure and implicated a role of DNA methylation. However, the relationship between methylation and ischemic stroke has not yet been clarified. We conducted a large-sample sequencing study to identify blood leukocyte DNA methylations as novel biomarkers for ischemic stroke risk and prognosis based on previously identified genetic loci.MethodsMethylation levels of 17 genes were measured by sequencing in 271 ischemic stroke cases and 323 controls, and the significant associations were validated in another independent sample of 852 cases and 925 controls. The associations between methylation levels and ischemic stroke risk and prognosis were evaluated.ResultsMethylation of AMH, C17orf82, HDAC9, IGFBP3, LRRC10B, PDE3A, PRDM6, SYT7 and TBX2 was significantly associated with ischemic stroke. Compared to participants without any hypomethylated targets, the odds ratio (OR) (95% confidence interval, CI) for those with 9 hypomethylated genes was 1.41 (1.33–1.51) for ischemic stroke. Adding methylation levels of the 9 genes to the basic model of traditional risk factors significantly improved the risk stratification for ischemic stroke. Associations between AMH, HDAC9, IGFBP3, PDE3A and PRDM6 gene methylation and modified Rankin Scale scores were significant after adjustment for covariates. Lower methylation levels of AMH, C17orf82, PRDM6 and TBX2 were significantly associated with increased 3-month mortality. Compared to patients without any hypomethylated targets, the OR (95% CI) for those with 4 hypomethylated targets was 1.12 (1.08–1.15) for 3-month mortality (P = 2.28 × 10−10).ConclusionThe present study identified blood leukocyte DNA methylations as potential factors affecting ischemic stroke risk and prognosis among Han Chinese individuals.
Collapse
Affiliation(s)
- Huan Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Xingbo Mo
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Aili Wang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Hao Peng
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Daoxia Guo
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Chongke Zhong
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Zhengbao Zhu
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Tan Xu
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Yonghong Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
- *Correspondence: Yonghong Zhang
| |
Collapse
|
586
|
Ko S, German CA, Jensen A, Shen J, Wang A, Mehrotra DV, Sun YV, Sinsheimer JS, Zhou H, Zhou JJ. GWAS of longitudinal trajectories at biobank scale. Am J Hum Genet 2022; 109:433-445. [PMID: 35196515 PMCID: PMC8948167 DOI: 10.1016/j.ajhg.2022.01.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 01/25/2022] [Indexed: 12/12/2022] Open
Abstract
Biobanks linked to massive, longitudinal electronic health record (EHR) data make numerous new genetic research questions feasible. One among these is the study of biomarker trajectories. For example, high blood pressure measurements over visits strongly predict stroke onset, and consistently high fasting glucose and Hb1Ac levels define diabetes. Recent research reveals that not only the mean level of biomarker trajectories but also their fluctuations, or within-subject (WS) variability, are risk factors for many diseases. Glycemic variation, for instance, is recently considered an important clinical metric in diabetes management. It is crucial to identify the genetic factors that shift the mean or alter the WS variability of a biomarker trajectory. Compared to traditional cross-sectional studies, trajectory analysis utilizes more data points and captures a complete picture of the impact of time-varying factors, including medication history and lifestyle. Currently, there are no efficient tools for genome-wide association studies (GWASs) of biomarker trajectories at the biobank scale, even for just mean effects. We propose TrajGWAS, a linear mixed effect model-based method for testing genetic effects that shift the mean or alter the WS variability of a biomarker trajectory. It is scalable to biobank data with 100,000 to 1,000,000 individuals and many longitudinal measurements and robust to distributional assumptions. Simulation studies corroborate that TrajGWAS controls the type I error rate and is powerful. Analysis of eleven biomarkers measured longitudinally and extracted from UK Biobank primary care data for more than 150,000 participants with 1,800,000 observations reveals loci that significantly alter the mean or WS variability.
Collapse
Affiliation(s)
- Seyoon Ko
- Department of Computational Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Christopher A German
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Aubrey Jensen
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Judong Shen
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Anran Wang
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Devan V Mehrotra
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Yan V Sun
- Department of Epidemiology, Emory University, Atlanta, GA 30322, USA
| | - Janet S Sinsheimer
- Department of Computational Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Hua Zhou
- Department of Computational Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Jin J Zhou
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Epidemiology and Biostatistics, University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
587
|
Zhu X, Zhu L, Wang H, Cooper RS, Chakravarti A. Genome-wide pleiotropy analysis identifies novel blood pressure variants and improves its polygenic risk scores. Genet Epidemiol 2022; 46:105-121. [PMID: 34989438 PMCID: PMC8863647 DOI: 10.1002/gepi.22440] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/07/2021] [Indexed: 01/21/2023]
Abstract
Systolic and diastolic blood pressure (S/DBP) are highly correlated modifiable risk factors for cardiovascular disease (CVD). We report here a bidirectional Mendelian Randomization (MR) and horizontal pleiotropy analysis of S/DBP summary statistics from the UK Biobank (UKB)-International Consortium for Blood Pressure (ICBP) (UKB-ICBP) BP genome-wide association study and construct a composite genetic risk score (GRS) by including pleiotropic variants. The composite GRS captures greater (1.11-3.26 fold) heritability for BP traits and increases (1.09- and 2.01-fold) Nagelkerke's R2 for hypertension and CVD. We replicated 118 novel BP horizontal pleiotropic variants including 18 novel BP loci using summary statistics from the Million Veteran Program (MVP) study. An additional 219 novel BP signals and 40 novel loci were identified after a meta-analysis of the UKB-ICBP and MVP summary statistics but without further independent replication. Our study provides further insight into BP regulation and provides a novel way to construct a GRS by including pleiotropic variants for other complex diseases.
