101
|
Kurniansyah N, Goodman MO, Khan AT, Wang J, Feofanova E, Bis JC, Wiggins KL, Huffman JE, Kelly T, Elfassy T, Guo X, Palmas W, Lin HJ, Hwang SJ, Gao Y, Young K, Kinney GL, Smith JA, Yu B, Liu S, Wassertheil-Smoller S, Manson JE, Zhu X, Chen YDI, Lee IT, Gu CC, Lloyd-Jones DM, Zöllner S, Fornage M, Kooperberg C, Correa A, Psaty BM, Arnett DK, Isasi CR, Rich SS, Kaplan RC, Redline S, Mitchell BD, Franceschini N, Levy D, Rotter JI, Morrison AC, Sofer T. Evaluating the use of blood pressure polygenic risk scores across race/ethnic background groups. Nat Commun 2023; 14:3202. [PMID: 37268629 PMCID: PMC10238525 DOI: 10.1038/s41467-023-38990-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 05/24/2023] [Indexed: 06/04/2023] Open
Abstract
We assess performance and limitations of polygenic risk scores (PRSs) for multiple blood pressure (BP) phenotypes in diverse population groups. We compare "clumping-and-thresholding" (PRSice2) and LD-based (LDPred2) methods to construct PRSs from each of multiple GWAS, as well as multi-PRS approaches that sum PRSs with and without weights, including PRS-CSx. We use datasets from the MGB Biobank, TOPMed study, UK biobank, and from All of Us to train, assess, and validate PRSs in groups defined by self-reported race/ethnic background (Asian, Black, Hispanic/Latino, and White). For both SBP and DBP, the PRS-CSx based PRS, constructed as a weighted sum of PRSs developed from multiple independent GWAS, perform best across all race/ethnic backgrounds. Stratified analysis in All of Us shows that PRSs are better predictive of BP in females compared to males, individuals without obesity, and middle-aged (40-60 years) compared to older and younger individuals.
Collapse
Affiliation(s)
- Nuzulul Kurniansyah
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Matthew O Goodman
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Alyna T Khan
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Jiongming Wang
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Elena Feofanova
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Kerri L Wiggins
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jennifer E Huffman
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
| | - Tanika Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Tali Elfassy
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Xiuqing Guo
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Walter Palmas
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Henry J Lin
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Shih-Jen Hwang
- The Population Sciences Branch of the National Heart, Lung and Blood Institute, Bethesda, MD, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - Yan Gao
- The Jackson Heart Study, University of Mississippi Medical Center, Jackson, MS, USA
| | - Kendra Young
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
| | - Gregory L Kinney
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
| | - Jennifer A Smith
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Bing Yu
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Simin Liu
- Center for Global Cardiometabolic Health, Department of Epidemiology, Brown University, Providence, RI, USA
- Department of Medicine, Brown University, Providence, RI, USA
| | - Sylvia Wassertheil-Smoller
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - JoAnn E Manson
- Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Xiaofeng Zhu
- Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Yii-Der Ida Chen
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - I-Te Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung City, 40705, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - C Charles Gu
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Sebastian Zöllner
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Myriam Fornage
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Adolfo Correa
- Departments of Medicine and Pediatrics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Bruce M Psaty
- Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
- Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Donna K Arnett
- Office of the Provost, University of South Carolina, Columbia, SC, USA
| | - Carmen R Isasi
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Robert C Kaplan
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Braxton D Mitchell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Daniel Levy
- The Population Sciences Branch of the National Heart, Lung and Blood Institute, Bethesda, MD, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - Jerome I Rotter
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|
102
|
Phulka JS, Ashraf M, Bajwa BK, Pare G, Laksman Z. Current State and Future of Polygenic Risk Scores in Cardiometabolic Disease: A Scoping Review. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:286-313. [PMID: 37035923 DOI: 10.1161/circgen.122.003834] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
A polygenic risk score (PRS) is derived from a genome-wide association study and represents an aggregate of thousands of single-nucleotide polymorphisms that provide a baseline estimate of an individual's genetic risk for a specific disease or trait at birth. However, it remains unclear how PRSs can be used in clinical practice. We provide an overview of the PRSs related to cardiometabolic disease and discuss the evidence supporting their clinical applications and limitations. The Preferred Reporting Items For Systematic Reviews and Meta-Analysis Extension for Scoping Reviews protocol was used to conduct a scoping review of the MEDLINE, EMBASE, and CENTRAL databases. Across the 4863 studies screened, 82 articles met the inclusion criteria. The most common PRS related to coronary artery disease, followed by hypertension and cerebrovascular disease. Limited ancestral diversity was observed in the study sample populations. Most studies included only individuals of European ancestry. The predictive performance of most PRSs was similar to or superior to traditional risk factors. More than half of the included studies reported an integrated risk model combining a derived PRS and clinical risk tools such as the Framingham Risk Score and Pooled Cohort Equations. The inclusion of a PRS into a clinical risk model tended to improve predictive accuracy consistently. This scoping review is the first of its kind and reports strong evidence for the clinical utility of PRSs in coronary artery disease, hypertension, cerebrovascular disease, and atrial fibrillation. However, most PRSs are generated in cohorts of European ancestry, which likely contributes to a lack of PRS transferability across different ancestral groups. Future prospective studies should focus on further establishing the clinical utility of PRSs and ensuring diversity is incorporated into genome-wide association study cohorts.
Collapse
Affiliation(s)
- Jobanjit S Phulka
- Heart Rhythm Services & Center for Cardiovascular Innovation, Division of Cardiology, University of British Columbia, Vancouver (J.S.P., M.A., B.K.B., Z.L.)
| | - Mishal Ashraf
- Heart Rhythm Services & Center for Cardiovascular Innovation, Division of Cardiology, University of British Columbia, Vancouver (J.S.P., M.A., B.K.B., Z.L.)
| | - Beenu K Bajwa
- Heart Rhythm Services & Center for Cardiovascular Innovation, Division of Cardiology, University of British Columbia, Vancouver (J.S.P., M.A., B.K.B., Z.L.)
| | - Guillaume Pare
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute; Thrombosis and Atherosclerosis Research Institute, Department of Health Research Methods, Evidence, and Impact, Department of Pathology & Molecular Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada (G.P.)
| | - Zachary Laksman
- Heart Rhythm Services & Center for Cardiovascular Innovation, Division of Cardiology, University of British Columbia, Vancouver (J.S.P., M.A., B.K.B., Z.L.)
| |
Collapse
|
103
|
Pérez-Gimeno G, Seral-Cortes M, Sabroso-Lasa S, Esteban LM, Lurbe E, Béghin L, Gottrand F, Meirhaeghe A, Muntaner M, Kafatos A, Molnár D, Leclercq C, Widhalm K, Kersting M, Nova E, Salazar-Tortosa DF, Gonzalez-Gross M, Breidenassel C, Sinningen K, De Ruyter T, Labayen I, Rupérez AI, Bueno-Lozano G, Moreno LA. Development of a genetic risk score to predict the risk of hypertension in European adolescents from the HELENA study. Front Cardiovasc Med 2023; 10:1118919. [PMID: 37324619 PMCID: PMC10267871 DOI: 10.3389/fcvm.2023.1118919] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 05/11/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction From genome wide association study (GWAS) a large number of single nucleotide polymorphisms (SNPs) have previously been associated with blood pressure (BP) levels. A combination of SNPs, forming a genetic risk score (GRS) could be considered as a useful genetic tool to identify individuals at risk of developing hypertension from early stages in life. Therefore, the aim of our study was to build a GRS being able to predict the genetic predisposition to hypertension (HTN) in European adolescents. Methods Data were extracted from the Healthy Lifestyle in Europe by Nutrition in Adolescence (HELENA) cross-sectional study. A total of 869 adolescents (53% female), aged 12.5-17.5, with complete genetic and BP information were included. The sample was divided into altered (≥130 mmHg for systolic and/or ≥80 mmHg for diastolic) or normal BP. Based on the literature, a total of 1.534 SNPs from 57 candidate genes related with BP were selected from the HELENA GWAS database. Results From 1,534 SNPs available, An initial screening of SNPs univariately associated with HTN (p < 0.10) was established, to finally obtain a number of 16 SNPs significantly associated with HTN (p < 0.05) in the multivariate model. The unweighted GRS (uGRS) and weighted GRS (wGRS) were estimated. To validate the GRSs, the area under the curve (AUC) was explored using ten-fold internal cross-validation for uGRS (0.802) and wGRS (0.777). Further covariates of interest were added to the analyses, obtaining a higher predictive ability (AUC values of uGRS: 0.879; wGRS: 0.881 for BMI z-score). Furthermore, the differences between AUCs obtained with and without the addition of covariates were statistically significant (p < 0.05). Conclusions Both GRSs, the uGRS and wGRS, could be useful to evaluate the predisposition to hypertension in European adolescents.
Collapse
Affiliation(s)
- Gloria Pérez-Gimeno
- Growth, Exercise, NUtrition and Development (GENUD), Research Group, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria Aragón (IIS Aragón), Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
| | - Miguel Seral-Cortes
- Growth, Exercise, NUtrition and Development (GENUD), Research Group, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria Aragón (IIS Aragón), Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
| | - Sergio Sabroso-Lasa
- Genetic and Molecular Epidemiology Group (GMEG), Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - Empar Lurbe
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- INCLIVA Biomedical Research Institute, Pediatric Department, Consorcio Hospital General, University of Valencia, Valencia, Spain
| | - Laurent Béghin
- Université Lille, Inserm, CHU Lille, INFINITE—Institute for Translational Research in Inflammation, Lille, France
| | - Frederic Gottrand
- Université Lille, Inserm, CHU Lille, INFINITE—Institute for Translational Research in Inflammation, Lille, France
| | - Aline Meirhaeghe
- Risk Factors and Molecular Determinants of Aging-Related Diseases (RID-AGE), Centre Hosp. Univ Lille, Institut Pasteur de Lille, Université de Lille, Lille, France
| | - Manon Muntaner
- Risk Factors and Molecular Determinants of Aging-Related Diseases (RID-AGE), Centre Hosp. Univ Lille, Institut Pasteur de Lille, Université de Lille, Lille, France
| | - Anthony Kafatos
- Department of Social Medicine, Preventive Medicine and Nutrition Clinic, University of Crete School of Medicine, Heraklion, Greece
| | - Dénes Molnár
- Department of Pediatrics, University of Pecs, Pecs, Hungary
| | - Catherine Leclercq
- INRAN, National Research Institute for Food and Nutrition, Food and Nutrition Research Centre-Council for Agricultural Research and Economics, Rome, Italy
| | - Kurt Widhalm
- Division of Clinical Nutrition and Prevention, Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Mathilde Kersting
- Departement of Nutrition—Human Nutrition, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Esther Nova
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN), CSIC, Madrid, Spain
| | - Diego F. Salazar-Tortosa
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ, United States
- PROFITH ‘PROmoting FITness and Health Through Physical Activity’ Research Group, Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain
| | - Marcela Gonzalez-Gross
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- ImFine Research Group, Department of Health and Human Performance, Facultad de Ciencias de la Actividad Física y del Deporte-INEF, Universidad Politécnica de Madrid, Madrid, Spain
| | - Christina Breidenassel
- Departement of Nutrition—Human Nutrition, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
- ImFine Research Group, Department of Health and Human Performance, Facultad de Ciencias de la Actividad Física y del Deporte-INEF, Universidad Politécnica de Madrid, Madrid, Spain
| | - Kathrin Sinningen
- Research Department of Child Nutrition, University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Thaïs De Ruyter
- Department of Public Health and Primary Care, Ghent University, Ghent, Belgium
| | - Idoia Labayen
- Department of Health Sciences, Institute for Innovation & Sustainable Food Chain Development, Public University of Navarra, Pamplona, Spain
| | - Azahara I. Rupérez
- Growth, Exercise, NUtrition and Development (GENUD), Research Group, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria Aragón (IIS Aragón), Universidad de Zaragoza, Zaragoza, Spain
| | - Gloria Bueno-Lozano
- Growth, Exercise, NUtrition and Development (GENUD), Research Group, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria Aragón (IIS Aragón), Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
| | - Luis A. Moreno
- Growth, Exercise, NUtrition and Development (GENUD), Research Group, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria Aragón (IIS Aragón), Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
104
|
Maitusong B, Laguzzi F, Strawbridge RJ, Baldassarre D, Veglia F, Humphries SE, Savonen K, Kurl S, Pirro M, Smit AJ, Giral P, Silveira A, Tremoli E, Hamsten A, de Faire U, Gigante B, Leander K. Cross-Sectional Gene-Smoking Interaction Analysis in Relation to Subclinical Atherosclerosis-Results From the IMPROVE Study. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:236-247. [PMID: 37021583 PMCID: PMC10284137 DOI: 10.1161/circgen.122.003710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/29/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Smoking is associated with carotid intima-media thickness (C-IMT). However, knowledge about how genetics may influence this association is limited. We aimed to perform nonhypothesis driven gene-smoking interaction analyses to identify potential genetic variants, among those included in immune and metabolic platforms, that may modify the effect of smoking on carotid intima-media thickness. METHODS We used baseline data from 1551 men and 1700 women, aged 55 to 79, included in a European multi-center study. Carotid intima-media thickness maximum, the maximum of values measured at different locations of the carotid tree, was dichotomized with cut point values ≥75, respectively. Genetic data were retrieved through use of the Illumina Cardio-Metabo- and Immuno- Chips. Gene-smoking interactions were evaluated through calculations of Synergy index (S). After adjustments for multiple testing, P values of <2.4×10-7 for S were considered significant. The models were adjusted for age, sex, education, physical activity, type of diet, and population stratification. RESULTS Our screening of 207 586 SNPs available for analysis, resulted in the identification of 47 significant gene-smoking synergistic interactions in relation to carotid intima-media thickness maximum. Among the significant SNPs, 28 were in protein coding genes, 2 in noncoding RNA and the remaining 17 in intergenic regions. CONCLUSIONS Through nonhypothesis-driven analyses of gene-smoking interactions, several significant results were observed. These may stimulate further research on the role of specific genes in the process that determines the effect of smoking habits on the development of carotid atherosclerosis.
Collapse
Affiliation(s)
- Buamina Maitusong
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China (B.M.)
| | - Federica Laguzzi
- Unit of Cardiovascular & Nutritional Epidemiology, Institute of Environmental Medicine (F.L., U.d.F., K.L.), Karolinska Institutet, Stockholm, Sweden
| | - Rona J. Strawbridge
- Cardiovascular Medicine Unit, Department of Medicine Solna (R.J.S., B.G.), Karolinska Institutet, Stockholm, Sweden
- Mental Health & Wellbeing, Institute of Mental Health & Wellbeing, University of Glasgow (R.J.S.)
- Health Data Research, United Kingdom (R.J.S.)
| | - Damiano Baldassarre
- Department of Medical Biotechnology & Translational Medicine, Università degli Studi di Milano (D.B.)
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.)
| | - Fabrizio Veglia
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.)
| | - Steve E. Humphries
- Cardiovascular Genetics, Institute Cardiovascular Science, University College London, United Kingdom (S.E.H.)
| | - Kai Savonen
- Foundation for Research in Health Exercise & Nutrition, Kuopio & Research Institute of Exercise Medicine, Kuopio, Finland (K.S.)
