51
|
Cañadas-Garre M, Anderson K, Cappa R, Skelly R, Smyth LJ, McKnight AJ, Maxwell AP. Genetic Susceptibility to Chronic Kidney Disease - Some More Pieces for the Heritability Puzzle. Front Genet 2019; 10:453. [PMID: 31214239 PMCID: PMC6554557 DOI: 10.3389/fgene.2019.00453] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/30/2019] [Indexed: 12/12/2022] Open
Abstract
Chronic kidney disease (CKD) is a major global health problem with an increasing prevalence partly driven by aging population structure. Both genomic and environmental factors contribute to this complex heterogeneous disease. CKD heritability is estimated to be high (30-75%). Genome-wide association studies (GWAS) and GWAS meta-analyses have identified several genetic loci associated with CKD, including variants in UMOD, SHROOM3, solute carriers, and E3 ubiquitin ligases. However, these genetic markers do not account for all the susceptibility to CKD, and the causal pathways remain incompletely understood; other factors must be contributing to the missing heritability. Less investigated biological factors such as telomere length; mitochondrial proteins, encoded by nuclear genes or specific mitochondrial DNA (mtDNA) encoded genes; structural variants, such as copy number variants (CNVs), insertions, deletions, inversions and translocations are poorly covered and may explain some of the missing heritability. The sex chromosomes, often excluded from GWAS studies, may also help explain gender imbalances in CKD. In this review, we outline recent findings on molecular biomarkers for CKD (telomeres, CNVs, mtDNA variants, sex chromosomes) that typically have received less attention than gene polymorphisms. Shorter telomere length has been associated with renal dysfunction and CKD progression, however, most publications report small numbers of subjects with conflicting findings. CNVs have been linked to congenital anomalies of the kidney and urinary tract, posterior urethral valves, nephronophthisis and immunoglobulin A nephropathy. Information on mtDNA biomarkers for CKD comes primarily from case reports, therefore the data are scarce and diverse. The most consistent finding is the A3243G mutation in the MT-TL1 gene, mainly associated with focal segmental glomerulosclerosis. Only one GWAS has found associations between X-chromosome and renal function (rs12845465 and rs5987107). No loci in the Y-chromosome have reached genome-wide significance. In conclusion, despite the efforts to find the genetic basis of CKD, it remains challenging to explain all of the heritability with currently available methods and datasets. Although additional biomarkers have been investigated in less common suspects such as telomeres, CNVs, mtDNA and sex chromosomes, hidden heritability in CKD remains elusive, and more comprehensive approaches, particularly through the integration of multiple -"omics" data, are needed.
Collapse
Affiliation(s)
- Marisa Cañadas-Garre
- Epidemiology and Public Health Research Group, Centre for Public Health, Queen’s University of Belfast, Belfast, United Kingdom
| | - Kerry Anderson
- Epidemiology and Public Health Research Group, Centre for Public Health, Queen’s University of Belfast, Belfast, United Kingdom
| | - Ruaidhri Cappa
- Epidemiology and Public Health Research Group, Centre for Public Health, Queen’s University of Belfast, Belfast, United Kingdom
| | - Ryan Skelly
- Epidemiology and Public Health Research Group, Centre for Public Health, Queen’s University of Belfast, Belfast, United Kingdom
| | - Laura Jane Smyth
- Epidemiology and Public Health Research Group, Centre for Public Health, Queen’s University of Belfast, Belfast, United Kingdom
| | - Amy Jayne McKnight
- Epidemiology and Public Health Research Group, Centre for Public Health, Queen’s University of Belfast, Belfast, United Kingdom
| | - Alexander Peter Maxwell
- Epidemiology and Public Health Research Group, Centre for Public Health, Queen’s University of Belfast, Belfast, United Kingdom
- Regional Nephrology Unit, Belfast City Hospital, Belfast, United Kingdom
| |
Collapse
|
52
|
Huang S, Sheng X, Susztak K. The kidney transcriptome, from single cells to whole organs and back. Curr Opin Nephrol Hypertens 2019; 28:219-226. [PMID: 30844884 PMCID: PMC6761926 DOI: 10.1097/mnh.0000000000000495] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE OF REVIEW Transcriptome analysis of human kidney samples provides an integrated output of genetic, physiological, or environmental inputs. This review summarizes recent findings including gene expression and genetic variation integration, bulk and single cell gene expression analysis, and describes how such studies have improved our understanding of kidney disease development. RECENT FINDINGS Bulk or whole tissue analysis of patient kidney samples identified a large number of genes, whose levels correlate with kidney function and/or structural damage. These genes were enriched for metabolic and immune functions. Using expression quantitative trait analysis, genetic variations-driven gene expression can be identified. Recent developments in single cell sequencing defined cell-type-specific gene expression changes and highlighted specific cell types for disease development. SUMMARY Recent advancement in whole tissue transcriptomics, specifically incorporating genotype information and single cell data have been powerful to identify kidney disease-associated genes, pathways, and cell types.
Collapse
Affiliation(s)
- Shizheng Huang
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
| | - Xin Sheng
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
| | - Katalin Susztak
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
53
|
The mercapturomic profile of health and non-communicable diseases. High Throughput 2019; 8:ht8020010. [PMID: 31018482 PMCID: PMC6630208 DOI: 10.3390/ht8020010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 04/15/2019] [Accepted: 04/17/2019] [Indexed: 12/17/2022] Open
Abstract
The mercapturate pathway is a unique metabolic circuitry that detoxifies electrophiles upon adducts formation with glutathione. Since its discovery over a century ago, most of the knowledge on the mercapturate pathway has been provided from biomonitoring studies on environmental exposure to toxicants. However, the mercapturate pathway-related metabolites that is formed in humans—the mercapturomic profile—in health and disease is yet to be established. In this paper, we put forward the hypothesis that these metabolites are key pathophysiologic factors behind the onset and development of non-communicable chronic inflammatory diseases. This review goes from the evidence in the formation of endogenous metabolites undergoing the mercapturate pathway to the methodologies for their assessment and their association with cancer and respiratory, neurologic and cardiometabolic diseases.
Collapse
|
54
|
Graham SE, Nielsen JB, Zawistowski M, Zhou W, Fritsche LG, Gabrielsen ME, Skogholt AH, Surakka I, Hornsby WE, Fermin D, Larach DB, Kheterpal S, Brummett CM, Lee S, Kang HM, Abecasis GR, Romundstad S, Hallan S, Sampson MG, Hveem K, Willer CJ. Sex-specific and pleiotropic effects underlying kidney function identified from GWAS meta-analysis. Nat Commun 2019; 10:1847. [PMID: 31015462 PMCID: PMC6478837 DOI: 10.1038/s41467-019-09861-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 04/03/2019] [Indexed: 12/19/2022] Open
Abstract
Chronic kidney disease (CKD) is a growing health burden currently affecting 10–15% of adults worldwide. Estimated glomerular filtration rate (eGFR) as a marker of kidney function is commonly used to diagnose CKD. We analyze eGFR data from the Nord-Trøndelag Health Study and Michigan Genomics Initiative and perform a GWAS meta-analysis with public summary statistics, more than doubling the sample size of previous meta-analyses. We identify 147 loci (53 novel) associated with eGFR, including genes involved in transcriptional regulation, kidney development, cellular signaling, metabolism, and solute transport. Additionally, sex-stratified analysis identifies one locus with more significant effects in women than men. Using genetic risk scores constructed from these eGFR meta-analysis results, we show that associated variants are generally predictive of CKD with only modest improvements in detection compared with other known clinical risk factors. Collectively, these results yield additional insight into the genetic factors underlying kidney function and progression to CKD. Estimated glomerular filtration rate (eGFR) is a measure of kidney function and used to characterize chronic kidney disease. Here, Graham et al. identify 53 novel loci for eGFR in a GWAS meta-analysis, a subset of which are associated with other common diseases, such as diabetes and hypertension, based on PheWAS.
Collapse
Affiliation(s)
- Sarah E Graham
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Jonas B Nielsen
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Matthew Zawistowski
- Department of Biostatistics: Center for Statistical Genetics, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Wei Zhou
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Lars G Fritsche
- Department of Biostatistics: Center for Statistical Genetics, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Maiken E Gabrielsen
- K.G. Jebsen Center for Genetic Epidemiology, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, 7491, Norway.,Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, 7491, Norway
| | - Anne Heidi Skogholt
- K.G. Jebsen Center for Genetic Epidemiology, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, 7491, Norway.,Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, 7491, Norway.,Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, 7491, Norway
| | - Ida Surakka
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Whitney E Hornsby
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Damian Fermin
- Department of Pediatrics: Pediatric Nephrology, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Daniel B Larach
- Department of Anesthesiology, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Sachin Kheterpal
- Department of Anesthesiology, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Chad M Brummett
- Department of Anesthesiology, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Seunggeun Lee
- Department of Biostatistics: Center for Statistical Genetics, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Hyun Min Kang
- Department of Biostatistics: Center for Statistical Genetics, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Goncalo R Abecasis
- Department of Biostatistics: Center for Statistical Genetics, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Solfrid Romundstad
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, 7491, Norway.,Department of Internal Medicine, Levanger Hospital, Health Trust Nord-Trøndelag, Levanger, 7600, Norway
| | - Stein Hallan
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, 7491, Norway.,Department of Nephrology, St Olav Hospital, Trondheim, 7491, Norway
| | - Matthew G Sampson
- Department of Pediatrics: Pediatric Nephrology, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, 7491, Norway. .,Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, 7491, Norway. .,HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, 7600, Norway.
| | - Cristen J Willer
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, 48109, MI, USA. .,Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, 48109, MI, USA. .,Department of Human Genetics, University of Michigan, Ann Arbor, 48109, MI, USA.
| |
Collapse
|
55
|
Maydan O, McDade PG, Liu Y, Wu XR, Matsell DG, Eddy AA. Uromodulin deficiency alters tubular injury and interstitial inflammation but not fibrosis in experimental obstructive nephropathy. Physiol Rep 2019; 6:e13654. [PMID: 29595914 PMCID: PMC5875544 DOI: 10.14814/phy2.13654] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 02/07/2018] [Accepted: 02/10/2018] [Indexed: 12/11/2022] Open
Abstract
Human GWAS and Mendelian genetic studies have linked polymorphic variants and mutations in the human uromodulin gene (UMOD) with chronic kidney disease. The primary function of this kidney‐specific and secreted protein remains elusive. This study investigated whether UMOD deficiency modified responses to unilateral ureteral obstruction (UUO)‐induced kidney injury. Kidneys harvested from groups of wild‐type (UMOD+/+) and knockout (UMOD−/−) male mice (n = 7–10 each) were studied on days 7, 14, and 21. Compared to sham kidneys, UMOD protein levels increased 9–13x after UUO and were associated with increased urinary UMOD levels. Kidney KIM‐1 protein levels were higher in the UMOD−/− groups at all time‐points (4–14x). The UMOD−/− groups also had higher KIM‐1 kidney‐to‐urine relative ratios (5–35x). In vitro studies using KIM‐1 expressing 769‐P cells showed lower KIM‐1 levels in the presence of UMOD protein. Levels of proapoptotic genes and the epithelial cell apoptotic protein marker M30 were significantly lower in the UMOD−/− groups. Both M30 and KIM‐1 colocalized with intraluminal UMOD protein deposits. Interstitial inflammation was less intense in the UMOD−/− groups. Renal fibrosis severity (kidney collagen mRNA and protein) was similar in both genotypic groups on days 7, 14, and 21. Our findings suggest a role for UMOD‐dependent inhibition of KIM‐1 expression and its apoptotic cell scavenging responses during chronic obstruction‐associated tubular injury.
Collapse
Affiliation(s)
- Olena Maydan
- Department of Pediatrics, University of British Columbia and British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Paul G McDade
- Department of Pediatrics, University of British Columbia and British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Yan Liu
- Department of Urology, New York University, New York, New York
| | - Xue-Ru Wu
- Department of Urology, New York University, New York, New York
| | - Douglas G Matsell
- Department of Pediatrics, University of British Columbia and British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Allison A Eddy
- Department of Pediatrics, University of British Columbia and British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
56
|
Cañadas-Garre M, Anderson K, McGoldrick J, Maxwell AP, McKnight AJ. Proteomic and metabolomic approaches in the search for biomarkers in chronic kidney disease. J Proteomics 2019; 193:93-122. [PMID: 30292816 DOI: 10.1016/j.jprot.2018.09.020] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 09/20/2018] [Accepted: 09/30/2018] [Indexed: 12/15/2022]
Abstract
Chronic kidney disease (CKD) is an aging-related disorder that represents a major global public health burden. Current biochemical biomarkers, such as serum creatinine and urinary albumin, have important limitations when used to identify the earliest indication of CKD or in tracking the progression to more advanced CKD. These issues underline the importance of finding and testing new molecular biomarkers that are capable of successfully meeting this clinical need. The measurement of changes in nature and/or levels of proteins and metabolites in biological samples from patients provide insights into pathophysiological processes. Proteomic and metabolomic techniques provide opportunities to record dynamic chemical signatures in patients over time. This review article presents an overview of the recent developments in the fields of metabolomics and proteomics in relation to CKD. Among the many different proteomic biomarkers proposed, there is particular interest in the CKD273 classifier, a urinary proteome biomarker reported to predict CKD progression and with implementation potential. Other individual non-invasive peptidomic biomarkers that are potentially relevant for CKD detection include type 1 collagen, uromodulin and mucin-1. Despite the limited sample sizes and variability of the metabolomics studies, some metabolites such as trimethylamine N-oxide, kynurenine and citrulline stand out as potential biomarkers in CKD.
Collapse
Affiliation(s)
- M Cañadas-Garre
- Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University of Belfast, Regional Genetics Centre, Level A, Tower Block, Belfast City Hospital, Lisburn Road, Belfast BT9 7AB, United Kingdom; Regional Nephrology Unit, Belfast City Hospital, Belfast, United Kingdom.
| | - K Anderson
- Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University of Belfast, Regional Genetics Centre, Level A, Tower Block, Belfast City Hospital, Lisburn Road, Belfast BT9 7AB, United Kingdom; Regional Nephrology Unit, Belfast City Hospital, Belfast, United Kingdom.
| | - J McGoldrick
- Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University of Belfast, Regional Genetics Centre, Level A, Tower Block, Belfast City Hospital, Lisburn Road, Belfast BT9 7AB, United Kingdom; Regional Nephrology Unit, Belfast City Hospital, Belfast, United Kingdom.
| | - A P Maxwell
- Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University of Belfast, Regional Genetics Centre, Level A, Tower Block, Belfast City Hospital, Lisburn Road, Belfast BT9 7AB, United Kingdom; Regional Nephrology Unit, Belfast City Hospital, Belfast, United Kingdom.
| | - A J McKnight
- Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University of Belfast, Regional Genetics Centre, Level A, Tower Block, Belfast City Hospital, Lisburn Road, Belfast BT9 7AB, United Kingdom; Regional Nephrology Unit, Belfast City Hospital, Belfast, United Kingdom.
| |
Collapse
|
57
|
Taglieri N, Nanni C, Ghetti G, Bonfiglioli R, Saia F, Buia F, Lima GM, Marco V, Bruno AG, Prati F, Fanti S, Rapezzi C. Multi-Imaging Investigation to Evaluate the Relationship between Serum Cystatin C and Features of Atherosclerosis in Non-ST-Segment Elevation Acute Coronary Syndrome. APPLIED SCIENCES 2019; 9:657. [DOI: 10.3390/app9040657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Objectives: High cystatin C(CysC) levels are associated with impaired cardiovascular outcome. Whether CysC levels are independently related to the atherosclerosis burden is still controversial. Methods: We enrolled 31 non-ST-segment elevation acute coronary syndrome patients undergoing percutaneous coronary intervention. Patients were divided into 2 groups on the basis of median value of serum CysC. Using the high CysC group as a dependent variable, univariable and multivariable analyses were used to evaluate the association between CysC and three different features of atherosclerosis: 1) coronary plaque vulnerability as assessed by optical coherence tomography (OCT), 2) coronary artery calcium (CAC) by means of computed tomography scan, and 3) aortic wall metabolic activity, as assessed using 18F-Fluorodeoxyglucose-positron emission tomography (18F-FDG-PET). Results: After univariable and multivariable analyses, 18F-FDG uptake in the descending aorta (DA) was independently associated with a low level of CysC [(Odds Ratio = 0.02; 95%CI 0.0004–0.89; p = 0.044; 18F-FDG uptake measured as averaged maximum target to blood ratio); (Odds Ratio = 0.89; 95%CI 0.82–0.98, p = 0.025; 18F-FDG uptake measured as number of active slices)]. No trend was found for the association between CysC and characteristics of OCT-assessed coronary plaque vulnerability or CAC score. Conclusions: In patients with non-ST-segment elevation acute coronary syndrome (NSTE-ACS), 18F-FDG uptake in the DA was associated with a low level of serum CysC. There was no relation between CysC levels and OCT-assessed coronary plaque vulnerability or CAC score. These findings suggest that high levels of CysC may not be considered as independent markers of atherosclerosis.
Collapse
Affiliation(s)
- Nevio Taglieri
- Polo Cardio-Toraco-Vascolare, Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Alma Mater Studiorum Università di Bologna, 40138 Bologna, Italy
| | - Cristina Nanni
- Istituto di Medicina Nucleare, Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Alma Mater Studiorum Università di Bologna, 40138 Bologna, Italy
| | - Gabriele Ghetti
- Polo Cardio-Toraco-Vascolare, Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Alma Mater Studiorum Università di Bologna, 40138 Bologna, Italy
| | - Rachele Bonfiglioli
- Istituto di Medicina Nucleare, Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Alma Mater Studiorum Università di Bologna, 40138 Bologna, Italy
| | - Francesco Saia
- Polo Cardio-Toraco-Vascolare, Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Alma Mater Studiorum Università di Bologna, 40138 Bologna, Italy
| | - Francesco Buia
- Istituto di Radiologia, Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Alma Mater Studiorum Università di Bologna, 40138 Bologna, Italy
| | - Giacomo Maria Lima
- Istituto di Medicina Nucleare, Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Alma Mater Studiorum Università di Bologna, 40138 Bologna, Italy
| | | | - Antonio Giulio Bruno
- Polo Cardio-Toraco-Vascolare, Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Alma Mater Studiorum Università di Bologna, 40138 Bologna, Italy
| | - Francesco Prati
- CLI Foundation, 00182 Rome, Italy
- GVM Care & Research, Ettore Sansavini Health Science Foundation, 48033 Cotignola, Italy
| | - Stefano Fanti
- Istituto di Medicina Nucleare, Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Alma Mater Studiorum Università di Bologna, 40138 Bologna, Italy
| | - Claudio Rapezzi
- Polo Cardio-Toraco-Vascolare, Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Alma Mater Studiorum Università di Bologna, 40138 Bologna, Italy
| |
Collapse
|
58
|
Köttgen A, Raffler J, Sekula P, Kastenmüller G. Genome-Wide Association Studies of Metabolite Concentrations (mGWAS): Relevance for Nephrology. Semin Nephrol 2019; 38:151-174. [PMID: 29602398 DOI: 10.1016/j.semnephrol.2018.01.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Metabolites are small molecules that are intermediates or products of metabolism, many of which are freely filtered by the kidneys. In addition, the kidneys have a central role in metabolite anabolism and catabolism, as well as in active metabolite reabsorption and/or secretion during tubular passage. This review article illustrates how the coupling of genomics and metabolomics in genome-wide association analyses of metabolites can be used to illuminate mechanisms underlying human metabolism, with a special focus on insights relevant to nephrology. First, genetic susceptibility loci for reduced kidney function and chronic kidney disease (CKD) were reviewed systematically for their associations with metabolite concentrations in metabolomics studies of blood and urine. Second, kidney function and CKD-associated metabolites reported from observational studies were interrogated for metabolite-associated genetic variants to generate and discuss complementary insights. Finally, insights originating from the simultaneous study of both blood and urine or by modeling intermetabolite relationships are summarized. We also discuss methodologic questions related to the study of metabolite concentrations in urine as well as among CKD patients. In summary, genome-wide association analyses of metabolites using metabolite concentrations quantified from blood and/or urine are a promising avenue of research to illuminate physiological and pathophysiological functions of the kidney.
