101
|
Song YM, Sung J, Lee K. Genetic and environmental influences on the associations between change in kidney function and changes in cardiometabolic factors in Koreans. Clin Exp Nephrol 2016; 21:474-480. [DOI: 10.1007/s10157-016-1295-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 06/08/2016] [Indexed: 12/24/2022]
|
102
|
Luttropp K, Debowska M, Lukaszuk T, Bobrowski L, Carrero JJ, Qureshi AR, Stenvinkel P, Lindholm B, Waniewski J, Nordfors L. Genotypic and phenotypic predictors of inflammation in patients with chronic kidney disease. Nephrol Dial Transplant 2016; 31:2033-2040. [PMID: 27190335 DOI: 10.1093/ndt/gfw066] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 03/07/2016] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND In complex diseases such as chronic kidney disease (CKD), the risk of clinical complications is determined by interactions between phenotypic and genotypic factors. However, clinical epidemiological studies rarely attempt to analyse the combined effect of large numbers of phenotype and genotype features. We have recently shown that the relaxed linear separability (RLS) model of feature selection can address such complex issues. Here, it is applied to identify risk factors for inflammation in CKD. METHODS The RLS model was applied in 225 CKD stage 5 patients sampled in conjunction with dialysis initiation. Fifty-seven anthropometric or biochemical measurements and 79 genetic polymorphisms were entered into the model. The model was asked to identify phenotypes and genotypes that, when combined, could separate inflamed from non-inflamed patients. Inflammation was defined as a high-sensitivity C-reactive protein concentration above the median (5 mg/L). RESULTS Among the 60 genotypic and phenotypic features predicting inflammation, 31 were genetic. Among the 10 strongest predictors of inflammation, 8 were single nucleotide polymorphisms located in the NAMPT, CIITA, BMP2 and PIK3CB genes, whereas fibrinogen and bone mineral density were the only phenotypic biomarkers. CONCLUSION These results indicate a larger involvement of hereditary factors in inflammation than might have been expected and suggest that inclusion of genotype features in risk assessment studies is critical. The RLS model demonstrates that inflammation in CKD is determined by an extensive panel of factors and may prove to be a suitable tool that could enable a much-needed multifactorial approach as opposed to the commonly utilized single-factor analysis.
Collapse
Affiliation(s)
- Karin Luttropp
- Department of Molecular Medicine and Surgery, Neurogenetics Division, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Malgorzata Debowska
- Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | | | - Leon Bobrowski
- Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland.,Bialystok University of Technology, Bialystok, Poland
| | - Juan Jesus Carrero
- Department of Molecular Medicine and Surgery, Neurogenetics Division, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Abdul Rashid Qureshi
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Peter Stenvinkel
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Lindholm
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Jacek Waniewski
- Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Louise Nordfors
- Department of Molecular Medicine and Surgery, Neurogenetics Division, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
103
|
Identification of genetic loci stratified by diabetic status and microRNA related SNPs influencing kidney function in Korean populations. Genes Genomics 2016. [DOI: 10.1007/s13258-016-0411-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
104
|
Conserved differences in protein sequence determine the human pathogenicity of Ebolaviruses. Sci Rep 2016; 6:23743. [PMID: 27009368 PMCID: PMC4806318 DOI: 10.1038/srep23743] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 03/14/2016] [Indexed: 12/23/2022] Open
Abstract
Reston viruses are the only Ebolaviruses that are not pathogenic in humans. We analyzed 196 Ebolavirus genomes and identified specificity determining positions (SDPs) in all nine Ebolavirus proteins that distinguish Reston viruses from the four human pathogenic Ebolaviruses. A subset of these SDPs will explain the differences in human pathogenicity between Reston and the other four ebolavirus species. Structural analysis was performed to identify those SDPs that are likely to have a functional effect. This analysis revealed novel functional insights in particular for Ebolavirus proteins VP40 and VP24. The VP40 SDP P85T interferes with VP40 function by altering octamer formation. The VP40 SDP Q245P affects the structure and hydrophobic core of the protein and consequently protein function. Three VP24 SDPs (T131S, M136L, Q139R) are likely to impair VP24 binding to human karyopherin alpha5 (KPNA5) and therefore inhibition of interferon signaling. Since VP24 is critical for Ebolavirus adaptation to novel hosts, and only a few SDPs distinguish Reston virus VP24 from VP24 of other Ebolaviruses, human pathogenic Reston viruses may emerge. This is of concern since Reston viruses circulate in domestic pigs and can infect humans, possibly via airborne transmission.
Collapse
|
105
|
Liu Y, Zhou J, Luo X, Yang C, Zhang Y, Shi S. Association of RAC1 Gene Polymorphisms with Primary End-Stage Renal Disease in Chinese Renal Recipients. PLoS One 2016; 11:e0148270. [PMID: 26841219 PMCID: PMC4739498 DOI: 10.1371/journal.pone.0148270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 01/15/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND/OBJECTIVE RAC1 gene could influence susceptibility to renal failure by altering the activity and expression of Rac1, which is a member of the Rho family of small GTP-binding proteins. In clinical practice, renal transplantation provides the optimal treatment for people with end-stage renal disease (ESRD). The objective of this present study was to determine whether the RAC1 gene polymorphisms were associated with primary ESRD susceptibility in Chinese renal recipients. METHODS Six single nucleotide polymorphisms (SNPs) of RAC1 gene, including rs836488 T>C, rs702482 A>T, rs10951982 G>A, rs702483 A>G, rs6954996 G>A, and rs9374 G>A, were genotyped in 300 renal transplant recipients (cases) and 998 healthy Chinese subjects (controls) by using TaqMan SNP genotyping assay. Allele, genotype, and haplotype frequencies of the six SNPs were compared between cases and controls. Odds ratios (OR) and 95% confidence intervals (CI) were calculated in logistic regression models to evaluate the associations of the six SNPs with ESRD risk. RESULTS The genotype distributions for the six SNPs in controls were consistent with Hardy-Weinberg equilibrium (P > 0.05). Association analysis revealed that three SNPs were significantly associated with ESRD risk. Positive associations with ESRD risk were found for the rs836488, rs702482, and rs702483 in the co-dominant model (minor allele homozygotes versus major allele homozygotes); specifically, the frequencies of the minor allele homozygotes and the minor allele for the three SNPs were higher in the cases than in the controls. In addition, these three SNPs also had associations with increased ESRD risk under the additive model (P < 0.05), and positive associations were also found for the rs836488 in the dominant model (P < 0.05) and for the rs702483 in the recessive model (P < 0.05). All these associations were independent of confounding factors. The other three SNPs (rs10951982, rs6954996, and rs9374), in all comparison models, were not associated with ESRD risk (P > 0.05). In haplotype analysis, carriers with "C-T-G-G-G-G" haplotype had a significantly higher risk of ESRD compared with the most common haplotype "T-A-G-A-G-G" (P = 0.011, OR = 1.46, 95% CI = 1.09-1.94). CONCLUSION This study suggested that polymorphisms of RAC1 gene might influence the susceptibility to ESRD in Chinese Han population. Further studies are necessary to confirm our findings.
Collapse
Affiliation(s)
- Yani Liu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiali Zhou
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaomei Luo
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunxiao Yang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaojun Shi
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- * E-mail:
| |
Collapse
|
106
|
Wuttke M, Wong CS, Wühl E, Epting D, Luo L, Hoppmann A, Doyon A, Li Y, CKDGen Consortium, Sözeri B, Thurn D, Helmstädter M, Huber TB, Blydt-Hansen TD, Kramer-Zucker A, Mehls O, Melk A, Querfeld U, Furth SL, Warady BA, Schaefer F, Köttgen A. Genetic loci associated with renal function measures and chronic kidney disease in children: the Pediatric Investigation for Genetic Factors Linked with Renal Progression Consortium. Nephrol Dial Transplant 2016; 31:262-9. [PMID: 26420894 PMCID: PMC4829056 DOI: 10.1093/ndt/gfv342] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 08/26/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) in children is characterized by rapid progression and a high incidence of end-stage renal disease and therefore constitutes an important health problem. While unbiased genetic screens have identified common risk variants influencing renal function and CKD in adults, the presence and identity of such variants in pediatric CKD are unknown. METHODS The international Pediatric Investigation for Genetic Factors Linked with Renal Progression (PediGFR) Consortium comprises three pediatric CKD cohorts: Chronic Kidney Disease in Children (CKiD), Effect of Strict Blood Pressure Control and ACE Inhibition on the Progression of CRF in Pediatric Patients (ESCAPE) and Cardiovascular Comorbidity in Children with CKD (4C). Clean genotype data from > 10 million genotyped or imputed single-nucleotide polymorphisms (SNPs) were available for 1136 patients with measurements of serum creatinine at study enrollment. Genome-wide association studies were conducted to relate the SNPs to creatinine-based estimated glomerular filtration rate (eGFR crea) and proteinuria (urinary albumin- or protein-to-creatinine ratio ≥ 300 and ≥ 500 mg/g, respectively). In addition, European-ancestry PediGFR patients (cases) were compared with 1347 European-ancestry children without kidney disease (controls) to identify genetic variants associated with the presence of CKD. RESULTS SNPs with suggestive association P-values < 1 × 10(-5) were identified in 10 regions for eGFR crea, four regions for proteinuria and six regions for CKD including some plausible biological candidates. No SNP was associated at genome-wide significance (P < 5 × 10(-8)). Investigation of the candidate genes for proteinuria in adults from the general population provided support for a region on chromosome 15 near RSL24D1/UNC13C/RAB27A. Conversely, targeted investigation of genes harboring GFR-associated variants in adults from the general population did not reveal significantly associated SNPs in children with CKD. CONCLUSIONS Our findings suggest that larger collaborative efforts will be needed to draw reliable conclusions about the presence and identity of common variants associated with eGFR, proteinuria and CKD in pediatric populations.
Collapse
Affiliation(s)
- Matthias Wuttke
- Renal Division, Department of Internal Medicine, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Craig S. Wong
- Division of Pediatric Nephrology, University of New Mexico Children's Hospital, Albuquerque, NM, USA
| | - Elke Wühl
- Division of Pediatric Nephrology, University Medical Center Heidelberg, Heidelberg, Germany
| | - Daniel Epting
- Renal Division, Department of Internal Medicine, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Li Luo
- Division of Epidemiology, Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Anselm Hoppmann
- Renal Division, Department of Internal Medicine, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Anke Doyon
- Division of Pediatric Nephrology, University Medical Center Heidelberg, Heidelberg, Germany
| | - Yong Li
- Renal Division, Department of Internal Medicine, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - CKDGen Consortium
- Renal Division, Department of Internal Medicine, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
- Division of Pediatric Nephrology, University of New Mexico Children's Hospital, Albuquerque, NM, USA
- Division of Pediatric Nephrology, University Medical Center Heidelberg, Heidelberg, Germany
- Division of Epidemiology, Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
- Faculty of Medicine, Ege University, Izmir, Turkey
- Department of Pediatric Nephrology, Hannover Medical School, Hannover, Germany
- University of Manitoba, Winnipeg, Manitoba, Canada
- Charite Universitätsmedizin Berlin, Berlin, Germany
- Departments of Pediatrics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, Childrens Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Pediatric Nephrology, Children's Mercy Hospital, Kansas City, MO, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Betül Sözeri
- Faculty of Medicine, Ege University, Izmir, Turkey
| | - Daniela Thurn
- Department of Pediatric Nephrology, Hannover Medical School, Hannover, Germany
| | - Martin Helmstädter
- Renal Division, Department of Internal Medicine, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Tobias B. Huber
- Renal Division, Department of Internal Medicine, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | | | - Albrecht Kramer-Zucker
- Renal Division, Department of Internal Medicine, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Otto Mehls
- Division of Pediatric Nephrology, University Medical Center Heidelberg, Heidelberg, Germany
| | - Anette Melk
- Department of Pediatric Nephrology, Hannover Medical School, Hannover, Germany
| | - Uwe Querfeld
- Charite Universitätsmedizin Berlin, Berlin, Germany
| | - Susan L. Furth
- Departments of Pediatrics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, Childrens Hospital of Philadelphia, Philadelphia, PA, USA
| | - Bradley A. Warady
- Division of Pediatric Nephrology, Children's Mercy Hospital, Kansas City, MO, USA
| | - Franz Schaefer
- Division of Pediatric Nephrology, University Medical Center Heidelberg, Heidelberg, Germany
| | - Anna Köttgen
- Renal Division, Department of Internal Medicine, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
107
|
Pattaro C, Teumer A, Gorski M, Chu AY, Li M, Mijatovic V, Garnaas M, Tin A, Sorice R, Li Y, Taliun D, Olden M, Foster M, Yang Q, Chen MH, Pers TH, Johnson AD, Ko YA, Fuchsberger C, Tayo B, Nalls M, Feitosa MF, Isaacs A, Dehghan A, d'Adamo P, Adeyemo A, Dieffenbach AK, Zonderman AB, Nolte IM, van der Most PJ, Wright AF, Shuldiner AR, Morrison AC, Hofman A, Smith AV, Dreisbach AW, Franke A, Uitterlinden AG, Metspalu A, Tonjes A, Lupo A, Robino A, Johansson Å, Demirkan A, Kollerits B, Freedman BI, Ponte B, Oostra BA, Paulweber B, Krämer BK, Mitchell BD, Buckley BM, Peralta CA, Hayward C, Helmer C, Rotimi CN, Shaffer CM, Müller C, Sala C, van Duijn CM, Saint-Pierre A, Ackermann D, Shriner D, Ruggiero D, Toniolo D, Lu Y, Cusi D, Czamara D, Ellinghaus D, Siscovick DS, Ruderfer D, Gieger C, Grallert H, Rochtchina E, Atkinson EJ, Holliday EG, Boerwinkle E, Salvi E, Bottinger EP, Murgia F, Rivadeneira F, Ernst F, Kronenberg F, Hu FB, Navis GJ, Curhan GC, Ehret GB, Homuth G, Coassin S, Thun GA, Pistis G, Gambaro G, Malerba G, Montgomery GW, Eiriksdottir G, Jacobs G, Li G, Wichmann HE, Campbell H, Schmidt H, et alPattaro C, Teumer A, Gorski M, Chu AY, Li M, Mijatovic V, Garnaas M, Tin A, Sorice R, Li Y, Taliun D, Olden M, Foster M, Yang Q, Chen MH, Pers TH, Johnson AD, Ko YA, Fuchsberger C, Tayo B, Nalls M, Feitosa MF, Isaacs A, Dehghan A, d'Adamo P, Adeyemo A, Dieffenbach AK, Zonderman AB, Nolte IM, van der Most PJ, Wright AF, Shuldiner AR, Morrison AC, Hofman A, Smith AV, Dreisbach AW, Franke A, Uitterlinden AG, Metspalu A, Tonjes A, Lupo A, Robino A, Johansson Å, Demirkan A, Kollerits B, Freedman BI, Ponte B, Oostra BA, Paulweber B, Krämer BK, Mitchell BD, Buckley BM, Peralta CA, Hayward C, Helmer C, Rotimi CN, Shaffer CM, Müller C, Sala C, van Duijn CM, Saint-Pierre A, Ackermann D, Shriner D, Ruggiero D, Toniolo D, Lu Y, Cusi D, Czamara D, Ellinghaus D, Siscovick DS, Ruderfer D, Gieger C, Grallert H, Rochtchina E, Atkinson EJ, Holliday EG, Boerwinkle E, Salvi E, Bottinger EP, Murgia F, Rivadeneira F, Ernst F, Kronenberg F, Hu FB, Navis GJ, Curhan GC, Ehret GB, Homuth G, Coassin S, Thun GA, Pistis G, Gambaro G, Malerba G, Montgomery GW, Eiriksdottir G, Jacobs G, Li G, Wichmann HE, Campbell H, Schmidt H, Wallaschofski H, Völzke H, Brenner H, Kroemer HK, Kramer H, Lin H, Leach IM, Ford I, Guessous I, Rudan I, Prokopenko I, Borecki I, Heid IM, Kolcic I, Persico I, Jukema JW, Wilson JF, Felix JF, Divers J, Lambert JC, Stafford JM, Gaspoz JM, Smith JA, Faul JD, Wang JJ, Ding J, Hirschhorn JN, Attia J, Whitfield JB, Chalmers J, Viikari J, Coresh J, Denny JC, Karjalainen J, Fernandes JK, Endlich K, Butterbach K, Keene KL, Lohman K, Portas L, Launer LJ, Lyytikäinen LP, Yengo L, Franke L, Ferrucci L, Rose LM, Kedenko L, Rao M, Struchalin M, Kleber ME, Cavalieri M, Haun M, Cornelis MC, Ciullo M, Pirastu M, de Andrade M, McEvoy MA, Woodward M, Adam M, Cocca M, Nauck M, Imboden M, Waldenberger M, Pruijm M, Metzger M, Stumvoll M, Evans MK, Sale MM, Kähönen M, Boban M, Bochud M, Rheinberger M, Verweij N, Bouatia-Naji N, Martin NG, Hastie N, Probst-Hensch N, Soranzo N, Devuyst O, Raitakari O, Gottesman O, Franco OH, Polasek O, Gasparini P, Munroe PB, Ridker PM, Mitchell P, Muntner P, Meisinger C, Smit JH, Kovacs P, Wild PS, Froguel P, Rettig R, Mägi R, Biffar R, Schmidt R, Middelberg RPS, Carroll RJ, Penninx BW, Scott RJ, Katz R, Sedaghat S, Wild SH, Kardia SLR, Ulivi S, Hwang SJ, Enroth S, Kloiber S, Trompet S, Stengel B, Hancock SJ, Turner ST, Rosas SE, Stracke S, Harris TB, Zeller T, Zemunik T, Lehtimäki T, Illig T, Aspelund T, Nikopensius T, Esko T, Tanaka T, Gyllensten U, Völker U, Emilsson V, Vitart V, Aalto V, Gudnason V, Chouraki V, Chen WM, Igl W, März W, Koenig W, Lieb W, Loos RJF, Liu Y, Snieder H, Pramstaller PP, Parsa A, O'Connell JR, Susztak K, Hamet P, Tremblay J, de Boer IH, Böger CA, Goessling W, Chasman DI, Köttgen A, Kao WHL, Fox CS. Genetic associations at 53 loci highlight cell types and biological pathways relevant for kidney function. Nat Commun 2016; 7:10023. [PMID: 26831199 PMCID: PMC4735748 DOI: 10.1038/ncomms10023] [Show More Authors] [Citation(s) in RCA: 403] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 10/27/2015] [Indexed: 11/23/2022] Open
Abstract
Reduced glomerular filtration rate defines chronic kidney disease and is associated with cardiovascular and all-cause mortality. We conducted a meta-analysis of genome-wide association studies for estimated glomerular filtration rate (eGFR), combining data across 133,413 individuals with replication in up to 42,166 individuals. We identify 24 new and confirm 29 previously identified loci. Of these 53 loci, 19 associate with eGFR among individuals with diabetes. Using bioinformatics, we show that identified genes at eGFR loci are enriched for expression in kidney tissues and in pathways relevant for kidney development and transmembrane transporter activity, kidney structure, and regulation of glucose metabolism. Chromatin state mapping and DNase I hypersensitivity analyses across adult tissues demonstrate preferential mapping of associated variants to regulatory regions in kidney but not extra-renal tissues. These findings suggest that genetic determinants of eGFR are mediated largely through direct effects within the kidney and highlight important cell types and biological pathways.
