201
|
|
202
|
Abstract
In this paper, we propose two mixed-effects models for Genetic Analysis Workshop 18 (GAW18) longitudinal blood pressure data. The first method extends EMMA, an efficient mixed-model association-mapping algorithm. EMMA corrects for population structure and genetic relatedness using a kinship similarity matrix. We replace the kinship similarity matrix in EMMA with an estimated correlation matrix for modeling the dependence structure of repeated measurements. Our second approach is a Bayesian multiple association-mapping algorithm based on a mixed-effects model with a built-in variable selection feature. It models multiple single-nucleotide polymorphisms (SNPs) simultaneously and allows for SNP-SNP interactions and SNP-environment interactions. We applied these two methods to the longitudinal systolic blood pressure (SBP) and diastolic blood pressure (DBP) data from GAW18. The extended EMMA method identified a single SNP on Chr5:75506197 (p-value = 4.67 × 10(-7)) for SBP and three SNPs on Chr3:23715851 (p-value = 9.00 × 10(-8)), Chr 17:54834217 (p-value = 1.98 × 10(-7)), and Chr21:18744081 (p-value = 4.95 × 10(-7)) for DBP. The Bayesian method identified several additional SNPs on Chr1:17876090 (Bayes factor [BF] = 102), Chr3:197469358 (BF = 69), Chr15:87675666 (BF = 43), and Chr19:41642807 (BF = 33) for SBP. Furthermore, for SBP, we found a single SNP on Chr3:197469358 (BF = 69) that has a strong interaction with age. We further evaluated the performances of the proposed methods by simulations.
Collapse
Affiliation(s)
- Wonil Chung
- Department of Biostatistics, University of North Carolina at Chapel Hill, 3101 McGavran-Greenberg Hall, Chapel Hill, NC 27599, USA
| | - Fei Zou
- Department of Biostatistics, University of North Carolina at Chapel Hill, 3101 McGavran-Greenberg Hall, Chapel Hill, NC 27599, USA
| |
Collapse
|
203
|
Chiang KM, Yang HC, Liang YJ, Chen JW, Hwang SM, Ho HY, Ting CT, Lin TH, Sheu SH, Tsai WC, Chen JH, Leu HB, Yin WH, Chiu TY, Chen CI, Lin SJ, Thomas GN, Tomlinson B, Guo Y, Gui HS, Sham PC, Lam TH, Pan WH. A three-stage genome-wide association study combining multilocus test and gene expression analysis for young-onset hypertension in Taiwan Han Chinese. Am J Hypertens 2014; 27:819-27. [PMID: 24413707 DOI: 10.1093/ajh/hpt239] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Although many large-scale genome-wide association studies (GWASs) have been performed, only a few studies have successfully identified replicable, large-impact hypertension loci; even fewer studies have been done on Chinese subjects. Young-onset hypertension (YOH) is considered to be a more promising target disorder to investigate than late-onset hypertension because of its stronger genetic component. METHODS To map YOH genetic variants, we performed a 3-stage study combining 1st-stage multilocus GWASs, 2nd-stage gene expression analysis, and 3rd-stage multilocus confirmatory study. RESULTS In the 1st stage, Illumina550K data from 400 case-control pairs were used, and 22 genes flanked by 14 single nucleotide polymorphism (SNP) septets (P values adjusted for false discovery rate (pFDR) < 3.16×10(-7)) were identified. In the 2nd stage, differential gene expression analysis was carried out for these genes, and 5 genes were selected (pFDR < 0.05). In the 3rd stage, we re-examined the finding with an independent set of 592 case-control pairs and with the joint samples (n = 992 case-control pairs). A total of 6 SNP septets flanking C1orf135, GSN, LARS, and ACTN4 remained significant in all 3 stages. Among them, the same septet flanking ACTN4 was also associated with blood pressure traits in the Hong Kong Hypertension Study (HKHS) and in the Wellcome Trust Case-Control Consortium Hypertension Study (WTCCCHS). LARS was detected in the HKHS, but not in the WTCCCHS. GSN may be specific to Taiwanese individuals because it was not found by either the HKHS or the WTCCCHS. CONCLUSIONS Our study identified 4 previously unknown YOH loci in Han Chinese. Identification of these genes enriches the hypertension susceptibility gene list, thereby shedding light on the etiology of hypertension in Han Chinese.
Collapse
Affiliation(s)
- Kuang-Mao Chiang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
204
|
He D, Fu M, Miao S, Hotta K, Chandak GR, Xi B. FTO gene variant and risk of hypertension: a meta-analysis of 57,464 hypertensive cases and 41,256 controls. Metabolism 2014; 63:633-9. [PMID: 24641884 DOI: 10.1016/j.metabol.2014.02.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 02/09/2014] [Accepted: 02/10/2014] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Recent studies have suggested that fat mass and obesity-associated gene (FTO) may predispose individuals to develop hypertension. However, the results have been inconsistent. We performed a meta-analysis to investigate the relationship of FTO gene variant with risk of hypertension and influence of body mass index (BMI) on this risk. MATERIALS/METHODS A systematic literature search in PubMed, Embase and ISI web of science databases was performed to identify eligible published literatures. Pooled odds ratios (ORs) with 95% confidence intervals (CIs) were calculated. RESULTS A total of seven studies comprising 57,464 hypertensive cases and 41,256 controls met the inclusion criteria and were included in the meta-analysis. The FTO gene variant(s) showed significant association with the risk of hypertension (OR=1.16, 95% CI=1.07-1.25, P<0.001) which disappeared on adjustment for BMI (OR=1.04, 95% CI=0.98-1.10, P=0.162). In addition, stratified analysis demonstrated a significant association of the FTO variant with the risk of hypertension in obese subjects (OR=1.10, 95% CI=1.01-1.19, P=0.032) but not in non-obese individuals (OR=1.00, 95% CI=0.97-1.03, P=0.832). Subgroup analysis based on ethnicity showed significant association between FTO variant and hypertension in both European (OR=1.07, 95% CI=1.01-1.14, P=0.028) and Asian populations (OR=1.37, 95% CI=1.23-1.53, P<0.001). However, the association remained significant only in Asians (OR=1.17, 95% CI=1.01-1.35, P=0.035) but not in the Europeans (OR=1.02, 95% CI=0.97-1.07, P=0.390) on adjustment for BMI. CONCLUSIONS The present meta-analysis confirms that FTO genotype mediates obesity-related hypertension.
Collapse
Affiliation(s)
- Dan He
- Department of Maternal and Child Health Care, School of Public Health, Shandong University, Jinan, People's Republic of China
| | - Maosun Fu
- Department of Maternal and Child Health Care, School of Public Health, Shandong University, Jinan, People's Republic of China
| | - Song Miao
- Shandong Medical College (Jinan), Jinan, People's Republic of China
| | - Kikuko Hotta
- EBM Research Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Bo Xi
- Department of Maternal and Child Health Care, School of Public Health, Shandong University, Jinan, People's Republic of China.
| |
Collapse
|
205
|
ENGER TB, PLEYM H, STENSETH R, WAHBA A, VIDEM V. Genetic and clinical risk factors for fluid overload following open-heart surgery. Acta Anaesthesiol Scand 2014; 58:539-48. [PMID: 24628133 DOI: 10.1111/aas.12310] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2014] [Indexed: 11/29/2022]
Abstract
BACKGROUND Post-operative fluid overload following cardiac surgery is associated with increased morbidity and mortality. We hypothesised that genetic variations and pre-operative clinical factors predispose some patients to post-operative fluid overload. METHODS Perioperative variables were collected prospectively for 1026 consecutive adults undergoing open-heart surgery at St. Olavs University Hospital, Norway from 2008-2010. Post-operative fluid overload was defined as a post-operative fluid balance/kg ≥ the 90th percentile of the study population. Genotyping was performed for 31 single-nucleotide polymorphisms related to inflammatory/vascular responses or previously associated with complications following open-heart surgery. Data were analysed using logistic regression modelling, and the findings were internally validated by bootstrapping (n = 100). RESULTS Homozygous carriers of the common G allele of rs12917707 in the UMOD gene had a 2.2 times greater risk of post-operative fluid overload (P = 0.005) after adjustment for significant clinical variables (age, duration of cardiopulmonary bypass, and intraoperative red cell transfusion). A genetic risk score including 14 single-nucleotide polymorphisms was independently associated with post-operative fluid overload (P = 0.001). The number of risk alleles was linearly associated with the frequency of fluid overload (odds ratio per risk allele 1.153, 95 % confidence interval 1.056-1.258). Nagelkerke's R(2) increased with 7.5% to a total of 25% for the combined clinical and genetic model. Hemofiltration did not reduce the risk. CONCLUSION A common variation in the UMOD gene previously shown to be related to renal function was associated with increased risk of post-operative fluid overload following cardiac surgery. Our findings support a genetic susceptibility to disturbed fluid handling following cardiac surgery.
Collapse
Affiliation(s)
- T. B. ENGER
- Department of Laboratory Medicine, Children's and Women's Health; Faculty of Medicine; Norwegian University of Science and Technology; Trondheim Norway
| | - H. PLEYM
- Department of Circulation and Medical Imaging; Faculty of Medicine; Norwegian University of Science and Technology; Trondheim Norway
- Department of Cardiothoracic Anaesthesia and Intensive Care; St. Olavs University Hospital; Trondheim Norway
| | - R. STENSETH
- Department of Circulation and Medical Imaging; Faculty of Medicine; Norwegian University of Science and Technology; Trondheim Norway
- Department of Cardiothoracic Anaesthesia and Intensive Care; St. Olavs University Hospital; Trondheim Norway
| | - A. WAHBA
- Department of Circulation and Medical Imaging; Faculty of Medicine; Norwegian University of Science and Technology; Trondheim Norway
- Department of Cardiothoracic Surgery; St. Olavs University Hospital; Trondheim Norway
| | - V. VIDEM
- Department of Laboratory Medicine, Children's and Women's Health; Faculty of Medicine; Norwegian University of Science and Technology; Trondheim Norway
- Department of Immunology and Transfusion Medicine; St. Olavs University Hospital; Trondheim Norway
| |
Collapse
|
206
|
Simino J, Shi G, Weder A, Boerwinkle E, Hunt SC, Rao DC. Body mass index modulates blood pressure heritability: the Family Blood Pressure Program. Am J Hypertens 2014; 27:610-9. [PMID: 24029162 PMCID: PMC3958601 DOI: 10.1093/ajh/hpt144] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 07/22/2013] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Candidate gene and twin studies suggest that interactions between body mass index (BMI) and genes contribute to the variability of blood pressure (BP). To determine whether there is evidence for gene-BMI interactions, we investigated the modulation of BP heritability by BMI using 4,153 blacks, 1,538 Asians, 4,013 whites, and 2,199 Hispanic Americans from the Family Blood Pressure Program. METHODS To capture the BP heritability dependence on BMI, we employed a generalized variance components model incorporating linear and Gaussian interactions between BMI and the genetic component. Within each race and network subgroup, we used the Akaike information criterion and likelihood ratio test to select the appropriate interaction function for each BP trait (systolic BP (SBP), diastolic BP (DBP), mean arterial pressure (MAP), and pulse pressure (PP)) and determine interaction significance, respectively. RESULTS BP heritabilities were significantly modified by BMI in the GenNet and SAPPHIRe Networks, which contained the youngest and least-obese participants, respectively. GenNet Whites had unimodal SBP, MAP, and PP heritabilities that peaked between BMI values of 33 and 37kg/m(2). The SBP and MAP heritabilities in GenNet Hispanic Americans, as well as the PP heritability in GenNet blacks, were increasing functions of BMI. The DBP and SBP heritabilities in the SAPPHIRe Chinese and Japanese, respectively, were decreasing functions of BMI. CONCLUSIONS BP heritability differed by BMI in the youngest and least-obese networks, although the shape of this dependence differed by race. Use of nonlinear gene-BMI interactions may enhance BP gene discovery efforts in individuals of European ancestry.