Collapse
Affiliation(s)
- Anna Köttgen
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany.
| | - Johannes Raffler
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Peggy Sekula
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| |
Collapse
|
59
|
Osman WM, Jelinek HF, Tay GK, Khandoker AH, Khalaf K, Almahmeed W, Hassan MH, Alsafar HS. Clinical and genetic associations of renal function and diabetic kidney disease in the United Arab Emirates: a cross-sectional study. BMJ Open 2018; 8:e020759. [PMID: 30552240 PMCID: PMC6303615 DOI: 10.1136/bmjopen-2017-020759] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Within the Emirati population, risk factors and genetic predisposition to diabetic kidney disease (DKD) have not yet been investigated. The aim of this research was to determine potential clinical, laboratory and reported genetic loci as risk factors for DKD. RESEARCH DESIGN AND METHODS Four hundred and ninety unrelated Emirati nationals with type 2 diabetes mellitus (T2DM) were recruited with and without DKD, and clinical and laboratory data were obtained. Following adjustments for possible confounders, a logistic regression model was developed to test the associations of 63 single nucleotide polymorphisms (SNPs) in 43 genetic loci with DKD (145 patients with DKD and 265 without DKD). Linear regression models, adjusted for age and gender, were then used to study the genetic associations of five renal function traits, including 83 SNPs with albumin-to-creatinine ratio, 92 SNPs with vitamin D (25-OH cholecalciferol), 288 SNPs with estimated glomerular filtration rate (eGFR), 363 SNPs with serum creatinine and 73 SNPs with blood urea. RESULTS Patients with DKD, as compared with those without the disease, were mostly men (52%vs38% for controls), older (67vs59 years) and had significant rates of hypertension and dyslipidaemia. Furthermore, patients with DKD had T2DM for a longer duration of time (16vs10 years), which in an additive manner was the single factor that significantly contributed to the development of DKD (p=0.02, OR=3.12, 95% CI 1.21 to 8.02). Among the replicated associations of the genetic loci with different renal function traits, the most notable included SHROOM3 with levels of serum creatinine, eGFR and DKD (Padjusted=0.04, OR=1.46); CASR, GC and CYP2R1 with vitamin D levels; as well as WDR72 with serum creatinine and eGFR levels. CONCLUSIONS Associations were found between several genetic loci and risk markers for DKD, which may influence kidney function traits and DKD in a population of Arab ancestry.
Collapse
Affiliation(s)
- Wael M Osman
- Center of Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Herbert F Jelinek
- School of Community Health, Charles Sturt University, Albury, New South Wales, Australia
- Clinical Medicine, Macquarie University, Sydney, New South Wales, Australia
| | - Guan K Tay
- Center of Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
- School of Health and Medical Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, Western Australia, Australia
- Biomedical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Ahsan H Khandoker
- Biomedical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Kinda Khalaf
- Biomedical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Wael Almahmeed
- Institute of Cardiac Science, Sheikh Khalifa Medical City, Abu Dhabi, United Arab Emirates
- Heart and Vascular Institute, Cleveland Clinic, Abu Dhabi, United Arab Emirates
| | - Mohamed H Hassan
- Medical Institute, Sheikh Khalifa Medical City, Abu Dhabi, United Arab Emirates
| | - Habiba S Alsafar
- Center of Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
- Biomedical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
| |
Collapse
|
60
|
Wang E, Zhao H, Zhao D, Li L, Du L. Functional Prediction of Chronic Kidney Disease Susceptibility Gene PRKAG2 by Comprehensively Bioinformatics Analysis. Front Genet 2018; 9:573. [PMID: 30559760 PMCID: PMC6287114 DOI: 10.3389/fgene.2018.00573] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 11/08/2018] [Indexed: 02/01/2023] Open
Abstract
The genetic predisposition to chronic kidney disease (CKD) has been widely evaluated especially using the genome-wide association studies, which highlighted some novel genetic susceptibility variants in many genes, and estimated glomerular filtration rate to diagnose and stage CKD. Of these variants, rs7805747 in PRKAG2 was identified to be significantly associated with both serum creatinine and CKD with genome wide significance level. Until now, the potential mechanism by which rs7805747 affects CKD risk is still unclear. Here, we performed a functional analysis of rs7805747 variant using multiple bioinformatics software and databases. Using RegulomeDB and HaploReg (version 4.1), rs7805747 was predicated to locate in enhancer histone marks (Liver, Duodenum Mucosa, Fetal Intestine Large, Fetal Intestine Small, and Right Ventricle tissues). Using GWAS analysis in PhenoScanner, we showed that rs7805747 is not only associated with CKD, but also is significantly associated with other diseases or phenotypes. Using metabolite analysis in PhenoScanner, rs7805747 is identified to be significantly associated with not only the serum creatinine, but also with other 16 metabolites. Using eQTL analysis in PhenoScanner, rs7805747 is identified to be significantly associated with gene expression in multiple human tissues and multiple genes including PRKAG2. The gene expression analysis of PRKAG2 using 53 tissues from GTEx RNA-Seq of 8555 samples (570 donors) in GTEx showed that PRKAG2 had the highest median expression in Heart-Atrial Appendage. Using the gene expression profiles in human CKD, we further identified different expression of PRKAG2 gene in CKD cases compared with control samples. In summary, our findings provide new insight into the underlying susceptibility of PRKAG2 gene to CKD.
Collapse
Affiliation(s)
- Ermin Wang
- Department of Nephrology, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, China
| | - Hainan Zhao
- Department of Nephrology, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, China
| | - Deyan Zhao
- Department of Nephrology, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, China
| | - Lijing Li
- Department of Nephrology, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, China
| | - Limin Du
- Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
61
|
Hurtado Del Pozo C, Garreta E, Izpisúa Belmonte JC, Montserrat N. Modeling epigenetic modifications in renal development and disease with organoids and genome editing. Dis Model Mech 2018; 11:dmm035048. [PMID: 30459215 PMCID: PMC6262817 DOI: 10.1242/dmm.035048] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Understanding epigenetic mechanisms is crucial to our comprehension of gene regulation in development and disease. In the past decades, different studies have shown the role of epigenetic modifications and modifiers in renal disease, especially during its progression towards chronic and end-stage renal disease. Thus, the identification of genetic variation associated with chronic kidney disease has resulted in better clinical management of patients. Despite the importance of these findings, the translation of genotype-phenotype data into gene-based medicine in chronic kidney disease populations still lacks faithful cellular or animal models that recapitulate the key aspects of the human kidney. The latest advances in the field of stem cells have shown that it is possible to emulate kidney development and function with organoids derived from human pluripotent stem cells. These have successfully recapitulated not only kidney differentiation, but also the specific phenotypical traits related to kidney function. The combination of this methodology with CRISPR/Cas9 genome editing has already helped researchers to model different genetic kidney disorders. Nowadays, CRISPR/Cas9-based approaches also allow epigenetic modifications, and thus represent an unprecedented tool for the screening of genetic variants, epigenetic modifications or even changes in chromatin structure that are altered in renal disease. In this Review, we discuss these technical advances in kidney modeling, and offer an overview of the role of epigenetic regulation in kidney development and disease.
Collapse
Affiliation(s)
- Carmen Hurtado Del Pozo
- Pluripotency for organ regeneration. Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), 08028 Barcelona, Spain
| | - Elena Garreta
- Pluripotency for organ regeneration. Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), 08028 Barcelona, Spain
| | | | - Nuria Montserrat
- Pluripotency for organ regeneration. Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), 08028 Barcelona, Spain
| |
Collapse
|
62
|
Jaykumar AB, Caceres PS, King-Medina KN, Liao TD, Datta I, Maskey D, Naggert JK, Mendez M, Beierwaltes WH, Ortiz PA. Role of Alström syndrome 1 in the regulation of blood pressure and renal function. JCI Insight 2018; 3:95076. [PMID: 30385718 DOI: 10.1172/jci.insight.95076] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 09/26/2018] [Indexed: 01/22/2023] Open
Abstract
Elevated blood pressure (BP) and renal dysfunction are complex traits representing major global health problems. Single nucleotide polymorphisms identified by genome-wide association studies have identified the Alström syndrome 1 (ALMS1) gene locus to render susceptibility for renal dysfunction, hypertension, and chronic kidney disease (CKD). Mutations in the ALMS1 gene in humans causes Alström syndrome, characterized by progressive metabolic alterations including hypertension and CKD. Despite compelling genetic evidence, the underlying biological mechanism by which mutations in the ALMS1 gene lead to the above-mentioned pathophysiology is not understood. We modeled this effect in a KO rat model and showed that ALMS1 genetic deletion leads to hypertension. We demonstrate that the link between ALMS1 and hypertension involves the activation of the renal Na+/K+/2Cl- cotransporter NKCC2, mediated by regulation of its endocytosis. Our findings establish a link between the genetic susceptibility to hypertension, CKD, and the expression of ALMS1 through its role in a salt-reabsorbing tubular segment of the kidney. These data point to ALMS1 as a potentially novel gene involved in BP and renal function regulation.
Collapse
Affiliation(s)
- Ankita Bachhawat Jaykumar
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Paulo S Caceres
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Keyona N King-Medina
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Tang-Dong Liao
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA
| | - Indrani Datta
- Department of Public Health Sciences and.,Center for Bioinformatics, Henry Ford Health System, Detroit, Michigan, USA
| | - Dipak Maskey
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA
| | | | - Mariela Mendez
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA
| | - William H Beierwaltes
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Pablo A Ortiz
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
63
|
Cañadas-Garre M, Anderson K, McGoldrick J, Maxwell AP, McKnight AJ. Genomic approaches in the search for molecular biomarkers in chronic kidney disease. J Transl Med 2018; 16:292. [PMID: 30359254 PMCID: PMC6203198 DOI: 10.1186/s12967-018-1664-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 10/14/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is recognised as a global public health problem, more prevalent in older persons and associated with multiple co-morbidities. Diabetes mellitus and hypertension are common aetiologies for CKD, but IgA glomerulonephritis, membranous glomerulonephritis, lupus nephritis and autosomal dominant polycystic kidney disease are also common causes of CKD. MAIN BODY Conventional biomarkers for CKD involving the use of estimated glomerular filtration rate (eGFR) derived from four variables (serum creatinine, age, gender and ethnicity) are recommended by clinical guidelines for the evaluation, classification, and stratification of CKD. However, these clinical biomarkers present some limitations, especially for early stages of CKD, elderly individuals, extreme body mass index values (serum creatinine), or are influenced by inflammation, steroid treatment and thyroid dysfunction (serum cystatin C). There is therefore a need to identify additional non-invasive biomarkers that are useful in clinical practice to help improve CKD diagnosis, inform prognosis and guide therapeutic management. CONCLUSION CKD is a multifactorial disease with associated genetic and environmental risk factors. Hence, many studies have employed genetic, epigenetic and transcriptomic approaches to identify biomarkers for kidney disease. In this review, we have summarised the most important studies in humans investigating genomic biomarkers for CKD in the last decade. Several genes, including UMOD, SHROOM3 and ELMO1 have been strongly associated with renal diseases, and some of their traits, such as eGFR and serum creatinine. The role of epigenetic and transcriptomic biomarkers in CKD and related diseases is still unclear. The combination of multiple biomarkers into classifiers, including genomic, and/or epigenomic, may give a more complete picture of kidney diseases.
Collapse
Affiliation(s)
- M. Cañadas-Garre
- Epidemiology and Public Health Research Group, Centre for Public Health, Belfast City Hospital, Queen’s University of Belfast, c/o University Floor, Level A, Tower Block, Lisburn Road, Belfast, BT9 7AB Northern Ireland UK
| | - K. Anderson
- Epidemiology and Public Health Research Group, Centre for Public Health, Belfast City Hospital, Queen’s University of Belfast, c/o University Floor, Level A, Tower Block, Lisburn Road, Belfast, BT9 7AB Northern Ireland UK
| | - J. McGoldrick
- Epidemiology and Public Health Research Group, Centre for Public Health, Belfast City Hospital, Queen’s University of Belfast, c/o University Floor, Level A, Tower Block, Lisburn Road, Belfast, BT9 7AB Northern Ireland UK
| | - A. P. Maxwell
- Epidemiology and Public Health Research Group, Centre for Public Health, Belfast City Hospital, Queen’s University of Belfast, c/o University Floor, Level A, Tower Block, Lisburn Road, Belfast, BT9 7AB Northern Ireland UK
- Regional Nephrology Unit, Belfast City Hospital, Belfast, UK
| | - A. J. McKnight
- Epidemiology and Public Health Research Group, Centre for Public Health, Belfast City Hospital, Queen’s University of Belfast, c/o University Floor, Level A, Tower Block, Lisburn Road, Belfast, BT9 7AB Northern Ireland UK
| |
Collapse
|
64
|
Zusi C, Trombetta M, Bonetti S, Dauriz M, Boselli ML, Trabetti E, Malerba G, Penno G, Zoppini G, Bonora E, Solini A, Bonadonna RC. A renal genetic risk score (GRS) is associated with kidney dysfunction in people with type 2 diabetes. Diabetes Res Clin Pract 2018; 144:137-143. [PMID: 30153470 DOI: 10.1016/j.diabres.2018.08.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 07/12/2018] [Accepted: 08/22/2018] [Indexed: 11/29/2022]
Abstract
This study aims to investigate whether renal and cardiovascular phenotypes in Italian patients with type 2 diabetes (T2D) could be influenced by a number of disease risk SNPs recently found in genome-wide association studies (GWAS). In 1591 Italian subjects with T2D: (1) 47 SNPs associated to kidney function and/or chronic kidney disease (CKD) and 49 SNPs associated to cardiovascular disease (CVD) risk were genotyped; (2) urinary albumin/creatinine (A/C) ratio, glomerular filtration rate (eGFR) and lipid profile were assessed; (3) a standard electrocardiogram was performed; (4) two genotype risk scores (GRS) were computed (a renal GRS calculated selecting 39 SNPs associated with intermediate traits of kidney damage and a cardiovascular GRS determined selecting 42 SNPs associated to CVD risk phenotypes). After correction for multiple comparisons, the renal GRS was not associated to A/C ratio (p = 0.33), but it was significantly related to decreased eGFR (p = 0.005). No association between the cardiovascular GRS and electrocardiogram was detected. Thus, in Italian patients with T2D a renal GRS might predict the decline in glomerular function, suggesting that the clock of diabetes associated CKD starts ticking long before hyperglycemia. Our data support the feasibility of gene-based prediction of complications in people with T2D.
Collapse
Affiliation(s)
- Chiara Zusi
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Verona, Verona, Italy
| | - Maddalena Trombetta
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Verona, Verona, Italy; Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Hospital Trust of Verona, Verona, Italy.
| | - Sara Bonetti
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Verona, Verona, Italy
| | - Marco Dauriz
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Verona, Verona, Italy
| | - Maria L Boselli
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Verona, Verona, Italy
| | - Elisabetta Trabetti
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biology and Genetics, University of Verona, Verona, Italy
| | - Giovanni Malerba
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biology and Genetics, University of Verona, Verona, Italy
| | - Giuseppe Penno
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giacomo Zoppini
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Verona, Verona, Italy
| | - Enzo Bonora
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Verona, Verona, Italy; Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Hospital Trust of Verona, Verona, Italy
| | - Anna Solini
- Department of Surgical, Medical, Molecular and Critical Area Pathology, University of Pisa, Pisa, Italy
| | - Riccardo C Bonadonna
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Division of Endocrinology and Metabolic Diseases, Azienda Ospedaliera Universitaria di Parma, Parma, Italy
| |
Collapse
|
65
|
Yamada Y, Kato K, Oguri M, Horibe H, Fujimaki T, Yasukochi Y, Takeuchi I, Sakuma J. Identification of 13 novel susceptibility loci for early-onset myocardial infarction, hypertension, or chronic kidney disease. Int J Mol Med 2018; 42:2415-2436. [PMID: 30226566 PMCID: PMC6192728 DOI: 10.3892/ijmm.2018.3852] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 08/13/2018] [Indexed: 12/28/2022] Open
Abstract
Early-onset cardiovascular and renal diseases have a strong genetic component. In the present study, exome-wide association studies (EWASs) were performed to identify genetic variants that confer susceptibility to early-onset myocardial infarction (MI), hypertension, or chronic kidney disease (CKD) in Japanese individuals. A total of 8,093 individuals aged ≤65 years was enrolled in the study. The EWASs for MI, hypertension, and CKD were performed in 6,926 subjects (1,152 cases, 5,774 controls), 8,080 subjects (3,444 cases, 4,636 controls), and 2,556 subjects (1,051 cases, 1,505 controls), respectively. Genotyping of single nucleotide polymorphisms (SNPs) was performed with Illumina Human Exome-12 DNA Analysis BeadChip or Infinium Exome-24 BeadChip arrays. The associations of allele frequencies for 31,245, 31,276, or 31,514 SNPs that passed quality control to MI, hypertension, and CKD, respectively, was examined with Fisher's exact test. Bonferroni's correction for statistical significance of association was applied to compensate for multiple comparisons of genotypes with MI, hypertension, or CKD. The EWASs of allele frequencies revealed that 25, 11, and 11 SNPs were significantly associated with MI (P<1.60×10−6), hypertension (P<1.60×10−6), or CKD (P<1.59×10−6), respectively. Multivariable logistic regression analysis with adjustment for covariates showed that all 25, 11, and 11 SNPs were significantly associated with MI (P<0.0005), hypertension (P<0.0011), or CKD (P<0.0011), respectively. On examination of the results from previous genome-wide association studies and linkage disequilibrium of the identified SNPs, 11 loci (TMOD4, COL6A3, ADGRL3-CXCL8-MARCH1, OR52E4, TCHP-GIT2, CCDC63, 12q24.1, OAS3, PLCB2-VPS33B, GOSR2, ZNF77), six loci (MOB3C-TMOD4, COL6A3, COL6A5, CXCL8-MARCH1, NFKBIL1-6p21.3-NCR3, PLCB2-VPS33B), and seven loci (MOB3C-TMOD4, COL6A3, COL6A5, ADGRL3-CXCL8-MARCH1, MUC17, PLCB2-VPS33B, ZNF77) were identified as novel loci significantly associated with MI, hypertension, and CKD, respectively. Furthermore, six genes (TMOD4, COL6A3, CXCL8, MARCH1, PLCB2, VPS33B) were significantly associated with MI, hypertension and CKD; two genes (ADGRL3, ZNF77) with MI and CKD; and two genes (COL6A5, MOB3C) with hypertension and CKD. Therefore, 13 novel loci (MOB3C-TMOD4, COL6A3, ADGRL3-CXCL8-MARCH1, OR52E4, TCHP- GIT2, CCDC63, 12q24.1, OAS3, PLCB2-VPS33B, ZNF77, COL6A5, NFKBIL1-NCR3, MUC17) were identified that confer susceptibility to early-onset MI, hypertension, or CKD. The determination of genotypes for the SNPs at these loci may provide informative for assessment of the genetic risk for MI, hypertension, or CKD.
Collapse
Affiliation(s)
- Yoshiji Yamada
- Department of Human Functional Genomics, Advanced Science Research Promotion Center, Mie University, Tsu, Mie 514‑8507, Japan
| | - Kimihiko Kato
- Department of Human Functional Genomics, Advanced Science Research Promotion Center, Mie University, Tsu, Mie 514‑8507, Japan
| | - Mitsutoshi Oguri
- Department of Human Functional Genomics, Advanced Science Research Promotion Center, Mie University, Tsu, Mie 514‑8507, Japan
| | - Hideki Horibe
- Department of Cardiovascular Medicine, Gifu Prefectural Tajimi Hospital, Tajimi, Gifu 507‑8522, Japan
| | - Tetsuo Fujimaki
- Department of Cardiovascular Medicine, Northern Mie Medical Center Inabe General Hospital, Inabe, Mie 511‑0428, Japan
| | - Yoshiki Yasukochi
- Department of Human Functional Genomics, Advanced Science Research Promotion Center, Mie University, Tsu, Mie 514‑8507, Japan
| | - Ichiro Takeuchi
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama 332‑0012, Japan
| | - Jun Sakuma
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama 332‑0012, Japan
| |
Collapse
|
66
|
Kobayashi H, Otsuka H, Yanai M, Hara M, Hishiki M, Soma M, Abe M. Adiponectin Receptor gene Polymorphisms are Associated with Kidney Function in Elderly Japanese Populations. J Atheroscler Thromb 2018; 26:328-339. [PMID: 30135333 PMCID: PMC6456456 DOI: 10.5551/jat.45609] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Aim: Adiponectin exhibits its biological effects through adiponectin receptors (AdipoR1 and AdipoR2), which are distributed in the kidneys, and activation of those receptors could prevent or ameliorate diabetic nephropathy. This study aimed to evaluate the associations between AdipoR single nucleotide polymorphisms (SNPs) and kidney function in an elderly Japanese population. Methods: A total of 271 elderly Japanese volunteers underwent anthropometric and laboratory tests (cystatin C-based eGFR and total and high molecular weight adiponectin levels at baseline and a follow-up visit). Genotype data were obtained for the selected 7 and 5 AdipoR1 and AdipoR2 SNPs, respectively. Results: In a cross-sectional analysis at baseline, we found a significant association between the AdipoR2 SNP rs12230440 and kidney function; eGFRcys tended to increase as the number of carriers of T alleles increased after adjustment for covariates and Bonferroni correction, although the association of the SNP and annual eGFR decline could not be identified in the longitudinal data. Regarding the variants rs16850797, rs11061925, and rs10773983, each of the allele G, allele C, and allele G showed nominally significant associations with higher eGFRcys. However, this failed to reach significance after Bonferroni correction. Conclusion: Here, an AdipoR2 SNP was associated with kidney function, suggesting that the effects of this polymorphism on adiponectin receptor may affect kidney function in the elderly Japanese population.