Collapse
Affiliation(s)
- Xiaofeng Zhu
- Department of Population and Quantitative Health SciencesCase Western Reserve UniversityClevelandOhioUSA
| | - Luke Zhu
- Department of Medicine, Center for Human Genetics & GenomicsNew York University Langone HealthNew YorkNew YorkUSA
| | - Heming Wang
- Division of Sleep and Circadian DisordersBrigham and Women's HospitalBostonMassachusettsUSA
| | - Richard S. Cooper
- Department of Public Health Sciences, Stritch School of MedicineLoyola University ChicagoMaywoodIllinoisUSA
| | - Aravinda Chakravarti
- Department of Medicine, Center for Human Genetics & GenomicsNew York University Langone HealthNew YorkNew YorkUSA
| |
Collapse
|
588
|
Groenland EH, Heidemann BE, van der Laan SW, van Setten J, Koopal C, Bots ML, Asselbergs FW, Visseren FLJ, Spiering W. Genetic variants associated with low-density lipoprotein cholesterol and systolic blood pressure and the risk of recurrent cardiovascular disease in patients with established vascular disease. Atherosclerosis 2022; 350:102-108. [DOI: 10.1016/j.atherosclerosis.2022.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/09/2022] [Accepted: 03/03/2022] [Indexed: 01/09/2023]
|
589
|
Barbu MC, Huider F, Campbell A, Amador C, Adams MJ, Lynall ME, Howard DM, Walker RM, Morris SW, Van Dongen J, Porteous DJ, Evans KL, Bullmore E, Willemsen G, Boomsma DI, Whalley HC, McIntosh AM. Methylome-wide association study of antidepressant use in Generation Scotland and the Netherlands Twin Register implicates the innate immune system. Mol Psychiatry 2022; 27:1647-1657. [PMID: 34880450 PMCID: PMC9095457 DOI: 10.1038/s41380-021-01412-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 10/11/2021] [Accepted: 11/26/2021] [Indexed: 12/28/2022]
Abstract
Antidepressants are an effective treatment for major depressive disorder (MDD), although individual response is unpredictable and highly variable. Whilst the mode of action of antidepressants is incompletely understood, many medications are associated with changes in DNA methylation in genes that are plausibly linked to their mechanisms. Studies of DNA methylation may therefore reveal the biological processes underpinning the efficacy and side effects of antidepressants. We performed a methylome-wide association study (MWAS) of self-reported antidepressant use accounting for lifestyle factors and MDD in Generation Scotland (GS:SFHS, N = 6428, EPIC array) and the Netherlands Twin Register (NTR, N = 2449, 450 K array) and ran a meta-analysis of antidepressant use across these two cohorts. We found ten CpG sites significantly associated with self-reported antidepressant use in GS:SFHS, with the top CpG located within a gene previously associated with mental health disorders, ATP6V1B2 (β = -0.055, pcorrected = 0.005). Other top loci were annotated to genes including CASP10, TMBIM1, MAPKAPK3, and HEBP2, which have previously been implicated in the innate immune response. Next, using penalised regression, we trained a methylation-based score of self-reported antidepressant use in a subset of 3799 GS:SFHS individuals that predicted antidepressant use in a second subset of GS:SFHS (N = 3360, β = 0.377, p = 3.12 × 10-11, R2 = 2.12%). In an MWAS analysis of prescribed selective serotonin reuptake inhibitors, we showed convergent findings with those based on self-report. In NTR, we did not find any CpGs significantly associated with antidepressant use. The meta-analysis identified the two CpGs of the ten above that were common to the two arrays used as being significantly associated with antidepressant use, although the effect was in the opposite direction for one of them. Antidepressants were associated with epigenetic alterations in loci previously associated with mental health disorders and the innate immune system. These changes predicted self-reported antidepressant use in a subset of GS:SFHS and identified processes that may be relevant to our mechanistic understanding of clinically relevant antidepressant drug actions and side effects.