Collapse
Affiliation(s)
- Jeannette Simino
- Division of Biostatistics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Gang Shi
- Division of Biostatistics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Alan Weder
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Sciences Center, Houston, Texas
| | - Steven C. Hunt
- Cardiovascular Genetics Division, Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Dabeeru C. Rao
- Division of Biostatistics, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| |
Collapse
|
207
|
Rampoldi L, Köttgen A, Devuyst O. The effect of common uromodulin variants on urinary protein level and gene transcription. Kidney Int 2014; 84:410-1. [PMID: 23903422 DOI: 10.1038/ki.2013.162] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
208
|
Tragante V, Barnes MR, Ganesh SK, Lanktree MB, Guo W, Franceschini N, Smith EN, Johnson T, Holmes MV, Padmanabhan S, Karczewski KJ, Almoguera B, Barnard J, Baumert J, Chang YPC, Elbers CC, Farrall M, Fischer ME, Gaunt TR, Gho JMIH, Gieger C, Goel A, Gong Y, Isaacs A, Kleber ME, Mateo Leach I, McDonough CW, Meijs MFL, Melander O, Nelson CP, Nolte IM, Pankratz N, Price TS, Shaffer J, Shah S, Tomaszewski M, van der Most PJ, Van Iperen EPA, Vonk JM, Witkowska K, Wong COL, Zhang L, Beitelshees AL, Berenson GS, Bhatt DL, Brown M, Burt A, Cooper-DeHoff RM, Connell JM, Cruickshanks KJ, Curtis SP, Davey-Smith G, Delles C, Gansevoort RT, Guo X, Haiqing S, Hastie CE, Hofker MH, Hovingh GK, Kim DS, Kirkland SA, Klein BE, Klein R, Li YR, Maiwald S, Newton-Cheh C, O'Brien ET, Onland-Moret NC, Palmas W, Parsa A, Penninx BW, Pettinger M, Vasan RS, Ranchalis JE, M Ridker P, Rose LM, Sever P, Shimbo D, Steele L, Stolk RP, Thorand B, Trip MD, van Duijn CM, Verschuren WM, Wijmenga C, Wyatt S, Young JH, Zwinderman AH, Bezzina CR, Boerwinkle E, Casas JP, Caulfield MJ, Chakravarti A, Chasman DI, Davidson KW, Doevendans PA, Dominiczak AF, FitzGerald GA, Gums JG, Fornage M, et alTragante V, Barnes MR, Ganesh SK, Lanktree MB, Guo W, Franceschini N, Smith EN, Johnson T, Holmes MV, Padmanabhan S, Karczewski KJ, Almoguera B, Barnard J, Baumert J, Chang YPC, Elbers CC, Farrall M, Fischer ME, Gaunt TR, Gho JMIH, Gieger C, Goel A, Gong Y, Isaacs A, Kleber ME, Mateo Leach I, McDonough CW, Meijs MFL, Melander O, Nelson CP, Nolte IM, Pankratz N, Price TS, Shaffer J, Shah S, Tomaszewski M, van der Most PJ, Van Iperen EPA, Vonk JM, Witkowska K, Wong COL, Zhang L, Beitelshees AL, Berenson GS, Bhatt DL, Brown M, Burt A, Cooper-DeHoff RM, Connell JM, Cruickshanks KJ, Curtis SP, Davey-Smith G, Delles C, Gansevoort RT, Guo X, Haiqing S, Hastie CE, Hofker MH, Hovingh GK, Kim DS, Kirkland SA, Klein BE, Klein R, Li YR, Maiwald S, Newton-Cheh C, O'Brien ET, Onland-Moret NC, Palmas W, Parsa A, Penninx BW, Pettinger M, Vasan RS, Ranchalis JE, M Ridker P, Rose LM, Sever P, Shimbo D, Steele L, Stolk RP, Thorand B, Trip MD, van Duijn CM, Verschuren WM, Wijmenga C, Wyatt S, Young JH, Zwinderman AH, Bezzina CR, Boerwinkle E, Casas JP, Caulfield MJ, Chakravarti A, Chasman DI, Davidson KW, Doevendans PA, Dominiczak AF, FitzGerald GA, Gums JG, Fornage M, Hakonarson H, Halder I, Hillege HL, Illig T, Jarvik GP, Johnson JA, Kastelein JJP, Koenig W, Kumari M, März W, Murray SS, O'Connell JR, Oldehinkel AJ, Pankow JS, Rader DJ, Redline S, Reilly MP, Schadt EE, Kottke-Marchant K, Snieder H, Snyder M, Stanton AV, Tobin MD, Uitterlinden AG, van der Harst P, van der Schouw YT, Samani NJ, Watkins H, Johnson AD, Reiner AP, Zhu X, de Bakker PIW, Levy D, Asselbergs FW, Munroe PB, Keating BJ. Gene-centric meta-analysis in 87,736 individuals of European ancestry identifies multiple blood-pressure-related loci. Am J Hum Genet 2014; 94:349-60. [PMID: 24560520 DOI: 10.1016/j.ajhg.2013.12.016] [Show More Authors] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 12/20/2013] [Indexed: 11/29/2022] Open
Abstract
Blood pressure (BP) is a heritable risk factor for cardiovascular disease. To investigate genetic associations with systolic BP (SBP), diastolic BP (DBP), mean arterial pressure (MAP), and pulse pressure (PP), we genotyped ~50,000 SNPs in up to 87,736 individuals of European ancestry and combined these in a meta-analysis. We replicated findings in an independent set of 68,368 individuals of European ancestry. Our analyses identified 11 previously undescribed associations in independent loci containing 31 genes including PDE1A, HLA-DQB1, CDK6, PRKAG2, VCL, H19, NUCB2, RELA, HOXC@ complex, FBN1, and NFAT5 at the Bonferroni-corrected array-wide significance threshold (p < 6 × 10(-7)) and confirmed 27 previously reported associations. Bioinformatic analysis of the 11 loci provided support for a putative role in hypertension of several genes, such as CDK6 and NUCB2. Analysis of potential pharmacological targets in databases of small molecules showed that ten of the genes are predicted to be a target for small molecules. In summary, we identified previously unknown loci associated with BP. Our findings extend our understanding of genes involved in BP regulation, which may provide new targets for therapeutic intervention or drug response stratification.
Collapse
Affiliation(s)
- Vinicius Tragante
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands; Department of Medical Genetics, Biomedical Genetics, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Michael R Barnes
- William Harvey Research Institute National Institute for Health Biomedical Research Unit, Barts and the London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK
| | - Santhi K Ganesh
- Division of Cardiovascular Medicine, Departments of Internal Medicine and Human Genetics, University of Michigan Health System, Ann Arbor, MI 48109, USA
| | - Matthew B Lanktree
- Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Wei Guo
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Erin N Smith
- Department of Pediatrics and Rady's Children's Hospital, University of California at San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Toby Johnson
- Clinical Pharmacology and Barts and The London Genome Centre, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Michael V Holmes
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sandosh Padmanabhan
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, UK
| | - Konrad J Karczewski
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Berta Almoguera
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - John Barnard
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jens Baumert
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Yen-Pei Christy Chang
- Departments of Medicine and Epidemiology & Public Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Clara C Elbers
- Department of Medical Genetics, Biomedical Genetics, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands; Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Martin Farrall
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Mary E Fischer
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI 53726, USA
| | - Tom R Gaunt
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Johannes M I H Gho
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Anuj Goel
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Yan Gong
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL 32610, USA
| | - Aaron Isaacs
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center, 3015 GE Rotterdam, the Netherlands
| | - Marcus E Kleber
- Medical Clinic V, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Irene Mateo Leach
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - Caitrin W McDonough
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL 32610, USA
| | - Matthijs F L Meijs
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Olle Melander
- Hypertension and Cardiovascular Disease, Department of Clinical Sciences, Lund University, Malmö 20502, Sweden; Centre of Emergency Medicine, Skåne University Hospital, Malmö 20502, Sweden
| | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester, Leicester LE3 9QP, UK; NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Ilja M Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - Nathan Pankratz
- Institute of Human Genetics, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Tom S Price
- MRC SGDP Centre, Institute of Psychiatry, London SE5 8AF, UK
| | - Jonathan Shaffer
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Sonia Shah
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, Kathleen Lonsdale Building, Gower Place, London WC1E 6BT, UK
| | - Maciej Tomaszewski
- Department of Cardiovascular Sciences, University of Leicester, Leicester LE3 9QP, UK
| | - Peter J van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - Erik P A Van Iperen
- Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, 3511 GC Utrecht, the Netherlands; Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, 1105 AZ Amsterdam, the Netherlands
| | - Judith M Vonk
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - Kate Witkowska
- Clinical Pharmacology and Barts and The London Genome Centre, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Caroline O L Wong
- Clinical Pharmacology and Barts and The London Genome Centre, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Li Zhang
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Amber L Beitelshees
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Gerald S Berenson
- Department of Epidemiology, Tulane University, New Orleans, LA 70118, USA
| | - Deepak L Bhatt
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Morris Brown
- Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Hills Road, Cambridge CB2 2QQ, UK
| | - Amber Burt
- Department of Medicine (Medical Genetics), University of Washington, Seattle, WA 98195, USA
| | - Rhonda M Cooper-DeHoff
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL 32610, USA
| | - John M Connell
- University of Dundee, Ninewells Hospital &Medical School, Dundee DD1 9SY, UK
| | - Karen J Cruickshanks
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI 53726, USA; Department of Population Health Sciences, University of Wisconsin, Madison, WI 53726, USA
| | - Sean P Curtis
- Merck Research Laboratories, P.O. Box 2000, Rahway, NJ 07065, USA
| | - George Davey-Smith
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Ron T Gansevoort
- Division of Nephrology, Department of Medicine, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - Xiuqing Guo
- Cedars-Sinai Med Ctr-PEDS, Los Angeles, CA 90048, USA
| | - Shen Haiqing
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Claire E Hastie
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Marten H Hofker
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands; Department Pathology and Medical Biology, Medical Biology Division, Molecular Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - G Kees Hovingh
- Department of Vascular Medicine, Academic Medical Center, 1105 AZ Amsterdam, the Netherlands
| | - Daniel S Kim
- Departments of Medicine (Medical Genetics) and Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Susan A Kirkland
- Department of Community Health and Epidemiology, Dalhousie University, Halifax, NS B3H 1V7, Canada
| | - Barbara E Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI 53726, USA
| | - Ronald Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI 53726, USA
| | - Yun R Li
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Steffi Maiwald
- Department of Vascular Medicine, Academic Medical Center, 1105 AZ Amsterdam, the Netherlands
| | | | - Eoin T O'Brien
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - N Charlotte Onland-Moret
- Department of Medical Genetics, Biomedical Genetics, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands; Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Walter Palmas
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Afshin Parsa
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Brenda W Penninx
- Department of Psychiatry/EMGO Institute, VU University Medical Centre, 1081 BT Amsterdam, the Netherlands
| | - Mary Pettinger
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Ramachandran S Vasan
- Department of Medicine, Boston University School of Medicine, Framingham, MA 02118, USA
| | - Jane E Ranchalis
- Department of Medicine (Medical Genetics), University of Washington, Seattle, WA 98195, USA
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Lynda M Rose
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Peter Sever
- International Centre for Circulatory Health, Imperial College London, W2 1LA UK
| | - Daichi Shimbo
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Laura Steele
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ronald P Stolk
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - Barbara Thorand
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Mieke D Trip
- Department of Cardiology, Academic Medical Center, 1105 AZ Amsterdam, the Netherlands
| | - Cornelia M van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center, 3015 GE Rotterdam, the Netherlands
| | - W Monique Verschuren
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands; National Institute for Public Health and the Environment (RIVM), 3720 BA Bilthoven, the Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - Sharon Wyatt
- Schools of Nursing and Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - J Hunter Young
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Aeilko H Zwinderman
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, 1105 AZ Amsterdam, the Netherlands
| | - Connie R Bezzina
- Heart Failure Research Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, 1105 AZ Amsterdam, the Netherlands; Molecular and Experimental Cardiology Group, Academic Medical Centre, 1105 AZ Amsterdam, the Netherlands
| | - Eric Boerwinkle
- Human Genetics Center and Institute of Molecular Medicine and Division of Epidemiology, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Juan P Casas
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK; Genetic Epidemiology Group, Department of Epidemiology and Public Health, University College London, London WC1E 6BT, UK
| | - Mark J Caulfield
- Clinical Pharmacology and Barts and The London Genome Centre, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Aravinda Chakravarti
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Karina W Davidson
- Departments of Medicine & Psychiatry, Columbia University, New York, NY 10032, USA
| | - Pieter A Doevendans
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Anna F Dominiczak
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, UK
| | - Garret A FitzGerald
- The Institute for Translational Medicine and Therapeutics, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John G Gums
- Departments of Pharmacotherapy and Translational Research and Community Health and Family Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Myriam Fornage
- Institute of Molecular Medicine and School of Public Health Division of Epidemiology Human Genetics and Environmental Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Indrani Halder
- School of Medicine, University of Pittsburgh, PA 15261, USA
| | - Hans L Hillege
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - Thomas Illig
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany; Hannover Unified Biobank, Hannover Medical School, Hannover 30625, Germany
| | - Gail P Jarvik
- International Centre for Circulatory Health, Imperial College London, W2 1LA UK
| | - Julie A Johnson
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL 32610, USA
| | - John J P Kastelein
- Department of Vascular Medicine, Academic Medical Center, 1105 AZ Amsterdam, the Netherlands
| | - Wolfgang Koenig
- Department of Internal Medicine II - Cardiology, University of Ulm Medical Centre, Ulm 89081, Germany
| | - Meena Kumari
- Department of Epidemiology and Public Health, Division of Population Health, University College London, Torrington Place, London WC1E 7HB, UK
| | - Winfried März
- Medical Clinic V, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany; Synlab Academy, Synlab Services GmbH, Mannheim 69214, Germany; Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz 8036, Austria
| | - Sarah S Murray
- Department of Pathology, University of California San Diego, La Jolla, CA 92037, USA
| | - Jeffery R O'Connell
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Albertine J Oldehinkel
- Interdisciplinary Center Psychopathology and Emotion Regulation, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - James S Pankow
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN 55454, USA
| | - Daniel J Rader
- Cardiovascular Institute, the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Susan Redline
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Muredach P Reilly
- Cardiovascular Institute, the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eric E Schadt
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - Michael Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alice V Stanton
- Molecular & Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland
| | - Martin D Tobin
- Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - André G Uitterlinden
- Departments of Epidemiology and Internal Medicine, Erasmus Medical Center, 3015 GE Rotterdam, the Netherlands
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands; Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, 3511 GC Utrecht, the Netherlands; Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - Yvonne T van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester LE3 9QP, UK; NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Hugh Watkins
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Andrew D Johnson
- National Heart, Lung and Blood Institute Framingham Heart Study, Framingham, MA 01702, USA
| | - Alex P Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Xiaofeng Zhu
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Paul I W de Bakker
- Department of Medical Genetics, Biomedical Genetics, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands; Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands; Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA and Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Daniel Levy
- Center for Population Studies, National Heart, Lung, and Blood Institute, Framingham, MA 01702, USA
| | - Folkert W Asselbergs
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands; Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, 3511 GC Utrecht, the Netherlands; Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London WC1E 6BT, UK
| | - Patricia B Munroe
- Clinical Pharmacology and Barts and The London Genome Centre, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Brendan J Keating
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
209
|
Graham LA, Padmanabhan S, Fraser NJ, Kumar S, Bates JM, Raffi HS, Welsh P, Beattie W, Hao S, Leh S, Hultstrom M, Ferreri NR, Dominiczak AF, Graham D, McBride MW. Validation of Uromodulin as a Candidate Gene for Human Essential Hypertension. Hypertension 2014; 63:551-8. [DOI: 10.1161/hypertensionaha.113.01423] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A recent genome-wide association study identified a locus on chromosome 16 in the promoter region of the uromodulin (
UMOD
) gene that is associated with hypertension. Here, we examined the hypertension signal with functional studies in Umod knockout (KO) mice. Systolic blood pressure was significantly lower in KO versus wild-type (WT) mice under basal conditions (KO: 116.6±0.3 mm Hg versus WT: 136.2±0.4 mm Hg;
P
<0.0001). Administration of 2% NaCl did not alter systolic blood pressure in KO mice, whereas it increased in WT mice by ≈33%,
P
<0.001. The average 24-hour urinary sodium excretion in the KO was greater than that of WT mice (
P
<0.001). Chronic renal function curves demonstrate a leftward shift in KO mice, suggesting that the relationship between UMOD and blood pressure is affected by sodium. Creatinine clearance was increased during salt loading with 2% NaCl in the KO mice, leading to augmented filtered Na
+
excretion and further Na
+
loss. The difference in sodium uptake that exists between WT and KO strains was explored at the molecular level. Urinary tumor necrosis factor-α levels were significantly higher in KO mice compared with WT mice (
P
<0.0001). Stimulation of primary thick ascending limb of the loop of Henle cells with exogenous tumor necrosis factor-α caused a reduction in NKCC2A expression (
P
<0.001) with a concurrent rise in the levels of UMOD mRNA (
P
<0.001). Collectively, we demonstrate that UMOD regulates sodium uptake in the thick ascending limb of the loop of Henle by modulating the effect of tumor necrosis factor-α on NKCC2A expression, making UMOD an important determinant of blood pressure control.