Collapse
Affiliation(s)
- Hiroki Kobayashi
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine
| | - Hiromasa Otsuka
- Division of General Medicine, Department of Internal Medicine, Nihon University School of Medicine
| | - Mitsuru Yanai
- Division of General Medicine, Department of Internal Medicine, Nihon University School of Medicine
| | - Motohiko Hara
- Department of Nursing, School of Health and Social Services, Saitama Prefectural University
| | - Mikano Hishiki
- Department of Diabetes and Endocrinology, Tokyo Metropolitan Hiroo Hospital
| | - Masayoshi Soma
- Division of General Medicine, Department of Internal Medicine, Nihon University School of Medicine
| | - Masanori Abe
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine
| |
Collapse
|
67
|
Cellular and molecular mechanisms of kidney fibrosis. Mol Aspects Med 2018; 65:16-36. [PMID: 29909119 DOI: 10.1016/j.mam.2018.06.002] [Citation(s) in RCA: 329] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/12/2018] [Indexed: 12/14/2022]
Abstract
Renal fibrosis is the final pathological process common to any ongoing, chronic kidney injury or maladaptive repair. It is considered as the underlying pathological process of chronic kidney disease (CKD), which affects more than 10% of world population and for which treatment options are limited. Renal fibrosis is defined by excessive deposition of extracellular matrix, which disrupts and replaces the functional parenchyma that leads to organ failure. Kidney's histological structure can be divided into three main compartments, all of which can be affected by fibrosis, specifically termed glomerulosclerosis in glomeruli, interstitial fibrosis in tubulointerstitium and arteriosclerosis and perivascular fibrosis in vasculature. In this review, we summarized the different appearance, cellular origin and major emerging processes and mediators of fibrosis in each compartment. We also depicted and discussed the challenges in translation of anti-fibrotic treatment to clinical practice and discuss possible solutions and future directions.
Collapse
|
68
|
Cyrus C, Al-Mueilo S, Vatte C, Chathoth S, Li YR, Qutub H, Al Ali R, Al-Muhanna F, Lanktree MB, Alkharsah KR, Al-Rubaish A, Kim-Mozeleski B, Keating B, Al Ali A. Assessing known chronic kidney disease associated genetic variants in Saudi Arabian populations. BMC Nephrol 2018; 19:88. [PMID: 29665793 PMCID: PMC5905143 DOI: 10.1186/s12882-018-0890-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 04/05/2018] [Indexed: 01/13/2023] Open
Abstract
Background Genome wide association studies of patients with European descent have identified common variants associated with risk of reduced estimated glomerular filtration rate (eGFR). A panel of eight variants were selected to evaluate their association and prevalence in a Saudi Arabian patient cohort with chronic kidney disease (CKD). Methods Eight genetic variants in four genes (SHROOM3, MYH9, SLC7A9, and CST3) were genotyped in 160 CKD patients and 189 ethnicity-matched healthy controls. Genetic variants were tested for association with the development of CKD (eGFR < 60 ml/min/1.73m2) and effects were compared with results obtained from 133,413 participants in the CKD genetics consortium. Multivariable regression was used to evaluate the role of these eight variants in improving prediction of CKD development. Results All eight variants were present in Saudi populations with minor allele frequency ranging from 16 to 46%. The risk variant in all four genes demonstrated the same direction of effect as observed in European populations. One variant, rs4821480, in MYH9 was significantly associated with increased risk of development of CKD (OR = 1.69, 95% CI 1.22–2.36, P = 0.002), but the additional variants were not statistically significant given our modest sample size. Conclusions CKD risk variants identified in European populations are present in Saudis. We did not find evidence to suggest heterogeneity of effect size compared to previously published estimates in European populations. Multivariable logistic regression analysis showed a statistically significant improvement in predicting the CKD using models with either FGF23 and vitamin D or FGF23, vitamin D level, and MYH9 genotypes (AUC = 0.93, 95% CI 0.90–0.95, P < 0.0001). Electronic supplementary material The online version of this article (10.1186/s12882-018-0890-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Cyril Cyrus
- Institute for Research and Medical Consultation, Imam Abdulrahman bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia.
| | - Samir Al-Mueilo
- King Fahd Hospital of the University, Imam Abdulrahman bin Faisal University , Alkhobar, Saudi Arabia
| | - Chittibabu Vatte
- Institute for Research and Medical Consultation, Imam Abdulrahman bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| | - Shahanas Chathoth
- Institute for Research and Medical Consultation, Imam Abdulrahman bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| | - Yun R Li
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hatem Qutub
- King Fahd Hospital of the University, Imam Abdulrahman bin Faisal University , Alkhobar, Saudi Arabia.,Al-Omran Scientific Chair for hematological diseases, King Faisal University, Al Hassa, Saudi Arabia
| | - Rudaynah Al Ali
- King Fahd Hospital of the University, Imam Abdulrahman bin Faisal University , Alkhobar, Saudi Arabia
| | - Fahad Al-Muhanna
- King Fahd Hospital of the University, Imam Abdulrahman bin Faisal University , Alkhobar, Saudi Arabia
| | - Matthew B Lanktree
- Nephrology Division, Department of Medicine, McMaster University, Hamilton, ON, L8N 4A6, Canada
| | - Khaled Riyad Alkharsah
- Institute for Research and Medical Consultation, Imam Abdulrahman bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| | - Abdullah Al-Rubaish
- King Fahd Hospital of the University, Imam Abdulrahman bin Faisal University , Alkhobar, Saudi Arabia
| | - Brian Kim-Mozeleski
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Brendan Keating
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amein Al Ali
- Institute for Research and Medical Consultation, Imam Abdulrahman bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia.,Al-Omran Scientific Chair for hematological diseases, King Faisal University, Al Hassa, Saudi Arabia
| |
Collapse
|
69
|
Kumar V, Yadav AK, Kumar V, Bhansali A, Jha V. Uromodulin rs4293393 T>C variation is associated with kidney disease in patients with type 2 diabetes. Indian J Med Res 2018; 146:S15-S21. [PMID: 29578190 PMCID: PMC5890591 DOI: 10.4103/ijmr.ijmr_919_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Background & objectives Uromodulin, a UMOD gene encoded glycoprotein is synthesized exclusively in renal tubular cells and released into urine. Mutations lead to uromodulin misfolding and retention in the kidney, where it might stimulate cells of immune system to cause inflammation and progression of kidney disease. Genome-wide association studies (GWAS) have identified UMOD locus to be associated with hypertension and diabetic nephropathy (DN). In this study, we investigated the association between rs4293393 variation in UMOD gene and susceptibility to kidney disease in individuals with type 2 diabetes mellitus (T2DM). Methods A total of 646 individuals, 208 with T2DM without evidence of kidney disease (DM), 221 with DN and 217 healthy controls (HC) were genotyped for UMOD variant rs4293393T>C by restriction fragment length polymorphism. Serum uromodulin levels were quantified by enzyme-linked immunosorbent assay. Results A significant difference was found in genotype and allelic frequency among DM, DN and HC. TC+CC genotype and C allele were found more frequently in DN compared to HC (33.9 vs 23.0%, P=0.011 and 20.1 vs 12.9%, P=0.004, respectively). Compared to DM, C allele was found to be more frequent in individuals with DN (20.1 vs 14.7%, P=0.034). Those with DN had higher serum uromodulin levels compared to those with DM (P=0.001). Serum uromodulin levels showed a positive correlation with serum creatinine (r=0.431; P<0.001) and negative correlation with estimated glomerular filtration rate (r=-0.423; P<0.001). Interpretation & conclusions The frequency of UMOD rs4293393 variant with C allele was significantly higher in individuals with DN. UMOD rs4293393 T>C variation might have a bearing on susceptibility to nephropathy in north Indian individuals with type 2 diabetes.
Collapse
Affiliation(s)
- Vinod Kumar
- Department of Nephrology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Ashok Kumar Yadav
- Department of Nephrology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Vivek Kumar
- Department of Nephrology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Anil Bhansali
- Department of Endocrinology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Vivekanand Jha
- Department of Nephrology, Postgraduate Institute of Medical Education & Research, Chandigarh; George Institute for Global Health, New Delhi, India; James Martin Fellow, University of Oxford, Oxford, UK
| |
Collapse
|
70
|
Uromodulin associates with cardiorenal function in patients with hypertension and cardiovascular disease. J Hypertens 2018; 35:2053-2058. [PMID: 28598953 DOI: 10.1097/hjh.0000000000001432] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Common genetic variants in the gene encoding uromodulin (UMOD) have been associated with renal function, blood pressure (BP) and hypertension. We investigated the associations between an important single nucleotide polymorphism (SNP) in UMOD, that is rs12917707-G>T, and estimated glomerular filtration rate (eGFR), BP and cardiac organ damage as determined by echocardiography in patients with arterial hypertension. METHODS A cohort of 1218 treated high-risk patients (mean age 58.5 years, 83% men) with documented cardiovascular disease (81% with coronary heart disease) was analysed. RESULTS The mean values for 24-h SBP and DBP were 124.7 ± 14.7 and 73.9 ± 9.4 mmHg; mean eGFR was 77.5 ± 18.3 ml/min per 1.73 m, mean left ventricular ejection fraction was 59.3 ± 9.9% and mean left ventricular mass index in men and women was 53.9 ± 23.2 and 54.9 ± 23.7 g/m with 50.4% of patients having left ventricular hypertrophy. A significant association between rs12917707 and eGFR was observed with T-allele carriers showing significantly higher eGFR values (+2.6 ml/min per 1.73 m, P = 0.006) than noncarriers. This SNP associated also with left atrial diameter (P = 0.007); homozygous carriers of the T-allele had smaller left atrial diameter (-1.5 mm) than other genotype groups (P = 0.040). No significant associations between rs12917707 and other cardiac or BP phenotypes were observed. CONCLUSIONS These findings extend the previously documented role of UMOD for renal function also to treated high-risk patients with arterial hypertension and reveal a novel association with left atrial remodelling and thus a potential cardiorenal link modulated by UMOD.
Collapse
|
71
|
Lee J, Lee Y, Park B, Won S, Han JS, Heo NJ. Genome-wide association analysis identifies multiple loci associated with kidney disease-related traits in Korean populations. PLoS One 2018; 13:e0194044. [PMID: 29558500 PMCID: PMC5860731 DOI: 10.1371/journal.pone.0194044] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/25/2018] [Indexed: 12/19/2022] Open
Abstract
Chronic kidney disease (CKD) is an important social health problem characterized by a decrease in the kidney glomerular filtration rate (GFR). In this study, we analyzed genome-wide association studies for kidney disease-related traits using data from a Korean adult health screening cohort comprising 7,064 participants. Kidney disease-related traits analyzed include blood urea nitrogen (BUN), serum creatinine, estimated GFR, and uric acid levels. We detected two genetic loci (SLC14A2 and an intergenic region) and 8 single nucleotide polymorphisms (SNPs) associated with BUN, 3 genetic loci (BCAS3, C17orf82, ALDH2) and 6 SNPs associated with serum creatinine, 3 genetic loci (BCAS3, C17orf82/TBX2, LRP2) and 7 SNPs associated with GFR, and 14 genetic loci (3 in ABCG2/PKD2, 2 in SLC2A9, 3 in intergenic regions on chromosome 4; OTUB1, NRXN2/SLC22A12, CDC42BPG, RPS6KA4, SLC22A9, and MAP4K2 on chromosome 11) and 84 SNPs associated with uric acid levels. By comparing significant genetic loci associated with serum creatinine levels and GFR, rs9895661 in BCAS3 and rs757608 in C17orf82 were simultaneously associated with both traits. The SNPs rs11710227 in intergenic regions on chromosome 3 showing significant association with BUN is newly discovered. Genetic variations of multiple gene loci are associated with kidney disease-related traits, and differences in associations between kidney disease-related traits and genetic variation are dependent on the population. The meanings of the mutations identified in this study will need to be reaffirmed in other population groups in the future.
Collapse
Affiliation(s)
- Jeonghwan Lee
- Department of Internal Medicine, Hallym University Hangang Sacred Heart Hospital, Seoul, Korea
| | - Young Lee
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, Korea
| | - Boram Park
- Department of Public Health Science, Seoul National University, Seoul, Korea
| | - Sungho Won
- Department of Public Health Science, Seoul National University, Seoul, Korea
- Interdisciplinary Program of Bioinformatics, Seoul National University, Seoul, Korea
- Institute of Health and Environment, Seoul National University, Seoul, Korea
| | - Jin Suk Han
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Nam Ju Heo
- Division of Nephrology, Department of Internal Medicine, Healthcare System Gangnam Center, Seoul National University Hospital, Seoul, Korea
- * E-mail:
| |
Collapse
|
72
|
Prokop JW, Yeo NC, Ottmann C, Chhetri SB, Florus KL, Ross EJ, Sosonkina N, Link BA, Freedman BI, Coppola CJ, McDermott-Roe C, Leysen S, Milroy LG, Meijer FA, Geurts AM, Rauscher FJ, Ramaker R, Flister MJ, Jacob HJ, Mendenhall EM, Lazar J. Characterization of Coding/Noncoding Variants for SHROOM3 in Patients with CKD. J Am Soc Nephrol 2018; 29:1525-1535. [PMID: 29476007 DOI: 10.1681/asn.2017080856] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 01/19/2018] [Indexed: 12/16/2022] Open
Abstract
Background Interpreting genetic variants is one of the greatest challenges impeding analysis of rapidly increasing volumes of genomic data from patients. For example, SHROOM3 is an associated risk gene for CKD, yet causative mechanism(s) of SHROOM3 allele(s) are unknown.Methods We used our analytic pipeline that integrates genetic, computational, biochemical, CRISPR/Cas9 editing, molecular, and physiologic data to characterize coding and noncoding variants to study the human SHROOM3 risk locus for CKD.Results We identified a novel SHROOM3 transcriptional start site, which results in a shorter isoform lacking the PDZ domain and is regulated by a common noncoding sequence variant associated with CKD (rs17319721, allele frequency: 0.35). This variant disrupted allele binding to the transcription factor TCF7L2 in podocyte cell nuclear extracts and altered transcription levels of SHROOM3 in cultured cells, potentially through the loss of repressive looping between rs17319721 and the novel start site. Although common variant mechanisms are of high utility, sequencing is beginning to identify rare variants involved in disease; therefore, we used our biophysical tools to analyze an average of 112,849 individual human genome sequences for rare SHROOM3 missense variants, revealing 35 high-effect variants. The high-effect alleles include a coding variant (P1244L) previously associated with CKD (P=0.01, odds ratio=7.95; 95% CI, 1.53 to 41.46) that we find to be present in East Asian individuals at an allele frequency of 0.0027. We determined that P1244L attenuates the interaction of SHROOM3 with 14-3-3, suggesting alterations to the Hippo pathway, a known mediator of CKD.Conclusions These data demonstrate multiple new SHROOM3-dependent genetic/molecular mechanisms that likely affect CKD.
Collapse
Affiliation(s)
- Jeremy W Prokop
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama; .,Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, Michigan
| | - Nan Cher Yeo
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
| | - Christian Ottmann
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands.,Department of Chemistry, University of Duisburg-Essen, Essen, Germany
| | - Surya B Chhetri
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama.,Department of Biological Sciences, The University of Alabama in Huntsville, Huntsville, Alabama
| | - Kacie L Florus
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama
| | - Emily J Ross
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama.,Department of Chemical and Physical Biology, Vanderbilt University, Nashville, Tennessee
| | | | - Brian A Link
- Department of Cell Biology, Neurobiology and Anatomy and
| | - Barry I Freedman
- Section on Nephrology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina; and
| | - Candice J Coppola
- Department of Biological Sciences, The University of Alabama in Huntsville, Huntsville, Alabama
| | - Chris McDermott-Roe
- Department of Physiology, Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Seppe Leysen
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Lech-Gustav Milroy
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Femke A Meijer
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Aron M Geurts
- Department of Physiology, Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Frank J Rauscher
- Gene Expression & Regulation Program, Wistar Institute, Philadelphia, Pennsylvania
| | - Ryne Ramaker
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama
| | - Michael J Flister
- Department of Physiology, Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Howard J Jacob
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama
| | - Eric M Mendenhall
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama.,Department of Biological Sciences, The University of Alabama in Huntsville, Huntsville, Alabama
| | - Jozef Lazar
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama;
| |
Collapse
|
73
|
Xin G, Chen R, Zhang X. Identification of key microRNAs, transcription factors and genes associated with congenital obstructive nephropathy in a mouse model of megabladder. Gene 2018; 650:77-85. [PMID: 29410288 DOI: 10.1016/j.gene.2018.01.063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 11/21/2017] [Accepted: 01/17/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The present study aimed to investigate the molecular mechanism underlying congenital obstructive nephropathy (CON). METHODS The microarray dataset GSE70879 was downloaded from the Gene Expression Omnibus, including 3 kidney samples of megabladder mice and 4 control kidneys. Using this dataset, differentially expressed miRNAs (DEMs) were identified between the kidney samples from megabladder mice and controls, followed by identification of the target genes for these DEMs and construction of a DEM and target gene interaction network. Additionally, the target genes were subjected to Gene Ontology and pathway enrichment analyses, and were used for construction of a protein-protein interaction (PPI) network. Finally, regulatory networks were constructed to analyze transcription factors for the key miRNAs. RESULTS From 17 DEMs identified between kidney samples of megabladder mice and controls, 3 key miRNAs were screened, including mmu-miR-150-5p, mmu-miR-374b-5p and mmu-miR-126a-5p. The regulatory networks identified vascular endothelial growth factor A (Vegfa) as the common target gene of mmu-miR-150-5p and five transcription factors, including nuclear receptor subfamily 4, group A, member 2 (Nr4a2), Jun dimerisation protein 2 (Jdp2), Kruppel-like factor 6 (Klf6), Neurexophilin-3 (Nxph3) and RNA binding motif protein 17 (Rbm17). The gene encoding phosphatase and tensin homolog (Pten) was found to be co-regulated by mmu-miR-374b-5p and high mobility group protein A1 (Hmga1), whereas the kirsten rat sarcoma viral oncogene (Kras) was identified as a common target gene of mmu-miR-126a-5p and paired box 6 (Pax6). CONCLUSIONS In summary, the above-listed key miRNAs, transcription factors and key genes may be involved in the development of CON.
Collapse
Affiliation(s)
- Guangda Xin
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Rui Chen
- Department of Pediatrics, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Xiaofei Zhang
- Department of Pediatrics, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| |
Collapse
|
74
|
Ebbels TMD, Rodriguez-Martinez A, Dumas ME, Keun HC. Advances in Computational Analysis of Metabolomic NMR Data. NMR-BASED METABOLOMICS 2018. [DOI: 10.1039/9781782627937-00310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
In this chapter we discuss some of the more recent developments in preprocessing and statistical analysis of NMR spectra in metabolomics. Bayesian methods for analyzing NMR spectra are summarized and we describe one particular approach, BATMAN, in more detail. We consider techniques based on statistical associations, such as correlation spectroscopy (e.g. STOCSY and recent variants), as well as approaches that model the associations as a network and how these change under different biological conditions. The link between metabolism and genotype is explored by looking at metabolic GWAS and related techniques. Finally, we describe the relevance and current status of data standards for NMR metabolomics.
Collapse
Affiliation(s)
- Timothy M. D. Ebbels
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London London SW7 2AZ UK
| | - Andrea Rodriguez-Martinez
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London London SW7 2AZ UK
| | - Marc-Emmanuel Dumas
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London London SW7 2AZ UK
| | - Hector C. Keun
- Division of Cancer, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London London W12 0NN UK
| |
Collapse
|
75
|
Limou S, Vince N, Parsa A. Lessons from CKD-Related Genetic Association Studies-Moving Forward. Clin J Am Soc Nephrol 2018; 13:140-152. [PMID: 29242368 PMCID: PMC5753320 DOI: 10.2215/cjn.09030817] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Over the past decade, genetic association studies have uncovered numerous determinants of kidney function in the general, diabetic, hypertensive, CKD, ESRD, and GN-based study populations (e.g., IgA nephropathy, membranous nephropathy, FSGS). These studies have led to numerous novel and unanticipated findings, which are helping improve our understanding of factors and pathways affecting both normal and pathologic kidney function. In this review, we report on major discoveries and advances resulting from this rapidly progressing research domain. We also predict some of the next steps the nephrology community should embrace to accelerate the identification of genetic and molecular processes leading to kidney dysfunction, pathophysiologically based disease subgroups, and specific therapeutic targets, as we attempt to transition toward a more precision-based medicine approach.