Collapse
Affiliation(s)
- Miruna C Barbu
- Division of Psychiatry, The University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK.
| | - Floris Huider
- Faculty of Behavioural and Movement Sciences, Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, The Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Carmen Amador
- MRC Human Genetics Unit, The Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Mark J Adams
- Division of Psychiatry, The University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK
| | | | - David M Howard
- Division of Psychiatry, The University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Rosie M Walker
- Centre for Genomic and Experimental Medicine, The Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Stewart W Morris
- Centre for Genomic and Experimental Medicine, The Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Jenny Van Dongen
- Faculty of Behavioural and Movement Sciences, Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - David J Porteous
- Centre for Genomic and Experimental Medicine, The Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Kathryn L Evans
- Centre for Genomic and Experimental Medicine, The Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Edward Bullmore
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Gonneke Willemsen
- Faculty of Behavioural and Movement Sciences, Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Dorret I Boomsma
- Faculty of Behavioural and Movement Sciences, Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Heather C Whalley
- Division of Psychiatry, The University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK
| | - Andrew M McIntosh
- Division of Psychiatry, The University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK
| |
Collapse
|
590
|
Kerns SL, Amidon Morlang A, Lee SM, Peterson DR, Marples B, Zhang H, Bylund K, Rosenzweig D, Hall W, De Ruyck K, Rosenstein BS, Stock RG, Gómez-Caamaño A, Vega A, Sosa-Fajardo P, Taboada-Valladares B, Aguado-Barrera ME, Parker C, Veldeman L, Fonteyne V, Bultijnck R, Talbot CJ, Symonds RP, Johnson K, Rattay T, Webb A, Lambrecht M, de Ruysscher D, Vanneste B, Choudhury A, Elliott RM, Sperk E, Herskind C, Veldwijk MR, Rancati T, Avuzzi B, Valdagni R, Azria D, Farcy Jacquet MP, Chang-Claude J, Seibold P, West C, Janelsins M, Chen Y, Messing E, Morrow G. Use of angiotensin converting enzyme inhibitors is associated with reduced risk of late bladder toxicity following radiotherapy for prostate cancer. Radiother Oncol 2022; 168:75-82. [PMID: 35077710 PMCID: PMC8986577 DOI: 10.1016/j.radonc.2022.01.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND PURPOSE Genome-wide association studies (GWAS) of late hematuria following prostate cancer radiotherapy identified single nucleotide polymorphisms (SNPs) near AGT, encoding angiotensinogen. We tested the hypothesis that patients taking angiotensin converting enzyme inhibitors (ACEi) have a reduced risk of late hematuria. We additionally tested genetically-defined hypertension. MATERIALS AND METHODS Prostate cancer patients undergoing potentially-curative radiotherapy were enrolled onto two multi-center observational studies, URWCI (N = 256) and REQUITE (N = 1,437). Patients were assessed pre-radiotherapy and followed prospectively for development of toxicity for up to four years. The cumulative probability of hematuria was estimated by the Kaplan-Meier method. Multivariable grouped relative risk models assessed the effect of ACEi on time to hematuria adjusting for clinical factors and stratified by enrollment site. A polygenic risk score (PRS) for blood pressure was tested for association with hematuria in REQUITE and our Radiogenomics Consortium GWAS. RESULTS Patients taking ACEi during radiotherapy had a reduced risk of hematuria (HR 0.51, 95%CI 0.28 to 0.94, p = 0.030) after adjusting for prior transurethral prostate and/or bladder resection, heart disease, pelvic node radiotherapy, and bladder volume receiving 70 Gy, which are associated with hematuria. A blood pressure PRS was associated with hypertension (odds ratio per standard deviation 1.38, 95%CI 1.31 to 1.46, n = 5,288, p < 0.001) but not hematuria (HR per standard deviation 0.96, 95%CI 0.87 to 1.06, n = 5,126, p = 0.41). CONCLUSIONS Our study is the first to show a radioprotective effect of ACEi on bladder in an international, multi-site study of patients receiving pelvic radiotherapy. Mechanistic studies are needed to understand how targeting the angiotensin pathway protects the bladder.
Collapse
Affiliation(s)
- Sarah L Kerns
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, United States.
| | - Ashley Amidon Morlang
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, United States
| | - Sharon M Lee
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, United States
| | - Derick R Peterson
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, United States
| | - Brian Marples
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, United States
| | - Hong Zhang
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, United States
| | - Kevin Bylund
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, United States
| | - Doug Rosenzweig
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, United States
| | - William Hall
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, United States
| | - Kim De Ruyck
- Department of Radiation Oncology, Ghent University Hospital and Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Barry S Rosenstein
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, United States; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Richard G Stock
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Antonio Gómez-Caamaño
- Department of Radiation Oncology, Complexo Hospitalario Universitario de Santiago, SERGAS, Santiago de Compostela, Spain; Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Ana Vega
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, Spain; Fundación Pública Galega de Medicina Xenómica-Servizo Galego de Saude (SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Santiago de Compostela, Spain
| | - Paloma Sosa-Fajardo