Collapse
Affiliation(s)
- Cristian Pattaro
- Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC), affiliated to the University of Lübeck, Via Galvani 31, Bolzano 39100, Italy
| | - Alexander Teumer
- Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt-University Greifswald, Friedrich-Loeffler-Straße 15a, Greifswald 17487, Germany
- Institute for Community Medicine, University of Greifswald, Walther-Rathenau-Strasse 48, Greifswald 17487, Germany
| | - Mathias Gorski
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg 93053, Germany
- Department of Nephrology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg 93053, Germany
| | - Audrey Y. Chu
- Preventive Medicine, Brigham and Women's Hospital, 900 Commonwealth Avenue East, Boston, Massachusetts 02215, USA
| | - Man Li
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, Maryland 21205, USA
| | - Vladan Mijatovic
- Department of Life and Reproduction Sciences, University of Verona, Strada Le Grazie 8, Verona 37134, Italy
| | - Maija Garnaas
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, New Research Building77 Avenue Louis Pasteur, Room 458, Boston, Massachusetts 02115, USA
| | - Adrienne Tin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, Maryland 21205, USA
| | - Rossella Sorice
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso"—CNR, Via P. Castellino 111, Napoli 80131, Italy
| | - Yong Li
- Department of Internal Medicine IV, University Hospital Freiburg, Berliner Allee 29, Freiburg 79110, Germany
| | - Daniel Taliun
- Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC), affiliated to the University of Lübeck, Via Galvani 31, Bolzano 39100, Italy
| | - Matthias Olden
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg 93053, Germany
- Department of Nephrology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg 93053, Germany
| | - Meredith Foster
- Division of Nephrology/Tufts Evidence Practice Center, Tufts University School of Medicine, Tufts Medical Center, Boston, Massachusetts 02111, USA
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, 715 Albany Street, Boston, Massachusetts 02118, USA
| | - Ming-Huei Chen
- Department of Biostatistics, Boston University School of Public Health, 715 Albany Street, Boston, Massachusetts 02118, USA
- Department of Neurology, Boston University School of Medicine, 72 East Concord ST B603, Boston, Massachusetts 02118, USA
| | - Tune H. Pers
- Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 2142, USA
| | - Andrew D. Johnson
- NHLBI's Framingham Heart Study and the Center for Population Studies, 73 Mt Wayte Avenue, Suite 2, Framingham, Massachusetts 01702, USA
| | - Yi-An Ko
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, 405B Clinical Research Building, Philadelphia, Pennsylvania 19104-4539, USA
| | - Christian Fuchsberger
- Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC), affiliated to the University of Lübeck, Via Galvani 31, Bolzano 39100, Italy
| | - Bamidele Tayo
- Department of Public Health Sciences, Loyola Medical Center, 2160 S First Avenue, Maywood, Illinois 60153, USA
| | - Michael Nalls
- Laboratory of Neurogenetics, Building 35—Porter Building, 1A1015, National Institute on Aging/NIH, Bethesda, Maryland 20892, USA
| | - Mary F. Feitosa
- Division of Statistical Genomics, Washington University School of Medicine, 4444 Forest Park Boulevard, Box 8506, St Louis, Missouri 63108, USA
| | - Aaron Isaacs
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Dr Molewaterplein 50, PO Box 2040, Rotterdam 3000 CA, The Netherlands
- Centre for Medical Systems Biology Leiden, Dr Molewaterplein 50, PO Box 2040, Rotterdam 3000 CA, The Netherlands
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus University Medical Center, PO Box 2040, Rotterdam 3000 CA, The Netherlands
| | - Pio d'Adamo
- Institute for Maternal and Child Health—IRCCS "Burlo Garofolo" and University of Trieste, via dell'Istria 65/1, Trieste 34137, Italy
| | - Adebowale Adeyemo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, Building 12A, Room 4047, 12 South Dr, MSC 5635, Bethesda, Maryland 20892-5635, USA
| | - Aida Karina Dieffenbach
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, Heidelberg 69120, Germany
- German Cancer Consortium (DKTK), Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Alan B. Zonderman
- Laboratory of Personality and Cognition, National Institute on Aging, National Institutes of Health, NIH Biomedical Center, 251 Bayview Boulevard, Suite 100, Baltimore, Maryland 21224, USA
| | - Ilja M. Nolte
- Unit of Genetic Epidemiology and Bioinformatics, Department of Epidemiology, University Medical Center Groningen, PO Box 30001, Groningen 9700 RB, The Netherlands
| | - Peter J. van der Most
- Unit of Genetic Epidemiology and Bioinformatics, Department of Epidemiology, University Medical Center Groningen, PO Box 30001, Groningen 9700 RB, The Netherlands
| | - Alan F. Wright
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Alan R. Shuldiner
- Department of Medicine, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, Maryland 21201, USA
- Geriatric Research and Education Clinical Center, Veterans Administration Medical Center, 10 North Greene Street, Baltimore, Maryland 21201, USA
| | - Alanna C. Morrison
- Human Genetics Center, University of Texas Health Science Center at Houston, 1200 Pressler St Suite 453E, Houston, Texas 77030, USA
| | - Albert Hofman
- Department of Epidemiology, Erasmus University Medical Center, PO Box 2040, Rotterdam 3000 CA, The Netherlands
| | - Albert V. Smith
- Icelandic Heart Association, Research Institute, Holtasmari 1, Kopavogur 201, Iceland
- University of Iceland, Sæmundargötu 2, Reykjavik 101, Iceland
| | - Albert W. Dreisbach
- Division of Nephrology, University of Mississippi, 2500 North State Street, Jackson, Mississippi 39216, USA
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts University of Kiel, Schittenhelmstraße 12, Kiel 24105, Germany
| | - Andre G. Uitterlinden
- Department of Internal Medicine, Erasmus University Medical Center, PO Box 1738, Rotterdam 3000 DR, The Netherlands
| | - Andres Metspalu
- Estonian Genome Center of University of Tartu (EGCUT), Riia 23B, Tartu 51010, Estonia
- Institute of Molecular and Cell Biology, University of Tartu and Estonian Biocenter, Riia 23, Tartu 51010, Estonia
| | - Anke Tonjes
- Department of Medicine, University of Leipzig, Liebigstraße 18, Leipzig 04103, Germany
| | - Antonio Lupo
- Division of Nephrology, Department of Medicine, University of Verona, Piazzale Aristide Stefani 1, Verona 37126, Italy
| | - Antonietta Robino
- Institute for Maternal and Child Health—IRCCS "Burlo Garofolo" and University of Trieste, via dell'Istria 65/1, Trieste 34137, Italy
| | - Åsa Johansson
- Uppsala University, Department of Immunology, Genetics and Pathology, Biomedical Center, SciLifeLab, Uppsala University, Uppsala SE- 75108, Sweden
| | - Ayse Demirkan
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Dr Molewaterplein 50, PO Box 2040, Rotterdam 3000 CA, The Netherlands
| | - Barbara Kollerits
- Innsbruck Medical University, Division of Genetic Epidemiology, Schoepfstraße 41, Innsbruck 6020, Austria
| | - Barry I. Freedman
- Internal Medicine Department, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157-1053, USA
| | - Belen Ponte
- Nephrology Division, Department of Specialties of Internal Medicine, Geneva University Hospital, 4 rue Gabrielle-Perret-Gentil, Geneve 1211, Switzerland
| | - Ben A. Oostra
- Department of Clinical Genetics, Erasmus University Medical Center, Dr Molewaterplein 50, PO Box 2040, Rotterdam 3000 CA, The Netherlands
| | - Bernhard Paulweber
- First Department of Internal Medicine, Paracelsus Medical University/Salzburger Landeskliniken, Müllner Hauptstraße 48, Salzburg 5020, Austria
| | - Bernhard K. Krämer
- University Medical Centre Mannheim, 5th Department of Medicine, University of Heidelberg, Theodor Kutzer Ufer 1-3, Mannheim 68167, Germany
| | - Braxton D. Mitchell
- Department of Medicine, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, Maryland 21201, USA
- Geriatric Research and Education Clinical Center, Veterans Administration Medical Center, 10 North Greene Street, Baltimore, Maryland 21201, USA
| | - Brendan M. Buckley
- Department of Pharmacology and Therapeutics, University College Cork, Clinical Investigations Building, Western Rd, Cork, Ireland
| | - Carmen A. Peralta
- Division of Nephrology, University of California, San Francisco Medical School and San Francisco VA Medical Center, 4150 Clement Street, San Francisco, California 94121, USA
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Catherine Helmer
- INSERM, ISPED, Centre INSERM U897—Epidemiologie-Biostatistique, Bordeaux F-33000, France
- Université Bordeaux, ISPED, Centre INSERM U897-Epidemiologie-Biostatistique, Bordeaux F-33000, France
| | - Charles N. Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, Building 12A, Room 4047, 12 South Dr, MSC 5635, Bethesda, Maryland 20892-5635, USA
| | - Christian M. Shaffer
- Vanderbilt University School of Medicine, 2215-B Garland Avenue 1224—MRB4 (Light Hall)Nashville, Tennessee 37232, USA
| | - Christian Müller
- University Heart Center Hamburg, Clinic for general and interventional cardiology, Martinistraße 52, Hamburg 20246, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, Martinistraße 52, Hamburg 20246, Germany
| | - Cinzia Sala
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Via Olgettina 58, Milano 20132, Italy
| | - Cornelia M. van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Dr Molewaterplein 50, PO Box 2040, Rotterdam 3000 CA, The Netherlands
| | - Aude Saint-Pierre
- Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC), affiliated to the University of Lübeck, Via Galvani 31, Bolzano 39100, Italy
- INSERM U1078, Etablissement Français du Sang, 46 rue Félix Le Dantec, CS 51819, Brest Cedex 2 29218, France
| | - Daniel Ackermann
- Nephrology Division, Department of Specialties of Internal Medicine, Geneva University Hospital, 4 rue Gabrielle-Perret-Gentil, Geneve 1211, Switzerland
| | - Daniel Shriner
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, Building 12A, Room 4047, 12 South Dr, MSC 5635, Bethesda, Maryland 20892-5635, USA
| | - Daniela Ruggiero
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso"—CNR, Via P. Castellino 111, Napoli 80131, Italy
| | - Daniela Toniolo
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Via Olgettina 58, Milano 20132, Italy
- Institute of Molecular Genetics-CNR, Via Abbiategrasso 207, Pavia 27100, Italy
| | - Yingchang Lu
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Daniele Cusi
- Department of Health Sciences, University of Milano, Via Antonio di Rudinì 8, Milano 20142, Italy
| | - Darina Czamara
- Max Planck Institute of Psychiatry, Kraepelinstraße 2–10, Munich 80804, Germany
| | - David Ellinghaus
- Institute of Clinical Molecular Biology, Christian-Albrechts University of Kiel, Schittenhelmstraße 12, Kiel 24105, Germany
| | - David S. Siscovick
- Cardiovascular Health Research Unit, Departments of Epidemiology and Medicine, University of Washington, 1730 Minor Ave, Suite 1360, Seattle, Washington 98101, USA
| | - Douglas Ruderfer
- Division of Psychiatric Genomics, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstraße 1, 85764 Neuherberg, Germany
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstraße 1, 85764 Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), Ingolstaedter Landstraße 1, Neuherberg 85764, Germany
| | - Elena Rochtchina
- Westmead Millennium Institute, Centre for Vision Research, University of Sydney, C24 Westmead Hospital, New South Wales 2145, Australia
| | - Elizabeth J. Atkinson
- Division of Biomedical Statistics and Informatics, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, USA
| | - Elizabeth G. Holliday
- Centre for Clinical Epidemiology and Biostatistics, School of Medicine and Public Health, University of Newcastle, HMRI Building1, Kookaburra Circuit, New Lambton New South Wales 2305, Australia
- Clinical Research Design, Information Technology and Statistical Support, Hunter Medical Research Institute, Newcastle, 1 Kookaburra Circuit, New Lambton Heights, New South Wales 2305, Australia
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Science Center at Houston, 1200 Pressler St Suite 453E, Houston, Texas 77030, USA
| | - Erika Salvi
- Department of Health Sciences, University of Milano, Via Antonio di Rudinì 8, Milano 20142, Italy
| | - Erwin P. Bottinger
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Federico Murgia
- Institute of Population Genetics—CNR, Traversa La Crucca 3, 07040 Reg. Baldinca, Li Punti, Sassari, Italy
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus University Medical Center, PO Box 1738, Rotterdam 3000 DR, The Netherlands
| | - Florian Ernst
- Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt-University Greifswald, Friedrich-Loeffler-Straße 15a, Greifswald 17487, Germany
| | - Florian Kronenberg
- Innsbruck Medical University, Division of Genetic Epidemiology, Schoepfstraße 41, Innsbruck 6020, Austria
| | - Frank B. Hu
- Department of Nutrition, Harvard School of Public Health, 665 Huntington Avenue, Building 2, Boston, Massachusetts 02115, USA
| | - Gerjan J. Navis
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen 9713 GZ, The Netherlands
| | - Gary C. Curhan
- Brigham and Women's Hospital and Channing Laboratory, Harvard Medical School, 181 Longwood Avenue, Boston, Massachusetts 02115, USA
| | - George B. Ehret
- Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 4, Geneva 1205, Switzerland
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt-University Greifswald, Friedrich-Loeffler-Straße 15a, Greifswald 17487, Germany
| | - Stefan Coassin
- Innsbruck Medical University, Division of Genetic Epidemiology, Schoepfstraße 41, Innsbruck 6020, Austria
| | - Gian-Andri Thun
- Swiss Tropical and Public Health Institute, PO Box 4002, Basel, Switzerland
- University of Basel, Petersplatz 1, Basel 4003, Switzerland
| | - Giorgio Pistis
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Via Olgettina 58, Milano 20132, Italy
| | - Giovanni Gambaro
- Division of Nephrology, Department of Internal Medicine and Medical Specialties, Columbus-Gemelli University Hospital, Catholic University, Via Moscati 31, Rome 00168, Italy
| | - Giovanni Malerba
- Department of Life and Reproduction Sciences, University of Verona, Strada Le Grazie 8, Verona 37134, Italy
| | - Grant W. Montgomery
- Genetic Epidemiology, Queensland Institute of Medical Research, QIMR, PO Royal Brisbane Hospital, Queensland 4029, Australia
| | - Gudny Eiriksdottir
- Icelandic Heart Association, Research Institute, Holtasmari 1, Kopavogur 201, Iceland
| | - Gunnar Jacobs
- Institute of Epidemiology and Biobank popgen, Christian-Albrechts University, Niemannsweg 11, Kiel 24105, Germany
| | - Guo Li
- Cardiovascular Health Research Unit, Departments of Epidemiology and Medicine, University of Washington, 1730 Minor Ave, Suite 1360, Seattle, Washington 98101, USA
| | - H-Erich Wichmann
- Institute of Epidemiology I, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, Neuherberg 85764, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-Universität, Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany
- Klinikum Grosshadern, Ingolstädter Landstraße 1, Neuherberg 85764, Germany
| | - Harry Campbell
- Centre for Population Health Sciences, University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG Scotland, UK
| | - Helena Schmidt
- Austrian Stroke Prevention Study, Institute of Molecular Biology and Biochemistry, Department of Neurology, Medical University Graz, Harrachgasse 21, Graz 8010, Austria
| | - Henri Wallaschofski
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, Greifswald 17475, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Greifswald, Ferdinand-Sauerbruch-Straße, Greifswald 17475, Germany
| | - Henry Völzke
- Institute for Community Medicine, University of Greifswald, Walther-Rathenau-Strasse 48, Greifswald 17487, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Greifswald, Ferdinand-Sauerbruch-Straße, Greifswald 17475, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, Heidelberg 69120, Germany
- German Cancer Consortium (DKTK), Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Heyo K. Kroemer
- Institute of Pharmacology, University of Greifswald, Friedrich-Loeffler-Straße 23d, Greifswald 17487, Germany
| | - Holly Kramer
- Department of Public Health Sciences, Loyola Medical Center, 2160 S First Avenue, Maywood, Illinois 60153, USA
| | - Honghuang Lin
- Boston University School of Medicine, 72 East Concord Street, B-616, Boston, Massachusetts 02118, USA
| | - I. Mateo Leach
- Department of Cardiology, University Medical Center Groningen, University of Groningen, PO Box 30.001, Groningen 9700 RB, The Netherlands
| | - Ian Ford
- Robertson Centre for Biostatistics, University of Glasgow, R1122B Level 11, Robertson Centre, Boyd Orr Building, Glasgow G12 8QQ, UK
| | - Idris Guessous
- Division of Primary Care Medicine, Department of Community Medicine, Primary Care and Emergency Medicine, Geneva University Hospitals, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland
- Community Prevention Unit, University Institute of Social and Preventive Medicine, Lausanne University Hospital, Route de la Corniche 10, Lausanne 1010, Switzerland
- Department of Epidemiology, Rollins School of Public Health, Emory University, 1518 Clifton Road, NE, Atlanta, Georgia 30322, USA
| | - Igor Rudan
- Centre for Population Health Sciences, University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG Scotland, UK
| | - Inga Prokopenko
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, London W12 0NN, UK
| | - Ingrid Borecki
- Division of Statistical Genomics, Washington University School of Medicine, 4444 Forest Park Boulevard, Box 8506, St Louis, Missouri 63108, USA
| | - Iris M. Heid
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg 93053, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstraße 1, 85764 Neuherberg, Germany
| | - Ivana Kolcic
- Croatian Centre for Global Health, University of Split Medical School, Šoltanska 2, Split 21000, Croatia
| | - Ivana Persico
- Institute of Population Genetics—CNR, Traversa La Crucca 3, 07040 Reg. Baldinca, Li Punti, Sassari, Italy
| | - J. Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, PO Box 9600, Leiden 2300 RC, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands (ICIN), Moreelsepark 1, Utrecht 3511 EP, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
- Durrer Center for Cardiogenetic Research, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
| | - James F. Wilson
- Centre for Population Health Sciences, University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG Scotland, UK
| | - Janine F. Felix
- Department of Epidemiology, Erasmus University Medical Center, PO Box 2040, Rotterdam 3000 CA, The Netherlands
| | - Jasmin Divers
- Division of Public Health Sciences, Department of Biostatistical Sciences, Wake Forest University Health Sciences, 2326 Medical Center Boulevard, Winston-Salem, North Carolina 27157-1063, USA
| | - Jean-Charles Lambert
- INSERM U744, Institut Pasteur de Lille, 1 rue du Pr, Calmette, Lille Cédex 59019, France
| | - Jeanette M. Stafford
- Division of Public Health Sciences, Department of Biostatistical Sciences, Wake Forest University Health Sciences, 2326 Medical Center Boulevard, Winston-Salem, North Carolina 27157-1063, USA
| | - Jean-Michel Gaspoz
- Division of Primary Care Medicine, Department of Community Medicine, Primary Care and Emergency Medicine, Geneva University Hospitals, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland
| | - Jennifer A. Smith
- Department of Epidemiology, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, Michigan 48109-2029, USA
| | - Jessica D. Faul
- Survey Research Center, Institute for Social Research, University of Michigan, 426 Thompson Street, #3456, Ann Arbor, Michigan 48104, USA
| | - Jie Jin Wang
- Centre for Vision Research, Westmead Millennium Institute, University of Sydney, C24 Westmead Hospital, New South Wales 2145, Australia
| | - Jingzhong Ding
- Wake Forest School of Medicine, Department of Internal Medicine/Geriatrics, Medical center Boulevard, Winston-Salem, North Carolina 27157, USA
| | - Joel N. Hirschhorn
- Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 2142, USA
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, NRB 0330, Boston, Massachusetts 02115, USA
| | - John Attia
- Division of Biomedical Statistics and Informatics, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, USA
- Centre for Clinical Epidemiology and Biostatistics, School of Medicine and Public Health, University of Newcastle, HMRI Building1, Kookaburra Circuit, New Lambton New South Wales 2305, Australia
| | - John B. Whitfield
- Genetic Epidemiology, Queensland Institute of Medical Research, QIMR, PO Royal Brisbane Hospital, Queensland 4029, Australia
| | - John Chalmers
- University of Sydney, The George Institute for Global Health, Level 10, King George V Building, 83-117 Missenden Road, Camperdown, New South Wales 2050, Australia
| | - Jorma Viikari
- Department of Medicine, University of Turku, Turku University Hospital, PO Box 52, Turku 20521, Finland
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, Maryland 21205, USA
- Welch Center for Prevention, Epidemiology and Clinical Research, 2024 East Monument St, Suite 2-600, Baltimore, Maryland 21287, USA
| | - Joshua C. Denny
- Vanderbilt University School of Medicine, 448 Eskind Biomedical Library, 2209 Garland Avenue, Nashville, Tennessee 37212, USA
| | - Juha Karjalainen
- Department of Genetics, University of Groningen, University Medical Centre Groningen, PO Box 72, Groningen 9700 AB, The Netherlands
| | - Jyotika K. Fernandes
- Division of Endocrinology, Medical University of South Carolina, 171 Ashley Avenue, Charleston, South Carolina 29425, USA
| | - Karlhans Endlich
- Institute of Anatomy and Cell Biology, University of Greifswald, Friedrich-Loeffler-Straße 23c, Greifswald 17487, Germany
| | - Katja Butterbach
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Keith L. Keene
- Center for Health Disparities, Department of Biology, East Carolina University, 1001 East 10th Street, N209 Howell Science Complex Mailstop 551, Greenville, North Carolina 27858, USA
| | - Kurt Lohman
- Internal Medicine Department, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157-1053, USA
| | - Laura Portas
- Institute of Population Genetics—CNR, Traversa La Crucca 3, 07040 Reg. Baldinca, Li Punti, Sassari, Italy
| | - Lenore J. Launer
- Intramural Research Program, Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, Gateway Building, 3C309, 7201 Winsconsin Avenue, Bethesda, Maryland 20892-9205, USA
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere, School of Medicine, Tampere 33520, Finland
| | - Loic Yengo
- CNRS UMR 8199, 1 Rue du Professeur Calmette, Lille 59000, France
- Lille Pasteur Institute, 1 Rue du Professeur Calmette, Lille 59000, France
- Lille II University, 42 Rue paul Duez, Lille 59000, France
| | - Lude Franke
- Department of Genetics, University of Groningen, University Medical Centre Groningen, PO Box 72, Groningen 9700 AB, The Netherlands
| | - Luigi Ferrucci
- Clinical Research Branch, National Institute on Aging, 251 Bayview Blvd, Baltimore, Maryland 21250, USA
| | - Lynda M. Rose
- Preventive Medicine, Brigham and Women's Hospital, 900 Commonwealth Avenue East, Boston, Massachusetts 02215, USA
| | - Lyudmyla Kedenko
- First Department of Internal Medicine, Paracelsus Medical University/Salzburger Landeskliniken, Müllner Hauptstraße 48, Salzburg 5020, Austria
| | - Madhumathi Rao
- Division of Nephrology/Tufts Evidence Practice Center, Tufts University School of Medicine, Tufts Medical Center, Boston, Massachusetts 02111, USA
| | - Maksim Struchalin
- Department of Epidemiology and Biostatistics, Erasmus University Medical Center, Dr Molewaterplein, Rotterdam 50-603015 GE, The Netherlands
- Department of Forensic Molecular Biology, Erasmus University Medical Center, Dr Molewaterplein, Rotterdam 50-603015 GE, The Netherlands
| | - Marcus E. Kleber
- Medical Clinic V, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, Mannheim 68167, Germany
| | - Margherita Cavalieri
- Austrian Stroke Prevention Study, Department of Neurology, Division of Special Neurology, Medical University Graz, Auenbruggerplatz 22, Graz 8036, Austria
| | - Margot Haun