Collapse
Affiliation(s)
- Sophie Limou
- Centre de Recherche en Transplantation et Immunologie Unité Mixte de Recherche 1064, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Nantes, Nantes, France
- Institut de Transplantation Urologie et Néphrologie, Centre Hospitalier Universitaire Nantes, Nantes, France
- Ecole Centrale de Nantes, Nantes, France
- Basic Science Program, Basic Research Laboratory, National Cancer Institute/National Institutes of Health, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, Maryland
| | - Nicolas Vince
- Centre de Recherche en Transplantation et Immunologie Unité Mixte de Recherche 1064, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Nantes, Nantes, France
- Institut de Transplantation Urologie et Néphrologie, Centre Hospitalier Universitaire Nantes, Nantes, France
| | - Afshin Parsa
- Division of Nephrology, University of Maryland School of Medicine, Baltimore, Maryland; and
- Department of Medicine, Baltimore VA Medical Center, Baltimore, Maryland
| |
Collapse
|
76
|
Chu CP, Hokamp JA, Cianciolo RE, Dabney AR, Brinkmeyer-Langford C, Lees GE, Nabity MB. RNA-seq of serial kidney biopsies obtained during progression of chronic kidney disease from dogs with X-linked hereditary nephropathy. Sci Rep 2017; 7:16776. [PMID: 29196624 PMCID: PMC5711945 DOI: 10.1038/s41598-017-16603-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 10/25/2017] [Indexed: 12/24/2022] Open
Abstract
Dogs with X-linked hereditary nephropathy (XLHN) have a glomerular basement membrane defect that leads to progressive juvenile-onset renal failure. Their disease is analogous to Alport syndrome in humans, and they also serve as a good model of progressive chronic kidney disease (CKD). However, the gene expression profile that affects progression in this disease has only been partially characterized. To help fill this gap, we used RNA sequencing to identify differentially expressed genes (DEGs), over-represented pathways, and upstream regulators that contribute to kidney disease progression. Total RNA from kidney biopsies was isolated at 3 clinical time points from 3 males with rapidly-progressing CKD, 3 males with slowly-progressing CKD, and 2 age-matched controls. We identified 70 DEGs by comparing rapid and slow groups at specific time points. Based on time course analysis, 1,947 DEGs were identified over the 3 time points revealing upregulation of inflammatory pathways: integrin signaling, T cell activation, and chemokine and cytokine signaling pathways. T cell infiltration was verified by immunohistochemistry. TGF-β1 was identified as the primary upstream regulator. These results provide new insights into the underlying molecular mechanisms of disease progression in XLHN, and the identified DEGs can be potential biomarkers and therapeutic targets translatable to all CKDs.
Collapse
Affiliation(s)
- Candice P Chu
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Jessica A Hokamp
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Rachel E Cianciolo
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Alan R Dabney
- Department of Statistics, College of Science, Texas A&M University, College Station, TX, USA
| | - Candice Brinkmeyer-Langford
- Department of Veterinary Integrative Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - George E Lees
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Mary B Nabity
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
77
|
Devuyst O, Pattaro C. The UMOD Locus: Insights into the Pathogenesis and Prognosis of Kidney Disease. J Am Soc Nephrol 2017; 29:713-726. [PMID: 29180396 DOI: 10.1681/asn.2017070716] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The identification of genetic factors associated with kidney disease has the potential to provide critical insights into disease mechanisms. Genome-wide association studies have uncovered genomic regions associated with renal function metrics and risk of CKD. UMOD is among the most outstanding loci associated with CKD in the general population, because it has a large effect on eGFR and CKD risk that is consistent across different ethnic groups. The relevance of UMOD for CKD is clear, because the encoded protein, uromodulin (Tamm-Horsfall protein), is exclusively produced by the kidney tubule and has specific biochemical properties that mediate important functions in the kidney and urine. Rare mutations in UMOD are the major cause of autosomal dominant tubulointerstitial kidney disease, a condition that leads to CKD and ESRD. In this brief review, we use the UMOD paradigm to describe how population genetic studies can yield insight into the pathogenesis and prognosis of kidney diseases.
Collapse
Affiliation(s)
- Olivier Devuyst
- Institute of Physiology, University of Zurich, Zurich, Switzerland; and
| | - Cristian Pattaro
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| |
Collapse
|
78
|
Piras D, Zoledziewska M, Cucca F, Pani A. Genome-Wide Analysis Studies and Chronic Kidney Disease. KIDNEY DISEASES 2017; 3:106-110. [PMID: 29344505 DOI: 10.1159/000481886] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 10/02/2017] [Indexed: 11/19/2022]
Abstract
In recent years, the very high worldwide prevalence of chronic kidney disease (CKD) has led some authors to talk of an "epidemic." The progression of CKD varies considerably among individuals despite similar aetiologies, optimal blood pressure, and glycaemic control. Over the last decade, through genome-wide association studies (GWAS), more than 50 genetic loci have been identified in association with CKD. Understanding the genetic basis of CKD could provide a better knowledge of the biology of the involved pathways, thus potentially leading to novel tools for the diagnosis, prevention, and therapy of CKD. In this review, we will analyse the role of GWAS in the study of CKD.
Collapse
Affiliation(s)
- Doloretta Piras
- Divisione di Nefrologia e Dialisi, Azienda Ospedaliera G. Brotzu, Cagliari, Italy
| | | | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica (IRGB-CNR), Cagliari, Italy.,Università degli Studi di Sassari, Sassari, Italy
| | - Antonello Pani
- Divisione di Nefrologia e Dialisi, Azienda Ospedaliera G. Brotzu, Cagliari, Italy
| |
Collapse
|
79
|
Identification of CDC42BPG as a novel susceptibility locus for hyperuricemia in a Japanese population. Mol Genet Genomics 2017; 293:371-379. [PMID: 29124443 PMCID: PMC5854719 DOI: 10.1007/s00438-017-1394-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/04/2017] [Indexed: 12/29/2022]
Abstract
Chronic kidney disease and hyperuricemia are serious global health problems. Recent genome-wide association studies have identified various genetic variants related to these disorders. However, most studies have been conducted in a cross-sectional manner. To identify novel susceptibility loci for chronic kidney disease or hyperuricemia, we performed longitudinal exome-wide association studies (EWASs), using ~ 244,000 genetic variants and clinical data of Japanese individuals who had undergone annual health checkups for several years. After establishing quality controls, the association of renal function-related traits in 5648 subjects (excluding patients with dialysis and population outliers) with 24,579 single nucleotide variants (SNVs) for three genetic models (P < 3.39 × 10− 7) was tested using generalized estimating equation models. The longitudinal EWASs revealed novel relations of five SNVs to renal function-related traits. Cross-sectional data for renal function-related traits in 7699 Japanese subjects were examined in a replication study. Among the five SNVs, rs55975541 in CDC42BPG was significantly (P < 4.90 × 10− 4) related to the serum concentration of uric acid in the replication cohort. We also examined the SNVs detected in our longitudinal EWASs with the information on P values in GKDGEN meta-analysis data. Four SNVs in SLC15A2 were significantly associated with the estimated glomerular filtration rate in European ancestry populations, although these SNVs were related to the serum concentration of uric acid with borderline significance in our longitudinal EWASs. Our findings indicate that CDC42BPG may be a novel susceptibility locus for hyperuricemia.
Collapse
|
80
|
Brandt MM, Meddens CA, Louzao-Martinez L, van den Dungen NAM, Lansu NR, Nieuwenhuis EES, Duncker DJ, Verhaar MC, Joles JA, Mokry M, Cheng C. Chromatin Conformation Links Distal Target Genes to CKD Loci. J Am Soc Nephrol 2017; 29:462-476. [PMID: 29093029 DOI: 10.1681/asn.2016080875] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 09/09/2017] [Indexed: 11/03/2022] Open
Abstract
Genome-wide association studies (GWASs) have identified many genetic risk factors for CKD. However, linking common variants to genes that are causal for CKD etiology remains challenging. By adapting self-transcribing active regulatory region sequencing, we evaluated the effect of genetic variation on DNA regulatory elements (DREs). Variants in linkage with the CKD-associated single-nucleotide polymorphism rs11959928 were shown to affect DRE function, illustrating that genes regulated by DREs colocalizing with CKD-associated variation can be dysregulated and therefore, considered as CKD candidate genes. To identify target genes of these DREs, we used circular chromosome conformation capture (4C) sequencing on glomerular endothelial cells and renal tubular epithelial cells. Our 4C analyses revealed interactions of CKD-associated susceptibility regions with the transcriptional start sites of 304 target genes. Overlap with multiple databases confirmed that many of these target genes are involved in kidney homeostasis. Expression quantitative trait loci analysis revealed that mRNA levels of many target genes are genotype dependent. Pathway analyses showed that target genes were enriched in processes crucial for renal function, identifying dysregulated geranylgeranyl diphosphate biosynthesis as a potential disease mechanism. Overall, our data annotated multiple genes to previously reported CKD-associated single-nucleotide polymorphisms and provided evidence for interaction between these loci and target genes. This pipeline provides a novel technique for hypothesis generation and complements classic GWAS interpretation. Future studies are required to specify the implications of our dataset and further reveal the complex roles that common variants have in complex diseases, such as CKD.
Collapse
Affiliation(s)
- Maarten M Brandt
- Experimental Cardiology, Department of Cardiology, Thoraxcenter Erasmus University Medical Center, Rotterdam, The Netherlands; and
| | - Claartje A Meddens
- Department of Pediatrics, Wilhelmina Children's Hospital.,Regenerative Medicine Center Utrecht, Department of Pediatrics
| | - Laura Louzao-Martinez
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology
| | - Noortje A M van den Dungen
- Department of Cardiology, Division Heart and Lungs, and.,Epigenomics Facility, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nico R Lansu
- Department of Pediatrics, Wilhelmina Children's Hospital.,Regenerative Medicine Center Utrecht, Department of Pediatrics.,Epigenomics Facility, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Dirk J Duncker
- Experimental Cardiology, Department of Cardiology, Thoraxcenter Erasmus University Medical Center, Rotterdam, The Netherlands; and
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology
| | - Jaap A Joles
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology
| | - Michal Mokry
- Department of Pediatrics, Wilhelmina Children's Hospital.,Regenerative Medicine Center Utrecht, Department of Pediatrics.,Epigenomics Facility, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Caroline Cheng
- Experimental Cardiology, Department of Cardiology, Thoraxcenter Erasmus University Medical Center, Rotterdam, The Netherlands; and .,Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology
| |
Collapse
|
81
|
Ainsworth HC, Langefeld CD, Freedman BI. Genetic epidemiology in kidney disease. Nephrol Dial Transplant 2017; 32:ii159-ii169. [PMID: 28201750 DOI: 10.1093/ndt/gfw270] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Accepted: 06/04/2016] [Indexed: 12/20/2022] Open
Abstract
Familial aggregation of chronic kidney disease and its component phenotypes-reduced glomerular filtration rate, proteinuria and renal histologic changes-has long been recognized. Rates of severe kidney disease are also known to differ markedly between populations based on ancestry. These epidemiologic observations support the existence of nephropathy susceptibility genes. Several molecular genetic technologies are now available to identify causative loci. The present article summarizes available strategies useful for identifying nephropathy susceptibility genes, including candidate gene association, family-based linkage, genome-wide association and admixture mapping (mapping by admixture linkage disequilibrium) approaches. Examples of loci detected using these techniques are provided. Epigenetic studies and future directions are also discussed. The identification of nephropathy susceptibility genes, coupled with modifiable environmental triggers impacting their function, is likely to improve risk prediction and transform care. Development of novel therapies to prevent progression of kidney disease will follow.
Collapse
Affiliation(s)
- Hannah C Ainsworth
- Department of Biostatistical Sciences, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Carl D Langefeld
- Department of Biostatistical Sciences, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Barry I Freedman
- Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Internal Medicine, Section on Nephrology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| |
Collapse
|
82
|
Devuyst O, Olinger E, Rampoldi L. Uromodulin: from physiology to rare and complex kidney disorders. Nat Rev Nephrol 2017; 13:525-544. [PMID: 28781372 DOI: 10.1038/nrneph.2017.101] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Uromodulin (also known as Tamm-Horsfall protein) is exclusively produced in the kidney and is the most abundant protein in normal urine. The function of uromodulin remains elusive, but the available data suggest that this protein might regulate salt transport, protect against urinary tract infection and kidney stones, and have roles in kidney injury and innate immunity. Interest in uromodulin was boosted by genetic studies that reported involvement of the UMOD gene, which encodes uromodulin, in a spectrum of rare and common kidney diseases. Rare mutations in UMOD cause autosomal dominant tubulointerstitial kidney disease (ADTKD), which leads to chronic kidney disease (CKD). Moreover, genome-wide association studies have identified common variants in UMOD that are strongly associated with risk of CKD and also with hypertension and kidney stones in the general population. These findings have opened up a new field of kidney research. In this Review we summarize biochemical, physiological, genetic and pathological insights into the roles of uromodulin; the mechanisms by which UMOD mutations cause ADTKD, and the association of common UMOD variants with complex disorders.
Collapse
Affiliation(s)
- Olivier Devuyst
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Eric Olinger
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Luca Rampoldi
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| |
Collapse
|
83
|
Rodrigues AD, Taskar KS, Kusuhara H, Sugiyama Y. Endogenous Probes for Drug Transporters: Balancing Vision With Reality. Clin Pharmacol Ther 2017; 103:434-448. [DOI: 10.1002/cpt.749] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 05/04/2017] [Accepted: 05/15/2017] [Indexed: 12/17/2022]
Affiliation(s)
- AD Rodrigues
- Pharmacokinetics; Dynamics & Metabolism, Medicine Design, Pfizer Inc.; Groton Connecticut USA
| | - KS Taskar
- Mechanistic Safety and Disposition; IVIVT, GlaxoSmithKline; Ware Hertfordshire UK
| | - H Kusuhara
- Laboratory of Molecular Pharmacokinetics; Graduate School of Pharmaceutical Sciences, University of Tokyo; Tokyo Japan
| | - Y Sugiyama
- RIKEN Innovation Center; Research Cluster for Innovation; RIKEN Kanagawa Japan
| |
Collapse
|
84
|
Yamada Y, Sakuma J, Takeuchi I, Yasukochi Y, Kato K, Oguri M, Fujimaki T, Horibe H, Muramatsu M, Sawabe M, Fujiwara Y, Taniguchi Y, Obuchi S, Kawai H, Shinkai S, Mori S, Arai T, Tanaka M. Identification of C21orf59 and ATG2A as novel determinants of renal function-related traits in Japanese by exome-wide association studies. Oncotarget 2017; 8:45259-45273. [PMID: 28410202 PMCID: PMC5542184 DOI: 10.18632/oncotarget.16696] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 03/08/2017] [Indexed: 11/25/2022] Open
Abstract
We have performed exome-wide association studies to identify genetic variants that influence renal function-related traits or confer susceptibility to chronic kidney disease or hyperuricemia in Japanese. Exome-wide association studies for estimated glomerular filtration rate and the serum concentration of creatinine were performed with 12,565 individuals, that for the serum concentration of uric acid with 9934 individuals, and those for chronic kidney disease or hyperuricemia with 5161 individuals (3270 cases, 1891 controls) or 11,686 individuals (2045 cases, 9641 controls), respectively. The relation of genotypes of single nucleotide polymorphisms to estimated glomerular filtration rate or the serum concentrations of creatinine or uric acid was examined by linear regression analysis, and that of allele frequencies of single nucleotide polymorphisms to chronic kidney disease or hyperuricemia was examined with Fisher's exact test. The exome-wide association studies revealed that 25, seven, and six single nucleotide polymorphisms were significantly (P <1.21 × 10-6) associated with estimated glomerular filtration rate or the serum concentrations of creatinine or uric acid, respectively, and that 49 and 35 polymorphisms were significantly associated with chronic kidney disease or hyperuricemia, respectively. Subsequent multivariable logistic regression analysis with adjustment for covariates revealed that four and three single nucleotide polymorphisms were related (P < 0.05) to chronic kidney disease or hyperuricemia, respectively. Among polymorphisms identified in the present study, rs76974938 [C/T (D67N)] of C21orf59 and rs188780113 [G/A (R478C)] of ATG2A may be novel determinants of estimated glomerular filtration rate and chronic kidney disease or of the serum concentration of uric acid, respectively.
Collapse
Affiliation(s)
- Yoshiji Yamada
- Department of Human Functional Genomics, Advanced Science Research Promotion Center, Mie University, Tsu, Japan
- CREST, Japan Science and Technology Agency, Kawaguchi, Japan
| | - Jun Sakuma
- CREST, Japan Science and Technology Agency, Kawaguchi, Japan
- Computer Science Department, College of Information Science, University of Tsukuba, Tsukuba, Japan
- RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
| | - Ichiro Takeuchi
- CREST, Japan Science and Technology Agency, Kawaguchi, Japan
- RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
- Department of Computer Science, Nagoya Institute of Technology, Nagoya, Japan
| | - Yoshiki Yasukochi
- Department of Human Functional Genomics, Advanced Science Research Promotion Center, Mie University, Tsu, Japan
- CREST, Japan Science and Technology Agency, Kawaguchi, Japan
| | - Kimihiko Kato
- Department of Human Functional Genomics, Advanced Science Research Promotion Center, Mie University, Tsu, Japan
- Department of Internal Medicine, Meitoh Hospital, Nagoya, Japan
| | - Mitsutoshi Oguri
- Department of Human Functional Genomics, Advanced Science Research Promotion Center, Mie University, Tsu, Japan
- Department of Cardiology, Kasugai Municipal Hospital, Kasugai, Japan
| | - Tetsuo Fujimaki
- Department of Cardiovascular Medicine, Inabe General Hospital, Inabe, Japan
| | - Hideki Horibe
- Department of Cardiovascular Medicine, Gifu Prefectural Tajimi Hospital, Tajimi, Japan
| | - Masaaki Muramatsu
- Department of Molecular Epidemiology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Motoji Sawabe
- Section of Molecular Pathology, Graduate School of Health Care Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshinori Fujiwara
- Research Team for Social Participation and Community Health, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Yu Taniguchi
- Research Team for Social Participation and Community Health, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Shuichi Obuchi
- Research Team for Promoting Support System for Home Care, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Hisashi Kawai
- Research Team for Promoting Support System for Home Care, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Shoji Shinkai
- Research Team for Social Participation and Health Promotion, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Seijiro Mori
- Center for Promotion of Clinical Investigation, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Tomio Arai
- Department of Pathology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Masashi Tanaka
- Department of Clinical Laboratory, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| |
Collapse
|
85
|
van der Laan SW, Fall T, Soumaré A, Teumer A, Sedaghat S, Baumert J, Zabaneh D, van Setten J, Isgum I, Galesloot TE, Arpegård J, Amouyel P, Trompet S, Waldenberger M, Dörr M, Magnusson PK, Giedraitis V, Larsson A, Morris AP, Felix JF, Morrison AC, Franceschini N, Bis JC, Kavousi M, O'Donnell C, Drenos F, Tragante V, Munroe PB, Malik R, Dichgans M, Worrall BB, Erdmann J, Nelson CP, Samani NJ, Schunkert H, Marchini J, Patel RS, Hingorani AD, Lind L, Pedersen NL, de Graaf J, Kiemeney LALM, Baumeister SE, Franco OH, Hofman A, Uitterlinden AG, Koenig W, Meisinger C, Peters A, Thorand B, Jukema JW, Eriksen BO, Toft I, Wilsgaard T, Onland-Moret NC, van der Schouw YT, Debette S, Kumari M, Svensson P, van der Harst P, Kivimaki M, Keating BJ, Sattar N, Dehghan A, Reiner AP, Ingelsson E, den Ruijter HM, de Bakker PIW, Pasterkamp G, Ärnlöv J, Holmes MV, Asselbergs FW. Cystatin C and Cardiovascular Disease: A Mendelian Randomization Study. J Am Coll Cardiol 2017; 68:934-45. [PMID: 27561768 PMCID: PMC5451109 DOI: 10.1016/j.jacc.2016.05.092] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 05/12/2016] [Accepted: 05/18/2016] [Indexed: 01/09/2023]
Abstract
BACKGROUND Epidemiological studies show that high circulating cystatin C is associated with risk of cardiovascular disease (CVD), independent of creatinine-based renal function measurements. It is unclear whether this relationship is causal, arises from residual confounding, and/or is a consequence of reverse causation. OBJECTIVES The aim of this study was to use Mendelian randomization to investigate whether cystatin C is causally related to CVD in the general population. METHODS We incorporated participant data from 16 prospective cohorts (n = 76,481) with 37,126 measures of cystatin C and added genetic data from 43 studies (n = 252,216) with 63,292 CVD events. We used the common variant rs911119 in CST3 as an instrumental variable to investigate the causal role of cystatin C in CVD, including coronary heart disease, ischemic stroke, and heart failure. RESULTS Cystatin C concentrations were associated with CVD risk after adjusting for age, sex, and traditional risk factors (relative risk: 1.82 per doubling of cystatin C; 95% confidence interval [CI]: 1.56 to 2.13; p = 2.12 × 10−14). The minor allele of rs911119 was associated with decreased serum cystatin C (6.13% per allele; 95% CI: 5.75 to 6.50; p = 5.95 × 10−211), explaining 2.8% of the observed variation in cystatin C. Mendelian randomization analysis did not provide evidence for a causal role of cystatin C, with a causal relative risk for CVD of 1.00 per doubling cystatin C (95% CI: 0.82 to 1.22; p = 0.994), which was statistically different from the observational estimate (p = 1.6 × 10−5). A causal effect of cystatin C was not detected for any individual component of CVD. CONCLUSIONS Mendelian randomization analyses did not support a causal role of cystatin C in the etiology of CVD. As such, therapeutics targeted at lowering circulating cystatin C are unlikely to be effective in preventing CVD.