- Department of Radiation Oncology, Complexo Hospitalario Universitario de Santiago, SERGAS, Santiago de Compostela, Spain; Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, Spain; Fundación Pública Galega de Medicina Xenómica-Servizo Galego de Saude (SERGAS), Santiago de Compostela, Spain
| | - Begoña Taboada-Valladares
- Department of Radiation Oncology, Complexo Hospitalario Universitario de Santiago, SERGAS, Santiago de Compostela, Spain; Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Miguel E Aguado-Barrera
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, Spain; Fundación Pública Galega de Medicina Xenómica-Servizo Galego de Saude (SERGAS), Santiago de Compostela, Spain
| | - Chris Parker
- Department of Uro-oncology, The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, Sutton, United Kingdom
| | - Liv Veldeman
- Department of Radiation Oncology, Ghent University Hospital and Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Valérie Fonteyne
- Department of Radiation Oncology, Ghent University Hospital and Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Renée Bultijnck
- Department of Radiation Oncology, Ghent University Hospital and Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | | | - R Paul Symonds
- Leicester Cancer Research Centre, University of Leicester, United Kingdom
| | - Kerstie Johnson
- Leicester Cancer Research Centre, University of Leicester, United Kingdom
| | - Tim Rattay
- Leicester Cancer Research Centre, University of Leicester, United Kingdom
| | - Adam Webb
- Department of Genetics and Genome Biology, University of Leicester, United Kingdom
| | | | - Dirk de Ruysscher
- KU Leuven, Radiation Oncology, Leuven, Belgium; Maastricht University Medical Center, Department of Radiation Oncology (Maastro Clinic), GROW School for Oncology and Developmental Biology, Maastricht, the Netherlands
| | - Ben Vanneste
- KU Leuven, Radiation Oncology, Leuven, Belgium; Maastricht University Medical Center, Department of Radiation Oncology (Maastro Clinic), GROW School for Oncology and Developmental Biology, Maastricht, the Netherlands
| | - Ananya Choudhury
- Division of Cancer Sciences, the University of Manchester, Manchester Academic Health Science Centre, Christie Hospital, Manchester, the Netherlands
| | - Rebecca M Elliott
- Division of Cancer Sciences, the University of Manchester, Manchester Academic Health Science Centre, Christie Hospital, Manchester, the Netherlands
| | - Elena Sperk
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Carsten Herskind
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Marlon R Veldwijk
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Tiziana Rancati
- Prostate Cancer Program, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Barbara Avuzzi
- Department of Radiation Oncology 1, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Riccardo Valdagni
- Prostate Cancer Program, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Department of Radiation Oncology 1, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Department of Oncology and Hemato-oncology, Università degli Studi di Milano, Milan, Italy
| | - David Azria
- Department of Radiation Oncology, Montpellier Cancer Institute, Université Montpellier, Inserm U1194, France
| | | | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Cancer Epidemiology Group, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Germany
| | - Petra Seibold
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Catharine West
- Division of Cancer Sciences, the University of Manchester, Manchester Academic Health Science Centre, Christie Hospital, Manchester, the Netherlands
| | - Michelle Janelsins
- Department of Surgery, University of Rochester Medical Center, Rochester, United States
| | - Yuhchyau Chen
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, United States
| | - Edward Messing
- Department of Urology, University of Rochester Medical Center, Rochester, United States
| | - Gary Morrow
- Department of Surgery, University of Rochester Medical Center, Rochester, United States
| |
Collapse
|
591
|
High blood pressure in children and adolescents: current perspectives and strategies to improve future kidney and cardiovascular health. Kidney Int Rep 2022; 7:954-970. [PMID: 35570999 PMCID: PMC9091586 DOI: 10.1016/j.ekir.2022.02.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 02/15/2022] [Accepted: 02/21/2022] [Indexed: 02/08/2023] Open
Abstract
Hypertension is one of the most common causes of preventable death worldwide. The prevalence of pediatric hypertension has increased significantly in recent decades. The cause of this is likely multifactorial, related to increasing childhood obesity, high dietary sodium intake, sedentary lifestyles, perinatal factors, familial aggregation, socioeconomic factors, and ethnic blood pressure (BP) differences. Pediatric hypertension represents a major public health threat. Uncontrolled pediatric hypertension is associated with subclinical cardiovascular disease and adult-onset hypertension. In children with chronic kidney disease (CKD), hypertension is also a strong risk factor for progression to kidney failure. Despite these risks, current rates of pediatric BP screening, hypertension detection, treatment, and control remain suboptimal. Contributing to these shortcomings are the challenges of accurately measuring pediatric BP, limited access to validated pediatric equipment and hypertension specialists, complex interpretation of pediatric BP measurements, problematic normative BP data, and conflicting society guidelines for pediatric hypertension. To date, limited pediatric hypertension research has been conducted to help address these challenges. However, there are several promising signs in the field of pediatric hypertension. There is greater attention being drawn on the cardiovascular risks of pediatric hypertension, more emphasis on the need for childhood BP screening and management, new public health initiatives being implemented, and increasing research interest and funding. This article summarizes what is currently known about pediatric hypertension, the existing knowledge-practice gaps, and ongoing research aimed at improving future kidney and cardiovascular health.