- Innsbruck Medical University, Division of Genetic Epidemiology, Schoepfstraße 41, Innsbruck 6020, Austria
| | - Marilyn C. Cornelis
- Department of Nutrition, Harvard School of Public Health, 665 Huntington Avenue, Building 2, Boston, Massachusetts 02115, USA
| | - Marina Ciullo
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso"—CNR, Via P. Castellino 111, Napoli 80131, Italy
| | - Mario Pirastu
- Institute of Population Genetics—CNR, Traversa La Crucca 3, 07040 Reg. Baldinca, Li Punti, Sassari, Italy
| | - Mariza de Andrade
- Division of Biomedical Statistics and Informatics, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, USA
| | - Mark A. McEvoy
- Centre for Clinical Epidemiology and Biostatistics, University of Newcastle, Hunter Medical Research Institute, John Hunter Hospital, Locked Bag 1, HRMC, New South Wales 2310, Australia
| | - Mark Woodward
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, Maryland 21205, USA
- University of Sydney, The George Institute for Global Health, Level 10, King George V Building, 83-117 Missenden Road, Camperdown, New South Wales 2050, Australia
- Department of Medicine, University of Turku, Turku University Hospital, PO Box 52, Turku 20521, Finland
- The George Institute for Global Health, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7LF, UK
| | - Martin Adam
- Swiss Tropical and Public Health Institute, PO Box 4002, Basel, Switzerland
- University of Basel, Petersplatz 1, Basel 4003, Switzerland
| | - Massimiliano Cocca
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Via Olgettina 58, Milano 20132, Italy
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, Greifswald 17475, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Greifswald, Ferdinand-Sauerbruch-Straße, Greifswald 17475, Germany
| | - Medea Imboden
- Swiss Tropical and Public Health Institute, PO Box 4002, Basel, Switzerland
- University of Basel, Petersplatz 1, Basel 4003, Switzerland
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstraße 1, 85764 Neuherberg, Germany
| | - Menno Pruijm
- Service of Nephrology, Lausanne University Hospital, Rue du Bugnon 17, Lausanne 1005, Switzerland
| | - Marie Metzger
- Inserm UMRS 1018, CESP Team 10, Université Paris-Sud, 16 avenue Paul Vaillant Couturier, Villejuif 94807, France
| | - Michael Stumvoll
- Department of Medicine, University of Leipzig, Liebigstraße 18, Leipzig 04103, Germany
| | - Michele K. Evans
- Health Disparities Research Section, Clinical Research Branch, National Institute on Aging, National Institutes of Health, NIH Biomedical Center, 251 Bayview Boulevard, Suite 100, Baltimore, Maryland 21224, USA
| | - Michele M. Sale
- Center for Public Health Genomics, Department of Medicine (Cardiovascular Medicine), University of Virginia, PO Box 800717, Charlottesville, Virginia 22908, USA
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, University of Tampere, School of Medicine, Tampere 33521, Finland
| | - Mladen Boban
- Croatian Centre for Global Health, University of Split Medical School, Šoltanska 2, Split 21000, Croatia
| | - Murielle Bochud
- University Institute of Social and Preventive Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Route de la Corniche 2, Epalinges CH-1066, Switzerland
| | - Myriam Rheinberger
- Department of Nephrology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg 93053, Germany
| | - Niek Verweij
- Department of Cardiology, University Medical Center Groningen, University of Groningen, PO Box 30.001, Groningen 9700 RB, The Netherlands
| | - Nabila Bouatia-Naji
- INSERM UMR970, Paris Cardiovascular Research Center (PARCC), 56 rue Leblanc, Paris F-75015, France
- Paris Descartes University, Faculty of medicine, Paris Cité Sorbonne, 12 Rue de l'école de Médecine, Paris F-75006, France
| | - Nicholas G. Martin
- Genetic Epidemiology, Queensland Institute of Medical Research, QIMR, PO Royal Brisbane Hospital, Queensland 4029, Australia
- Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, Massachusetts 02115, USA
| | - Nick Hastie
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Nicole Probst-Hensch
- Swiss Tropical and Public Health Institute, PO Box 4002, Basel, Switzerland
- University of Basel, Petersplatz 1, Basel 4003, Switzerland
| | | | - Olivier Devuyst
- University of Zurich, Institute of Physiology, Mechanisms of Inherited Kidney Disorders Group, Winterthurerstrasse 190, Zürich 8057, Switzerland
| | - Olli Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku University Hospital, Department of Clinical Physiology, PO Box 52, Turku 20521, Finland
| | - Omri Gottesman
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Oscar H. Franco
- Department of Epidemiology, Erasmus University Medical Center, PO Box 2040, Rotterdam 3000 CA, The Netherlands
| | - Ozren Polasek
- Croatian Centre for Global Health, University of Split Medical School, Šoltanska 2, Split 21000, Croatia
| | - Paolo Gasparini
- Institute for Maternal and Child Health—IRCCS "Burlo Garofolo" and University of Trieste, via dell'Istria 65/1, Trieste 34137, Italy
| | - Patricia B. Munroe
- Department Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
- NIHR Barts Cardiovascular Biomedical Research Unit, Queen Mary University of London, London EC1M 6BQ, UK
| | - Paul M. Ridker
- Harvard Medical School, 900 Commonwealth Avenue East, Boston, Massachusetts 02115, USA
| | - Paul Mitchell
- Centre for Vision Research, Westmead Millennium Institute, University of Sydney, C24 Westmead Hospital, New South Wales 2145, Australia
| | - Paul Muntner
- University of Alabama at Birmingham, Department of Medicine, 1530 3rd Avenue, South Birmingham, Alabama 35294-0022, USA
- University of Alabama at Birmingham, Department of Epidemiology, 1530 3rd Avenue, South Birmingham, Alabama 35294-0022, USA
| | - Christa Meisinger
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Johannes H. Smit
- Department of Psychiatry and EMGO+ Institute, VU University Medical Center, A.J. Ernststraat 1187, Amsterdam 1081 HL, The Netherlands
| | - Peter Kovacs
- IFB AdiposityDiseases, University of Leipzig, Liebigstraße 21, Leipzig 04103, Germany
| | - Philipp S. Wild
- Medical University Center Mainz, Langenbeckstraße 1, Mainz 55131, Germany
| | - Philippe Froguel
- CNRS UMR 8199, 1 Rue du Professeur Calmette, Lille 59000, France
- Lille Pasteur Institute, 1 Rue du Professeur Calmette, Lille 59000, France
- Lille II University, 42 Rue paul Duez, Lille 59000, France
| | - Rainer Rettig
- Institute of Physiology, University of Greifswald, Greifswald 17487, Germany
| | - Reedik Mägi
- Estonian Genome Center of University of Tartu (EGCUT), Riia 23B, Tartu 51010, Estonia
| | - Reiner Biffar
- Clinic for Prosthodontic Dentistry, Gerostomatology and Material Science, University of Greifswald, Rotgerberstraße 8, Greifswald 17475, Germany
| | - Reinhold Schmidt
- Austrian Stroke Prevention Study, Department of Neurology, Division of Special Neurology, Medical University Graz, Auenbruggerplatz 22, Graz 8036, Austria
| | - Rita P. S. Middelberg
- Genetic Epidemiology, Queensland Institute of Medical Research, QIMR, PO Royal Brisbane Hospital, Queensland 4029, Australia
| | - Robert J. Carroll
- Vanderbilt University School of Medicine, 448 Eskind Biomedical Library, 2209 Garland Avenue, Nashville, Tennessee 37212, USA
| | - Brenda W. Penninx
- Department of Psychiatry and EMGO+ Institute, VU University Medical Center, A.J. Ernststraat 1187, Amsterdam 1081 HL, The Netherlands
| | - Rodney J. Scott
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Hunter Medical Research Institute, John Hunter Hospital, Locked Bag 1, HRMC, New South Wales 2310, Australia
| | - Ronit Katz
- Kidney Research Institute, University of Washington, Box 359606, 325 9th Avenue, Seattle, Washington 98104, USA
| | - Sanaz Sedaghat
- Department of Epidemiology, Erasmus University Medical Center, PO Box 2040, Rotterdam 3000 CA, The Netherlands
| | - Sarah H. Wild
- Centre for Population Health Sciences, University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG Scotland, UK
| | - Sharon L. R. Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, Michigan 48109-2029, USA
| | - Sheila Ulivi
- Institute for Maternal and Child Health—IRCCS "Burlo Garofolo", Via dell'Istria 65, Trieste 34137, Italy
| | - Shih-Jen Hwang
- NHLBI's Framingham Heart Study and the Center for Population Studies, 73 Mt Wayte Avenue, Suite 2, Framingham, Massachusetts 01702, USA
| | - Stefan Enroth
- Uppsala University, Department of Immunology, Genetics and Pathology, Biomedical Center, SciLifeLab, Uppsala University, Uppsala SE- 75108, Sweden
| | - Stefan Kloiber
- Max Planck Institute of Psychiatry, Kraepelinstraße 2–10, Munich 80804, Germany
| | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center, PO Box 9600, Leiden 2300 RC, The Netherlands
| | - Benedicte Stengel
- Inserm UMRS 1018, CESP Team 10, Université Paris-Sud, 16 avenue Paul Vaillant Couturier, Villejuif 94807, France
| | - Stephen J. Hancock
- Centre for Clinical Epidemiology and Biostatistics, School of Medicine and Public Health, University of Newcastle, HMRI Building1, Kookaburra Circuit, New Lambton New South Wales 2305, Australia
- Clinical Research Design, Information Technology and Statistical Support, Hunter Medical Research Institute, Newcastle, 1 Kookaburra Circuit, New Lambton Heights, New South Wales 2305, Australia
| | - Stephen T. Turner
- Department of Internal Medicine, Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, USA
| | - Sylvia E. Rosas
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, 405B Clinical Research Building, Philadelphia, Pennsylvania 19104-4539, USA
| | - Sylvia Stracke
- INSERM U744, Institut Pasteur de Lille, 1 rue du Pr, Calmette, Lille Cédex 59019, France
- Clinic for Internal Medicine A, University of Greifswald, Friedrich-Loeffler-Straße 23a, Greifswald 17475, Germany
| | - Tamara B. Harris
- Intramural Research Program, Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, Gateway Building, 3C309, 7201 Winsconsin Avenue, Bethesda, Maryland 20892-9205, USA
| | - Tanja Zeller
- University Heart Center Hamburg, Clinic for general and interventional cardiology, Martinistraße 52, Hamburg 20246, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, Martinistraße 52, Hamburg 20246, Germany
| | - Tatijana Zemunik
- Croatian Centre for Global Health, University of Split Medical School, Šoltanska 2, Split 21000, Croatia
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere, School of Medicine, Tampere 33520, Finland
| | - Thomas Illig
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Thor Aspelund
- Icelandic Heart Association, Research Institute, Holtasmari 1, Kopavogur 201, Iceland
- University of Iceland, Sæmundargötu 2, Reykjavik 101, Iceland
| | - Tiit Nikopensius
- Estonian Genome Center of University of Tartu (EGCUT), Riia 23B, Tartu 51010, Estonia
- Institute of Molecular and Cell Biology, University of Tartu and Estonian Biocenter, Riia 23, Tartu 51010, Estonia
| | - Tonu Esko
- Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Estonian Genome Center of University of Tartu (EGCUT), Riia 23B, Tartu 51010, Estonia
- Institute of Molecular and Cell Biology, University of Tartu and Estonian Biocenter, Riia 23, Tartu 51010, Estonia
| | - Toshiko Tanaka
- Clinical Research Branch, National Institute on Aging, 251 Bayview Blvd, Baltimore, Maryland 21250, USA
| | - Ulf Gyllensten
- Uppsala University, Department of Immunology, Genetics and Pathology, Biomedical Center, SciLifeLab, Uppsala University, Uppsala SE- 75108, Sweden
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt-University Greifswald, Friedrich-Loeffler-Straße 15a, Greifswald 17487, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Greifswald, Ferdinand-Sauerbruch-Straße, Greifswald 17475, Germany
| | - Valur Emilsson
- Icelandic Heart Association, Research Institute, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Pharmaceutical Sciences, University of Iceland, Sæmundargata 2, Reykjavik 101, Iceland
| | - Veronique Vitart
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Ville Aalto
- Department of Clinical Physiology, Turku University Hospital, Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, PO Box 52, Turku 20521, Finland
| | - Vilmundur Gudnason
- Icelandic Heart Association, Research Institute, Holtasmari 1, Kopavogur 201, Iceland
- University of Iceland, Sæmundargötu 2, Reykjavik 101, Iceland
| | - Vincent Chouraki
- INSERM U744, Institut Pasteur de Lille, 1 rue du Pr, Calmette, Lille Cédex 59019, France
| | - Wei-Min Chen
- Center for Public Health Genomics, Department of Medicine (Cardiovascular Medicine), University of Virginia, PO Box 800717, Charlottesville, Virginia 22908, USA
| | - Wilmar Igl
- Uppsala University, Department of Immunology, Genetics and Pathology, Biomedical Center, SciLifeLab, Uppsala University, Uppsala SE- 75108, Sweden
| | - Winfried März
- Synlab Academy, Synlab Services GmbH, Oberer Eselsberg 45, Ulm 89081, Germany
| | - Wolfgang Koenig
- Department of Internal Medicine II—Cardiology, University of Ulm Medical Centre, Albert-Einstein-Allee 23, Ulm 89081, Germany
| | - Wolfgang Lieb
- Institute of Epidemiology and Biobank popgen, Christian-Albrechts University, Niemannsweg 11, Kiel 24105, Germany
| | - Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- The Mindich Child Health and Development Institute, Ichan School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Yongmei Liu
- Internal Medicine Department, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157-1053, USA
| | - Harold Snieder
- Unit of Genetic Epidemiology and Bioinformatics, Department of Epidemiology, University Medical Center Groningen, PO Box 30001, Groningen 9700 RB, The Netherlands
| | - Peter P. Pramstaller
- Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC), affiliated to the University of Lübeck, Via Galvani 31, Bolzano 39100, Italy
- Department of Neurology, General Central Hospital, Via Lorenz Bohler 5, Bolzano 39100, Italy
- Department of Neurology, University of Lübeck, Ratzeburger Allee 160, Lübeck 23538, Germany
| | - Afshin Parsa
- University of Maryland Medical School, Division of Nephrology, 685 W. Baltimore Street, MSTF 314, Baltimore, Maryland 21201, USA
| | - Jeffrey R. O'Connell
- Department of Medicine, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, Maryland 21201, USA
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, 405B Clinical Research Building, Philadelphia, Pennsylvania 19104-4539, USA
| | - Pavel Hamet
- CRCHUM, University of Montreal, CHUM Research Center, Technopôle Angus, 900 Saint-Denis, Montreal, Québec, Canada H2X 0A9
| | - Johanne Tremblay
- CRCHUM, University of Montreal, CHUM Research Center, Technopôle Angus, 900 Saint-Denis, Montreal, Québec, Canada H2X 0A9
| | - Ian H. de Boer
- Kidney Research Institute, University of Washington, Box 359606, 325 9th Avenue, Seattle, Washington 98104, USA
| | - Carsten A. Böger
- Department of Nephrology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg 93053, Germany
| | - Wolfram Goessling
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, New Research Building77 Avenue Louis Pasteur, Room 458, Boston, Massachusetts 02115, USA
| | - Daniel I. Chasman
- Preventive Medicine, Brigham and Women's Hospital, 900 Commonwealth Avenue East, Boston, Massachusetts 02215, USA
- Harvard Medical School, 900 Commonwealth Avenue East, Boston, Massachusetts 02115, USA
| | - Anna Köttgen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, Maryland 21205, USA
- Department of Internal Medicine IV, University Hospital Freiburg, Berliner Allee 29, Freiburg 79110, Germany
| | - W. H. Linda Kao
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, Maryland 21205, USA
- Welch Center for Prevention, Epidemiology and Clinical Research, 2024 East Monument St, Suite 2-600, Baltimore, Maryland 21287, USA
| | - Caroline S. Fox
- NHLBI's Framingham Heart Study and the Center for Population Studies, 73 Mt Wayte Avenue, Suite 2, Framingham, Massachusetts 01702, USA
- Division of Endocrinology, Brigham and Women s Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, Massachusetts 02115, USA
| |
Collapse
|
108
|
Troyanov S, Delmas-Frenette C, Bollée G, Youhanna S, Bruat V, Awadalla P, Devuyst O, Madore F. Clinical, Genetic, and Urinary Factors Associated with Uromodulin Excretion. Clin J Am Soc Nephrol 2015; 11:62-9. [PMID: 26683887 DOI: 10.2215/cjn.04770415] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 09/24/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND OBJECTIVES The urinary excretion of uromodulin is influenced by common variants in the UMOD gene, and it may be related to NaCl retention and hypertension. Levels of uromodulin are also dependent of the renal function, but other determinants remain unknown. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS We tested associations between the urinary excretion of uromodulin; medical history and medication; serum and urinary levels of electrolytes, glucose, and uric acid; and the genotype at the UMOD/Protein Disulfide Isomerase-Like, Testis Expressed locus (rs4293393 and rs12446492); 943 participants from the CARTaGENE Cohort, a random sample from the Canadian population of 20,004 individuals, were analyzed. Participants with available genotyping were obtained from a substudy addressing associations between common variants and cardiovascular disease in paired participants with high and low Framingham risk scores and vascular rigidity indexes. RESULTS The population studied was 54±9 years old, with 51% women and eGFR of 9±14 ml/min per 1.73 m(2). Uromodulin excretion was 25 (11-42) mg/g creatinine. Using linear regression, it was independently higher among patients with higher eGFR, the TT genotype of rs4293393, and the TT genotype of rs12446492. The fractional excretions of urate and sodium showed a strong positive correlation with uromodulin, likely linked to the extracellular volume status. The presence of glycosuria and the use of uricosuric drugs, which both increased the fraction excretion of urate, were independently associated with a lower uromodulin excretion, suggesting novel interactions between uric acid and uromodulin excretion. CONCLUSIONS In this large cohort, the excretion of uromodulin correlates with clinical, genetic, and urinary factors. The strongest associations were between uric acid, sodium, and uromodulin excretions and are likely linked to the extracellular volume status.
Collapse
Affiliation(s)
- Stéphan Troyanov
- Nephrology Division, Sacré-Coeur Hospital, Montreal, Quebec, Canada;
| | | | - Guillaume Bollée
- Nephrology Division, Hospital Center for the University of Montreal (CHUM), Montreal, Quebec, Canada
| | - Sonia Youhanna
- Institute of Physiology, Zurich Center for Integrative Human Physiology, Zurich, Switzerland
| | - Vanessa Bruat
- Medical and Population Genomics Laboratory, Research Center, Ste-Justine Hospital, Montreal, Quebec, Canada; and Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Philip Awadalla
- Medical and Population Genomics Laboratory, Research Center, Ste-Justine Hospital, Montreal, Quebec, Canada; and Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Olivier Devuyst
- Institute of Physiology, Zurich Center for Integrative Human Physiology, Zurich, Switzerland;
| | - François Madore
- Nephrology Division, Sacré-Coeur Hospital, Montreal, Quebec, Canada
| |
Collapse
|
109
|
|
110
|
Kato N, Loh M, Takeuchi F, Verweij N, Wang X, Zhang W, Kelly TN, Saleheen D, Lehne B, Leach IM, Drong AW, Abbott J, Wahl S, Tan ST, Scott WR, Campanella G, Chadeau-Hyam M, Afzal U, Ahluwalia TS, Bonder MJ, Chen P, Dehghan A, Edwards TL, Esko T, Go MJ, Harris SE, Hartiala J, Kasela S, Kasturiratne A, Khor CC, Kleber ME, Li H, Yu Mok Z, Nakatochi M, Sapari NS, Saxena R, Stewart AFR, Stolk L, Tabara Y, Teh AL, Wu Y, Wu JY, Zhang Y, Aits I, Da Silva Couto Alves A, Das S, Dorajoo R, Hopewell JC, Kim YK, Koivula RW, Luan J, Lyytikäinen LP, Nguyen QN, Pereira MA, Postmus I, Raitakari OT, Bryan MS, Scott RA, Sorice R, Tragante V, Traglia M, White J, Yamamoto K, Zhang Y, Adair LS, Ahmed A, Akiyama K, Asif R, Aung T, Barroso I, Bjonnes A, Braun TR, Cai H, Chang LC, Chen CH, Cheng CY, Chong YS, Collins R, Courtney R, Davies G, Delgado G, Do LD, Doevendans PA, Gansevoort RT, Gao YT, Grammer TB, Grarup N, Grewal J, Gu D, Wander GS, Hartikainen AL, Hazen SL, He J, Heng CK, Hixson JE, Hofman A, Hsu C, Huang W, Husemoen LLN, Hwang JY, et alKato N, Loh M, Takeuchi F, Verweij N, Wang X, Zhang W, Kelly TN, Saleheen D, Lehne B, Leach IM, Drong AW, Abbott J, Wahl S, Tan ST, Scott WR, Campanella G, Chadeau-Hyam M, Afzal U, Ahluwalia TS, Bonder MJ, Chen P, Dehghan A, Edwards TL, Esko T, Go MJ, Harris SE, Hartiala J, Kasela S, Kasturiratne A, Khor CC, Kleber ME, Li H, Yu Mok Z, Nakatochi M, Sapari NS, Saxena R, Stewart AFR, Stolk L, Tabara Y, Teh AL, Wu Y, Wu JY, Zhang Y, Aits I, Da Silva Couto Alves A, Das S, Dorajoo R, Hopewell JC, Kim YK, Koivula RW, Luan J, Lyytikäinen LP, Nguyen QN, Pereira MA, Postmus I, Raitakari OT, Bryan MS, Scott RA, Sorice R, Tragante V, Traglia M, White J, Yamamoto K, Zhang Y, Adair LS, Ahmed A, Akiyama K, Asif R, Aung T, Barroso I, Bjonnes A, Braun TR, Cai H, Chang LC, Chen CH, Cheng CY, Chong YS, Collins R, Courtney R, Davies G, Delgado G, Do LD, Doevendans PA, Gansevoort RT, Gao YT, Grammer TB, Grarup N, Grewal J, Gu D, Wander GS, Hartikainen AL, Hazen SL, He J, Heng CK, Hixson JE, Hofman A, Hsu C, Huang W, Husemoen LLN, Hwang JY, Ichihara S, Igase M, Isono M, Justesen JM, Katsuya T, Kibriya MG, Kim YJ, Kishimoto M, Koh WP, Kohara K, Kumari M, Kwek K, Lee NR, Lee J, Liao J, Lieb W, Liewald DCM, Matsubara T, Matsushita Y, Meitinger T, Mihailov E, Milani L, Mills R, Mononen N, Müller-Nurasyid M, Nabika T, Nakashima E, Ng HK, Nikus K, Nutile T, Ohkubo T, Ohnaka K, Parish S, Paternoster L, Peng H, Peters A, Pham ST, Pinidiyapathirage MJ, Rahman M, Rakugi H, Rolandsson O, Ann Rozario M, Ruggiero D, Sala CF, Sarju R, Shimokawa K, Snieder H, Sparsø T, Spiering W, Starr JM, Stott DJ, Stram DO, Sugiyama T, Szymczak S, Tang WHW, Tong L, Trompet S, Turjanmaa V, Ueshima H, Uitterlinden AG, Umemura S, Vaarasmaki M, van Dam RM, van Gilst WH, van Veldhuisen DJ, Viikari JS, Waldenberger M, Wang Y, Wang A, Wilson R, Wong TY, Xiang YB, Yamaguchi S, Ye X, Young RD, Young TL, Yuan JM, Zhou X, Asselbergs FW, Ciullo M, Clarke R, Deloukas P, Franke A, Franks PW, Franks S, Friedlander Y, Gross MD, Guo Z, Hansen T, Jarvelin MR, Jørgensen T, Jukema JW, kähönen M, Kajio H, Kivimaki M, Lee JY, Lehtimäki T, Linneberg A, Miki T, Pedersen O, Samani NJ, Sørensen TIA, Takayanagi R, Toniolo D, BIOS-consortium, CARDIo GRAMplusCD, LifeLines Cohort Study, The InterAct Consortium, Ahsan H, Allayee H, Chen YT, Danesh J, Deary IJ, Franco OH, Franke L, Heijman BT, Holbrook JD, Isaacs A, Kim BJ, Lin X, Liu J, März W, Metspalu A, Mohlke KL, Sanghera DK, Shu XO, van Meurs JBJ, Vithana E, Wickremasinghe AR, Wijmenga C, Wolffenbuttel BHW, Yokota M, Zheng W, Zhu D, Vineis P, Kyrtopoulos SA, Kleinjans JCS, McCarthy MI, Soong R, Gieger C, Scott J, Teo YY, He J, Elliott P, Tai ES, van der Harst P, Kooner JS, Chambers JC. Trans-ancestry genome-wide association study identifies 12 genetic loci influencing blood pressure and implicates a role for DNA methylation. Nat Genet 2015; 47:1282-1293. [PMID: 26390057 PMCID: PMC4719169 DOI: 10.1038/ng.3405] [Show More Authors] [Citation(s) in RCA: 256] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 08/21/2015] [Indexed: 12/17/2022]
Abstract
We carried out a trans-ancestry genome-wide association and replication study of blood pressure phenotypes among up to 320,251 individuals of East Asian, European and South Asian ancestry. We find genetic variants at 12 new loci to be associated with blood pressure (P = 3.9 × 10(-11) to 5.0 × 10(-21)). The sentinel blood pressure SNPs are enriched for association with DNA methylation at multiple nearby CpG sites, suggesting that, at some of the loci identified, DNA methylation may lie on the regulatory pathway linking sequence variation to blood pressure. The sentinel SNPs at the 12 new loci point to genes involved in vascular smooth muscle (IGFBP3, KCNK3, PDE3A and PRDM6) and renal (ARHGAP24, OSR1, SLC22A7 and TBX2) function. The new and known genetic variants predict increased left ventricular mass, circulating levels of NT-proBNP, and cardiovascular and all-cause mortality (P = 0.04 to 8.6 × 10(-6)). Our results provide new evidence for the role of DNA methylation in blood pressure regulation.