- Department of Clinical Physiology & Nuclear Medicine, Kuopio University Hospital (K.S.)
| | - Sudhir Kurl
- Institute of Public Health & Clinical Nutrition, University of Eastern Finland, Kuopio (S.K.)
| | - Matteo Pirro
- Unit of Internal Medicine, Angiology & Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Italy (M.P.)
| | - Andries J. Smit
- Department of Medicine, University Medical Center Groningen, the Netherlands (A.J.S.)
| | - Philippe Giral
- Unités de Prévention Cardiovasculaire, Assistance Publique-Hôpitaux de Paris, Service Endocrinologie-Métabolisme, Groupe Hospitalier Pitié-Salpétrière, France (P.G.)
| | - Angela Silveira
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet & Karolinska Hospital, Stockholm, Sweden (A.S., A.H.)
| | - Elena Tremoli
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.)
| | - Anders Hamsten
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet & Karolinska Hospital, Stockholm, Sweden (A.S., A.H.)
| | - Ulf de Faire
- Unit of Cardiovascular & Nutritional Epidemiology, Institute of Environmental Medicine (F.L., U.d.F., K.L.), Karolinska Institutet, Stockholm, Sweden
| | - Bruna Gigante
- Cardiovascular Medicine Unit, Department of Medicine Solna (R.J.S., B.G.), Karolinska Institutet, Stockholm, Sweden
| | - Karin Leander
- Unit of Cardiovascular & Nutritional Epidemiology, Institute of Environmental Medicine (F.L., U.d.F., K.L.), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
105
|
De Beer D, Mels CMC, Schutte AE, Delles C, Mary S, Mullen W, Latosinska A, Mischak H, Kruger R. Identifying a urinary peptidomics profile for hypertension in young adults: The African-PREDICT study: Urinary peptidomics and hypertension: Urinary peptidomics and hypertension. Proteomics 2023; 23:e2200444. [PMID: 36943111 DOI: 10.1002/pmic.202200444] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/27/2023] [Accepted: 03/02/2023] [Indexed: 03/23/2023]
Abstract
Hypertension is one of the most important and complex risk factors for cardiovascular diseases (CVDs). By using urinary peptidomics analyses, we aimed to identify peptides associated with hypertension, building a framework for future research towards improved prediction and prevention of premature development of CVD. We included 78 hypertensive and 79 normotensive participants from the African-PREDICT study (aged 20-30 years), matched for sex (51% male) and ethnicity (49% black and 51% white). Urinary peptidomics data were acquired using capillary-electrophoresis-time-of-flight-mass-spectrometry. Hypertension-associated peptides were identified and combined into a support vector machine-based multidimensional classifier. When comparing the peptide data between the normotensive and hypertensive groups, 129 peptides were nominally differentially abundant (Wilcoxon p < 0.05). Nonetheless, only three peptides, all derived from collagen alpha-1(III), remained significantly different after rigorous adjustments for multiple comparisons. The 37 most significant peptides (all p ≤ 0.001) served as basis for the development of a classifier, with 20 peptides being combined into a unifying score, resulting in an AUC of 0.85 in the ROC analysis (p < 0.001), with 83% sensitivity at 80% specificity. Our study suggests potential value of urinary peptides in the classification of hypertension, which could enable earlier diagnosis and better understanding of the pathophysiology of hypertension and premature cardiovascular disease development.
Collapse
Affiliation(s)
- Dalene De Beer
- Hypertension in Africa Research Team (HART), North-West University (Potchefstroom Campus), Potchefstroom, South Africa
| | - Catharina M C Mels
- Hypertension in Africa Research Team (HART), North-West University (Potchefstroom Campus), Potchefstroom, South Africa
- MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Aletta E Schutte
- Hypertension in Africa Research Team (HART), North-West University (Potchefstroom Campus), Potchefstroom, South Africa
- MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
- School of Population Health, The George Institute for Global Health, University of New South Wales, Sydney, Australia
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Sheon Mary
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - William Mullen
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | | | | | - Ruan Kruger
- Hypertension in Africa Research Team (HART), North-West University (Potchefstroom Campus), Potchefstroom, South Africa
- MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| |
Collapse
|
106
|
Aryal S, Manandhar I, Mei X, Yeoh BS, Tummala R, Saha P, Osman I, Zubcevic J, Durgan DJ, Vijay-Kumar M, Joe B. Combating hypertension beyond genome-wide association studies: Microbiome and artificial intelligence as opportunities for precision medicine. CAMBRIDGE PRISMS. PRECISION MEDICINE 2023; 1:e26. [PMID: 38550938 PMCID: PMC10953772 DOI: 10.1017/pcm.2023.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 03/15/2023] [Accepted: 04/04/2023] [Indexed: 11/03/2024]
Abstract
The single largest contributor to human mortality is cardiovascular disease, the top risk factor for which is hypertension (HTN). The last two decades have placed much emphasis on the identification of genetic factors contributing to HTN. As a result, over 1,500 genetic alleles have been associated with human HTN. Mapping studies using genetic models of HTN have yielded hundreds of blood pressure (BP) loci but their individual effects on BP are minor, which limits opportunities to target them in the clinic. The value of collecting genome-wide association data is evident in ongoing research, which is beginning to utilize these data at individual-level genetic disparities combined with artificial intelligence (AI) strategies to develop a polygenic risk score (PRS) for the prediction of HTN. However, PRS alone may or may not be sufficient to account for the incidence and progression of HTN because genetics is responsible for <30% of the risk factors influencing the etiology of HTN pathogenesis. Therefore, integrating data from other nongenetic factors influencing BP regulation will be important to enhance the power of PRS. One such factor is the composition of gut microbiota, which constitute a more recently discovered important contributor to HTN. Studies to-date have clearly demonstrated that the transition from normal BP homeostasis to a state of elevated BP is linked to compositional changes in gut microbiota and its interaction with the host. Here, we first document evidence from studies on gut dysbiosis in animal models and patients with HTN followed by a discussion on the prospects of using microbiota data to develop a metagenomic risk score (MRS) for HTN to be combined with PRS and a clinical risk score (CRS). Finally, we propose that integrating AI to learn from the combined PRS, MRS and CRS may further enhance predictive power for the susceptibility and progression of HTN.
Collapse
Affiliation(s)
- Sachin Aryal
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Ishan Manandhar
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Xue Mei
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Beng S. Yeoh
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Ramakumar Tummala
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Piu Saha
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Islam Osman
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Jasenka Zubcevic
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - David J. Durgan
- Integrative Physiology & Anesthesiology, Baylor College of Medicine, Houston, TX, USA
| | - Matam Vijay-Kumar
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Bina Joe
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| |
Collapse
|
107
|
Ivanova T, Churnosova M, Abramova M, Ponomarenko I, Reshetnikov E, Aristova I, Sorokina I, Churnosov M. Risk Effects of rs1799945 Polymorphism of the HFE Gene and Intergenic Interactions of GWAS-Significant Loci for Arterial Hypertension in the Caucasian Population of Central Russia. Int J Mol Sci 2023; 24:ijms24098309. [PMID: 37176017 PMCID: PMC10179076 DOI: 10.3390/ijms24098309] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
The aim of this case-control replicative study was to investigate the link between GWAS-impact for arterial hypertension (AH) and/or blood pressure (BP) gene polymorphisms and AH risk in Russian subjects (Caucasian population of Central Russia). AH (n = 939) and control (n = 466) cohorts were examined for ten GWAS AH/BP risk loci. The genotypes/alleles of these SNP and their combinations (SNP-SNP interactions) were tested for their association with the AH development using a logistic regression statistical procedure. The genotype GG of the SNP rs1799945 (C/G) HFE was strongly linked with an increased AH risk (ORrecGG = 2.53; 95%CIrecGG1.03-6.23; ppermGG = 0.045). The seven SNPs such as rs1173771 (G/A) AC026703.1, rs1799945 (C/G) HFE, rs805303 (G/A) BAG6, rs932764 (A/G) PLCE1, rs4387287 (C/A) OBFC1, rs7302981 (G/A) CERS5, rs167479 (T/G) RGL3, out of ten regarded loci, were related with AH within eight SNP-SNP interaction models (<0.001 ≤ pperm-interaction ≤ 0.047). Three polymorphisms such as rs8068318 (T/C) TBX2, rs633185 (C/G) ARHGAP42, and rs2681472 (A/G) ATP2B1 were not linked with AH. The pairwise rs805303 (G/A) BAG6-rs7302981 (G/A) CERS5 combination was a priority in determining the susceptibility to AH (included in six out of eight SNP-SNP interaction models [75%] and described 0.82% AH entropy). AH-associated variants are conjecturally functional for 101 genes involved in processes related to the immune system (major histocompatibility complex protein, processing/presentation of antigens, immune system process regulation, etc.). In conclusion, the rs1799945 polymorphism of the HFE gene and intergenic interactions of BAG6, CERS5, AC026703.1, HFE, PLCE1, OBFC1, RGL3 have been linked with AH risky in the Caucasian population of Central Russia.
Collapse
Affiliation(s)
- Tatiana Ivanova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Maria Churnosova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Maria Abramova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Irina Ponomarenko
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Evgeny Reshetnikov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Inna Aristova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Inna Sorokina
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Mikhail Churnosov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| |
Collapse
|
108
|
Ivanova T, Churnosova M, Abramova M, Plotnikov D, Ponomarenko I, Reshetnikov E, Aristova I, Sorokina I, Churnosov M. Sex-Specific Features of the Correlation between GWAS-Noticeable Polymorphisms and Hypertension in Europeans of Russia. Int J Mol Sci 2023; 24:ijms24097799. [PMID: 37175507 PMCID: PMC10178435 DOI: 10.3390/ijms24097799] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/13/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
The aim of the study was directed at studying the sex-specific features of the correlation between genome-wide association studies (GWAS)-noticeable polymorphisms and hypertension (HTN). In two groups of European subjects of Russia (n = 1405 in total), such as men (n = 821 in total: n = 564 HTN, n = 257 control) and women (n = 584 in total: n = 375 HTN, n = 209 control), the distribution of ten specially selected polymorphisms (they have confirmed associations of GWAS level with blood pressure (BP) parameters and/or HTN in Europeans) has been considered. The list of studied loci was as follows: (PLCE1) rs932764 A > G, (AC026703.1) rs1173771 G > A, (CERS5) rs7302981 G > A, (HFE) rs1799945 C > G, (OBFC1) rs4387287 C > A, (BAG6) rs805303 G > A, (RGL3) rs167479 T > G, (ARHGAP42) rs633185 C > G, (TBX2) rs8068318 T > C, and (ATP2B1) rs2681472 A > G. The contribution of individual loci and their inter-locus interactions to the HTN susceptibility with bioinformatic interpretation of associative links was evaluated separately in men's and women's cohorts. The men-women differences in involvement in the disease of the BP/HTN-associated GWAS SNPs were detected. Among women, the HTN risk has been associated with HFE rs1799945 C > G (genotype GG was risky; ORGG = 11.15 ppermGG = 0.014) and inter-locus interactions of all 10 examined SNPs as part of 26 intergenic interactions models. In men, the polymorphism BAG6 rs805303 G > A (genotype AA was protective; ORAA = 0.30 ppermAA = 0.0008) and inter-SNPs interactions of eight loci in only seven models have been founded as HTN-correlated. HTN-linked loci and strongly linked SNPs were characterized by pronounced polyvector functionality in both men and women, but at the same time, signaling pathways of HTN-linked genes/SNPs in women and men were similar and were represented mainly by immune mechanisms. As a result, the present study has demonstrated a more pronounced contribution of BP/HTN-associated GWAS SNPs to the HTN susceptibility (due to weightier intergenic interactions) in European women than in men.
Collapse
Affiliation(s)
- Tatiana Ivanova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Maria Churnosova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Maria Abramova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Denis Plotnikov
- Genetic Epidemiology Lab, Kazan State Medical University, 420012 Kazan, Russia
| | - Irina Ponomarenko
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Evgeny Reshetnikov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Inna Aristova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Inna Sorokina
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Mikhail Churnosov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| |
Collapse
|
109
|
Sallis HM, Palmer T, Tilling K, Davey Smith G, Munafò MR. Using allele scores to identify confounding by reverse causation: studies of alcohol consumption as an exemplar. Int J Epidemiol 2023; 52:536-544. [PMID: 35980022 PMCID: PMC10114122 DOI: 10.1093/ije/dyac165] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 08/04/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Mendelian randomization (MR) is a form of instrumental variable analysis used to investigate causality using observational data. Another important, although less frequently applied, use of this technique is to investigate confounding due to reverse causality. METHODS We used a form of reverse MR and data from UK Biobank in a proof-of-principle study to investigate confounding due to reverse causation. Here we focus on the association between alcohol consumption (exposure) and outcomes including educational attainment, and physical and mental health. First, we examined the observational relationship between alcohol consumption and these outcomes. Allele scores were then derived for educational attainment, and physical and mental health, and the association with alcohol consumption (as the outcome) was explored. Sample sizes ranged from 114 941-336 473 in observational analyses and 142 093-336 818 in genetic analyses. RESULTS Conventional observational analyses indicated associations between alcohol consumption and a number of outcomes (e.g. neuroticism, body mass index, educational attainment). Analyses using allele scores suggested evidence of reverse causation for several of these relationships (in particular physical health and educational attainment). CONCLUSION Allele scores allow us to investigate reverse causation in observational studies. Our findings suggest that observed associations implying beneficial effects of alcohol consumption may be due to confounding by reverse causation in many cases.
Collapse
Affiliation(s)
- Hannah M Sallis
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- School of Psychological Science, University of Bristol, Bristol, UK
| | - Tom Palmer
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Kate Tilling
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - George Davey Smith
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Marcus R Munafò
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- School of Psychological Science, University of Bristol, Bristol, UK
- NIHR Biomedical Research Centre, University Hospitals Bristol NHS Foundation Trust and University of Bristol, Bristol, UK
| |
Collapse
|
110
|
Pandey AK, Loscalzo J. Network medicine: an approach to complex kidney disease phenotypes. Nat Rev Nephrol 2023:10.1038/s41581-023-00705-0. [PMID: 37041415 DOI: 10.1038/s41581-023-00705-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2023] [Indexed: 04/13/2023]
Abstract
Scientific reductionism has been the basis of disease classification and understanding for more than a century. However, the reductionist approach of characterizing diseases from a limited set of clinical observations and laboratory evaluations has proven insufficient in the face of an exponential growth in data generated from transcriptomics, proteomics, metabolomics and deep phenotyping. A new systematic method is necessary to organize these datasets and build new definitions of what constitutes a disease that incorporates both biological and environmental factors to more precisely describe the ever-growing complexity of phenotypes and their underlying molecular determinants. Network medicine provides such a conceptual framework to bridge these vast quantities of data while providing an individualized understanding of disease. The modern application of network medicine principles is yielding new insights into the pathobiology of chronic kidney diseases and renovascular disorders by expanding the understanding of pathogenic mediators, novel biomarkers and new options for renal therapeutics. These efforts affirm network medicine as a robust paradigm for elucidating new advances in the diagnosis and treatment of kidney disorders.
Collapse
Affiliation(s)
- Arvind K Pandey
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
111
|
Cai L, Wang D, Gui T, Wang X, Zhao L, Boron WF, Chen LM, Liu Y. Dietary sodium enhances the expression of SLC4 family transporters, IRBIT, L-IRBIT, and PP1 in rat kidney: Insights into the molecular mechanism for renal sodium handling. Front Physiol 2023; 14:1154694. [PMID: 37082243 PMCID: PMC10111226 DOI: 10.3389/fphys.2023.1154694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/24/2023] [Indexed: 04/07/2023] Open
Abstract
The kidney plays a central role in maintaining the fluid and electrolyte homeostasis in the body. Bicarbonate transporters NBCn1, NBCn2, and AE2 are expressed at the basolateral membrane of the medullary thick ascending limb (mTAL). In a previous study, NBCn1, NBCn2, and AE2 are proposed to play as a regulatory pathway to decrease NaCl reabsorption in the mTAL under high salt condition. When heterologously expressed, the activity of these transporters could be stimulated by the InsP3R binding protein released with inositol 1,4,5-trisphosphate (IRBIT), L-IRBIT (collectively the IRBITs), or protein phosphatase PP1. In the present study, we characterized by immunofluorescence the expression and localization of the IRBITs, and PP1 in rat kidney. Our data showed that the IRBITs were predominantly expressed from the mTAL through the distal renal tubules. PP1 was predominantly expressed in the TAL, but is also present in high abundance from the distal convoluted tubule through the medullary collecting duct. Western blotting analyses showed that the abundances of NBCn1, NBCn2, and AE2 as well as the IRBITs and PP1 were greatly upregulated in rat kidney by dietary sodium. Co-immunoprecipitation study provided the evidence for protein interaction between NBCn1 and L-IRBIT in rat kidney. Taken together, our data suggest that the IRBITs and PP1 play an important role in sodium handling in the kidney. We propose that the IRBITs and PP1 stimulates NBCn1, NBCn2, and AE2 in the basolateral mTAL to inhibit sodium reabsorption under high sodium condition. Our study provides important insights into understanding the molecular mechanism for the regulation of sodium homeostasis in the body.