Collapse
|
592
|
Nuotio ML, Sánez Tähtisalo H, Lahtinen A, Donner K, Fyhrquist F, Perola M, Kontula KK, Hiltunen TP. Pharmacoepigenetics of hypertension: genome-wide methylation analysis of responsiveness to four classes of antihypertensive drugs using a double-blind crossover study design. Epigenetics 2022; 17:1432-1445. [PMID: 35213289 PMCID: PMC9586691 DOI: 10.1080/15592294.2022.2038418] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Essential hypertension remains the leading risk factor of global disease burden, but its treatment goals are often not met. We investigated whether DNA methylation is associated with antihypertensive responses to a diuretic, a beta-blocker, a calcium channel blocker or an angiotensin receptor antagonist. In addition, since we previously showed an SNP at the transcription start site (TSS) of the catecholamine biosynthesis-related ACY3 gene to associate with blood pressure (BP) response to beta-blockers, we specifically analysed the association of methylation sites close to the ACY3 TSS with BP responses to beta-blockers. We conducted an epigenome-wide association study between leukocyte DNA methylation and BP responses to antihypertensive monotherapies in two hypertensive Finnish cohorts: the GENRES (https://clinicaltrials.gov/ct2/show/NCT03276598; amlodipine 5 mg, bisoprolol 5 mg, hydrochlorothiazide 25 mg, or losartan 50 mg daily) and the LIFE-Fin studies (https://clinicaltrials.gov/ct2/show/NCT00338260; atenolol 50 mg or losartan 50 mg daily). The monotherapy groups consisted of approximately 200 individuals each. We identified 64 methylation sites to suggestively associate (P < 1E-5) with either systolic or diastolic BP responses to a particular study drug in GENRES. These associations did not replicate in LIFE-Fin . Three methylation sites close to the ACY3 TSS were associated with systolic BP responses to bisoprolol in GENRES but not genome-wide significantly (P < 0.05). No robust associations between DNA methylation and BP responses to four different antihypertensive drugs were identified. However, the findings on the methylation sites close to the ACY3 TSS may support the role of ACY3 genetic and epigenetic variation in BP response to bisoprolol.
Collapse
Affiliation(s)
- Marja-Liisa Nuotio
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Public Health Solutions, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Heini Sánez Tähtisalo
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Alexandra Lahtinen
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kati Donner
- Technology Centre, Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Frej Fyhrquist
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Markus Perola
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Public Health Solutions, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Kimmo K Kontula
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Timo P Hiltunen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
593
|
Geng TT, Jafar TH. Hypertension Pharmacogenomics in CKD: The Clinical Relevance and Public Health Implications. KIDNEY360 2022; 3:204-207. [PMID: 35373121 PMCID: PMC8967644 DOI: 10.34067/kid.0007792021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 01/19/2022] [Indexed: 04/11/2023]
Affiliation(s)
- Ting-Ting Geng
- Department of Epidemiology and Biostatistics, Huazhong University of Science and Technology, Wuhan, China
| | - Tazeen H. Jafar
- Programme in Health Services and Systems Research, Duke-NUS Medical School, Singapore, Singapore
- Department of Renal Medicine, Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
594
|
Toropainen A, Stolze LK, Örd T, Whalen MB, Torrell PM, Link VM, Kaikkonen MU, Romanoski CE. Functional noncoding SNPs in human endothelial cells fine-map vascular trait associations. Genome Res 2022; 32:409-424. [PMID: 35193936 PMCID: PMC8896458 DOI: 10.1101/gr.276064.121] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 01/06/2022] [Indexed: 11/25/2022]
Abstract
Functional consequences of genetic variation in the noncoding human genome are difficult to ascertain despite demonstrated associations to common, complex disease traits. To elucidate properties of functional noncoding SNPs with effects in human endothelial cells (ECs), we utilized our previous molecular quantitative trait locus (molQTL) analysis for transcription factor binding, chromatin accessibility, and H3K27 acetylation to nominate a set of likely functional noncoding SNPs. Together with information from genome-wide association studies (GWASs) for vascular disease traits, we tested the ability of 34,344 variants to perturb enhancer function in ECs using the highly multiplexed STARR-seq assay. Of these, 5711 variants validated, whose enriched attributes included: (1) mutations to TF binding motifs for ETS or AP-1 that are regulators of the EC state; (2) location in accessible and H3K27ac-marked EC chromatin; and (3) molQTL associations whereby alleles associate with differences in chromatin accessibility and TF binding across genetically diverse ECs. Next, using pro-inflammatory IL1B as an activator of cell state, we observed robust evidence (>50%) of context-specific SNP effects, underscoring the prevalence of noncoding gene-by-environment (GxE) effects. Lastly, using these cumulative data, we fine-mapped vascular disease loci and highlighted evidence suggesting mechanisms by which noncoding SNPs at two loci affect risk for pulse pressure/large artery stroke and abdominal aortic aneurysm through respective effects on transcriptional regulation of POU4F1 and LDAH. Together, we highlight the attributes and context dependence of functional noncoding SNPs and provide new mechanisms underlying vascular disease risk.