Collapse
Affiliation(s)
- Norihiro Kato
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Marie Loh
- Institute of Health Sciences, University of Oulu, Oulu, Finland
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Fumihiko Takeuchi
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Niek Verweij
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Xu Wang
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Ealing Hospital National Health Service (NHS) Trust, Middlesex, UK
| | - Tanika N Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Danish Saleheen
- Center for Non-Communicable Diseases, Karachi, Pakistan
- Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
- Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Benjamin Lehne
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Irene Mateo Leach
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Alexander W Drong
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - James Abbott
- Bioinformatics Support Service, Imperial College London, London, UK
| | - Simone Wahl
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Sian-Tsung Tan
- Ealing Hospital National Health Service (NHS) Trust, Middlesex, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - William R Scott
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Gianluca Campanella
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Marc Chadeau-Hyam
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Uzma Afzal
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Ealing Hospital National Health Service (NHS) Trust, Middlesex, UK
| | - Tarunveer S Ahluwalia
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Copenhagen Prospective Studies on Asthma in Childhood (COSPAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center, Gentofte, Denmark
| | - Marc Jan Bonder
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Peng Chen
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Todd L Edwards
- Vanderbilt Epidemiology Center, Center for Human Genetics Research, Division of Epidemiology, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Tõnu Esko
- Estonian Genome Center, University of Tartu, Tartu, Estonia
- Division of Endocrinology, Children’s Hospital Boston, Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA
| | - Min Jin Go
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, Republic of Korea
| | - Sarah E Harris
- Medical Genetics Section, University of Edinburgh Centre for Genomic and Experimental Medicine and Medical Research Council (MRC) Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, UK
- Centre for Cognitive Aging and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Jaana Hartiala
- Department of Preventive Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, USA
- Institute for Genetic Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Silva Kasela
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | | | - Chiea-Chuen Khor
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
- Genome Institute of Singapore, A*STAR, Singapore
- Department of Ophthalmology, National University of Singapore, Singapore
- Department of Paediatrics, National University of Singapore, Singapore
| | - Marcus E Kleber
- Medical Clinic V, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Huaixing Li
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zuan Yu Mok
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Masahiro Nakatochi
- Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Japan
| | - Nur Sabrina Sapari
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Richa Saxena
- Center for Human Genetic Research, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alexandre F R Stewart
- University of Ottawa Heart Institute, Cardiovascular Research Methods Centre, Ottawa, Ontario, Canada
- Ruddy Canadian Cardiovascular Genetics Centre, Ottawa, Ontario, Canada
| | - Lisette Stolk
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Yasuharu Tabara
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ai Ling Teh
- Singapore Institute for Clinical Sciences (SICS), A*STAR, Singapore
| | - Ying Wu
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jer-Yuarn Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Yi Zhang
- State Key Laboratory of Medical Genetics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Hypertension, Shanghai, China
| | - Imke Aits
- Institute of Epidemiology and Biobank popgen, Christian Albrechts University of Kiel, Kiel, Germany
| | - Alexessander Da Silva Couto Alves
- Department of Epidemiology and Biostatistics, MRC Health Protection Agency (PHE) Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | - Shikta Das
- Department of Epidemiology and Biostatistics, MRC Health Protection Agency (PHE) Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | | | - Jemma C Hopewell
- Clinical Trials Support Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Yun Kyoung Kim
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, Republic of Korea
| | - Robert W Koivula
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Skåne University Hospital Malmö, Malmö, Sweden
| | - Jian’an Luan
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere, Finland
| | - Quang N Nguyen
- Vietnam National Heart Institute, Bach Mai Hospital, Hanoi, Vietnam
| | - Mark A Pereira
- School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Iris Postmus
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
- Netherlands Consortium for Healthy Ageing, Leiden, the Netherlands
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Molly Scannell Bryan
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois, USA
| | - Robert A Scott
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Rossella Sorice
- Institute of Genetics and Biophysics A Buzzati-Traverso, CNR, Naples, Italy
| | - Vinicius Tragante
- Division of Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Michela Traglia
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
- Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) ‘Burlo Garofolo’, Trieste, Italy
| | - Jon White
- University College London Genetics Institute, Department of Genetics, Environment and Evolution, University College London, London, UK
| | - Ken Yamamoto
- Division of Genomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yonghong Zhang
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Linda S Adair
- Department of Nutrition, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | - Koichi Akiyama
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Rasheed Asif
- Center for Non-Communicable Diseases, Karachi, Pakistan
| | - Tin Aung
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Inês Barroso
- Metabolic Disease Group, Wellcome Trust Sanger Institute, Cambridge, UK
- National Institute for Health Research (NIHR) Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| | - Andrew Bjonnes
- Center for Human Genetic Research, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Timothy R Braun
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Hui Cai
- Vanderbilt Epidemiology Center, Division of Epidemiology, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Division of Epidemiology, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Li-Ching Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chien-Hsiun Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Ching-Yu Cheng
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
- Department of Ophthalmology, National University of Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Centre for Quantitative Medicine, Office of Clinical Sciences, Duke–National University of Singapore Graduate Medical School, Singapore
| | - Yap-Seng Chong
- Singapore Institute for Clinical Sciences (SICS), A*STAR, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Rory Collins
- Clinical Trials Support Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Regina Courtney
- Vanderbilt Epidemiology Center, Division of Epidemiology, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Division of Epidemiology, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Gail Davies
- Centre for Cognitive Aging and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Graciela Delgado
- Medical Clinic V, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Loi D Do
- Vietnam National Heart Institute, Bach Mai Hospital, Hanoi, Vietnam
| | - Pieter A Doevendans
- Division of Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ron T Gansevoort
- Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Yu-Tang Gao
- Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China
| | - Tanja B Grammer
- Medical Clinic V, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Niels Grarup
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jagvir Grewal
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Ealing Hospital National Health Service (NHS) Trust, Middlesex, UK
| | - Dongfeng Gu
- Fu Wai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gurpreet S Wander
- Dayanand Medical College and Hospital Unit, Hero DMC Heart Institute, Ludhiana, India
| | - Anna-Liisa Hartikainen
- Department of Obstetrics and Gynecology, Oulu University Hospital, Oulu, Finland
- Medical Research Center, University of Oulu, Oulu, Finland
- Unit of Primary Care, Oulu University Hospital, Oulu, Finland
| | - Stanley L Hazen
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jing He
- Department of Biostatistics, Vanderbilt University, Nashville, Tennessee, USA
| | - Chew-Kiat Heng
- Department of Paediatrics, Yong Loo Lin School of Medicine, Singapore
| | - James E Hixson
- Human Genetics Center, University of Texas School of Public Health at Houston, Houston, Texas, USA
| | - Albert Hofman
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Chris Hsu
- University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Wei Huang
- Department of Genetics, Chinese National Human Genomic Center, Shanghai, China
| | - Lise L N Husemoen
- Research Centre for Prevention and Health, Glostrup University Hospital, Glostrup, Denmark
| | - Joo-Yeon Hwang
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, Republic of Korea
| | - Sahoko Ichihara
- Graduate School of Regional Innovation Studies, Mie University, Tsu, Japan
| | - Michiya Igase
- Department of Geriatric Medicine, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Masato Isono
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Johanne M Justesen
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tomohiro Katsuya
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Geriatric Medicine and Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Muhammad G Kibriya
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois, USA
| | - Young Jin Kim
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, Republic of Korea
| | | | - Woon-Puay Koh
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
- Duke–National University of Singapore Graduate Medical School, Singapore
| | - Katsuhiko Kohara
- Department of Geriatric Medicine, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Meena Kumari
- Department of Epidemiology and Public Health, University College London, London, UK
| | | | - Nanette R Lee
- University of San Carlos Office of Population Studies Foundation, University of San Carlos, Cebu City, Philippines
- Department of Anthropology, Sociology and History, University of San Carlos, Cebu City, Philippines
| | - Jeannette Lee
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Jiemin Liao
- Department of Ophthalmology, National University of Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Wolfgang Lieb
- Institute of Epidemiology and Biobank popgen, Christian Albrechts University of Kiel, Kiel, Germany
| | - David C M Liewald
- Centre for Cognitive Aging and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Tatsuaki Matsubara
- Department of Internal Medicine, Aichi-Gakuin University School of Dentistry, Nagoya, Japan
| | - Yumi Matsushita
- National Center for Global Health and Medicine, Toyama, Japan
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Human Genetics, Technische Universität München, Munich, Germany
| | | | - Lili Milani
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Rebecca Mills
- Ealing Hospital National Health Service (NHS) Trust, Middlesex, UK
| | - Nina Mononen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere, Finland
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- Department of Medicine I, Ludwig Maximilians University Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Toru Nabika
- Department of Functional Pathology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Eitaro Nakashima
- Division of Endocrinology and Diabetes, Department of Internal Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Diabetes and Endocrinology, Chubu Rosai Hospital, Nagoya, Japan
| | - Hong Kiat Ng
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Kjell Nikus
- Heart Centre, Department of Cardiology, Tampere University Hospital and University of Tampere School of Medicine, Tampere, Finland
| | - Teresa Nutile
- Institute of Genetics and Biophysics A Buzzati-Traverso, CNR, Naples, Italy
| | - Takayoshi Ohkubo
- Department of Hygiene and Public Health, Teikyo University School of Medicine, Tokyo, Japan
| | - Keizo Ohnaka
- Department of Geriatric Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Sarah Parish
- Clinical Trials Support Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | | | - Hao Peng
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Son T Pham
- Vietnam National Heart Institute, Bach Mai Hospital, Hanoi, Vietnam
| | | | - Mahfuzar Rahman
- UChicago Research Bangladesh, Uttara, Dhaka, Bangladesh
- Research and Evaluation Division, Bangladesh Rehabilitation Assistance Committee (BRAC), Dhaka, Bangladesh
| | - Hiromi Rakugi
- Department of Geriatric Medicine and Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Olov Rolandsson
- Department of Public Health and Clinical Medicine, Section for Family Medicine, Umeå Universitet, Umeå, Sweden
| | - Michelle Ann Rozario
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Daniela Ruggiero
- Institute of Genetics and Biophysics A Buzzati-Traverso, CNR, Naples, Italy
| | - Cinzia F Sala
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Ralhan Sarju
- Dayanand Medical College and Hospital Unit, Hero DMC Heart Institute, Ludhiana, India
| | - Kazuro Shimokawa
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Thomas Sparsø
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Wilko Spiering
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
| | - John M Starr
- Centre for Cognitive Aging and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
| | - David J Stott
- Academic Section of Geriatric Medicine, Institute of Cardiovascular and Medical Sciences, Faculty of Medicine, University of Glasgow, Glasgow, UK
| | - Daniel O Stram
- University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Takao Sugiyama
- Institute for Adult Diseases, Asahi Life Foundation, Tokyo, Japan
| | - Silke Szymczak
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - W H Wilson Tang
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Lin Tong
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois, USA
| | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Väinö Turjanmaa
- Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
- Department of Clinical Physiology, University of Tampere School of Medicine, Tampere, Finland
| | - Hirotsugu Ueshima
- Department of Health Science, Shiga University of Medical Science, Otsu, Japan
- Center for Epidemiologic Research in Asia, Shiga University of Medical Science, Otsu, Japan
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Satoshi Umemura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Marja Vaarasmaki
- Department of Obstetrics and Gynecology, Oulu University Hospital, Oulu, Finland
- Medical Research Center, University of Oulu, Oulu, Finland
- Unit of Primary Care, Oulu University Hospital, Oulu, Finland
| | - Rob M van Dam
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Wiek H van Gilst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Dirk J van Veldhuisen
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jorma S Viikari
- Division of Medicine, Turku University Hospital, Turku, Finland
- Department of Medicine, University of Turku, Turku, Finland
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
| | - Yiqin Wang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Aili Wang
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Rory Wilson
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
| | - Tien-Yin Wong
- Department of Ophthalmology, National University of Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Yong-Bing Xiang
- Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China
| | - Shuhei Yamaguchi
- Third Department of Internal Medicine, Shimane University Faculty of Medicine, Izumo, Japan
| | - Xingwang Ye
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Robin D Young
- Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Terri L Young
- Neuroscience and Behavioural Disorders (NBD) Program, Duke–National University of Singapore Graduate Medical School, Singapore
- Duke Eye Center, Duke University Medical Center, Durham, North Carolina, USA
| | - Jian-Min Yuan
- Cancer Control and Population Sciences, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | - Xueya Zhou
- Bioinformatics Division, Tsinghua National Laboratory for Informatics Science and Technology (TNLIST), Ministry of Education Key Laboratory of Bioinformatics, Department of Automation, Tsinghua University, Beijing, China
- Center for Synthetic and Systems Biology, TNLIST, Ministry of Education Key Laboratory of Bioinformatics, Department of Automation, Tsinghua University, Beijing, China
- Department of Psychiatry, University of Hong Kong, Hong Kong
| | - Folkert W Asselbergs
- Division of Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
- Durrer Center for Cardiogenetic Research, Interuniversity Cardiology Institute of the Netherlands (ICIN)–Netherlands Heart Institute, Utrecht, the Netherlands
- Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, UK
| | - Marina Ciullo
- Institute of Genetics and Biophysics A Buzzati-Traverso, CNR, Naples, Italy
| | - Robert Clarke
- Clinical Trials Support Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Panos Deloukas
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- King Abdulaziz University, Jeddah, Saudi Arabia
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - Paul W Franks
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Skåne University Hospital Malmö, Malmö, Sweden
- Department of Public Health and Clinical Medicine, Section for Family Medicine, Umeå Universitet, Umeå, Sweden
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Steve Franks
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Hospital, London, UK
| | | | - Myron D Gross
- School of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Zhirong Guo
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Torben Hansen
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marjo-Riitta Jarvelin
- Institute of Health Sciences, University of Oulu, Oulu, Finland
- Department of Epidemiology and Biostatistics, MRC Health Protection Agency (PHE) Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- Unit of Primary Care, Oulu University Hospital, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Center for Life Course Epidemiology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Torben Jørgensen
- Research Centre for Prevention and Health, Glostrup University Hospital, Glostrup, Denmark
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Durrer Center for Cardiogenetic Research, Interuniversity Cardiology Institute of the Netherlands (ICIN)–Netherlands Heart Institute, Utrecht, the Netherlands
- ICIN, Utrecht, the Netherlands
| | - Mika kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
- Department of Clinical Physiology, University of Tampere School of Medicine, Tampere, Finland
| | - Hiroshi Kajio
- National Center for Global Health and Medicine, Toyama, Japan
| | - Mika Kivimaki
- Department of Epidemiology and Public Health, University College London, London, UK
| | - Jong-Young Lee
- Ministry of Health and Welfare, Seoul, Republic of Korea
- THERAGEN ETEX Bio Institute, Suwon, Republic of Korea
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere, Finland
| | - Allan Linneberg
- Research Centre for Prevention and Health, Glostrup University Hospital, Glostrup, Denmark
- Department of Clinical Experimental Research, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tetsuro Miki
- Department of Geriatric Medicine, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Oluf Pedersen
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, UK
- NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, UK
| | - Thorkild I A Sørensen
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Institute of Preventive Medicine, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Ryoichi Takayanagi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Kyushu, Japan
| | - Daniela Toniolo
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
- Institute of Molecular Genetics, National Research Council (CNR), Pavia, Italy
| | | | | | | | | | - Habibul Ahsan
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois, USA
| | - Hooman Allayee
- Department of Preventive Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, USA
- Institute for Genetic Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Yuan-Tsong Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - John Danesh
- Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
- Wellcome Trust Sanger Institute, Hinxton, UK
| | - Ian J Deary
- Centre for Cognitive Aging and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Oscar H Franco
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Lude Franke
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Bastiaan T Heijman
- Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Aaron Isaacs
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Bong-Jo Kim
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, Republic of Korea
| | - Xu Lin
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jianjun Liu
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
- Genome Institute of Singapore, A*STAR, Singapore
| | - Winfried März
- Medical Clinic V, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
- Synlab Academy, Synlab Services, Mannheim, Germany
| | | | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Dharambir K Sanghera
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Xiao-Ou Shu
- Vanderbilt Epidemiology Center, Division of Epidemiology, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Division of Epidemiology, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Joyce B J van Meurs
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Eranga Vithana
- Department of Ophthalmology, National University of Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Neuroscience and Behavioural Disorders (NBD) Program, Duke–National University of Singapore Graduate Medical School, Singapore
| | | | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Bruce H W Wolffenbuttel
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Mitsuhiro Yokota
- Department of Genome Science, Aichi-Gakuin University School of Dentistry, Nagoya, Japan
| | - Wei Zheng
- Vanderbilt Epidemiology Center, Division of Epidemiology, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Division of Epidemiology, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Dingliang Zhu
- State Key Laboratory of Medical Genetics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Hypertension, Shanghai, China
| | - Paolo Vineis
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Soterios A Kyrtopoulos
- National Hellenic Research Foundation, Institute of Biology, Medicinal Chemistry and Biotechnology, Athens, Greece
| | - Jos C S Kleinjans
- Department of Toxicogenomics, Maastricht University, Maastricht, the Netherlands
| | - Mark I McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Richie Soong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- Department of Pathology, National University of Singapore, Singapore
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
| | - James Scott
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Yik-Ying Teo
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
- Genome Institute of Singapore, A*STAR, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- National University of Singapore Graduate School for Integrative Science and Engineering, National University of Singapore, Singapore
- Life Sciences Institute, National University of Singapore, Singapore
- Department of Statistics and Applied Probability, National University of Singapore, Singapore
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - E Shyong Tai
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Durrer Center for Cardiogenetic Research, Interuniversity Cardiology Institute of the Netherlands (ICIN)–Netherlands Heart Institute, Utrecht, the Netherlands
| | - Jaspal S Kooner
- Ealing Hospital National Health Service (NHS) Trust, Middlesex, UK
- National Heart and Lung Institute, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - John C Chambers
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Ealing Hospital National Health Service (NHS) Trust, Middlesex, UK
- Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
111
|
Böger CA, Hayward C. The PediGFR consortium: studying the genetics of complex kidney traits in children. Nephrol Dial Transplant 2015; 31:173-5. [PMID: 26516202 DOI: 10.1093/ndt/gfv356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 09/09/2015] [Indexed: 11/13/2022] Open
Affiliation(s)
- Carsten A Böger
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Caroline Hayward
- MRCHGU, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
112
|
Sekula P, Goek ON, Quaye L, Barrios C, Levey AS, Römisch-Margl W, Menni C, Yet I, Gieger C, Inker LA, Adamski J, Gronwald W, Illig T, Dettmer K, Krumsiek J, Oefner PJ, Valdes AM, Meisinger C, Coresh J, Spector TD, Mohney RP, Suhre K, Kastenmüller G, Köttgen A. A Metabolome-Wide Association Study of Kidney Function and Disease in the General Population. J Am Soc Nephrol 2015; 27:1175-88. [PMID: 26449609 DOI: 10.1681/asn.2014111099] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 07/28/2015] [Indexed: 12/25/2022] Open
Abstract
Small molecules are extensively metabolized and cleared by the kidney. Changes in serum metabolite concentrations may result from impaired kidney function and can be used to estimate filtration (e.g., the established marker creatinine) or may precede and potentially contribute to CKD development. Here, we applied a nontargeted metabolomics approach using gas and liquid chromatography coupled to mass spectrometry to quantify 493 small molecules in human serum. The associations of these molecules with GFR estimated on the basis of creatinine (eGFRcr) and cystatin C levels were assessed in ≤1735 participants in the KORA F4 study, followed by replication in 1164 individuals in the TwinsUK registry. After correction for multiple testing, 54 replicated metabolites significantly associated with eGFRcr, and six of these showed pairwise correlation (r≥0.50) with established kidney function measures: C-mannosyltryptophan, pseudouridine, N-acetylalanine, erythronate, myo-inositol, and N-acetylcarnosine. Higher C-mannosyltryptophan, pseudouridine, and O-sulfo-L-tyrosine concentrations associated with incident CKD (eGFRcr <60 ml/min per 1.73 m(2)) in the KORA F4 study. In contrast with serum creatinine, C-mannosyltryptophan and pseudouridine concentrations showed little dependence on sex. Furthermore, correlation with measured GFR in 200 participants in the AASK study was 0.78 for both C-mannosyltryptophan and pseudouridine concentration, and highly significant associations of both metabolites with incident ESRD disappeared upon adjustment for measured GFR. Thus, these molecules may be alternative or complementary markers of kidney function. In conclusion, our study provides a comprehensive list of kidney function-associated metabolites and highlights potential novel filtration markers that may help to improve the estimation of GFR.
Collapse
Affiliation(s)
- Peggy Sekula
- Division of Nephrology and Center for Medical Biometry and Medical Informatics, Medical Center-University of Freiburg, Freiburg, Germany
| | | | - Lydia Quaye
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Clara Barrios
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom; Department of Nephrology, Hospital del Mar, Institut Mar d'Investigacions Mediques, Barcelona, Spain
| | - Andrew S Levey
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | | | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Idil Yet
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | | | - Lesley A Inker
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Jerzy Adamski
- Experimental Genetics, Genome Analysis Center, German Center for Diabetes Research, Neuherberg, Germany; Institute of Experimental Genetics, Technical University of Munich, Freising-Weihenstephan, Germany
| | - Wolfram Gronwald
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Thomas Illig
- Research Unit of Molecular Epidemiology and Hannover Unified Biobank and Institute for Human Genetics, Hannover Medical School, Hannover, Germany
| | - Katja Dettmer
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | | | - Peter J Oefner
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Ana M Valdes
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom; Academic Rheumatology, University of Nottingham, Nottingham, United Kingdom
| | - Christa Meisinger
- Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Josef Coresh
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | | | - Karsten Suhre
- Institutes of Bioinformatics and Systems Biology, Department of Physiology and Biophysics, Weill Cornell Medical College-Qatar, Doha, Qatar
| | - Gabi Kastenmüller
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom; Institutes of Bioinformatics and Systems Biology, German Center for Diabetes Research, Neuherberg, Germany;
| | | |
Collapse
|
113
|
Burkhardt R, Kirsten H, Beutner F, Holdt LM, Gross A, Teren A, Tönjes A, Becker S, Krohn K, Kovacs P, Stumvoll M, Teupser D, Thiery J, Ceglarek U, Scholz M. Integration of Genome-Wide SNP Data and Gene-Expression Profiles Reveals Six Novel Loci and Regulatory Mechanisms for Amino Acids and Acylcarnitines in Whole Blood. PLoS Genet 2015; 11:e1005510. [PMID: 26401656 PMCID: PMC4581711 DOI: 10.1371/journal.pgen.1005510] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 08/17/2015] [Indexed: 01/23/2023] Open
Abstract
Profiling amino acids and acylcarnitines in whole blood spots is a powerful tool in the laboratory diagnosis of several inborn errors of metabolism. Emerging data suggests that altered blood levels of amino acids and acylcarnitines are also associated with common metabolic diseases in adults. Thus, the identification of common genetic determinants for blood metabolites might shed light on pathways contributing to human physiology and common diseases. We applied a targeted mass-spectrometry-based method to analyze whole blood concentrations of 96 amino acids, acylcarnitines and pathway associated metabolite ratios in a Central European cohort of 2,107 adults and performed genome-wide association (GWA) to identify genetic modifiers of metabolite concentrations. We discovered and replicated six novel loci associated with blood levels of total acylcarnitine, arginine (both on chromosome 6; rs12210538, rs17657775), propionylcarnitine (chromosome 10; rs12779637), 2-hydroxyisovalerylcarnitine (chromosome 21; rs1571700), stearoylcarnitine (chromosome 1; rs3811444), and aspartic acid traits (chromosome 8; rs750472). Based on an integrative analysis of expression quantitative trait loci in blood mononuclear cells and correlations between gene expressions and metabolite levels, we provide evidence for putative causative genes: SLC22A16 for total acylcarnitines, ARG1 for arginine, HLCS for 2-hydroxyisovalerylcarnitine, JAM3 for stearoylcarnitine via a trans-effect at chromosome 1, and PPP1R16A for aspartic acid traits. Further, we report replication and provide additional functional evidence for ten loci that have previously been published for metabolites measured in plasma, serum or urine. In conclusion, our integrative analysis of SNP, gene-expression and metabolite data points to novel genetic factors that may be involved in the regulation of human metabolism. At several loci, we provide evidence for metabolite regulation via gene-expression and observed overlaps with GWAS loci for common diseases. These results form a strong rationale for subsequent functional and disease-related studies.
Collapse
Affiliation(s)
- Ralph Burkhardt
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Holger Kirsten
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- Department for Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Frank Beutner
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Heart Center Leipzig, Leipzig, Germany
| | - Lesca M. Holdt
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute for Laboratory Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Arnd Gross
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Andrej Teren
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Heart Center Leipzig, Leipzig, Germany
| | - Anke Tönjes
- Medical Department, Clinic for Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
| | - Susen Becker
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Knut Krohn
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Interdisciplinary Centre for Clinical Research, University of Leipzig, Leipzig, Germany
| | - Peter Kovacs
- Integrated Research and Treatment Center Adiposity Diseases, University of Leipzig, Leipzig Germany
| | - Michael Stumvoll
- Medical Department, Clinic for Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
- Integrated Research and Treatment Center Adiposity Diseases, University of Leipzig, Leipzig Germany
| | - Daniel Teupser
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute for Laboratory Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Joachim Thiery
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Uta Ceglarek
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Markus Scholz
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- * E-mail:
| |
Collapse
|
114
|
Genome-Wide Association Study with Targeted and Non-targeted NMR Metabolomics Identifies 15 Novel Loci of Urinary Human Metabolic Individuality. PLoS Genet 2015; 11:e1005487. [PMID: 26352407 PMCID: PMC4564198 DOI: 10.1371/journal.pgen.1005487] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 08/06/2015] [Indexed: 12/24/2022] Open
Abstract
Genome-wide association studies with metabolic traits (mGWAS) uncovered many genetic variants that influence human metabolism. These genetically influenced metabotypes (GIMs) contribute to our metabolic individuality, our capacity to respond to environmental challenges, and our susceptibility to specific diseases. While metabolic homeostasis in blood is a well investigated topic in large mGWAS with over 150 known loci, metabolic detoxification through urinary excretion has only been addressed by few small mGWAS with only 11 associated loci so far. Here we report the largest mGWAS to date, combining targeted and non-targeted 1H NMR analysis of urine samples from 3,861 participants of the SHIP-0 cohort and 1,691 subjects of the KORA F4 cohort. We identified and replicated 22 loci with significant associations with urinary traits, 15 of which are new (HIBCH, CPS1, AGXT, XYLB, TKT, ETNPPL, SLC6A19, DMGDH, SLC36A2, GLDC, SLC6A13, ACSM3, SLC5A11, PNMT, SLC13A3). Two-thirds of the urinary loci also have a metabolite association in blood. For all but one of the 6 loci where significant associations target the same metabolite in blood and urine, the genetic effects have the same direction in both fluids. In contrast, for the SLC5A11 locus, we found increased levels of myo-inositol in urine whereas mGWAS in blood reported decreased levels for the same genetic variant. This might indicate less effective re-absorption of myo-inositol in the kidneys of carriers. In summary, our study more than doubles the number of known loci that influence urinary phenotypes. It thus allows novel insights into the relationship between blood homeostasis and its regulation through excretion. The newly discovered loci also include variants previously linked to chronic kidney disease (CPS1, SLC6A13), pulmonary hypertension (CPS1), and ischemic stroke (XYLB). By establishing connections from gene to disease via metabolic traits our results provide novel hypotheses about molecular mechanisms involved in the etiology of diseases. Human metabolism is influenced by genetic and environmental factors defining a person’s metabolic individuality. This individuality is linked to personal differences in the ability to react on metabolic challenges and in the susceptibility to specific diseases. By investigating how common variants in genetic regions (loci) affect individual blood metabolite levels, the substantial contribution of genetic inheritance to metabolic individuality has been demonstrated previously. Meanwhile, more than 150 loci influencing metabolic homeostasis in blood are known. Here we shift the focus to genetic variants that modulate urinary metabolite excretion, for which only 11 loci were reported so far. In the largest genetic study on urinary metabolites to date, we identified 15 additional loci. Most of the 26 loci also affect blood metabolite levels. This shows that the metabolic individuality seen in blood is also reflected in urine, which is expected when urine is regarded as “diluted blood”. Nonetheless, we also found loci that appear to primarily influence metabolite excretion. For instance, we identified genetic variants near a gene of a transporter that change the capability for renal re-absorption of the transporter’s substrate. Thus, our findings could help to elucidate molecular mechanisms influencing kidney function and the body’s detoxification capabilities.