Collapse
Affiliation(s)
- Sander W van der Laan
- Laboratory of Experimental Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, the Netherlands.
| | - Tove Fall
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Aicha Soumaré
- INSERM U1219 Team Vintage, University of Bordeaux, Bordeaux, France
| | - Alexander Teumer
- Department SHIP-KEF, Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany; Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK, German Centre for Cardiovascular Research) partner site, Greifswald, Germany
| | - Sanaz Sedaghat
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jens Baumert
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Delilah Zabaneh
- Department of Genetics, Environment and Evolution, University College London, London, United Kingdom; Genetics Institute, University College London, London, United Kingdom
| | - Jessica van Setten
- Laboratory of Experimental Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ivana Isgum
- Image Sciences Institute, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Tessel E Galesloot
- Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Johannes Arpegård
- Department of Emergency Medicine, Karolinska University Hospital-Solna, Stockholm, Sweden; Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Philippe Amouyel
- INSERM, University of Lille, Lille, France; Institut Pasteur de Lille, Lille, France
| | - Stella Trompet
- Department of Cardiology C5-P, Leiden University Medical Center, Leiden, the Netherlands; Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Melanie Waldenberger
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; Research Unit of Molecular Epidemiology Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Marcus Dörr
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK, German Centre for Cardiovascular Research) partner site, Greifswald, Germany; Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Patrik K Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | | | - Anders Larsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Andrew P Morris
- Department of Biostatistics, University of Liverpool, Liverpool, United Kingdom; Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Janine F Felix
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Alanna C Morrison
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center, Houston, Texas
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Christopher O'Donnell
- Department of Cardiology, Boston Veterans Administration Healthcare, West Roxbury, Massachusetts; National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, Massachusetts
| | - Fotios Drenos
- Centre for Cardiovascular Genetics, Institute of Cardiovascular Sciences; University College London, London, United Kingdom; MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Vinicius Tragante
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Patricia B Munroe
- National Institute for Health Research Cardiovascular Biomedical Research Unit, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Rainer Malik
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Bradford B Worrall
- Departments of Neurology and Health Evaluation Sciences, University of Virginia, Charlottesville, Virginia
| | - Jeanette Erdmann
- Institute for Integrative and Experimental Genomics, University of Lübeck, Lübeck, Germany
| | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester, British Heart Foundation Cardiovascular Research Centre, Glenfield Hospital, Leicester, United Kingdom; National Institute for Health Research Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, British Heart Foundation Cardiovascular Research Centre, Glenfield Hospital, Leicester, United Kingdom; National Institute for Health Research Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany; DZHK, German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Munich, Germany
| | - Jonathan Marchini
- Department of Statistics, University of Oxford, Oxford, United Kingdom
| | - Riyaz S Patel
- The Genetic Epidemiology Research Group, Institute of Cardiovascular Science, University College London, London, United Kingdom; Bart's Heart Centre, London, United Kingdom; Farr Institute of Health Informatics, University College London, London, United Kingdom
| | - Aroon D Hingorani
- The Genetic Epidemiology Research Group, Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jacqueline de Graaf
- Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Lambertus A L M Kiemeney
- Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Sebastian E Baumeister
- Department SHIP-KEF, Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany; Institute for Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
| | - Oscar H Franco
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Wolfgang Koenig
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK, German Centre for Cardiovascular Research) partner site, Greifswald, Germany; Deutsches Herzzentrum München, Technische Universität München, Munich, Germany; Department of Internal Medicine II-Cardiology, University of Ulm Medical Center, Ulm, Germany
| | - Christa Meisinger
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Annette Peters
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK, German Centre for Cardiovascular Research) partner site, Greifswald, Germany; Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Barbara Thorand
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - J Wouter Jukema
- Department of Cardiology C5-P, Leiden University Medical Center, Leiden, the Netherlands; Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, the Netherlands
| | - Bjørn Odvar Eriksen
- Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway; Section of Nephrology, University Hospital of North Norway, Tromsø, Norway
| | - Ingrid Toft
- Section of Nephrology, University Hospital of North Norway, Tromsø, Norway
| | - Tom Wilsgaard
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - N Charlotte Onland-Moret
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Yvonne T van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Meena Kumari
- Biological and Social Epidemiology, Institute for Social and Economic Research, University of Essex, Essex, United Kingdom
| | - Per Svensson
- Department of Emergency Medicine, Karolinska University Hospital-Solna, Stockholm, Sweden; Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Pim van der Harst
- Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, the Netherlands; Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Mika Kivimaki
- Department of Epidemiology and Public Health, University College London, London, United Kingdom
| | - Brendan J Keating
- Department of Surgery, Division of Transplantation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Abbas Dehghan
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Alex P Reiner
- Department of Epidemiology, University of Washington, Seattle, Washington
| | - Erik Ingelsson
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California
| | - Hester M den Ruijter
- Laboratory of Experimental Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Paul I W de Bakker
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Medical Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Gerard Pasterkamp
- Laboratory of Experimental Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, the Netherlands; Laboratory of Clinical Chemistry and Hematology, Division of Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Johan Ärnlöv
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Michael V Holmes
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom.
| | - Folkert W Asselbergs
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht, the Netherlands; Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, the Netherlands; Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, United Kingdom.
| |
Collapse
|
86
|
Abstract
Acute kidney injury (AKI) is characterized by an acute decline in renal function and is associated to increased mortality rate, hospitalization time, and total health-related costs. The severity of this ‘fearsome’ clinical complication might depend on, or even be worsened by, the late detection of AKI, when the diagnosis is based on the elevation of serum creatinine (SCr). For these reasons, in recent years a great number of new tools, biomarkers and predictive models have been proposed to clinicians in order to improve diagnosis and prevent the development of AKI. The purpose of this narrative paper is to review the current state of the art in prediction and early detection of AKI and outline future challenges.
Collapse
Affiliation(s)
- Simona Pozzoli
- Chair of Nephrology - IRCCS San Raffaele Scientific Institute, Genomics of Renal Diseases and Hypertension Unit, Università Vita Salute San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Marco Simonini
- Chair of Nephrology - IRCCS San Raffaele Scientific Institute, Genomics of Renal Diseases and Hypertension Unit, Università Vita Salute San Raffaele, Via Olgettina 60, 20132, Milan, Italy.
| | - Paolo Manunta
- Chair of Nephrology - IRCCS San Raffaele Scientific Institute, Genomics of Renal Diseases and Hypertension Unit, Università Vita Salute San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| |
Collapse
|
87
|
Bailie C, Kilner J, Maxwell AP, McKnight AJ. Development of next generation sequencing panel for UMOD and association with kidney disease. PLoS One 2017; 12:e0178321. [PMID: 28609449 PMCID: PMC5469457 DOI: 10.1371/journal.pone.0178321] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 05/11/2017] [Indexed: 11/19/2022] Open
Abstract
Chronic kidney disease (CKD) has a prevalence of approximately 10% in adult populations. CKD can progress to end-stage renal disease (ESRD) and this is usually fatal unless some form of renal replacement therapy (chronic dialysis or renal transplantation) is provided. There is an inherited predisposition to CKD with several genetic risk markers now identified. The UMOD gene has been associated with CKD of varying aetiologies. An AmpliSeq next generation sequencing panel was developed to facilitate comprehensive sequencing of the UMOD gene, covering exonic and regulatory regions. SNPs and CpG sites in the genomic region encompassing UMOD were evaluated for association with CKD in two studies; the UK Wellcome Trust Case-Control 3 Renal Transplant Dysfunction Study (n = 1088) and UK-ROI GENIE GWAS (n = 1726). A technological comparison of two Ion Torrent machines revealed 100% allele call concordance between S5 XL™ and PGM™ machines. One SNP (rs183962941), located in a non-coding region of UMOD, was nominally associated with ESRD (p = 0.008). No association was identified between UMOD variants and estimated glomerular filtration rate. Analysis of methylation data for over 480,000 CpG sites revealed differential methylation patterns within UMOD, the most significant of these was cg03140788 p = 3.7 x 10-10.
Collapse
Affiliation(s)
- Caitlin Bailie
- Nephrology Research, Centre for Public Health, Queen’s University of Belfast, Belfast City Hospital, Belfast, Northern Ireland
| | - Jill Kilner
- Nephrology Research, Centre for Public Health, Queen’s University of Belfast, Belfast City Hospital, Belfast, Northern Ireland
| | - Alexander P. Maxwell
- Nephrology Research, Centre for Public Health, Queen’s University of Belfast, Belfast City Hospital, Belfast, Northern Ireland
| | - Amy Jayne McKnight
- Nephrology Research, Centre for Public Health, Queen’s University of Belfast, Belfast City Hospital, Belfast, Northern Ireland
| |
Collapse
|
88
|
1000 Genomes-based meta-analysis identifies 10 novel loci for kidney function. Sci Rep 2017; 7:45040. [PMID: 28452372 PMCID: PMC5408227 DOI: 10.1038/srep45040] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 02/20/2017] [Indexed: 12/31/2022] Open
Abstract
HapMap imputed genome-wide association studies (GWAS) have revealed >50 loci at which common variants with minor allele frequency >5% are associated with kidney function. GWAS using more complete reference sets for imputation, such as those from The 1000 Genomes project, promise to identify novel loci that have been missed by previous efforts. To investigate the value of such a more complete variant catalog, we conducted a GWAS meta-analysis of kidney function based on the estimated glomerular filtration rate (eGFR) in 110,517 European ancestry participants using 1000 Genomes imputed data. We identified 10 novel loci with p-value < 5 × 10-8 previously missed by HapMap-based GWAS. Six of these loci (HOXD8, ARL15, PIK3R1, EYA4, ASTN2, and EPB41L3) are tagged by common SNPs unique to the 1000 Genomes reference panel. Using pathway analysis, we identified 39 significant (FDR < 0.05) genes and 127 significantly (FDR < 0.05) enriched gene sets, which were missed by our previous analyses. Among those, the 10 identified novel genes are part of pathways of kidney development, carbohydrate metabolism, cardiac septum development and glucose metabolism. These results highlight the utility of re-imputing from denser reference panels, until whole-genome sequencing becomes feasible in large samples.
Collapse
|
89
|
Abdel-Hady Algharably E, Beige J, Kreutz R, Bolbrinker J. Effect of UMOD genotype on long-term graft survival after kidney transplantation in patients treated with cyclosporine-based therapy. THE PHARMACOGENOMICS JOURNAL 2017; 18:227-231. [PMID: 28418009 DOI: 10.1038/tpj.2017.14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/31/2017] [Accepted: 02/14/2017] [Indexed: 11/09/2022]
Abstract
The genetic rs12917707-G>T variant in uromodulin (UMOD) has been associated with renal function, chronic kidney disease and hypertension with the minor T-allele showing a protective effect. Hypertension and nephrotoxicity are adverse effects of chronic cyclosporine treatment. We tested whether UMOD rs12917707-T in donor kidneys associates with long-term graft survival in 393 Caucasian patients with stable graft function for more than 10 weeks after kidney transplantation treated with a cyclosporine-based maintenance therapy (mean graft survival 9 years). Presence of the donor T-allele had no effect on blood pressure, serum creatinine 1 year after transplantation, and on number of acute graft rejections during the first year. No significant effect on overall graft survival was observed in Kaplan-Meier analysis (P=0.65). In death-censored adjusted multivariate analysis, presence of donor T-allele associated with a significant lower hazard ratio of 0.67 (95% confidence interval: 0.46-0.97, P=0.05) for graft loss. This protective effect of the donor T-allele on graft loss observed in multivariate adjusted analysis justifies further investigations including patients treated with similar or other immunosuppressive regimens.
Collapse
Affiliation(s)
- E Abdel-Hady Algharably
- Institute of Clinical Pharmacology and Toxicology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - J Beige
- Faculty of Medicine, Martin-Luther-University Halle/Wittenberg, Halle, Germany.,Department of Medicine Nephrology, Klinikum St. Georg, Leipzig, Germany
| | - R Kreutz
- Institute of Clinical Pharmacology and Toxicology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - J Bolbrinker
- Institute of Clinical Pharmacology and Toxicology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
90
|
Nolte IM, van der Most PJ, Alizadeh BZ, de Bakker PI, Boezen HM, Bruinenberg M, Franke L, van der Harst P, Navis G, Postma DS, Rots MG, Stolk RP, Swertz MA, Wolffenbuttel BH, Wijmenga C, Snieder H. Missing heritability: is the gap closing? An analysis of 32 complex traits in the Lifelines Cohort Study. Eur J Hum Genet 2017; 25:877-885. [PMID: 28401901 DOI: 10.1038/ejhg.2017.50] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 02/03/2017] [Accepted: 02/14/2017] [Indexed: 01/08/2023] Open
Abstract
Despite the recent explosive rise in number of genetic markers for complex disease traits identified in genome-wide association studies, there is still a large gap between the known heritability of these traits and the part explained by these markers. To gauge whether this 'heritability gap' is closing, we first identified genome-wide significant SNPs from the literature and performed replication analyses for 32 highly relevant traits from five broad disease areas in 13 436 subjects of the Lifelines Cohort. Next, we calculated the variance explained by multi-SNP genetic risk scores (GRSs) for each trait, and compared it to their broad- and narrow-sense heritabilities captured by all common SNPs. The majority of all previously-associated SNPs (median=75%) were significantly associated with their respective traits. All GRSs were significant, with unweighted GRSs generally explaining less phenotypic variance than weighted GRSs, for which the explained variance was highest for height (15.5%) and varied between 0.02 and 6.7% for the other traits. Broad-sense common-SNP heritability estimates were significant for all traits, with the additive effect of common SNPs explaining 48.9% of the variance for height and between 5.6 and 39.2% for the other traits. Dominance effects were uniformly small (0-1.5%) and not significant. On average, the variance explained by the weighted GRSs accounted for only 10.7% of the common-SNP heritability of the 32 traits. These results indicate that GRSs may not yet be ready for accurate personalized prediction of complex disease traits limiting widespread adoption in clinical practice.
Collapse
Affiliation(s)
- Ilja M Nolte
- Unit of Genetic Epidemiology and Bioinformatics, Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter J van der Most
- Unit of Genetic Epidemiology and Bioinformatics, Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Behrooz Z Alizadeh
- Unit of Genetic Epidemiology and Bioinformatics, Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Paul Iw de Bakker
- Department of Medical Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - H Marike Boezen
- Unit of Genetic Epidemiology and Bioinformatics, Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Lude Franke
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gerjan Navis
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dirkje S Postma
- Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marianne G Rots
- Department of Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ronald P Stolk
- Unit of Genetic Epidemiology and Bioinformatics, Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Morris A Swertz
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Bruce Hr Wolffenbuttel
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Harold Snieder
- Unit of Genetic Epidemiology and Bioinformatics, Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
91
|
Chittoor G, Haack K, Mehta NR, Laston S, Cole SA, Comuzzie AG, Butte NF, Voruganti VS. Genetic variation underlying renal uric acid excretion in Hispanic children: the Viva La Familia Study. BMC MEDICAL GENETICS 2017; 18:6. [PMID: 28095793 PMCID: PMC5240212 DOI: 10.1186/s12881-016-0366-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 12/30/2016] [Indexed: 01/01/2023]
Abstract
Background Reduced renal excretion of uric acid plays a significant role in the development of hyperuricemia and gout in adults. Hyperuricemia has been associated with chronic kidney disease and cardiovascular disease in children and adults. There are limited genome-wide association studies associating genetic polymorphisms with renal urate excretion measures. Therefore, we investigated the genetic factors that influence the excretion of uric acid and related indices in 768 Hispanic children of the Viva La Familia Study. Methods We performed a genome-wide association analysis for 24-h urinary excretion measures such as urinary uric acid/urinary creatinine ratio, uric acid clearance, fractional excretion of uric acid, and glomerular load of uric acid in SOLAR, while accounting for non-independence among family members. Results All renal urate excretion measures were significantly heritable (p <2 × 10−6) and ranged from 0.41 to 0.74. Empirical threshold for genome-wide significance was set at p <1 × 10−7. We observed a strong association (p < 8 × 10−8) of uric acid clearance with a single nucleotide polymorphism (SNP) in zinc finger protein 446 (ZNF446) (rs2033711 (A/G), MAF: 0.30). The minor allele (G) was associated with increased uric acid clearance. Also, we found suggestive associations of uric acid clearance with SNPs in ZNF324, ZNF584, and ZNF132 (in a 72 kb region of 19q13; p <1 × 10−6, MAFs: 0.28–0.31). Conclusion For the first time, we showed the importance of 19q13 region in the regulation of renal urate excretion in Hispanic children. Our findings indicate differences in inherent genetic architecture and shared environmental risk factors between our cohort and other pediatric and adult populations. Electronic supplementary material The online version of this article (doi:10.1186/s12881-016-0366-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Geetha Chittoor
- Department of Nutrition and UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, 500 Laureate Way, Kannapolis, NC, 28081, USA
| | - Karin Haack
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Nitesh R Mehta
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Sandra Laston
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Shelley A Cole
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Anthony G Comuzzie
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Nancy F Butte
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - V Saroja Voruganti
- Department of Nutrition and UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, 500 Laureate Way, Kannapolis, NC, 28081, USA.
| |
Collapse
|
92
|
Chen T, Wang Q, Li G, Wang L. A single nucleotide polymorphism in the UMOD promoter is associated with end stage renal disease. BMC MEDICAL GENETICS 2016; 17:95. [PMID: 27938332 PMCID: PMC5148830 DOI: 10.1186/s12881-016-0358-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 12/02/2016] [Indexed: 01/15/2023]
Abstract
BACKGROUND Several genome-wide association studies revealed that several variants of UMOD gene were related to the estimated glomerular filtration rate (eGFR), CKD or hypertension. In this study, we investigated the association between a common variant rs13333226 in the promoter region of UMOD gene and end stage renal disease (ESRD). METHODS Variant rs13333226 of UMOD gene was genotyped by using the ABI Real time TaqMan allelic discrimination assay in a case-control study including 638 unrelated patients with ESRD and 366 controls. RESULTS The frequency of UMOD SNP rs13333226 GG/GA genotype was significantly higher (36.83% vs. 20.22%, P = 4.02 × 10-8) and the frequency of G allele was much higher (19.04% vs. 11.20%, P = 4.00 × 10-6) in the patients with ESRD than in the controls. The G allele was associated with an increased risk of ESRD (odds ratio 2.30, 95% confidence interval 1.70-3.11, P = 6.10 × 10-8). And G allele (odds ratio 2.33, 95% confidence interval 1.32-4.13, P = 3.65 × 10-3) was associated independently with ESRD. CONCLUSIONS A common variation rs13333226 in the promoter region of UMOD gene was independently associated with ESRD in Han Chinese.