Collapse
Affiliation(s)
- Anu Toropainen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio 70211, Finland
| | - Lindsey K Stolze
- The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona 85721, USA.,The Genetics Interdisciplinary Graduate Program, The University of Arizona, Tucson, Arizona 85721, USA
| | - Tiit Örd
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio 70211, Finland
| | - Michael B Whalen
- The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona 85721, USA
| | - Paula Martí Torrell
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio 70211, Finland
| | - Verena M Link
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Minna U Kaikkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio 70211, Finland
| | - Casey E Romanoski
- The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona 85721, USA.,The Genetics Interdisciplinary Graduate Program, The University of Arizona, Tucson, Arizona 85721, USA
| |
Collapse
|
595
|
Hartmann K, Seweryn M, Sadee W. Interpreting coronary artery disease GWAS results: A functional genomics approach assessing biological significance. PLoS One 2022; 17:e0244904. [PMID: 35192625 PMCID: PMC8863290 DOI: 10.1371/journal.pone.0244904] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/01/2022] [Indexed: 01/09/2023] Open
Abstract
Genome-wide association studies (GWAS) have implicated 58 loci in coronary artery disease (CAD). However, the biological basis for these associations, the relevant genes, and causative variants often remain uncertain. Since the vast majority of GWAS loci reside outside coding regions, most exert regulatory functions. Here we explore the complexity of each of these loci, using tissue specific RNA sequencing data from GTEx to identify genes that exhibit altered expression patterns in the context of GWAS-significant loci, expanding the list of candidate genes from the 75 currently annotated by GWAS to 245, with almost half of these transcripts being non-coding. Tissue specific allelic expression imbalance data, also from GTEx, allows us to uncover GWAS variants that mark functional variation in a locus, e.g., rs7528419 residing in the SORT1 locus, in liver specifically, and rs72689147 in the GUYC1A1 locus, across a variety of tissues. We consider the GWAS variant rs1412444 in the LIPA locus in more detail as an example, probing tissue and transcript specific effects of genetic variation in the region. By evaluating linkage disequilibrium (LD) between tissue specific eQTLs, we reveal evidence for multiple functional variants within loci. We identify 3 variants (rs1412444, rs1051338, rs2250781) that when considered together, each improve the ability to account for LIPA gene expression, suggesting multiple interacting factors. These results refine the assignment of 58 GWAS loci to likely causative variants in a handful of cases and for the remainder help to re-prioritize associated genes and RNA isoforms, suggesting that ncRNAs maybe a relevant transcript in almost half of CAD GWAS results. Our findings support a multi-factorial system where a single variant can influence multiple genes and each genes is regulated by multiple variants.
Collapse
Affiliation(s)
- Katherine Hartmann
- Department of Cancer Biology and Genetics, Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus, OH, United States of America
| | - Michał Seweryn
- Biobank Lab, Department of Molecular Biophysics, University of Lodz, Lodz, Poland
| | - Wolfgang Sadee
- Department of Cancer Biology and Genetics, Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus, OH, United States of America
| |
Collapse
|
596
|
Tsao CW, Aday AW, Almarzooq ZI, Alonso A, Beaton AZ, Bittencourt MS, Boehme AK, Buxton AE, Carson AP, Commodore-Mensah Y, Elkind MSV, Evenson KR, Eze-Nliam C, Ferguson JF, Generoso G, Ho JE, Kalani R, Khan SS, Kissela BM, Knutson KL, Levine DA, Lewis TT, Liu J, Loop MS, Ma J, Mussolino ME, Navaneethan SD, Perak AM, Poudel R, Rezk-Hanna M, Roth GA, Schroeder EB, Shah SH, Thacker EL, VanWagner LB, Virani SS, Voecks JH, Wang NY, Yaffe K, Martin SS. Heart Disease and Stroke Statistics-2022 Update: A Report From the American Heart Association. Circulation 2022; 145:e153-e639. [PMID: 35078371 DOI: 10.1161/cir.0000000000001052] [Citation(s) in RCA: 3174] [Impact Index Per Article: 1058.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND The American Heart Association, in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, diet, and weight) and health factors (cholesterol, blood pressure, and glucose control) that contribute to cardiovascular health. The Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure, valvular disease, venous disease, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The American Heart Association, through its Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States to provide the most current information available in the annual Statistical Update. The 2022 Statistical Update is the product of a full year's worth of effort by dedicated volunteer clinicians and scientists, committed government professionals, and American Heart Association staff members. This year's edition includes data on the monitoring and benefits of cardiovascular health in the population and an enhanced focus on social determinants of health, adverse pregnancy outcomes, vascular contributions to brain health, and the global burden of cardiovascular disease and healthy life expectancy. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
597
|
Vaura F, Palmu J, Aittokallio J, Kauko A, Niiranen T. Genetic, Molecular, and Cellular Determinants of Sex-Specific Cardiovascular Traits. Circ Res 2022; 130:611-631. [PMID: 35175841 DOI: 10.1161/circresaha.121.319891] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Despite the well-known sex dimorphism in cardiovascular disease traits, the exact genetic, molecular, and cellular underpinnings of these differences are not well understood. A growing body of evidence currently points at the links between cardiovascular disease traits and the genome, epigenome, transcriptome, and metabolome. However, the sex-specific differences in these links remain largely unstudied due to challenges in bioinformatic methods, inadequate statistical power, analytic costs, and paucity of valid experimental models. This review article provides an overview of the literature on sex differences in genetic architecture, heritability, epigenetic changes, transcriptomic signatures, and metabolomic profiles in relation to cardiovascular disease traits. We also review the literature on the associations between sex hormones and cardiovascular disease traits and discuss the potential mechanisms underlying these associations, focusing on human studies.