Collapse
Affiliation(s)
- Lu Cai
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dengke Wang
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Tianxiang Gui
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoyu Wang
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lingyu Zhao
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Walter F. Boron
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Li-Ming Chen
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
- *Correspondence: Li-Ming Chen, ; Ying Liu,
| | - Ying Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
- *Correspondence: Li-Ming Chen, ; Ying Liu,
| |
Collapse
|
112
|
Adua E. Decoding the mechanism of hypertension through multiomics profiling. J Hum Hypertens 2023; 37:253-264. [PMID: 36329155 PMCID: PMC10063442 DOI: 10.1038/s41371-022-00769-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 08/24/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Hypertension, characterised by a constant high blood pressure, is the primary risk factor for multiple cardiovascular events and a major cause of death in adults. Excitingly, innovations in high-throughput technologies have enabled the global exploration of the whole genome (genomics), revealing dysregulated genes that are linked to hypertension. Moreover, post-genomic biomarkers, from the emerging fields of transcriptomics, proteomics, glycomics and lipidomics, have provided new insights into the molecular underpinnings of hypertension. In this paper, we review the pathophysiology of hypertension, and highlight the multi-omics approaches for hypertension prediction and diagnosis.
Collapse
Affiliation(s)
- Eric Adua
- School of Clinical Medicine, Medicine & Health, Rural Clinical Campus, University of New South Wales, Wagga Wagga, NSW, Australia.
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.
| |
Collapse
|
113
|
Fehrenbach DJ, Nguyen B, Alexander MR, Madhur MS. Modulating T Cell Phenotype and Function to Treat Hypertension. KIDNEY360 2023; 4:e534-e543. [PMID: 36951464 PMCID: PMC10278787 DOI: 10.34067/kid.0000000000000090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 01/25/2023] [Indexed: 03/24/2023]
Abstract
Hypertension is the leading modifiable risk factor of worldwide morbidity and mortality because of its effects on cardiovascular and renal end-organ damage. Unfortunately, BP control is not sufficient to fully reduce the risks of hypertension, underscoring the need for novel therapies that address end-organ damage in hypertension. Over the past several decades, the link between immune activation and hypertension has been well established, but there are still no therapies for hypertension that specifically target the immune system. In this review, we describe the critical role played by T cells in hypertension and hypertensive end-organ damage and outline potential therapeutic targets to modulate T-cell phenotype and function in hypertension without causing global immunosuppression.
Collapse
Affiliation(s)
- Daniel J. Fehrenbach
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee
| | - Bianca Nguyen
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
| | - Matthew R. Alexander
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, Tennessee
| | - Meena S. Madhur
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, Tennessee
| |
Collapse
|
114
|
Sajal IH, Biswas S. Bivariate quantitative Bayesian LASSO for detecting association of rare haplotypes with two correlated continuous phenotypes. Front Genet 2023; 14:1104727. [PMID: 36968609 PMCID: PMC10033866 DOI: 10.3389/fgene.2023.1104727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/21/2023] [Indexed: 03/12/2023] Open
Abstract
In genetic association studies, the multivariate analysis of correlated phenotypes offers statistical and biological advantages compared to analyzing one phenotype at a time. The joint analysis utilizes additional information contained in the correlation and avoids multiple testing. It also provides an opportunity to investigate and understand shared genetic mechanisms of multiple phenotypes. Bivariate logistic Bayesian LASSO (LBL) was proposed earlier to detect rare haplotypes associated with two binary phenotypes or one binary and one continuous phenotype jointly. There is currently no haplotype association test available that can handle multiple continuous phenotypes. In this study, by employing the framework of bivariate LBL, we propose bivariate quantitative Bayesian LASSO (QBL) to detect rare haplotypes associated with two continuous phenotypes. Bivariate QBL removes unassociated haplotypes by regularizing the regression coefficients and utilizing a latent variable to model correlation between two phenotypes. We carry out extensive simulations to investigate the performance of bivariate QBL and compare it with that of a standard (univariate) haplotype association test, Haplo.score (applied twice to two phenotypes individually). Bivariate QBL performs better than Haplo.score in all simulations with varying degrees of power gain. We analyze Genetic Analysis Workshop 19 exome sequencing data on systolic and diastolic blood pressures and detect several rare haplotypes associated with the two phenotypes.
Collapse
Affiliation(s)
| | - Swati Biswas
- Department of Mathematical Sciences, University of Texas at Dallas, Richardson, TX, United States
| |
Collapse
|
115
|
Nurkkala J, Kauko A, FinnGen, Laivuori H, Saarela T, Tyrmi JS, Vaura F, Cheng S, Bello NA, Aittokallio J, Niiranen T. Associations of polygenic risk scores for preeclampsia and blood pressure with hypertensive disorders of pregnancy. J Hypertens 2023; 41:380-387. [PMID: 36947680 PMCID: PMC9894151 DOI: 10.1097/hjh.0000000000003336] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/01/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Preexisting hypertension increases risk for preeclampsia. We examined whether a generic blood pressure polygenic risk score (BP-PRS), compared with a preeclampsia-specific polygenic risk score (PE-PRS), could better predict hypertensive disorders of pregnancy. METHODS Our study sample included 141 298 genotyped FinnGen study participants with at least one childbirth and followed from 1969 to 2021. We calculated PRSs for SBP and preeclampsia using summary statistics for greater than 1.1 million single nucleotide polymorphisms. RESULTS We observed 8488 cases of gestational hypertension (GHT) and 6643 cases of preeclampsia. BP-PRS was associated with GHT [multivariable-adjusted hazard ratio for 1SD increase in PRS (hazard ratio 1.38; 95% CI 1.35-1.41)] and preeclampsia (1.26, 1.23-1.29), respectively. The PE-PRS was also associated with GHT (1.16; 1.14-1.19) and preeclampsia (1.21, 1.18-1.24), but with statistically more modest magnitudes of effect (P = 0.01). The model c-statistic for preeclampsia improved when PE-PRS was added to clinical risk factors (P = 4.6 × 10-15). Additional increment in the c-statistic was observed when BP-PRS was added to a model already including both clinical risk factors and PE-PRS (P = 1.1 × 10-14). CONCLUSION BP-PRS is strongly associated with hypertensive disorders of pregnancy. Our current observations suggest that the BP-PRS could capture the genetic architecture of preeclampsia better than the current PE-PRSs. These findings also emphasize the common pathways in the development of all BP disorders. The clinical utility of a BP-PRS for preeclampsia prediction warrants further investigation.
Collapse
Affiliation(s)
- Jouko Nurkkala
- Division of Perioperative Services, Intensive Care and Pain Medicine, Turku University Hospital
- Department of Anesthesiology and Intensive Care
| | - Anni Kauko
- Department of Internal Medicine, University of Turku, Turku
| | | | - Hannele Laivuori
- Department of Obstetrics and Gynecology, Tampere University Hospital
- Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science, University of Helsinki
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki
| | - Tanja Saarela
- Department of Clinical Genetics, Kuopio University Hospital, Kuopio
| | - Jaakko S. Tyrmi
- Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland
| | - Felix Vaura
- Department of Internal Medicine, University of Turku, Turku
| | - Susan Cheng
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Division of Cardiology Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Natalie A. Bello
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jenni Aittokallio
- Division of Perioperative Services, Intensive Care and Pain Medicine, Turku University Hospital
- Department of Anesthesiology and Intensive Care
| | - Teemu Niiranen
- Department of Internal Medicine, University of Turku, Turku
- Division of Medicine, Turku University Hospital
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Turku, Finland
| |
Collapse
|
116
|
Notch Signaling in Acute Inflammation and Sepsis. Int J Mol Sci 2023; 24:ijms24043458. [PMID: 36834869 PMCID: PMC9967996 DOI: 10.3390/ijms24043458] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/27/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
Notch signaling, a highly conserved pathway in mammals, is crucial for differentiation and homeostasis of immune cells. Besides, this pathway is also directly involved in the transmission of immune signals. Notch signaling per se does not have a clear pro- or anti-inflammatory effect, but rather its impact is highly dependent on the immune cell type and the cellular environment, modulating several inflammatory conditions including sepsis, and therefore significantly impacts the course of disease. In this review, we will discuss the contribution of Notch signaling on the clinical picture of systemic inflammatory diseases, especially sepsis. Specifically, we will review its role during immune cell development and its contribution to the modulation of organ-specific immune responses. Finally, we will evaluate to what extent manipulation of the Notch signaling pathway could be a future therapeutic strategy.
Collapse
|
117
|
Tawfik SM, Elhosseiny AA, Galal AA, William MB, Qansuwa E, Elbaz RM, Salama M. Health inequity in genomic personalized medicine in underrepresented populations: a look at the current evidence. Funct Integr Genomics 2023; 23:54. [PMID: 36719510 DOI: 10.1007/s10142-023-00979-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/24/2023] [Accepted: 01/24/2023] [Indexed: 02/01/2023]
Abstract
Improvements in sequencing technology coupled with dramatic declines in the cost of genome sequencing have led to a proportional growth in the size and number of genetic datasets since the release of the human genetic sequence by The Human Genome Project (HGP) international consortium. The HGP was undeniably a significant scientific success, a turning point in human genetics and the beginning of human genomics. This burst of genetic information has led to a greater understanding of disease pathology and the potential of employing this data to deliver more precise patient care. Hence, the recognition of high-penetrance disease-causing mutations which encode drivers of disease has made the management of most diseases more specific. Nonetheless, while genetic scores are becoming more extensively used, their application in the real world is expected to be limited due to the lack of diversity in the data used to construct them. Underrepresented populations, such as racial and ethnic minorities, low-income individuals, and those living in rural areas, often experience greater health disparities and worse health outcomes compared to the general population. These disparities are often the result of systemic barriers, such as poverty, discrimination, and limited access to healthcare. Addressing health inequity in underrepresented populations requires addressing the underlying social determinants of health and implementing policies and programs which promoted health equity and reduce disparities. This can include expanding access to affordable healthcare, addressing poverty and unemployment, and promoting policies that combat discrimination and racism.
Collapse
Affiliation(s)
- Sherouk M Tawfik
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, Cairo, 11835, Egypt.,Department of Pharmacology and Biochemistry, Faculty of Pharmacy, The British University in Egypt (BUE), Cairo, 11837, Egypt
| | - Aliaa A Elhosseiny
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, Cairo, 11835, Egypt.,Department of Pharmacology and Biochemistry, Faculty of Pharmacy, The British University in Egypt (BUE), Cairo, 11837, Egypt
| | - Aya A Galal
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, Cairo, 11835, Egypt.,Systems Genomics Laboratory, The American University in Cairo, New Cairo, Egypt
| | - Martina B William
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, Cairo, 11835, Egypt.,Department of Clinical Pharmacy, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Esraa Qansuwa
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, Cairo, 11835, Egypt
| | - Rana M Elbaz
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, Cairo, 11835, Egypt
| | - Mohamed Salama
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, Cairo, 11835, Egypt. .,Faculty of Medicine, Mansoura University, Mansoura, Egypt. .,Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
118
|
Zhao D, Sun H, Li H, Li C, Zhou B. A prediction model for the impact of environmental and genetic factors on cardiovascular events: development in a salt substitutes population. J Transl Med 2023; 21:62. [PMID: 36717874 PMCID: PMC9887817 DOI: 10.1186/s12967-023-03899-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/17/2023] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Cardiovascular disease (CVD) has evolved into a serious public health issue that demands the use of suitable methods to estimate the risk of the disease. As a result, in a sample of individuals who completed a 3-year low-sodium salt or conventional salt intervention in a hypertensive environment, we constructed a 13-year cardiovascular (CV) event risk prediction model with a 10-year follow-up. METHODS A Cox proportional hazards model was used to build a prediction model based on data from 306 participants who matched the inclusion criteria. Both the discriminating power and the calibration of the prediction models were assessed. The discriminative power of the prediction model was measured using the area under the curve (AUC). Brier scores and calibration plots were used to assess the prediction model's calibration. The model was internally validated using the tenfold cross-validation method. The nomogram served as a tool for visualising the model. RESULTS Among the 306 total individuals, there were 100 cases and 206 control. In the model, there were six predictors including age, smoking, LDL-C (low-density lipoprotein cholesterol), baseline SBP (systolic blood pressure), CVD (cardiovascular history), and CNV (genomic copy number variation) nsv483076. The fitted model has an AUC of 0.788, showing strong model discrimination, and a Brier score of 0.166, indicating that it was well-calibrated. According to the results of internal validation, the prediction model utilised in this study had a good level of repeatability. According to the model integrating the interaction of CNVs and baseline blood pressure, the effect of baseline SBP on CV events may be greater when nsv483076 was normal double copies than when nsv483076 was copy number variation. CONCLUSIONS The efficacy of risk prediction models for CV events that include environmental and genetic components is excellent, and they may be utilised as risk assessment tools for CV events in specific groups to offer a foundation for tailored intervention strategies.
Collapse
Affiliation(s)
- Dan Zhao
- grid.412636.40000 0004 1757 9485Department of Clinical Epidemiology and Evidence-Based Medicine, The First Hospital of China Medical University, No.155, Nanjing North Street, Heping District, Shenyang, Liaoning China ,grid.412449.e0000 0000 9678 1884School of Public Health, China Medical University, Shenyang, Liaoning China
| | - Hao Sun
- grid.412636.40000 0004 1757 9485Department of Clinical Epidemiology and Evidence-Based Medicine, The First Hospital of China Medical University, No.155, Nanjing North Street, Heping District, Shenyang, Liaoning China
| | - Huamin Li
- grid.412636.40000 0004 1757 9485Department of Clinical Epidemiology and Evidence-Based Medicine, The First Hospital of China Medical University, No.155, Nanjing North Street, Heping District, Shenyang, Liaoning China ,grid.412449.e0000 0000 9678 1884School of Public Health, China Medical University, Shenyang, Liaoning China
| | - Chaoxiu Li
- grid.412636.40000 0004 1757 9485Department of Clinical Epidemiology and Evidence-Based Medicine, The First Hospital of China Medical University, No.155, Nanjing North Street, Heping District, Shenyang, Liaoning China ,grid.412449.e0000 0000 9678 1884School of Public Health, China Medical University, Shenyang, Liaoning China
| | - Bo Zhou
- Department of Clinical Epidemiology and Evidence-Based Medicine, The First Hospital of China Medical University, No.155, Nanjing North Street, Heping District, Shenyang, Liaoning, China.
| |
Collapse
|
119
|
Udosen B, Soremekun O, Kamiza A, Machipisa T, Cheickna C, Omotuyi O, Soliman M, Wélé M, Nashiru O, Chikowore T, Fatumo S. Meta-Analysis and Multivariate GWAS Analyses in 80,950 Individuals of African Ancestry Identify Novel Variants Associated with Blood Pressure Traits. Int J Mol Sci 2023; 24:2164. [PMID: 36768488 PMCID: PMC9916484 DOI: 10.3390/ijms24032164] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/24/2022] [Accepted: 01/03/2023] [Indexed: 01/25/2023] Open
Abstract
High blood pressure (HBP) has been implicated as a major risk factor for cardiovascular diseases in several populations, including individuals of African ancestry. Despite the elevated burden of HBP-induced cardiovascular diseases in Africa and other populations of African descent, limited genetic studies have been carried out to explore the genetic mechanism driving this phenomenon. We performed genome-wide association univariate and multivariate analyses of both systolic (SBP) and diastolic blood pressure (DBP) traits in 80,950 individuals of African ancestry. We used summary statistics data from six independent cohorts, including the African Partnership for Chronic Disease Research (APCDR), the UK Biobank, and the Million Veteran Program (MVP). FUMA was used to annotate, prioritize, visualize, and interpret our findings to gain a better understanding of the molecular mechanism(s) underlying the genetics of BP traits. Finally, we undertook a Bayesian fine-mapping analysis to identify potential causal variants. Our meta-analysis identified 10 independent variants associated with SBP and 9 with DBP traits. Whilst our multivariate GWAS method identified 21 independent signals, 18 of these SNPs have been previously identified. SBP was linked to gene sets involved in biological processes such as synapse assembly and cell-cell adhesion via plasma membrane adhesion. Of the 19 independent SNPs identified in the BP meta-analysis, only 11 variants had posterior probability (PP) of > 50%, including one novel variant: rs562545 (MOBP, PP = 77%). To facilitate further research and fine-mapping of high-risk loci/variants in highly susceptible groups for cardiovascular disease and other related traits, large-scale genomic datasets are needed. Our findings highlight the importance of including ancestrally diverse populations in large GWASs and the need for diversity in genetic research.