Collapse
Affiliation(s)
- Lesley A. Graham
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Sandosh Padmanabhan
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Niall J. Fraser
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Satish Kumar
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - James M. Bates
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Hajamohideen S. Raffi
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Paul Welsh
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Wendy Beattie
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Shoujin Hao
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Sabine Leh
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Michael Hultstrom
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Nicholas R. Ferreri
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Anna F. Dominiczak
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Delyth Graham
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Martin W. McBride
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
210
|
Kelishadi R, Poursafa P. A review on the genetic, environmental, and lifestyle aspects of the early-life origins of cardiovascular disease. Curr Probl Pediatr Adolesc Health Care 2014; 44:54-72. [PMID: 24607261 DOI: 10.1016/j.cppeds.2013.12.005] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 12/04/2013] [Indexed: 12/29/2022]
Abstract
This article is a comprehensive review on developmental origins of health and disease regarding various factors related to the origins of cardiovascular diseases from early life. It presents a summary of the impacts of various factors such as epigenetics; gene-environment interaction; ethnic predisposition to cardiovascular diseases and their underlying risk factors; prenatal factors; fetal programming; maternal weight status and weight gain during pregnancy; type of feeding during infancy; growth pattern during childhood; obesity; stunting; socioeconomic status; dietary and physical activity habits; active, secondhand, and thirdhand smoking, as well as environmental factors including air pollution and global climate change on the development and progress of cardiovascular diseases and their risk factors. The importance of early identification of predisposing factors for cardiovascular diseases for primordial and primary prevention of cardiovascular diseases from early life is highlighted.
Collapse
Affiliation(s)
- Roya Kelishadi
- Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Child Growth and Development Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parinaz Poursafa
- Environment Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
211
|
Olden M, Corre T, Hayward C, Toniolo D, Ulivi S, Gasparini P, Pistis G, Hwang SJ, Bergmann S, Campbell H, Cocca M, Gandin I, Girotto G, Glaudemans B, Hastie ND, Loffing J, Polasek O, Rampoldi L, Rudan I, Sala C, Traglia M, Vollenweider P, Vuckovic D, Youhanna S, Weber J, Wright AF, Kutalik Z, Bochud M, Fox CS, Devuyst O. Common variants in UMOD associate with urinary uromodulin levels: a meta-analysis. J Am Soc Nephrol 2014; 25:1869-82. [PMID: 24578125 DOI: 10.1681/asn.2013070781] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Uromodulin is expressed exclusively in the thick ascending limb and is the most abundant protein excreted in normal urine. Variants in UMOD, which encodes uromodulin, are associated with renal function, and urinary uromodulin levels may be a biomarker for kidney disease. However, the genetic factors regulating uromodulin excretion are unknown. We conducted a meta-analysis of urinary uromodulin levels to identify associated common genetic variants in the general population. We included 10,884 individuals of European descent from three genetic isolates and three urban cohorts. Each study measured uromodulin indexed to creatinine and conducted linear regression analysis of approximately 2.5 million single nucleotide polymorphisms using an additive model. We also tested whether variants in genes expressed in the thick ascending limb associate with uromodulin levels. rs12917707, located near UMOD and previously associated with renal function and CKD, had the strongest association with urinary uromodulin levels (P<0.001). In all cohorts, carriers of a G allele of this variant had higher uromodulin levels than noncarriers did (geometric means 10.24, 14.05, and 17.67 μg/g creatinine for zero, one, or two copies of the G allele). rs12446492 in the adjacent gene PDILT (protein disulfide isomerase-like, testis expressed) also reached genome-wide significance (P<0.001). Regarding genes expressed in the thick ascending limb, variants in KCNJ1, SORL1, and CAB39 associated with urinary uromodulin levels. These data indicate that common variants in the UMOD promoter region may influence urinary uromodulin levels. They also provide insights into uromodulin biology and the association of UMOD variants with renal function.
Collapse
Affiliation(s)
- Matthias Olden
- National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts; Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
| | - Tanguy Corre
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Caroline Hayward
- Institute of Genetics and Molecular Medicine, Western General Hospital, and
| | - Daniela Toniolo
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy; Institute of Molecular Genetics, National Research Council, Pavia, Italy
| | - Sheila Ulivi
- Institute for Maternal and Child Health, Burlo Garofolo Pediatric Institute Trieste, Italy
| | - Paolo Gasparini
- Institute for Maternal and Child Health, Burlo Garofolo Pediatric Institute Trieste, Italy; Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Giorgio Pistis
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Shih-Jen Hwang
- National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts
| | - Sven Bergmann
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Harry Campbell
- Dulbecco Telethon Institute, San Raffaele Scientific Institute, Milan, Italy
| | - Massimiliano Cocca
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Ilaria Gandin
- Institute for Maternal and Child Health, Burlo Garofolo Pediatric Institute Trieste, Italy; Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Giorgia Girotto
- Institute for Maternal and Child Health, Burlo Garofolo Pediatric Institute Trieste, Italy; Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Bob Glaudemans
- Institute of Physiology, Zurich Center for Integrative Human Physiology, and
| | - Nicholas D Hastie
- Institute of Genetics and Molecular Medicine, Western General Hospital, and
| | | | - Ozren Polasek
- Department of Public Health, Faculty of Medicine, University of Split, Croatia
| | - Luca Rampoldi
- Dulbecco Telethon Institute, San Raffaele Scientific Institute, Milan, Italy
| | - Igor Rudan
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
| | - Cinzia Sala
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Michela Traglia
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | | | - Dragana Vuckovic
- Institute for Maternal and Child Health, Burlo Garofolo Pediatric Institute Trieste, Italy; Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Sonia Youhanna
- Institute of Physiology, Zurich Center for Integrative Human Physiology, and Division of Nephrology, Catholic University of Louvain Medical School, Brussels, Belgium
| | - Julien Weber
- Institute of Physiology, Zurich Center for Integrative Human Physiology, and Division of Nephrology, Catholic University of Louvain Medical School, Brussels, Belgium
| | - Alan F Wright
- Institute of Genetics and Molecular Medicine, Western General Hospital, and
| | - Zoltán Kutalik
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland; Institute of Social and Preventive Medicine, Lausanne University Hospital, Lausanne, Switzerland; and
| | - Murielle Bochud
- Institute of Social and Preventive Medicine, Lausanne University Hospital, Lausanne, Switzerland; and
| | - Caroline S Fox
- National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts; Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Olivier Devuyst
- Institute of Physiology, Zurich Center for Integrative Human Physiology, and Division of Nephrology, Catholic University of Louvain Medical School, Brussels, Belgium;
| |
Collapse
|
212
|
|
213
|
Franceschini N, Le TH. Genetics of hypertension: discoveries from the bench to human populations. Am J Physiol Renal Physiol 2014; 306:F1-F11. [PMID: 24133117 PMCID: PMC3921821 DOI: 10.1152/ajprenal.00334.2013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 10/11/2013] [Indexed: 12/20/2022] Open
Abstract
Hypertension is a complex trait that is influenced by both heritable and environmental factors. The search for genes accounting for the susceptibility to hypertension has driven parallel efforts in human research and in research using experimental animals in controlled environmental settings. Evidence from rodent models of genetic hypertension and human Mendelian forms of hypertension and hypotension have yielded mechanistic insights into the pathways that are perturbed in blood pressure homeostasis, most of which converge at the level of renal sodium reabsorption. However, the bridging of evidence from these very diverse approaches to identify mechanisms underlying hypertension susceptibility and the translation of these findings to human populations and public health remain a challenge. Furthermore, findings from genome-wide association studies still require functional validation in experimental models. In this review, we highlight results and implications from key studies in experimental and clinical hypertension to date.
Collapse
|
214
|
Simino J, Sung YJ, Kume R, Schwander K, Rao DC. Gene-alcohol interactions identify several novel blood pressure loci including a promising locus near SLC16A9. Front Genet 2013; 4:277. [PMID: 24376456 PMCID: PMC3860258 DOI: 10.3389/fgene.2013.00277] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Accepted: 11/22/2013] [Indexed: 01/11/2023] Open
Abstract
Alcohol consumption is a known risk factor for hypertension, with recent candidate studies implicating gene-alcohol interactions in blood pressure (BP) regulation. We used 6882 (predominantly) Caucasian participants aged 20-80 years from the Framingham SNP Health Association Resource (SHARe) to perform a genome-wide analysis of SNP-alcohol interactions on BP traits. We used a two-step approach in the ABEL suite to examine genetic interactions with three alcohol measures (ounces of alcohol consumed per week, drinks consumed per week, and the number of days drinking alcohol per week) on four BP traits [systolic (SBP), diastolic (DBP), mean arterial (MAP), and pulse (PP) pressure]. In the first step, we fit a linear mixed model of each BP trait onto age, sex, BMI, and antihypertensive medication while accounting for the phenotypic correlation among relatives. In the second step, we conducted 1 degree-of-freedom (df) score tests of the SNP main effect, alcohol main effect, and SNP-alcohol interaction using the maximum likelihood estimates (MLE) of the parameters from the first step. We then calculated the joint 2 df score test of the SNP main effect and SNP-alcohol interaction using MixABEL. The effect of SNP rs10826334 (near SLC16A9) on SBP was significantly modulated by both the number of alcoholic drinks and the ounces of alcohol consumed per week (p-values of 1.27E-08 and 3.92E-08, respectively). Each copy of the G-allele decreased SBP by 3.79 mmHg in those consuming 14 drinks per week vs. a 0.461 mmHg decrease in non-drinkers. Index SNPs in 20 other loci exhibited suggestive (p-value ≤ 1E-06) associations with BP traits by the 1 df interaction test or joint 2 df test, including 3 rare variants, one low-frequency variant, and SNPs near/in genes ESRRG, FAM179A, CRIPT-SOCS5, KAT2B, ADCY2, GLI3, ZNF716, SLIT1, PDE3A, KERA-LUM, RNF219-AS1, CLEC3A, FBXO15, and IGSF5. SNP-alcohol interactions may enhance discovery of novel variants with large effects that can be targeted with lifestyle modifications.
Collapse
Affiliation(s)
- Jeannette Simino
- Division of Biostatistics, Washington University School of MedicineSt. Louis, MO, USA
| | | | | | | | | |
Collapse
|
215
|
|
216
|
Trudu M, Janas S, Lanzani C, Debaix H, Schaeffer C, Ikehata M, Citterio L, Demaretz S, Trevisani F, Ristagno G, Glaudemans B, Laghmani K, Dell'Antonio G, Loffing J, Rastaldi MP, Manunta P, Devuyst O, Rampoldi L. Common noncoding UMOD gene variants induce salt-sensitive hypertension and kidney damage by increasing uromodulin expression. Nat Med 2013; 19:1655-1660. [PMID: 24185693 PMCID: PMC3856354 DOI: 10.1038/nm.3384] [Citation(s) in RCA: 288] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 09/19/2013] [Indexed: 12/21/2022]
Abstract
Hypertension and chronic kidney disease (CKD) are complex traits representing major global health problems. Multiple genome-wide association studies have identified common variants in the promoter of the UMOD gene, which encodes uromodulin, the major protein secreted in normal urine, that cause independent susceptibility to CKD and hypertension. Despite compelling genetic evidence for the association between UMOD risk variants and disease susceptibility in the general population, the underlying biological mechanism is not understood. Here, we demonstrate that UMOD risk variants increased UMOD expression in vitro and in vivo. Uromodulin overexpression in transgenic mice led to salt-sensitive hypertension and to the presence of age-dependent renal lesions similar to those observed in elderly individuals homozygous for UMOD promoter risk variants. The link between uromodulin and hypertension is due to activation of the renal sodium cotransporter NKCC2. We demonstrated the relevance of this mechanism in humans by showing that pharmacological inhibition of NKCC2 was more effective in lowering blood pressure in hypertensive patients who are homozygous for UMOD promoter risk variants than in other hypertensive patients. Our findings link genetic susceptibility to hypertension and CKD to the level of uromodulin expression and uromodulin's effect on salt reabsorption in the kidney. These findings point to uromodulin as a therapeutic target for lowering blood pressure and preserving renal function.
Collapse
Affiliation(s)
- Matteo Trudu
- Dulbecco Telethon Institute, c/o Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Sylvie Janas
- Institute of Physiology, Zurich Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
- Division of Nephrology, UCL Medical School, Brussels, Belgium
| | - Chiara Lanzani
- Division of Nephrology and Dialysis, San Raffaele Scientific Institute, Milan, Italy
| | - Huguette Debaix
- Institute of Physiology, Zurich Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
- Division of Nephrology, UCL Medical School, Brussels, Belgium
| | - Céline Schaeffer
- Dulbecco Telethon Institute, c/o Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Masami Ikehata
- Renal Research Laboratory, Fondazione IRCCS Ospedale Maggiore Policlinico & Fondazione D'Amico per la Ricerca sulle Malattie Renali, Milan, Italy
| | - Lorena Citterio
- Division of Nephrology and Dialysis, San Raffaele Scientific Institute, Milan, Italy
| | | | | | - Giuseppe Ristagno
- Department of Cardiovascular Research, Istituto di Ricerca Farmacologica Mario Negri, Milan, Italy
| | - Bob Glaudemans
- Institute of Physiology, Zurich Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | | | | | - Johannes Loffing
- Institute of Anatomy, Zurich Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Maria P Rastaldi
- Renal Research Laboratory, Fondazione IRCCS Ospedale Maggiore Policlinico & Fondazione D'Amico per la Ricerca sulle Malattie Renali, Milan, Italy
| | | | - Olivier Devuyst
- Institute of Physiology, Zurich Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Luca Rampoldi
- Dulbecco Telethon Institute, c/o Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
217
|
Ganesh SK, Arnett DK, Assimes TL, Basson CT, Chakravarti A, Ellinor PT, Engler MB, Goldmuntz E, Herrington DM, Hershberger RE, Hong Y, Johnson JA, Kittner SJ, McDermott DA, Meschia JF, Mestroni L, O’Donnell CJ, Psaty BM, Vasan RS, Ruel M, Shen WK, Terzic A, Waldman SA. Genetics and Genomics for the Prevention and Treatment of Cardiovascular Disease: Update. Circulation 2013; 128:2813-51. [DOI: 10.1161/01.cir.0000437913.98912.1d] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
218
|
Common noncoding UMOD gene variants induce salt-sensitive hypertension and kidney damage by increasing uromodulin expression. Nat Med 2013. [PMID: 24185693 DOI: 10.1038/nm.3384.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Hypertension and chronic kidney disease (CKD) are complex traits representing major global health problems. Multiple genome-wide association studies have identified common variants in the promoter of the UMOD gene, which encodes uromodulin, the major protein secreted in normal urine, that cause independent susceptibility to CKD and hypertension. Despite compelling genetic evidence for the association between UMOD risk variants and disease susceptibility in the general population, the underlying biological mechanism is not understood. Here, we demonstrate that UMOD risk variants increased UMOD expression in vitro and in vivo. Uromodulin overexpression in transgenic mice led to salt-sensitive hypertension and to the presence of age-dependent renal lesions similar to those observed in elderly individuals homozygous for UMOD promoter risk variants. The link between uromodulin and hypertension is due to activation of the renal sodium cotransporter NKCC2. We demonstrated the relevance of this mechanism in humans by showing that pharmacological inhibition of NKCC2 was more effective in lowering blood pressure in hypertensive patients who are homozygous for UMOD promoter risk variants than in other hypertensive patients. Our findings link genetic susceptibility to hypertension and CKD to the level of uromodulin expression and uromodulin's effect on salt reabsorption in the kidney. These findings point to uromodulin as a therapeutic target for lowering blood pressure and preserving renal function.