Collapse
|
115
|
Ma RCW. Genetics of cardiovascular and renal complications in diabetes. J Diabetes Investig 2015; 7:139-54. [PMID: 27042264 PMCID: PMC4773661 DOI: 10.1111/jdi.12391] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 06/29/2015] [Accepted: 06/29/2015] [Indexed: 01/29/2023] Open
Abstract
The development of debilitating complications represents a major heathcare burden associated with the treatment of diabetes. Despite advances in new therapies for controlling hyperglycemia, the burden associated with diabetic complications remains high, especially in relation to cardiovascular and renal complications. Furthermore, an increasing proportion of patients develop type 2 diabetes at a younger age, putting them at higher risk of developing complications as a result of the increased exposure to hyperglycemia. Diabetes has become the main contributing cause to end‐stage renal disease in most countries. Although there has been important breakthroughs in our understanding of the genetics of type 1 and type 2 diabetes, bringing important insights towards the pathogenesis of diabetes, there has been comparatively less progress in our understanding of the genetic basis of diabetic complications. Genome‐wide association studies are beginning to expand our understanding of the genetic architecture relating to diabetic complications. Improved understanding of the genetic basis of diabetic cardiorenal complications might provide an opportunity for improved risk prediction, as well as the development of new therapies.
Collapse
Affiliation(s)
- Ronald C W Ma
- Department of Medicine and Therapeutics The Chinese University of Hong Kong Hong Kong; Hong Kong Institute of Diabetes and Obesity The Chinese University of Hong Kong Hong Kong; Li Ka Shing Institute of Health Sciences The Chinese University of Hong Kong Hong Kong
| |
Collapse
|
116
|
Breit M, Weinberger KM. Metabolic biomarkers for chronic kidney disease. Arch Biochem Biophys 2015; 589:62-80. [PMID: 26235490 DOI: 10.1016/j.abb.2015.07.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 07/11/2015] [Accepted: 07/26/2015] [Indexed: 01/28/2023]
Abstract
Chronic kidney disease (CKD) is an increasingly recognized burden for patients and health care systems with high (and growing) global incidence and prevalence, significant mortality, and disproportionately high treatment costs. Yet, the available diagnostic tools are either impractical in clinical routine or have serious shortcomings impeding a well-informed disease management although optimized treatment strategies with proven benefits for the patients have become available. Advances in bioanalytical technologies have facilitated studies that identified genomic, proteomic, and metabolic biomarker candidates, and confirmed some of them in independent cohorts. This review summarizes the CKD-related markers discovered so far, and focuses on compounds and pathways, for which there is quantitative data, substantiating evidence from translational research, and a mechanistic understanding of the processes involved. Also, multiparametric marker panels have been suggested that showed promising diagnostic and prognostic performance in initial analyses although the data basis from prospective trials is very limited. Large-scale studies, however, are underway and will provide the information for validating a set of parameters and discarding others. Finally, the path from clinical research to a routine application is discussed, focusing on potential obstacles such as the use of mass spectrometry, and the feasibility of obtaining regulatory approval for targeted metabolomics assays.
Collapse
Affiliation(s)
- Marc Breit
- Research Group for Clinical Bioinformatics, Institute of Electrical and Biomedical Engineering (IEBE), University for Health Sciences, Medical Informatics and Technology (UMIT), 6060 Hall in Tirol, Austria
| | - Klaus M Weinberger
- Research Group for Clinical Bioinformatics, Institute of Electrical and Biomedical Engineering (IEBE), University for Health Sciences, Medical Informatics and Technology (UMIT), 6060 Hall in Tirol, Austria; sAnalytiCo Ltd., Forsyth House, Cromac Square, Belfast BT2 8LA, United Kingdom.
| |
Collapse
|
117
|
Kastenmüller G, Raffler J, Gieger C, Suhre K. Genetics of human metabolism: an update. Hum Mol Genet 2015; 24:R93-R101. [PMID: 26160913 PMCID: PMC4572003 DOI: 10.1093/hmg/ddv263] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 07/06/2015] [Indexed: 01/01/2023] Open
Abstract
Genome-wide association studies with metabolomics (mGWAS) identify genetically influenced metabotypes (GIMs), their ensemble defining the heritable part of every human's metabolic individuality. Knowledge of genetic variation in metabolism has many applications of biomedical and pharmaceutical interests, including the functional understanding of genetic associations with clinical end points, design of strategies to correct dysregulations in metabolic disorders and the identification of genetic effect modifiers of metabolic disease biomarkers. Furthermore, it has been shown that GIMs provide testable hypotheses for functional genomics and metabolomics and for the identification of novel gene functions and metabolite identities. mGWAS with growing sample sizes and increasingly complex metabolic trait panels are being conducted, allowing for more comprehensive and systems-based downstream analyses. The generated large datasets of genetic associations can now be mined by the biomedical research community and provide valuable resources for hypothesis-driven studies. In this review, we provide a brief summary of the key aspects of mGWAS, followed by an update of recently published mGWAS. We then discuss new approaches of integrating and exploring mGWAS results and finish by presenting selected applications of GIMs in recent studies.
Collapse
Affiliation(s)
- Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany, German Center for Diabetes Research, Neuherberg, Germany and
| | - Johannes Raffler
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany and Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Karsten Suhre
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany, Department of Physiology and Biophysics, Weill Cornell Medical College-Qatar, Doha, Qatar
| |
Collapse
|
118
|
|
119
|
Li H, Chen H, Liu F, Ren C, Wang S, Bo X, Shu W. Functional annotation of HOT regions in the human genome: implications for human disease and cancer. Sci Rep 2015; 5:11633. [PMID: 26113264 PMCID: PMC4481521 DOI: 10.1038/srep11633] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 06/01/2015] [Indexed: 12/17/2022] Open
Abstract
Advances in genome-wide association studies (GWAS) and large-scale sequencing studies have resulted in an impressive and growing list of disease- and trait-associated genetic variants. Most studies have emphasised the discovery of genetic variation in coding sequences, however, the noncoding regulatory effects responsible for human disease and cancer biology have been substantially understudied. To better characterise the cis-regulatory effects of noncoding variation, we performed a comprehensive analysis of the genetic variants in HOT (high-occupancy target) regions, which are considered to be one of the most intriguing findings of recent large-scale sequencing studies. We observed that GWAS variants that map to HOT regions undergo a substantial net decrease and illustrate development-specific localisation during haematopoiesis. Additionally, genetic risk variants are disproportionally enriched in HOT regions compared with LOT (low-occupancy target) regions in both disease-relevant and cancer cells. Importantly, this enrichment is biased toward disease- or cancer-specific cell types. Furthermore, we observed that cancer cells generally acquire cancer-specific HOT regions at oncogenes through diverse mechanisms of cancer pathogenesis. Collectively, our findings demonstrate the key roles of HOT regions in human disease and cancer and represent a critical step toward further understanding disease biology, diagnosis, and therapy.
Collapse
Affiliation(s)
- Hao Li
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Hebing Chen
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Feng Liu
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Chao Ren
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Shengqi Wang
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xiaochen Bo
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Wenjie Shu
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| |
Collapse
|
120
|
Stafford-Smith M, Li YJ, Mathew JP, Li YW, Ji Y, Phillips-Bute BG, Milano CA, Newman MF, Kraus WE, Kertai MD, Shah SH, Podgoreanu MV. Genome-wide association study of acute kidney injury after coronary bypass graft surgery identifies susceptibility loci. Kidney Int 2015; 88:823-32. [PMID: 26083657 PMCID: PMC4589439 DOI: 10.1038/ki.2015.161] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 03/18/2015] [Accepted: 04/09/2015] [Indexed: 12/25/2022]
Abstract
Acute kidney injury (AKI) is a common, serious complication of cardiac surgery. Since prior studies have supported a genetic basis for postoperative AKI, we conducted a genome-wide association study (GWAS) for AKI following coronary bypass graft (CABG) surgery. The discovery dataset consisted of 873 non-emergent CABG surgery patients with cardiopulmonary bypass (PEGASUS), while a replication dataset had 380 cardiac surgical patients (CATHGEN). Single nucleotide polymorphism (SNP) data were based on Illumina Human610-Quad (PEGASUS) and OMNI1-Quad (CATHGEN) BeadChips. We used linear regression with adjustment for a clinical AKI risk score to test SNP associations with the postoperative peak rise relative to preoperative serum creatinine concentration as a quantitative AKI trait. Nine SNPs meeting significance in the discovery set were detected. The rs13317787 in GRM7|LMCD1-AS1 intergenic region (3p21.6) and rs10262995 in BBS9 (7p14.3) were replicated with significance in the CATHGEN data set and exhibited significantly strong overall association following meta-analysis. Additional fine-mapping using imputed SNPs across these two regions and meta-analysis found genome wide significance at the GRM7|LMCD1-AS1 locus and a significantly strong association at BBS9. Thus, through an unbiased GWAS approach, we found two new loci associated with post-CABG AKI providing new insights into the pathogenesis of perioperative AKI.
Collapse
Affiliation(s)
- Mark Stafford-Smith
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Yi-Ju Li
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina, USA.,Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Joseph P Mathew
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Yen-Wei Li
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina, USA
| | - Yunqi Ji
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina, USA
| | | | - Carmelo A Milano
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Mark F Newman
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - William E Kraus
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, USA.,Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Miklos D Kertai
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Svati H Shah
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, USA.,Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Mihai V Podgoreanu
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | | |
Collapse
|
121
|
Van Driest SL, McGregor TL, Velez Edwards DR, Saville BR, Kitchner TE, Hebbring SJ, Brilliant M, Jouni H, Kullo IJ, Creech CB, Kannankeril PJ, Vear SI, Brothers KB, Bowton EA, Shaffer CM, Patel N, Delaney JT, Bradford Y, Wilson S, Olson LM, Crawford DC, Potts AL, Ho RH, Roden DM, Denny JC. Genome-Wide Association Study of Serum Creatinine Levels during Vancomycin Therapy. PLoS One 2015; 10:e0127791. [PMID: 26030142 PMCID: PMC4452656 DOI: 10.1371/journal.pone.0127791] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 04/18/2015] [Indexed: 11/18/2022] Open
Abstract
Vancomycin, a commonly used antibiotic, can be nephrotoxic. Known risk factors such as age, creatinine clearance, vancomycin dose / dosing interval, and concurrent nephrotoxic medications fail to accurately predict nephrotoxicity. To identify potential genomic risk factors, we performed a genome-wide association study (GWAS) of serum creatinine levels while on vancomycin in 489 European American individuals and validated findings in three independent cohorts totaling 439 European American individuals. In primary analyses, the chromosome 6q22.31 locus was associated with increased serum creatinine levels while on vancomycin therapy (most significant variant rs2789047, risk allele A, β = -0.06, p = 1.1 x 10-7). SNPs in this region had consistent directions of effect in the validation cohorts, with a meta-p of 1.1 x 10-7. Variation in this region on chromosome 6, which includes the genes TBC1D32/C6orf170 and GJA1 (encoding connexin43), may modulate risk of vancomycin-induced kidney injury.
Collapse
Affiliation(s)
- Sara L. Van Driest
- Department of Pediatrics, Vanderbilt University School of Medicine and the Monroe Carell Jr. Children’s Hospital at Vanderbilt, Nashville, Tennessee, United States of America
- * E-mail:
| | - Tracy L. McGregor
- Department of Pediatrics, Vanderbilt University School of Medicine and the Monroe Carell Jr. Children’s Hospital at Vanderbilt, Nashville, Tennessee, United States of America
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Digna R. Velez Edwards
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Obstetrics and Gynecology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Ben R. Saville
- Department of Biostatistics, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Terrie E. Kitchner
- Center for Human Genetics, Marshfield Clinic, Marshfield, Wisconsin, United States of America
| | - Scott J. Hebbring
- Center for Human Genetics, Marshfield Clinic, Marshfield, Wisconsin, United States of America
| | - Murray Brilliant
- Center for Human Genetics, Marshfield Clinic, Marshfield, Wisconsin, United States of America
| | - Hayan Jouni
- Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
| | - Iftikhar J. Kullo
- Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
| | - C. Buddy Creech
- Department of Pediatrics, Vanderbilt University School of Medicine and the Monroe Carell Jr. Children’s Hospital at Vanderbilt, Nashville, Tennessee, United States of America
| | - Prince J. Kannankeril
- Department of Pediatrics, Vanderbilt University School of Medicine and the Monroe Carell Jr. Children’s Hospital at Vanderbilt, Nashville, Tennessee, United States of America
| | - Susan I. Vear
- Department of Pediatrics, Vanderbilt University School of Medicine and the Monroe Carell Jr. Children’s Hospital at Vanderbilt, Nashville, Tennessee, United States of America
| | - Kyle B. Brothers
- Department of Pediatrics, Vanderbilt University School of Medicine and the Monroe Carell Jr. Children’s Hospital at Vanderbilt, Nashville, Tennessee, United States of America
| | - Erica A. Bowton
- Institute for Clinical and Translational Research, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Christian M. Shaffer
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Neelam Patel
- School of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Jessica T. Delaney
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Yuki Bradford
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Sarah Wilson
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Lana M. Olson
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Dana C. Crawford
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Amy L. Potts
- Department of Pharmaceutical Services, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Richard H. Ho
- Department of Pediatrics, Vanderbilt University School of Medicine and the Monroe Carell Jr. Children’s Hospital at Vanderbilt, Nashville, Tennessee, United States of America
| | - Dan M. Roden
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Josh C. Denny
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Biomedical Informatics, Vanderbilt University, Nashville, Tennessee, United States of America
| |
Collapse
|
122
|
Association of homoarginine and methylarginines with liver dysfunction and mortality in chronic liver disease. Amino Acids 2015; 47:1817-26. [PMID: 25952253 DOI: 10.1007/s00726-015-2000-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/27/2015] [Indexed: 01/18/2023]
Abstract
Previous studies on arginine metabolites reported an association of asymmetric dimethylarginine (ADMA) and symmetric dimethylarginine (SDMA) with liver dysfunction and an inverse relation of homoarginine (hArg) with cardiovascular risk. The aim of the present study was to investigate the relationships between hArg, ADMA, SDMA, and the dimethylarginine score (DAS, i.e., ADMA + SDMA) and liver dysfunction and survival in chronic liver disease. In 94 consecutive cirrhotic patients admitted to our outpatient liver clinic, serum levels of hArg, ADMA, and SDMA were measured by HPLC at baseline. Patients were followed with respect to mortality. In the entire study cohort (age 58.5 ± 11.2 years; 31 % females), the serum concentrations were 1.94 ± 0.90 µM for homoarginine, 0.90 ± 0.22 µM for ADMA, and 0.70 (0.60-0.93) µM for SDMA. ADMA correlated with both Child-Pugh and MELD scores, while SDMA, DAS, and hArg correlated with MELD score only. Thirty patients (32 %) died during a median follow-up of 3.5 years. Age- and sex-adjusted Cox proportional hazard ratios (HR) per µM (with 95 % confidence intervals) showed that hArg was associated with decreased mortality [HR 0.59 (0.37-0.96)], whereas mortality was increased in patients with higher ADMA [HR 3.78 (0.98-14.60)], SDMA [HR 6.54 (3.15-13.59)] and DAS [HR 4.13 (2.26-7.56)]. Only SDMA and DAS remained significantly associated with mortality after additional adjustments for either Child-Pugh stage or MELD score. In conclusion, in cirrhotic patients seen in an outpatient liver clinic, hArg as well as the dimethylarginines ADMA and SDMA was related to long-term mortality. In particular, SDMA predicts mortality independently of both Child-Pugh stage and MELD score.
Collapse
|
123
|
Gorski M, Tin A, Garnaas M, McMahon GM, Chu AY, Tayo BO, Pattaro C, Teumer A, Chasman DI, Chalmers J, Hamet P, Tremblay J, Woodward M, Aspelund T, Eiriksdottir G, Gudnason V, Harris TB, Launer LJ, Smith AV, Mitchell BD, O'Connell JR, Shuldiner AR, Coresh J, Li M, Freudenberger P, Hofer E, Schmidt H, Schmidt R, Holliday EG, Mitchell P, Wang JJ, de Boer IH, Li G, Siscovick DS, Kutalik Z, Corre T, Vollenweider P, Waeber G, Gupta J, Kanetsky PA, Hwang SJ, Olden M, Yang Q, de Andrade M, Atkinson EJ, Kardia SLR, Turner ST, Stafford JM, Ding J, Liu Y, Barlassina C, Cusi D, Salvi E, Staessen JA, Ridker PM, Grallert H, Meisinger C, Müller-Nurasyid M, Krämer BK, Kramer H, Rosas SE, Nolte IM, Penninx BW, Snieder H, Fabiola Del Greco M, Franke A, Nöthlings U, Lieb W, Bakker SJL, Gansevoort RT, van der Harst P, Dehghan A, Franco OH, Hofman A, Rivadeneira F, Sedaghat S, Uitterlinden AG, Coassin S, Haun M, Kollerits B, Kronenberg F, Paulweber B, Aumann N, Endlich K, Pietzner M, Völker U, Rettig R, Chouraki V, Helmer C, Lambert JC, Metzger M, Stengel B, Lehtimäki T, Lyytikäinen LP, Raitakari O, Johnson A, Parsa A, Bochud M, Heid IM, Goessling W, et alGorski M, Tin A, Garnaas M, McMahon GM, Chu AY, Tayo BO, Pattaro C, Teumer A, Chasman DI, Chalmers J, Hamet P, Tremblay J, Woodward M, Aspelund T, Eiriksdottir G, Gudnason V, Harris TB, Launer LJ, Smith AV, Mitchell BD, O'Connell JR, Shuldiner AR, Coresh J, Li M, Freudenberger P, Hofer E, Schmidt H, Schmidt R, Holliday EG, Mitchell P, Wang JJ, de Boer IH, Li G, Siscovick DS, Kutalik Z, Corre T, Vollenweider P, Waeber G, Gupta J, Kanetsky PA, Hwang SJ, Olden M, Yang Q, de Andrade M, Atkinson EJ, Kardia SLR, Turner ST, Stafford JM, Ding J, Liu Y, Barlassina C, Cusi D, Salvi E, Staessen JA, Ridker PM, Grallert H, Meisinger C, Müller-Nurasyid M, Krämer BK, Kramer H, Rosas SE, Nolte IM, Penninx BW, Snieder H, Fabiola Del Greco M, Franke A, Nöthlings U, Lieb W, Bakker SJL, Gansevoort RT, van der Harst P, Dehghan A, Franco OH, Hofman A, Rivadeneira F, Sedaghat S, Uitterlinden AG, Coassin S, Haun M, Kollerits B, Kronenberg F, Paulweber B, Aumann N, Endlich K, Pietzner M, Völker U, Rettig R, Chouraki V, Helmer C, Lambert JC, Metzger M, Stengel B, Lehtimäki T, Lyytikäinen LP, Raitakari O, Johnson A, Parsa A, Bochud M, Heid IM, Goessling W, Köttgen A, Kao WHL, Fox CS, Böger CA. Genome-wide association study of kidney function decline in individuals of European descent. Kidney Int 2015; 87:1017-29. [PMID: 25493955 PMCID: PMC4425568 DOI: 10.1038/ki.2014.361] [Show More Authors] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 09/04/2014] [Accepted: 09/11/2014] [Indexed: 11/08/2022]
Abstract
Genome-wide association studies (GWASs) have identified multiple loci associated with cross-sectional eGFR, but a systematic genetic analysis of kidney function decline over time is missing. Here we conducted a GWAS meta-analysis among 63,558 participants of European descent, initially from 16 cohorts with serial kidney function measurements within the CKDGen Consortium, followed by independent replication among additional participants from 13 cohorts. In stage 1 GWAS meta-analysis, single-nucleotide polymorphisms (SNPs) at MEOX2, GALNT11, IL1RAP, NPPA, HPCAL1, and CDH23 showed the strongest associations for at least one trait, in addition to the known UMOD locus, which showed genome-wide significance with an annual change in eGFR. In stage 2 meta-analysis, the significant association at UMOD was replicated. Associations at GALNT11 with Rapid Decline (annual eGFR decline of 3 ml/min per 1.73 m(2) or more), and CDH23 with eGFR change among those with CKD showed significant suggestive evidence of replication. Combined stage 1 and 2 meta-analyses showed significance for UMOD, GALNT11, and CDH23. Morpholino knockdowns of galnt11 and cdh23 in zebrafish embryos each had signs of severe edema 72 h after gentamicin treatment compared with controls, but no gross morphological renal abnormalities before gentamicin administration. Thus, our results suggest a role in the deterioration of kidney function for the loci GALNT11 and CDH23, and show that the UMOD locus is significantly associated with kidney function decline.