Collapse
Affiliation(s)
- Felix Vaura
- Department of Internal Medicine (F.V., J.P., A.K., T.N.), University of Turku, Finland
| | - Joonatan Palmu
- Department of Internal Medicine (F.V., J.P., A.K., T.N.), University of Turku, Finland
| | - Jenni Aittokallio
- Department of Anesthesiology and Intensive Care (J.A.), University of Turku, Finland.,Division of Perioperative Services, Intensive Care and Pain Medicine (J.A.), Turku University Hospital, Finland
| | - Anni Kauko
- Department of Internal Medicine (F.V., J.P., A.K., T.N.), University of Turku, Finland
| | - Teemu Niiranen
- Department of Internal Medicine (F.V., J.P., A.K., T.N.), University of Turku, Finland.,Division of Medicine (T.N.), Turku University Hospital, Finland.,Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland (T.N.)
| |
Collapse
|
598
|
Zheng Z, Hua R, Xu G, Yang H, Shi P. Gene losses may contribute to subterranean adaptations in naked mole-rat and blind mole-rat. BMC Biol 2022; 20:44. [PMID: 35172813 PMCID: PMC8851862 DOI: 10.1186/s12915-022-01243-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/28/2022] [Indexed: 01/18/2023] Open
Abstract
Background Naked mole-rats (Heterocephalus glaber, NMRs) and blind mole-rats (Spalax galili, BMRs) are representative subterranean rodents that have evolved many extraordinary traits, including hypoxia tolerance, longevity, and cancer resistance. Although multiple candidate loci responsible for these traits have been uncovered by genomic studies, many of them are limited to functional changes to amino acid sequence and little is known about the contributions of other genetic events. To address this issue, we focused on gene losses (unitary pseudogenes) and systematically analyzed gene losses in NMRs and BMRs, aiming to elucidate the potential roles of pseudogenes in their adaptation to subterranean lifestyle. Results We obtained the pseudogene repertoires in NMRs and BMRs, as well as their respective aboveground relatives, guinea pigs and rats, on a genome-wide scale. As a result, 167, 139, 341, and 112 pseudogenes were identified in NMRs, BMRs, guinea pigs, and rats, respectively. Functional enrichment analysis identified 4 shared and 2 species-specific enriched functional groups (EFGs) in subterranean lineages. Notably, the pseudogenes in these EFGs might be associated with either regressive (e.g., visual system) or adaptive (e.g., altered DNA damage response) traits. In addition, several pseudogenes including TNNI3K and PDE5A might be associated with specific cardiac features observed in subterranean lineages. Interestingly, we observed 20 convergent gene losses in NMRs and BMRs. Given that the functional investigations of these genes are generally scarce, we provided functional evidence that independent loss of TRIM17 in NMRs and BMRs might be beneficial for neuronal survival under hypoxia, supporting the positive role of eliminating TRIM17 function in hypoxia adaptation. Our results also suggested that pseudogenes, together with positively selected genes, reinforced subterranean adaptations cooperatively. Conclusions Our study provides new insights into the molecular underpinnings of subterranean adaptations and highlights the importance of gene losses in mammalian evolution. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01243-0.
Collapse
Affiliation(s)
- Zhizhong Zheng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China
| | - Rong Hua
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China.,Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Guoqiang Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.,Joint Laboratory of Animal Models for Human Diseases and Drug Development, Soochow University and Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Hui Yang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China.
| | - Peng Shi
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China. .,Joint Laboratory of Animal Models for Human Diseases and Drug Development, Soochow University and Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China. .,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China. .,School of Future Technology, University of Chinese Academy of Sciences, Beijing, 101408, China.
| |
Collapse
|
599
|
Maj C, Salvi E, Citterio L, Borisov O, Simonini M, Glorioso V, Barlassina C, Glorioso N, Thijs L, Kuznetsova T, Cappuccio FP, Zhang ZY, Staessen JA, Cusi D, Lanzani C, Manunta P. Dissecting the Polygenic Basis of Primary Hypertension: Identification of Key Pathway-Specific Components. Front Cardiovasc Med 2022; 9:814502. [PMID: 35252394 PMCID: PMC8888857 DOI: 10.3389/fcvm.2022.814502] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 01/19/2022] [Indexed: 12/11/2022] Open
Abstract
Introduction and Objectives Genome-wide association studies have identified a high number of genetic loci associated with hypertension suggesting the presence of an underlying polygenic architecture. In this study, we aimed to dissect the polygenic component of primary hypertension searching also for pathway-specific components. Methods The polygenic risk score (PRS) models, based on the UK biobank genetic signals for hypertension status, were obtained on a target Italian case/control cohort including 561 cases and 731 hyper-normal controls from HYPERGENES, and were then applied to an independent validation cohort composed by multi-countries European-based samples including 1,284 cases and 960 hyper-normal controls. Results The resulting genome-wide PRS was capable of stratifying the individuals for hypertension risk by comparing between individuals in the last PRS decile and the median decile: we observed an odds ratio (OR) of 3.62, CI = [2.01, 6.32] (P = 9.01E-07) and 3.22, 95% CI = [2.06, 5.10] (P = 6.47E-08) in the target and validation cohorts, respectively. The relatively high case/control ORs across PRS quantiles corroborates the presence of strong polygenic components which could be driven by an enrichment of risk alleles within the cases but also by potential enrichment of protective alleles in the old normotensive controls. Moreover, novel pathway-specific PRS revealed an enrichment of the polygenic signal attributable to specific biological pathways. Among those the most significantly associated with hypertension status was the calcium signaling pathway together with other mainly related such as the phosphatidylinositol/inositol phosphate pathways. Conclusions The development of pathway-specific PRS could prioritize biological mechanisms, according to their contribution to the genetic susceptibility, whose regulations might be a potential pharmacological preventive target.