Collapse
|
219
|
Kemter E, Prückl P, Rathkolb B, Micklich K, Adler T, Becker L, Beckers J, Busch DH, Götz AA, Hans W, Horsch M, Ivandic B, Klingenspor M, Klopstock T, Rozman J, Schrewe A, Schulz H, Fuchs H, Gailus-Durner V, Hrabé de Angelis M, Wolf E, Aigner B. Standardized, systemic phenotypic analysis of Umod(C93F) and Umod(A227T) mutant mice. PLoS One 2013; 8:e78337. [PMID: 24205203 PMCID: PMC3813435 DOI: 10.1371/journal.pone.0078337] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 09/18/2013] [Indexed: 12/31/2022] Open
Abstract
Uromodulin-associated kidney disease (UAKD) summarizes different clinical features of an autosomal dominant heritable disease syndrome in humans with a proven uromodulin (UMOD) mutation involved. It is often characterized by hyperuricemia, gout, alteration of urine concentrating ability, as well as a variable rate of disease progression inconstantly leading to renal failure and histological alterations of the kidneys. We recently established the two Umod mutant mouse lines Umod(C93F) and Umod(A227T) on the C3H inbred genetic background both showing kidney defects analogous to those found in human UAKD patients. In addition, disease symptoms were revealed that were not yet described in other published mouse models of UAKD. To examine if further organ systems and/or metabolic pathways are affected by Umod mutations as primary or secondary effects, we describe a standardized, systemic phenotypic analysis of the two mutant mouse lines Umod(A227T) and Umod(C93F) in the German Mouse Clinic. Different genotypes as well as different ages were tested. Beside the already published changes in body weight, body composition and bone metabolism, the influence of the Umod mutation on energy metabolism was confirmed. Hematological analysis revealed a moderate microcytic and erythropenic anemia in older Umod mutant mice. Data of the other analyses in 7-10 month-old mutant mice showed single small additional effects.
Collapse
Affiliation(s)
- Elisabeth Kemter
- Chair for Molecular Animal Breeding and Biotechnology, and Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU, Munich, Munich, Germany
| | - Petra Prückl
- Chair for Molecular Animal Breeding and Biotechnology, and Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU, Munich, Munich, Germany
| | - Birgit Rathkolb
- Chair for Molecular Animal Breeding and Biotechnology, and Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU, Munich, Munich, Germany
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Kateryna Micklich
- Chair for Molecular Animal Breeding and Biotechnology, and Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU, Munich, Munich, Germany
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Thure Adler
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Medical Microbiology, Immunology, and Hygiene, TU, Munich, Munich, Germany
| | - Lore Becker
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Neurology, Friedrich-Baur-Institut, LMU, Munich, Munich, Germany
| | - Johannes Beckers
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Member of German Center for Diabetes Research (DZD), Neuherberg, Germany
- Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, TU, Munich, Freising-Weihenstephan, Germany
| | - Dirk H. Busch
- Institute of Medical Microbiology, Immunology, and Hygiene, TU, Munich, Munich, Germany
| | - Alexander A. Götz
- German Mouse Clinic, Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Wolfgang Hans
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Marion Horsch
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Boris Ivandic
- Department of Medicine III, Division of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - Martin Klingenspor
- Molecular Nutritional Medicine, Else Kröner-Fresenius Center, TU, Munich, Freising-Weihenstephan, Germany
| | - Thomas Klopstock
- Department of Neurology, Friedrich-Baur-Institut, LMU, Munich, Munich, Germany
- German Center for Vertigo and Balance Disorders, University Hospital Munich, Campus Grosshadern, Munich, Germany
| | - Jan Rozman
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Member of German Center for Diabetes Research (DZD), Neuherberg, Germany
- Molecular Nutritional Medicine, Else Kröner-Fresenius Center, TU, Munich, Freising-Weihenstephan, Germany
| | - Anja Schrewe
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Medicine III, Division of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - Holger Schulz
- Institute of Epidemiology I, Helmholtz Zentrum München, Neuherberg, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Member of German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Valérie Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Member of German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Martin Hrabé de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Member of German Center for Diabetes Research (DZD), Neuherberg, Germany
- Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, TU, Munich, Freising-Weihenstephan, Germany
- German Center for Vertigo and Balance Disorders, University Hospital Munich, Campus Grosshadern, Munich, Germany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, and Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU, Munich, Munich, Germany
| | - Bernhard Aigner
- Chair for Molecular Animal Breeding and Biotechnology, and Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU, Munich, Munich, Germany
- * E-mail:
| |
Collapse
|
220
|
Youhanna S, Weber J, Beaujean V, Glaudemans B, Sobek J, Devuyst O. Determination of uromodulin in human urine: influence of storage and processing. Nephrol Dial Transplant 2013; 29:136-45. [PMID: 24097801 DOI: 10.1093/ndt/gft345] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Uromodulin (Tamm-Horsfall protein) is the most abundant protein excreted in the urine under physiological conditions. It is exclusively produced in the kidney and secreted into the urine via proteolytic cleavage. The involvement of UMOD, the gene that encodes uromodulin, in rare autosomal dominant diseases, and its robust genome-wide association with the risk of chronic kidney disease suggest that the level of uromodulin in urine could represent a critical biomarker for kidney function. The structure of uromodulin is complex, with multiple disulfide bonds and typical domains of extracellular proteins. METHODS Thus far, the conditions influencing stability and measurement of uromodulin in human urine have not been systematically investigated, giving inconsistent results. In this study, we used a robust, in-house ELISA to characterize the conditions of sampling and storage necessary to provide a faithful dosage of uromodulin in the urine. RESULTS The levels of uromodulin in human urine were significantly affected by centrifugation and vortexing, as well as by the conditions and duration of storage. CONCLUSIONS These results validate a simple, low-cost ELISA and document the optimal conditions of processing and storage for measuring uromodulin in human urine.
Collapse
Affiliation(s)
- Sonia Youhanna
- Institute of Physiology, Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
221
|
Nalls MA, Saad M, Noyce AJ, Keller MF, Schrag A, Bestwick JP, Traynor BJ, Gibbs JR, Hernandez DG, Cookson MR, Morris HR, Williams N, Gasser T, Heutink P, Wood N, Hardy J, Martinez M, Singleton AB. Genetic comorbidities in Parkinson's disease. Hum Mol Genet 2013; 23:831-41. [PMID: 24057672 DOI: 10.1093/hmg/ddt465] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Parkinson's disease (PD) has a number of known genetic risk factors. Clinical and epidemiological studies have suggested the existence of intermediate factors that may be associated with additional risk of PD. We construct genetic risk profiles for additional epidemiological and clinical factors using known genome-wide association studies (GWAS) loci related to these specific phenotypes to estimate genetic comorbidity in a systematic review. We identify genetic risk profiles based on GWAS variants associated with schizophrenia and Crohn's disease as significantly associated with risk of PD. Conditional analyses adjusting for SNPs near loci associated with PD and schizophrenia or PD and Crohn's disease suggest that spatially overlapping loci associated with schizophrenia and PD account for most of the shared comorbidity, while variation outside of known proximal loci shared by PD and Crohn's disease accounts for their shared genetic comorbidity. We examine brain methylation and expression signatures proximal to schizophrenia and Crohn's disease loci to infer functional changes in the brain associated with the variants contributing to genetic comorbidity. We compare our results with a systematic review of epidemiological literature, while the findings are dissimilar to a degree; marginal genetic associations corroborate the directionality of associations across genetic and epidemiological data. We show a strong genetically defined level of comorbidity between PD and Crohn's disease as well as between PD and schizophrenia, with likely functional consequences of associated variants occurring in brain.
Collapse
Affiliation(s)
- Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
222
|
Zhou J, Chen Y, Liu Y, Shi S, Wang S, Li X, Zhang H, Wang H. Urinary uromodulin excretion predicts progression of chronic kidney disease resulting from IgA nephropathy. PLoS One 2013; 8:e71023. [PMID: 23990922 PMCID: PMC3750049 DOI: 10.1371/journal.pone.0071023] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 06/28/2013] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Uromodulin, or Tamm-Horsfall protein, is the most abundant urinary protein in healthy individuals. Recent studies have suggested that uromodulin may play a role in chronic kidney diseases. We examined an IgA nephropathy cohort to determine whether uromodulin plays a role in the progression of IgA nephropathy. METHODS A total of 344 IgA nephropathy patients were involved in this study. Morphological changes were evaluated with the Oxford classification of IgA nephropathy. Enzyme Linked Immunosorbent Assay (ELISA) measured the urinary uromodulin level on the renal biopsy day. Follow up was done regularly on 185 patients. Time-average blood pressure, time-average proteinuria, estimated glomerular filtration rate (eGFR) and eGFR decline rate were caculated. Association between the urinary uromodulin level and the eGFR decline rate was analyzed with SPSS 13.0. RESULTS We found that lower baseline urinary uromodulin levels (P = 0.03) and higher time-average proteinuria (P = 0.04) were risk factors for rapid eGFR decline in a follow-up subgroup of the IgA nephropathy cohort. Urinary uromodulin level was correlated with tubulointerstitial lesions (P = 0.016). Patients that had more tubular atrophy/interstitial fibrosis on the surface had lower urinary uromodulin levels (P = 0.02). CONCLUSIONS Urinary uromodulin level is associated with interstitial fibrosis/tubular atrophy and contributes to eGFR decline in IgA nephropathy.
Collapse
Affiliation(s)
- Jingjing Zhou
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
| | - Yuqing Chen
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- * E-mail:
| | - Ying Liu
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
| | - Sufang Shi
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
| | - Suxia Wang
- Laboratory of Electron Microscopy, Peking University First Hospital, Peking University First Hospital, Beijing, China
| | - Xueying Li
- Department of Statistics, Peking University First Hospital, Beijing, China
| | - Hong Zhang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
| | - Haiyan Wang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
| |
Collapse
|
223
|
DING YU, XIA BOHOU, YU JINFANG, LENG JIANHANG, HUANG JINYU. Mitochondrial DNA mutations and essential hypertension (Review). Int J Mol Med 2013; 32:768-74. [DOI: 10.3892/ijmm.2013.1459] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 07/22/2013] [Indexed: 11/06/2022] Open
|
224
|
Abstract
The elucidation of genes implicated in Mendelian forms of hypertension demonstrates rare variants with substantial effects are responsible, and often these genes lie within pathways managing sodium homeostasis. More recently with advances in affordable high-throughput genotyping strategies, multiple common genetic variants with modest effects on blood pressure (<1 mmHg systolic) have been discovered in the population. In aggregate, these common variants explain <3% of the variance of blood pressure. Although these findings may offer new mechanistic insights into the biology of blood pressure, a key question is can these findings translate into patient benefit? It is timely to reflect on recent advances in genomics, and the use of new resources, such as the 1000 Genomes Project and the Encyclopedia of DNA Elements, to annotate likely causal variants, and their relevance to cardiovascular disease. In this review, we discuss the advances in relation to our knowledge of the genetic architecture of blood pressure, and whether gene discoveries might influence cardiovascular risk assessment, help to stratify patient response to medicine, or identify new biological pathways for novel therapeutic targets.
Collapse
Affiliation(s)
- Patricia B Munroe
- William Harvey Research Institute and Barts National Institute for Health Research Cardiovascular Biomedical Research Unit, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ United Kingdom
| | | | | |
Collapse
|
225
|
Hypertension and kidneys: unraveling complex molecular mechanisms underlying hypertensive renal damage. J Hum Hypertens 2013; 28:74-9. [PMID: 23803592 DOI: 10.1038/jhh.2013.55] [Citation(s) in RCA: 198] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 04/12/2013] [Accepted: 05/28/2013] [Indexed: 01/18/2023]
Abstract
Kidney damage represents a frequent event in the course of hypertension, ranging from a benign to a malignant form of nephropathy depending on several factors, that is, individual susceptibility, degree of hypertension, type of etiology and underlying kidney disease. Multiple mechanisms are involved in determination of kidney glomerular, tubular and interstitial injuries in hypertension. The present review article discusses relevant contributory molecular mechanisms underpinning the promotion of hypertensive renal damage, such as the renin-angiotensin-aldosterone system (RAAS), oxidative stress, endothelial dysfunction, and genetic and epigenetic determinants. We highlighted major pathways involved in the progression of inflammation and fibrosis leading to glomerular sclerosis, tubular atrophy and interstitial fibrosis, thus providing a state of the art review of the pathogenetic background useful for a better understanding of current and future therapeutic strategies toward hypertensive nephropathy. An adequate control of high blood pressure, obtained through an appropriate therapeutic intervention, still represents the key strategy to achieve a satisfactory control of renal damage in hypertension. In this regard, we reviewed the impact of currently available antihypertensive pharmacological treatment on kidney damage, with particular regard to RAAS inhibitors. Notably, recent findings underscored the ability of the kidneys to regenerate and to repair tissue injuries through the differentiation of resident embryonic stem cells. Pharmacological modulation of the renal endogenous reparative process (that is, with angiotensin-converting enzyme inhibitors and AT1 angiotensin II receptor blockers), as well as future therapeutic strategies targeted to the renopoietic system, offers interesting perspectives for the management of hypertensive nephropathy.