Collapse
Affiliation(s)
- Mathias Gorski
- 1] Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany [2] Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Adrienne Tin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Maija Garnaas
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gearoid M McMahon
- 1] Division of Nephrology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA [2] NHLBI's Framingham Heart Study, National Heart, Lung and Blood Institute, Framingham, Massachusetts, USA
| | - Audrey Y Chu
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Bamidele O Tayo
- Department of Public Health Services, Loyola Medical Center, Loyola University Chicago, Maywood, Illinois, USA
| | - Cristian Pattaro
- Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC), affiliated to the University of Lübeck, Bolzano, Italy
| | - Alexander Teumer
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - John Chalmers
- George Institute for Global Health, University of Sydney, Sydney, New South Wales, Australia
| | - Pavel Hamet
- Centre de recherche du Centre hospitalier de l'Université de Montréal, University of Montreal, Montreal, Quebec, Canada
| | - Johanne Tremblay
- CHUM Research Center- Technopôle Angus, Montreal, Québec, Canada
| | - Marc Woodward
- George Institute for Global Health, University of Sydney, Sydney, New South Wales, Australia
| | - Thor Aspelund
- 1] Icelandic Heart Association, Research Institute, Kopavogur, Iceland [2] University of Iceland, Reykjavik, Iceland
| | | | - Vilmundur Gudnason
- 1] Icelandic Heart Association, Research Institute, Kopavogur, Iceland [2] University of Iceland, Reykjavik, Iceland
| | - Tamara B Harris
- Intramural Research Program, Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, Bethesda, Maryland, USA
| | - Lenore J Launer
- Intramural Research Program, Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, Bethesda, Maryland, USA
| | - Albert V Smith
- 1] Icelandic Heart Association, Research Institute, Kopavogur, Iceland [2] University of Iceland, Reykjavik, Iceland
| | - Braxton D Mitchell
- 1] Department of Medicine and Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA [2] Geriatric Research and Education Clinical Center, Veterans Administration Medical Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jeffrey R O'Connell
- Department of Medicine and Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Alan R Shuldiner
- 1] Department of Medicine and Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA [2] Geriatric Research and Education Clinical Center, Veterans Administration Medical Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Josef Coresh
- 1] Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA [2] Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, Maryland, USA
| | - Man Li
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Paul Freudenberger
- Institute of Molecular Biology and Biochemistry, Medical University Graz, Graz, Austria
| | - Edith Hofer
- Department of Neurology, Institute for Medical Informatics, Statistics and Documentation, Medical University Graz, Graz, Austria
| | - Helena Schmidt
- Institute of Molecular Biology and Biochemistry, Medical University Graz, Graz, Austria
| | | | - Elizabeth G Holliday
- Centre for Clinical Epidemiology and Biostatistics, University of Newcastle, CReDITSS, HMRI, Callaghan, New South Wales, Australia
| | - Paul Mitchell
- Centre for Vision Research, Westmead Millennium Institute, University of Sydney, Westmead Hospital, Sydney, New South Wales, Australia
| | - Jie Jin Wang
- Centre for Vision Research, Westmead Millennium Institute, University of Sydney, Westmead Hospital, Sydney, New South Wales, Australia
| | | | - Guo Li
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington, USA
| | - David S Siscovick
- 1] Cardiovascular Health Research Unit, University of Washington, Seattle, Washington, USA [2] New York Academy of Medicine, New York, New York, USA
| | - Zoltan Kutalik
- 1] Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland [2] Department of Medical Genetics, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Tanguy Corre
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
| | - Peter Vollenweider
- Internal Medicine Department, Lausanne University Hospital, Lausanne, Switzerland
| | - Gérard Waeber
- Internal Medicine Department, Lausanne University Hospital, Lausanne, Switzerland
| | - Jayanta Gupta
- Perelman School of Medicine at the University of Pennsylvania, Center for Clinical Epidemiology and Biostatistics
| | - Peter A Kanetsky
- Perelman School of Medicine at the University of Pennsylvania, Center for Clinical Epidemiology and Biostatistics
| | - Shih-Jen Hwang
- NHLBI's Framingham Heart Study, National Heart, Lung and Blood Institute, Framingham, Massachusetts, USA
| | - Matthias Olden
- 1] Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany [2] NHLBI's Framingham Heart Study, National Heart, Lung and Blood Institute, Framingham, Massachusetts, USA
| | - Qiong Yang
- 1] NHLBI's Framingham Heart Study, National Heart, Lung and Blood Institute, Framingham, Massachusetts, USA [2] Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | | | | | | | | | - Jeanette M Stafford
- Department of Biostatistical Sciences, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Jingzhong Ding
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Yongmei Liu
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | | | - Daniele Cusi
- 1] Department of Health Science, University of Milano, Milano, Italy [2] Division of Nephrology, San Paolo Hospital, Milano, Italy
| | - Erika Salvi
- Department of Health Science, University of Milano, Milano, Italy
| | - Jan A Staessen
- 1] Department of Epidemiology, Maastricht University, Maastricht, The Netherlands [2] Studies Coordinating Centre, Division of Hypertension and Cardiovascular Rehabilitation, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Harald Grallert
- 1] Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany [2] Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany [3] German Center for Diabetes Research, Neuherberg, Germany
| | - Christa Meisinger
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Martina Müller-Nurasyid
- 1] DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany [2] Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany [3] Institute of Medical Informatics, Biometry, and Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany [4] Department of Medicine I, University Hospital Grosshadern, Ludwig-Maximilians-Universität, Munich, Germany
| | - Bernhard K Krämer
- University Medical Centre Mannheim, 5th Department of Medicine, University of Heidelberg, Mannheim, Germany
| | - Holly Kramer
- Department of Public Health Services, Loyola Medical Center, Loyola University Chicago, Maywood, Illinois, USA
| | - Sylvia E Rosas
- Joslin Diabetes Center and Beth Israel Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ilja M Nolte
- 1] Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands [2] Unit of Genetic Epidemiology and Bioinformatics, Department of Epidemiology (FA40), University Medical Center Groningen, Groningen, The Netherlands
| | - Brenda W Penninx
- 1] Department of Psychiatry/EMGO Institute/Neuroscience Campus, VU University Medical Centre, Amsterdam, The Netherlands [2] EMGO Institute Vumc, NESDA, Amsterdam, The Netherlands
| | - Harold Snieder
- 1] Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands [2] Unit of Genetic Epidemiology and Bioinformatics, Department of Epidemiology (FA40), University Medical Center Groningen, Groningen, The Netherlands
| | - M Fabiola Del Greco
- Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC), affiliated to the University of Lübeck, Bolzano, Italy
| | - Andre Franke
- Institute of Clinical Molecular Biology, Kiel, Germany
| | - Ute Nöthlings
- 1] Popgen Biobank, University Hospital Schleswig-Holstein, Kiel, Germany [2] Section for Epidemiology, Institute for Experimental Medicine, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Wolfgang Lieb
- Institute of Epidemiology and Biobank popgen, Christian-Albrechts University, Kiel, Germany
| | - Stephan J L Bakker
- University Medical Center Groningen, Department of Nephrology, University of Groningen, Groningen, The Netherlands
| | - Ron T Gansevoort
- University Medical Center Groningen, Department of Nephrology, University of Groningen, Groningen, The Netherlands
| | - Pim van der Harst
- University Medical Center Groningen, Department of Cardiology, University of Groningen, Groningen, The Netherlands
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Oscar H Franco
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Sanaz Sedaghat
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Stefan Coassin
- Division of Genetic Epidemiology, Innsbruck Medical University, Innsbruck, Austria
| | - Margot Haun
- Division of Genetic Epidemiology, Innsbruck Medical University, Innsbruck, Austria
| | - Barbara Kollerits
- Division of Genetic Epidemiology, Innsbruck Medical University, Innsbruck, Austria
| | - Florian Kronenberg
- Division of Genetic Epidemiology, Innsbruck Medical University, Innsbruck, Austria
| | - Bernhard Paulweber
- First Department of Internal Medicine, Paracelsus Private Medical University Salzburg, Salzburg, Austria
| | - Nicole Aumann
- Department SHIP/KEF, Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Karlhans Endlich
- Institute of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Mike Pietzner
- Institute for Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Rainer Rettig
- Institute of Physiology, University of Greifswald, Greifswald-Karlsburg, Germany
| | - Vincent Chouraki
- Inserm, U744, Institut Pasteur de Lille, Université Lille-Nord de France, CHR&U de Lille, Service d'épidémiologie régional, CHRU, Lille, France
| | - Catherine Helmer
- Inserm, U897, Université Bordeaux 2, ISPED, ISPED, Université Bordeaux 2, Bordeaux, France
| | - Jean-Charles Lambert
- Inserm, U744, Institut Pasteur de Lille, Université Lille-Nord de France, Institut Pasteur, Lille, France
| | - Marie Metzger
- Inserm, U1018, University Paris-Sud, CESP Team 10, Villejuif, France
| | - Benedicte Stengel
- Inserm, U1018, University Paris-Sud, CESP Team 10, Villejuif, France
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | | | - Olli Raitakari
- 1] Department of Clinical Physiology, Turku University Hospital, Turku, Finland [2] Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Andrew Johnson
- NHLBI Cardiovascular Epidemiology and Human Genomics Branch, Framingham Heart Study, National Heart, Lung and Blood Institute, Framingham, Massachusetts, USA
| | - Afshin Parsa
- Department of Medicine and Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Murielle Bochud
- Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital, Epalinges, Switzerland
| | - Iris M Heid
- 1] Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany [2] Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Wolfram Goessling
- 1] Divisions of Genetics and Gastroenterology, Department of Medicine, Brigham and Women's Hospital, and Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA [2] Harvard Stem Cell Institute, Harvard University and Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Anna Köttgen
- 1] Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA [2] Renal Division, Freiburg University Clinic, Germany, Freiburg, Germany
| | - W H Linda Kao
- 1] Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA [2] Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, Maryland, USA
| | - Caroline S Fox
- 1] NHLBI's Framingham Heart Study, National Heart, Lung and Blood Institute, Framingham, Massachusetts, USA [2] Department of Endocrinology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Carsten A Böger
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
124
|
Homoarginine in the renal and cardiovascular systems. Amino Acids 2015; 47:1703-13. [DOI: 10.1007/s00726-015-1993-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/18/2015] [Indexed: 10/23/2022]
|
125
|
Boor P, Floege J. Renal allograft fibrosis: biology and therapeutic targets. Am J Transplant 2015; 15:863-86. [PMID: 25691290 DOI: 10.1111/ajt.13180] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 11/30/2014] [Accepted: 12/19/2014] [Indexed: 01/25/2023]
Abstract
Renal tubulointerstitial fibrosis is the final common pathway of progressive renal diseases. In allografts, it is assessed with tubular atrophy as interstitial fibrosis/tubular atrophy (IF/TA). IF/TA occurs in about 40% of kidney allografts at 3-6 months after transplantation, increasing to 65% at 2 years. The origin of renal fibrosis in the allograft is complex and includes donor-related factors, in particular in case of expanded criteria donors, ischemia-reperfusion injury, immune-mediated damage, recurrence of underlying diseases, hypertensive damage, nephrotoxicity of immunosuppressants, recurrent graft infections, postrenal obstruction, etc. Based largely on studies in the non-transplant setting, there is a large body of literature on the role of different cell types, be it intrinsic to the kidney or bone marrow derived, in mediating renal fibrosis, and the number of mediator systems contributing to fibrotic changes is growing steadily. Here we review the most important cellular processes and mediators involved in the progress of renal fibrosis, with a focus on the allograft situation, and discuss some of the challenges in translating experimental insights into clinical trials, in particular fibrosis biomarkers or imaging modalities.
Collapse
Affiliation(s)
- P Boor
- Division of Nephrology and Clinical Immunology, RWTH University of Aachen, Aachen, Germany; Department of Pathology, RWTH University of Aachen, Aachen, Germany; Institute of Molecular Biomedicine, Bratislava, Slovakia
| | | |
Collapse
|
126
|
Yamada Y, Matsui K, Takeuchi I, Fujimaki T. Association of genetic variants with dyslipidemia and chronic kidney disease in a longitudinal population-based genetic epidemiological study. Int J Mol Med 2015; 35:1290-300. [PMID: 25813695 PMCID: PMC4380205 DOI: 10.3892/ijmm.2015.2152] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 03/20/2015] [Indexed: 12/21/2022] Open
Abstract
We previously identified 9 genes and chromosomal region 3q28 as susceptibility loci for myocardial infarction, ischemic stroke, or chronic kidney disease (CKD) in Japanese individuals by genome-wide or candidate gene association studies. In the present study, we examined the association of 13 polymorphisms at these 10 loci with the prevalence of hypertriglyceridemia, hyper-low-density lipoprotein (LDL) cholesterolemia, hypo-high-density lipoprotein (HDL) cholesterolemia, or CKD in community-dwelling Japanese individuals. The study subjects comprised 6,027 individuals who were recruited to the Inabe Health and Longevity Study, a longitudinal genetic epidemiological study of atherosclerotic, cardiovascular and metabolic diseases. The subjects were recruited from individuals who visited the Health Care Center at Inabe General Hospital for an annual health checkup, and they were followed up each year (mean follow‑up period, 5 years). Longitudinal analysis with a generalized estimating equation and with adjustment for covariates revealed that rs6929846 of butyrophilin, subfamily 2, member A1 gene (BTN2A1) was significantly associated with the prevalence of hypertriglyceridemia (P=0.0001), hyper-LDL cholesterolemia (P=0.0004), and CKD (P=0.0007); rs2569512 of interleukin enhancer binding factor 3 (ILF3) was associated with hyper-LDL cholesterolemia (P=0.0029); and rs2074379 (P=0.0019) and rs2074388 (P=0.0029) of alpha-kinase 1 (ALPK1) were associated with CKD. Longitudinal analysis with a generalized linear mixed-effect model and with adjustment for covariates among all individuals revealed that rs6929846 of BTN2A1 was significantly associated with the serum concentrations of triglycerides (P=0.0011), LDL cholesterol (P=3.3 x 10(-5)), and creatinine (P=0.0006), as well as with the estimated glomerular filtration rate (eGFR) (P=0.0004); rs2569512 of ILF3 was shown to be associated with the serum concentration of LDL cholesterol (P=0.0221); and rs2074379 (P=0.0302) and rs2074388 (P=0.0336) of ALPK1 were shown to be associated with the serum concentration of creatinine. Similar analysis among individuals not taking any anti‑dyslipidemic medication revealed that rs6929846 of BTN2A1 was significantly associated with the serum concentrations of triglycerides (P=8.3 x 10‑5) and LDL cholesterol (P=0.0004), and that rs2569512 of ILF3 was associated with the serum concentration of LDL cholesterol (P=0.0010). BTN2A1 may thus be a susceptibility gene for hypertriglyceridemia, hyper‑LDL cholesterolemia and CKD in Japanese individuals.
Collapse
Affiliation(s)
- Yoshiji Yamada
- Department of Human Functional Genomics, Life Science Research Center, Mie University, Tsu, Mie 514‑8507, Japan
| | - Kota Matsui
- Core Research for Evolutionary Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102‑0076, Japan
| | - Ichiro Takeuchi
- Core Research for Evolutionary Science and Technology (CREST), Japan Science and Technology Agency, Tokyo 102‑0076, Japan
| | - Tetsuo Fujimaki
- Department of Cardiovascular Medicine, Inabe General Hospital, Inabe, Mie 511‑0428, Japan
| |
Collapse
|
127
|
Song YM, Sung J, Lee K. Longitudinal relationships of metabolic syndrome and obesity with kidney function: Healthy Twin Study. Clin Exp Nephrol 2015; 19:887-94. [PMID: 25634252 DOI: 10.1007/s10157-015-1083-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 01/13/2015] [Indexed: 01/22/2023]
Abstract
BACKGROUND To examine cross-sectional and longitudinal relationships, including genetic and environmental correlations, of metabolic syndrome (MetS) and obesity with kidney function. METHODS Subjects were 3,437 Korean adults of the Healthy Twin Study for cross-sectional relationships and 1,881 participants for longitudinal relationships (follow-up interval 3.7 ± 1.4 years). Obesity (body mass index ≥ 25 vs. <25 kg/m(2)), MetS, and chronic kidney disease [CKD; estimated glomerular filtration rate (eGFR) (<60 mL/min/1.73 m(2) using the modification of diet in renal disease study equation)] were categorized at baseline and follow-up. RESULTS The prevalence and incidence of chronic kidney disease were 2.5 and 3.3 %, respectively. Compared to individuals without obesity and MetS, prevalent CKD was associated with MetS regardless of weight status [adjusted odds ratio (AOR) 4.19 for those with MetS but without obesity; AOR 4.63 for those with MetS and obesity]. Incident CKD was associated with obesity regardless of baseline metabolic status (AOR 2.03 for those with obesity but without MetS; AOR 2.85 for those with obesity and MetS). MetS at follow-up was associated with incident CKD regardless of baseline MetS (AOR 2.42-2.52). Sex-adjusted bivariate analyses show inverse environmental correlations of the number of MetS components and BMI at baseline, with eGFR at baseline and follow-up (ρ E, -0.26 to -0.42, P < 0.001). CONCLUSIONS MetS predicts prevalent CKD regardless of obesity, and obesity predicts incident CKD regardless of baseline MetS. Incident CKD is also associated with MetS at follow-up regardless of baseline MetS. These associations appear to be explained by shared environmental factors.
Collapse
Affiliation(s)
- Yun-Mi Song
- Department of Family Medicine, Samsung Medical Center, Sungkyunkwan University, School of Medicine, Seoul, South Korea
| | - Joohon Sung
- Department of Epidemiology, School of Public Health, Seoul National University, Seoul, South Korea.,Institute of Health and Environment, Seoul National University, Seoul, South Korea
| | - Kayoung Lee
- Department of Family Medicine, Busan Paik Hospital, Inje University College of Medicine, 633-165 Gaegum-dong, Busan Jin-Gu, Busan, 614-735, South Korea.
| |
Collapse
|
128
|
Genome-wide association study reveals a polymorphism in the podocyte receptor RANK for the decline of renal function in coronary patients. PLoS One 2014; 9:e114240. [PMID: 25478860 PMCID: PMC4257683 DOI: 10.1371/journal.pone.0114240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 11/04/2014] [Indexed: 11/19/2022] Open
Abstract
Impaired kidney function is a significant health problem and a major concern in clinical routine and is routinely determined by decreased glomerular filtration rate (GFR). In contrast to single assessment of a patients' kidney function providing only limited information on patients' health, serial measurements of GFR clearly improves the validity of diagnosis. The decline of kidney function has recently been reported to be predictive for mortality and vascular events in coronary patients. However, it has not been investigated for genetic association in GWA studies. This study investigates for the first time the association of cardiometabolic polymorphisms with the decline of estimated GFR during a 4 year follow up in 583 coronary patients, using the Cardio-Metabo Chip. We revealed a suggestive association with 3 polymorphisms, surpassing genome-wide significance (p = 4.0 e-7). The top hit rs17069906 (p = 5.6 e-10) is located within the genomic region of RANK, recently demonstrated to be an important player in the adaptive recovery response in podocytes and suggested as a promising therapeutic target in glomerular diseases.
Collapse
|
129
|
Bergen AW, Javitz HS, Krasnow R, Michel M, Nishita D, Conti DV, Edlund CK, Kwok PY, McClure JB, Kim RB, Hall SM, Tyndale RF, Baker TB, Benowitz NL, Swan GE. Organic cation transporter variation and response to smoking cessation therapies. Nicotine Tob Res 2014; 16:1638-46. [PMID: 25143296 PMCID: PMC4296186 DOI: 10.1093/ntr/ntu161] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 08/09/2014] [Indexed: 12/20/2022]
Abstract
INTRODUCTION We evaluated chr6q25.3 organic cation transporter gene (SLC22A1, SLC22A2, SLC22A3) variation and response to smoking cessation therapies. The corresponding proteins are low-affinity transporters of choline, acetylcholine and monoamines, and smoking cessation pharmacotherapies expressed in multiple tissues. METHODS We selected 7 common polymorphisms for mega-regression analysis. We assessed additive model association of polymorphisms with 7-day point prevalence abstinence overall and by assigned pharmacotherapy at end of treatment and at 6 months among European-ancestry participants of 7 randomized controlled trials adjusted for demographic, population genetic, and trial covariates. RESULTS Initial results were obtained in 6 trials with 1,839 participants. Nominally statistically significant associations of 2 SLC22A2 polymorphisms were observed: (1) with rs316019 at 6 months, overall ([c.808T>G; p.Ser270Ala], OR = 1.306, 95% CI = 1.034-1.649, p = .025), and among those randomized to nicotine replacement therapy (NRT) (OR = 1.784, 95% CI = 1.072-2.970, p = .026); and (2) with rs316006 (c.1502-529A>T) among those randomized to varenicline (OR = 1.420, 95% CI = 1.038-1.944, p = .028, OR = 1.362, 95% CI = 1.001-1.853, p = .04) at end of treatment and 6 months. Individuals randomized to NRT from a seventh trial were genotyped for rs316019; rs316019 was associated with a nominally statistically significant effect on abstinence overall at 6 months among 2,233 participants (OR = 1.249, 95% CI = 1.007-1.550, p = .043). CONCLUSIONS The functional OCT2 Ser270Ala polymorphism is nominally statistically significantly associated with abstinence among European-ancestry treatment-seeking smokers after adjustments for pharmacotherapy, demographics, population genetics, and without adjustment for multiple testing of 7 SNPs. Replication of these preliminary findings in additional randomized controlled trials of smoking cessation therapies and from multiple continental populations would describe another pharmacogenetic role for SLC22A2/OCT2.
Collapse
Affiliation(s)
- Andrew W Bergen
- Center for Health Sciences, SRI International, Menlo Park, CA;
| | - Harold S Javitz
- Center for Health Sciences, SRI International, Menlo Park, CA
| | - Ruth Krasnow
- Center for Health Sciences, SRI International, Menlo Park, CA
| | - Martha Michel
- Center for Health Sciences, SRI International, Menlo Park, CA
| | - Denise Nishita
- Center for Health Sciences, SRI International, Menlo Park, CA
| | - David V Conti
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA
| | - Christopher K Edlund
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA; BioRealm, LLC, Monument, CO
| | - Pui-Yan Kwok
- Department of Dermatology, University of California, San Francisco, CA
| | | | - Richard B Kim
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Sharon M Hall
- Department of Psychiatry, University of California, San Francisco, CA
| | - Rachel F Tyndale
- Centre for Addiction and Mental Health, and Departments of Psychiatry, Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Timothy B Baker
- Center for Tobacco Research and Intervention, University of Wisconsin, Madison, WI
| | - Neal L Benowitz
- Departments of Medicine and Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA
| | - Gary E Swan
- Stanford Prevention Research Center, Stanford University School of Medicine, Palo Alto, CA
| |
Collapse
|
130
|
Genetic variants in nicotinic acetylcholine receptor genes jointly contribute to kidney function in American Indians: the Strong Heart Family Study. J Hypertens 2014; 32:1042-8; discussion 1049. [PMID: 24569419 DOI: 10.1097/hjh.0000000000000151] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Cigarette smoking negatively affects kidney function. Genetic variants in the nicotinic acetylcholine receptor (nAChR) genes have been associated with nicotine dependence, and are likely to influence renal function and related traits. Whereas each single variant may only exert a small effect, the joint contribution of multiple variants to the risk of disease could be substantial. METHODS Using a gene-family approach, we investigated the joint association of 61 tagging SNPs in seven genes encoding the nAChRs with kidney function in 3620 American Indians participating in the Strong Heart Family Study, independent of known risk factors. Kidney function was evaluated by estimated glomerular filtration rate, urinary albumin/creatinine ratio, albuminuria and chronic kidney disease. The joint impact of smoking-related variants was assessed using the weighted truncated product method. RESULTS Multiple SNPs showed marginal individual effect on renal function variability, and only a few survive multiple comparison correction. In contrast, a gene-family analysis considering the joint impact of all 61 SNPs reveals significant associations of the nAChR gene family with kidney function variables including estimated glomerular filtration rate, urinary albumin/creatinine ratio, and albuminuria (all Ps ≤ 0.0001) after adjusting for established risk factors including cigarette smoking. CONCLUSION Genetic variants in nAChR genes jointly contribute to renal function or kidney damage in American Indians. The effects of these genetic variants on kidney function or damage are independent of traditional risk factors including cigarette smoking per se.
Collapse
|
131
|
Smyth LJ, Duffy S, Maxwell AP, McKnight AJ. Genetic and epigenetic factors influencing chronic kidney disease. Am J Physiol Renal Physiol 2014; 307:F757-76. [PMID: 25080522 DOI: 10.1152/ajprenal.00306.2014] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Chronic kidney disease (CKD) has become a serious public health problem because of its associated morbidity, premature mortality, and attendant healthcare costs. The rising number of persons with CKD is linked with the aging population structure and an increased prevalence of diabetes, hypertension, and obesity. There is an inherited risk associated with developing CKD, as evidenced by familial clustering and differing prevalence rates across ethnic groups. Previous studies to determine the inherited risk factors for CKD rarely identified genetic variants that were robustly replicated. However, improvements in genotyping technologies and analytic methods are now helping to identify promising genetic loci aided by international collaboration and multiconsortia efforts. More recently, epigenetic modifications have been proposed to play a role in both the inherited susceptibility to CKD and, importantly, to explain how the environment dynamically interacts with the genome to alter an individual's disease risk. Genome-wide, epigenome-wide, and whole transcriptome studies have been performed, and optimal approaches for integrative analysis are being developed. This review summarizes recent research and the current status of genetic and epigenetic risk factors influencing CKD using population-based information.
Collapse
Affiliation(s)
- L J Smyth
- Nephrology Research, Centre for Public Health, Queen's University of Belfast, Belfast, Northern Ireland
| | - S Duffy
- Nephrology Research, Centre for Public Health, Queen's University of Belfast, Belfast, Northern Ireland
| | - A P Maxwell
- Nephrology Research, Centre for Public Health, Queen's University of Belfast, Belfast, Northern Ireland
| | - A J McKnight
- Nephrology Research, Centre for Public Health, Queen's University of Belfast, Belfast, Northern Ireland
| |
Collapse
|
132
|
Ledo N, Ko YA, Park ASD, Kang HM, Han SY, Choi P, Susztak K. Functional genomic annotation of genetic risk loci highlights inflammation and epithelial biology networks in CKD. J Am Soc Nephrol 2014; 26:692-714. [PMID: 25231882 DOI: 10.1681/asn.2014010028] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Genome-wide association studies (GWASs) have identified multiple loci associated with the risk of CKD. Almost all risk variants are localized to the noncoding region of the genome; therefore, the role of these variants in CKD development is largely unknown. We hypothesized that polymorphisms alter transcription factor binding, thereby influencing the expression of nearby genes. Here, we examined the regulation of transcripts in the vicinity of CKD-associated polymorphisms in control and diseased human kidney samples and used systems biology approaches to identify potentially causal genes for prioritization. We interrogated the expression and regulation of 226 transcripts in the vicinity of 44 single nucleotide polymorphisms using RNA sequencing and gene expression arrays from 95 microdissected control and diseased tubule samples and 51 glomerular samples. Gene expression analysis from 41 tubule samples served for external validation. 92 transcripts in the tubule compartment and 34 transcripts in glomeruli showed statistically significant correlation with eGFR. Many novel genes, including ACSM2A/2B, FAM47E, and PLXDC1, were identified. We observed that the expression of multiple genes in the vicinity of any single CKD risk allele correlated with renal function, potentially indicating that genetic variants influence multiple transcripts. Network analysis of GFR-correlating transcripts highlighted two major clusters; a positive correlation with epithelial and vascular functions and an inverse correlation with inflammatory gene cluster. In summary, our functional genomics analysis highlighted novel genes and critical pathways associated with kidney function for future analysis.