Collapse
Affiliation(s)
- Brenda Udosen
- The African Computational Genomics (TACG) Research Group, MRC/UVRI and LSHTM, Entebbe 7545, Uganda; (B.U.); (O.S.); (A.K.)
- The African Center of Excellence in Bioinformatics of Bamako (ACE-B), University of Sciences, Techniques and Technologies of Bamako, Bamako 3206, Mali; (C.C.); (M.W.)
- H3Africa Bioinformatics Network (H3ABioNet) Node, Centre for Genomics Research and Innovation, NABDA/FMST, Abuja 901101, Nigeria;
| | - Opeyemi Soremekun
- The African Computational Genomics (TACG) Research Group, MRC/UVRI and LSHTM, Entebbe 7545, Uganda; (B.U.); (O.S.); (A.K.)
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, Westville Campus, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Abram Kamiza
- The African Computational Genomics (TACG) Research Group, MRC/UVRI and LSHTM, Entebbe 7545, Uganda; (B.U.); (O.S.); (A.K.)
- Malawi Epidemiology and Intervention Research Unit, Lilongwe P.O. Box 46, Malawi
| | - Tafadzwa Machipisa
- Hatter Institute for Cardiovascular Diseases Research in Africa (HICRA), Department of Medicine, University of Cape Town, Cape Town 7701, South Africa;
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, ON L8L 2X2, Canada
| | - Cisse Cheickna
- The African Center of Excellence in Bioinformatics of Bamako (ACE-B), University of Sciences, Techniques and Technologies of Bamako, Bamako 3206, Mali; (C.C.); (M.W.)
- Department of Biological Sciences, Faculty of Sciences and Techniques, University of Sciences, Techniques and Technologies of Bamako, Bamako 3206, Mali
| | - Olaposi Omotuyi
- Institute for Drug Research and Development, S.E. Bogoro Center, Afe Babalola University, Ado Ekiti 360101, Nigeria;
| | - Mahmoud Soliman
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, Westville Campus, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Mamadou Wélé
- The African Center of Excellence in Bioinformatics of Bamako (ACE-B), University of Sciences, Techniques and Technologies of Bamako, Bamako 3206, Mali; (C.C.); (M.W.)
- Department of Biological Sciences, Faculty of Sciences and Techniques, University of Sciences, Techniques and Technologies of Bamako, Bamako 3206, Mali
| | - Oyekanmi Nashiru
- H3Africa Bioinformatics Network (H3ABioNet) Node, Centre for Genomics Research and Innovation, NABDA/FMST, Abuja 901101, Nigeria;
| | - Tinashe Chikowore
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2050, South Africa;
- MRC/Wits Developmental Pathways for Health Research Unit, Department of Pediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2050, South Africa
| | - Segun Fatumo
- The African Computational Genomics (TACG) Research Group, MRC/UVRI and LSHTM, Entebbe 7545, Uganda; (B.U.); (O.S.); (A.K.)
- H3Africa Bioinformatics Network (H3ABioNet) Node, Centre for Genomics Research and Innovation, NABDA/FMST, Abuja 901101, Nigeria;
- Segun Fatumo, Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| |
Collapse
|
120
|
Limonova AS, Ershova AI, Kiseleva AV, Ramensky VE, Vyatkin YV, Kutsenko VA, Meshkov AN, Drapkina OM. Assessment of polygenic risk of hypertension. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2023. [DOI: 10.15829/1728-8800-2022-3464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Hypertension (HTN) is a leading risk factor for the development of cardiovascular diseases. In recent decades, the rapid development of genetic tests, in particular genome-wide association study (GWAS), has made it possible to identify hundreds of nucleotide sequence variants associated with the development of HTN. One approach to improve the predictive power of genetic testing is to combine information about many nucleotide sequence variants into a single risk assessment system, often referred to as a genetic risk score. Within the framework of this review, the most significant publications on the study of the genetic risk score for HTN will be considered, and the features of their development and application will be discussed.
Collapse
Affiliation(s)
- A. S. Limonova
- National Medical Research Center for Therapy and Preventive Medicine
| | - A. I. Ershova
- National Medical Research Center for Therapy and Preventive Medicine
| | - A. V. Kiseleva
- National Medical Research Center for Therapy and Preventive Medicine
| | - V. E. Ramensky
- National Medical Research Center for Therapy and Preventive Medicine; Lomonosov Moscow State University
| | - Yu. V. Vyatkin
- National Medical Research Center for Therapy and Preventive Medicine; Novosibirsk National Research State University
| | - V. A. Kutsenko
- National Medical Research Center for Therapy and Preventive Medicine; Faculty of Mechanics and Mathematics, Lomonosov Moscow State University
| | - A. N. Meshkov
- National Medical Research Center for Therapy and Preventive Medicine; Pirogov Russian National Research Medical University
| | - O. M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine
| |
Collapse
|
121
|
Yang J, Nie D, Chen Y, Liu Z, Li M, Gong C, Liu Q. The role of smoking and alcohol in mediating the effect of gastroesophageal reflux disease on lung cancer: A Mendelian randomization study. Front Genet 2023; 13:1054132. [PMID: 36726719 PMCID: PMC9885128 DOI: 10.3389/fgene.2022.1054132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/07/2022] [Indexed: 01/19/2023] Open
Abstract
Observational studies have suggested a positive association between gastroesophageal reflux disease and lung cancer, but due to the existence of confounders, it remains undetermined whether gastroesophageal reflux disease (GERD) has a causal association with lung cancer. Therefore, Mendelian randomization (MR) analyses were applied to investigate the relationship between the two conditions. Two-sample Mendelian randomization analysis was utilized with summary genetic data from the European Bioinformatics Institute (602,604 individuals) and International Lung Cancer Consortium, which provides information on lung cancer and its histological subgroups. Furthermore, we used two-step Mendelian randomization and multivariable Mendelian randomization to estimate whether smoking initiation (311,629 cases and 321,173 controls) and alcohol intake frequency (n = 462,346) mediate any effect of gastroesophageal reflux disease on lung cancer risk. The Mendelian randomization analyses indicated that gastroesophageal reflux disease was associated with and significantly increased the risk of lung cancer (ORIVW = 1.35, 95% CI = 1.18-1.54; p = 1.36 × 10-5). Smoking initiation and alcohol intake frequency mediated 35% and 3% of the total effect of gastroesophageal reflux disease on lung cancer, respectively. The combined effect of these two factors accounted for 60% of the total effect. In conclusion, gastroesophageal reflux disease is associated with an increased risk of lung cancer, and interventions to reduce smoking and alcohol intake may reduce the incidence of lung cancer.
Collapse
Affiliation(s)
- Jing Yang
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Duorui Nie
- Graduate school of Hunan University of Chinese Medicine, Changsha, China
| | - Yujing Chen
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zixing Liu
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Mengzhao Li
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chun Gong
- The First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Qiong Liu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
122
|
Soremekun O, Dib MJ, Rajasundaram S, Fatumo S, Gill D. Genetic heterogeneity in cardiovascular disease across ancestries: Insights for mechanisms and therapeutic intervention. CAMBRIDGE PRISMS. PRECISION MEDICINE 2023; 1:e8. [PMID: 38550935 PMCID: PMC10953756 DOI: 10.1017/pcm.2022.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 11/03/2024]
Abstract
Cardiovascular diseases (CVDs) are complex in their aetiology, arising due to a combination of genetics, lifestyle and environmental factors. By nature of this complexity, different CVDs vary in their molecular mechanisms, clinical presentation and progression. Although extensive efforts are being made to develop novel therapeutics for CVDs, genetic heterogeneity is often overlooked in the development process. By considering molecular mechanisms at an individual and ancestral level, a richer understanding of the influence of environmental and lifestyle factors can be gained and more refined therapeutic interventions can be developed. It is therefore expedient to understand the molecular and clinical heterogeneity in CVDs that exists across different populations. In this review, we highlight how the mechanisms underlying CVDs vary across diverse population ancestry groups due to genetic heterogeneity. We then discuss how such genetic heterogeneity is being leveraged to inform therapeutic interventions and personalised medicine, highlighting examples across the CVD spectrum. Finally, we present an overview of how polygenic risk scores and Mendelian randomisation can foster more robust insight into disease mechanisms and therapeutic intervention in diverse populations. Fulfilment of the vision of precision medicine requires more exhaustive leveraging of the genetic variability across diverse ancestry populations to improve our understanding of disease onset, progression and response to therapeutic intervention.
Collapse
Affiliation(s)
- Opeyemi Soremekun
- The African Computational Genomics (TACG) Research Group, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| | - Marie-Joe Dib
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- British Heart Foundation Centre of Excellence, Imperial College London, London, UK
| | - Skanda Rajasundaram
- Centre for Evidence-Based Medicine, University of Oxford, Oxford, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Segun Fatumo
- The African Computational Genomics (TACG) Research Group, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
- Department of Non-Communicable Disease Epidemiology (NCDE), London School of Hygiene and Tropical Medicine, London, UK
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- British Heart Foundation Centre of Excellence, Imperial College London, London, UK
| |
Collapse
|
123
|
Zsichla L, Müller V. Risk Factors of Severe COVID-19: A Review of Host, Viral and Environmental Factors. Viruses 2023; 15:175. [PMID: 36680215 PMCID: PMC9863423 DOI: 10.3390/v15010175] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/04/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
The clinical course and outcome of COVID-19 are highly variable, ranging from asymptomatic infections to severe disease and death. Understanding the risk factors of severe COVID-19 is relevant both in the clinical setting and at the epidemiological level. Here, we provide an overview of host, viral and environmental factors that have been shown or (in some cases) hypothesized to be associated with severe clinical outcomes. The factors considered in detail include the age and frailty, genetic polymorphisms, biological sex (and pregnancy), co- and superinfections, non-communicable comorbidities, immunological history, microbiota, and lifestyle of the patient; viral genetic variation and infecting dose; socioeconomic factors; and air pollution. For each category, we compile (sometimes conflicting) evidence for the association of the factor with COVID-19 outcomes (including the strength of the effect) and outline possible action mechanisms. We also discuss the complex interactions between the various risk factors.
Collapse
Affiliation(s)
- Levente Zsichla
- Institute of Biology, Eötvös Loránd University, 1117 Budapest, Hungary
- National Laboratory for Health Security, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Viktor Müller
- Institute of Biology, Eötvös Loránd University, 1117 Budapest, Hungary
- National Laboratory for Health Security, Eötvös Loránd University, 1117 Budapest, Hungary
| |
Collapse
|
124
|
O'Sullivan JW, Ashley EA, Elliott PM. Polygenic risk scores for the prediction of cardiometabolic disease. Eur Heart J 2023; 44:89-99. [PMID: 36478054 DOI: 10.1093/eurheartj/ehac648] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 08/28/2022] [Accepted: 10/27/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiometabolic diseases contribute more to global morbidity and mortality than any other group of disorders. Polygenic risk scores (PRSs), the weighted summation of individually small-effect genetic variants, represent an advance in our ability to predict the development and complications of cardiometabolic diseases. This article reviews the evidence supporting the use of PRS in seven common cardiometabolic diseases: coronary artery disease (CAD), stroke, hypertension, heart failure and cardiomyopathies, obesity, atrial fibrillation (AF), and type 2 diabetes mellitus (T2DM). Data suggest that PRS for CAD, AF, and T2DM consistently improves prediction when incorporated into existing clinical risk tools. In other areas such as ischaemic stroke and hypertension, clinical application appears premature but emerging evidence suggests that the study of larger and more diverse populations coupled with more granular phenotyping will propel the translation of PRS into practical clinical prediction tools.
Collapse
Affiliation(s)
- Jack W O'Sullivan
- Stanford Center for Inherited Cardiovascular Disease, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Euan A Ashley
- Stanford Center for Inherited Cardiovascular Disease, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Perry M Elliott
- UCL Institute of Cardiovascular Science, Gower Street, London WC1E 6BT, UK
- St. Bartholomew's Hospital, W Smithfield, London EC1A 7BE, UK
| |
Collapse
|
125
|
mRNA Metabolism and Hypertension. Biomedicines 2023; 11:biomedicines11010118. [PMID: 36672629 PMCID: PMC9855994 DOI: 10.3390/biomedicines11010118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/28/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023] Open
Abstract
Hypertension is the most frequent cardiovascular risk factor all over the world. It remains a leading contributor to the risk of cardiovascular events and death. In the year 2015, about 1.5 billion of adult people worldwide had hypertension (as defined by office systolic blood pressure ≥ 140 mmHg or office diastolic blood pressure ≥ 90 mmHg). Moreover, the number of hypertensive patients with age ranging from 30 to 79 years doubled in the last 30 years (from 317 million men and 331 million women in the year 1990 to 652 million men and 626 million women in 2019) despite stable age-standardized prevalence worldwide. Despite such impressive growth, the proportion of controlled hypertension is very low. A better understanding of the pathogenesis of hypertension may contribute to the development of innovative therapeutic strategies. In this context, alterations of the messenger RNA metabolism have been recently evaluated as contributors to the pathogenesis of hypertension, and pharmacological modulation of RNA metabolism is under investigation as potential and novel therapeutic armamentarium in hypertension.
Collapse
|
126
|
Cao Y, Li P, Zhang Y, Qiu M, Li J, Ma S, Yan Y, Li Y, Han Y. Association of systemic immune inflammatory index with all-cause and cause-specific mortality in hypertensive individuals: Results from NHANES. Front Immunol 2023; 14:1087345. [PMID: 36817427 PMCID: PMC9932782 DOI: 10.3389/fimmu.2023.1087345] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
Background The relationship between the systemic immune inflammatory index (SII) and the prognosis of hypertensive patients is unclear. This study aims to explore the association of SII with all-cause and cause-specific mortality in patients with hypertension. Methods This study included 8524 adults with hypertension from the National Health and Nutritional Examination Surveys (NHANES) 2011-2018, and followed for survival through December 31, 2019. Cox proportional hazards models were used to investigate the associations between SII and mortality from all causes, cardiovascular disease (CVD), and cancer. Restricted cubic spline, piecewise linear regression, subgroup and sensitivity analyses were also used. Results During a median follow-up of 4.58 years, 872 all-cause deaths occurred. After adjusting for covariates, higher SII was significantly associated with an elevated risk of CVD mortality. There was a 102% increased risk of CVD mortality per one-unit increment in natural log-transformed SII (lnSII) (P < 0.001). Consistent results were also observed when SII was examined as categorical variable (quartiles). The associations of SII with all-cause and cancer mortality were detected as U-shaped with threshold values of 5.97 and 6.18 for lnSII respectively. Below thresholds, higher SII was significantly associated with lower all-cause mortality (HR=0.79, 95%CI=0.64-0.97) and cancer mortality (HR=0.73, 95%CI=0.53-1.00). Above thresholds, SII was significantly positive associated with all-cause mortality (HR=1.93, 95%CI=1.55-2.40) and cancer mortality (HR=1.93, 95%CI=1.22-3.05). The results were robust in subgroup and sensitivity analyses. Conclusion Higher SII (either as a continuous or categorical variable) were significantly associated with a higher risk of CVD mortality. The U-shaped associations were observed between SII and all-cause and cancer mortality.