Collapse
Affiliation(s)
- Tingyu Chen
- Renal Division and Institute of Nephrology, Sichuan Provincial People's Hospital, No. 32, West 2nd Duan, 1st Circle Rd., Qingyang District, Chengdu, Sichuan, 610072, People's Republic of China
| | - Qianliao Wang
- Renal Division and Institute of Nephrology, Sichuan Provincial People's Hospital, No. 32, West 2nd Duan, 1st Circle Rd., Qingyang District, Chengdu, Sichuan, 610072, People's Republic of China
| | - Guisen Li
- Renal Division and Institute of Nephrology, Sichuan Provincial People's Hospital, No. 32, West 2nd Duan, 1st Circle Rd., Qingyang District, Chengdu, Sichuan, 610072, People's Republic of China. .,School of Medicine, University of Electronic Science and Technology of China, No. 32, West 2nd Duan, 1st Circle Rd., Qingyang District, Chengdu, Sichuan, 610072, People's Republic of China.
| | - Li Wang
- Renal Division and Institute of Nephrology, Sichuan Provincial People's Hospital, No. 32, West 2nd Duan, 1st Circle Rd., Qingyang District, Chengdu, Sichuan, 610072, People's Republic of China.,School of Medicine, University of Electronic Science and Technology of China, No. 32, West 2nd Duan, 1st Circle Rd., Qingyang District, Chengdu, Sichuan, 610072, People's Republic of China
| |
Collapse
|
93
|
Tsermpini EE, Zhang Y, Niola P, Chillotti C, Ardau R, Bocchetta A, Patrinos GP, Del Zompo M, Severino G, Lee MTM, Squassina A. Pharmacogenetics of lithium effects on glomerular function in bipolar disorder patients under chronic lithium treatment: a pilot study. Neurosci Lett 2016; 638:1-4. [PMID: 27923663 DOI: 10.1016/j.neulet.2016.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 11/30/2016] [Accepted: 12/01/2016] [Indexed: 11/25/2022]
Abstract
Bipolar disorder (BD) is a psychiatric disease characterized by alternating episodes of mania and depression. Lithium (Li) represents the mainstay treatment for BD, although a significant proportion of patients shows insufficient or no response. Li is also associated with potentially severe side effects, including renal effects. Several studies reported that Li may induce reduction of glomerular filtration rate (GFR) in patients under long-term treatment. The biological systems and the genetic factors involved in susceptibility to Li-induced renal-side effects have been scarcely explored. The aim of our study was to test the contribution of putatively risk genetic variants in Li-induced reduction in estimated GFR (eGFR) in BD patients under long-term Li treatment. Tag SNPs, located in genes previously shown to be associated with kidney dysfunction or Li mechanism of action, were selected and genotyped in a sample of 70 BD patients of Sardinian origin. SNP rs378448, located in Acid Sensing Ion Channel Neurona-1 (ACCN1) gene, showed a significant interaction with duration of Li treatment on eGFR (F2=3.623, p=0.033). Our preliminary findings suggest that rs378448 could predispose BD subjects to a detrimental effect of chronic Li treatment on kidney functioning.
Collapse
Affiliation(s)
- Evangelia Eirini Tsermpini
- University of Patras, School of Health Science, Department of Pharmacy, Laboratory of Molecular Biology and Immunology, Greece
| | - Yanfei Zhang
- RIKEN Center for Integrative Medical Sciences, Laboratory for International Alliance on Genomic Research, Yokohama, Japan; Genomic Medicine Institute, Geisinger Health System, Danville, PA, USA
| | - Paola Niola
- Section of Neurosciences and Clinical Pharmacology, Department of Biomedical Sciences, School of Medicine, University of Cagliari, Cagliari, Italy
| | - Caterina Chillotti
- Unit of Clinical Pharmacology of the University Hospital of Cagliari, Italy
| | - Raffaella Ardau
- Unit of Clinical Pharmacology of the University Hospital of Cagliari, Italy
| | - Alberto Bocchetta
- Unit of Clinical Pharmacology of the University Hospital of Cagliari, Italy
| | - George P Patrinos
- University of Patras, School of Health Science, Department of Pharmacy, Laboratory of Molecular Biology and Immunology, Greece
| | - Maria Del Zompo
- Section of Neurosciences and Clinical Pharmacology, Department of Biomedical Sciences, School of Medicine, University of Cagliari, Cagliari, Italy; Unit of Clinical Pharmacology of the University Hospital of Cagliari, Italy
| | - Giovanni Severino
- Section of Neurosciences and Clinical Pharmacology, Department of Biomedical Sciences, School of Medicine, University of Cagliari, Cagliari, Italy.
| | - Ming Ta Michael Lee
- RIKEN Center for Integrative Medical Sciences, Laboratory for International Alliance on Genomic Research, Yokohama, Japan; Genomic Medicine Institute, Geisinger Health System, Danville, PA, USA.
| | - Alessio Squassina
- Section of Neurosciences and Clinical Pharmacology, Department of Biomedical Sciences, School of Medicine, University of Cagliari, Cagliari, Italy.
| |
Collapse
|
94
|
Zaharenko L, Kalnina I, Geldnere K, Konrade I, Grinberga S, Židzik J, Javorský M, Lejnieks A, Nikitina-Zake L, Fridmanis D, Peculis R, Radovica-Spalvina I, Hartmane D, Pugovics O, Tkáč I, Klimčáková L, Pīrāgs V, Klovins J. Single nucleotide polymorphisms in the intergenic region between metformin transporter OCT2 and OCT3 coding genes are associated with short-term response to metformin monotherapy in type 2 diabetes mellitus patients. Eur J Endocrinol 2016; 175:531-540. [PMID: 27609360 DOI: 10.1530/eje-16-0347] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 08/31/2016] [Accepted: 09/08/2016] [Indexed: 12/22/2022]
Abstract
OBJECTIVES High variability in clinical response to metformin is often observed in type 2 diabetes (T2D) patients, and it highlights the need for identification of genetic components affecting the efficiency of metformin therapy. Aim of this observational study is to evaluate the role of tagSNPs (tagging single nucleotide polymorphisms) from genomic regions coding for six metformin transporter genes with respect to the short-term efficiency. DESIGN 102 tagSNPs in 6 genes coding for metformin transporters were genotyped in the group of 102 T2D patients treated with metformin for 3 months. METHODS Most significant hits were analyzed in the group of 131 T2D patients from Slovakia. Pharmacokinetic study in 25 healthy nondiabetic volunteers was conducted to investigate the effects of identified polymorphisms. RESULTS In the discovery group of 102 patients, minor alleles of rs3119309, rs7757336 and rs2481030 were significantly nominally associated with metformin inefficiency (P = 1.9 × 10-6 to 8.1 × 10-6). Effects of rs2481030 and rs7757336 did not replicate in the group of 131 T2DM patients from Slovakia alone, whereas rs7757336 was significantly associated with a reduced metformin response in combined group. In pharmacokinetic study, group of individuals harboring risk alleles of rs7757336 and rs2481030 displayed significantly reduced AUC∞ of metformin in plasma. CONCLUSIONS For the first time, we have identified an association between the lack of metformin response and SNPs rs3119309 and rs7757336 located in the 5' flanking region of the genes coding for Organic cation transporter 2 and rs2481030 located in the 5' flanking region of Organic cation transporter 3 that was supported by the results of a pharmacokinetic study on 25 healthy volunteers.
Collapse
Affiliation(s)
| | - Ineta Kalnina
- Latvian Biomedical Research and Study CentreRiga, Latvia
| | - Kristine Geldnere
- Pauls Stradins Clinical University HospitalRiga, Latvia
- Faculty of MedicineUniversity of Latvia, Riga, Latvia
| | - Ilze Konrade
- Riga East Clinical University HospitalRiga, Latvia
- Riga Stradins UniversityRiga, Latvia
| | | | - Jozef Židzik
- Faculty of MedicineP. J. Šafárik University, Košice, Slovakia
| | - Martin Javorský
- Faculty of MedicineP. J. Šafárik University, Košice, Slovakia
| | - Aivars Lejnieks
- Riga East Clinical University HospitalRiga, Latvia
- Riga Stradins UniversityRiga, Latvia
| | | | | | - Raitis Peculis
- Latvian Biomedical Research and Study CentreRiga, Latvia
| | | | | | | | - Ivan Tkáč
- Faculty of MedicineP. J. Šafárik University, Košice, Slovakia
| | | | - Valdis Pīrāgs
- Pauls Stradins Clinical University HospitalRiga, Latvia
- Faculty of MedicineUniversity of Latvia, Riga, Latvia
| | - Janis Klovins
- Latvian Biomedical Research and Study CentreRiga, Latvia
| |
Collapse
|
95
|
Ried JS, Jeff M. J, Chu AY, Bragg-Gresham JL, van Dongen J, Huffman JE, Ahluwalia TS, Cadby G, Eklund N, Eriksson J, Esko T, Feitosa MF, Goel A, Gorski M, Hayward C, Heard-Costa NL, Jackson AU, Jokinen E, Kanoni S, Kristiansson K, Kutalik Z, Lahti J, Luan J, Mägi R, Mahajan A, Mangino M, Medina-Gomez C, Monda KL, Nolte IM, Pérusse L, Prokopenko I, Qi L, Rose LM, Salvi E, Smith MT, Snieder H, Stančáková A, Ju Sung Y, Tachmazidou I, Teumer A, Thorleifsson G, van der Harst P, Walker RW, Wang SR, Wild SH, Willems SM, Wong A, Zhang W, Albrecht E, Couto Alves A, Bakker SJL, Barlassina C, Bartz TM, Beilby J, Bellis C, Bergman RN, Bergmann S, Blangero J, Blüher M, Boerwinkle E, Bonnycastle LL, Bornstein SR, Bruinenberg M, Campbell H, Chen YDI, Chiang CWK, Chines PS, Collins FS, Cucca F, Cupples LA, D'Avila F, de Geus EJ.C, Dedoussis G, Dimitriou M, Döring A, Eriksson JG, Farmaki AE, Farrall M, Ferreira T, Fischer K, Forouhi NG, Friedrich N, Gjesing AP, Glorioso N, Graff M, Grallert H, Grarup N, Gräßler J, Grewal J, Hamsten A, Harder MN, Hartman CA, Hassinen M, Hastie N, Hattersley AT, Havulinna AS, Heliövaara M, Hillege H, Hofman A, Holmen O, et alRied JS, Jeff M. J, Chu AY, Bragg-Gresham JL, van Dongen J, Huffman JE, Ahluwalia TS, Cadby G, Eklund N, Eriksson J, Esko T, Feitosa MF, Goel A, Gorski M, Hayward C, Heard-Costa NL, Jackson AU, Jokinen E, Kanoni S, Kristiansson K, Kutalik Z, Lahti J, Luan J, Mägi R, Mahajan A, Mangino M, Medina-Gomez C, Monda KL, Nolte IM, Pérusse L, Prokopenko I, Qi L, Rose LM, Salvi E, Smith MT, Snieder H, Stančáková A, Ju Sung Y, Tachmazidou I, Teumer A, Thorleifsson G, van der Harst P, Walker RW, Wang SR, Wild SH, Willems SM, Wong A, Zhang W, Albrecht E, Couto Alves A, Bakker SJL, Barlassina C, Bartz TM, Beilby J, Bellis C, Bergman RN, Bergmann S, Blangero J, Blüher M, Boerwinkle E, Bonnycastle LL, Bornstein SR, Bruinenberg M, Campbell H, Chen YDI, Chiang CWK, Chines PS, Collins FS, Cucca F, Cupples LA, D'Avila F, de Geus EJ.C, Dedoussis G, Dimitriou M, Döring A, Eriksson JG, Farmaki AE, Farrall M, Ferreira T, Fischer K, Forouhi NG, Friedrich N, Gjesing AP, Glorioso N, Graff M, Grallert H, Grarup N, Gräßler J, Grewal J, Hamsten A, Harder MN, Hartman CA, Hassinen M, Hastie N, Hattersley AT, Havulinna AS, Heliövaara M, Hillege H, Hofman A, Holmen O, Homuth G, Hottenga JJ, Hui J, Husemoen LL, Hysi PG, Isaacs A, Ittermann T, Jalilzadeh S, James AL, Jørgensen T, Jousilahti P, Jula A, Marie Justesen J, Justice AE, Kähönen M, Karaleftheri M, Tee Khaw K, Keinanen-Kiukaanniemi SM, Kinnunen L, Knekt PB, Koistinen HA, Kolcic I, Kooner IK, Koskinen S, Kovacs P, Kyriakou T, Laitinen T, Langenberg C, Lewin AM, Lichtner P, Lindgren CM, Lindström J, Linneberg A, Lorbeer R, Lorentzon M, Luben R, Lyssenko V, Männistö S, Manunta P, Leach IM, McArdle WL, Mcknight B, Mohlke KL, Mihailov E, Milani L, Mills R, Montasser ME, Morris AP, Müller G, Musk AW, Narisu N, Ong KK, Oostra BA, Osmond C, Palotie A, Pankow JS, Paternoster L, Penninx BW, Pichler I, Pilia MG, Polašek O, Pramstaller PP, Raitakari OT, Rankinen T, Rao DC, Rayner NW, Ribel-Madsen R, Rice TK, Richards M, Ridker PM, Rivadeneira F, Ryan KA, Sanna S, Sarzynski MA, Scholtens S, Scott RA, Sebert S, Southam L, Sparsø TH, Steinthorsdottir V, Stirrups K, Stolk RP, Strauch K, Stringham HM, Swertz MA, Swift AJ, Tönjes A, Tsafantakis E, van der Most PJ, Van Vliet-Ostaptchouk JV, Vandenput L, Vartiainen E, Venturini C, Verweij N, Viikari JS, Vitart V, Vohl MC, Vonk JM, Waeber G, Widén E, Willemsen G, Wilsgaard T, Winkler TW, Wright AF, Yerges-Armstrong LM, Hua Zhao J, Carola Zillikens M, Boomsma DI, Bouchard C, Chambers JC, Chasman DI, Cusi D, Gansevoort RT, Gieger C, Hansen T, Hicks AA, Hu F, Hveem K, Jarvelin MR, Kajantie E, Kooner JS, Kuh D, Kuusisto J, Laakso M, Lakka TA, Lehtimäki T, Metspalu A, Njølstad I, Ohlsson C, Oldehinkel AJ, Palmer LJ, Pedersen O, Perola M, Peters A, Psaty BM, Puolijoki H, Rauramaa R, Rudan I, Salomaa V, Schwarz PEH, Shudiner AR, Smit JH, Sørensen TIA, Spector TD, Stefansson K, Stumvoll M, Tremblay A, Tuomilehto J, Uitterlinden AG, Uusitupa M, Völker U, Vollenweider P, Wareham NJ, Watkins H, Wilson JF, Zeggini E, Abecasis GR, Boehnke M, Borecki IB, Deloukas P, van Duijn CM, Fox C, Groop LC, Heid IM, Hunter DJ, Kaplan RC, McCarthy MI, North KE, O'Connell JR, Schlessinger D, Thorsteinsdottir U, Strachan DP, Frayling T, Hirschhorn JN, Müller-Nurasyid M, Loos RJF. A principal component meta-analysis on multiple anthropometric traits identifies novel loci for body shape. Nat Commun 2016; 7:13357. [PMID: 27876822 PMCID: PMC5114527 DOI: 10.1038/ncomms13357] [Show More Authors] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 09/21/2016] [Indexed: 01/15/2023] Open
Abstract
Large consortia have revealed hundreds of genetic loci associated with anthropometric traits, one trait at a time. We examined whether genetic variants affect body shape as a composite phenotype that is represented by a combination of anthropometric traits. We developed an approach that calculates averaged PCs (AvPCs) representing body shape derived from six anthropometric traits (body mass index, height, weight, waist and hip circumference, waist-to-hip ratio). The first four AvPCs explain >99% of the variability, are heritable, and associate with cardiometabolic outcomes. We performed genome-wide association analyses for each body shape composite phenotype across 65 studies and meta-analysed summary statistics. We identify six novel loci: LEMD2 and CD47 for AvPC1, RPS6KA5/C14orf159 and GANAB for AvPC3, and ARL15 and ANP32 for AvPC4. Our findings highlight the value of using multiple traits to define complex phenotypes for discovery, which are not captured by single-trait analyses, and may shed light onto new pathways.
Collapse
Affiliation(s)
- Janina S. Ried
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Janina Jeff M.