Collapse
Affiliation(s)
- Carlo Maj
- Institute for Genomic Statistics and Bioinformatics, Medical Faculty, University of Bonn, Bonn, Germany
- *Correspondence: Carlo Maj
| | - Erika Salvi
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico “Carlo Besta”, Milan, Italy
| | - Lorena Citterio
- Genomics of Renal Diseases and Hypertension Unit, Istituto di Ricovero e Cura a Carattere Scientifico IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Oleg Borisov
- Institute for Genomic Statistics and Bioinformatics, Medical Faculty, University of Bonn, Bonn, Germany
| | - Marco Simonini
- Genomics of Renal Diseases and Hypertension Unit, Istituto di Ricovero e Cura a Carattere Scientifico IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Valeria Glorioso
- Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milan, Italy
| | | | - Nicola Glorioso
- Department of Clinical and Experimental Medicine, Hypertension and Related Diseases Centre, University of Sassari, Sassari, Italy
| | - Lutgarde Thijs
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Tatiana Kuznetsova
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Francesco P. Cappuccio
- Warwick Medical School, and UHCW NHS Trust, University of Warwick, Coventry, United Kingdom
| | - Zhen-Yu Zhang
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Jan A. Staessen
- Research Institute Alliance for the Promotion of Preventive Medicine (APPREMED), Mechelen, Belgium
- Biomedical Science Group, Faculty of Medicine, University of Leuven, Leuven, Belgium
| | - Daniele Cusi
- Institute of Biomedical Technologies Milano National Research Council of Italy (CNR), Milano, Italy
- Bio4Dreams Scientific Unit, Bio4Dreams-Business Nursery for Life Sciences, Milano, Italy
| | - Chiara Lanzani
- Genomics of Renal Diseases and Hypertension Unit, Istituto di Ricovero e Cura a Carattere Scientifico IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Paolo Manunta
- Genomics of Renal Diseases and Hypertension Unit, Istituto di Ricovero e Cura a Carattere Scientifico IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
600
|
Xia K, Zhang L, Tang L, Huang T, Fan D. Assessing the role of blood pressure in amyotrophic lateral sclerosis: a Mendelian randomization study. Orphanet J Rare Dis 2022; 17:56. [PMID: 35172853 PMCID: PMC8848798 DOI: 10.1186/s13023-022-02212-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 02/06/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Observational studies have suggested a close but controversial relationship between blood pressure (BP) and amyotrophic lateral sclerosis (ALS). It remains unclear whether this association is causal. The authors employed a bidirectional two-sample Mendelian randomization (MR) approach to evaluate the causal relationship between BP and ALS. Genetic proxies for systolic blood pressure (SBP), diastolic blood pressure (DBP), antihypertensive drugs (AHDs), ALS, and their corresponding genome-wide association study (GWAS) summary datasets were obtained from the most recent studies with the largest sample sizes. The inverse variance weighted (IVW) method was adopted as the main approach to examine the effect of BP on ALS and four other MR methods were used for sensitivity analyses. To exclude the interference between SBP and DBP, a multivariable MR approach was used. RESULTS We found that genetically determined increased DBP was a protective factor for ALS (OR = 0.978, 95% CI 0.960-0.996, P = 0.017) and that increased SBP was an independent risk factor for ALS (OR = 1.014, 95% CI 1.003-1.025, P = 0.015), which is supported by sensitivity analyses. The use of calcium channel blocker (CCB) showed a causal relationship with ALS (OR = 0.985, 95% CI 0.971-1.000, P = 0.049). No evidence was revealed that ALS caused changes in BP. CONCLUSIONS This study provides genetic support for a causal effect of BP and ALS that increased DBP has a protective effect on ALS, and increased SBP is a risk factor for ALS, which may be related to sympathetic excitability. Blood pressure management is essential in ALS, and CCB may be a promising candidate.
Collapse
Affiliation(s)
- Kailin Xia
- Department of Neurology, Peking University Third Hospital, Garden North Road No. 49, Beijing, 100191, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Linjing Zhang
- Department of Neurology, Peking University Third Hospital, Garden North Road No. 49, Beijing, 100191, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Lu Tang
- Department of Neurology, Peking University Third Hospital, Garden North Road No. 49, Beijing, 100191, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Tao Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China. .,Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, Beijing, China.
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Garden North Road No. 49, Beijing, 100191, China. .,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China. .,Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing, China.
| |
Collapse
|