Collapse
Affiliation(s)
- Yang Cao
- The Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China.,The Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shanxi, China
| | - Pengxiao Li
- The Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China.,The Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shanxi, China
| | - Yan Zhang
- The Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shanxi, China
| | - Miaohan Qiu
- The Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Jing Li
- The Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Sicong Ma
- The Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Yudong Yan
- The Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China.,The Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shanxi, China
| | - Yi Li
- The Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Yaling Han
- The Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| |
Collapse
|
127
|
Kutumova E, Kiselev I, Sharipov R, Lifshits G, Kolpakov F. Mathematical modeling of antihypertensive therapy. Front Physiol 2022; 13:1070115. [PMID: 36589434 PMCID: PMC9795234 DOI: 10.3389/fphys.2022.1070115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Hypertension is a multifactorial disease arising from complex pathophysiological pathways. Individual characteristics of patients result in different responses to various classes of antihypertensive medications. Therefore, evaluating the efficacy of therapy based on in silico predictions is an important task. This study is a continuation of research on the modular agent-based model of the cardiovascular and renal systems (presented in the previously published article). In the current work, we included in the model equations simulating the response to antihypertensive therapies with different mechanisms of action. For this, we used the pharmacodynamic effects of the angiotensin II receptor blocker losartan, the calcium channel blocker amlodipine, the angiotensin-converting enzyme inhibitor enalapril, the direct renin inhibitor aliskiren, the thiazide diuretic hydrochlorothiazide, and the β-blocker bisoprolol. We fitted therapy parameters based on known clinical trials for all considered medications, and then tested the model's ability to show reasonable dynamics (expected by clinical observations) after treatment with individual drugs and their dual combinations in a group of virtual patients with hypertension. The extended model paves the way for the next step in personalized medicine that is adapting the model parameters to a real patient and predicting his response to antihypertensive therapy. The model is implemented in the BioUML software and is available at https://gitlab.sirius-web.org/virtual-patient/antihypertensive-treatment-modeling.
Collapse
Affiliation(s)
- Elena Kutumova
- Department of Computational Biology, Sirius University of Science and Technology, Sochi, Russia,Laboratory of Bioinformatics, Federal Research Center for Information and Computational Technologies, Novosibirsk, Russia,Biosoft.Ru, Ltd., Novosibirsk, Russia,*Correspondence: Elena Kutumova,
| | - Ilya Kiselev
- Department of Computational Biology, Sirius University of Science and Technology, Sochi, Russia,Laboratory of Bioinformatics, Federal Research Center for Information and Computational Technologies, Novosibirsk, Russia,Biosoft.Ru, Ltd., Novosibirsk, Russia
| | - Ruslan Sharipov
- Department of Computational Biology, Sirius University of Science and Technology, Sochi, Russia,Laboratory of Bioinformatics, Federal Research Center for Information and Computational Technologies, Novosibirsk, Russia,Biosoft.Ru, Ltd., Novosibirsk, Russia,Specialized Educational Scientific Center, Novosibirsk State University, Novosibirsk, Russia
| | - Galina Lifshits
- Laboratory for Personalized Medicine, Center of New Medical Technologies, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | - Fedor Kolpakov
- Department of Computational Biology, Sirius University of Science and Technology, Sochi, Russia,Laboratory of Bioinformatics, Federal Research Center for Information and Computational Technologies, Novosibirsk, Russia,Biosoft.Ru, Ltd., Novosibirsk, Russia
| |
Collapse
|
128
|
Tsuo K, Zhou W, Wang Y, Kanai M, Namba S, Gupta R, Majara L, Nkambule LL, Morisaki T, Okada Y, Neale BM, Global Biobank Meta-analysis Initiative, Daly MJ, Martin AR. Multi-ancestry meta-analysis of asthma identifies novel associations and highlights the value of increased power and diversity. CELL GENOMICS 2022; 2:100212. [PMID: 36778051 PMCID: PMC9903683 DOI: 10.1016/j.xgen.2022.100212] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 09/01/2022] [Accepted: 10/12/2022] [Indexed: 11/09/2022]
Abstract
Asthma is a complex disease that varies widely in prevalence across populations. The extent to which genetic variation contributes to these disparities is unclear, as the genetics underlying asthma have been investigated primarily in populations of European descent. As part of the Global Biobank Meta-analysis Initiative, we conducted a large-scale genome-wide association study of asthma (153,763 cases and 1,647,022 controls) via meta-analysis across 22 biobanks spanning multiple ancestries. We discovered 179 asthma-associated loci, 49 of which were not previously reported. Despite the wide range in asthma prevalence among biobanks, we found largely consistent genetic effects across biobanks and ancestries. The meta-analysis also improved polygenic risk prediction in non-European populations compared with previous studies. Additionally, we found considerable genetic overlap between age-of-onset subtypes and between asthma and comorbid diseases. Our work underscores the multi-factorial nature of asthma development and offers insight into its shared genetic architecture.
Collapse
Affiliation(s)
- Kristin Tsuo
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Wei Zhou
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ying Wang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Masahiro Kanai
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shinichi Namba
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Rahul Gupta
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Lerato Majara
- Department of Psychiatry and Mental Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Lethukuthula L. Nkambule
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Takayuki Morisaki
- Division of Molecular Pathology, The Institute of Medical Science, The University of Tokyo, Minatu-ku, Tokyo, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita 565-0871, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita 565-0871, Japan
| | - Benjamin M. Neale
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Global Biobank Meta-analysis Initiative
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry and Mental Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Division of Molecular Pathology, The Institute of Medical Science, The University of Tokyo, Minatu-ku, Tokyo, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita 565-0871, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita 565-0871, Japan
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Mark J. Daly
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Alicia R. Martin
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
129
|
Abramova M, Churnosova M, Efremova O, Aristova I, Reshetnikov E, Polonikov A, Churnosov M, Ponomarenko I. Effects of Pre-Pregnancy Overweight/Obesity on the Pattern of Association of Hypertension Susceptibility Genes with Preeclampsia. Life (Basel) 2022; 12:2018. [PMID: 36556383 PMCID: PMC9784908 DOI: 10.3390/life12122018] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/21/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
The aim of this study was to explore the effects of pre-pregnancy overweight/obesity on the pattern of association of hypertension susceptibility genes with preeclampsia (PE). Ten single-nucleotide polymorphisms (SNPs) of the 10 genome-wide association studies (GWAS)-significant hypertension/blood pressure (BP) candidate genes were genotyped in 950 pregnant women divided into two cohorts according to their pre-pregnancy body mass index (preBMI): preBMI ≥ 25 (162 with PE and 159 control) and preBMI < 25 (290 with PE and 339 control). The PLINK software package was utilized to study the association (analyzed four genetic models using logistic regression). The functionality of PE-correlated loci was analyzed by performing an in silico database analysis. Two SNP hypertension/BP genes, rs805303 BAG6 (OR: 0.36−0.66) and rs167479 RGL3 (OR: 1.86), in subjects with preBMI ≥ 25 were associated with PE. No association between the studied SNPs and PE in the preBMI < 25 group was determined. Further analysis showed that two PE-associated SNPs are functional (have weighty eQTL, sQTL, regulatory, and missense values) and could be potentially implicated in PE development. In conclusion, this study was the first to discover the modifying influence of overweight/obesity on the pattern of association of GWAS-significant hypertension/BP susceptibility genes with PE: these genes are linked with PE in preBMI ≥ 25 pregnant women and are not PE-involved in the preBMI < 25 group.
Collapse
Affiliation(s)
- Maria Abramova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Maria Churnosova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Olesya Efremova
- Department of Medical Genetics, Kharkiv National Medical University, 61022 Kharkov, Ukraine
- Grishchenko Clinic of Reproductive Medicine, 61052 Kharkov, Ukraine
| | - Inna Aristova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Evgeny Reshetnikov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Alexey Polonikov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
- Department of Biology, Medical Genetics and Ecology and Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
| | - Mikhail Churnosov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Irina Ponomarenko
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| |
Collapse
|
130
|
Preudhomme LK, Gellman MD, Franceschini N, Perreira KM, Fernández-Rhodes LE, Gallo LC, Isasi CR, Smoller S, Castañeda SF, Daviglus M, Hutten C, Cooper RS, Cai J, Schneiderman N, Llabre MM. Genetic and stress influences on the prevalence of hypertension among hispanics/latinos in the hispanic community health study/study of latinos (HCHS/SOL). Blood Press 2022; 31:155-163. [PMID: 35762607 DOI: 10.1080/08037051.2022.2091977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
PURPOSE The current study examined the effects of chronic stress and a genetic risk score on the presence of hypertension and elevated systolic blood pressure and diastolic blood pressure among Hispanics/Latinos in the target population of Hispanic Community Health Study/Study of Latinos. MATERIALS AND METHODS Of the participants (N = 11,623) assessed during two clinic visits (Visit 1 2008-2013 & Visit 2 2014-2018), we analysed data from 7,429 adults (50.4% female), aged 18-74, who were genotyped and responded to chronic stress questionnaires. We calculated an unweighted genetic risk score using blood pressure increasing single nucleotide polymorphisms (SNPs) found to be generalisable to Hispanics/Latinos (10 SNPs). Linear and logistic regression models were used to estimate associations between chronic stress and genetic risk score and their interaction, with prevalent Visit 2 SBP or DBP, and hypertension, respectively. Models accounted for sampling weights, stratification, and cluster design. RESULTS Chronic stress (adjusted OR = 1.18, 95%CI:1.15,1.22) and hypertension genetic risk score (adjusted OR = 1.04, 95%CI:1.01,1.07) were significantly associated with prevalent hypertension, but there was no significant interaction between the chronic stress and genetic risk score on hypertension (p = .49). genetic risk score (b = .32, 95%CI:.08, .55, R2 = .02) and chronic stress (b = .45, 95%CI:.19, .72, R2 = .11) were related to DBP, with no significant interaction (p = .62). Genetic risk score (b = .42, 95%CI:.08, .76, R2 = .01) and chronic stress (b = .80, 95%CI:.34,1.26, R2 = .11) were also related to SBP, with no significant interaction (p = .51). CONCLUSION Results demonstrate the utility of a genetic risk score for blood pressure and are consistent with literature suggesting chronic stress has a strong, direct association with elevated blood pressure among U.S. Hispanics/Latinos.
Collapse
Affiliation(s)
| | - Marc D Gellman
- University of Miami, Department of Psychology, Coral Gables, USA
| | | | | | - Lindsay E Fernández-Rhodes
- Pennsylvania State University, Biobehavioral Health - BBH Epidemiology and Genetics across Populations & Societies, University Park, USA
| | - Linda C Gallo
- San Diego State University, Department of Psychology, San Diego, USA
| | - Carmen R Isasi
- Albert Einstein College of Medicine, Department of Epidemiology & Population Health, Bronx, USA
| | - Sylvia Smoller
- Albert Einstein College of Medicine, Department of Epidemiology & Population Health, Bronx, USA
| | | | - Martha Daviglus
- University of Illinois at Chicago, Department of Medicine, Chicago, USA
| | - Christina Hutten
- University of Illinois at Chicago, Department of Epidemiology and Biostatistics, Chicago, USA
| | - Richard S Cooper
- Loyola University-Chicago, Department of Public Health Sciences, Chicago, USA
| | - Jianwen Cai
- UNC-Chapel Hill, Department of Epidemiology, Chapel Hill, USA
| | | | - Maria M Llabre
- University of Miami, Department of Psychology, Coral Gables, USA
| |
Collapse
|
131
|
Identifying susceptibility genes for essential hypertension by transcriptome-wide association study. Biochem Biophys Rep 2022; 32:101387. [DOI: 10.1016/j.bbrep.2022.101387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/29/2022] [Accepted: 11/12/2022] [Indexed: 11/22/2022] Open
|
132
|
Abstract
Primary hypertension (PH) is most common during adolescence with increasing prevalence globally, alongside the epidemic of obesity. Unlike in adults, there are no data on children with uncontrolled hypertension and their future risk of hard cardiovascular and cerebrovascular outcomes. However, hypertension in childhood is linked to hypertensive-mediated organ damage (HMOD) which is often reversible if treated appropriately. Despite differing guidelines regarding the threshold for defining hypertension, there is consensus that early recognition and prompt management with lifestyle modification escalating to antihypertensive medication is required to ameliorate adverse outcomes. Unfortunately, many unknowns remain regarding pathophysiology and optimum treatment of childhood hypertension.
Collapse
Affiliation(s)
- Emily Haseler
- Department of Paediatric Nephrology, Evelina London Children's Hospital, Guys & St Thomas NHS Foundation Trust, Westminster Bridge Road, 3rd Floor Beckett House, London SE1 7EH, United Kingdom; Kings College London, United Kingdom
| | - Manish D Sinha
- Department of Paediatric Nephrology, Evelina London Children's Hospital, Guys & St Thomas NHS Foundation Trust, Westminster Bridge Road, 3rd Floor Beckett House, London SE1 7EH, United Kingdom; Kings College London, United Kingdom.
| |
Collapse
|
133
|
Katz AE, Yang ML, Levin MG, Tcheandjieu C, Mathis M, Hunker K, Blackburn S, Eliason JL, Coleman DM, Fendrikova-Mahlay N, Gornik HL, Karmakar M, Hill H, Xu C, Zawistowski M, Brummett CM, Zoellner S, Zhou X, O'Donnell CJ, Douglas JA, Assimes TL, Tsao PS, Li JZ, Damrauer SM, Stanley JC, Ganesh SK. Fibromuscular Dysplasia and Abdominal Aortic Aneurysms Are Dimorphic Sex-Specific Diseases With Shared Complex Genetic Architecture. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2022; 15:e003496. [PMID: 36374587 PMCID: PMC9772208 DOI: 10.1161/circgen.121.003496] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/26/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND The risk of arterial diseases may be elevated among family members of individuals having multifocal fibromuscular dysplasia (FMD). We sought to investigate the risk of arterial diseases in families of individuals with FMD. METHODS Family histories for 73 probands with FMD were obtained, which included an analysis of 463 total first-degree relatives focusing on FMD and related arterial disorders. A polygenic risk score for FMD (PRSFMD) was constructed from prior genome-wide association findings of 584 FMD cases and 7139 controls and evaluated for association with an abdominal aortic aneurysm (AAA) in a cohort of 9693 AAA cases and 294 049 controls. A previously published PRSAAA was also assessed among the FMD cases and controls. RESULTS Of all first degree relatives of probands, 9.3% were diagnosed with FMD, aneurysms, and dissections. Aneurysmal disease occurred in 60.5% of affected relatives and 5.6% of all relatives. Among 227 female first-degree relatives of probands, 4.8% (11) had FMD, representing a relative risk (RR)FMD of 1.5 ([95% CI, 0.75-2.8]; P=0.19) compared with the estimated population prevalence of 3.3%, though not of statistical significance. Of all fathers of FMD probands, 11% had AAAs resulting in a RRAAA of 2.3 ([95% CI, 1.12-4.6]; P=0.014) compared with population estimates. The PRSFMD was found to be associated with an AAA (odds ratio, 1.03 [95% CI, 1.01-1.05]; P=2.6×10-3), and the PRSAAA was found to be associated with FMD (odds ratio, 1.53 [95% CI, 1.2-1.9]; P=9.0×10-5) as well. CONCLUSIONS FMD and AAAs seem to be sex-dimorphic manifestations of a heritable arterial disease with a partially shared complex genetic architecture. Excess risk of having an AAA according to a family history of FMD may justify screening in family members of individuals having FMD.