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Audrey Y. Chu
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02215, USA
| | - Jennifer L. Bragg-Gresham
- Kidney Epidemiology and Cost Center, Internal Medicine-Nephrology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Jenny van Dongen
- Department of Biological Psychology, VU University, 1081BT Amsterdam, The Netherlands
| | - Jennifer E. Huffman
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XU Edinburgh, Scotland
| | - Tarunveer S. Ahluwalia
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
- Steno Diabetes Center A/S, DK-2820 Gentofte, Denmark
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Ledreborg Allé 34, DK-2820 Copenhagen, Denmark
| | - Gemma Cadby
- Centre for Genetic Origins of Health and Disease, University of Western Australia, Crawley, Western Australia 6009, Australia
| | - Niina Eklund
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Joel Eriksson
- Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Tõnu Esko
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 2142, USA
- Divisions of Endocrinology and Genetics and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Mary F. Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63108, USA
| | - Anuj Goel
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Mathias Gorski
- Department of Nephrology, University Hospital Regensburg, 93042 Regensburg, Germany
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, 93053 Regensburg, Germany
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XU Edinburgh, Scotland
| | - Nancy L. Heard-Costa
- National Heart, Lung, and Blood Institute, the Framingham Heart Study, Framingham, Massachusetts 01702, USA
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Anne U. Jackson
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Eero Jokinen
- Hospital for Children and Adolescents, University of Helsinki, FI-00290 Helsinki, Finland
| | - Stavroula Kanoni
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge CB10 1SA, UK
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Kati Kristiansson
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00290 Helsinki, Finland
| | - Zoltán Kutalik
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
- Department of Medical Genetics, University of Lausanne, Lausanne, 1005, Switzerland
- Institute of Social and Preventive Medicine, University Hospital Lausanne (CHUV), 1010 Lausanne, Switzerland
| | - Jari Lahti
- Folkhälsan Research Centre, FI-00290 Helsinki, Finland
- Institute of Behavioural Sciences, University of Helsinki, FI-00014 Helsinki, Finland
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Carolina Medina-Gomez
- Department of Epidemiology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | - Keri L. Monda
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- The Center for Observational Research, Amgen Inc., Thousand Oaks, California 91320-1799, USA
| | - Ilja M. Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Louis Pérusse
- Department of Kinesiology, Laval University, Québec, Québec, Canada G1V 0A6
- Institute of Nutrition and Functional Foods, Laval University, Québec, Québec, Canada G1V 0A6
| | - Inga Prokopenko
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, London W12 0NN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Lu Qi
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | - Lynda M. Rose
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02215, USA
| | - Erika Salvi
- Department of Health Sciences, University of Milano at San Paolo Hospital, 20139 Milano, Italy
- Filarete Foundation, Genomic and Bioinformatics Unit, Milano 20139, Italy
| | - Megan T. Smith
- Department of Biostatistics, University of Washington, Seattle, Washington 98195, USA
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Alena Stančáková
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Yun Ju Sung
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Ioanna Tachmazidou
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge CB10 1SA, UK
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany
| | | | - Pim van der Harst
- Durrer Center for Cardiogenetic Research, Interuniversity Cardiology Institute Netherlands-Netherlands Heart Institute, 3501 DG Utrecht, The Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, Netherlands
| | - Ryan W. Walker
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- The Department of Preventive Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Sophie R. Wang
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Divisions of Genetics and Endocrinology and Program in Genomics, Boston's Children's Hospital, Boston, Massachusetts 02115, USA
| | - Sarah H. Wild
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, EH8 9AG Teviot Place, Edinburgh, Scotland
| | - Sara M. Willems
- Department of Epidemiology, Genetic Epidemiology Unit, Erasmus University Medical Center, 3015GE Rotterdam, The Netherlands
| | - Andrew Wong
- MRC Unit for Lifelong Health & Ageing at UCL, London WC1B 5JU, UK
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK
- Ealing Hospital NHS Trust, Middlesex UB1 3HW, UK
| | - Eva Albrecht
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Alexessander Couto Alves
- Department of Epidemiology and Biostatistics, MRC Health Protection Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London W12 0NN, UK
| | - Stephan J. L. Bakker
- Department of Medicine, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, Netherlands
| | - Cristina Barlassina
- Department of Health Sciences, University of Milano at San Paolo Hospital, 20139 Milano, Italy
- Filarete Foundation, Genomic and Bioinformatics Unit, Milano 20139, Italy
| | - Traci M. Bartz
- Department of Biostatistics, University of Washington, Seattle, Washington 98195, USA
- Department of Medicine, University of Washington, Seattle, Washington 98101, USA
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington 98101, USA
| | - John Beilby
- Pathwest Laboratory Medicine of Western Australia, Nedlands, Western Australia 6009, Australia
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Claire Bellis
- Genomics Research Centre, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland 4001, Australia
- Human Genetics, Genome Institute of Singapore, Agency for Science, Technology and Research of Singapore, Singapore 138672, Singapore
| | - Richard N. Bergman
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | - Sven Bergmann
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
- Department of Medical Genetics, University of Lausanne, Lausanne, 1005, Switzerland
| | - John Blangero
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Brownsville, Texas 78520, USA
| | - Matthias Blüher
- University of Leipzig, IFB Adiposity Diseases, 04103 Leipzig, Germany
- Department of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Eric Boerwinkle
- Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Lori L. Bonnycastle
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Stefan R. Bornstein
- Medical Faculty Carl Gustav Carus, Department of Medicine III, University of Dresden, 01307 Dresden, Germany
| | - Marcel Bruinenberg
- University of Groningen, University Medical Center Groningen, The LifeLines Cohort Study, 9700 RB Groningen, The Netherlands
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, EH8 9AG Teviot Place, Edinburgh, Scotland
| | - Yii-Der Ida Chen
- Los Angeles BioMedical Resesarch Institute at Harbor-UCLA Medical Center, Torrance, California 90502, USA
| | | | - Peter S. Chines
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Francis S Collins
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | | | - L Adrienne Cupples
- National Heart, Lung, and Blood Institute, the Framingham Heart Study, Framingham, Massachusetts 01702, USA
| | - Francesca D'Avila
- Department of Health Sciences, University of Milano at San Paolo Hospital, 20139 Milano, Italy
- Filarete Foundation, Genomic and Bioinformatics Unit, Milano 20139, Italy
| | - Eco J .C. de Geus
- Department of Biological Psychology, VU University, 1081BT Amsterdam, The Netherlands
- EMGO Institute for Health and Care Research, VU University Medical Center, 1081 BT Amsterdam, The Netherlands
| | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece
| | - Maria Dimitriou
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge CB10 1SA, UK
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece
| | - Angela Döring
- Institute of Epidemiology I, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Johan G. Eriksson
- Folkhälsan Research Centre, FI-00290 Helsinki, Finland
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, FI-00271 Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, FI-00014 Helsinki, Finland
| | - Aliki-Eleni Farmaki
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece
| | - Martin Farrall
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Teresa Ferreira
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Krista Fischer
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Nita G. Forouhi
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Anette Prior Gjesing
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Nicola Glorioso
- Hypertension and Related Disease Centre, AOU-University of Sassari, 7100 Sassari, Italy
| | - Mariaelisa Graff
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Harald Grallert
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Niels Grarup
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Jürgen Gräßler
- Department of Medicine III, Pathobiochemistry, Technische Universitaet, 01307 Dresden, Germany
| | - Jagvir Grewal
- Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK
- Ealing Hospital NHS Trust, Middlesex UB1 3HW, UK
| | - Anders Hamsten
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine Solna, Atherosclerosis Research Unit, Karolinska Institutet, 17176 Stockholm 17176, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Marie Neergaard Harder
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Catharina A. Hartman
- University of Groningen, University Medical Center Groningen, Interdisciplinary Center Psychopathology and Emotion Regulation, 9700 RB Groningen, The Netherlands
| | - Maija Hassinen
- Kuopio Research Institute of Exercise Medicine, 70100 Kuopio, Finland
| | - Nicholas Hastie
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XU Edinburgh, Scotland
| | - Andrew Tym Hattersley
- Institue of Biomedical & Clinical Science, University of Exeter, Barrack Road, Exeter EX2 5DW, UK
| | - Aki S. Havulinna
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Markku Heliövaara
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Hans Hillege
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | - Oddgeir Holmen
- Department of Public Health and General Practice, Norwegian University of Science and Technology, 7489 Trondheim, Norway
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, VU University, 1081BT Amsterdam, The Netherlands
| | - Jennie Hui
- Pathwest Laboratory Medicine of Western Australia, Nedlands, Western Australia 6009, Australia
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, Western Australia 6009, Australia
- School of Population Health, University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Lise Lotte Husemoen
- Research Centre for Prevention and Health, Glostrup Hospital, 2600 Glostrup, Denmark
| | - Pirro G. Hysi
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Aaron Isaacs
- Department of Epidemiology, Genetic Epidemiology Unit, Erasmus University Medical Center, 3015GE Rotterdam, The Netherlands
| | - Till Ittermann
- Institute for Community Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Shapour Jalilzadeh
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Alan L. James
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia 6009, Australia
| | - Torben Jørgensen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Faculty of Medicine, University of Aalborg, 9220 Aalborg, Denmark
- Research Centre for Prevention and Health, Capital Region of Denmark, DK2600 Glostrup, Denmark
| | - Pekka Jousilahti
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Antti Jula
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Johanne Marie Justesen
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Anne E. Justice
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, FI-33521 Tampere, Finland
- Department of Clinical Physiology, University of Tampere School of Medicine, FI-33014 Tampere, Finland
| | | | - Kay Tee Khaw
- Clinical Gerontology Unit, Box 251, Addenbrooke's Hospital, Hills Road, Cambridge CB2 2QQ, UK
| | - Sirkka M. Keinanen-Kiukaanniemi
- Faculty of Medicine, Institute of Health Sciences, University of Oulu, Oulu F1-90014, Finland
- Unit of General Practice, Oulu University Hospital, Oulu FI-90029, Finland
| | - Leena Kinnunen
- National Institute for Health and Welfare, FI-00271 Helsinki, Finland
| | - Paul B. Knekt
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Heikki A. Koistinen
- National Institute for Health and Welfare, FI-00271 Helsinki, Finland
- Department of Medicine and Abdominal Center: Endocrinology, University of Helsinki and Helsinki University Central Hospital,, 00029 Helsinki, Finland
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland
| | - Ivana Kolcic
- Department of Public Health, Faculty of Medicine, University of Split, 21000 Split, Croatia
| | | | - Seppo Koskinen
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Peter Kovacs
- University of Leipzig, IFB Adiposity Diseases, 04103 Leipzig, Germany
| | - Theodosios Kyriakou
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Tomi Laitinen
- Kuopio University Hospital, 70029 Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
- Department of Epidemiology and Public Health, UCL, London WC1E 6BT, UK
| | - Alexandra M. Lewin
- Department of Epidemiology and Biostatistics, MRC Health Protection Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London W12 0NN, UK
| | - Peter Lichtner
- Institute of Human Genetics, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Cecilia M. Lindgren
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 2142, USA
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- The Big Data Institute, University of Oxford, Oxford OX3 7LJ, UK
| | - Jaana Lindström
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Allan Linneberg
- Research Centre for Prevention and Health, Glostrup Hospital, 2600 Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Clinical Experimental Research, Rigshospitalet, 2600 Glostrup, Denmark
| | - Roberto Lorbeer
- Institute for Community Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Mattias Lorentzon
- Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Robert Luben
- Strangeways Research Laboratory Wort's Causeway, Cambridge CB1 8RN, UK
| | - Valeriya Lyssenko
- Steno Diabetes Center A/S, DK-2820 Gentofte, Denmark
- Lund University Diabetes Centre and Department of Clinical Science, Diabetes & Endocrinology Unit, Lund University, 221 00 Malmö, Sweden
| | - Satu Männistö
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Paolo Manunta
- Chair of Nephrology, Università Vita Salute San Raffaele and Genomics of Renal Diseases and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Milan 20139, Italy
| | - Irene Mateo Leach
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, Netherlands
| | - Wendy L. McArdle
- School of Social and Community Medicine, University of Bristol, Bristol BS82BN, UK
| | - Barbara Mcknight
- Department of Biostatistics, University of Washington, Seattle, Washington 98195, USA
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington 98101, USA
- Divison of Public Health Sciences, Program in Biostatistics and Biomathematics, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Karen L. Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Evelin Mihailov
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Lili Milani
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | | | - May E. Montasser
- Division of Endocrinology, Diabetes & Nutrition, Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Andrew P. Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Department of Biostatistics, University of Liverpool, Liverpool L69 3GA, UK
| | - Gabriele Müller
- Center for Evidence Based Healthcare, University of Dresden, Medical Faculty Carl Gustav Carus, Dresden, 01307, Germany
| | - Arthur W. Musk
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Nedlands, West Australia 6009, Australia
| | - Narisu Narisu
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Ken K. Ong
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
- MRC Unit for Lifelong Health & Ageing at UCL, London WC1B 5JU, UK
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Ben A. Oostra
- Department of Epidemiology, Genetic Epidemiology Unit, Erasmus University Medical Center, 3015GE Rotterdam, The Netherlands
| | - Clive Osmond
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Aarno Palotie
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00290 Helsinki, Finland
- Massachusetts General Hospital, Center for Human Genetic Research, Psychiatric and Neurodevelopmental Genetics Unit, Boston, Massachusetts 02114, USA
| | - James S. Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota 55455-0381, USA
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 1TH, UK
| | - Brenda W. Penninx
- Department of Psychiatry and EMGO Institute for Health and Care Research, VU University Medical Center, AJ Ernstraat 1887, 1081 HL Amsterdam, The Netherlands
| | - Irene Pichler
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), 39100 Bolzano, Italy
- Affiliated Institute of the University of Lübeck, 23562 Lübeck, Germany
| | - Maria G. Pilia
- Istituto di Ricerca Genetica e Biomedica, CNR, 9042 Monserrato, Italy
| | - Ozren Polašek
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, EH8 9AG Teviot Place, Edinburgh, Scotland
- Department of Public Health, Faculty of Medicine, University of Split, 21000 Split, Croatia
| | - Peter P. Pramstaller
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), 39100 Bolzano, Italy
- Affiliated Institute of the University of Lübeck, 23562 Lübeck, Germany
- Department of Neurology, University of Lübeck, 23562 Lübeck, Germany
- Department of Neurology, General Central Hospital, 39100 Bolzano, Italy
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, FI-20521 Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, FI-20520 Turku, Finland
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808, USA
| | - D. C. Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Nigel W. Rayner
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton CB10 1HH, UK
| | - Rasmus Ribel-Madsen
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Treva K. Rice
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Marcus Richards
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
- MRC Unit for Lifelong Health & Ageing at UCL, London WC1B 5JU, UK
| | - Paul M. Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02215, USA
- Harvard Medical School, Boston, Massachusetts, 02115, USA
| | - Fernando Rivadeneira
- Department of Epidemiology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | - Kathy A. Ryan
- Division of Endocrinology, Diabetes & Nutrition, Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Serena Sanna
- Istituto di Ricerca Genetica e Biomedica, CNR, 9042 Monserrato, Italy
| | - Mark A. Sarzynski
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808, USA
| | - Salome Scholtens
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Robert A. Scott
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Sylvain Sebert
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, London W12 0NN, UK
- Biocenter Oulu, University of Oulu, Oulu FI-90014, Finland
- Center For Life-Course Health Research, University of Oulu, FI-90014 Oulu, Finland
| | - Lorraine Southam
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge CB10 1SA, UK
| | - Thomas Hempel Sparsø
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
| | | | - Kathleen Stirrups
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge CB10 1SA, UK
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Ronald P. Stolk
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Heather M. Stringham
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Morris A. Swertz
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Amy J. Swift
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Anke Tönjes
- Department of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | | | - Peter J. van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Jana V. Van Vliet-Ostaptchouk
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Liesbeth Vandenput
- Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Erkki Vartiainen
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Cristina Venturini
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Niek Verweij
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, Netherlands
| | - Jorma S. Viikari
- Department of Medicine, University of Turku, FI-20521 Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Veronique Vitart
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XU Edinburgh, Scotland
| | - Marie-Claude Vohl
- Institute of Nutrition and Functional Foods, Laval University, Québec, Québec, Canada G1V 0A6
- School of Nutrition, Laval University, Québec, Québec, Canada G1V 0A6
| | - Judith M. Vonk
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Gérard Waeber
- Department of Internal Medicine, University Hospital Lausanne (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland
| | - Elisabeth Widén
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00290 Helsinki, Finland
| | - Gonneke Willemsen
- Department of Biological Psychology, VU University, 1081BT Amsterdam, The Netherlands
| | - Tom Wilsgaard
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, 9037 Tromsø, Norway
| | - Thomas W. Winkler
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, 93053 Regensburg, Germany
| | - Alan F. Wright
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XU Edinburgh, Scotland
| | - Laura M. Yerges-Armstrong
- Division of Endocrinology, Diabetes & Nutrition, Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Jing Hua Zhao
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - M. Carola Zillikens
- Department of Internal Medicine, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | - Dorret I. Boomsma
- Department of Biological Psychology, VU University, 1081BT Amsterdam, The Netherlands
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808, USA
| | - John C. Chambers
- Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK
- Ealing Hospital NHS Trust, Middlesex UB1 3HW, UK
- Imperial College Healthcare NHS Trust, London W12 0HS, UK
| | - Daniel I. Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02215, USA
- Harvard Medical School, Boston, Massachusetts, 02115, USA
| | - Daniele Cusi
- Department of Health Sciences, University of Milano at San Paolo Hospital, 20139 Milano, Italy
- Filarete Foundation, Genomic and Bioinformatics Unit, Milano 20139, Italy
| | - Ron T. Gansevoort
- Department of Medicine, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, Netherlands
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Torben Hansen
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
- Faculty of Health Sciences, University of Southern Denmark, 5000 Odense, Denmark
| | - Andrew A. Hicks
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), 39100 Bolzano, Italy
- Affiliated Institute of the University of Lübeck, 23562 Lübeck, Germany
| | - Frank Hu
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | - Kristian Hveem
- Department of Public Health and General Practice, Norwegian University of Science and Technology, 7489 Trondheim, Norway
| | - Marjo-Riitta Jarvelin
- Biocenter Oulu, University of Oulu, Oulu FI-90014, Finland
- Unit of Primary Care, Oulu University Hospital, 90029 OYS Oulu, Finland
- Department of Epidemiology and Biostatistics, MRC–PHE Centre for Environment & Health, School of Public Health, Imperial College London W12 0NN, UK
- Faculty of Medicine, Center for Life Course Epidemiology, University of Oulu, P.O.Box 5000, FI-90014 Oulu, Finland
| | - Eero Kajantie
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, FI-00271 Helsinki, Finland
- Children's Hospital, Helsinki University Hospital and University of Helsinki, FI-00029 Helsinki, Finland
- Department of Obstetrics and Gynecology, MRC Oulu, Oulu University Hospital and University of Oulu, FI-90029 Oulu, Finland
| | - Jaspal S. Kooner
- Ealing Hospital NHS Trust, Middlesex UB1 3HW, UK
- Imperial College Healthcare NHS Trust, London W12 0HS, UK
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Diana Kuh
- MRC Unit for Lifelong Health & Ageing at UCL, London WC1B 5JU, UK
| | - Johanna Kuusisto
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Timo A. Lakka
- Kuopio Research Institute of Exercise Medicine, 70100 Kuopio, Finland
- Department of Physiology, Institute of Biomedicine, University of Eastern Finland, Kuopio Campus, 70210 Kuopio, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, University of Tampere School of Medicine, FI-33014 Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories and School of Medicine, University of Tampere, FI-33520 Tampere, Finland
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Inger Njølstad
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, 9037 Tromsø, Norway
- Department of Clinical Medicine, Faculty of Health Sciences, University of Tromsø, 9037 Tromsø, Norway
| | - Claes Ohlsson
- Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Albertine J. Oldehinkel
- University of Groningen, University Medical Center Groningen, Interdisciplinary Center Psychopathology and Emotion Regulation, 9700 RB Groningen, The Netherlands
| | - Lyle J. Palmer
- School of Public Health, University of Adelaide, Adelaide, South Australia 5005, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Oluf Pedersen
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Markus Perola
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00290 Helsinki, Finland
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- DZHK (German Centre for Cardiovascular Research), partnersite Munich Heart Alliance, 80802 Munich, Germany
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington 98101, USA
- Department of Medicine, University of Washington, Seattle, Washington 98101, USA
- Departments of Epidemiology and Health Services, University of Washington, Seattle, Washington 98101, USA
- Group Health Research Institute, Group Health Cooperative, Seatte, Washington 98101, USA
| | - Hannu Puolijoki
- South Ostrobothnia Central Hospital, Seinäjoki Fi-60220, Finland
| | - Rainer Rauramaa
- Kuopio Research Institute of Exercise Medicine, 70100 Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, 70211 Kuopio, Finland
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, EH8 9AG Teviot Place, Edinburgh, Scotland
| | - Veikko Salomaa
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Peter E. H. Schwarz
- Medical Faculty Carl Gustav Carus, Department of Medicine III, University of Dresden, 01307 Dresden, Germany
- Paul Langerhans Institute Dresden, German Center for Diabetes Research (DZD), Dresden 01307, Germany
| | - Alan R. Shudiner
- Division of Endocrinology, Diabetes & Nutrition, Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
- Geriatric Research and Education Clinical Center, Vetrans Administration Medical Center, Baltimore, Maryland 21042, USA
| | - Jan H. Smit
- Department of Psychiatry and EMGO Institute for Health and Care Research, VU University Medical Center, AJ Ernstraat 1887, 1081 HL Amsterdam, The Netherlands
| | - Thorkild I. A. Sørensen
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS82BN, UK
- Institute of Preventive Medicine, Bispebjerg and Frederiksberg Hospital, The Capital Region, 2000 Frederiksberg, Denmark
| | - Timothy D. Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Kari Stefansson
- deCODE Genetics, Amgen inc., 101 Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Michael Stumvoll
- University of Leipzig, IFB Adiposity Diseases, 04103 Leipzig, Germany
- Department of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Angelo Tremblay
- Department of Kinesiology, Laval University, Québec, Québec, Canada G1V 0A6
| | - Jaakko Tuomilehto
- Diabetes Prevention Unit, National Institute for Health and Welfare, FI-00271 Helsinki, Finland
- Centre for Vascular Prevention, Danube-University Krems, 3500 Krems, Austria
- Diabetes Research Group, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - André G. Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | - Matti Uusitupa
- Department of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland
- Research Unit, Kuopio University Hospital, 70029 Kuopio, Finland
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, 17475 Greifswald, Germany
| | - Peter Vollenweider
- Department of Internal Medicine, University Hospital Lausanne (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland
| | - Nicholas J. Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - James F. Wilson
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XU Edinburgh, Scotland
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, EH8 9AG Teviot Place, Edinburgh, Scotland
| | - Eleftheria Zeggini
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge CB10 1SA, UK
| | - Goncalo R. Abecasis
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Michael Boehnke
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Ingrid B. Borecki
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63108, USA
| | - Panos Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton CB10 1HH, UK
- Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Cornelia M. van Duijn
- Department of Epidemiology, Genetic Epidemiology Unit, Erasmus University Medical Center, 3015GE Rotterdam, The Netherlands
- Center for Medical Systems Biology, 2300 Leiden, The Netherlands
| | - Caroline Fox
- National Heart, Lung, and Blood Institute, the Framingham Heart Study, Framingham, Massachusetts 01702, USA
- Harvard Medical School, Boston, Massachusetts, 02115, USA
| | - Leif C. Groop
- Lund University Diabetes Centre and Department of Clinical Science, Diabetes & Endocrinology Unit, Lund University, 221 00 Malmö, Sweden
- Finnish Institute for Molecular Medicine (FIMM), Helsinki University, 00014 Helsinki, Finland
| | - Iris M. Heid
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, 93053 Regensburg, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - David J. Hunter
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 2142, USA
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts 02115, USA
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | - Robert C. Kaplan
- Department of Epidemiology and Popualtion Health, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Mark I. McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
- Oxford NIHR Biomedical Research Centre, Oxford OX3 7LJ, UK
| | - Kari E. North
- Department of Epidemiology, Carolina Center for Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7400, USA
| | - Jeffrey R. O'Connell
- Division of Endocrinology, Diabetes & Nutrition, Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - David Schlessinger
- National Institute on Aging, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Unnur Thorsteinsdottir
- deCODE Genetics, Amgen inc., 101 Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - David P. Strachan
- Population Health Research Institute, St George's, University of London, London SW17 0RE, UK
| | - Timothy Frayling
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter EX1 2LU, UK
| | - Joel N. Hirschhorn
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 2142, USA
- Divisions of Endocrinology and Genetics and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Divisions of Genetics and Endocrinology and Program in Genomics, Boston's Children's Hospital, Boston, Massachusetts 02115, USA
- Metabolism Initiative, Broad Institute, Cambridge, Massachusetts 02142, USA
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, 81377 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partnersite Munich Heart Alliance, 80802 Munich, Germany
- Department of Medicine I, University Hospital Grosshadern, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
- The Department of Preventive Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- The Genetics of Obesity and Related Metabolic Traits Program, The Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- The Mindich Child Health and Development Institute, The Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|
96
|
Duffy DL, McDonald SP, Hayhurst B, Panagiotopoulos S, Smith TJ, Wang XL, Wilcken DE, Duarte NL, Mathews J, Hoy WE. Familial aggregation of albuminuria and arterial hypertension in an Aboriginal Australian community and the contribution of variants in ACE and TP53. BMC Nephrol 2016; 17:183. [PMID: 27871254 PMCID: PMC5117595 DOI: 10.1186/s12882-016-0396-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 11/09/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Aboriginal Australians are at high risk of cardiovascular, metabolic and renal diseases, resulting in a marked reduction in life expectancy when compared to the rest of the Australian population. This is partly due to recognized environmental and lifestyle risk factors, but a contribution of genetic susceptibility is also likely. METHODS Using results from a comprehensive survey of one community (N = 1350 examined individuals), we have tested for familial aggregation of plasma glucose, arterial blood pressure, albuminuria (measured as urinary albumin to creatinine ratio, UACR) and estimated glomerular filtration rate (eGFR), and quantified the contribution of variation at four candidate genes (ACE; TP53; ENOS3; MTHFR). RESULTS In the subsample of 357 individuals with complete genotype and phenotype data we showed that both UACR (h2 = 64%) and blood pressure (sBP h2 = 29%, dBP, h2 = 11%) were significantly heritable. The ACE insertion-deletion (P = 0.0009) and TP53 codon72 polymorphisms (P = 0.003) together contributed approximately 15% of the total heritability of UACR, with an effect of ACE genotype on BP also clearly evident. CONCLUSIONS While the effects of the ACE insertion-deletion on risk of renal disease (especially in the setting of diabetes) are well recognized, this is only the second study to implicate p53 genotype as a risk factor for albuminuria - the other being an earlier study we performed in a different Aboriginal community (McDonald et al., J Am Soc Nephrol 13: 677-83, 2002). We conclude that there are significant genetic contributions to the high prevalence of chronic diseases observed in this population.