Collapse
|
226
|
Delles C. Genetics of hypertension. Hypertension 2013. [DOI: 10.2217/ebo.12.196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Christian Delles
- Christian Delles graduated from the University of Freiburg (Baden-Württemberg, Germany) and trained in internal medicine and nephrology at the Department of Nephrology, University of Erlangen-Nürnberg (Erlangen, Germany), where he worked in Roland Schmieder’s group. In 2003, he was awarded a fellowship by the Deutsche Forschungsgemeinschaft for studies into reactive oxygen species in hypertension in Anna Dominiczak’s group in Glasgow (UK). He is currently a Reader and Honorary Consultant Physician at the
| |
Collapse
|
227
|
Bochud M, Guessous I. Gene-environment interactions of selected pharmacogenes in arterial hypertension. Expert Rev Clin Pharmacol 2013; 5:677-86. [PMID: 23234325 DOI: 10.1586/ecp.12.58] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Hypertension affects approximately 1 billion people worldwide. Owing to population aging, hypertension-related cardiovascular burden is expected to rise in the near future. In addition to genetic variants influencing the blood pressure response to antihypertensive drugs, several genes encoding for drug-metabolizing or -transporting enzymes have been associated with blood pressure and/or hypertension in humans (e.g., ACE, CYP1A2, CYP3A5, ABCB1 and MTHFR) regardless of drug treatment. These genes are also involved in the metabolism and transport of endogenous substances and their effects may be modified by selected environmental factors, such as diet or lifestyle. However, little is currently known on the complex interplay between environmental factors, endogenous factors, genetic variants and drugs on blood pressure control. This review will discuss the respective role of population-based primary prevention and personalized medicine for arterial hypertension, taking a pharmacogenomics' perspective focusing on selected pharmacogenes.
Collapse
Affiliation(s)
- Murielle Bochud
- Institute of Social and Preventive Medicine, Lausanne University Hospital, Switzerland.
| | | |
Collapse
|
228
|
Larsson E, Wahlstrand B, Hedblad B, Hedner T, Kjeldsen SE, Melander O, Lindahl P. Hypertension and genetic variation in endothelial-specific genes. PLoS One 2013; 8:e62035. [PMID: 23637959 PMCID: PMC3639261 DOI: 10.1371/journal.pone.0062035] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 03/18/2013] [Indexed: 01/11/2023] Open
Abstract
Genome-wide association (GWA) studies usually detect common genetic variants with low-to-medium effect sizes. Many contributing variants are not revealed, since they fail to reach significance after strong correction for multiple comparisons. The WTCCC study for hypertension, for example, failed to identify genome-wide significant associations. We hypothesized that genetic variation in genes expressed specifically in the endothelium may be important for hypertension development. Results from the WTCCC study were combined with previously published gene expression data from mice to specifically investigate SNPs located within endothelial-specific genes, bypassing the requirement for genome-wide significance. Six SNPs from the WTCCC study were selected for independent replication in 5205 hypertensive patients and 5320 population-based controls, and successively in a cohort of 16537 individuals. A common variant (rs10860812) in the DRAM (damage-regulated autophagy modulator) locus showed association with hypertension (P = 0.008) in the replication study. The minor allele (A) had a protective effect (OR = 0.93; 95% CI 0.88–0.98 per A-allele), which replicates the association in the WTCCC GWA study. However, a second follow-up, in the larger cohort, failed to reveal an association with blood pressure. We further tested the endothelial-specific genes for co-localization with a panel of newly discovered SNPs from large meta-GWAS on hypertension or blood pressure. There was no significant overlap between those genes and hypertension or blood pressure loci. The result does not support the hypothesis that genetic variation in genes expressed in endothelium plays an important role for hypertension development. Moreover, the discordant association of rs10860812 with blood pressure in the case control study versus the larger Malmö Preventive Project–study highlights the importance of rigorous replication in multiple large independent studies.
Collapse
Affiliation(s)
- Erik Larsson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- * E-mail: (EL); (PL)
| | - Björn Wahlstrand
- Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Bo Hedblad
- Clinical research Center (CRC), Malmö University Hospital, Malmö, Sweden
| | - Thomas Hedner
- Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Sverre E. Kjeldsen
- Department of Cardiology/Cardiovascular and Renal Research Center, Ullevaal University Hospital, Oslo, Norway
| | - Olle Melander
- Clinical research Center (CRC), Malmö University Hospital, Malmö, Sweden
| | - Per Lindahl
- Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- * E-mail: (EL); (PL)
| |
Collapse
|
229
|
Ganesh SK, Tragante V, Guo W, Guo Y, Lanktree MB, Smith EN, Johnson T, Castillo BA, Barnard J, Baumert J, Chang YPC, Elbers CC, Farrall M, Fischer ME, Franceschini N, Gaunt TR, Gho JM, Gieger C, Gong Y, Isaacs A, Kleber ME, Leach IM, McDonough CW, Meijs MF, Mellander O, Molony CM, Nolte IM, Padmanabhan S, Price TS, Rajagopalan R, Shaffer J, Shah S, Shen H, Soranzo N, van der Most PJ, Van Iperen EP, Van Setten JA, Vonk JM, Zhang L, Beitelshees AL, Berenson GS, Bhatt DL, Boer JM, Boerwinkle E, Burkley B, Burt A, Chakravarti A, Chen W, Cooper-DeHoff RM, Curtis SP, Dreisbach A, Duggan D, Ehret GB, Fabsitz RR, Fornage M, Fox E, Furlong CE, Gansevoort RT, Hofker MH, Hovingh GK, Kirkland SA, Kottke-Marchant K, Kutlar A, LaCroix AZ, Langaee TY, Li YR, Lin H, Liu K, Maiwald S, Malik R, CARDIOGRAM, METASTROKE, Murugesan G, Newton-Cheh C, O'Connell JR, Onland-Moret NC, Ouwehand WH, Palmas W, Penninx BW, Pepine CJ, Pettinger M, Polak JF, Ramachandran VS, Ranchalis J, Redline S, Ridker PM, Rose LM, Scharnag H, Schork NJ, Shimbo D, Shuldiner AR, Srinivasan SR, Stolk RP, Taylor HA, Thorand B, Trip MD, van Duijn CM, Verschuren WM, Wijmenga C, Winkelmann BR, Wyatt S, et alGanesh SK, Tragante V, Guo W, Guo Y, Lanktree MB, Smith EN, Johnson T, Castillo BA, Barnard J, Baumert J, Chang YPC, Elbers CC, Farrall M, Fischer ME, Franceschini N, Gaunt TR, Gho JM, Gieger C, Gong Y, Isaacs A, Kleber ME, Leach IM, McDonough CW, Meijs MF, Mellander O, Molony CM, Nolte IM, Padmanabhan S, Price TS, Rajagopalan R, Shaffer J, Shah S, Shen H, Soranzo N, van der Most PJ, Van Iperen EP, Van Setten JA, Vonk JM, Zhang L, Beitelshees AL, Berenson GS, Bhatt DL, Boer JM, Boerwinkle E, Burkley B, Burt A, Chakravarti A, Chen W, Cooper-DeHoff RM, Curtis SP, Dreisbach A, Duggan D, Ehret GB, Fabsitz RR, Fornage M, Fox E, Furlong CE, Gansevoort RT, Hofker MH, Hovingh GK, Kirkland SA, Kottke-Marchant K, Kutlar A, LaCroix AZ, Langaee TY, Li YR, Lin H, Liu K, Maiwald S, Malik R, CARDIOGRAM, METASTROKE, Murugesan G, Newton-Cheh C, O'Connell JR, Onland-Moret NC, Ouwehand WH, Palmas W, Penninx BW, Pepine CJ, Pettinger M, Polak JF, Ramachandran VS, Ranchalis J, Redline S, Ridker PM, Rose LM, Scharnag H, Schork NJ, Shimbo D, Shuldiner AR, Srinivasan SR, Stolk RP, Taylor HA, Thorand B, Trip MD, van Duijn CM, Verschuren WM, Wijmenga C, Winkelmann BR, Wyatt S, Young JH, Boehm BO, Caulfield MJ, Chasman DI, Davidson KW, Doevendans PA, FitzGerald GA, Gums JG, Hakonarson H, Hillege HL, Illig T, Jarvik GP, Johnson JA, Kastelein JJ, Koenig W, LifeLines Cohort Study, März W, Mitchell BD, Murray SS, Oldehinkel AJ, Rader DJ, Reilly MP, Reiner AP, Schadt EE, Silverstein RL, Snieder H, Stanton AV, Uitterlinden AG, van der Harst P, van der Schouw YT, Samani NJ, Johnson AD, Munroe PB, de Bakker PI, Zhu X, Levy D, Keating BJ, Asselbergs FW. Loci influencing blood pressure identified using a cardiovascular gene-centric array. Hum Mol Genet 2013; 22:1663-78. [PMID: 23303523 PMCID: PMC3657476 DOI: 10.1093/hmg/dds555] [Show More Authors] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 11/30/2012] [Accepted: 12/27/2012] [Indexed: 01/11/2023] Open
Abstract
Blood pressure (BP) is a heritable determinant of risk for cardiovascular disease (CVD). To investigate genetic associations with systolic BP (SBP), diastolic BP (DBP), mean arterial pressure (MAP) and pulse pressure (PP), we genotyped ∼50 000 single-nucleotide polymorphisms (SNPs) that capture variation in ∼2100 candidate genes for cardiovascular phenotypes in 61 619 individuals of European ancestry from cohort studies in the USA and Europe. We identified novel associations between rs347591 and SBP (chromosome 3p25.3, in an intron of HRH1) and between rs2169137 and DBP (chromosome1q32.1 in an intron of MDM4) and between rs2014408 and SBP (chromosome 11p15 in an intron of SOX6), previously reported to be associated with MAP. We also confirmed 10 previously known loci associated with SBP, DBP, MAP or PP (ADRB1, ATP2B1, SH2B3/ATXN2, CSK, CYP17A1, FURIN, HFE, LSP1, MTHFR, SOX6) at array-wide significance (P < 2.4 × 10(-6)). We then replicated these associations in an independent set of 65 886 individuals of European ancestry. The findings from expression QTL (eQTL) analysis showed associations of SNPs in the MDM4 region with MDM4 expression. We did not find any evidence of association of the two novel SNPs in MDM4 and HRH1 with sequelae of high BP including coronary artery disease (CAD), left ventricular hypertrophy (LVH) or stroke. In summary, we identified two novel loci associated with BP and confirmed multiple previously reported associations. Our findings extend our understanding of genes involved in BP regulation, some of which may eventually provide new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Santhi K. Ganesh
- Division of Cardiovascular Medicine, University of Michigan Health System, Ann Arbor, MI, USA
| | - Vinicius Tragante
- Department of Cardiology, Division Heart and Lungs
- Department of Medical Genetics and
| | - Wei Guo
- Department of Epidemiology and Biostatistics, School of Medicine and
| | - Yiran Guo
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Matthew B. Lanktree
- Departments of Medicine and Biochemistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Erin N. Smith
- Department of Pediatrics and Rady's Children's Hospital, School of Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Toby Johnson
- Clinical Pharmacology and Barts and The London Genome Centre, William Harvey Research Institute, Barts and
- The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Berta Almoguera Castillo
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - John Barnard
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Yen-Pei Christy Chang
- Department of Medicine and
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Clara C. Elbers
- Department of Medical Genetics and
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Martin Farrall
- Department of Cardiovascular Medicine, The Wellcome Trust Centre for Human Genetics, University of Oxford, OxfordOX3 7BN, UK
| | - Mary E. Fischer
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI, USA
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tom R. Gaunt
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | | | | | - Yan Gong
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and
| | - Aaron Isaacs
- Genetic Epidemiology Unit, Department of Epidemiology and
| | - Marcus E. Kleber
- LURIC Study nonprofit LLC, Freiburg, Germany
- Mannheim Institute of Public Health, Social and Preventive Medicine, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| | | | - Caitrin W. McDonough
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and
| | | | - Olle Mellander
- Hypertension and Cardiovascular Disease, Department of Clinical Sciences, Lund University, Malmö, Sweden
- Centre of Emergency Medicine, Skåne University Hospital, Malmö, Sweden
| | | | | | - Sandosh Padmanabhan
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Tom S. Price
- MRC SGDP Centre, Institute of Psychiatry, London, UK
| | - Ramakrishnan Rajagopalan
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, USA
| | | | - Sonia Shah
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, Kathleen Lonsdale Building, Gower Place, London WC1E 6BT, UK
| | | | | | | | - Erik P.A. Van Iperen
- Durrer Center for Cardiogenetic Research
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics
| | | | | | - Li Zhang
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Amber L. Beitelshees
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Gerald S. Berenson
- Department of Epidemiology, Tulane University, 1440 Canal Street, Suite 1829, New Orleans, LA, USA
| | - Deepak L. Bhatt
- VA Boston Healthcare System, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA
| | - Jolanda M.A. Boer
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Eric Boerwinkle
- Human Genetics Center and Institute of Molecular Medicine and Division of Epidemiology, University of Texas Health Science Center, Houston, TX, USA
| | - Ben Burkley
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and
| | - Amber Burt
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, USA
| | - Aravinda Chakravarti
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine and
| | - Wei Chen
- Department of Epidemiology, Tulane University, 1440 Canal Street, Suite 1829, New Orleans, LA, USA
| | - Rhonda M. Cooper-DeHoff
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and
| | - Sean P. Curtis
- Merck Research Laboratories, PO Box 2000, Rahway, NJ 07065, USA
| | | | - David Duggan
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Georg B. Ehret
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine and
| | - Richard R. Fabsitz
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Myriam Fornage
- Human Genetics Center and Institute of Molecular Medicine and Division of Epidemiology, University of Texas Health Science Center, Houston, TX, USA
| | | | - Clement E. Furlong
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, USA
| | | | - Marten H. Hofker
- Molecular Genetics, Medical Biology Section, Department of Pathology and Medical Biology
| | | | - Susan A. Kirkland
- Department of Community Health and Epidemiology, Dalhousie University, Canada
| | | | | | - Andrea Z. LaCroix
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Taimour Y. Langaee
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and
| | - Yun R. Li
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Honghuang Lin
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Kiang Liu
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Steffi Maiwald
- Department of Vascular Medicine, University of Amsterdam, Amsterdam, The Netherlands
| | - Rainer Malik
- Institute for Stroke and Dementia Research and
- Neurologische Klinik, Klinikum Grosshadern, Ludwig-Maximilians-Universität, München, Germany
| | | | - Gurunathan Murugesan
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Christopher Newton-Cheh
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA, USA
| | - Jeffery R. O'Connell
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
| | - N. Charlotte Onland-Moret
- Department of Medical Genetics and
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Willem H. Ouwehand
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge and Human Genetics, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Walter Palmas
- Department of Medicine, Columbia University, New York, NY, USA
| | - Brenda W. Penninx
- Department of Psychiatry/EMGO Institute, VU University Medical Centre, Amsterdam, The Netherlands
| | - Carl J. Pepine
- Division of Cardiovascular Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Mary Pettinger
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | | | - Vasan S. Ramachandran
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- National Heart, Lung and Blood Institute's Framingham Heart Study, 73 Mt.Wayte Avenue Suite #2, Framingham, MA, USA
| | - Jane Ranchalis
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, USA
| | - Susan Redline
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School and
| | - Paul M. Ridker