Collapse
Affiliation(s)
- Alexander E Katz
- Department of Internal Medicine, Division of Cardiovascular Medicine (A.E.K., M.-L.Y., K.H., H.H., S.K.G.), University of Michigan, Ann Arbor
- Department of Human Genetics (A.E.K., M.-L.Y., K.H., H.H., J.A.D., J.Z.L., S.K.G.), University of Michigan, Ann Arbor
- Medical Genomics & Metabolic Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD (A.E.K.)
| | - Min-Lee Yang
- Department of Internal Medicine, Division of Cardiovascular Medicine (A.E.K., M.-L.Y., K.H., H.H., S.K.G.), University of Michigan, Ann Arbor
- Department of Human Genetics (A.E.K., M.-L.Y., K.H., H.H., J.A.D., J.Z.L., S.K.G.), University of Michigan, Ann Arbor
- Department of Computational Medicine and Bioinformatics (M.-L.Y.), University of Michigan, Ann Arbor
| | - Michael G Levin
- Corporal Michael J. Crescenz Philadelphia VA Medical Center (M.G.L., S.M.D.)
- Division of Cardiovascular Medicine, Department of Medicine (M.G.L.)
| | - Catherine Tcheandjieu
- Gladstone Institute of data science and Biotechnology, Gladstone Institutes; and Department of epidemiology and biostatistics, University of California at San Francisco, CA. (C.T.)
| | - Michael Mathis
- Department of Anesthesiology, Michigan Medicine (M.M., C.M.B.), University of Michigan, Ann Arbor
| | - Kristina Hunker
- Department of Internal Medicine, Division of Cardiovascular Medicine (A.E.K., M.-L.Y., K.H., H.H., S.K.G.), University of Michigan, Ann Arbor
- Department of Human Genetics (A.E.K., M.-L.Y., K.H., H.H., J.A.D., J.Z.L., S.K.G.), University of Michigan, Ann Arbor
| | - Susan Blackburn
- Department of Surgery, Section of Vascular Surgery (S.B., J.L.E., D.M.C., M.K., J.C.S.), University of Michigan, Ann Arbor
| | - Jonathan L Eliason
- Department of Surgery, Section of Vascular Surgery (S.B., J.L.E., D.M.C., M.K., J.C.S.), University of Michigan, Ann Arbor
| | - Dawn M Coleman
- Department of Surgery, Section of Vascular Surgery (S.B., J.L.E., D.M.C., M.K., J.C.S.), University of Michigan, Ann Arbor
| | | | - Heather L Gornik
- Harrington Heart and Vascular Institute, University Hospitals, Cleveland, OH (H.L.G.)
| | - Monita Karmakar
- Department of Surgery, Section of Vascular Surgery (S.B., J.L.E., D.M.C., M.K., J.C.S.), University of Michigan, Ann Arbor
| | - Hannah Hill
- Department of Internal Medicine, Division of Cardiovascular Medicine (A.E.K., M.-L.Y., K.H., H.H., S.K.G.), University of Michigan, Ann Arbor
- Department of Human Genetics (A.E.K., M.-L.Y., K.H., H.H., J.A.D., J.Z.L., S.K.G.), University of Michigan, Ann Arbor
| | - Chang Xu
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor (C.X., M.Z., S.Z., X.Z.)
| | - Matthew Zawistowski
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor (C.X., M.Z., S.Z., X.Z.)
| | - Chad M Brummett
- Department of Anesthesiology, Michigan Medicine (M.M., C.M.B.), University of Michigan, Ann Arbor
| | - Sebastian Zoellner
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor (C.X., M.Z., S.Z., X.Z.)
| | - Xiang Zhou
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor (C.X., M.Z., S.Z., X.Z.)
| | - Christopher J O'Donnell
- VA Boston Healthcare System (C.O.)
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (C.O.)
| | - Julie A Douglas
- Department of Human Genetics (A.E.K., M.-L.Y., K.H., H.H., J.A.D., J.Z.L., S.K.G.), University of Michigan, Ann Arbor
| | - Themistocles L Assimes
- VA Palo Alto Health Care System (T.L.A., P.S.T.)
- Division of Cardiovascular Medicine, Department of Medicine (T.L.A.), Stanford University School of Medicine, CA
| | | | - Jun Z Li
- Department of Human Genetics (A.E.K., M.-L.Y., K.H., H.H., J.A.D., J.Z.L., S.K.G.), University of Michigan, Ann Arbor
| | - Scott M Damrauer
- Corporal Michael J. Crescenz Philadelphia VA Medical Center (M.G.L., S.M.D.)
- Department of Surgery and Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia (S.M.D.)
| | - James C Stanley
- Department of Surgery, Section of Vascular Surgery (S.B., J.L.E., D.M.C., M.K., J.C.S.), University of Michigan, Ann Arbor
| | - Santhi K Ganesh
- Department of Internal Medicine, Division of Cardiovascular Medicine (A.E.K., M.-L.Y., K.H., H.H., S.K.G.), University of Michigan, Ann Arbor
- Department of Human Genetics (A.E.K., M.-L.Y., K.H., H.H., J.A.D., J.Z.L., S.K.G.), University of Michigan, Ann Arbor
| |
Collapse
|
134
|
Shah R, St-Onge MP, Emin M, Gao S, Sampogna RV, Aggarwal B, Wei Y, Jelic S. Sleep Deprivation Impairs Vascular Function in Healthy Women: A Clinical Trial. Ann Am Thorac Soc 2022; 19:2097-2100. [PMID: 36112109 PMCID: PMC9743480 DOI: 10.1513/annalsats.202205-406rl] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Affiliation(s)
| | | | | | - Su Gao
- Columbia UniversityNew York, New York
| | | | | | - Ying Wei
- Columbia UniversityNew York, New York
| | | |
Collapse
|
135
|
Parcha V, Pampana A, Shetty NS, Irvin MR, Natarajan P, Lin HJ, Guo X, Rich SS, Rotter JI, Li P, Oparil S, Arora G, Arora P. Association of a Multiancestry Genome-Wide Blood Pressure Polygenic Risk Score With Adverse Cardiovascular Events. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2022; 15:e003946. [PMID: 36334310 PMCID: PMC9812363 DOI: 10.1161/circgen.122.003946] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/04/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND Traditional cardiovascular risk factors and the underlying genetic risk of elevated blood pressure (BP) determine an individual's composite risk of developing adverse cardiovascular events. We sought to evaluate the relative contributions of the traditional cardiovascular risk factors to the development of adverse cardiovascular events in the context of varying BP genetic risk profiles. METHODS Genome-wide polygenic risk score (PRS) was computed using multiancestry genome-wide association estimates among US adults who underwent whole-genome sequencing in the Trans-Omics for Precision program. Individuals were stratified into high, intermediate, and low genetic risk groups (>80th, 20-80th, and <20th centiles of systolic BP [SBP] PRS). Based on the ACC/AHA Pooled Cohort Equations, participants were stratified into low and high (10 year-atherosclerotic cardiovascular disease [CVD] risk: <10% or ≥10%) cardiovascular risk factor profile groups. The primary study outcome was incident cardiovascular event (composite of incident heart failure, incident stroke, and incident coronary heart disease). RESULTS Among 21 897 US adults (median age: 56 years; 56.0% women; 35.8% non-White race/ethnicity), 1 SD increase in the SBP PRS, computed using 1.08 million variants, was associated with SBP (β: 4.39 [95% CI, 4.13-4.65]) and hypertension (odds ratio, 1.50 [95% CI, 1.46-1.55]), respectively. This association was robustly seen across racial/ethnic groups. Each SD increase in SBP PRS was associated with a higher risk of the incident CVD (multivariable-adjusted hazards ratio, 1.07 [95% CI, 1.04-1.10]) after controlling for ACC/AHA Pooled Cohort Equations risk scores. Among individuals with a high SBP PRS, low atherosclerotic CVD risk was associated with a 58% lower hazard for incident CVD (multivariable-adjusted hazards ratio, 0.42 [95% CI, 0.36-0.50]) compared to those with high atherosclerotic CVD risk. A similar pattern was noted in intermediate and low genetic risk groups. CONCLUSIONS In a multiancestry cohort of >21 000 US adults, genome-wide SBP PRS was associated with BP traits and adverse cardiovascular events. Adequate control of modifiable cardiovascular risk factors may reduce the predisposition to adverse cardiovascular events among those with a high SBP PRS.
Collapse
Affiliation(s)
- Vibhu Parcha
- Division of Cardiovascular Disease, Univ of Alabama at Birmingham, Birmingham, AL
| | - Akhil Pampana
- Division of Cardiovascular Disease, Univ of Alabama at Birmingham, Birmingham, AL
| | - Naman S. Shetty
- Division of Cardiovascular Disease, Univ of Alabama at Birmingham, Birmingham, AL
| | - Marguerite R. Irvin
- Dept of Epidemiology, School of Public Health, Univ of Alabama at Birmingham, Birmingham, AL
| | - Pradeep Natarajan
- Cardiology Division, Dept of Medicine, Massachusetts General Hospital
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
- Program in Medical & Population Genetics, Broad Institute of Harvard & MIT, Cambridge, MA
| | - Henry J. Lin
- The Institute for Translational Genomics & Population Sciences, Dept of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Xiuqing Guo
- The Institute for Translational Genomics & Population Sciences, Dept of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Stephen S. Rich
- Center for Public Health, Univ of Virginia, Charlottesville, VA
| | - Jerome I. Rotter
- The Institute for Translational Genomics & Population Sciences, Dept of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Peng Li
- School of Nursing, Univ of Alabama at Birmingham, Birmingham, AL
| | - Suzanne Oparil
- Division of Cardiovascular Disease, Univ of Alabama at Birmingham, Birmingham, AL
| | - Garima Arora
- Division of Cardiovascular Disease, Univ of Alabama at Birmingham, Birmingham, AL
| | - Pankaj Arora
- Division of Cardiovascular Disease, Univ of Alabama at Birmingham, Birmingham, AL
- Section of Cardiology, Birmingham Veterans Affairs Medical Center, Birmingham, AL
| |
Collapse
|
136
|
The Role of Certain Polymorphic Variants in Genes, Previously Associated with Blood Pressure Values, with Reference to the Risk of Development of Coronary Artery Disease. ACTA MEDICA BULGARICA 2022. [DOI: 10.2478/amb-2022-0034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
Abstract
The aim of the study was to analyze the effect of polymorphic variants previously associated with arterial hypertension (AH) in Genome Wide Association Studies (GWASs) in/next to genes and locuses CYP7A1 and PLEKHA7 on the development of coronary artery disease (CAD) in Bulgarian patients. A hundred and nine consecutive patients with angiographically documented CAD were studied. The genotyping was done with 7900 HT Fast Real-Time PCR (Applied Biosystems) with TaqMan® method. The control group consisted of 192 healthy population controls, selected from the bio- bank of the Molecular Medicine Center. SPSS and PLINK were used for the statistical analysis with level of significance < 0.05 and confidence interval 95%. The mean age of the studied patients was 63.71 ± 9.35 years; 35 (35%) females. Previous myocardial infarction (MI) had 38(38%); one-vessel – 39 (39%); two-vessel – 28 (28%); three-vessel disease – 34 (34%); 43 (43%) were with diabetes mellitus; 92 (92%) – with arterial hypertension (AH); 77 (77%) – with dyslipidemia; 42 (42%) were smokers; 25 (25%) were obese. We did not find any significant association between CAD and poly- morphism rs11191548 near CYP17A1 and only a tendency for genotype of rs381815 in PLEKHA7 (p = 0.06; OR 0.64; CI 0.40-1.02 for CAD) under dominant model. This is of practical importance both for studying the genetic aspects of CAD in the future and for enlargement of the current database.
Collapse
|
137
|
Kumar S, Yadav S, Kumar A. Oscillometric Waveform Evaluation for Blood Pressure Devices. BIOMEDICAL ENGINEERING ADVANCES 2022. [DOI: 10.1016/j.bea.2022.100046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
138
|
Abramova MY, Ponomarenko IV, Churnosov MI. The Polymorphic Locus rs167479 of the RGL3 Gene Is Associated with the Risk of Severe Preeclampsia. RUSS J GENET+ 2022. [DOI: 10.1134/s102279542212002x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
139
|
Xiao X, Li R, Wu C, Yan Y, Yuan M, Cui B, Zhang Y, Zhang C, Zhang X, Zhang W, Hui R, Wang Y. A genome-wide association study identifies a novel association between SDC3 and apparent treatment-resistant hypertension. BMC Med 2022; 20:463. [PMID: 36447229 PMCID: PMC9710180 DOI: 10.1186/s12916-022-02665-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 11/14/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Compared with patients who require fewer antihypertensive agents, those with apparent treatment-resistant hypertension (aTRH) are at increased risk for cardiovascular and all-cause mortality, independent of blood pressure control. However, the etiopathogenesis of aTRH is still poorly elucidated. METHODS We performed a genome-wide association study (GWAS) in first cohort including 586 aTRHs and 871 healthy controls. Next, expression quantitative trait locus (eQTL) analysis was used to identify genes that are regulated by single nucleotide polymorphisms (SNPs) derived from the GWAS. Then, we verified the genes obtained from the eQTL analysis in the validation cohort including 65 aTRHs, 96 hypertensives, and 100 healthy controls through gene expression profiling analysis and real-time quantitative polymerase chain reaction (RT-qPCR) assay. RESULTS The GWAS in first cohort revealed four suggestive loci (1p35, 4q13.2-21.1, 5q22-23.2, and 15q11.1-q12) represented by 23 SNPs. The 23 significant SNPs were in or near LAPTM5, SDC3, UGT2A1, FTMT, and NIPA1. eQTL analysis uncovered 14 SNPs in 1p35 locus all had same regulation directions for SDC3 and LAPTM5. The disease susceptible alleles of SNPs in 1p35 locus were associated with lower gene expression for SDC3 and higher gene expression for LAPTM5. The disease susceptible alleles of SNPs in 4q13.2-21.1 were associated with higher gene expression for UGT2B4. GTEx database did not show any statistically significant eQTLs between the SNPs in 5q22-23.2 and 15q11.1-q12 loci and their influenced genes. Then, gene expression profiling analysis in the validation cohort confirmed lower expression of SDC3 in aTRH but no significant differences on LAPTM5 and UGT2B4, when compared with controls and hypertensives, respectively. RT-qPCR assay further verified the lower expression of SDC3 in aTRH. CONCLUSIONS Our study identified a novel association of SDC3 with aTRH, which contributes to the elucidation of its etiopathogenesis and provides a promising therapeutic target.