Collapse
Affiliation(s)
- Nora Ledo
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yi-An Ko
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ae-Seo Deok Park
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hyun-Mi Kang
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sang-Youb Han
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Peter Choi
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
133
|
Scolari F, Izzi C, Ghiggeri GM. Uromodulin: from monogenic to multifactorial diseases: FIGURE 1:. Nephrol Dial Transplant 2014; 30:1250-6. [DOI: 10.1093/ndt/gfu300] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 08/21/2014] [Indexed: 12/30/2022] Open
|
134
|
Creatinine, eGFR and association with myocardial infarction, ischemic heart disease and early death in the general population. Atherosclerosis 2014; 237:67-75. [PMID: 25222342 DOI: 10.1016/j.atherosclerosis.2014.08.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 08/15/2014] [Accepted: 08/16/2014] [Indexed: 11/22/2022]
Abstract
OBJECTIVE We tested the hypothesis that moderately elevated plasma creatinine levels and decreased levels of estimated glomerular filtration rate (eGFR) are associated with increased risk of myocardial infarction, ischemic heart disease, and early death in the general population. METHODS We studied 10,489 individuals with a plasma creatinine measurement and calculated eGFR from the Danish general population, of which 1498 developed myocardial infarction, 3001 ischemic heart disease, and 7573 died during 32 years follow-up. RESULTS Cumulative incidences of myocardial infarction and ischemic heart disease as a function of age increased with increasing levels of creatinine, and survival decreased (log-rank trends: <0.001). The median survival age was 78.7 (95%CI: 78.0-79.2) years for persons with creatinine levels <90th percentile, 78.1 (76.3-79.5) years for 90th-94th percentiles, and 74.8 (72.8-76.7) years for ≥95th percentile. Hazard ratios for myocardial infarction and plasma creatinine levels of 90th-94th percentiles and ≥95th percentile versus <50th percentile were 2.06 (95%CI: 1.67-2.56) and 1.90 (1.56-2.31) adjusted for gender and age, and 1.35 (1.09-1.68) and 1.11 (0.90-1.36) adjusted multifactorially, respectively. Corresponding estimates for creatinine and ischemic heart disease were 1.57 (1.33-1.85) and 1.64 (1.42-1.89) adjusted for gender and age, and 1.16 (0.98-1.37) and 1.11 (0.95-1.29) adjusted multifactorially. Finally, corresponding values for early death were 1.18 (1.06-1.32) and 1.43 (1.30-1.57), and 0.97 (0.87-1.09) and 1.13 (1.02-1.24), respectively. Low eGFR did not associate consistently with increased risk of these endpoints. CONCLUSION In the general population, moderately elevated plasma creatinine was associated with increased risk of myocardial infarction, ischemic heart disease, and early death, while low eGFR was not.
Collapse
|
135
|
Dinic M, Ghisdal L, Racapé J, Thibaudin L, Gatault P, Essig M, Le Meur Y, Noël C, Touchard G, Merville P, Ajarchouh Z, Mariat C, Abramowicz M, Abramowicz D, Alamartine E. UMOD polymorphism rs12917707 is not associated with severe or stable IgA nephropathy in a large Caucasian cohort. BMC Nephrol 2014; 15:138. [PMID: 25163389 PMCID: PMC4236674 DOI: 10.1186/1471-2369-15-138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 05/22/2014] [Indexed: 01/12/2023] Open
Abstract
Background Genetic factors are suspected in the pathogenesis of IgA nephropathy, as well as in the course of IgA nephropathy progression towards end stage renal failure. UMOD polymorphism rs12917707 is known to associate with end stage renal failure of mixed aetiologies. Methods We tested a large cohort of Caucasian patients for association of rs12917707 with IgA nephropathy showing a benign, stable course and with IgA nephropathy that progressed toward end stage renal failure. Results No association was observed between either groups, and a non-significant trend was observed for more severe IgA nephropathy with the allele reported to protect against end stage renal failure of mixed aetiologies. Conclusion We conclude that UMOD is unlikely to play a role in IgA nephropathy pathogenesis nor progression to end stage renal failure, and suggest that UMOD effects are restricted to some causes of renal disease, e.g. diabetes or hypertension.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Eric Alamartine
- Nephrology-Renal Transplantation Department, CHU de Saint Etienne & EA3064, GIMAP, Université Jean Monnet, Saint Etienne 42055 Cedex 02, France.
| |
Collapse
|
136
|
Sveinbjornsson G, Mikaelsdottir E, Palsson R, Indridason OS, Holm H, Jonasdottir A, Helgason A, Sigurdsson S, Jonasdottir A, Sigurdsson A, Eyjolfsson GI, Sigurdardottir O, Magnusson OT, Kong A, Masson G, Sulem P, Olafsson I, Thorsteinsdottir U, Gudbjartsson DF, Stefansson K. Rare mutations associating with serum creatinine and chronic kidney disease. Hum Mol Genet 2014; 23:6935-43. [PMID: 25082825 DOI: 10.1093/hmg/ddu399] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chronic kidney disease (CKD) is a complex disorder with a strong genetic component. A number of common sequence variants have been found to associate with serum creatinine (SCr), estimated glomerular filtration rate (eGFR) and/or CKD. We imputed 24 million single-nucleotide polymorphisms and insertions/deletions identified by whole-genome sequencing of 2230 Icelanders into 81 656 chip-typed individuals and 112 630 relatives of genotyped individuals over the age of 18 with SCr measurements. The large set of sequenced individuals allowed accurate imputation of variants to a minor allele frequency (MAF) of 0.1%. We tested the imputed variants for association with SCr. In addition to replicating established loci, we discovered missense and loss-of-function variants associating with SCr in three solute carriers (SLC6A19, SLC25A45 and SLC47A1) and two E3 ubiquitin ligases (RNF186 and RNF128). All the variants are within coding sequences and all but one are rare (MAF <2%) with SCr effects between 0.085 and 0.129 standard deviations. These rare variants have a larger effect on SCr than previously reported common variants, explaining 0.5% of the variability of SCr in Icelanders in addition to the 1% already accounted for. We tested the five variants associating with SCr for association with CKD in an Icelandic sample of 15 594 cases and 291 428 controls. Three of the variants also associated with CKD. These variants may either affect kidney function or creatinine synthesis and excretion. Of note were four mutations in SLC6A19 that associate with reduced SCr, three of which have been shown to cause Hartnup disease.
Collapse
Affiliation(s)
| | | | | | | | - Hilma Holm
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | | | - Agnar Helgason
- deCODE Genetics, 101 Reykjavik, Iceland Department of Anthropology
| | | | | | | | | | - Olof Sigurdardottir
- Department of Clinical Biochemistry, Akureyri Hospital, 600 Akureyri, Iceland
| | | | - Augustine Kong
- deCODE Genetics, 101 Reykjavik, Iceland School of Engineering and Natural Sciences, University of Iceland, 101 Reykjavik, Iceland
| | | | | | - Isleifur Olafsson
- Department of Clinical Biochemistry, Landspitali - The National University Hospital of Iceland, 101 Reykjavík, Iceland
| | | | - Daniel F Gudbjartsson
- deCODE Genetics, 101 Reykjavik, Iceland School of Engineering and Natural Sciences, University of Iceland, 101 Reykjavik, Iceland
| | | |
Collapse
|
137
|
Fu J, Zhang H, Zhuang Y, Liu H, Shi Q, Li D, Ju X. The role of N-acetyltransferase 8 in mesenchymal stem cell-based therapy for liver ischemia/reperfusion injury in rats. PLoS One 2014; 9:e103355. [PMID: 25057902 PMCID: PMC4109999 DOI: 10.1371/journal.pone.0103355] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Accepted: 06/29/2014] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE To evaluate the impact of mesenchymal stem cells (MSCs) against hepatic I/R injury and explore the role of N-acetyltransferase 8 (NAT8) in the process. METHODS We investigated the potential of injected MSCs systemically via the tail vein in healing injuried liver of the SD rat model of 70% hepatic I/R injury by measuring the biochemical and pathologic alterations. Subsequently, we evaluated the expression levels of NAT8 by western blotting in vivo. Concurrently, hydrogen peroxide (H2O2)-induced apoptosis in the human normal liver cell line L02 was performed in vitro to evaluate the protective effects of MSC conditioned medium (MSC-CM) on L02 cells. In addition, we downregulated and upregulated NAT8 expression in L02 cells and induced apoptosis by using H2O2 to study the protective role of NAT8. RESULTS MSCs implantation led to a significant reduced liver enzyme levels, an advanced protection in the histopathological findings of the acutely injured liver and a significantly lower percentage of TUNEL-positive cells, which were increased after I/R injury. In vitro assays, MSC-CM inhibited hepatocyte apoptosis induced by H2O2. Moreover, overexpression or downregulation of NAT8 prevented or aggravated hepatocyte apoptosis induced by H2O2, respectively. CONCLUSIONS MSC transplantation provides support to the I/R-injured liver by inhibiting hepatocellular apoptosis and stimulating NAT8 regeneration.
Collapse
Affiliation(s)
- Jinqiu Fu
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Haiyan Zhang
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Yong Zhuang
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Huan Liu
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Qing Shi
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Dong Li
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, P.R. China
- * E-mail: (XJ); (DL)
| | - Xiuli Ju
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, P.R. China
- * E-mail: (XJ); (DL)
| |
Collapse
|
138
|
Jäger G, Peltzer A, Nieselt K. inPHAP: interactive visualization of genotype and phased haplotype data. BMC Bioinformatics 2014; 15:200. [PMID: 25002076 PMCID: PMC4083868 DOI: 10.1186/1471-2105-15-200] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 06/10/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To understand individual genomes it is necessary to look at the variations that lead to changes in phenotype and possibly to disease. However, genotype information alone is often not sufficient and additional knowledge regarding the phase of the variation is needed to make correct interpretations. Interactive visualizations, that allow the user to explore the data in various ways, can be of great assistance in the process of making well informed decisions. But, currently there is a lack for visualizations that are able to deal with phased haplotype data. RESULTS We present inPHAP, an interactive visualization tool for genotype and phased haplotype data. inPHAP features a variety of interaction possibilities such as zooming, sorting, filtering and aggregation of rows in order to explore patterns hidden in large genetic data sets. As a proof of concept, we apply inPHAP to the phased haplotype data set of Phase 1 of the 1000 Genomes Project. Thereby, inPHAP's ability to show genetic variations on the population as well as on the individuals level is demonstrated for several disease related loci. CONCLUSIONS As of today, inPHAP is the only visual analytical tool that allows the user to explore unphased and phased haplotype data interactively. Due to its highly scalable design, inPHAP can be applied to large datasets with up to 100 GB of data, enabling users to visualize even large scale input data. inPHAP closes the gap between common visualization tools for unphased genotype data and introduces several new features, such as the visualization of phased data. inPHAP is available for download at http://bit.ly/1iJgKmX.
Collapse
Affiliation(s)
- Günter Jäger
- Integrative Transcriptomics, Center for Bioinformatics, University of Tübingen, Sand 14, 72076 Tübingen, Germany
| | - Alexander Peltzer
- Integrative Transcriptomics, Center for Bioinformatics, University of Tübingen, Sand 14, 72076 Tübingen, Germany
| | - Kay Nieselt
- Integrative Transcriptomics, Center for Bioinformatics, University of Tübingen, Sand 14, 72076 Tübingen, Germany
| |
Collapse
|
139
|
Wing MR, Ramezani A, Gill HS, Devaney JM, Raj DS. Epigenetics of progression of chronic kidney disease: fact or fantasy? Semin Nephrol 2014; 33:363-74. [PMID: 24011578 DOI: 10.1016/j.semnephrol.2013.05.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Epigenetic modifications are important in the normal functioning of the cell, from regulating dynamic expression of essential genes and associated proteins to repressing those that are unneeded. Epigenetic changes are essential for development and functioning of the kidney, and aberrant methylation, histone modifications, and expression of microRNA could lead to chronic kidney disease (CKD). Here, epigenetic modifications modulate transforming growth factor β signaling, inflammation, profibrotic genes, and the epithelial-to-mesenchymal transition, promoting renal fibrosis and progression of CKD. Identification of these epigenetic changes is important because they are potentially reversible and may serve as therapeutic targets in the future to prevent subsequent renal fibrosis and CKD. In this review we discuss the different types of epigenetic control, methods to study epigenetic modifications, and how epigenetics promotes progression of CKD.
Collapse
Affiliation(s)
- Maria R Wing
- Division of Renal Disease and Hypertension, The George Washington University, Washington, DC
| | | | | | | | | |
Collapse
|
140
|
Mussap M, Noto A, Fanos V, Van Den Anker JN. Emerging biomarkers and metabolomics for assessing toxic nephropathy and acute kidney injury (AKI) in neonatology. BIOMED RESEARCH INTERNATIONAL 2014; 2014:602526. [PMID: 25013791 PMCID: PMC4071811 DOI: 10.1155/2014/602526] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 03/25/2014] [Indexed: 01/07/2023]
Abstract
Identification of novel drug-induced toxic nephropathy and acute kidney injury (AKI) biomarkers has been designated as a top priority by the American Society of Nephrology. Increasing knowledge in the science of biology and medicine is leading to the discovery of still more new biomarkers and of their roles in molecular pathways triggered by physiological and pathological conditions. Concomitantly, the development of the so-called "omics" allows the progressive clinical utilization of a multitude of information, from those related to the human genome (genomics) and proteome (proteomics), including the emerging epigenomics, to those related to metabolites (metabolomics). In preterm newborns, one of the most important factors causing the pathogenesis and the progression of AKI is the interaction between the individual genetic code, the environment, the gestational age, and the disease. By analyzing a small urine sample, metabolomics allows to identify instantly any change in phenotype, including changes due to genetic modifications. The role of liquid chromatography-mass spectrometry (LC-MS), proton nuclear magnetic resonance (1H NMR), and other emerging technologies is strategic, contributing basically to the sudden development of new biochemical and molecular tests. Urine neutrophil gelatinase-associated lipocalin (uNGAL) and kidney injury molecule-1 (KIM-1) are closely correlated with the severity of kidney injury, representing noninvasive sensitive surrogate biomarkers for diagnosing, monitoring, and quantifying kidney damage. To become routine tests, uNGAL and KIM-1 should be carefully tested in multicenter clinical trials and should be measured in biological fluids by robust, standardized analytical methods.
Collapse
Affiliation(s)
- M. Mussap
- Department of Laboratory Medicine, IRCCS San Martino-IST, University Hospital, National Institute for Cancer Research, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - A. Noto
- Department of Pediatrics and Clinical Medicine, Section of Neonatal Intensive Care Unit, Puericulture Institute and Neonatal Section, Azienda Mista and University of Cagliari, 09042 Cagliari, Italy
| | - V. Fanos
- Department of Pediatrics and Clinical Medicine, Section of Neonatal Intensive Care Unit, Puericulture Institute and Neonatal Section, Azienda Mista and University of Cagliari, 09042 Cagliari, Italy
| | - J. N. Van Den Anker
- Division of Pediatric Clinical Pharmacology, Children's National Medical Center, Washington, DC 20010, USA
| |
Collapse
|
141
|
Abstract
Unravelling the genotype–phenotype relationship in humans remains a challenging task in genomics studies. Recent advances in sequencing technologies mean there are now thousands of sequenced human genomes, revealing millions of single nucleotide variants (SNVs). For non-synonymous SNVs present in proteins the difficulties of the problem lie in first identifying those nsSNVs that result in a functional change in the protein among the many non-functional variants and in turn linking this functional change to phenotype. Here we present VarMod (Variant Modeller) a method that utilises both protein sequence and structural features to predict nsSNVs that alter protein function. VarMod develops recent observations that functional nsSNVs are enriched at protein–protein interfaces and protein–ligand binding sites and uses these characteristics to make predictions. In benchmarking on a set of nearly 3000 nsSNVs VarMod performance is comparable to an existing state of the art method. The VarMod web server provides extensive resources to investigate the sequence and structural features associated with the predictions including visualisation of protein models and complexes via an interactive JSmol molecular viewer. VarMod is available for use at http://www.wasslab.org/varmod.
Collapse
Affiliation(s)
- Morena Pappalardo
- Centre for Molecular Processing, School of Biosciences, University of Kent, CT2 7NH, UK
| | - Mark N Wass
- Centre for Molecular Processing, School of Biosciences, University of Kent, CT2 7NH, UK
| |
Collapse
|
142
|
Schramm K, Marzi C, Schurmann C, Carstensen M, Reinmaa E, Biffar R, Eckstein G, Gieger C, Grabe HJ, Homuth G, Kastenmüller G, Mägi R, Metspalu A, Mihailov E, Peters A, Petersmann A, Roden M, Strauch K, Suhre K, Teumer A, Völker U, Völzke H, Wang-Sattler R, Waldenberger M, Meitinger T, Illig T, Herder C, Grallert H, Prokisch H. Mapping the genetic architecture of gene regulation in whole blood. PLoS One 2014; 9:e93844. [PMID: 24740359 PMCID: PMC3989189 DOI: 10.1371/journal.pone.0093844] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 03/07/2014] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND We aimed to assess whether whole blood expression quantitative trait loci (eQTLs) with effects in cis and trans are robust and can be used to identify regulatory pathways affecting disease susceptibility. MATERIALS AND METHODS We performed whole-genome eQTL analyses in 890 participants of the KORA F4 study and in two independent replication samples (SHIP-TREND, N = 976 and EGCUT, N = 842) using linear regression models and Bonferroni correction. RESULTS In the KORA F4 study, 4,116 cis-eQTLs (defined as SNP-probe pairs where the SNP is located within a 500 kb window around the transcription unit) and 94 trans-eQTLs reached genome-wide significance and overall 91% (92% of cis-, 84% of trans-eQTLs) were confirmed in at least one of the two replication studies. Different study designs including distinct laboratory reagents (PAXgene™ vs. Tempus™ tubes) did not affect reproducibility (separate overall replication overlap: 78% and 82%). Immune response pathways were enriched in cis- and trans-eQTLs and significant cis-eQTLs were partly coexistent in other tissues (cross-tissue similarity 40-70%). Furthermore, four chromosomal regions displayed simultaneous impact on multiple gene expression levels in trans, and 746 eQTL-SNPs have been previously reported to have clinical relevance. We demonstrated cross-associations between eQTL-SNPs, gene expression levels in trans, and clinical phenotypes as well as a link between eQTLs and human metabolic traits via modification of gene regulation in cis. CONCLUSIONS Our data suggest that whole blood is a robust tissue for eQTL analysis and may be used both for biomarker studies and to enhance our understanding of molecular mechanisms underlying gene-disease associations.
Collapse
Affiliation(s)
- Katharina Schramm
- Institute of Human Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Human Genetics, Technical University Munich, München, Germany
| | - Carola Marzi
- Research Unit of Molecular Epidemiology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Claudia Schurmann
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Maren Carstensen
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), partner site Düsseldorf, Germany
| | - Eva Reinmaa
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Reiner Biffar
- Department of Prosthetic Dentistry, Gerostomatology and Dental Materials, University Medicine Greifswald, Greifswald, Germany
| | - Gertrud Eckstein
- Institute of Human Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Hans-Jörgen Grabe
- Department of Psychiatry and Psychotherapy, Helios Hospital Stralsund, University Medicine of Greifswald, Greifswald, Germany
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Gabriele Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Andres Metspalu
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Evelin Mihailov
- Estonian Genome Center, University of Tartu, Tartu, Estonia
- Estonian Biocentre, Tartu, Estonia
| | - Annette Peters
- Institute of Human Genetics, Technical University Munich, München, Germany
| | - Astrid Petersmann
- Institute of Clinical Chemistry and Laboratory Medicine, Greifswald, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), partner site Düsseldorf, Germany
- Division of Endocrinology and Diabetology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-Universität Munich, Neuherberg, Germany
| | - Karsten Suhre
- Institute of Bioinformatics and Systems Biology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Physiology and Biophysics, Weill Cornell Medical College in Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Alexander Teumer
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Rui Wang-Sattler
- Research Unit of Molecular Epidemiology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Human Genetics, Technical University Munich, München, Germany
- Munich Heart Alliance, München, Germany
| | - Thomas Illig
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), partner site Düsseldorf, Germany
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Holger Prokisch
- Institute of Human Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Human Genetics, Technical University Munich, München, Germany
- * E-mail:
| |
Collapse
|
143
|
Yu B, Zheng Y, Alexander D, Morrison AC, Coresh J, Boerwinkle E. Genetic determinants influencing human serum metabolome among African Americans. PLoS Genet 2014; 10:e1004212. [PMID: 24625756 PMCID: PMC3952826 DOI: 10.1371/journal.pgen.1004212] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 01/13/2014] [Indexed: 11/18/2022] Open
Abstract
Phenotypes proximal to gene action generally reflect larger genetic effect sizes than those that are distant. The human metabolome, a result of multiple cellular and biological processes, are functional intermediate phenotypes proximal to gene action. Here, we present a genome-wide association study of 308 untargeted metabolite levels among African Americans from the Atherosclerosis Risk in Communities (ARIC) Study. Nineteen significant common variant-metabolite associations were identified, including 13 novel loci (p<1.6×10−10). These loci were associated with 7–50% of the difference in metabolite levels per allele, and the variance explained ranged from 4% to 20%. Fourteen genes were identified within the nineteen loci, and four of them contained non-synonymous substitutions in four enzyme-encoding genes (KLKB1, SIAE, CPS1, and NAT8); the other significant loci consist of eight other enzyme-encoding genes (ACE, GATM, ACY3, ACSM2B, THEM4, ADH4, UGT1A, TREH), a transporter gene (SLC6A13) and a polycystin protein gene (PKD2L1). In addition, four potential disease-associated paths were identified, including two direct longitudinal predictive relationships: NAT8 with N-acetylornithine, N-acetyl-1-methylhistidine and incident chronic kidney disease, and TREH with trehalose and incident diabetes. These results highlight the value of using endophenotypes proximal to gene function to discover new insights into biology and disease pathology. Most contemporary GWAS studies have achieved increased power by increasing the size of the discovery sample to tens of thousands of individuals. An alternative approach for detecting the effects of novel loci is to measure phenotypes that more immediately reflect the effects of gene function. The metabolome consists of a collection of small molecules resulting from a variety of cellular and biologic processes, which can be considered intermediate phenotypes proximal to gene function. Here, we report a genome-wide association study identifying nineteen genetic loci influencing untargeted metabolomes traits among African Americans in the Atherosclerosis Risk in Communities (ARIC) Study. Fourteen genes mapped within nineteen loci, including twelve enzyme-encoding genes (KLKB1, SIAE, CPS1, NAT8, ACE, GATM, ACY3, ACSM2B, THEM4, ADH4, UGT1A and TREH), a transporter gene (SLC6A13) and a polycystin protein gene (PKD2L1). In addition, four potential disease-associated paths were identified, including two direct longitudinal predictive relationships: NAT8 with N-acetylornithine, N-acetyl-1-methylhistidine and incident chronic kidney disease, and TREH with trehalose and incident diabetes. These results highlight the value of using phenotypes proximal to gene function to promote novel gene discovery.
Collapse
Affiliation(s)
- Bing Yu
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Yan Zheng
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Danny Alexander
- Metabolon, Inc., Durham, North Carolina, United States of America
| | - Alanna C. Morrison
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
144
|
|
145
|
Fisel P, Renner O, Nies AT, Schwab M, Schaeffeler E. Solute carrier transporter and drug-related nephrotoxicity: the impact of proximal tubule cell models for preclinical research. Expert Opin Drug Metab Toxicol 2014; 10:395-408. [PMID: 24397389 DOI: 10.1517/17425255.2014.876990] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION The final excretion step of several drugs is facilitated by membrane transporters of the Solute carrier (SLC) family expressed in the proximal tubules of the kidney. Membrane transporters contribute substantially to the pharmacokinetic profile of drugs and play important roles in drug-induced nephrotoxicity. Different cell models have been applied as tools for the assessment of nephrotoxic effects caused by drugs. AREAS COVERED This review gives an overview over clinically relevant SLC transporters involved in the renal elimination of drug agents and their specific role in drug-induced nephrotoxicity. Most widely applied cell models are described and their advantages and limitations are outlined. EXPERT OPINION In vitro cell culture models (e.g., continuous and primary renal cell lines, polarized cell monolayers) represent valuable tools for early assessment of the nephrotoxic potential of drugs. Since SLC transporters contribute to drug excretion in a large part, in vitro cell culture models might be very helpful to study transport pathways and/or potential drug-drug interactions at an early stage of the drug development process to predict nephrotoxic effects.