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School and
| | - Lynda M. Rose
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School and
| | - Hubert Scharnag
- Division of Cardiovascular Medicine, University of Michigan Health System, Ann Arbor, MI, USA
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Austria
| | - Nicholas J. Schork
- The Scripps Translational Science Institute and The Scripps Research Institute, 3344 N. Torrey Pines Ct. Ste 300, La Jolla, CA, USA
| | - Daichi Shimbo
- Department of Medicine, Columbia University, New York, NY, USA
| | - Alan R. Shuldiner
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
- Geriatric Research and Education Clinical Center, Veterans Administration Medical Center, Baltimore, MD, USA
| | - Sathanur R. Srinivasan
- Department of Epidemiology, Tulane University, 1440 Canal Street, Suite 1829, New Orleans, LA, USA
| | | | | | | | - Mieke D. Trip
- Department of Cardiology, AMC, Amsterdam, The Netherlands
| | | | - W. Monique Verschuren
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | | | | | - Sharon Wyatt
- School of Nursing, University of Mississippi Medical Center, Jackson, MS, USA
| | - J. Hunter Young
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Mark J. Caulfield
- Clinical Pharmacology and Barts and The London Genome Centre, William Harvey Research Institute, Barts and
| | - Daniel I. Chasman
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School and
| | - Karina W. Davidson
- Departments of Medicine & Psychiatry, Columbia University, New York, NY, USA
| | | | - Garret A. FitzGerald
- The Institute for Translational Medicine and Therapeutics, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John G. Gums
- Departments of Pharmacotherapy and Translational Research and Community Health and Family Medicine, University of Florida, Gainesville, FL, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | - Thomas Illig
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - Gail P. Jarvik
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, USA
| | - Julie A. Johnson
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and
| | | | - Wolfgang Koenig
- Department of Internal Medicine I—Cardiology, University of Ulm Medical Centre, Ulm, Germany
| | | | - Winfried März
- Mannheim Institute of Public Health, Social and Preventive Medicine, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
- Synlab Academy, Mannheim, Germany
| | - Braxton D. Mitchell
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sarah S. Murray
- Scripps Translational Science Institute and Scripps Health, 3344 N. Torrey Pines Ct. Ste 300, La Jolla, CA, USA
| | - Albertine J. Oldehinkel
- Interdisciplinary Center Psychopathology and Emotion Regulation, Groningen University, University Medical Center Groningen, Groningen, The Netherlands
| | - Daniel J. Rader
- Cardiovascular Institute, The Perelman School of Medicine at the University of Pennsylvania, PA, USA
| | - Muredach P. Reilly
- Cardiovascular Institute, The Perelman School of Medicine at the University of Pennsylvania, PA, USA
| | - Alex P. Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Eric E. Schadt
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY, USA
| | - Roy L. Silverstein
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic
| | | | - Alice V. Stanton
- Molecular & Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland
| | - André G. Uitterlinden
- Departments of Epidemiology and Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Yvonne T. van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nilesh J. Samani
- Department of Cardiovascular Sciences, University of Leicester and
- Leicester NIHR Biomedical Research Unit in Cardiovascular Disease, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Andrew D. Johnson
- National Heart, Lung and Blood Institute's Framingham Heart Study, 73 Mt.Wayte Avenue Suite #2, Framingham, MA, USA
| | - Patricia B. Munroe
- Clinical Pharmacology and Barts and The London Genome Centre, William Harvey Research Institute, Barts and
- The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Paul I.W. de Bakker
- Department of Medical Genetics and
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaofeng Zhu
- Department of Epidemiology and Biostatistics, School of Medicine and
| | - Daniel Levy
- Center for Population Studies, National Heart, Lung, and Blood Institute, Framingham, MA, USA
| | - Brendan J. Keating
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Folkert W. Asselbergs
- Department of Cardiology, Division Heart and Lungs
- Department of Medical Genetics and
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
- Durrer Center for Cardiogenetic Research
| |
Collapse
|
230
|
Havulinna AS, Kettunen J, Ukkola O, Osmond C, Eriksson JG, Kesäniemi YA, Jula A, Peltonen L, Kontula K, Salomaa V, Newton-Cheh C. A blood pressure genetic risk score is a significant predictor of incident cardiovascular events in 32,669 individuals. Hypertension 2013; 61:987-94. [PMID: 23509078 DOI: 10.1161/hypertensionaha.111.00649] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Recent genome-wide association studies have identified genetic variants associated with blood pressure (BP). We investigated whether genetic risk scores (GRSs) constructed of these variants would predict incident cardiovascular disease (CVD) events. We genotyped 32 common single nucleotide polymorphisms in several Finnish cohorts, with up to 32,669 individuals after exclusion of prevalent CVD cases. The median follow-up was 9.8 years, during which 2295 incident CVD events occurred. We created GRSs separately for systolic BP and diastolic BP by multiplying the risk allele count of each single nucleotide polymorphism by the effect size estimated in published genome-wide association studies. We performed Cox regression analyses with and without adjustment for clinical factors, including BP at baseline in each cohort. The results were combined by inverse variance-weighted fixed-effects meta-analysis. The GRSs were strongly associated with systolic BP and diastolic BP, and baseline hypertension (all P<10(-62)). Hazard ratios comparing the highest quintiles of systolic BP and diastolic BP GRSs with the lowest quintiles after adjustment for age, age squared, and sex were 1.25 (1.07-1.46; P=0.006) and 1.23 (1.05-1.43; P=0.01), respectively, for incident coronary heart disease; 1.24 (1.01-1.53; P=0.04) and 1.35 (1.09-1.66; P=0.005), respectively, for incident stroke; and 1.23 (1.08-1.40; P=2 × 10(-6)) and 1.26 (1.11-1.44; P=5 × 10(-4)), respectively, for composite CVD. In conclusion, BP findings from genome-wide association studies are strongly replicated. GRSs comprising bona fide BP-single nucleotide polymorphisms predicted CVD risk, consistent with a lifelong effect on BP of these variants collectively.
Collapse
Affiliation(s)
- Aki S Havulinna
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
231
|
Abstract
TwinsUK is a nation-wide registry of volunteer twins in the United Kingdom, with about 12,000 registered twins (83% female, equal number of monozygotic and dizygotic twins, predominantly middle-aged and older). Over the last 20 years, questionnaire and blood/urine/tissue samples have been collected on over 7,000 subjects, as well as three comprehensive phenotyping assessments in the clinical facilities of the Department of Twin Research and Genetic Epidemiology, King's College London. The primary focus of study has been the genetic basis of healthy aging process and complex diseases, including cardiovascular, metabolic, musculoskeletal, and ophthalmologic disorders. Alongside the detailed clinical, biochemical, behavioral, and socio-economic characterization of the study population, the major strength of TwinsUK is availability of several 'omics' technologies for the participants. These include genome-wide scans of single nucleotide variants, next-generation sequencing, exome sequencing, epigenetic markers (MeDIP sequencing), gene expression arrays and RNA sequencing, telomere length measures, metabolomic profiles, and gut flora microbiomics. The scientific community now can freely access parts of the phenotype data from the 'TwinsUK', and interested researchers are encouraged to contact us via our Web site (www.twinsuk.ac.uk) for future collaborations.
Collapse
Affiliation(s)
- Alireza Moayyeri
- Department of Twin Research and Genetic Epidemiology, St. Thomas' Hospital, London, UK
| | | | | | | |
Collapse
|
232
|
Common genetic variants of the human uromodulin gene regulate transcription and predict plasma uric acid levels. Kidney Int 2013; 83:733-40. [PMID: 23344472 DOI: 10.1038/ki.2012.449] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Uromodulin (UMOD) genetic variants cause familial juvenile hyperuricemic nephropathy, characterized by hyperuricemia with decreased renal excretion of UMOD and uric acid, suggesting a role for UMOD in the regulation of plasma uric acid. To determine this, we screened common variants across the UMOD locus in one community-based Chinese population of 1000 individuals and the other population from 642 American twins and siblings of European and Hispanic ancestry. Transcriptional activity of promoter variants was estimated in luciferase reporter plasmids transfected into HEK-293 cells and mIMCD3 cells. In the primary Chinese population, we found that carriers of the GCC haplotype had higher plasma uric acid, and three promoter variants were associated with plasma uric acid. UMOD promoter variants displayed reciprocal effects on urine uric acid excretion and plasma uric acid concentration, suggesting a primary effect on renal tubular handling of urate. These UMOD genetic marker-on-trait associations for uric acid were replicated in the independent American cohort. Site-directed mutagenesis at trait-associated UMOD promoter variants altered promoter activity in transfected luciferase reporter plasmids. Thus, UMOD promoter variants seem to initiate a cascade of transcriptional and biochemical changes influencing UMOD secretion, leading to altered plasma uric acid levels.
Collapse
|
233
|
Abstract
Hypertension (HTN) is quantitatively the major cardiovascular risk factor and responsible for ∼50% of cardiovascular morbidity and mortality. Blood pressure (BP) is also a classical complex genetic trait with heritability estimates of 30-50%. Although much is known about BP regulation, the intrinsic origin of essential HTN remains obscure although many environmental factors are known. Analyses of rare monogenic syndromes of HTN have focused attention on pathways that involve renal sodium handling, and steroid hormone metabolism including the mineralocorticoid receptor activity. The genetic basis of common essential HTN on the other hand is only just becoming accessible through high-throughput approaches. Unbiased genome-wide analyses of BP genomics have identified 43 genetic variants associated with systolic, diastolic BP, and HTN. It is highly likely based on current findings that there are hundreds of such loci with small effects on BP, opening a perspective on the genetic architecture of BP that was unknown before. It is our hope that the knowledge of these and further loci will lead to improved understanding of BP pathophysiology and to the identification of new targets for drug therapy.
Collapse
Affiliation(s)
- Georg B Ehret
- Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland
| | | |
Collapse
|
234
|
Alvarez-Madrazo S, MacKenzie SM, Davies E, Fraser R, Lee WK, Brown M, Caulfield MJ, Dominiczak AF, Farrall M, Lathrop M, Hedner T, Melander O, Munroe PB, Samani N, Stewart PM, Wahlstrand B, Webster J, Palmer CN, Padmanabhan S, Connell JM. Common Polymorphisms in the
CYP11B1
and
CYP11B2
Genes: Evidence for a Digenic Influence on Hypertension. Hypertension 2013; 61:232-9. [DOI: 10.1161/hypertensionaha.112.200741] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Samantha Alvarez-Madrazo
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (S.A-M., S.M.M., E.D., R.F., W-K.L., A.F.D., S.P.); Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (M.B.); Clinical Pharmacology, William Harvey Research Institute, Barts and the London Medical and Dental School, Queen Mary University of London, London, United Kingdom (M.J.C., P.B.M.)
| | - Scott M. MacKenzie
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (S.A-M., S.M.M., E.D., R.F., W-K.L., A.F.D., S.P.); Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (M.B.); Clinical Pharmacology, William Harvey Research Institute, Barts and the London Medical and Dental School, Queen Mary University of London, London, United Kingdom (M.J.C., P.B.M.)
| | - Eleanor Davies
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (S.A-M., S.M.M., E.D., R.F., W-K.L., A.F.D., S.P.); Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (M.B.); Clinical Pharmacology, William Harvey Research Institute, Barts and the London Medical and Dental School, Queen Mary University of London, London, United Kingdom (M.J.C., P.B.M.)
| | - Robert Fraser
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (S.A-M., S.M.M., E.D., R.F., W-K.L., A.F.D., S.P.); Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (M.B.); Clinical Pharmacology, William Harvey Research Institute, Barts and the London Medical and Dental School, Queen Mary University of London, London, United Kingdom (M.J.C., P.B.M.)
| | - Wai-Kwong Lee
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (S.A-M., S.M.M., E.D., R.F., W-K.L., A.F.D., S.P.); Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (M.B.); Clinical Pharmacology, William Harvey Research Institute, Barts and the London Medical and Dental School, Queen Mary University of London, London, United Kingdom (M.J.C., P.B.M.)
| | - Morris Brown
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (S.A-M., S.M.M., E.D., R.F., W-K.L., A.F.D., S.P.); Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (M.B.); Clinical Pharmacology, William Harvey Research Institute, Barts and the London Medical and Dental School, Queen Mary University of London, London, United Kingdom (M.J.C., P.B.M.)
| | - Mark J. Caulfield
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (S.A-M., S.M.M., E.D., R.F., W-K.L., A.F.D., S.P.); Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (M.B.); Clinical Pharmacology, William Harvey Research Institute, Barts and the London Medical and Dental School, Queen Mary University of London, London, United Kingdom (M.J.C., P.B.M.)
| | - Anna F. Dominiczak
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (S.A-M., S.M.M., E.D., R.F., W-K.L., A.F.D., S.P.); Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (M.B.); Clinical Pharmacology, William Harvey Research Institute, Barts and the London Medical and Dental School, Queen Mary University of London, London, United Kingdom (M.J.C., P.B.M.)
| | - Martin Farrall
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (S.A-M., S.M.M., E.D., R.F., W-K.L., A.F.D., S.P.); Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (M.B.); Clinical Pharmacology, William Harvey Research Institute, Barts and the London Medical and Dental School, Queen Mary University of London, London, United Kingdom (M.J.C., P.B.M.)
| | - Mark Lathrop
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (S.A-M., S.M.M., E.D., R.F., W-K.L., A.F.D., S.P.); Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (M.B.); Clinical Pharmacology, William Harvey Research Institute, Barts and the London Medical and Dental School, Queen Mary University of London, London, United Kingdom (M.J.C., P.B.M.)
| | - Thomas Hedner
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (S.A-M., S.M.M., E.D., R.F., W-K.L., A.F.D., S.P.); Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (M.B.); Clinical Pharmacology, William Harvey Research Institute, Barts and the London Medical and Dental School, Queen Mary University of London, London, United Kingdom (M.J.C., P.B.M.)
| | - Olle Melander
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (S.A-M., S.M.M., E.D., R.F., W-K.L., A.F.D., S.P.); Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (M.B.); Clinical Pharmacology, William Harvey Research Institute, Barts and the London Medical and Dental School, Queen Mary University of London, London, United Kingdom (M.J.C., P.B.M.)