Collapse
Affiliation(s)
- Xiao Xiao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Rd, Beijing, China
| | - Rui Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Rd, Beijing, China
| | - Cunjin Wu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Rd, Beijing, China
| | - Yupeng Yan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Rd, Beijing, China
| | - Mengmeng Yuan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Rd, Beijing, China
| | - Bing Cui
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Rd, Beijing, China
| | - Yu Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Rd, Beijing, China
| | - Channa Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Rd, Beijing, China
| | - Xiaoxia Zhang
- Department of Pharmacy, The First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, China
| | - Weili Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Rd, Beijing, China
| | - Rutai Hui
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Rd, Beijing, China
| | - Yibo Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Rd, Beijing, China.
| |
Collapse
|
140
|
Changalidis AI, Maksiutenko EM, Barbitoff YA, Tkachenko AA, Vashukova ES, Pachuliia OV, Nasykhova YA, Glotov AS. Aggregation of Genome-Wide Association Data from FinnGen and UK Biobank Replicates Multiple Risk Loci for Pregnancy Complications. Genes (Basel) 2022; 13:genes13122255. [PMID: 36553520 PMCID: PMC9777867 DOI: 10.3390/genes13122255] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/20/2022] [Accepted: 11/23/2022] [Indexed: 12/02/2022] Open
Abstract
Complications endangering mother or fetus affect around one in seven pregnant women. Investigation of the genetic susceptibility to such diseases is of high importance for better understanding of the disease biology as well as for prediction of individual risk. In this study, we collected and analyzed GWAS summary statistics from the FinnGen cohort and UK Biobank for 24 pregnancy complications. In FinnGen, we identified 11 loci associated with pregnancy hypertension, excessive vomiting, and gestational diabetes. When UK Biobank and FinnGen data were combined, we discovered six loci reaching genome-wide significance in the meta-analysis. These include rs35954793 in FGF5 (p=6.1×10-9), rs10882398 in PLCE1 (p=8.9×10-9), and rs167479 in RGL3 (p=5.2×10-9) for pregnancy hypertension, rs10830963 in MTNR1B (p=4.5×10-41) and rs36090025 in TCF7L2 (p=3.4×10-15) for gestational diabetes, and rs2963457 in the EBF1 locus (p=6.5×10-9) for preterm birth. In addition to the identified genome-wide associations, we also replicated 14 out of 40 previously reported GWAS markers for pregnancy complications, including four more preeclampsia-related variants. Finally, annotation of the GWAS results identified a causal relationship between gene expression in the cervix and gestational hypertension, as well as both known and previously uncharacterized genetic correlations between pregnancy complications and other traits. These results suggest new prospects for research into the etiology and pathogenesis of pregnancy complications, as well as early risk prediction for these disorders.
Collapse
Affiliation(s)
- Anton I. Changalidis
- Dpt. of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynaecology, and Reproductology, 199034 St. Petersburg, Russia
- Faculty of Software Engineering and Computer Systems, ITMO University, 197101 St. Petersburg, Russia
| | - Evgeniia M. Maksiutenko
- Dpt. of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynaecology, and Reproductology, 199034 St. Petersburg, Russia
- Dpt. of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Yury A. Barbitoff
- Dpt. of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynaecology, and Reproductology, 199034 St. Petersburg, Russia
- Dpt. of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia
- Correspondence: (Y.A.B.); (A.S.G.)
| | - Alexander A. Tkachenko
- Dpt. of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynaecology, and Reproductology, 199034 St. Petersburg, Russia
| | - Elena S. Vashukova
- Dpt. of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynaecology, and Reproductology, 199034 St. Petersburg, Russia
| | - Olga V. Pachuliia
- Dpt. of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynaecology, and Reproductology, 199034 St. Petersburg, Russia
| | - Yulia A. Nasykhova
- Dpt. of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynaecology, and Reproductology, 199034 St. Petersburg, Russia
| | - Andrey S. Glotov
- Dpt. of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynaecology, and Reproductology, 199034 St. Petersburg, Russia
- Correspondence: (Y.A.B.); (A.S.G.)
| |
Collapse
|
141
|
Mun S, Park S, Whang S, Whang M. Effects of Temporary Respiration Exercise with Individual Harmonic Frequency on Blood Pressure and Autonomic Balance. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:15676. [PMID: 36497750 PMCID: PMC9736073 DOI: 10.3390/ijerph192315676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 06/17/2023]
Abstract
This study investigated the effects of modulated respiration on blood pressure and autonomic balance to develop a healthcare application system for stabilizing autonomic balance. Thirty-two participants were asked to perform self-regulated tasks with 18 different respiration sequences, and their electrocardiograms (ECG) and blood pressure were measured. Changes in cardiovascular system functions and blood pressure were compared between free-breathing and various respiration conditions. Systolic and diastolic blood pressures stabilized after individual harmonic breathing. Autonomic balance, characterized by heart rate variability, was also stabilized with brief respiration training according to harmonic frequency. Five machine-learning algorithms were used to classify the two opposing factors between the free and modulated breathing conditions. The random forest models outperformed the other classifiers in the training data of systolic blood pressure and heart rate variability. The mean areas under the curves (AUCs) were 0.82 for systolic blood pressure and 0.98 for heart rate variability. Our findings lend support that blood pressure and autonomic balance were improved by temporary harmonic frequency respiration. This study provides a self-regulated respiration system that can control and help stabilize blood pressure and autonomic balance, which would help reduce mental stress and enhance human task performance in various fields.
Collapse
Affiliation(s)
- Sungchul Mun
- Department of Industrial Engineering, Jeonju Universtiy, Jeonju 55069, Republic of Korea
| | - Sangin Park
- Industry-Academy Cooperation Team, Hanyang University, Seoul 04763, Republic of Korea
| | - Sungyop Whang
- Rotary and Mission Systems, Lockheed Martin, 199 Borton Landing Road, Moorestown, NJ 08054, USA
| | - Mincheol Whang
- Department of Human-Centered Artificial Intelligence, Sangmyung University, Seoul 03016, Republic of Korea
| |
Collapse
|
142
|
Hypertensive disorders of pregnancy share common cfDNA methylation profiles. Sci Rep 2022; 12:19837. [PMID: 36400896 PMCID: PMC9674847 DOI: 10.1038/s41598-022-24348-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
Hypertensive disorders of pregnancy (HDP) contribute substantially to perinatal morbidity and mortality. Epigenetic changes point towards cardio-metabolic dysregulation for these vascular disorders. In early pregnancy, epigenetic changes using cell free DNA (cfDNA) are largely unexplored. We aimed to investigate these in HDP between 11 and 14 weeks of gestation by analysis of cfDNA methylation profiles in patients with hypertensive disorders. We identified patients without chronic hypertension but with subsequent development of preeclampsia (PE) (n = 11), with chronic hypertension (HT) but without PE development (n = 14), and lacking both PE and HT (n = 422). We matched patients according to PE risk factors into three groups (n = 5 each group): (1) PE: no HT but PE development, (2) HT: chronic hypertension but no PE and (3) Control: no PE or HT. We successfully optimized our cfDNA isolation process prior to whole genome bisulfite sequencing. Analysis of cfDNA methylation changes indicate a common predisposition in PE and HT groups, chiefly of maternal origin. Assessment of significant differentially methylated regions and annotated genes point towards a common cardiovascular predisposition in preeclampsia and hypertension groups in the first trimester. We postulate the pivotal role of the maternal cardiovascular system in HDP, which is already evident in the first trimester.
Collapse
|
143
|
Nyberg M, Terzic D, Ludvigsen TP, Mark PD, Michaelsen NB, Abildstrøm SZ, Engelmann M, Richards AM, Goetze JP. Review A State of Natriuretic Peptide Deficiency. Endocr Rev 2022; 44:379-392. [PMID: 36346821 PMCID: PMC10166265 DOI: 10.1210/endrev/bnac029] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/13/2022] [Accepted: 11/04/2022] [Indexed: 11/10/2022]
Abstract
Measurement of natriuretic peptides (NPs) has proven its clinical value as biomarker, especially in the context of heart failure (HF). In contrast, a state partial NP deficiency appears integral to several conditions in which lower NP concentrations in plasma presage overt cardiometabolic disease. Here, obesity and type 2 diabetes have attracted considerable attention. Other factors - including age, sex, race, genetics, and diurnal regulation - affect the NP "armory" and may leave some individuals more prone to development of cardiovascular disease. The molecular maturation of NPs has also proven complex with highly variable O-glycosylation within the biosynthetic precursors. The relevance of this regulatory step in post-translational propeptide maturation has recently become recognized in biomarker measurement/interpretation and cardiovascular pathophysiology. An important proportion of people appear to have reduced effective net NP bioactivity in terms of receptor activation and physiological effects. The state of NP deficiency, then, both entails a potential for further biomarker development and could also offer novel pharmacological possibilities. Alleviating the state of NP deficiency before development of overt cardiometabolic disease in selected patients could be a future path for improving precision medicine.
Collapse
Affiliation(s)
| | - Dijana Terzic
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | - Peter D Mark
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | | | | | - A Mark Richards
- Division of Cardiology, National University Heart Centre, National University Hospital, Singapore
| | - Jens P Goetze
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Department of Biomedical Sciences, Faculty of Health, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
144
|
Handelman SK, Puentes YM, Kuppa A, Chen Y, Du X, Feitosa MF, Palmer ND, Speliotes EK. Population-based meta-analysis and gene-set enrichment identifies FXR/RXR pathway as common to fatty liver disease and serum lipids. Hepatol Commun 2022; 6:3120-3131. [PMID: 36098472 PMCID: PMC9592792 DOI: 10.1002/hep4.2066] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 02/03/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is prevalent worldwide. NAFLD is associated with elevated serum triglycerides (TG), low-density lipoprotein cholesterol (LDL), and reduced high-density lipoprotein cholesterol (HDL). Both NAFLD and blood lipid levels are genetically influenced and may share a common genetic etiology. We used genome-wide association studies (GWAS)-ranked genes and gene-set enrichment analysis to identify pathways that affect serum lipids and NAFLD. We identified credible genes in these pathways and characterized missense variants in these for effects on serum traits. We used MAGENTA to identify 58 enriched pathways from publicly available TG, LDL, and HDL GWAS (n = 99,000). Three of these pathways were also enriched for associations with European-ancestry NAFLD GWAS (n = 7176). One pathway, farnesoid X receptor (FXR)/retinoid X receptor (RXR) activation, was replicated for association in an African-ancestry NAFLD GWAS (n = 3214) and plays a role in serum lipids and NAFLD. Credible genes (proteins) in FXR/RXR activation include those associated with cholesterol/bile/bilirubin transport/absorption (ABCC2 (MRP2) [ATP binding cassette subfamily C member (multidrug resistance-associated protein 2)], ABCG5, ABCG8 [ATP-binding cassette (ABC) transporters G5 and G8], APOB (APOB) [apolipoprotein B], FABP6 (ILBP) [fatty acid binding protein 6 (ileal lipid-binding protein)], MTTP (MTP) [microsomal triglyceride transfer protein], SLC4A2 (AE2) [solute carrier family 4 member 2 (anion exchange protein 2)]), nuclear hormone-mediated control of metabolism (NR0B2 (SHP) [nuclear receptor subfamily 0 group B member 2 (small heterodimer partner)], NR1H4 (FXR) [nuclear receptor subfamily 1 group H member 4 (FXR)], PPARA (PPAR) [peroxisome proliferator activated receptor alpha], FOXO1 (FOXO1A) [forkhead box O1]), or other pathways (FETUB (FETUB) [fetuin B]). Missense variants in ABCC2 (MRP2), ABCG5 (ABCG5), ABCG8 (ABCG8), APOB (APOB), MTTP (MTP), NR0B2 (SHP), NR1H4 (FXR), and PPARA (PPAR) that associate with serum LDL levels also associate with serum liver function tests in UK Biobank. Conclusion: Genetic variants in NR1H4 (FXR) that protect against liver steatosis increase serum LDL cholesterol while variants in other members of the family have congruent effects on these traits. Human genetic pathway enrichment analysis can help guide therapeutic development by identifying effective targets for NAFLD/serum lipid manipulation while minimizing side effects. In addition, missense variants could be used in companion diagnostics to determine their influence on drug effectiveness.
Collapse
Affiliation(s)
- Samuel K. Handelman
- Division of Gastroenterology and HepatologyUniversity of Michigan Health SystemAnn ArborMichiganUSA
| | - Yindra M. Puentes
- Division of Gastroenterology and HepatologyUniversity of Michigan Health SystemAnn ArborMichiganUSA
- Department of Computational Medicine and BioinformaticsUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| | - Annapurna Kuppa
- Division of Gastroenterology and HepatologyUniversity of Michigan Health SystemAnn ArborMichiganUSA
| | - Yanhua Chen
- Division of Gastroenterology and HepatologyUniversity of Michigan Health SystemAnn ArborMichiganUSA
| | - Xiaomeng Du
- Division of Gastroenterology and HepatologyUniversity of Michigan Health SystemAnn ArborMichiganUSA
| | - Mary F. Feitosa
- Division of Statistical Genomics, Department of GeneticsWashington UniversitySt. LouisMissouriUSA
| | - Nicholette D. Palmer
- Department of BiochemistryWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Elizabeth K. Speliotes
- Division of Gastroenterology and HepatologyUniversity of Michigan Health SystemAnn ArborMichiganUSA
- Department of Computational Medicine and BioinformaticsUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| |
Collapse
|
145
|
Churnosov M, Abramova M, Reshetnikov E, Lyashenko IV, Efremova O, Churnosova M, Ponomarenko I. Polymorphisms of hypertension susceptibility genes as a risk factors of preeclampsia in the Caucasian population of central Russia. Placenta 2022; 129:51-61. [PMID: 36219912 DOI: 10.1016/j.placenta.2022.09.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 08/18/2022] [Accepted: 09/14/2022] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The study was designed to assess the effects of hypertension (HT) susceptibility genes polymorphisms in the development of preeclampsia (PE) in Caucasians from Central Russia. METHODS PE patients (n = 452) and women control group (n = 498) were genotyped for 10 polymorphisms of HT/blood pressure (BP) susceptibility genes (according to the previously published GWAS in Caucasian populations) including AC026703.1 (rs1173771), HFE (rs1799945), BAG6 (rs805303), PLCE1 (rs932764), OBFC1 (rs4387287), ARHGAP42 (rs633185), CERS5 (rs7302981), ATP2B1 (rs2681472), TBX2 (rs8068318) and RGL3 (rs167479). A logistic regression method was applied to search for associations between SNPs and PE. The relationship between SNP-SNP interactions and PE risk was analyzed by performing MB-MDR. RESULTS The rs1799945 gene in HFE significantly independently increased the risk of developing PE (OR = 2.24) and rs805303 in BAG6 was associated with a reduced risk in the occurrence of PE (OR = 0.55-0.78). Among the 10 SNPs examined, nine SNPs were associated with PEs within the 10 most significant SNP-SNP interaction models. Loci rs7302981 CERS5, rs805303 BAG6 and rs932764 PLCE1 contributed to the largest number of epistatic models (50% or more). DISCUSSION The present study is the first to report an association between polymorphisms of HT/BP susceptibility genes important for GWAS and the risk of PE in Caucasians from Central Russia. Our pathway-based functional annotation of the PE risk variants highlights the potential regulatory function (epigenetic/eQTL/sQTL/non-synonymous) that nine genetic risk markers and their 115 highly correlated variants exert on 155 genes. The study shows that these genes may function cooperatively in key signaling pathways in PE biology.