Collapse
Affiliation(s)
- Pascale Fisel
- Margarete Fischer-Bosch-Institute of Clinical Pharmacology , Auerbachstrasse 125, Stuttgart, 70376 , Germany
| | | | | | | | | |
Collapse
|
146
|
Tomaschitz A, Meinitzer A, Pilz S, Rus-Machan J, Genser B, Drechsler C, Grammer T, Krane V, Ritz E, Kleber ME, Pieske B, Kraigher-Krainer E, Fahrleitner-Pammer A, Wanner C, Boehm BO, Marz W. Homoarginine, kidney function and cardiovascular mortality risk. Nephrol Dial Transplant 2014; 29:663-71. [DOI: 10.1093/ndt/gft512] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
147
|
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
|
148
|
Giacopo AD, Rubio-Aliaga I, Cantone A, Artunc F, Rexhepaj R, Frey-Wagner I, Font-Llitjós M, Gehring N, Stange G, Jaenecke I, Mohebbi N, Closs EI, Palacín M, Nunes V, Daniel H, Lang F, Capasso G, Wagner CA. Differential cystine and dibasic amino acid handling after loss of function of the amino acid transporter b0,+AT (Slc7a9) in mice. Am J Physiol Renal Physiol 2013; 305:F1645-55. [DOI: 10.1152/ajprenal.00221.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cystinuria is an autosomal recessive disease caused by mutations in SLC3A1 ( rBAT) and SLC7A9 ( b 0,+ AT). Gene targeting of the catalytic subunit ( Slc7a9) in mice leads to excessive excretion of cystine, lysine, arginine, and ornithine. Here, we studied this non-type I cystinuria mouse model using gene expression analysis, Western blotting, clearance, and brush-border membrane vesicle (BBMV) uptake experiments to further characterize the renal and intestinal consequences of losing Slc7a9 function. The electrogenic and BBMV flux studies in the intestine suggested that arginine and ornithine are transported via other routes apart from system b0,+. No remarkable gene expression changes were observed in other amino acid transporters and the peptide transporters in the intestine and kidney. Furthermore, the glomerular filtration rate (GFR) was reduced by 30% in knockout animals compared with wild-type animals. The fractional excretion of arginine was increased as expected (∼100%), but fractional excretions of lysine (∼35%), ornithine (∼16%), and cystine (∼11%) were less affected. Loss of function of b0,+AT reduced transport of cystine and arginine in renal BBMVs and completely abolished the exchanger activity of dibasic amino acids with neutral amino acids. In conclusion, loss of Slc7a9 function decreases the GFR and increases the excretion of several amino acids to a lesser extent than expected with no clear regulation at the mRNA and protein level of alternative transporters and no increased renal epithelial uptake. These observations indicate that transporters located in distal segments of the kidney and/or metabolic pathways may partially compensate for Slc7a9 loss of function.
Collapse
Affiliation(s)
- Andrea Di Giacopo
- Institute of Physiology-Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland
| | - Isabel Rubio-Aliaga
- Institute of Physiology-Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland
| | - Alessandra Cantone
- Department of Internal Medicine, Chair of Nephrology, Second University of Naples, Naples, Italy
| | - Ferruh Artunc
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Rexhep Rexhepaj
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | | | - Mariona Font-Llitjós
- Medical and Molecular Genetics Center, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Spain
- U730 CIBERER, Barcelona, Spain
| | - Nicole Gehring
- Institute of Physiology-Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland
| | - Gerti Stange
- Institute of Physiology-Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland
| | - Isabel Jaenecke
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Nilufar Mohebbi
- Institute of Physiology-Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland
| | - Ellen I. Closs
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Manuel Palacín
- IRB Barcelona, Department of Biochemistry and Molecular Biology, University of Barcelona and U731 CIBERER, Barcelona, Spain
| | - Virginia Nunes
- Medical and Molecular Genetics Center, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Spain
- Department of Physiological Sciences II, University of Barcelona, Spain; and
- U730 CIBERER, Barcelona, Spain
| | - Hannelore Daniel
- Molecular Nutrition Unit, Technical University of Munich, Freising, Germany
| | - Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Giovambattista Capasso
- Department of Internal Medicine, Chair of Nephrology, Second University of Naples, Naples, Italy
| | - Carsten A. Wagner
- Institute of Physiology-Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland
| |
Collapse
|
149
|
Freedman BI, Divers J, Palmer ND. Population ancestry and genetic risk for diabetes and kidney, cardiovascular, and bone disease: modifiable environmental factors may produce the cures. Am J Kidney Dis 2013; 62:1165-75. [PMID: 23896482 PMCID: PMC3840048 DOI: 10.1053/j.ajkd.2013.05.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 05/24/2013] [Indexed: 12/22/2022]
Abstract
Variable rates of disease observed between members of different continental population groups may be mediated by inherited factors, environmental exposures, or their combination. This article provides evidence in support of differential allele frequency distributions that underlie the higher rates of nondiabetic kidney disease in the focal segmental glomerulosclerosis spectrum of disease and lower rates of coronary artery calcified atherosclerotic plaque and osteoporosis in populations of African ancestry. With recognition that these and other common complex diseases are affected by biological factors comes the realization that targeted manipulation of environmental exposures and pharmacologic treatments will have different effects based on genotype. The present era of precision medicine will couple one's genetic makeup with specific therapies to reduce rates of disease based on the presence of disease-specific alleles.
Collapse
Affiliation(s)
- Barry I Freedman
- Department of Internal Medicine-Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, NC; Center for Human Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC.
| | | | | |
Collapse
|
150
|
Parsa A, Fuchsberger C, Köttgen A, O’Seaghdha CM, Pattaro C, de Andrade M, Chasman DI, Teumer A, Endlich K, Olden M, Chen MH, Tin A, Kim YJ, Taliun D, Li M, Feitosa M, Gorski M, Yang Q, Hundertmark C, Foster MC, Glazer N, Isaacs A, Rao M, Smith AV, O’Connell JR, Struchalin M, Tanaka T, Li G, Hwang SJ, Atkinson EJ, Lohman K, Cornelis MC, Johansson Å, Tönjes A, Dehghan A, Couraki V, Holliday EG, Sorice R, Kutalik Z, Lehtimäki T, Esko T, Deshmukh H, Ulivi S, Chu AY, Murgia F, Trompet S, Imboden M, Kollerits B, Pistis G, Harris TB, Launer LJ, Aspelund T, Eiriksdottir G, Mitchell BD, Boerwinkle E, Schmidt H, Hofer E, Hu F, Demirkan A, Oostra BA, Turner ST, Ding J, Andrews JS, Freedman BI, Giulianini F, Koenig W, Illig T, Döring A, Wichmann HE, Zgaga L, Zemunik T, Boban M, Minelli C, Wheeler HE, Igl W, Zaboli G, Wild SH, Wright AF, Campbell H, Ellinghaus D, Nöthlings U, Jacobs G, Biffar R, Ernst F, Homuth G, Kroemer HK, Nauck M, Stracke S, Völker U, Völzke H, Kovacs P, Stumvoll M, Mägi R, Hofman A, Uitterlinden AG, Rivadeneira F, Aulchenko YS, Polasek O, Hastie N, Vitart V, et alParsa A, Fuchsberger C, Köttgen A, O’Seaghdha CM, Pattaro C, de Andrade M, Chasman DI, Teumer A, Endlich K, Olden M, Chen MH, Tin A, Kim YJ, Taliun D, Li M, Feitosa M, Gorski M, Yang Q, Hundertmark C, Foster MC, Glazer N, Isaacs A, Rao M, Smith AV, O’Connell JR, Struchalin M, Tanaka T, Li G, Hwang SJ, Atkinson EJ, Lohman K, Cornelis MC, Johansson Å, Tönjes A, Dehghan A, Couraki V, Holliday EG, Sorice R, Kutalik Z, Lehtimäki T, Esko T, Deshmukh H, Ulivi S, Chu AY, Murgia F, Trompet S, Imboden M, Kollerits B, Pistis G, Harris TB, Launer LJ, Aspelund T, Eiriksdottir G, Mitchell BD, Boerwinkle E, Schmidt H, Hofer E, Hu F, Demirkan A, Oostra BA, Turner ST, Ding J, Andrews JS, Freedman BI, Giulianini F, Koenig W, Illig T, Döring A, Wichmann HE, Zgaga L, Zemunik T, Boban M, Minelli C, Wheeler HE, Igl W, Zaboli G, Wild SH, Wright AF, Campbell H, Ellinghaus D, Nöthlings U, Jacobs G, Biffar R, Ernst F, Homuth G, Kroemer HK, Nauck M, Stracke S, Völker U, Völzke H, Kovacs P, Stumvoll M, Mägi R, Hofman A, Uitterlinden AG, Rivadeneira F, Aulchenko YS, Polasek O, Hastie N, Vitart V, Helmer C, Wang JJ, Stengel B, Ruggiero D, Bergmann S, Kähönen M, Viikari J, Nikopensius T, Province M, Colhoun H, Doney A, Robino A, Krämer BK, Portas L, Ford I, Buckley BM, Adam M, Thun GA, Paulweber B, Haun M, Sala C, Mitchell P, Ciullo M, Vollenweider P, Raitakari O, Metspalu A, Palmer C, Gasparini P, Pirastu M, Jukema JW, Probst-Hensch NM, Kronenberg F, Toniolo D, Gudnason V, Shuldiner AR, Coresh J, Schmidt R, Ferrucci L, van Duijn CM, Borecki I, Kardia SL, Liu Y, Curhan GC, Rudan I, Gyllensten U, Wilson JF, Franke A, Pramstaller PP, Rettig R, Prokopenko I, Witteman J, Hayward C, Ridker PM, Bochud M, Heid IM, Siscovick DS, Fox CS, Kao WL, Böger CA. Common variants in Mendelian kidney disease genes and their association with renal function. J Am Soc Nephrol 2013; 24:2105-17. [PMID: 24029420 PMCID: PMC3839542 DOI: 10.1681/asn.2012100983] [Show More Authors] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 07/10/2013] [Indexed: 12/28/2022] Open
Abstract
Many common genetic variants identified by genome-wide association studies for complex traits map to genes previously linked to rare inherited Mendelian disorders. A systematic analysis of common single-nucleotide polymorphisms (SNPs) in genes responsible for Mendelian diseases with kidney phenotypes has not been performed. We thus developed a comprehensive database of genes for Mendelian kidney conditions and evaluated the association between common genetic variants within these genes and kidney function in the general population. Using the Online Mendelian Inheritance in Man database, we identified 731 unique disease entries related to specific renal search terms and confirmed a kidney phenotype in 218 of these entries, corresponding to mutations in 258 genes. We interrogated common SNPs (minor allele frequency >5%) within these genes for association with the estimated GFR in 74,354 European-ancestry participants from the CKDGen Consortium. However, the top four candidate SNPs (rs6433115 at LRP2, rs1050700 at TSC1, rs249942 at PALB2, and rs9827843 at ROBO2) did not achieve significance in a stage 2 meta-analysis performed in 56,246 additional independent individuals, indicating that these common SNPs are not associated with estimated GFR. The effect of less common or rare variants in these genes on kidney function in the general population and disease-specific cohorts requires further research.
Collapse
Affiliation(s)
- Afshin Parsa
- Division of Nephrology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Christian Fuchsberger
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Anna Köttgen
- Renal Division, Freiburg University Clinic, Freiburg, Germany
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Conall M. O’Seaghdha
- National Heart, Lung, and Blood Institute's Framingham Heart Study and the Center for Population Studies, Framingham, Massachusetts
- Division of Nephrology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Cristian Pattaro
- Centre for Biomedicine, European Academy of Bozen/Bolzano, Bolzano, Italy
| | - Mariza de Andrade
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Daniel I. Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Alexander Teumer
- Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Karlhans Endlich
- Institute of Anatomy and Cell Biology, University of Greifswald, Greifswald, Germany
| | - Matthias Olden
- Division of Nephrology, Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
| | - Ming-Huei Chen
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Adrienne Tin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Young J. Kim
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
- Genomics Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Daniel Taliun
- Centre for Biomedicine, European Academy of Bozen/Bolzano, Bolzano, Italy
| | - Man Li
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Mary Feitosa
- Division of Statistical Genomics, Washington University School of Medicine, St. Louis, Missouri
| | - Mathias Gorski
- Division of Nephrology, Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | | | - Meredith C. Foster
- National Heart, Lung, and Blood Institute's Framingham Heart Study and the Center for Population Studies, Framingham, Massachusetts
| | - Nicole Glazer
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Aaron Isaacs
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Centre for Medical Systems Biology, Rotterdam, The Netherlands
| | - Madhumathi Rao
- Division of Nephrology, Tufts Evidence Practice Center, Tufts University School of Medicine, Tufts Medical Center, Boston, Massachusetts
| | - Albert V. Smith
- Research Institute, Icelandic Heart Association, Kopavogur, Iceland
- University of Iceland, Reykjavik, Iceland
| | - Jeffrey R. O’Connell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Maksim Struchalin
- Departments of Epidemiology and Biostatistics and Forensic Molecular Biology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Toshiko Tanaka
- Clinical Research Branch, National Institute of Aging, Baltimore Maryland
| | - Guo Li
- University of Washington, Seattle, Washington
| | - Shih-Jen Hwang
- National Heart, Lung, and Blood Institute's Framingham Heart Study and the Center for Population Studies, Framingham, Massachusetts
| | - Elizabeth J. Atkinson
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Kurt Lohman
- Department of Epidemiology and Prevention, Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Marilyn C. Cornelis
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts
| | - Åsa Johansson
- Rudbeck Laboratory, Department of Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Anke Tönjes
- Department of Medicine, University of Leipzig, Leipzig, Germany
- Adiposity Diseases Integrated Research and Treatment Center, University of Leipzig, Leipzig, Germany
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Elizabeth G. Holliday
- Centre for Clinical Epidemiology and Biostatistics, School of Medicine and Public Health, University of Newcastle, Newcastle, Australia
- Centre for Information-Based Medicine, Hunter Medical Research Institute, Newcastle, Australia
| | - Rossella Sorice
- Adriano-Buzzati Traverso-CNR Institute of Genetics and Biophysics, Naples, Italy
| | - Zoltan Kutalik
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Terho Lehtimäki
- Fimlab Laboratories, Department of Clinical Chemistry, School of Medicine, University of Tampere, Tampere, Finland
| | - Tõnu Esko
- Estonian Genome Center, University of Tartu, Tartu, Estonia
- Institute of Molecular and Cell Biology, Estonian Biocentre, University of Tartu, Tartu, Estonia
| | - Harshal Deshmukh
- Wellcome Trust Centre for Molecular Medicine, Clinical Research Centre, University of Dundee, Ninewells Hospital, Dundee, United Kingdom
| | - Sheila Ulivi
- IRCCS Burlo Garofolo Institute for Maternal and Child Health, University of Trieste, Trieste, Italy
| | - Audrey Y. Chu
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | | | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Medea Imboden
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Barbara Kollerits
- Division of Genetic Epidemiology, Innsbruck Medical University, Innsbruck, Austria
| | - Giorgio Pistis
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Tamara B. Harris
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, Bethesda, Maryland
| | - Lenore J. Launer
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, Bethesda, Maryland
| | - Thor Aspelund
- Research Institute, Icelandic Heart Association, Kopavogur, Iceland
- University of Iceland, Reykjavik, Iceland
| | | | - Braxton D. Mitchell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Science Center, Houston, Texas
| | - Helena Schmidt
- Austrian Stroke Prevention Study, Department of Neurology, Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Edith Hofer
- Austrian Stroke Prevention Study, Clinical Division of Neurogeriatrics, Department of Neurology, University Clinic of Neurology, Medical University of Graz, Graz, Austria
| | - Frank Hu
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts
| | - Ayse Demirkan
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ben A. Oostra
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Stephen T. Turner
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Jingzhong Ding
- Division of Geriatrics, Department of Internal Medicine, Wake Forest School of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - Jeanette S. Andrews
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - Barry I. Freedman
- Division of Nephrology, Department of Internal Medicine, Wake Forest School of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - Franco Giulianini
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Wolfgang Koenig
- Department of Internal Medicine II, Ulm University Clinic, University of Ulm, Ulm, Germany
| | - Thomas Illig
- Hanover Unified Biobank, Hanover Medical School, Hanover, Germany
- Research Unit of Molecular Epidemiology, German Research Center for Environmental Health, Neuherberg, Germany
| | - Angela Döring
- Institute of Epidemiology I and II, German Research Center for Environmental Health, Neuherberg, Germany
| | - H.-Erich Wichmann
- Institute of Epidemiology I and II, German Research Center for Environmental Health, Neuherberg, Germany
- Institute for Medical Informatics, Biometry, and Epidemiology, Ludwig-Maximilians-University, Munich, Germany
- Grosshadern Clinic, Neuherberg, Germany
| | - Lina Zgaga
- Center for Population Health Sciences, University of Edinburgh Medical School, Edinburgh, Scotland, United Kingdom
| | - Tatijana Zemunik
- Croatian Centre for Global Health, University of Split Medical School, Split, Croatia
| | - Mladen Boban
- Croatian Centre for Global Health, University of Split Medical School, Split, Croatia
| | - Cosetta Minelli
- Centre for Biomedicine, European Academy of Bozen/Bolzano, Bolzano, Italy
| | - Heather E. Wheeler
- Department of Genetics, Stanford University, Stanford, California
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Wilmar Igl
- Rudbeck Laboratory, Department of Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ghazal Zaboli
- Rudbeck Laboratory, Department of Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Sarah H. Wild
- Center for Population Health Sciences, University of Edinburgh Medical School, Edinburgh, Scotland, United Kingdom
| | - Alan F. Wright
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, United Kingdom
| | - Harry Campbell
- Center for Population Health Sciences, University of Edinburgh Medical School, Edinburgh, Scotland, United Kingdom
| | - David Ellinghaus
- Institute of Clinical Molecular Biology, Christian-Albrechts University, Kiel, Germany
| | - Ute Nöthlings
- PopGen Biobank, Schleswig-Holstein University Hospital, Kiel, Germany
- Institute for Epidemiology, University of Kiel, Kiel, Germany
- Department of Nutrition and Food Sciences, University of Bonn, Bonn, Germany
| | - Gunnar Jacobs
- PopGen Biobank, Schleswig-Holstein University Hospital, Kiel, Germany
- Institute for Epidemiology, University of Kiel, Kiel, Germany
| | - Reiner Biffar
- Clinic for Prosthodontic Dentistry, Gerostomatology, and Material Science, University of Greifswald, Greifswald, Germany
| | - Florian Ernst
- Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Heyo K. Kroemer
- Institute of Pharmacology, University of Greifswald, Greifswald, Germany
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University of Greifswald, Greifswald, Germany
| | - Sylvia Stracke
- Clinic for Internal Medicine A, University of Greifswald, Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Henry Völzke
- Institute for Community Medicine, University of Greifswald, Greifswald, Germany
| | - Peter Kovacs
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Michael Stumvoll
- Department of Medicine, University of Leipzig, Leipzig, Germany
- Adiposity Diseases Integrated Research and Treatment Center, University of Leipzig, Leipzig, Germany
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu, Estonia
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Yurii S. Aulchenko
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ozren Polasek
- Croatian Centre for Global Health, University of Split Medical School, Split, Croatia
| | - Nick Hastie
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, United Kingdom
| | - Veronique Vitart
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, United Kingdom
| | - Catherine Helmer
- INSERM U897, Institute of Public Health, Victor Segalen Bordeaux II University, Bordeaux, France
- Victor Segalen Bordeaux II University, Bordeaux, France
| | - Jie Jin Wang
- Centre for Vision Research, Westmead Millennium Institute, Westmead Hospital, University of Sydney, Sydney, Australia
- Centre for Eye Research Australia, University of Melbourne, East Melbourne, Australia
| | - Bénédicte Stengel
- INSERM UMRS 1018, Villejuif, France
- UMRS 1018, University of Paris-Sud, Paris, France
| | - Daniela Ruggiero
- Adriano-Buzzati Traverso-CNR Institute of Genetics and Biophysics, Naples, Italy
| | - Sven Bergmann
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, School of Medicine, University of Tampere, Tampere, Finland
| | - Jorma Viikari
- Department of Medicine, Turku University Hospital, University of Turku, Turku, Finland
| | - Tiit Nikopensius
- Institute of Molecular and Cell Biology, Estonian Biocentre, University of Tartu, Tartu, Estonia
| | - Michael Province
- Division of Statistical Genomics, Washington University School of Medicine, St. Louis, Missouri
| | - Helen Colhoun
- Wellcome Trust Centre for Molecular Medicine, Clinical Research Centre, University of Dundee, Ninewells Hospital, Dundee, United Kingdom
| | - Alex Doney
- National Health Service Tayside, Wellcome Trust Centre for Molecular Medicine, Clinical Research Centre, Ninewells Hospital, Dundee, United Kingdom
| | - Antonietta Robino
- IRCCS Burlo Garofolo Institute for Maternal and Child Health, University of Trieste, Trieste, Italy
| | - Bernhard K. Krämer
- Fifth Department of Medicine, Mannheim University Medical Centre, Mannheim, Germany
| | - Laura Portas
- CNR Institute of Population Genetics, Sassari, Italy
| | - Ian Ford
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, United Kingdom
| | - Brendan M. Buckley
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Martin Adam
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Gian-Andri Thun
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Bernhard Paulweber
- First Department of Internal Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Margot Haun
- Division of Genetic Epidemiology, Innsbruck Medical University, Innsbruck, Austria
| | - Cinzia Sala
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Paul Mitchell
- Centre for Vision Research, Westmead Millennium Institute, Westmead Hospital, University of Sydney, Sydney, Australia
| | - Marina Ciullo
- Adriano-Buzzati Traverso-CNR Institute of Genetics and Biophysics, Naples, Italy
| | - Peter Vollenweider
- Department of Internal Medicine, Vaudois University Hospital Center, University of Lausanne, Lausanne, Switzerland
| | - Olli Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu, Estonia
- Institute of Molecular and Cell Biology, Estonian Biocentre, University of Tartu, Tartu, Estonia
| | - Colin Palmer
- Biomedical Research Institute, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Paolo Gasparini
- IRCCS Burlo Garofolo Institute for Maternal and Child Health, University of Trieste, Trieste, Italy
| | - Mario Pirastu
- CNR Institute of Population Genetics, Sassari, Italy
| | - J. Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden, The Netherlands
- Durrer Center for Cardiogenetic Research, Amsterdam, The Netherlands
| | - Nicole M. Probst-Hensch
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Florian Kronenberg
- Division of Genetic Epidemiology, Innsbruck Medical University, Innsbruck, Austria
| | - Daniela Toniolo
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
- CNR Institute of Molecular Genetics, Pavia, Italy
| | - Vilmundur Gudnason
- Research Institute, Icelandic Heart Association, Kopavogur, Iceland
- University of Iceland, Reykjavik, Iceland
| | - Alan R. Shuldiner
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
- Geriatric Research and Education Clinical Center, Veterans Affairs Medical Center, Baltimore, Maryland
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- Welch Center for Prevention, Epidemiology, and Clinical Research, Baltimore, Maryland
| | - Reinhold Schmidt
- Austrian Stroke Prevention Study, Clinical Division of Neurogeriatrics, Department of Neurology, University Clinic of Neurology, Medical University of Graz, Graz, Austria
| | - Luigi Ferrucci
- Clinical Research Branch, National Institute of Aging, Baltimore Maryland
| | - Cornelia M. van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Centre for Medical Systems Biology, Rotterdam, The Netherlands
- Netherlands Consortium for Healthy Aging, Netherlands Genomics Initiative, Leiden, the Netherlands
| | - Ingrid Borecki
- Division of Statistical Genomics, Washington University School of Medicine, St. Louis, Missouri
| | - Sharon L.R. Kardia
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Yongmei Liu
- Department of Epidemiology and Prevention, Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Gary C. Curhan
- Channing Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Igor Rudan
- Center for Population Health Sciences, University of Edinburgh Medical School, Edinburgh, Scotland, United Kingdom
| | - Ulf Gyllensten
- Rudbeck Laboratory, Department of Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - James F. Wilson
- Center for Population Health Sciences, University of Edinburgh Medical School, Edinburgh, Scotland, United Kingdom
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts University, Kiel, Germany
| | | | - Rainer Rettig
- Institute of Physiology, University of Greifswald, Karlsburg, Germany
| | - Inga Prokopenko
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Jacqueline Witteman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, United Kingdom
| | - Paul M. Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Murielle Bochud
- University Institute of Social and Preventive Medicine, Vaudois University Hospital Center, University of Lausanne, Lausanne, Switzerland
| | - Iris M. Heid
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany; and
| | | | - Caroline S. Fox
- National Heart, Lung, and Blood Institute's Framingham Heart Study and the Center for Population Studies, Framingham, Massachusetts
- Division of Endocrinology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - W. Linda Kao
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- Welch Center for Prevention, Epidemiology, and Clinical Research, Baltimore, Maryland
| | - Carsten A. Böger
- Division of Nephrology, Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|