| | - Patricia B. Munroe
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (S.A-M., S.M.M., E.D., R.F., W-K.L., A.F.D., S.P.); Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (M.B.); Clinical Pharmacology, William Harvey Research Institute, Barts and the London Medical and Dental School, Queen Mary University of London, London, United Kingdom (M.J.C., P.B.M.)
| | - Nilesh Samani
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (S.A-M., S.M.M., E.D., R.F., W-K.L., A.F.D., S.P.); Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (M.B.); Clinical Pharmacology, William Harvey Research Institute, Barts and the London Medical and Dental School, Queen Mary University of London, London, United Kingdom (M.J.C., P.B.M.)
| | - Paul M. Stewart
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (S.A-M., S.M.M., E.D., R.F., W-K.L., A.F.D., S.P.); Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (M.B.); Clinical Pharmacology, William Harvey Research Institute, Barts and the London Medical and Dental School, Queen Mary University of London, London, United Kingdom (M.J.C., P.B.M.)
| | - Björn Wahlstrand
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (S.A-M., S.M.M., E.D., R.F., W-K.L., A.F.D., S.P.); Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (M.B.); Clinical Pharmacology, William Harvey Research Institute, Barts and the London Medical and Dental School, Queen Mary University of London, London, United Kingdom (M.J.C., P.B.M.)
| | - John Webster
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (S.A-M., S.M.M., E.D., R.F., W-K.L., A.F.D., S.P.); Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (M.B.); Clinical Pharmacology, William Harvey Research Institute, Barts and the London Medical and Dental School, Queen Mary University of London, London, United Kingdom (M.J.C., P.B.M.)
| | - Colin N.A. Palmer
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (S.A-M., S.M.M., E.D., R.F., W-K.L., A.F.D., S.P.); Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (M.B.); Clinical Pharmacology, William Harvey Research Institute, Barts and the London Medical and Dental School, Queen Mary University of London, London, United Kingdom (M.J.C., P.B.M.)
| | - Sandosh Padmanabhan
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (S.A-M., S.M.M., E.D., R.F., W-K.L., A.F.D., S.P.); Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (M.B.); Clinical Pharmacology, William Harvey Research Institute, Barts and the London Medical and Dental School, Queen Mary University of London, London, United Kingdom (M.J.C., P.B.M.)
| | - John M. Connell
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (S.A-M., S.M.M., E.D., R.F., W-K.L., A.F.D., S.P.); Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom (M.B.); Clinical Pharmacology, William Harvey Research Institute, Barts and the London Medical and Dental School, Queen Mary University of London, London, United Kingdom (M.J.C., P.B.M.)
| |
Collapse
|
235
|
Arenas IA, Tremblay J, Deslauriers B, Sandoval J, Šeda O, Gaudet D, Merlo E, Kotchen T, Cowley AW, Hamet P. Dynamic genetic linkage of intermediate blood pressure phenotypes during postural adaptations in a founder population. Physiol Genomics 2012; 45:138-50. [PMID: 23269701 DOI: 10.1152/physiolgenomics.00037.2012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Blood pressure (BP) is a dynamic phenotype that varies rapidly to adjust to changing environmental conditions. Standing upright is a recent evolutionary trait, and genetic factors that influence postural adaptations may contribute to BP variability. We studied the effect of posture on the genetics of BP and intermediate BP phenotypes. We included 384 sib-pairs in 64 sib-ships from families ascertained by early-onset hypertension and dyslipidemia. Blood pressure, three hemodynamic and seven neuroendocrine intermediate BP phenotypes were measured with subjects lying supine and standing upright. The effect of posture on estimates of heritability and genetic covariance was investigated in full pedigrees. Linkage was conducted on 196 candidate genes by sib-pair analyses, and empirical estimates of significance were obtained. A permutation algorithm was implemented to study the postural effect on linkage. ADRA1A, APO, CAST, CORIN, CRHR1, EDNRB, FGF2, GC, GJA1, KCNB2, MMP3, NPY, NR3C2, PLN, TGFBR2, TNFRSF6, and TRHR showed evidence of linkage with any phenotype in the supine position and not upon standing, whereas AKR1B1, CD36, EDNRA, F5, MMP9, PKD2, PON1, PPARG, PPARGC1A, PRKCA, and RET were specifically linked to standing phenotypes. Genetic profiling was undertaken to show genetic interactions among intermediate BP phenotypes and genes specific to each posture. When investigators perform genetic studies exclusively on a single posture, important genetic components of BP are missed. Supine and standing BPs have distinct genetic signatures. Standardized maneuvers influence the results of genetic investigations into BP, thus reflecting its dynamic regulation.
Collapse
Affiliation(s)
- I A Arenas
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
236
|
Lambers Heerspink HJ, de Borst MH, Bakker SJL, Navis GJ. Improving the efficacy of RAAS blockade in patients with chronic kidney disease. Nat Rev Nephrol 2012; 9:112-21. [PMID: 23247573 DOI: 10.1038/nrneph.2012.281] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Reduction of blood pressure and proteinuria by blockade of the renin-angiotensin-aldosterone system (RAAS) has been the cornerstone of renoprotective intervention for patients with chronic kidney disease (CKD) for many years. Despite the proven efficacy of RAAS blockade, however, the reduction in proteinuria is insufficient in many patients, and does not prevent further deterioration of renal function. Short-term studies have shown that a variety of treatment intensification strategies have a beneficial effect on blood pressure and proteinuria, including RAAS blockade using either dose escalation or multiple drugs, and restriction of dietary sodium. Large clinical trials have shown that RAAS blockade with multiple drugs does not improve patients' long-term renal or cardiovascular outcome. By contrast, two post-hoc analyses of landmark trials in nephrology show beneficial renal and cardiovascular effects from avoiding excessive dietary sodium intake during single-agent RAAS blockade therapy. The effects of dietary sodium restriction on renal or cardiovascular outcome still require prospective confirmation. However, current data support the implementation of lifestyle changes to reduce dietary sodium intake in combination with single-agent RAAS blockade, rather than dual-agent RAAS blockade, as a potent and feasible strategy to mitigate the burden of renal and cardiovascular disease in patients with CKD.
Collapse
Affiliation(s)
- Hiddo J Lambers Heerspink
- Department of Clinical Pharmacology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 9700 AD, Groningen, The Netherlands
| | | | | | | |
Collapse
|
237
|
Fava C, Sjögren M, Montagnana M, Danese E, Almgren P, Engström G, Nilsson P, Hedblad B, Guidi GC, Minuz P, Melander O. Prediction of blood pressure changes over time and incidence of hypertension by a genetic risk score in Swedes. Hypertension 2012; 61:319-26. [PMID: 23232644 DOI: 10.1161/hypertensionaha.112.202655] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Recent Genome-Wide Association Studies (GWAS) have pinpointed different single nucleotide polymorphisms consistently associated with blood pressure (BP) and hypertension prevalence. However, little data exist regarding single nucleotide polymorphisms predicting BP variation over time and hypertension incidence. The aim of this study was to confirm the association of a genetic risk score (GRS), based on 29 independent single nucleotide polymorphisms, with cross-sectional BP and hypertension prevalence and to challenge its prediction of BP change over time and hypertension incidence in >17 000 middle-aged Swedes participating in a prospective study, the Malmö Preventive Project, investigated at baseline and over a 23-year average period of follow-up. The GRS was associated with higher systolic and diastolic BP values both at baseline (β ± SEM, 0.968 ± 0.102 mm Hg and 0.585 ± 0.064 mm Hg; P<1E-19 for both) and at reinvestigation (β ± SEM, 1.333 ± 0.161 mm Hg and 0.724 ± 0.086 mm Hg; P<1E-15 for both) and with increased hypertension prevalence (odds ratio [95% CI], 1.192 [1.140-1.245] and 1.144 [1.107-1.183]; P<1E-15 for both). The GRS was positively associated with change (Δ) in BP (β ± SEM, 0.033 ± 0.008 mm Hg/y and 0.023 ± 0.004 mm Hg/y; P<1E-04 for both) and hypertension incidence (odds ratio [95% CI], 1.110 [1.065-1.156]; P=6.7 E-07), independently from traditional risk factors. The relative weight of the GRS was lower in magnitude than obesity or prehypertension, but comparable with diabetes mellitus or a positive family history of hypertension. A C-statistics analysis does not show any improvement in the prediction of incident hypertension on top of traditional risk factors. Our data from a large cohort study show that a GRS is independently associated with BP increase and incidence of hypertension.
Collapse
Affiliation(s)
- Cristiano Fava
- Department of Clinical Sciences, Lund University, University Hospital of Malmö, Sweden.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
238
|
Pal A, McCarthy MI. The genetics of type 2 diabetes and its clinical relevance. Clin Genet 2012; 83:297-306. [PMID: 23167659 DOI: 10.1111/cge.12055] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 11/01/2012] [Accepted: 11/01/2012] [Indexed: 12/13/2022]
Abstract
The increasing worldwide prevalence of type 2 diabetes (T2D) motivates efforts to use genetics to define key pathways involved in disease predisposition, and thereby to improve management of the disease. Research over the past 5 years has taken the total number of genetic loci implicated in T2D susceptibility beyond 60, and the emphasis is now shifting to the translation of these genetic insights into clinical value. Clinical translation may flow from the identification of novel therapeutic targets, but opportunities also exist with respect to individual prediction, diagnostic biomarkers and therapeutic optimization. To date, the main clinical impact has been seen for relatively rare, monogenic forms of diabetes rather than common T2D. However, the advent of high throughput sequencing approaches may herald discovery of rare and low frequency variants that offer greater translational potential.
Collapse
Affiliation(s)
- A Pal
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | | |
Collapse
|
239
|
Doris PA. Genetic susceptibility to hypertensive renal disease. Cell Mol Life Sci 2012; 69:3751-63. [PMID: 22562581 PMCID: PMC3422437 DOI: 10.1007/s00018-012-0996-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 04/04/2012] [Accepted: 04/06/2012] [Indexed: 12/21/2022]
Abstract
Hypertensive renal disease occurs at increased frequency among the relatives of patients with this disease compared to individuals who lack a family history of disease. This suggests a heritable risk in which genetic variation may play a role. These observations have motivated a search for genetic variation contributing to this risk in both experimental animal models and in human populations. Studies of animal models indicate the capacity of natural genetic variants to contribute to disease risk and have produced a few insights into the disease mechanism. In its current phase, human population genetic studies have sought to associate genetic variation with disease in large populations by testing genotypes at a large number of common genetic variations in the genome, expecting that common genetic variants contributing to renal disease risk will be identified. These genome-wide association studies (GWAS) have been productive and are a clear technical success; they have also identified narrowly defined loci and genes containing variation contributing to disease risk. Further extension and refinement of these GWAS are likely to extend this success. However, it is also clear that few additional variants with substantial effects accounting for the greatest part of heritability will be uncovered by GWAS. This raises an interesting biological question regarding where the remaining unaccounted heritable risk may be located. At present, much consideration is being given to this question and to the challenge of testing hypotheses that lead from the various alternative mechanisms under consideration. One result of the progress of GWAS is likely to be a renewed interest in mechanisms by which related individuals can share and transmit traits independently of Mendelian inheritance. This paper reviews the current progress in this area and considers other mechanisms by which familial aggregation of risk for renal disease may arise.
Collapse
Affiliation(s)
- Peter A Doris
- Institute of Molecular Medicine, University of Texas HSC at Houston, Houston, TX 77030, USA.
| |
Collapse
|
240
|
Langley SR, Bottolo L, Kunes J, Zicha J, Zidek V, Hubner N, Cook SA, Pravenec M, Aitman TJ, Petretto E. Systems-level approaches reveal conservation of trans-regulated genes in the rat and genetic determinants of blood pressure in humans. Cardiovasc Res 2012; 97:653-65. [PMID: 23118132 DOI: 10.1093/cvr/cvs329] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
AIMS Human genome-wide association studies (GWAS) of hypertension identified only few susceptibility loci with large effect that were replicated across populations. The vast majority of genes detected by GWAS has small effect and the regulatory mechanisms through which these genetic variants cause disease remain mostly unclear. Here, we used comparative genomics between human and an established rat model of hypertension to explore the transcriptional mechanisms mediating the effect of genes identified in 15 hypertension GWAS. METHODS AND RESULTS Time series analysis of radiotelemetric blood pressure (BP) was performed to assess 11 parameters of BP variation in recombinant inbred strains derived from the spontaneously hypertensive rat. BP data were integrated with ∼27 000 expression quantative trait loci (eQTLs) mapped across seven tissues, detecting >8000 significant associations between eQTL genes and BP variation in the rat. We then compiled a large catalogue of human genes from GWAS of hypertension and identified a subset of 2292 rat-human orthologous genes. Expression levels for 795 (34%) of these genes correlated with BP variation across rat tissues: 51 genes were cis-regulated, whereas 459 were trans-regulated and enriched for 'calcium signalling pathway' (P = 9.6 × 10(-6)) and 'ion channel' genes (P = 3.5 × 10(-7)), which are important determinants of hypertension. We identified 158 clusters of trans-eQTLs, annotated the underlying 'master regulator' genes and found significant over-representation in the human hypertension gene set (enrichment P = 5 × 10(-4)). CONCLUSION We showed extensive conservation of trans-regulated genes and their master regulators between rat and human hypertension. These findings reveal that small-effect genes associated with hypertension by human GWAS are likely to exert their action through coordinate regulation of pathogenic pathways.
Collapse
Affiliation(s)
- Sarah R Langley
- MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
241
|
Orozco G, Ioannidis JPA, Morris A, Zeggini E. Sex-specific differences in effect size estimates at established complex trait loci. Int J Epidemiol 2012; 41:1376-82. [PMID: 22825589 PMCID: PMC3465768 DOI: 10.1093/ije/dys104] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2012] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Genetic differences between men and women may contribute to sex differences in prevalence and progression of many common complex diseases. Using the WTCCC GWAS, we analysed whether there are sex-specific differences in effect size estimates at 142 established loci for seven complex diseases: rheumatoid arthritis, type 1 diabetes (T1D), Crohn's disease, type 2 diabetes (T2D), hypertension, coronary artery disease and bipolar disorder. METHODS For each Single nucleotide polymorphism (SNP), we calculated the per-allele odds ratio for each sex and the relative odds ratios (RORs; the effect size is higher in men with ROR greater than one). RORs were then meta-analysed across loci within each disease and across diseases. RESULTS For each disease, summary RORs were not different from one, but there was between-SNP heterogeneity in the RORs for T1D and T2D. Four loci in T1D, three in Crohn's disease and three in T2D showed differences in the genetic effect between men and women (P<0.05). We probed these differences in additional independent replication samples for T1D and T2D. The differences remained for the T1D loci CTSH, 17q21 and 20p13 and the T2D locus BCL11A, when WTCCC data and replication data were meta-analysed. Only CTSH showed different genetic effect between men and women in the replication data alone. CONCLUSION Our results exclude the presence of large and frequent differences in the effect size estimates between men and women for the established loci in the seven common diseases explored. Documenting small differences in genetic effects between men and women requires large studies and systematic evaluation.