Collapse
Affiliation(s)
- Mikhail Churnosov
- Belgorod State National Research University, Department of Medical Biological Disciplines, Belgorod, Russia.
| | - Maria Abramova
- Belgorod State National Research University, Department of Medical Biological Disciplines, Belgorod, Russia
| | - Evgeny Reshetnikov
- Belgorod State National Research University, Department of Medical Biological Disciplines, Belgorod, Russia
| | - Igor V Lyashenko
- Belgorod State National Research University, Department of English Philology and Cross-cultural Communication, Belgorod, Russia
| | - Olesya Efremova
- Kharkiv National Medical University, Department of Medical Genetics, Kharkov, Ukraine; Grishchenko Clinic of Reproductive Medicine, Kharkov, Ukraine
| | - Maria Churnosova
- Belgorod State National Research University, Department of Medical Biological Disciplines, Belgorod, Russia
| | - Irina Ponomarenko
- Belgorod State National Research University, Department of Medical Biological Disciplines, Belgorod, Russia
| |
Collapse
|
146
|
Nakaya N, Nakaya K, Tsuchiya N, Sone T, Kogure M, Hatanaka R, kanno I, Metoki H, Obara T, Ishikuro M, Hozawa A, Kuriyama S. Similarities in cardiometabolic risk factors among random male-female pairs: a large observational study in Japan. BMC Public Health 2022; 22:1978. [PMID: 36307801 PMCID: PMC9617423 DOI: 10.1186/s12889-022-14348-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 09/26/2022] [Accepted: 10/13/2022] [Indexed: 11/21/2022] Open
Abstract
Background: Previous observational studies have shown similarities in cardiometabolic risk factors between spouses. It is still possible that this result reflects the age similarity of spouses rather than environmental factors of spouses (e.g. cohabitation effect). To clarify the importance of mate cardiometabolic risk factors for similarity of environmental factors, it is necessary to examine whether they are observed in random male-female pairs while maintaining the age of the spousal pairs. This study aimed to determine whether the similarities found between spousal pairs for cardiometabolic risks were also observed between random male-female pairs. Methods: This cross-sectional study included 5,391 spouse pairs from Japan; data were obtained from a large biobank study. For pairings, women of the same age were randomly shuffled to create new male-female pairs of the same age as that of the original spouse pairs. Similarities in cardiometabolic risk factors between the random male-female pairs were analysed using Pearson’s correlation or age-adjusted logistic regression analyses. Results: The mean ages of the men and women were 63.2 and 60.4 years, respectively. Almost all cardiometabolic risk factors similarities were not noted in cardiometabolic risk factors, including the continuous risk factors (anthropometric traits, blood pressure, glycated haemoglobin level, and lipid traits); lifestyle habits (smoking, drinking, and physical activity); or diseases (hypertension, type 2 diabetes mellitus, and metabolic syndrome) between the random male-female pairs. The age-adjusted correlation coefficients ranged from − 0.007 for body mass index to 0.071 for total cholesterol. The age-adjusted odds ratio (95% confidence interval) for current drinkers was 0.94 (0.81 − 1.09); hypertension, 1.07 (0.93 − 1.23); and type 2 diabetes mellitus, 1.08 (0.77 − 1.50). Conclusion: In this study, few similarities in cardiometabolic risk factors were noted among the random male-female pairs. As spouse pairs may share environmental factors, intervention strategies targeting lifestyle habits and preventing lifestyle-related diseases may be effective.
Collapse
|
147
|
Lin C, Sun Z, Mei Z, Zeng H, Zhao M, Hu J, Xia M, Huang T, Wang C, Gao X, Zheng Y. The causal associations of circulating amino acids with blood pressure: a Mendelian randomization study. BMC Med 2022; 20:414. [PMID: 36307799 PMCID: PMC9615211 DOI: 10.1186/s12916-022-02612-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 10/17/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Circulating levels of amino acids were associated with blood pressure (BP) in observational studies. However, the causation of such associations has been hypothesized but is difficult to prove in human studies. Here, we aimed to use two-sample Mendelian randomization analyses to evaluate the potential causal associations of circulating levels of amino acids with BP and risk of hypertension. METHODS We generated genetic instruments for circulating levels of nine amino acids by conducting meta-analyses of genome-wide association study (GWAS) in UK Biobank participants with metabolomic data (n = 98,317) and another published metabolomics GWAS (n = 24,925). Data on the associations of the genetic variants with BP and hypertension were obtained in the UK Biobank participants without metabolomic data (n = 286,390). The causal effects were estimated using inverse-variance weighted method. RESULTS Significant evidence consistently supported the causal effects of increased branched-chain amino acids (BCAAs, i.e., leucine, isoleucine, and valine) levels on higher BP and risk of hypertension (all P < 0.006 after Bonferroni correction except for Pleucine-on-diastolicBP = 0.008). For example, per standard deviation higher of genetically predicted isoleucine levels were associated with 2.71 ± 0.78 mmHg higher systolic BP and 1.24 ± 0.34 mmHg higher diastolic BP, as well as with 7% higher risk of hypertension (odds ratio: 1.07, [95% CI: 1.04-1.10]). In addition, per standard deviation higher of genetically predicted glycine level was associated with lower systolic BP (- 0.70 ± 0.17 mmHg, P = 4.04 × 10-5) and a lower risk of hypertension (0.99 [0.98-0.99], P = 6.46 × 10-5). In the reverse direction, genetically predicted higher systolic BP was associated with lower circulating levels of glycine (- 0.025±0.008, P = 0.001). CONCLUSIONS This study provides evidence for causal impacts of genetically predicted circulating BCAAs and glycine levels on BP. Meanwhile, genetically predicted higher BP was associated with lower glycine levels. Further investigations are warranted to clarify the underlying mechanisms.
Collapse
Affiliation(s)
- Chenhao Lin
- State Key Laboratory of Genetic Engineering, Human Phenome Institute and School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200433, China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Zhonghan Sun
- State Key Laboratory of Genetic Engineering, Human Phenome Institute and School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200433, China
| | - Zhendong Mei
- State Key Laboratory of Genetic Engineering, Human Phenome Institute and School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200433, China
| | - Hailuan Zeng
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan Institute for Metabolic Diseases, and Human Phenome Institute, Fudan University, Shanghai, China
| | - Manying Zhao
- State Key Laboratory of Genetic Engineering, Human Phenome Institute and School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200433, China
| | - Jianying Hu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute and School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200433, China
| | - Mingfeng Xia
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan Institute for Metabolic Diseases, and Human Phenome Institute, Fudan University, Shanghai, China
| | - Tao Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Chaolong Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan Institute for Metabolic Diseases, and Human Phenome Institute, Fudan University, Shanghai, China
| | - Yan Zheng
- State Key Laboratory of Genetic Engineering, Human Phenome Institute and School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200433, China.
- Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China.
| |
Collapse
|
148
|
Alexander MR, Hank S, Dale BL, Himmel L, Zhong X, Smart CD, Fehrenbach DJ, Chen Y, Prabakaran N, Tirado B, Centrella M, Ao M, Du L, Shyr Y, Levy D, Madhur MS. A Single Nucleotide Polymorphism in SH2B3/LNK Promotes Hypertension Development and Renal Damage. Circ Res 2022; 131:731-747. [PMID: 36169218 PMCID: PMC9588739 DOI: 10.1161/circresaha.121.320625] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 09/15/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND SH2B3 (SH2B adaptor protein 3) is an adaptor protein that negatively regulates cytokine signaling and cell proliferation. A common missense single nucleotide polymorphism in SH2B3 (rs3184504) results in substitution of tryptophan (Trp) for arginine (Arg) at amino acid 262 and is a top association signal for hypertension in human genome-wide association studies. Whether this variant is causal for hypertension, and if so, the mechanism by which it impacts pathogenesis is unknown. METHODS We used CRISPR-Cas9 technology to create mice homozygous for the major (Arg/Arg) and minor (Trp/Trp) alleles of this SH2B3 polymorphism. Mice underwent angiotensin II (Ang II) infusion to evaluate differences in blood pressure (BP) elevation and end-organ damage including albuminuria and renal fibrosis. Cytokine production and Stat4 phosphorylation was also assessed in Arg/Arg and Trp/Trp T cells. RESULTS Trp/Trp mice exhibit 10 mmHg higher systolic BP during chronic Ang II infusion compared to Arg/Arg controls. Renal injury and perivascular fibrosis are exacerbated in Trp/Trp mice compared to Arg/Arg controls following Ang II infusion. Renal and ex vivo stimulated splenic CD8+ T cells from Ang II-infused Trp/Trp mice produce significantly more interferon gamma (IFNg) compared to Arg/Arg controls. Interleukin-12 (IL-12)-induced IFNg production is greater in Trp/Trp compared to Arg/Arg CD8+ T cells. In addition, IL-12 enhances Stat4 phosphorylation to a greater degree in Trp/Trp compared to Arg/Arg CD8+ T cells, suggesting that Trp-encoding SH2B3 exhibits less negative regulation of IL-12 signaling to promote IFNg production. Finally, we demonstrated that a multi-SNP model genetically predicting increased SH2B3 expression in lymphocytes is inversely associated with hypertension and hypertensive chronic kidney disease in humans.. CONCLUSIONS Taken together, these results suggest that the Trp encoding allele of rs3184504 is causal for BP elevation and renal dysfunction, in part through loss of SH2B3-mediated repression of T cell IL-12 signaling leading to enhanced IFNg production.
Collapse
Affiliation(s)
- Matthew R. Alexander
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Department of Medicine, Division of Cardiovascular Medicine, VUMC, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, TN, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Samuel Hank
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Bethany L. Dale
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Lauren Himmel
- Department of Pathology, Microbiology and Immunology, VUMC, Nashville, TN, USA
| | - Xue Zhong
- Department of Medicine, Division of Genetic Medicine, VUMC, Nashville, TN, USA
| | - Charles D. Smart
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Daniel J. Fehrenbach
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Yuhan Chen
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | | | | | - Megan Centrella
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Mingfang Ao
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Liping Du
- Department of Biostatistics, VUMC, Nashville, TN
| | - Yu Shyr
- Department of Biostatistics, VUMC, Nashville, TN
| | - Daniel Levy
- Framingham Heart Study, Framingham, MA and Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Meena S. Madhur
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Department of Medicine, Division of Cardiovascular Medicine, VUMC, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, TN, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
149
|
Portilla-Fernandez E, Klarin D, Hwang SJ, Biggs ML, Bis JC, Weiss S, Rospleszcz S, Natarajan P, Hoffmann U, Rogers IS, Truong QA, Völker U, Dörr M, Bülow R, Criqui MH, Allison M, Ganesh SK, Yao J, Waldenberger M, Bamberg F, Rice KM, Essers J, Kapteijn DMC, van der Laan SW, de Knegt RJ, Ghanbari M, Felix JF, Ikram MA, Kavousi M, Uitterlinden AG, Roks AJM, Danser AHJ, Tsao PS, Damrauer SM, Guo X, Rotter JI, Psaty BM, Kathiresan S, Völzke H, Peters A, Johnson C, Strauch K, Meitinger T, O’Donnell CJ, Dehghan A, VA Million Veteran Program. Genetic and clinical determinants of abdominal aortic diameter: genome-wide association studies, exome array data and Mendelian randomization study. Hum Mol Genet 2022; 31:3566-3579. [PMID: 35234888 PMCID: PMC9558840 DOI: 10.1093/hmg/ddac051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
Progressive dilation of the infrarenal aortic diameter is a consequence of the ageing process and is considered the main determinant of abdominal aortic aneurysm (AAA). We aimed to investigate the genetic and clinical determinants of abdominal aortic diameter (AAD). We conducted a meta-analysis of genome-wide association studies in 10 cohorts (n = 13 542) imputed to the 1000 Genome Project reference panel including 12 815 subjects in the discovery phase and 727 subjects [Partners Biobank cohort 1 (PBIO)] as replication. Maximum anterior-posterior diameter of the infrarenal aorta was used as AAD. We also included exome array data (n = 14 480) from seven epidemiologic studies. Single-variant and gene-based associations were done using SeqMeta package. A Mendelian randomization analysis was applied to investigate the causal effect of a number of clinical risk factors on AAD. In genome-wide association study (GWAS) on AAD, rs74448815 in the intronic region of LDLRAD4 reached genome-wide significance (beta = -0.02, SE = 0.004, P-value = 2.10 × 10-8). The association replicated in the PBIO1 cohort (P-value = 8.19 × 10-4). In exome-array single-variant analysis (P-value threshold = 9 × 10-7), the lowest P-value was found for rs239259 located in SLC22A20 (beta = 0.007, P-value = 1.2 × 10-5). In the gene-based analysis (P-value threshold = 1.85 × 10-6), PCSK5 showed an association with AAD (P-value = 8.03 × 10-7). Furthermore, in Mendelian randomization analyses, we found evidence for genetic association of pulse pressure (beta = -0.003, P-value = 0.02), triglycerides (beta = -0.16, P-value = 0.008) and height (beta = 0.03, P-value < 0.0001), known risk factors for AAA, consistent with a causal association with AAD. Our findings point to new biology as well as highlighting gene regions in mechanisms that have previously been implicated in the genetics of other vascular diseases.
Collapse
Affiliation(s)
- Eliana Portilla-Fernandez
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Derek Klarin
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Shih-Jen Hwang
- Population Sciences Branch, Division of Intramural Research, NHLBI/NIH, Bethesda MD, USA
- National Heart Lung and Blood Institute's Intramural Research Program's Framingham Heart Study, Framingham, MA, USA
| | - Mary L Biggs
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Stefan Weiss
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Susanne Rospleszcz
- Institute of Epidemiology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Pradeep Natarajan
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Udo Hoffmann
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Ian S Rogers
- Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA
| | - Quynh A Truong
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Marcus Dörr
- Department of Internal Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Robin Bülow
- Department of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Michael H Criqui
- Department of Family Medicine, University of California, San Diego, CA, USA
| | - Matthew Allison
- Department of Family Medicine, University of California, San Diego, CA, USA
| | - Santhi K Ganesh
- Department of Internal Medicine and Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Melanie Waldenberger
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Fabian Bamberg
- Department of Diagnostic and Interventional Radiology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kenneth M Rice
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Jeroen Essers
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Radiation Oncology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Vascular Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Daniek M C Kapteijn
- Laboratory of Experimental Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Sander W van der Laan
- Laboratory of Clinical Chemistry & Hematology, Division Laboratories, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Rob J de Knegt
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Janine F Felix
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Andre G Uitterlinden
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Anton J M Roks
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - A H Jan Danser
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Philip S Tsao
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
- School of Medicine, Stanford University, Stanford, CA, USA
| | - Scott M Damrauer
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Services, University of Washington, Seattle, WA, USA
| | - Sekar Kathiresan
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Henry Völzke
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
- Chair of Epidemiology, Institute for Medical Information Processing, Biometry, and Epidemiology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Craig Johnson
- Collaborative Health Studies Coordinating Center, Department of Biostatistics in the School of Public Health, University of Washington, Seattle, WA, USA
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
- Chair of Genetic Epidemiology, Institute for Medical Information Processing, Biometry, and Epidemiology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Thomas Meitinger
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Institute of Human Genetics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Human Genetics, Technische Universität München, München, Germany
| | - Christopher J O’Donnell
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | | |
Collapse
|
150
|
Jain VG, Monangi N, Zhang G, Muglia LJ. Genetics, epigenetics, and transcriptomics of preterm birth. Am J Reprod Immunol 2022; 88:e13600. [PMID: 35818963 PMCID: PMC9509423 DOI: 10.1111/aji.13600] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/13/2022] [Accepted: 07/06/2022] [Indexed: 11/29/2022] Open
Abstract
Preterm birth contributes significantly to neonatal mortality and morbidity. Despite its global significance, there has only been limited progress in preventing preterm birth. Spontaneous preterm birth (sPTB) results from a wide variety of pathological processes. Although many non-genetic risk factors influence the timing of gestation and labor, compelling evidence supports the role of substantial genetic and epigenetic influences and their interactions with the environment contributing to sPTB. To investigate a common and complex disease such as sPTB, various approaches such as genome-wide association studies, whole-exome sequencing, transcriptomics, and integrative approaches combining these with other 'omics studies have been used. However, many of these studies were typically small or focused on a single ethnicity or geographic region with limited data, particularly in populations at high risk for sPTB, or lacked a robust replication. These studies found many genes involved in the inflammation and immunity-related pathways that may affect sPTB. Recent studies also suggest the role of epigenetic modifications of gene expression by the environmental signals as a potential contributor to the risk of sPTB. Future genetic studies of sPTB should continue to consider the contributions of both maternal and fetal genomes as well as their interaction with the environment.
Collapse
Affiliation(s)
- Viral G. Jain
- Division of Neonatology, Department of Pediatrics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nagendra Monangi
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center and March of Dimes Prematurity Research Center Ohio Collaborative, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Ge Zhang
- Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center and March of Dimes Prematurity Research Center Ohio Collaborative, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Louis J. Muglia
- Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center and March of Dimes Prematurity Research Center Ohio Collaborative, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- Burroughs Wellcome Fund, Research Triangle Park, North Carolina, USA
| |
Collapse
|