Collapse
Affiliation(s)
- David L. Duffy
- Genetic Epidemiology Laboratory, QIMR Berghofer Institute of Medical Research, 300 Herston Rd, Brisbane, 4006 Australia
| | | | - Beverley Hayhurst
- Cradle Coast Authority, Tasmania, Formerly Menzies School of Health Research, Darwin, Australia
| | | | | | - Xing L. Wang
- Department of Genetics, Southwest Foundation for Biomedical Research, San Antonio, Texas Australia
| | - David E. Wilcken
- Cardiovascular Genetics Department, Prince of Wales Hospital, Sydney, Australia
| | - Natalia L. Duarte
- Cardiovascular Genetics Department, Prince of Wales Hospital, Sydney, Australia
| | - John Mathews
- Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Wendy E. Hoy
- Centre for Chronic Disease, The University of Queensland School of Medicine, Brisbane, Australia
- Centre for Chronic Disease, Central Clinical School, Royal Brisbane Hospital, Queensland, 4029 Australia
| |
Collapse
|
97
|
Liu C, Kraja AT, Smith JA, Brody JA, Franceschini N, Bis JC, Rice K, Morrison AC, Lu Y, Weiss S, Guo X, Palmas W, Martin LW, Chen YDI, Surendran P, Drenos F, Cook JP, Auer PL, Chu AY, Giri A, Zhao W, Jakobsdottir J, Lin LA, Stafford JM, Amin N, Mei H, Yao J, Voorman A, Larson MG, Grove ML, Smith AV, Hwang SJ, Chen H, Huan T, Kosova G, Stitziel NO, Kathiresan S, Samani N, Schunkert H, Deloukas P, Li M, Fuchsberger C, Pattaro C, Gorski M, Kooperberg C, Papanicolaou GJ, Rossouw JE, Faul JD, Kardia SLR, Bouchard C, Raffel LJ, Uitterlinden AG, Franco OH, Vasan RS, O'Donnell CJ, Taylor KD, Liu K, Bottinger EP, Gottesman O, Daw EW, Giulianini F, Ganesh S, Salfati E, Harris TB, Launer LJ, Dörr M, Felix SB, Rettig R, Völzke H, Kim E, Lee WJ, Lee IT, Sheu WHH, Tsosie KS, Edwards DRV, Liu Y, Correa A, Weir DR, Völker U, Ridker PM, Boerwinkle E, Gudnason V, Reiner AP, van Duijn CM, Borecki IB, Edwards TL, Chakravarti A, Rotter JI, Psaty BM, Loos RJF, Fornage M, Ehret GB, Newton-Cheh C, Levy D, Chasman DI. Meta-analysis identifies common and rare variants influencing blood pressure and overlapping with metabolic trait loci. Nat Genet 2016; 48:1162-70. [PMID: 27618448 PMCID: PMC5320952 DOI: 10.1038/ng.3660] [Citation(s) in RCA: 201] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 08/05/2016] [Indexed: 11/08/2022]
Abstract
Meta-analyses of association results for blood pressure using exome-centric single-variant and gene-based tests identified 31 new loci in a discovery stage among 146,562 individuals, with follow-up and meta-analysis in 180,726 additional individuals (total n = 327,288). These blood pressure-associated loci are enriched for known variants for cardiometabolic traits. Associations were also observed for the aggregation of rare and low-frequency missense variants in three genes, NPR1, DBH, and PTPMT1. In addition, blood pressure associations at 39 previously reported loci were confirmed. The identified variants implicate biological pathways related to cardiometabolic traits, vascular function, and development. Several new variants are inferred to have roles in transcription or as hubs in protein-protein interaction networks. Genetic risk scores constructed from the identified variants were strongly associated with coronary disease and myocardial infarction. This large collection of blood pressure-associated loci suggests new therapeutic strategies for hypertension, emphasizing a link with cardiometabolic risk.
Collapse
Affiliation(s)
- Chunyu Liu
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
- Department of Biostatistics, School of Public Health, Boston University, Boston, Massachusetts, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Aldi T Kraja
- Division of Statistical Genomics, Department of Genetics and Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Nora Franceschini
- Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Kenneth Rice
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Alanna C Morrison
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston Texas, USA
| | - Yingchang Lu
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Stefan Weiss
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst Moritz Arndt University Greifswald, Greifswald, Germany
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, USA
| | - Walter Palmas
- Division of General Medicine, Columbia University Medical Center, New York, New York, USA
| | - Lisa W Martin
- George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Yii-Der Ida Chen
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, USA
| | - Praveen Surendran
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Fotios Drenos
- Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, London, UK
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol, UK
| | - James P Cook
- Department of Biostatistics, University of Liverpool, Liverpool, UK
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Paul L Auer
- Joseph J. Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, USA
| | - Audrey Y Chu
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Ayush Giri
- Vanderbilt Epidemiology Center, Vanderbilt Genetics Institute, Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Li-An Lin
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Jeanette M Stafford
- Division of Public Health Sciences, Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Najaf Amin
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
| | - Hao Mei
- Department of Data Science, School of Population Health, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Jie Yao
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, USA
| | - Arend Voorman
- Bill and Melinda Gates Foundation, Seattle, Washington, USA
| | - Martin G Larson
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
- Department of Biostatistics, School of Public Health, Boston University, Boston, Massachusetts, USA
- Department of Mathematics and Statistics, Boston University, Boston, Massachusetts, USA
| | - Megan L Grove
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston Texas, USA
| | - Albert V Smith
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Shih-Jen Hwang
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Han Chen
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Tianxiao Huan
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Gulum Kosova
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, Massachusetts, USA
| | - Nathan O Stitziel
- Division of Cardiology, Department of Medicine and Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sekar Kathiresan
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, Massachusetts, USA
| | - Nilesh Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, UK
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| | - Panos Deloukas
- Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Man Li
- Department of Epidemiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Christian Fuchsberger
- Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC), Bolzano, Italy (affiliated with the University of Lübeck, Lübeck, Germany)
| | - Cristian Pattaro
- Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC), Bolzano, Italy (affiliated with the University of Lübeck, Lübeck, Germany)
| | - Mathias Gorski
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - George J Papanicolaou
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Jacques E Rossouw
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Claude Bouchard
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | - Leslie J Raffel
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Oscar H Franco
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
| | - Ramachandran S Vasan
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
- Department of Preventive Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Christopher J O'Donnell
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
- Cardiology Section, Department of Medicine, Boston Veterans Administration Healthcare, Boston, Massachusetts, USA
- Cardiovascular Division, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, USA
| | - Kiang Liu
- Northwestern University School of Medicine, Chicago, Illinois, USA
| | - Erwin P Bottinger
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Omri Gottesman
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - E Warwick Daw
- Division of Statistical Genomics, Department of Genetics and Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Franco Giulianini
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Santhi Ganesh
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Elias Salfati
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tamara B Harris
- Laboratory of Epidemiology, Demography and Biometry, National Institute on Aging, US National Institutes of Health, Bethesda, Maryland, USA
| | - Lenore J Launer
- Neuroepidemiology Section, National Institute on Aging, US National Institutes of Health, Bethesda, Maryland, USA
| | - Marcus Dörr
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Stephan B Felix
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Rainer Rettig
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Institute of Physiology, University of Greifswald, Greifswald, Germany
| | - Henry Völzke
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- DZD (German Center for Diabetes Research), site Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Eric Kim
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, USA
| | - Wen-Jane Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - I-Te Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Wayne H-H Sheu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Institute of Medical Technology, National Chung-Hsing University, Taichung, Taiwan
- School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Krystal S Tsosie
- Vanderbilt Epidemiology Center, Vanderbilt Genetics Institute, Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Digna R Velez Edwards
- Vanderbilt Epidemiology Center, Vanderbilt Genetics Institute, Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yongmei Liu
- Epidemiology and Prevention Center for Genomics and Personalized Medicine Research, Wake Forest Baptist Medical Center, Winston-Salem, North Carolina, USA
| | - Adolfo Correa
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Uwe Völker
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst Moritz Arndt University Greifswald, Greifswald, Germany
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston Texas, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Alexander P Reiner
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Cornelia M van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
| | - Ingrid B Borecki
- Division of Statistical Genomics, Department of Genetics and Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Todd L Edwards
- Vanderbilt Epidemiology Center, Vanderbilt Genetics Institute, Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Aravinda Chakravarti
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, USA
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Medicine, Harbor-UCLA Medical Center, Torrance, California, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
- Department of Health Services, University of Washington, Seattle, Washington, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ruth J F Loos
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Myriam Fornage
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Georg B Ehret
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Cardiology, Geneva University Hospitals, Geneva, Switzerland
| | - Christopher Newton-Cheh
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Daniel Levy
- Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
98
|
Pankratz N, Schick UM, Zhou Y, Zhou W, Ahluwalia TS, Allende ML, Auer PL, Bork-Jensen J, Brody JA, Chen MH, Clavo V, Eicher JD, Grarup N, Hagedorn EJ, Hu B, Hunker K, Johnson AD, Leusink M, Lu Y, Lyytikäinen LP, Manichaikul A, Marioni RE, Nalls MA, Pazoki R, Smith AV, van Rooij FJA, Yang ML, Zhang X, Zhang Y, Asselbergs FW, Boerwinkle E, Borecki IB, Bottinger EP, Cushman M, de Bakker PIW, Deary IJ, Dong L, Feitosa MF, Floyd JS, Franceschini N, Franco OH, Garcia ME, Grove ML, Gudnason V, Hansen T, Harris TB, Hofman A, Jackson RD, Jia J, Kähönen M, Launer LJ, Lehtimäki T, Liewald DC, Linneberg A, Liu Y, Loos RJF, Nguyen VM, Numans ME, Pedersen O, Psaty BM, Raitakari OT, Rich SS, Rivadeneira F, Di Sant AMR, Rotter JI, Starr JM, Taylor KD, Thuesen BH, Tracy RP, Uitterlinden AG, Wang J, Wang J, Dehghan A, Huo Y, Cupples LA, Wilson JG, Proia RL, Zon LI, O’Donnell CJ, Reiner AP, Ganesh SK. Meta-analysis of rare and common exome chip variants identifies S1PR4 and other loci influencing blood cell traits. Nat Genet 2016; 48:867-76. [PMID: 27399967 PMCID: PMC5145000 DOI: 10.1038/ng.3607] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 06/03/2016] [Indexed: 12/15/2022]
Abstract
Hematologic measures such as hematocrit and white blood cell (WBC) count are heritable and clinically relevant. We analyzed erythrocyte and WBC phenotypes in 52,531 individuals (37,775 of European ancestry, 11,589 African Americans, and 3,167 Hispanic Americans) from 16 population-based cohorts with Illumina HumanExome BeadChip genotypes. We then performed replication analyses of new discoveries in 18,018 European-American women and 5,261 Han Chinese. We identified and replicated four new erythrocyte trait-locus associations (CEP89, SHROOM3, FADS2, and APOE) and six new WBC loci for neutrophil count (S1PR4), monocyte count (BTBD8, NLRP12, and IL17RA), eosinophil count (IRF1), and total WBC count (MYB). The association of a rare missense variant in S1PR4 supports the role of sphingosine-1-phosphate signaling in leukocyte trafficking and circulating neutrophil counts. Loss-of-function experiments for S1pr4 in mouse and s1pr4 in zebrafish demonstrated phenotypes consistent with the association observed in humans and altered kinetics of neutrophil recruitment and resolution in response to tissue injury.
Collapse
Affiliation(s)
- Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Ursula M Schick
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Yi Zhou
- Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA, USA
| | - Wei Zhou
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Biology, University of Michigan, Ann Arbor, MI, USA
| | - Tarunveer Singh Ahluwalia
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center, Gentofte, Denmark
| | - Maria Laura Allende
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Paul L Auer
- School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Jette Bork-Jensen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Ming-Huei Chen
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, MA, USA
| | - Vinna Clavo
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - John D Eicher
- National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, MA, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Niels Grarup
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Elliott J Hagedorn
- Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA, USA
| | - Bella Hu
- Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA, USA
| | - Kristina Hunker
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Andrew D Johnson
- National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, MA, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Maarten Leusink
- Division Pharmacoepidemiology & Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Yingchang Lu
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Genetics of Obesity and Related Metabolic Traits Program, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories and University of Tampere School of Medicine, Tampere, Finland
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Riccardo E Marioni
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Raha Pazoki
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Albert Vernon Smith
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Frank J A van Rooij
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Min-Lee Yang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Xiaoling Zhang
- National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, MA, USA
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Yan Zhang
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Folkert W Asselbergs
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht, Netherlands
- Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, Netherlands
- Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, United Kingdom
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Ingrid B Borecki
- Department of Genetics, Division of Statistical Genomics, Washington University School of Medicine, St. Louis, MO, USA
| | - Erwin P Bottinger
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mary Cushman
- Department of Medicine, Division of Hematology/Oncology, University of Vermont, Burlington, VT, USA
| | - Paul I W de Bakker
- Department of Medical Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Liguang Dong
- Jin Ding Street Community Healthy Center, Peking University Shougang Hospital, Beijing, China
| | - Mary F Feitosa
- Department of Genetics, Division of Statistical Genomics, Washington University School of Medicine, St. Louis, MO, USA
| | - James S Floyd
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Nora Franceschini
- Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Oscar H Franco
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Melissa E Garcia
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Megan L Grove
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Albert Hofman
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Rebecca D Jackson
- Division of Endocrinology, Diabetes, and Metabolism, Ohio State University, Columbus, OH, USA
| | - Jia Jia
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital and University of Tampere School of Medicine, Tampere, Finland
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and University of Tampere School of Medicine, Tampere, Finland
| | - David C Liewald
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Allan Linneberg
- Research Centre for Prevention and Health, Capital Region of Denmark, Copenhagen, Denmark
- Department of Clinical Experimental Research, Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yongmei Liu
- Center for Human Genetics, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Genetics of Obesity and Related Metabolic Traits Program, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vy M Nguyen
- Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA, USA
| | - Mattijs E Numans
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
- Public Health and Primary Care, Leiden University Medical Centre, Leiden, Netherlands
| | - Oluf Pedersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Services, University of Washington, Seattle, WA, USA
- Group Health Research Institute, Group Health Cooperative, Seattle, WA, USA
| | - 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
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Fernando Rivadeneira
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Amanda M Rosa Di Sant
- Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA, USA
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute, Torrance, CA, USA
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - John M Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Geriatric Medicine unit, University of Edinburgh, Edinburgh, UK
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute, Torrance, CA, USA
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | | | - Russell P Tracy
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Colchester, VT, USA
- Department of Biochemistry, University of Vermont College of Medicine, Colchester, VT, USA
| | - Andre G Uitterlinden
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jiansong Wang
- Chronic Diseases Research Center, Peking University Shougang Hospital, Beijing, China
| | - Judy Wang
- Department of Genetics, Division of Statistical Genomics, Washington University School of Medicine, St. Louis, MO, USA
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Yong Huo
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - L Adrienne Cupples
- National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Richard L Proia
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Leonard I Zon
- Stem Cell and Regenerative Biology Department, Harvard University, Cambridge, MA, USA
| | - Christopher J O’Donnell
- National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, MA, USA
- Cardiovascular Epidemiology and Human Genomics Branch, National Heart, Lung, and Blood Institute, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
- Cardiology Section, Department of Medicine, Boston Veteran’s Administration Healthcare, Boston, MA, USA
| | - Alex P Reiner
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA, USA
| | - Santhi K Ganesh
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
99
|
Wuttke M, Köttgen A. Insights into kidney diseases from genome-wide association studies. Nat Rev Nephrol 2016; 12:549-62. [PMID: 27477491 DOI: 10.1038/nrneph.2016.107] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Over the past decade, genome-wide association studies (GWAS) have considerably improved our understanding of the genetic basis of kidney function and disease. Population-based studies, used to investigate traits that define chronic kidney disease (CKD), have identified >50 genomic regions in which common genetic variants associate with estimated glomerular filtration rate or urinary albumin-to-creatinine ratio. Case-control studies, used to study specific CKD aetiologies, have yielded risk loci for specific kidney diseases such as IgA nephropathy and membranous nephropathy. In this Review, we summarize important findings from GWAS and clinical and experimental follow-up studies. We also compare risk allele frequency, effect sizes, and specificity in GWAS of CKD-defining traits and GWAS of specific CKD aetiologies and the implications for study design. Genomic regions identified in GWAS of CKD-defining traits can contain causal genes for monogenic kidney diseases. Population-based research on kidney function traits can therefore generate insights into more severe forms of kidney diseases. Experimental follow-up studies have begun to identify causal genes and variants, which are potential therapeutic targets, and suggest mechanisms underlying the high allele frequency of causal variants. GWAS are thus a useful approach to advance knowledge in nephrology.
Collapse
Affiliation(s)
- Matthias Wuttke
- Division of Genetic Epidemiology, Institute for Medical Biometry and Statistics, Faculty of Medicine, and Medical Centre - University of Freiburg, Berliner Allee 29, 79110 Freiburg, Germany.,Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany
| | - Anna Köttgen
- Division of Genetic Epidemiology, Institute for Medical Biometry and Statistics, Faculty of Medicine, and Medical Centre - University of Freiburg, Berliner Allee 29, 79110 Freiburg, Germany.,Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany.,Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, Maryland, USA
| |
Collapse
|
100
|
Miliku K, Vogelezang S, Franco OH, Hofman A, Jaddoe VWV, Felix JF. Influence of common genetic variants on childhood kidney outcomes. Pediatr Res 2016; 80:60-6. [PMID: 26959481 PMCID: PMC5496666 DOI: 10.1038/pr.2016.44] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 12/15/2015] [Indexed: 11/09/2022]
Abstract
BACKGROUND Kidney measures in early life are associated with kidney disease in later life. We hypothesized that these associations are partly explained by common genetic variants that lead to both smaller kidneys with lower kidney function in early childhood and kidney disease in adulthood. METHODS We examined in a population-based prospective cohort study among 4,119 children the associations of a weighted genetic risk score combining 20 previously identified common genetic variants related to adult eGFRcreat with kidney outcomes in children aged 6.0 years (95% range 5.7-7.8). Childhood kidney outcomes included combined kidney volume, glomerular filtration rate (eGFR) based on creatinine levels, and microalbuminuria based on albumin and creatinine urine levels. RESULTS We observed that the genetic risk score based on variants related to impaired kidney function in adults was associated with a smaller combined kidney volume (P value 3.0 × 10(-3)) and with a lower eGFR (P value 4.0 × 10(-4)) in children. The genetic risk score was not associated with microalbuminuria. CONCLUSION Common genetic variants related to impaired kidney function in adults already lead to subclinical changes in childhood kidney outcomes. The well-known associations of kidney measures in early life with kidney disease in later life may at least be partly explained by common genetic variants.
Collapse
Affiliation(s)
- Kozeta Miliku
- The Generation R Study Group, Erasmus MC, University Medical Center, Rotterdam, the Netherlands,Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands,Department of Pediatrics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Suzanne Vogelezang
- The Generation R Study Group, Erasmus MC, University Medical Center, Rotterdam, the Netherlands,Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands,Department of Pediatrics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Oscar H. Franco
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Vincent WV Jaddoe
- The Generation R Study Group, Erasmus MC, University Medical Center, Rotterdam, the Netherlands,Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands,Department of Pediatrics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Janine F Felix
- The Generation R Study Group, Erasmus MC, University Medical Center, Rotterdam, the Netherlands,Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands,Department of Pediatrics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|