Collapse
Affiliation(s)
- Gisela Orozco
- Arthritis Research UK Epidemiology Unit, Manchester Academic Health Science Centre, The University of Manchester, UK.
| | | | | | | |
Collapse
|
242
|
El Shamieh S, Visvikis-Siest S. Genetic biomarkers of hypertension and future challenges integrating epigenomics. Clin Chim Acta 2012; 414:259-65. [PMID: 23010416 DOI: 10.1016/j.cca.2012.09.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 09/10/2012] [Accepted: 09/16/2012] [Indexed: 12/15/2022]
Abstract
Essential hypertension is a multifactorial disease, considered to be one of the world's greatest public health problems. Despite recent, major, technical advances aiming to elucidate its genetic component, the discovered biomarkers up to now were reported to have only small effects, explaining consequently a tiny fraction of its phenotypic variance and resulting in a large proportion of missing heritability. Likewise, little evidence is available with regard to the epigenetic regulation of essential hypertension, since no robust biomarkers have yet been reported. In the current review, we discuss the main approaches used exclusively to study the genetics and epigenetics of essential hypertension, the biomarkers identified, their clinical utility and the difficulties to be overcome. Furthermore, we propose a new category of functional genetic-epigenetic biomarkers, eMethSNPs, and we provide their hypothetical gene expression profiles for a genetic functional regulation of hypertension via DNA methylation. Though believed to be infrequent, eMethSNPs could constitute a new category of mechanistically-based genetic biomarkers predisposing to essential hypertension.
Collapse
Affiliation(s)
- Said El Shamieh
- Université de Lorraine, Génétique Cardiovasculaire, EA-4373, Nancy 54000, France.
| | | |
Collapse
|
243
|
Reznichenko A, Böger CA, Snieder H, van den Born J, de Borst MH, Damman J, van Dijk MCRF, van Goor H, Hepkema BG, Hillebrands JL, Leuvenink HGD, Niesing J, Bakker SJL, Seelen M, Navis G. UMOD as a susceptibility gene for end-stage renal disease. BMC MEDICAL GENETICS 2012; 13:78. [PMID: 22947327 PMCID: PMC3495046 DOI: 10.1186/1471-2350-13-78] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 08/31/2012] [Indexed: 11/10/2022]
Abstract
Background In recent genetic association studies, common variants including rs12917707 in the UMOD locus have shown strong evidence of association with eGFR, prevalent and incident chronic kidney disease and uromodulin urinary concentration in general population cohorts. The association of rs12917707 with end-stage renal disease (ESRD) in a recent case-control study was only nominally significant. Methods To investigate whether rs12917707 associates with ESRD, graft failure (GF) and urinary uromodulin levels in an independent cohort, we genotyped 1142 ESRD patients receiving a renal transplantation and 1184 kidney donors as controls. After transplantation, 1066 renal transplant recipients were followed up for GF. Urinary uromodulin concentration was measured at median [IQR] 4.2 [2.2-6.1] yrs after kidney transplantation. Results The rs12917707 minor allele showed association with lower risk of ESRD (OR 0.89 [0.76-1.03], p = 0.04) consistent in effect size and direction with the previous report (Böger et al, PLoS Genet 2011). Meta-analysis of these findings showed significant association of rs12917707 with ESRD (OR 0.91 [0.85-98], p = 0.008). In contrast, rs12917707 was not associated with incidence of GF. Urinary uromodulin concentration was lower in recipients-carriers of the donor rs12917707 minor allele as compared to non-carriers, again consistent with previous observations in general population cohorts. Conclusions Our study thus corroborates earlier evidence and independently confirms the association between UMOD and ESRD.
Collapse
Affiliation(s)
- Anna Reznichenko
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, A. Deusinglaan 1, Groningen 9713AV, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
244
|
Liu C, Yang Q, Hwang SJ, Sun F, Johnson AD, Shirihai OS, Vasan RS, Levy D, Schwartz F. Association of genetic variation in the mitochondrial genome with blood pressure and metabolic traits. Hypertension 2012; 60:949-56. [PMID: 22949535 DOI: 10.1161/hypertensionaha.112.196519] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Elevated blood pressure (BP) is a major risk factor for cardiovascular disease. Several studies have noted a consistent maternal effect on BP; consequently, mitochondrial DNA variation has become an additional target of investigation of the missing BP heritability. Analyses of common mitochondrial DNA polymorphisms, however, have not found evidence of association with hypertension. To explore associations of uncommon (frequency>5%) mitochon drial DNA variants with BP, we identified uncommon/rare variants through sequencing the entire mitochondrial genome in 32 unrelated individuals with extreme-high BP in the Framingham Heart Study and genotyped 40 mitochondrial single nucleotide polymorphisms in 7219 Framingham Heart Study participants. The nonsynonymous mitochondrial single nucleotide polymorphism 5913G>A (Asp4Asn) in the cytochrome c oxidase subunit 1 of respiratory complex IV demonstrated significant associations with BP and fasting blood glucose (FBG) levels. Individuals with the rare 5913A allele had, on average, 7-mm Hg higher systolic BP at baseline (Pempirical=0.05) and 17-mg/dL higher mean FBG over 25 years of follow-up (Pempirical=0.009). Significant associations with FBG levels were also detected for nonsynonymous mitochondrial single nucleotide polymorphism 3316G>A (Ala4Thr) in the NADH dehydrogenase subunit 1 of complex I. On average, individuals with rare allele 3316A had 17- and 25-mg/dL higher FBG at baseline (Pempirical=0.01) and over 25 years of follow-up (Pempirical=0.007). Our findings provide the first evidence of putative association of variants in the mitochondrial genome with systolic BP and FBG in the general population. Replication in independent samples, however, is needed to confirm these putative associations.
Collapse
Affiliation(s)
- Chunyu Liu
- National Heart, Lung, and Blood Institute’s Framingham Heart Study, 73 Mt Wayte Ave, Suite 2, Framingham, MA 01702, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
245
|
Taal HR, van den Hil LCL, Hofman A, van der Heijden AJ, Jaddoe VWV. Genetic variants associated with adult blood pressure and kidney function do not affect fetal kidney volume. The Generation R Study. Early Hum Dev 2012; 88:711-6. [PMID: 22445569 DOI: 10.1016/j.earlhumdev.2012.02.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 02/20/2012] [Accepted: 02/24/2012] [Indexed: 01/11/2023]
Abstract
BACKGROUND Smaller kidneys with reduced number of nephrons in early life lead to impaired kidney function and risk for hypertension and chronic kidney disease. These associations might be partly explained by common genetic variation. AIMS To assess the associations between common genetic variants, which have recently shown to be associated with blood pressure or kidney function, with fetal kidney volume. STUDY DESIGN A prospective population based cohort study in Rotterdam, The Netherlands. SUBJECTS 855 children, followed from early fetal life onwards (born 2003-2005). PREDICTOR Common genetic variants previously associated with blood pressure or kidney function. OUTCOME MEASURES Combined third trimester fetal kidney volume. RESULTS After taking into account multiple testing, only rs12940887 (near ZNF652) was significantly associated with fetal kidney volume (β: 0.88 (95% CI: 0.40; 1.37) cm(3) per minor allele, P-value<0.001), but the effect showed the opposite direction as expected. The remaining common genetic variants were not associated with fetal kidney volume. We also did not find associations of genetic variants previously shown to affect newborn kidney volume, with third trimester fetal kidney volume. CONCLUSIONS Our results suggest that common genetic variants, associated with kidney function or disease and blood pressure, do not affect the third trimester fetal kidney volume. Further studies are needed to elucidate the mechanisms underlying the associations between small kidney size and increased risks of hypertension and impaired kidney function in adulthood.
Collapse
Affiliation(s)
- H Rob Taal
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | | | | |
Collapse
|
246
|
Li-Na Pu, Ze Zhao, Yuan-Ting Zhang. Investigation on Cardiovascular Risk Prediction Using Genetic Information. ACTA ACUST UNITED AC 2012; 16:795-808. [DOI: 10.1109/titb.2012.2205009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
247
|
Hypertension, cardiovascular risk and polymorphisms in genes controlling the cytochrome P450 pathway of arachidonic acid: A sex-specific relation? Prostaglandins Other Lipid Mediat 2012; 98:75-85. [DOI: 10.1016/j.prostaglandins.2011.11.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2011] [Revised: 11/16/2011] [Accepted: 11/21/2011] [Indexed: 01/11/2023]
|
248
|
Fava C, Montagnana M, Guidi GC, Melander O. From circulating biomarkers to genomics and imaging in the prediction of cardiovascular events in the general population. Ann Med 2012; 44:433-47. [PMID: 21623699 DOI: 10.3109/07853890.2011.582511] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death and disability worldwide. In the last decades numerous markers have been considered and investigated for the prediction of CV events, but only a few of them resulted in improved global risk assessment beyond traditional risk factors when incorporated into coronary evaluation scores. Recent genetic studies have pointed out a few but consistent loci or genes which are independently associated with CV risk. The idea is fascinating that these genetic markers could lead to improved individual CV risk assessment and tailored pharmacological interventions. In this brief review we will not make a systematic review of all non-genetic and genetic markers of CV risk but we will try to make a brief overview of the most interesting ones with the aim to underline potential 'pros' and 'cons' of their implementation in clinical practice.
Collapse
Affiliation(s)
- Cristiano Fava
- Department of Clinical Sciences, Lund University, University Hospital of Malmö, Sweden
| | | | | | | |
Collapse
|
249
|
Abstract
Familial risk in hypertensive renal disease has stimulated a search for genetic variation contributing to this risk. The current phase of population genetic studies has sought to associate genetic variation with disease in large populations by testing genotypes at a large number of common genetic variations in the genome, expecting that common genetic variants contributing to renal disease risk will be identified. These genome-wide association studies (GWAS) have been productive and are a clear technical success. It is also clear that narrowly defined loci and genes containing variation contributing to disease risk have been identified. Further extension and refinement of these GWAS are likely to extend this success. However, it is also clear that few if any variants with substantial effects accounting for the greatest part of heritability will be uncovered by GWAS. This raises an interesting biological question regarding where the remaining heritable risk may be located. One result of the progress of GWAS is likely to be a renewed interest in mechanisms by which related individuals can share and transmit traits independently of Mendelian inheritance. This paper reviews current progress in this area and considers other mechanisms by which familial aggregation of risk for renal disease may arise.
Collapse
Affiliation(s)
- Michael C Braun
- Division of Pediatric Nephrology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX 77030, USA
| | | |
Collapse
|
250
|
Köttgen A, Yang Q, Shimmin LC, Tin A, Schaeffer C, Coresh J, Liu X, Rampoldi L, Hwang SJ, Boerwinkle E, Hixson JE, Kao WHL, Fox CS. Association of estimated glomerular filtration rate and urinary uromodulin concentrations with rare variants identified by UMOD gene region sequencing. PLoS One 2012; 7:e38311. [PMID: 22693617 PMCID: PMC3365030 DOI: 10.1371/journal.pone.0038311] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 05/08/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Recent genome-wide association studies (GWAS) have identified common variants in the UMOD region associated with kidney function and disease in the general population. To identify novel rare variants as well as common variants that may account for this GWAS signal, the exons and 4 kb upstream region of UMOD were sequenced. METHODOLOGY/PRINCIPAL FINDINGS Individuals (n = 485) were selected based on presence of the GWAS risk haplotype and chronic kidney disease (CKD) in the ARIC Study and on the extremes of of the UMOD gene product, uromodulin, in urine (Tamm Horsfall protein, THP) in the Framingham Heart Study (FHS). Targeted sequencing was conducted using capillary based Sanger sequencing (3730 DNA Analyzer). Variants were tested for association with THP concentrations and estimated glomerular filtration rate (eGFR), and identified non-synonymous coding variants were genotyped in up to 22,546 follow-up samples. Twenty-four and 63 variants were identified in the 285 ARIC and 200 FHS participants, respectively. In both studies combined, there were 33 common and 54 rare (MAF<0.05) variants. Five non-synonymous rare variants were identified in FHS; borderline enrichment of rare variants was found in the extremes of THP (SKAT p-value = 0.08). Only V458L was associated with THP in the FHS general-population validation sample (p = 9*10(-3), n = 2,522), but did not show direction-consistent and significant association with eGFR in both the ARIC (n = 14,635) and FHS (n = 7,520) validation samples. Pooling all non-synonymous rare variants except V458L together showed non-significant associations with THP and eGFR in the FHS validation sample. Functional studies of V458L revealed no alternations in protein trafficking. CONCLUSIONS/SIGNIFICANCE Multiple novel rare variants in the UMOD region were identified, but none were consistently associated with eGFR in two independent study samples. Only V458L had modest association with THP levels in the general population and thus could not account for the observed GWAS signal.
Collapse
Affiliation(s)
- Anna Köttgen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Renal Division, Freiburg University Clinic, Freiburg, Germany
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachussets, United States of America
| | - Lawrence C. Shimmin
- Human Genetics Center, Division of Epidemiology and Disease Control, UT-Houston School of Public Health, Houston, Texas, United States of America
| | - Adrienne Tin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Céline Schaeffer
- Dulbecco Telethon Institute and Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Xuan Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachussets, United States of America
| | - Luca Rampoldi
- Dulbecco Telethon Institute and Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Shih-Jen Hwang
- NHLBI's Framingham Heart Study and the Center for Population Studies, Framingham, Massachussets, United States of America
| | - Eric Boerwinkle
- Human Genetics Center, Division of Epidemiology and Disease Control, UT-Houston School of Public Health, Houston, Texas, United States of America
| | - James E. Hixson
- Human Genetics Center, Division of Epidemiology and Disease Control, UT-Houston School of Public Health, Houston, Texas, United States of America
| | - W. H. Linda Kao
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Caroline S. Fox
- NHLBI's Framingham Heart Study and the Center for Population Studies, Framingham, Massachussets, United States of America
- Division of Endocrinology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachussets, United States of America
| |
Collapse
|